ArticlePDF Available

Impaired detection of omicron by SARS-CoV-2 rapid antigen tests

Authors:

Abstract and Figures

Since autumn 2020, rapid antigen tests (RATs) have been implemented in several countries as an important pillar of the national testing strategy to rapidly screen for infections on site during the SARS-CoV-2 pandemic. The current surge in infection rates around the globe is driven by the variant of concern (VoC) omicron (B.1.1.529). Here, we evaluated the performance of nine SARS-CoV-2 RATs in a single-centre laboratory study. We examined a total of 115 SARS-CoV-2 PCR-negative and 166 SARS-CoV-2 PCR-positive respiratory swab samples (101 omicron, 65 delta (B.1.617.2)) collected from October 2021 until January 2022 as well as cell culture-expanded clinical isolates of both VoCs. In an assessment of the analytical sensitivity in clinical specimen, the 50% limit of detection (LoD50) ranged from 1.77 × 10 ⁶ to 7.03 × 10 ⁷ RNA copies subjected to the RAT for omicron compared to 1.32 × 10 ⁵ to 2.05 × 10 ⁶ for delta. To score positive in these point-of-care tests, up to 10-fold (LoD50) or 101-fold (LoD95) higher virus loads were required for omicron- compared to delta-containing samples. The rates of true positive test results for omicron samples in the highest virus load category (Ct values < 25) ranged between 31.4 and 77.8%, while they dropped to 0–8.3% for samples with intermediate Ct values (25–30). Of note, testing of expanded virus stocks suggested a comparable RAT sensitivity of both VoCs, questioning the predictive value of this type of in vitro-studies for clinical performance. Given their importance for national test strategies in the current omicron wave, awareness must be increased for the reduced detection rate of omicron infections by RATs and a short list of suitable RATs that fulfill the minimal requirements of performance should be rapidly disclosed.
Content may be subject to copyright.
Vol.:(0123456789)
1 3
Medical Microbiology and Immunology (2022) 211:105–117
https://doi.org/10.1007/s00430-022-00730-z
ORIGINAL ARTICLE
Impaired detection ofomicron bySARS‑CoV‑2 rapid antigen tests
AndreasOsterman1 · IrinaBadell1· ElifBasara1· MarcelStern1 · FabianKriesel1· MarwaEletreby1·
GamzeNazÖztan1· MelanieHuber1· HannaAutenrieth1· RicardaKnabe1· PatriciaM.Späth1·
MaximilianMuenchho1,2,3 · AlexanderGraf4 · StefanKrebs4 · HelmutBlum4 · JürgenDurner5,6·
LudwigCzibere5 · ChristopherDächert1,2 · LarsKaderali7 · Hanna‑MariBaldauf1 · OliverT.Keppler1,2,3
Received: 9 February 2022 / Accepted: 12 February 2022 / Published online: 20 February 2022
© The Author(s) 2022
Abstract
Since autumn 2020, rapid antigen tests (RATs) have been implemented in several countries as an important pillar of the
national testing strategy to rapidly screen for infections on site during the SARS-CoV-2 pandemic. The current surge in
infection rates around the globe is driven by the variant of concern (VoC) omicron (B.1.1.529). Here, we evaluated the
performance of nine SARS-CoV-2 RATs in a single-centre laboratory study. We examined a total of 115 SARS-CoV-2 PCR-
negative and 166 SARS-CoV-2 PCR-positive respiratory swab samples (101 omicron, 65 delta (B.1.617.2)) collected from
October 2021 until January 2022 as well as cell culture-expanded clinical isolates of both VoCs. In an assessment of the
analytical sensitivity in clinical specimen, the 50% limit of detection (LoD50) ranged from 1.77 × 106 to 7.03 × 107 RNA cop-
ies subjected to the RAT for omicron compared to 1.32 × 105 to 2.05 × 106 for delta. To score positive in these point-of-care
tests, up to 10-fold (LoD50) or 101-fold (LoD95) higher virus loads were required for omicron- compared to delta-containing
samples. The rates of true positive test results for omicron samples in the highest virus load category (Ct values < 25) ranged
between 31.4 and 77.8%, while they dropped to 0–8.3% for samples with intermediate Ct values (25–30). Of note, testing of
expanded virus stocks suggested a comparable RAT sensitivity of both VoCs, questioning the predictive value of this type
of invitro-studies for clinical performance. Given their importance for national test strategies in the current omicron wave,
awareness must be increased for the reduced detection rate of omicron infections by RATs and a short list of suitable RATs
that fulfill the minimal requirements of performance should be rapidly disclosed.
Keywords SARS-CoV-2 rapid antigen test· Nucleocapsid protein· Diagnostic test· Sensitivity· Specificity· VoC· Lateral
flow
Edited by: Matthias J. Reddehase.
* Lars Kaderali
lars.kaderali@uni-greifswald.de
* Hanna-Mari Baldauf
baldauf@mvp.lmu.de
* Oliver T. Keppler
keppler@mvp.lmu.de
1 Max Von Pettenkofer Institute andGene Center, Virology,
National Reference Center forRetroviruses, LMU München,
Feodor-Lynen-Str. 23, 81377Munich, Germany
2 German Center forInfection Research (DZIF), Partner Site,
Munich, Germany
3 COVID-19 Registry oftheLMU Munich (CORKUM),
University Hospital, LMU Munich, Munich, Germany
4 Laboratory forFunctional Genome Analysis, Gene Center,
LMU München, Munich, Germany
5 Labor Becker MVZ GbR, Munich, Germany
6 Department ofConservative Dentistry andPeriodontology,
University Hospital, LMU München, Goethestr. 70,
80336Munich, Germany
7 Institute ofBioinformatics, University Medicine Greifswald,
Felix-Hausdorff-Str. 8, 17475Greifswald, Germany
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
106 Medical Microbiology and Immunology (2022) 211:105–117
1 3
Introduction
During the SARS-CoV-2 pandemic, new variants of the
virus continue to emerge. The variant of concern (VoC)
omicron was first detected in November 2021 in Southern
Africa. Omicron’s ability for immune escape [15] and
high transmissibility in the population [610] have resulted
in omicron’s rapid spread around the globe [11, 12] and
have fueled the pandemic with numbers of new infections
reaching all-time highs despite increasing COVID-19 vac-
cination rates. In early February 2022, numbers of mostly
unvaccinated, at-risk COVID-19 patients with omicron
that require hospitalization or even intensive care treat-
ment are high in some countries, including Israel and the
United States. Omicron is characterized by a large number
of mutations in the spike protein [13] and currently has
two prominent sub-lineages based on Pangolin nomencla-
ture, namely BA.1 and BA.2.
In many countries, rapid antigen tests (RATs) for the
detection of SARS-CoV-2 continue to be a central com-
ponent of national testing strategies offering quick, inex-
pensive and laboratory-independent, point-of-care diagnos-
tics. However, the evaluation by independent laboratories
[1425] and a Cochrane meta-analysis [26] have indicated
a highly variable performance of RATs resulting in an ongo-
ing controversy over these tests’ utility for the detection of
acute SARS-CoV-2-infected individuals in different settings
relevant for clinical diagnosis and containment strategies.
Some of these RATs have been approved for use by lay-
person and are frequently used as a tool for public health
interventions. In light of the somewhat limited capacities of
laboratory-based RT-qPCR for the quantitative detection of
SARS-CoV-2 RNA, test results from RATs alone are cur-
rently being suggested by national health authorities both to
reliably diagnose COVID-19 or to document an “uninfected”
or “non-infectious” status for ending quarantine or isolation
of persons without symptoms, respectively.
The nucleocapsid protein is most commonly detected
by SARS-CoV-2 antigen tests [13]. Since the majority
of RATs was developed prior to the emergence of VoCs,
the latter carrying different mutational patterns in the
nucleocapsid protein [19] (Suppl. Table1), it is of utmost
importance to perform VoC-specific RAT evaluations. In
omicron, besides more than 30 non-synonymous muta-
tions in the spike protein, the nucleocapsid protein of the
BA.1 sub-lineage carries four mutations compared to the
reference sequence of the Wuhan-hu-1 virus, i.e. P13L,
DEL31/33, R203K and G204R, and the additional muta-
tion S413R is present in BA.2 (Suppl. Table1). Three of
these mutations (P13L, DEL31/33 and S413R) are unique
to VoC omicron compared to alpha, beta, gamma or delta,
rendering predictions of the performance of RATs difficult.
Notably, first reports on the clinical and analytical per-
formance of RATs for omicron are partly contradictory: a
high-risk occupational case cohort of 30 individuals with
daily testing during an omicron outbreak in December 2021
showed a median of two days of apparently false-negative
RAT results with four recorded transmission events during
the period preceding the positive PCR result [27]. A preprint
study reports on 296 persons seeking COVID-19 testing at a
walk-up San Francisco community site in January 2022, who
tested positive by RT-qPCR. In these individuals, simultane-
ous nasal rapid antigen testing with BinaxNOW™ detected
95.2% of high viral load-omicron cases and 65.2% of all
PCR-positive cases [28]. A comparative study on isolates
classified as delta or omicron, that had been expanded in
tissue culture, found no VoC-specific differences in the ana-
lytical sensitivity of ten RATs, concluding the effectiveness
of these antigen lateral flow tests for omicron [29]. Other
investigators have suggested that an analytical and com-
parative validation with cultured VoCs might be a proxy
for clinical accuracy, but also stated that this cannot replace
clinical evaluations [14].
The aim of the current study was to compare nine dif-
ferent SARS-CoV-2 RATs that all detect the nucleocapsid
protein of SARS-CoV-2, for their analytical performance
using both clinical respiratory material and cultured viruses.
We evaluated respiratory specimen, identified as either delta
or omicron that had been collected from patients seen at dif-
ferent hospitals, nursing homes, outpatient clinics, COVID-
19 testing centers or seen by primary care physicians in the
Greater Munich area during the fourth and fifth pandemic
wave in Germany in the period October 2021 until January
2022. In addition, dilution series of isolates of delta and
omicron that had been expanded in cell culture in a biosafety
level 3 laboratory, were assessed.
Materials andmethods
Respiratory swabs
In the period October 30, 2021 to January 17, 2022 (delta)
and November 26, 2021 to January 19, 2022 (omicron),
health care professionals collected respiratory swabs (naso-
pharyngeal or nasal) from individuals who were seen on
clinical units or in the emergency room of the LMU Klini-
kum, the second-largest University Hospital in Germany, and
three teaching hospitals of the LMU Munich (Helios Amper
Hospital Dachau, Helios Hospital München West and Helios
Hospital München Perlach). Furthermore, SARS-CoV-2 in
respiratory swabs identified as omicron by Labor Becker
MVZ GbR in Munich, Germany, a diagnostic laboratory
that receives samples from regional hospitals, COVID-19
testing centers, nursing homes and primary care physicians'
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
107Medical Microbiology and Immunology (2022) 211:105–117
1 3
offices, completed the study panel. All samples from this
period were randomly included in the study depending on
the availability of sufficient sample volume. No informa-
tion on the persons’ vaccination status, previous infections,
symptoms or clinical course were available. For this study,
flocked swabs were collected in different liquid transport
media and analyzed by RT-qPCR for SARS-CoV-2 RNA.
Dry swabs were resuspended in sterile 0.9% NaCl. Patient
samples in liquid transport medium with denaturing poten-
tial were excluded from the study. All samples with a Cp/
Ct value < 40 by RT-qPCR, which was quantified less than
24h after sample collection and under accredited diagnostic
laboratory conditions, were scored “SARS-CoV-2-positive”.
Original respiratory swabs and transport media were stored
at 2–8°C for up to 1week. If longer storage periods were
required, samples were frozen at − 20°C and thawed once
before antigen testing was performed. 166 SARS-CoV-
2-PCR-positive and 115 PCR-negative respiratory samples
were analyzed in this study.
SARS‑CoV‑2 antigen tests
The methodological procedure for the evaluation of the
RATs in this study follows the publication on the com-
parative sensitivity evaluation for CE-marked rapid diag-
nostic tests for SARS-CoV-2 antigen as "corresponding to
the current state-of-the-art" of the Paul-Ehrlich-Institute
[21]. For testing a particular specimen, a 50µl aliquot
was added and completely absorbed using the sampling
devices (swab) supplied with the respective kit. This cor-
responds to a spike-in after step 1 in Fig.1. Swabs were
then eluted in the test-specific buffer, following the respec-
tive manufacturer's instructions. After application of the
specimen/buffer solution to the test cassette, visual read-
ing of the control and target lines was performed after
a 15min-incubation period. The appearance of the RDT
control line was mandatory for a valid test result. Invalid
tests were documented and repeated if possible. Any vis-
ible colored band in the test area was considered a positive
result regardless of the intensity of the band. Scoring the
RAT was performed under constant lighting conditions
and by a trained person who was blinded to the preceding
RT-qPCR result of the sample. This procedure deviates in
part from manufacturer specifications regarding the use of
liquid transport medium rather than direct swabs and the
fact that the sample was not immediately inserted into the
assay. These study design-related adaptations in the pro-
tocol are unlikely to affect the comparative assessment of
the RATs for VoCs delta and omicron. For comparability,
this study reports the number of SARS-CoV-2 RNA copies
subjected to each test (determined by absolute quantifica-
tion of RT-qPCR—see below).
123
Extraction bufferSwab Positive test result
TC
represents > 1 Mio. RNA equivalents
Fig. 1 Schematic overview of the nasal swab sampling and testing
procedure. Step 1: viral particles are collected with the provided col-
lecting device (swab) from the anterior part of the nasal cavity. In
general, the test sensitivity is influenced by the amount of nucleocap-
sid antigen collected with the swab from nasal mucosa and secretions.
Step 2: antigen-containing sample material is eluted from the swab
into an extraction buffer. The sensitivity of the assay can be affected
by the efficiency of the physical elution of the antigen from the col-
lection device. Step 3: extracted nucleocapsid antigen is applied onto
the rapid antigen test cassette. In this step, manufacturer-specific rec-
ommendations for the volume of antigen-containing buffer that are
applied to the test cassette (known as “input ratio”) exist. The amount
of “RNA copies subjected to assay” investigated in this study refers to
an input equivalent at step 1
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
108 Medical Microbiology and Immunology (2022) 211:105–117
1 3
Eight of the RATs examined in this study are recom-
mended by the manufacturers for self-testing and were listed
as a CE-labeled antigen test by the Bundesinstitut für Arz-
neimittel und Medizinprodukte (BfArM) in Germany at the
time of purchase, fulfilling the minimum criteria required for
these test devices in Germany according to the information
provided by the manufacturers. Of note, these eight assays
investigated were among the first rapid SARS-CoV-2 antigen
tests approved by BfarM for lay use in March 2021.
In detail, the products and manufacturers are: for test
1: FUJIFILM COVID-19 Ag Test (Fujifilm Cooperation),
research-use-only kits were provided for this study by the
manufacturer. Except for the labels, these are identical to the
final product, which meets the above criteria and was intro-
duced after approval. Test 2: Novel Coronavirus 2019-nCoV
Antigen Test (Colloidal gold) (Beijing Hotgen Biotech Co.,
Ltd.); test 3: NanoRepro SARS-CoV-2 Antigen Schnelltest
(Viromed) (NanoRepro AG); test 4: CLINITEST Rapid
COVID-19 Antigen Test (Healgen Scientific LLC); test 5:
Lyher Novel Coronavirus (COVID-19) Antigen Test Kit
(Colloidal Gold) (Hangzhou Laihe Biotech Co., Ltd.); test
6: COVID-19 Ag BSS self-test (Biosynex Swiss SA); test 7:
rapid SARS-CoV-2 Antigen Test Card (Xiamen Boson Bio-
tech Co., Ltd.); test 8: rapid SARS-CoV-2 Antigen Test Card
(MP Biomedicals Germany GmbH) and test 9: Medicovid-
AG SARS-CoV-2 Antigen Rapid Test Card-nasal (Xiamen
Boson Biotech Co., Ltd.). Tests 7 and 9 appear to be the
same antigen cassette product from one manufacturer (Ref
No.: 1N40C5 and 1N40C5-4), but they may differ in other
kit components (swab and buffer) in addition to package
size. Eight of these nine RATs have been evaluated by the
Paul-Ehrlich-Institute [21] and the European Commission
[30]. An overview of the RATs used in the current study and
their characteristics is provided in Suppl. Table2.
Quantitative viral load determination
In the accredited routine diagnostics laboratory of the
Max von Pettenkofer Institute, the following RT-qPCR
assays were used and quantified as described previously
[19] using the respective formulas: the nucleocapsid
(N1) reaction (Center for Disease Control (CDC) proto-
col [31] (
x=e
(
y
48.597)
1.461 ) on a LightCycler 480 system, the
Roche Cobas SARS-CoV-2 E-Gen reaction on a Cobas
6800 system (
x=e
(
y
44.576)
1.401 ) or the Xpert Xpress SARS-
CoV-2 (
x=e
(
y
50.859)
1.887 ), Xpert Xpress SARS-CoV-2/Flu/
RSV (
x=e
(
y
45.904)
1.5 ) and Xpert Xpress CoV-2/Flu/RSVplus
(
x=e
(
y
46.747)
1.501 ) run on a GeneXpert System, with x equals Geq
per ml and y equals the Ct/Cq value. For nucleic acid extrac-
tion, the QIAsymphony DSP Virus/Pathogen Kit was used
with the QIAsymphony instrument from QIAGEN GmbH.
In general, the calculations for quantification do not take
into account variability between separate RT-qPCR runs.
However, since this variability applies to all study groups,
they do not affect the interpretation of the results in this
study.
SARS‑CoV‑2 variant‑specific PCR andwhole‑genome
sequencing
Protocols for SARS-CoV-2 variant-specific PCR have been
reported [32]. In brief, nucleic acids from patients’ respira-
tory samples were extracted and eluates used for melting
curve analyses using commercially available PCR kits from
two vendors. For whole genome sequencing, amplicon pools
covering the SARS-CoV-2 genome were prepared accord-
ing to the ARTIC network nCoV-2019 sequencing protocol
v2 and analyzed utilizing the Artic bioinformatics protocol
[33]. The consensus sequences and associated sample meta-
data were uploaded to the GISAID repository.
Expansion ofSARS‑CoV‑2 frompatient material
SARS-CoV-2 was expanded as reported [5]. In brief, for
generation of high titer SARS-CoV-2 virus stocks, Vero-E6
cells were challenged with respiratory specimens containing
either delta or omicron. Virus stocks were harvested three
days post infection when approximately 50% of cells were
detached displaying a strong cytopathic effect. Supernatants
were cleared and stored at −80°C. Expanded stocks were
analyzed by whole genome sequencing (B.1.617.2: GISAID
3233464; B.1.1.529: GISAID 7808190) and RNA copies per
mL were determined as the mean from three independent
biological experiments with technical unicates or duplicates.
Statistical analyses
Statistical analysis was performed in R version 4.1.2. Bino-
mial confidence intervals for sensitivities and specificities
were computed using the Wilson score interval. To further
analyze analytical sensitivities, we used logistic regression,
with viral loads and RNA copy numbers subjected to the test
as independent and test outcomes as the dependent variable,
yielding detection probabilities for each viral load level.
Results
Evaluation ofRAT specificity
To evaluate the specificity of all RATs in a comparable
experimental setting (Fig.1), test collection devices (swabs)
were individually incubated with 50µl each of the individual
115 PCR-negative nasal/nasopharyngeal swabs and applied
to the individual assay’s extraction buffer. Under these
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
109Medical Microbiology and Immunology (2022) 211:105–117
1 3
conditions, the specificity of all nine RATs was 100% (CI
96.3–100%) (Suppl. Table3).
Characterization ofrespiratory specimens
fromCOVID‑19 patients containing VoCs delta
oromicron
Next, we quantified the viral load in PCR-positive naso-
pharyngeal swabs, of which 65 were classified as delta
(B.1.617.2) and 101 as omicron (B.1.1.529), respectively, by
variant-specific PCR and next-generation sequencing [32].
The omicron sequences in all respiratory swabs were clas-
sified as belonging to the BA.1 sublineage. The viral loads
of delta specimen ranged from 1.74 × 104 to 6.72 × 108 Geq/
ml (median 6.67 × 106 Geq/ml, interquartile range 6.61 × 105
– 5.14 × 107 Geq/ml, median absolute deviation 9.73 × 106
Geq/ml) (Fig.2a, b, e), the viral loads of omicron speci-
men ranged from 9.3 × 102 to 2.26 × 109 Geq/ml (median
6.97 × 106 Geq/ml, interquartile range 9.45 × 105–5.14 × 107
Geq/ml, median absolute deviation 1.02 × 107 Geq/ml)
OmicronDelta
9
7
5
3
log10 viral load (Geq/ml)
6
10
2
0
10
20
30
2610
48
0
10
20
2610
48
log10 viral load (Geq/ml)
Number of samples
040 10020 60 80
log10 viral load (Geq/ml)
Number of samples
04020 60
Frequency
log10 viral load (Geq/ml)
Frequency
log10 viral load (Geq/ml)
e
ab
cd
n.s.
Delta
Omicron
6
10
2
Fig. 2 SARS-CoV-2 viral load distribution of respiratory samples
included in the study containing either delta or omicron. a, c Shown
is the log10 viral load (Geq/ml) distribution of all 65 delta (a) and
all 101 omicron (c) sorted by ascending magnitude from left to right.
Each dot indicates one patient and the sample ID is indicated. b, d
Histogram of the viral load distribution in specimen containing delta
(b) or omicron (d) categorized into the indicated ranges of log10 viral
load. Each bar depicts the number of samples in the respective viral
load range. e The horizontal line in the box plots shows the median of
the samples shown in a and c, bound between upper and lower quar-
tiles, and whiskers between minimum and maximum are indicated.
n.s. = not significant by Wilcoxon rank sum test with continuity cor-
rection and by two-sample Kolmogorov–Smirnov test
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
110 Medical Microbiology and Immunology (2022) 211:105–117
1 3
(Fig.2c, d, e). Importantly, the median and distribution of
viral loads were comparable and statistically indistinguish-
able between both groups of VoC-containing respiratory
samples (Fig.2e), allowing a direct comparison of the ana-
lytical sensitivity of RATs for the detection of these two
most recently emerged VoCs.
Comparative analysis ofanalytical RAT sensitivity
forVoCs delta andomicron inclinical specimens
We used this panel of SARS-CoV-2 RT-qPCR-positive res-
piratory specimen to determine the analytical sensitivity
of the nine RATs for the detection of delta and omicron
in a direct comparison. The overall sensitivities for delta-
containing specimen ranged between 34.92 and 58.46%
(Table1), corresponding to 22 of 63 and 38 of 65 samples,
respectively, that were tested true positive. For omicron-
containing specimens, the overall sensitivities ranged from
22.22 to 57.43% (Table2), corresponding to 22 of 99 and
58 of 101 SARS-CoV-2 PCR-positive samples, respectively,
that were correctly scored.
We determined the 50% (dotted line in pink vertical area)
and 95% (dotted line in yellow vertical area) limits of detec-
tion (LoD) based on a logistic regression model as recently
reported [16] (Fig.3a, b). For test 1, the RNA copy numbers
for LoD50 and LoD95 were 1.19 × 106 and 7.03 × 107 for delta-
containing specimen, respectively. The detection of omicron-
containing samples was in contrast impaired: the LoD50 and
LoD95 values corresponded to 1.11 × 107 and 7.12 × 109 RNA
copies, respectively, thus requiring 9- to 101-fold higher RNA
copy numbers than those for delta-containing specimens to
reach these detection thresholds. For test 2, the LoD50 and
LoD95 values for delta-containing samples corresponded to
1.54 × 105 and 3.86 × 106 RNA copy numbers, respectively.
Those for omicron-containing specimens were 9- to 17-fold
higher with 1.38 × 106 and 6.64 × 107 RNA copy numbers,
respectively. The performance of test 3 was comparable to test
2 with LoD50 and LoD95 values for delta-containing sam-
ples of 2.05 × 105 and 5.04 × 106 RNA copy numbers, respec-
tively, and for omicron-containing samples of 2.12 × 106 and
1.35 × 108 RNA copy numbers, respectively, i.e. ranging 10-
to 27-fold higher for the latter. The SARS-CoV-2 RNA copy
numbers corresponding to LoD50 and LoD95 for test 4 for
delta- versus omicron-containing specimens were 8- to ten-
fold higher for the latter with 1.77 × 105 and 4.52 × 106 versus
1.33 × 106 and 4.37 × 107, respectively. For test 5, the LoD50
and LoD95 values for delta-containing samples corresponded
to 1.34 × 105 and 3.08 × 106 RNA copies, respectively. These
LoD values were 3- and 5-times higher for omicron-containing
samples with 4.54 × 105 and 1.43 × 107 RNA copies, respec-
tively. The LoD50 and LoD95 values for test 6 were 1.82 × 105
and 3.02 × 106 for delta- and 6.93 × 105 and 2.08 × 107 for
omicron-containing specimen, respectively, corresponding to
a 4- to sevenfold difference. For test 7, the RNA copies with
50% and 95% detection rates determined for delta-containing
specimens were 3.31 × 105 and 1.77 × 106, respectively. Detec-
tion of omicron was similar to test 1 and strongly impaired
as LoD values were 8- to 99-fold higher with 2.62 × 106 and
1.75 × 108 RNA copies, respectively. The relative performance
of test 8 was among the best of the RATs tested in this study:
the LoD50 and LoD95 values for delta and omicron differed at
maximum by a factor of 2: values for delta-containing samples
corresponded to 1.32 × 105 (LoD50) and 3.86 × 106 (LoD95),
and for omicron samples 3.24 × 105 and 3.94 × 106 RNA cop-
ies, respectively. Test 9 performed similarly well in our analy-
sis: the LoD50 and LoD95 for delta samples were 1.82 × 105
and 3.02 × 106 RNA copies and for omicron samples 1.94 × 105
and 3.41 × 106, respectively.
Table 1 Determination of assay sensitivity for nine SARS-CoV-2
rapid antigen tests in SARS-CoV-2 PCR-positive respiratory swabs
classified as delta
Binomial confidence intervals were computed using the Wilson score
interval
Assay Sensitivity (%) 95% CI (%) True
positive/
total
Test 1 34.92 24.33–47.25 22/63
Test 2 56.25 44.09–67.71 36/64
Test 3 53.85 41.85–65.41 35/65
Test 4 55.38 43.34–66.83 36/65
Test 5 58.46 46.34–69.64 38/65
Test 6 55.38 43.34–66.83 36/65
Test 7 49.21 37.27–61.24 31/63
Test 8 58.46 46.34–69.64 38/65
Test 9 55.38 43.34–66.83 36/65
Table 2 Determination of assay sensitivity for nine SARS-CoV-2
rapid antigen tests in SARS-CoV-2 PCR-positive respiratory swabs
classified as omicron
Binomial confidence intervals were computed using the Wilson score
interval
Assay Sensitivity (%) 95% CI (%) True
positive/
total
Test 1 22.22 15.16–31.36 22/99
Test 2 35.64 26.99–45.35 36/101
Test 3 31.68 23.42–41.29 32/101
Test 4 35.64 26.99–45.35 36/101
Test 5 47.52 38.06–57.18 48/101
Test 6 43.00 33.73–52.78 43/100
Test 7 29.70 21.67–39.23 30/101
Test 8 51.49 41.86–61.00 52/101
Test 9 57.43 47.69–66.62 58/101
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
111Medical Microbiology and Immunology (2022) 211:105–117
1 3
In summary, a reduction in the analytical sensitivity for
the detection of omicron- compared to delta-containing res-
piratory specimens, determined by the LoD50 and LoD95,
was seen for eight of the nine RATs with factors of up to
10.3-fold and up to 101-fold more virus genome equiva-
lents, respectively, required to score positive, reaching sta-
tistical significance for tests 2, 3, 4 and 7. This suggests
a general impairment for a virus load-stratified detection
of an omicron infection compared to a delta infection by
SARS-CoV-2 antigen testing with a RAT-specific degree
of sensitivity reduction.
Analytical RAT sensitivity incell culture‑expanded
virus stocks
Based on an initial report of comparable sensitivity for
invitro-expanded virus stocks [29], we assessed in paral-
lel cell-culture expanded delta and omicron stocks. The
expanded delta isolate (GISAID 3233464) carries the D63G,
R203M and D377Y mutations in the nucleocapsid protein
as well as the commonly reported G215C mutation [34].
The expanded omicron isolate (GISAID 7808190; BA.1 sub-
lineage) carries the expected P13L, del31/33, R203K and
G204R mutations. Neither culture medium alone nor the
virus-inactivating detergent Triton X-100 had an impact on
the performance of the RATs (not shown).
As shown in Suppl. Figure1, rapid antigen tests 1, 5, 8
and 9 were able to detect delta and omicron virus stocks
down to 2.5 × 106 RNA copies, whereas the other five tests
were less sensitive requiring up to 8-times more SARS-
CoV-2 RNA to score positive. Interestingly, there was a
trend for a slightly better detection of cell-culture expanded
omicron compared to delta, in particular for tests 2, 4, 6 and
7. In summary, this analysis confirms first studies by other
laboratories [14, 35] indicating a comparable detection of
a
b
0
50
100
0
50
100
0
50
100
0
50
100
Test 6
Test 5Test 1Test 2
Test 7
Test 3
Test 8
Test 4
Test 9
Probability (%)Probability (%) Probability (%)Probability (%)
Delta
Omicron
Omicron
Delta
8624
log
10
RNA copies
subjected to test
log
10
RNA copies
subjected to test
log
10
RNA copies
subjected to test
log
10
RNA copies
subjected to test
10 8624 10 8624 10 8624 10
log
10
RNA copies
subjected to test
8624 108624
log
10
RNA copies
subjected to test
log
10
RNA copies
subjected to test
log
10
RNA copies
subjected to test
log
10
RNA copies
subjected to test
10 8624 10 8624 10 8624 10
Fig. 3 Limit of detection analyses of respiratory samples positive for
either delta (top panels) or omicron (bottom panels) by RT-qPCR for
nine SARS-CoV-2 RATs. a test #1–5, b test #6–9. The log10 RNA
copies subjected to the test of quantified samples on the x axis were
plotted against a positive (+ 1) or negative (0) test outcome on the y
axis. For readability of the figure, slight normal jitter was added to
the y values. Blue (delta) and red (omicron) curves, respectively,
show logistic regressions of the viral load on the test outcome; verti-
cal dashed lines indicate log10 RNA copies subjected to the test at
which 50% (LoD50) and 95% (LoD95), respectively, of the samples
are expected positive based on the regression results. Significant
differences for LoD50: test 2, 3, 4 and 7. Significant difference for
LoD95: test 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
112 Medical Microbiology and Immunology (2022) 211:105–117
1 3
invitro-expanded omicron and delta virus stocks by rapid
antigen testing. However, this stands in clear contrast to the
above evaluation of clinical respiratory specimens, which
show an impaired sensitivity for the detection of an omicron
infection in COVID-19 patients with comparable viral loads
in respiratory specimens.
Comparative, Ct value‑stratified evaluation
ofanalytical RAT sensitivity
Next, we sought to directly compare the analytical sensi-
tivities of the nine RATs for the detection of “non-delta and
non-omicron” SARS-CoV-2, evaluated and published by the
Paul-Ehrlich-Institute, to VoCs delta or omicron assessed in
our study in respiratory specimen in predefined corridors of
SARS-CoV-2 RNA copy numbers. To this end, we harmo-
nized the corresponding Ct/Cp values between our study
results and those reported by Puyskens etal. and Scheiblauer
etal. [21, 36]. This allowed us to plot and compare the per-
centages of test samples that scored positive in one of the
three categories of Ct/Cp values, i.e. < 25, 25–30 and > 30.
The following observations were made (Table3): (1) for
the seven RATs, for which all three variant data sets were
available, the overall sensitivity of the “non-delta and non-
omicron” SARS-CoV-2 scored highest in four RATs, closely
followed by three RATs for delta. (2) In eight out of nine
RATs, the overall sensitivity was higher for delta- compared
to omicron-containing respiratory samples. (3) In the highest
viral load category with Ct values < 25, omicron detected the
lowest percentage of cases in eight out of nine RATs with
test 9 posing the exception. (4) In the intermediate viral load
corridor reflected by Ct values 25–30, none of the respira-
tory specimen scored positive in six of the RATs, and with
low percentages of 4.2–8.3% for tests 5, 8 and 9. (5) Samples
with Ct values > 30 were generally not detected with a single
exception for the “non-delta and non-omicron” specimen
group (Table3). In summary, these results indicate that the
analytical sensitivity of RATs for omicron-containing respir-
atory samples cannot be deduced from previous evaluations
of other SARS-CoV-2 variants. For the majority of RATs
evaluated in this study, an impaired detection of the emer-
gent VoC omicron compared to previous variants, including
delta, was noted.
Discussion
In the current study, we evaluated the performance of nine
commercially available SARS-CoV-2 raid antigen tests that
were launched on the European market during the corona-
virus pandemic for the detection of the viral nucleocapsid
protein. We find that for the majority of RATs evaluated,
detection of omicron is impaired compared to delta, requir-
ing up to 10.3-fold and up to 101-fold more SARS-CoV-2
RNA of omicron in respiratory specimens to score positive
in LoD50 or LoD95 analyses, respectively.
We examined two groups of specimens: the first group
encompassed respiratory samples collected from patients in
the period October 2021 until January 2022. A total of 115
SARS-CoV-2 PCR-negative and 166 SARS-CoV-2 PCR-
positive specimen (65 delta, 101 omicron) were tested. The
second group consisted of cell culture-expanded clinical
isolates of these VoCs. The latter was important to assess
the predictive value of sensitivity evaluations of RATs using
SARS-CoV-2 variants expanded in tissue culture as a poten-
tially rapid proxy for their diagnostic performance.
Limitations of the current study include the lack of infor-
mation on the vaccination status, previous infections, symp-
toms or stage of COVID-19 of individuals from whom the
respiratory swabs were taken. We focused on the analytical
sensitivity in relation to the viral loads in respiratory swabs
to allow a direct comparison of specimens containing omi-
cron and delta (the current study) and “non-omicron/non-
delta” SARS-CoV-2 (studies by the Paul-Ehrlich-Institute
[21, 36]) in the assessment of RAT performance.
A potential confounder in the methodological procedure
for specificity assessment is the rather small volume of
biological material from the original swab that is actually
transferred to the RAT extraction buffer. Here, swab kits
containing 1ml virus transport medium were used, result-
ing up front in a 1:20 dilution of the input sample. This may
have potentially reduced biological components that may
trigger a false-positive result. Therefore, the RATs’ specific-
ity of 100% observed in the current study is only informative
for the chosen experimental setting to evaluate the analytical
sensitivity, but is unlikely to reflect the specificity of the
investigated RATs in a point-of-care setting.
The reasons underlying the discrepancy of the RAT per-
formance comparing delta and omicron between respiratory
samples, on the one hand, and expanded virus stocks, on the
other hand, are currently unclear. As proposed in a cryo-
electron tomography study [37], nucleocapsid assembly
of SARS-CoV-2 is driven by the intracellular interaction
between the nucleocapsid protein and the genomic viral
RNA. In a clinical setting, our data suggest that the relative
ratio of nucleocapsid protein to SARS-CoV-2 RNA may be
higher, on average, in the extracellular space and on the res-
piratory mucosa of COVID-19 patients infected with delta
compared to those infected with omicron.
Several scenarios can be envisioned that may under-
lie this difference: (1) a variant-specific degree of virus-
induced cell death or cytolysis triggered by the host’s
immune response may contribute to a VoC-specific level
of nucleocapsid protein found on the respiratory mucosa.
(2) A higher severity of disease has been associated with
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
113Medical Microbiology and Immunology (2022) 211:105–117
1 3
increased transcriptional levels of nucleocapsid RNA
[38], providing a potential explanation for an enhanced
nucleocapsid per viral RNA ratio in COVID-19 as a
consequence of a delta infection compared to an omi-
cron infection. (3) Tissue culture-expanded virus stocks
are harvested at a time point of considerable lysis of the
Table 3 Comparative evaluation
of the analytical sensitivity of
nine SARS-CoV-2 rapid antigen
tests stratified for Ct/Cp value
ranges based on studies by the
Paul-Ehrlich-Institute (non-
delta/non-omicron*) and the
current study for respiratory
samples containing VoCs delta
and omicron
* Data published by Puyskens etal. (collected between March and September 2020 (Panel 1V1; [36]) and
Scheiblauer etal. (additionally collected between October 2020 and January 2021 (Panel 1V2); [21]) and
“SARS-CoV-2 antigen rapid diagnostic tests passing the sensitivity criteria” of the Paul-Ehrlich-Institute
(date of access: 31.1.22)
1 The Ct values of test specimens in the study performed by the Paul-Ehrlich-Institute and our study were
harmonized to allow a direct comparison of data sets. Harmonization was achieved based on the publica-
tion by Scheiblauer etal. and the formula derived to convert virus loads into Ct values:
y = − 1.455lnx + 41.154 with x equals Geq per ml and y equals the Ct/Cq value. n.a. not available;– these
test results were no longer included in the latest data set released by the Paul-Ehrlich-Institute on January
31, 2022
NCt < 25
(%)1Ct 25–30
(%)1Ct > 30
(%)1Overall
sensitivity
(%)
Test 1
Non-Delta/non-Omicron* n.a 85.0 10.0 0 38.0
Delta 63 50.0 5.6 0 34.9
Omicron 99 31.4 0 0 22.2
Test 2
Non-Delta/non-Omicron* n.a 100 47.8 0 56.0
Delta 64 74.4 22.2 0 56.2
Omicron 101 50.0 0 0 35.6
Test 3
Non-Delta/non-Omicron* n.a 94.1
Delta 65 72.7 16.7 0 53.9
Omicron 101 44.4 0 0 31.7
Test 4
Non-Delta/non-Omicron* n.a 100 87 0 76.0
Delta 65 72.7 22.2 0 55.4
Omicron 101 50.0 0 0 35.6
Test 5
Non-Delta/non-Omicron* n.a 94.4 17.4 0 42.0
Delta 65 77.3 22.2 0 58.5
Omicron 101 63.9 8.3 0 47.5
Test 6
Non-Delta/non-Omicron* n.a 100 78.3 11.1 74.0
Delta 65 75.0 16.7 0 55.4
Omicron 100 60.6 0 0 43.0
Test 7
Non-Delta/non-Omicron* n.a 100 43.5 0 54.0
Delta 63 72.1 0 0 49.2
Omicron 101 41.7 0 0 29.7
Test 8
Non-Delta/non-Omicron* n.a 100 43.5 0 54.0
Delta 65 75.0 27.8 0 58.5
Omicron 101 70.8 4.2 0 51.5
Test 9
Non-Delta/non-Omicron* n.a – – – –
Delta 65 75.0 16.7 0 55.4
Omicron 101 77.8 8.3 0 57.4
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
114 Medical Microbiology and Immunology (2022) 211:105–117
1 3
virus-producing Vero-E6 cells, potentially leveling out
VoC-specific differences in the ratio of released nucle-
ocapsid protein relative to viral RNA compared to a more
physiological setting. (4) Moreover, one can speculate that
SARS-CoV-2-specific antibody responses in COVID-19
patients, either through vaccination or previous infections,
could differentially impact on the VoC-specific positivity
rate of RATs.
It is widely communicated that individuals with an omi-
cron infection with high viral loads are reliably detected
by RATs that are available on the German market, and that
these tests’ quality is sufficient to have a positive impact
on national pandemic management [39, 40]. These general
claims stand in contrast to the following aspects: (1) among
the approximately 580 RATs [41] currently available on the
German market not even half has been evaluated by either
the Paul-Ehrlich-Institute or independent diagnostic labo-
ratories for any SARS-CoV-2 variant, let alone omicron.
The Paul-Ehrlich-Institute stated that first results of their
omicron validations will be made publically available at the
end of February 2022; (2) general claims that reactivity in a
RAT reliably identifies the group of “truly” infectious indi-
viduals or individuals with super-spreader potential under
normal human interaction conditions are not substantiated
by published scientific literature. Examples of an apparent
super-spreader, long before the emergence of VoCs with
enhanced transmissibility, with Ct values of ≥ 27 [42] or cul-
tivation of SARS-CoV-2 from specimen with Ct values 35
[43, 44] have been reported. Similarly, experimental studies
have estimated that around 1000 virus particles may be suf-
ficient for infection of a new host [45, 46], while viral loads
required for a positive score in SARS-CoV-2 RATs range
more than 1.000-fold higher. (3) The determination of the
analytical sensitivity alone is not sufficient to draw conclu-
sions on the clinical performance. Preliminary reports indi-
cate that COVID-19 patients with omicron may shed even
less virus than individuals infected with previous VoCs [7].
This could further worsen the clinical performance in light
of the lower analytical sensitivity of the majority of RATs
examined in this study. Since omicron is considered more
contagious than previous VoCs based on epidemiological
findings [47, 48], these COVID-19 patients, with or without
symptoms, would be assumed to potentially be infectious
despite a lower viral load in respiratory swabs. Furthermore,
clinical studies are just beginning to assess the level of virus
shedding and infectivity in the context of omicron infections
and breakthrough infections in vaccinated individuals [79,
27].
Modeling studies suggest that the limited sensitivity of
RATs to identify infected and infectious individuals may
be compensated for by multiple repeated testing. This is
based, in part, on claims that a “recalibrated absolute sen-
sitivity” of RATs is around 80% [49], which is, however,
not supported by the majority of independent laboratory-
based or field studies [26]. On the other hand, especially in
the initial phase of symptomatic disease, a time window of
a few hours may be sufficient to repeat a rapid antigen test-
ing with low sensitivity in suspected disease to enhance
the likelihood of detecting an infection. A recent study
suggests that the viral load during this period can rapidly
and markedly increase for RATs to score positive [50].
Interestingly, epidemiological studies by the Robert
Koch-Institute suggested that 67% of individuals with a
negative RAT result believe that they are non-infectious
for at least 48h, potentially affecting their behavior regard-
ing preventive hygiene measures. In part, this perception
results from overly optimistic communications on the reli-
ability of this class of tests.
Our current study suggests that the emergence of future
VoCs of this pandemic β-coronavirus with an altered muta-
tional pattern in the nucleocapsid protein should result in
an immediate reassessment of the performance of RATs.
Since the rapidly spreading BA.2 sub-lineage of omicron
carries an additional S413R mutation in the nucleocapsid
protein, it is difficult to predict the RAT performance for
this VoC. The minimum requirements stated by interna-
tional organizations and the Paul-Ehrlich-Institute for the
performance of SARS-CoV-2 rapid antigen tests include
an overall diagnostic sensitivity of > 80% and a specific-
ity of > 97% [51, 52]. Independently validated RATs that
do not meet these criteria, should be taken off the market
immediately.
In light of first international reports [14, 29] and the
results from our present study, current communications by
the Paul-Ehrlich Institute [51] and the Bundesministerium
für Gesundheit [40] on the apparent reliability of RATs for
the detection of an omicron infection seem premature. More-
over, critical pre-analytical factors, including the timing of
the swab relative to the onset of symptoms, the vaccination
status, the swabbing practices and test procedures, in par-
ticular when not performed by trained health care profes-
sionals in COVID-19 testing centers, may also have a great
impact on the “real-world” diagnostic sensitivity of RATs.
The latter factors should underlie a constant optimization
and a regular and stringent assessment by health authorities.
Vaccination campaigns have achieved a substantial cover-
age of the adult population in first-world countries, from the
perspective of health care systems markedly improving the
starting point for the control of future VoCs that may arise
from the original evolutionary trait of SARS-CoV-2, from
which VoCs alpha, beta, gamma and delta arose. Alternative
testing concepts including sample pooling for PCR or iso-
thermal RT-LAMP nucleic acid-based detection of SARS-
CoV-2 should be optimized to reduce the potential burden
of infections and COVID-19 in the context of large social
events with conditions favoring SARS-CoV-2 transmission.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
115Medical Microbiology and Immunology (2022) 211:105–117
1 3
Supplementary Information The online version contains supplemen-
tary material available at https:// doi. org/ 10. 1007/ s00430- 022- 00730-z.
Acknowledgements We are grateful to members of the diagnostic
laboratory of virology at the Max von Pettenkofer Institute for support.
Author contributions AO and OTK designed the study. IB, EB, FK,
GNÖ, ME, RK, MH, PS, MS, HA, AG, SK, performed experiments.
MC and JD provided and characterized clinical samples. AO, MM, HB,
CD, LK, H-MB, OTK analyzed data. LK performed statistical analysis.
AO, MS, CD, LK, H-MB, and OTK wrote the manuscript. All authors
discussed the results and commented on the final manuscript.
Funding Open Access funding enabled and organized by Projekt
DEAL. This work was supported by the Free State of Bavaria (research
initiatives Bay-VOC (M.M., A.G., S.K., H.B., O.T.K.) and FOR-
COVID (M.M., O.T.K.)).
Declarations
Conflict of interest The authors declare that they have no conflict of
interest.
Ethics approval Ethical approval was not required because all samples
used were residual samples. Samples were identified in the laboratory
and assigned a number without reference to patient or clinical details.
Results from testing using the study assays did not impact on clinical
care.
Data and materials availability Not applicable.
Code availability Not applicable.
Open Access This article is licensed under a Creative Commons Attri-
bution 4.0 International License, which permits use, sharing, adapta-
tion, 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/.
References
1. Cele S, Jackson L, Khoury DS, Khan K, Moyo-Gwete T, Tegally
H etal (2021) Omicron extensively but incompletely escapes
Pfizer BNT162b2 neutralization. Nature. https:// doi. org/ 10. 1038/
s41586- 021- 04387-1
2. Planas D, Saunders N, Maes P, Guivel-Benhassine F, Planchais
C, Buchrieser J etal (2021) Considerable escape of SARS-CoV-2
Omicron to antibody neutralization. Nature. https:// doi. org/ 10.
1038/ s41586- 021- 04389-z
3. Schmidt F, Weisblum Y, Rutkowska M, Poston D, DaSilva J,
Zhang F etal (2021) High genetic barrier to SARS-CoV-2 poly-
clonal neutralizing antibody escape. Nature 600(7889):512–516.
https:// doi. org/ 10. 1038/ s41586- 021- 04005-0
4. Gruell H, Vanshylla K, Tober-Lau P, Hillus D, Schommers
P, Lehmann C etal (2022) mRNA booster immunization
elicits potent neutralizing serum activity against the SARS-
CoV-2 Omicron variant. Nat Med. https:// doi. org/ 10. 1038/
s41591- 021- 01676-0
5. Wratil PR, Stern M, Priller A, Willmann A, Almanzar G, Vogel
E etal (2022) Three exposures to the spike protein of SARS-
CoV-2 by either infection or vaccination elicit superior neutral-
izing immunity to all variants of concern. Nat Med. https:// doi.
org/ 10. 1038/ s41591- 022- 01715-4
6. Volz E, Mishra S, Chand M, Barrett JC, Johnson R, Geidelberg
L etal (2021) Assessing transmissibility of SARS-CoV-2 lineage
B.1.1.7 in England. Nature 593(7858):266–269. https:// doi. org/
10. 1038/ s41586- 021- 03470-x
7. Hay JA, Kissler SM, Fauver JR, Mack C, Tai CG, Samant RM
etal (2022) Viral dynamics and duration of PCR positivity of the
SARS-CoV-2 Omicron variant. medRxiv. https:// doi. org/ 10. 1101/
2022. 01. 13. 22269 257
8. Petros BA, Turcinovic J, Welch NL, White LF, Kolaczyk ED,
Bauer MR etal (2022) Early introduction and rise of the Omicron
SARS-CoV-2 variant in highly vaccinated university populations.
medRxiv. https:// doi. org/ 10. 1101/ 2022. 01. 27. 22269 787
9. Puhach O, Adea K, Hulo N, Sattonnet P, Genecand C, Iten A
etal (2022) Infectious viral load in unvaccinated and vaccinated
patients infected with SARS-CoV-2 WT, Delta and Omicron.
medRxiv. https:// doi. org/ 10. 1101/ 2022. 01. 10. 22269 010
10. Araf Y, Akter F, Tang Y-D, Fatemi R, Parvez MSA, Zheng C
etal (2022) Omicron variant of SARS-CoV-2: genomics, trans-
missibility, and responses to current COVID-19 vaccines. J Med
Virol. https:// doi. org/ 10. 1002/ jmv. 27588
11. Karim SSA, Karim QA (2021) Omicron SARS-CoV-2 vari-
ant: a new chapter in the COVID-19 pandemic. Lancet
398(10317):2126–2128. https:// doi. org/ 10. 1016/ s0140-
6736(21) 02758-6 ((Epub 2021/12/07))
12. Mohapatra RK, Sarangi AK, Kandi V, Azam M, Tiwari R,
Dhama K (2022) Omicron (B1.1.529 variant of SARS-CoV-2);
an emerging threat: current global scenario. J Med Virol. https://
doi. org/ 10. 1002/ jmv. 27561
13. Alaa Abdel Latif JLM, Manar A, Ginger T, Marco C, Emily
H, Jerry Z, Mark Z, Emory H, Nate M, Chunlei W, Kristian
GA, Andrew IS, Karthik G, Laura DH, and the Center for Viral
Systems Biology. http:// outbr eak. info. Acccessed 22 Jul 2021
14. Bekliz M, Adea K, Alvarez C, Essaidi-Laziosi M, Escadafal
C, Kaiser L etal (2021) Analytical sensitivity of seven SARS-
CoV-2 antigen-detecting rapid tests for Omicron variant.
medRxiv. https:// doi. org/ 10. 1101/ 2021. 12. 18. 21268 018
15. Bekliz M, Adea K, Essaidi-Laziosi M, Sacks JA, Escadafal C,
Kaiser L etal (2021) SARS-CoV-2 rapid diagnostic tests for
emerging variants. Lancet Microbe 2(8):e351. https:// doi. org/
10. 1016/ S2666- 5247(21) 00147-6
16. Corman VM, Haage VC, Bleicker T, Schmidt ML, Mühlemann
B, Zuchowski M etal (2021) Comparison of seven commercial
SARS-CoV-2 rapid point-of-care antigen tests: a single-centre
laboratory evaluation study. Lancet Microbe 2(7):e311–e319.
https:// doi. org/ 10. 1016/ S2666- 5247(21) 00056-2
17. Jungnick S, Hobmaier B, Mautner L, Hoyos M, Haase M, Bai-
ker A etal (2021) Invitro rapid antigen test performance with
the SARS-CoV-2 variants of concern B.1.1.7 (Alpha), B.1.351
(Beta), P.1 (Gamma), and B.1.617.2 (Delta). Microorganisms
9(9):1967. https:// doi. org/ 10. 3390/ micro organ isms9 091967
18. Osterman A, Baldauf HM, Eletreby M, Wettengel JM, Afridi
SQ, Fuchs T etal (2021) Evaluation of two rapid antigen tests
to detect SARS-CoV-2 in a hospital setting. Med Microbiol
Immunol 210(1):65–72. https:// doi. org/ 10. 1007/ s00430- 020-
00698-8 ((Epub 2021/01/17))
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
116 Medical Microbiology and Immunology (2022) 211:105–117
1 3
19. Osterman A, Iglhaut M, Lehner A, Späth P, Stern M, Autenri-
eth H etal (2021) Comparison of four commercial, automated
antigen tests to detect SARS-CoV-2 variants of concern. Med
Microbiol Immunol 210(5–6):263–275. https:// doi. org/ 10. 1007/
s00430- 021- 00719-0 ((Epub 2021/08/21))
20. Sakai-Tagawa Y, Yamayoshi S, Halfmann PJ, Kawaoka Y (2021)
Comparative sensitivity of rapid antigen tests for the delta vari-
ant (B.1.617.2) of SARS-CoV-2. Viruses 13(11):2183. https://
doi. org/ 10. 3390/ v1311 2183
21. Scheiblauer H, Filomena A, Nitsche A, Puyskens A, Corman
VM, Drosten C etal (2021) Comparative sensitivity evalua-
tion for 122 CE-marked rapid diagnostic tests for SARS-CoV-2
antigen, Germany, September 2020 to April 2021. Euro Surveill
26:44. https:// doi. org/ 10. 2807/ 1560- 7917. Es. 2021. 26. 44. 21004
41 ((Epub 2021/11/06))
22. Schildgen V, Demuth S, Lüsebrink J, Schildgen O (2021) Lim-
its and opportunities of SARS-CoV-2 antigen rapid tests: an
experienced-based perspective. Pathogens 10:1. https:// doi. org/
10. 3390/ PATHO GENS1 00100 38 ((Epub 2021/01/21))
23. Toptan T, Eckermann L, Pfeiffer AE, Hoehl S, Ciesek S, Dros-
ten C etal (2021) Evaluation of a SARS-CoV-2 rapid antigen
test: potential to help reduce community spread? J Clin Virol
135:104713. https:// doi. org/ 10. 1016/j. jcv. 2020. 104713 ((Epub
2020/12/23))
24. Bekliz M, Adea K, Essaidi-Laziosi M, Sacks JA, Escadafal C,
Kaiser L etal (2022) SARS-CoV-2 antigen-detecting rapid tests
for the delta variant. Lancet Microbe 3(2):e90. https:// doi. org/
10. 1016/ S2666- 5247(21) 00302-5
25. Krüger LJ, Tanuri A, Lindner AK, Gaeddert M, Köppel L,
Tobian F etal (2022) Accuracy and ease-of-use of seven point-
of-care SARS-CoV-2 antigen-detecting tests: a multi-centre
clinical evaluation. EBioMedicine 75:103774. https:// doi. org/
10. 1016/j. ebiom. 2021. 103774 ((Epub 2021/12/28))
26. Dinnes J, Deeks JJ, Berhane S, Taylor M, Adriano A, Davenport
C etal (2021) Rapid, point-of-care antigen and molecular-based
tests for diagnosis of SARS-CoV-2 infection. Cochrane Data-
base Syst Rev. https:// doi. org/ 10. 1002/ 14651 858. CD013 705.
pub2
27. Adamson B, Sikka R, Wyllie AL, Premsrirut P (2022) Discordant
SARS-CoV-2 PCR and rapid antigen test results when infectious:
a December 2021 occupational case series. medRxiv. https:// doi.
org/ 10. 1101/ 2022. 01. 04. 22268 770
28. Schrom J, Marquez C, Pilarowski G, Wang G, Mitchell A, Puc-
cinelli R etal (2022) Direct comparison of SARS Co-V-2 nasal
RT-PCR and rapid antigen test (BinaxNOW™) at a community
testing site during an Omicron surge. medRxiv. https:// doi. org/ 10.
1101/ 2022. 01. 08. 22268 954
29. Deerain J, Druce J, Tran T, Batty M, Yoga Y, Fennell M etal
(2021) Assessment of the analytical sensitivity of ten lateral flow
devices against the SARS-CoV-2 omicron variant. J Clin Micro-
biol. https:// doi. org/ 10. 1128/ jcm. 02479- 21 ((Epub 2021/12/23))
30. European Commission. EU health preparedness: A common list
of COVID-19 rapid antigen tests; A common standardised set
of data to be included in COVID-9 test result certificates; and
A common list of COVID-19 laboratory based antigenic assays.
https:// ec. europa. eu/ health/ system/ files/ 2022- 01/ covid- 19_ rat_
common- list_ en. pdf. Accessed 07 Jul 2022
31. Centers for Disease Control and Prevention (CDC) Information
for laboratories about coronavirus (COVID-19). https:// www. cdc.
gov/ coron avirus/ 2019- ncov/ lab/ index. html. Accessed 2 Jul 2022
32. Dächert C, Muenchhoff M, Graf A, Autenrieth H, Bender S,
Mairhofer H etal (2022) Rapid and sensitive identification of
omicron by variant-specific PCR and nanopore sequencing: par-
adigm for diagnostics of emerging SARS-CoV-2 variants. Med
Microbiol Immunol. https:// doi. org/ 10. 1007/ s00430- 022- 00728-7
((Epub 2022/01/22))
33. Weinberger T, Steffen J, Osterman A, Mueller TT, Muenchhoff
M, Wratil PR etal (2021) Prospective longitudinal serosurvey of
healthcare workers in the first wave of the severe acute respiratory
syndrome coronavirus 2 (SARS-CoV-2) pandemic in a quaternary
care hospital in Munich, Germany. Clin Infect Dis 73(9):e3055–
e3065. https:// doi. org/ 10. 1093/ cid/ ciaa1 935 ((Epub 2021/01/04))
34. Kannan SR, Spratt AN, Cohen AR, Naqvi SH, Chand HS, Quinn
TP etal (2021) Evolutionary analysis of the delta and delta plus
variants of the SARS-CoV-2 viruses. J Autoimmun 124:102715.
https:// doi. org/ 10. 1016/j. jaut. 2021. 102715 ((Epub 2021/08/11))
35. Stanley S, Hamel DJ, Wolf ID, Riedel S, Dutta S, Cheng A etal
(2022) Limit of detection for rapid antigen testing of the SARS-
CoV-2 Omicron variant. medRxiv. https:// doi. org/ 10. 1101/ 2022.
01. 28. 22269 968
36. Puyskens A, Krause E, Michel J, Nübling CM, Scheiblauer H,
Bourquain D etal (2021) Establishment of a specimen panel for
the decentralised technical evaluation of the sensitivity of 31 rapid
diagnostic tests for SARS-CoV-2 antigen, Germany, September
2020 to April 2021. Euro Surveill 26:44. https:// doi. org/ 10. 2807/
1560- 7917. Es. 2021. 26. 44. 21004 42 ((Epub 2021/11/06))
37. Klein S, Cortese M, Winter SL, Wachsmuth-Melm M, Neufeldt
CJ, Cerikan B etal (2020) SARS-CoV-2 structure and replication
characterized by insitu cryo-electron tomography. Nat Commun
11(1):5885. https:// doi. org/ 10. 1038/ s41467- 020- 19619-7
38. Liu T, Jia P, Fang B, Zhao Z (2020) Differential expression of
viral transcripts from single-cell RNA sequencing of moderate
and severe COVID-19 patients and its implications for case sever-
ity. Front Microbiol. https:// doi. org/ 10. 3389/ fmicb. 2020. 603509
39. Paul-Ehrlich-Institute. SARS-CoV-2-Antigentests für Nachweis
der Omikron-Infektion geeignet (in German). https:// www. pei. de/
DE/ newsr oom/ hp- meldu ngen/ 2021/ 211230- antig entes ts- omikr on-
varia nte. html. Accessed 2 Jul 2022
40. Gesundheitministerium für Gesundheit. Fragen und Antworten
zu Quarantäne- und Isolierungsregeln (in German). https:// www.
bunde sgesu ndhei tsmin ister ium. de/ coron avirus/ faq- quara ntaene-
isoli erung srege ln. html# c23632. Accessed 2 Jul 2022
41. Bundesinstitut für Arzneimittel und Medizinprodukte (BfarM).
Antigen-Tests auf SARS-CoV-2 zur Eigenanwendung, die Gegen-
stand des Anspruchs nach §1 Satz 1 Coronavirus-Testverordnung
(TestV) sind („Selbsttests“) (in German). https:// antig entest.
bfarm. de/ ords/f? p= ANTIG ENTES TS- AUF- SARS- COV-2:
TESTS- ZUR- EIGEN ANWEN DUNG- DURCH- LAIEN: 11537
84940 4532::::: & tz=1: 00. Accessed 2 Jun 2022
42. Lin J, Yan K, Zhang J, Cai T, Zheng J (2020) A super-spreader
of COVID-19 in Ningbo city in China. J Infect Public Health
13(7):935–937. https:// doi. org/ 10. 1016/j. jiph. 2020. 05. 023
43. Liu L-T, Tsai J-J, Chen C-H, Lin P-C, Tsai C-Y, Tsai Y-Y etal
(2022) Isolation and identification of a rare spike gene double-
deletion SARS-CoV-2 variant from the patient with high cycle
threshold value. Front Med. https:// doi. org/ 10. 3389/ fmed. 2021.
822633
44. Singanayagam A, Patel M, Charlett A, Lopez Bernal J, Saliba V,
Ellis J etal (2020) Duration of infectiousness and correlation with
RT-PCR cycle threshold values in cases of COVID-19, England,
January to May 2020. Euro Surveill 25:32. https:// doi. org/ 10.
2807/ 1560- 7917. Es. 2020. 25. 32. 20014 83 ((Epub 2020/08/15))
45. Imai M, Iwatsuki-Horimoto K, Hatta M, Loeber S, Halfmann PJ,
Nakajima N etal (2020) Syrian hamsters as a small animal model
for SARS-CoV-2 infection and countermeasure development. Proc
Natl Acad Sci 117(28):16587–16595. https:// doi. org/ 10. 1073/
pnas. 20097 99117
46. Popa A, Genger J-W, Nicholson MD, Penz T, Schmid D, Aberle
SW etal (2020) Genomic epidemiology of superspreading events
in Austria reveals mutational dynamics and transmission proper-
ties of SARS-CoV-2. Sci Transl Med 12(573):eabe2555. https://
doi. org/ 10. 1126/ scitr anslm ed. abe25 55
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
117Medical Microbiology and Immunology (2022) 211:105–117
1 3
47. Ito K, Piantham C, Nishiura H (2021) Relative instantaneous
reproduction number of Omicron SARS-CoV-2 variant with
respect to the Delta variant in Denmark. J Med Virol. https:// doi.
org/ 10. 1002/ jmv. 27560 ((Epub 2021/12/31))
48. Nishiura H, Ito K, Anzai A, Kobayashi T, Piantham C, Rodríguez-
Morales AJ (2021) Relative reproduction number of SARS-CoV-2
Omicron (B.1.1.529) compared with delta variant in South Africa.
J Clin Med 11:1. https:// doi. org/ 10. 3390/ jcm11 010030 ((Epub
2022/01/12))
49. Petersen I, Crozier A, Buchan I, Mina MJ, Bartlett JW (2021)
Recalibrating SARS-CoV-2 antigen rapid lateral flow test rela-
tive sensitivity from validation studies to absolute sensitivity for
indicating individuals shedding transmissible virus. Clin Epide-
miol 13:935–940. https:// doi. org/ 10. 2147/ clep. S3119 77 ((Epub
2021/10/28))
50. Ben K, Alex M, Mariya K, Alison B, Niluka G, Jie Z etal (2022).
Nat Portfolio. https:// doi. org/ 10. 21203/ rs.3. rs- 11219 93/ v1
51. Paul-Ehrlich-Institute. Mindestkriterien für SARS-CoV-2 Anti-
gentests im Sinne von § 1 Abs. 1 Satz 1 TestVO: antigenschnell-
tests (in German). https:// www. pei. de/ Share dDocs/ Downl oads/
DE/ newsr oom/ dossi ers/ minde stkri terien- sars- cov-2- antig entes
ts- 01- 12- 2020. pdf?__ blob= publi catio nFile. Accessed 2 Jul 2022.
52. World Health Organization (WHO) (2020) Antigen-detection in
the diagnosis of SARS-CoV-2 infection using rapid immunoas-
says. https:// www. who. int/ publi catio ns/i/ item/ antig en- detec tion-
in- the- diagn osis- of- sars- cov- 2infe ction- using- rapid- immun oassa
ys. Accessed 2 Jul 2022
Publisher's Note Springer Nature remains neutral with regard to
jurisdictional claims in published 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
... The rapid global emergence and dominance of the Omicron variant highlighted the importance of understanding the performance of ADTs in real-world settings. Some in vitro studies suggested that the performance of rapid ADTs did not differ between the Delta and Omicron variants [18,19], while studies using clinical specimens suggested a possible decrease in antigen tests' sensitivity for the Omicron variant [20][21][22][23]. ...
... The genome of Omicron subvariants contains more than 50 mutations [37], many of which have been associated with an increased transmissibility, variable disease severity, and the potential to evade immune responses acquired after SARS-CoV-2 vaccination or infection with a previous variant. Few studies have attempted to investigate the impact of mutations in the N protein on the diagnostic performance of ADTs, with conflicting results [8,[18][19][20][21][22]38]. Due to a possible change in the tropism, it has been suggested that the detection of the Omicron variant could be favoured in oral swabs compared to nasal swabs [24]. ...
... First, the lack of clinical characteristics of the patients included in the comparison of ADTs' sensitivity in Omicron vs. Delta infections. In fact, previous studies have shown a very low sensitivity of rapid antigen tests in asymptomatic patients, and only a moderate decrease in sensitivity for symptomatic Omicron infections [20][21][22]38,43]. Second, 6.8% of the analysed samples derived from multiple hospital accesses were from individuals who participated more than once in the study, so this could be a possible confounder. ...
Article
Full-text available
With the continuous spread of new SARS-CoV-2 variants of concern (VOCs), the monitoring of diagnostic test performances is mandatory. We evaluated the changes in antigen diagnostic tests’ (ADTs) accuracy along the Delta to Omicron VOCs transition, exploring the N protein mutations possibly affecting ADT sensitivity and assessing the best sampling site for the diagnosis of Omicron infections. In total, 5175 subjects were enrolled from 1 October 2021 to 15 July 2022. The inclusion criteria were SARS-CoV-2 ADT combined with a same-day RT-PCR swab test. For the sampling site analysis, 61 patients were prospectively recruited during the Omicron period for nasal and oral swab analyses by RT-PCR. Next-Generation Sequencing data were obtained to evaluate the different sublineages. Using RT-PCR as a reference, 387 subjects resulted in becoming infected and the overall sensitivity of the ADT decreased from 63% in the Delta period to 33% in the Omicron period. This decrease was highly statistically significant (p < 0.001), and no decrease in viral load was detected at the RNA level. The nasal site presented a significantly higher viral load than the oral site during the Omicron wave. The reduced detection rate of Omicron infections by ADT should be considered in the global testing strategy to preserve accurate diagnoses across the changing SARS-CoV-2 variants.
... We found most of the mutations that evolved during the second wave changed the hydrophobicity score of the corresponding epitope regions particularly mutations, N:D63G, N:R203M, and N:D377Y, inside nucleocapsid protein, S:L452R and S:P681R inside the spike proteins of the SARS-CoV-2. These mutations were found to have various roles in the spread of infection, N:D63G might increase immune escape abilities (22), N:D377Y might affect the antibody binding (23)(24)(25), S:L452R might be involved in decreased neutralization by MAbs (26), and S:P681R might be involved in furin cleavage (27). ...
Article
The evolution of viral variants and their impact on viral transmission have been an area of considerable importance in this pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We analyzed the viral variants in different phases of the pandemic in West Bengal, a state in India that is important geographically, and compared the variants with other states like Delhi, Maharashtra, and Karnataka, located in other regions of the country. We have identified 57 pango-lineages in 3,198 SARS-CoV-2 genomes, alteration in their distribution, as well as contrasting profiles of amino acid mutational dynamics across different waves in different states. The evolving characteristics of Delta (B.1.617.2) sublineages and alterations in hydrophobicity profiles of the viral proteins caused by these mutations were also studied. Additionally, implications of predictive host miRNA binding/unbinding to emerging spike or nucleocapsid mutations were highlighted. Our results throw considerable light on interesting aspects of the viral genomic variation and provide valuable information for improved understanding of wave-defining mutations in unfolding the pandemic. IMPORTANCE Multiple waves of infection were observed in many states in India during the coronavirus disease 2019 (COVID19) pandemic. Fine-scale evolution of major SARS-CoV-2 lineages and sublineages during four wave-window categories: Pre-Wave 1, Wave 1, Pre-Wave 2, and Wave 2 in four major states of India: Delhi (North), Maharashtra (West), Karnataka (South), and West Bengal (East) was studied using large-scale virus ge-nome sequencing data. Our comprehensive analysis reveals contrasting molecular profiles of the wave-defining mutations and their implications in host miRNA binding/ unbinding of the lineages in the major states of India.
... We found most of the mutations that evolved during the second wave changed the hydrophobicity score of the corresponding epitope regions particularly mutations, N:D63G, N:R203M, and N:D377Y, inside nucleocapsid protein, S:L452R and S:P681R inside the spike proteins of the SARS-CoV-2. These mutations were found to have various roles in the spread of infection, N:D63G might increase immune escape abilities (22), N:D377Y might affect the antibody binding (23)(24)(25), S:L452R might be involved in decreased neutralization by MAbs (26), and S:P681R might be involved in furin cleavage (27). ...
... COVID-19 emerged in late 2019, quickly spread around the world and was declared a global pandemic on March 11, 2021, by the World Health Organization (WHO) (1). Since its emergence multiple SARS-CoV-2 variants developed which mostly were characterized by mutations in the Spike protein but also within the Nucleocapsid protein (2)(3)(4)(5). Variants showing a decrease in the effectiveness of available diagnostic tests among other criteria are termed Variants of Concern (VOC) by the WHO (6). To date the WHO has listed 5 VOCs, namely: B.1.1.7 (alpha), B.1.351 ...
Article
Full-text available
Background This study is the extension of the COVAG study. We compared two RATs, the Panbio COVID-19 Ag Rapid Test (Abbott) and the SD Biosensor Q SARS-CoV-2 Rapid Antigen Test (Roche), against RT-PCR on the foil of new variants. Methods We included 888 all-comers at a diagnostic center between October 20, 2021, and March 18, 2022. RT-PCR-positive samples with a Ct value ≤32 were examined for SARS-CoV-2 variants. Findings The sensitivity of the Abbott-RAT and Roche-RAT were 65 and 67%, respectively. For both RATs, lower Ct values were significantly correlated with higher sensitivity. For samples with Ct values ≤25, the sensitivities of the Roche-RAT and of the Abbott-RAT were 96 and 95%, for Ct values 25–30 both were 19%, and for Ct values ≥30 they were 6 and 2%, respectively. The RATs had substantially higher sensitivities in symptomatic than asymptomatic participants (76, 77%, vs. 29, 31%, for Abbott-RAT, Roche-RAT, respectively) and in participants referred to testing by their primary care physician (84, 85%) compared to participants who sought testing due to referral by the health department (55, 58%) or a warning by the Corona-Warn-App (49, 49%). In persons with self-reported previous COVID-19 sensitivities were markedly lower than in patients without previous COVID-19: 27% vs. 75% for Roche-RAT and 27% vs. 73% for Abbott-RAT. We did not find significant correlation between vaccination status and sensitivity. The Omicron variant was detected with a sensitivity of 94 and 92%, the delta variant with a sensitivity of 80 and 80% for Abbott-RAT and Roche-RAT, respectively. This difference is attributable to the lower Ct values of the Omicron samples compared to the Delta samples. When adjusted for the Ct value, a multivariate logistic regression did not show a significant difference between Omicron and Delta. In terms of sensitivity, we found no significant difference between the wild-type and the Omicron and Delta variants, but a significantly lower sensitivity to the alpha variant compared to the other variants. The specificities were > 99% overall.
... [25,26] Evidence to date is heterogeneous that the Omicron VOC might reduce the test performance of RDTs. [16,19,21,[27][28][29][30] Data from an interim analysis of the study suggested that any deterioration in RDT test performance is not attributable to VOC itself but rather to the change in symptomatology mediated by the VOC throughout the course of the COVID-19 pandemic. [18] Further, regarding the potential influence of COVID-19 vaccination on RDT performance only very few studies have so far considered the aspect of COVID-19 vaccination revealing no influence of COVID-19 vaccination on RDT performance. ...
Preprint
Full-text available
Introduction During the COVID-19 pandemic, SARS-CoV-2 antigen rapid detection tests (RDTs) emerged as point-of-care diagnostics in addition to the RT-qPCR as the gold standard for SARS-CoV-2 diagnostics. Facing the course of the COVID-19 pandemic to an endemic characterised by several SARS-CoV-2 virus variants of concern (VOC) and an increasing public COVID-19 vaccination rate the aim of the study was to investigate the long-term test performance of SARS-CoV-2 RDT in large-scale, clinical screening use during and its influencing factors, above all SARS-CoV-2 VOC and COVID-19 vaccination. Methods In a prospective performance assessment conducted at a single centre tertiary care hospital, RDTs from three manufacturers (NADAL®, Panbio™, MEDsan®) were compared to RT-qPCR among individuals aged ≥6 month. The evaluation involved the determination of standardised viral load from oropharyngeal swabs as well as the evaluation of their influencing factors, especially the COVID-19 vaccination, for detecting SARS-CoV-2 in a clinical point-of-care environment spanning from 12 November 2020 to 30 June 2023 among patients, staff, and visitors of the hospital. Results Among the 78,798 RDT/RT-qPCR tandems analysed, 2,016 (2.6%) tandems tested positive for SARS-CoV-2, with an overall sensitivity of 34.5% (95% CI 32.4-36.6%). A logistic regression revealed that typical COVID-19 symptoms significantly declined over the course of the study and throughout the COVID-19 pandemic, and that among the vaccinated, significantly fewer presented with an infection exhibiting typical symptoms. The employed lasso regression model indicated that only higher viral load and typical COVID-19 symptoms significantly increase the likelihood of a positive RDT result in the case of a SARS-CoV-2 infection directly. Conclusion Our findings indicate that only viral load and COVID-19 symptoms directly influence RDT performance while the obtained effects of COVID-19 vaccination and Omicron VOC both reducing RDT performance were mediated by these two factors. RDTs remain an adequate diagnostic tool for detecting SARS-CoV-2 in individuals showing respiratory symptoms. RDTs show promise beyond SARS-CoV-2, proving adaptable for detecting other pathogens like Influenza and RSV, highlighting their ongoing importance in infection control and prevention efforts.
Article
Background The development of serodiagnostic tests and vaccines for COVID-19 depends on the identification of epitopes from the SARS-CoV-2 genome. An epitope is the specific part of an antigen that is recognized by the immune system and can elicit an immune response. However, when the genetic variants contained in epitopes are used to develop rapid antigen tests (Ag-RDTs) and DNA or RNA vaccines, test sensitivity and vaccine efficacy can be low. Methods Here, we developed a “variant on epitope (VOE)” software, a new Python script for identifying variants located on an epitope. Variant analysis and sensitivity calculation for seven recommended epitopes were processed by VOE. Variants in 1,011 Omicron SRA reads from two variant databases (BCFtools and SARS-CoV-2-Freebayes) were processed by VOE. Results A variant with HIGH or MODERATE impact was found on all epitopes from both variant databases except the epitopes KLNDLCFTNV, RVQPTES, LKPFERD, and ITLCFTLKRK on the S gene and ORF7a gene. All epitope variants from the BCFtools and SARS-CoV-2 Freebayes variant databases showed about 100% sensitivity except epitopes APGQTGK and DSKVGGNYN on the S gene, which showed respective sensitivities of 28.4866% and 6.8249%, and 87.7349% and 71.1177%. Conclusions Therefore, the epitopes KLNDLCFTNV, RVQPTES, LKPFERD, and ITLCFTLKRK may be useful for the development of an epitope-based peptide vaccine and GGDGKMKD on the N gene may be useful for the development of serodiagnostic tests. Moreover, VOE can also be used to analyze other epitopes, and a new variant database for VOE may be further established when a new variant of SARS-CoV-2 emerges.
Article
Post-market surveillance of test performance is a critical function of public health agencies and clinical researchers that ensures tests maintaining diagnostic characteristics following their regulatory approval. Changes in product quality, manufacturing processes over time, or the evolution of new variants may impact product performance. During the COVID-19 pandemic, a plethora of point-of-care tests (POCTs) was released onto the Canadian market. This study evaluated the performance characteristics of several of the most widely distributed POCTs in Canada, including four rapid antigen tests (Abbott Panbio, BTNX Rapid Response, SD Biosensor, and Quidel QuickVue) and two molecular tests (Abbott ID NOW and Lucira Check IT). All tests were challenged with 149 SARS-CoV-2 clinical positives, including multiple variants up to and including Omicron XBB.1.5, as well as 29 clinical negatives. Results were stratified based on whether the isolate was Omicron or pre-Omicron as well as by reverse transcriptase quantitative PCR Ct value. The test performance of each POCT was consistent with the manufacturers' claims and showed no significant decline in clinical performance against any of the variants tested. These findings provide continued confidence in the results of these POCTs as they continue to be used to support decentralized COVID-19 testing. This work demonstrates the essential role of post-market surveillance in ensuring reliability in diagnostic tools. IMPORTANCE Post-market surveillance of diagnostic test performance is critical to ensure their reliability after regulatory approval. This is especially critical in the context of the COVID-19 pandemic as the use of point-of-care tests (POCTs) became widespread. Our study focused on four rapid antigen tests (Abbott Panbio, BTNX Rapid Response, SD Biosensor, and Quidel QuickVue) and two molecular tests (Abbott ID NOW and Lucira Check IT) that were widely distributed across Canada, assessing their performance using many SARS-CoV-2 variants, including up to Omicron subvariant XBB.1.5. Overall, we found no significant difference in performance against any variant, reinforcing confidence in their use. As concerns in test efficacy have been raised by news outlets, particularly regarding the BTNX Rapid Response, this work is even more timely and crucial. Our research offers insights into the performance of widely used COVID-19 POCTs but also highlights the necessity for post-market surveillance.
Article
Full-text available
Background The Omicron variant of SARS-CoV-2 is highly transmissible in vaccinated and unvaccinated populations. The dynamics governing its establishment and propensity towards fixation (reaching 100% frequency in the SARS-CoV-2 population) in communities remain unknown. In this work, we describe the dynamics of Omicron at three institutions of higher education (IHEs) in the greater Boston area. Methods We use diagnostic and variant-specifying molecular assays and epidemiological analytical approaches to describe the rapid dominance of Omicron following its introduction to three IHEs with asymptomatic surveillance programs. Results We show that the establishment of Omicron at IHEs precedes that of the state and region, and that the time to fixation is shorter at IHEs (9.5-12.5 days) than in the state (14.8 days) or region. We show that the trajectory of Omicron fixation among university employees resembles that of students, with a 2-3 day delay. Finally, we compare cycle threshold (Ct) values in Omicron vs. Delta variant cases on college campuses, and identify lower viral loads among college affiliates harboring Omicron infections. Conclusions We document the rapid takeover of the Omicron variant at IHEs, reaching near-fixation within the span of 9.5-12.5 days despite lower viral loads, on average, than the previously dominant Delta variant. These findings highlight the transmissibility of Omicron, its propensity to rapidly dominate small populations, and the ability of robust asymptomatic surveillance programs to offer early insights into the dynamics of pathogen arrival and spread.
Article
Full-text available
Infectious viral load (VL) expelled as droplets and aerosols by infected individuals partly determines SARS-CoV-2 transmission. RNA VL measured by qRT-PCR is only a weak proxy for infectiousness. Studies on the kinetics of infectious VL are important to understand the mechanisms behind the different transmissibility of SARS-CoV-2 variants and the effect of vaccination on transmission, which allows to guide public health measures. In this study we quantified infectious VL in SARS-CoV-2 infected individuals during the first 5 symptomatic days by in vitro culturability assay in unvaccinated or vaccinated individuals infected with pre-variant of concern (pre-VOC) SARS-CoV-2, Delta, or Omicron. Unvaccinated individuals infected with pre-VOC SARS-CoV-2 had lower infectious VL compared to Delta-infected unvaccinated individuals. Full vaccination (defined as >2weeks after reception of 2nd dose during primary vaccination series) significantly reduced infectious VL for Delta breakthrough cases compared to unvaccinated individuals. For Omicron breakthrough cases, reduced infectious VL was only observed in boosted but not in fully vaccinated individuals compared to unvaccinated subjects. In addition, infectious VL was lower in fully vaccinated Omicron- compared to fully vaccinated Delta-infected individuals, suggesting that other mechanisms than increased infectious VL contribute to the high infectiousness of SARS-CoV-2 Omicron. Our findings indicate that vaccines may lower transmission risk and therefore have a public health benefit beyond the individual protection from severe disease. The infectious viral load of SARS-CoV-2 Omicron is lower than that of Delta in symptomatic breakthrough infections of recipients of two doses of a COVID-19 vaccine, suggesting that the higher transmission of Omicron is not linked to higher infectious viral load.
Article
Full-text available
To establish a novel SARS-CoV-2 human challenge model, 36 volunteers aged 18-29 years without evidence of previous infection or vaccination were inoculated with 10 TCID 50 of a wild-type virus (SARS-CoV-2/human/GBR/484861/2020) intranasally. Two participants were excluded from per protocol analysis due to seroconversion between screening and inoculation. Eighteen (~53%) became infected, with viral load (VL) rising steeply and peaking at ~5 days post-inoculation. Virus was first detected in the throat but rose to significantly higher levels in the nose, peaking at ~8.87 log 10 copies/ml (median, 95% CI [8.41,9.53). Viable virus was recoverable from the nose up to ~10 days post-inoculation, on average. There were no serious adverse events. Mild-to-moderate symptoms were reported by 16 (89%) infected individuals, beginning 2-4 days post-inoculation. Anosmia/dysosmia developed more gradually in 12 (67%) participants. No quantitative correlation was noted between VL and symptoms, with high VLs even in asymptomatic infection, followed by the development of serum spike-specific and neutralising antibodies. However, lateral flow results were strongly associated with viable virus and modelling showed that twice-weekly rapid tests could diagnose infection before 70-80% of viable virus had been generated. Thus, in this first SARS-CoV-2 human challenge study, no serious safety signals were detected and the detailed characteristics of early infection and their public health implications were shown. ClinicalTrials.gov identifier: NCT04865237.
Preprint
Full-text available
There has been debate in the literature about the ability of antigen tests to detect the SARS-CoV-2 Omicron variant including indication on the US Food and Drug administration website that antigen tests may have lower sensitivity for the Omicron variant without provision of data or the potential scale of the issue (see https://www.fda.gov/medical-devices/coronavirus-covid-19-and-medical-devices/sars-cov-2-viral-mutations-impact-covid-19-tests - omicronvariantimpact, accessed 1/27/2022). Here we determined the limit of detection (LoD) for the Omicron variant compared with the WA1 strain used for LoD studies described in the Instructions for Use for all Emergency Use Authorization (EUA)-approved antigen tests. Using live virus (to avoid artifactual findings potentially obtained with gamma-irradiated or heat-killed virus) quantified by plaque forming units (PFU), we examined the analytical sensitivity of three antigen tests widely used in the United States: the Abbott Binax Now, the AccessBio CareStart , and LumiraDx antigen tests. We found that the 95% detection threshold (LoD) for antigen tests was at least as good for Omicron as for the WA1 strain. Furthermore, the relationship of genome copies to plaque forming units for Omicron and WA1 overlap. Therefore, the LoD equivalency also applies if the quantitative comparator is genome copies determined from live virus preparations. Taken together, our data support the continued ability of the antigen tests examined to detect the Omicron variant.
Preprint
Full-text available
The Omicron variant of SARS-CoV-2 is transmissible in vaccinated and unvaccinated populations. Here, we describe the rapid dominance of Omicron following its introduction to three Massachusetts universities with asymptomatic surveillance programs. We find that Omicron was established and reached fixation earlier on these campuses than in Massachusetts or New England as a whole, rapidly outcompeting Delta despite its association with lower viral loads. These findings highlight the transmissibility of Omicron and its propensity to fixate in small populations, as well as the ability of robust asymptomatic surveillance programs to offer early insights into the dynamics of pathogen arrival and spread.
Article
Full-text available
Infection-neutralizing antibody responses after SARS-CoV-2 infection or COVID-19 vaccination are an essential component of antiviral immunity. Antibody-mediated protection is challenged by the emergence of SARS-CoV-2 variants of concern (VoCs) with immune escape properties, such as omicron (B.1.1.529) that is rapidly spreading worldwide. Here, we report neutralizing antibody dynamics in a longitudinal cohort of COVID-19 convalescent and infection-naive individuals vaccinated with mRNA BNT162b2 by quantifying anti-SARS-CoV-2-spike antibodies and determining their avidity and neutralization capacity in serum. Using live-virus neutralization assays, we show that a superior infection-neutralizing capacity against all VoCs, including omicron, developed after either two vaccinations in convalescents or after a third vaccination or breakthrough infection of twice-vaccinated, naive individuals. These three consecutive spike antigen exposures resulted in an increasing neutralization capacity per anti-spike antibody unit and were paralleled by stepwise increases in antibody avidity. We conclude that an infection-plus-vaccination-induced hybrid immunity or a triple immunization can induce high-quality antibodies with superior neutralization capacity against VoCs, including omicron.
Article
Full-text available
On November 26, 2021, the World Health Organization classified B.1.1.529 as a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant of concern (VoC), named omicron. Spike -gene dropouts in conventional SARS-CoV-2 PCR systems have been reported over the last weeks as indirect diagnostic evidence for the identification of omicron. Here, we report the combination of PCRs specific for heavily mutated sites in the spike gene and nanopore-based full-length genome sequencing for the rapid and sensitive identification of the first four COVID-19 patients diagnosed in Germany to be infected with omicron on November 28, 2021. This study will assist the unambiguous laboratory-based diagnosis and global surveillance for this highly contagious VoC with an unprecedented degree of humoral immune escape. Moreover, we propose that specialized diagnostic laboratories should continuously update their assays for variant-specific PCRs in the spike gene of SARS-CoV-2 to readily detect and diagnose emerging variants of interest and VoCs. The combination with established nanopore sequencing procedures allows both the rapid confirmation by whole genome sequencing as well as the sensitive identification of newly emerging variants of this pandemic β-coronavirus in years to come.
Article
A new definition of metabolic dysfunction-associated fatty liver disease (MAFLD) has recently been proposed. We aim to examine the associations of MAFLD, particularly its discordance from non-alcoholic fatty liver disease (NAFLD), with the progression of elevated brachial-ankle pulse wave velocity (baPWV) and albuminuria in a community-based study sample in Shanghai, China. After 4.3 years of follow-up, 778 participants developed elevated baPWV and 499 developed albuminuria. In comparison with the non-MAFLD group, the multivariable adjusted odds ratio (OR) of MAFLD group for new-onset elevated baPWV was 1.25 (95% confidence interval (CI) 1.01-1.55) and 1.35 (95% CI 1.07-1.70) for albuminuria. Participants without NAFLD but diagnosed according to MAFLD definition were associated with higher risk of incident albuminuria (OR 1.77; 95% CI 1.07-2.94). Patients with MAFLD with high value of hepamet fibrosis score or poor-controlled diabetes had higher risk of elevated baPWV or albuminuria. In conclusion, MAFLD was associated with new-onset elevated baPWV and albuminuria independently of body mass index, waist circumference, and hip circumference. Individuals without NAFLD but diagnosed as MAFLD had high risk of albuminuria, supporting that MAFLD criteria would be practical for the evaluation of long-term risk of subclinical atherosclerosis among fatty liver patients.
Article
Background: SARS-CoV-2 rapid antigen tests are an important public health tool. Objective: To evaluate field performance of the BinaxNOW rapid antigen test (Abbott) compared with reverse transcriptase polymerase chain reaction (RT-PCR) for detecting infection with the Omicron variant of SARS-CoV-2. Design: Cross-sectional surveillance study. Setting: Free, walk-up, outdoor, urban community testing and vaccine site led by Unidos en Salud, serving a predominantly Latinx community highly impacted by COVID-19. Participants: Persons seeking COVID-19 testing in January 2022. Measurements: Simultaneous BinaxNOW and RT-PCR from nasal, cheek, and throat swabs, including cycle threshold (Ct) measures; a lower Ct value is a surrogate for higher amounts of virus. Results: Among 731 persons tested with nasal swabs, there were 296 (40.5%) positive results on RT-PCR; 98.9% were the Omicron variant. BinaxNOW detected 95.2% (95% CI, 91% to 98%) of persons who tested positive on RT-PCR with a Ct value below 30, 82.1% (CI, 77% to 87%) of those who tested positive on RT-PCR with a Ct value below 35, and 65.2% (CI, 60% to 71%) of all who were positive on RT-PCR. Among 75 persons with simultaneous nasal and cheek swabs, BinaxNOW using a cheek swab failed to detect 91% (20 of 22) of specimens that were positive on BinaxNOW with a nasal swab. Among persons with simultaneous nasal and throat swabs who were positive on RT-PCR with a Ct value below 30, 42 of 49 (85.7%) were detected by nasal BinaxNOW, 23 of 49 (46.9%) by throat BinaxNOW, and 44 of 49 (89.8%) by either. Limitation: Participants were a cross-sectional sample from a community-based sentinel surveillance site, precluding study of viral or symptom dynamics. Conclusion: BinaxNOW detected persons with high SARS-CoV-2 levels during the Omicron surge, enabling rapid responses to positive test results. Cheek or throat swabs should not replace nasal swabs. As currently recommended, high-risk persons with an initial negative BinaxNOW result should have repeated testing. Primary funding source: University of California, San Francisco.
Article
Inducing durable and effective immunity against severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2) via vaccination is essential to combat the current pandemic of coronavirus disease 2019 (COVID-19). It has been noticed that the strength of anti-COVID-19 vaccination-induced immunity fades over time, which calls for an additional vaccination regime, as known as booster immunization, to restore immunity among previously vaccinated populations. Here we report a pilot open-label trial of a third dose of BBIBP-CorV, an inactivated SARS-CoV-2 vaccine (Vero cell), on 136 participants aged between 18 to 63 years. Safety and immunogenicity in terms of neutralizing antibody titers and cytokine/chemokine responses were analyzed as the main endpoint until day 28. While systemic reactogenicity was either absent or mild, SARS-CoV-2-specific neutralizing antibody titers rapidly arose in all participants within 4 weeks, surpassing the peak antibody titers elicited by the initial two-dose immunization regime. Broad increases of cellular immunity-associated cytokines and chemokines were also detected in the majority of participants after the third vaccination. Furthermore, in an exploratory study, a newly developed recombinant protein vaccine, NVSI-06-08 (CHO Cells), was found to be safe and even more effective than BBIBP-CorV in eliciting humoral immune responses in BBIBP-CorV-primed individuals. Together, these results indicate that a third immunization schedule with either homologous or heterologous vaccine showed favorable safety profiles and restored potent SARS-CoV-2-specific immunity, providing support for further trials of booster vaccination in larger populations.