You are on page 1of 20

The Lancet Regional Health - Americas

ed
Long-term analysis of antibodies elicited by Sputnik V in Tucumán, Argentina

iew
Rossana Elena Chahla1,†, Rodrigo Hernán Tomas-Grau2,†, Silvia Inés Cazorla3,†, Diego Ploper2,†,

Esteban Vera Pingitore2,†, Mónica Aguilar López4, Patricia Aznar4, María Elena Alcorta4, Eva

María del Mar Vélez4, Agustín Stagnetto2, César Luís Ávila2, Carolina Maldonado-Galdeano3,

ev
Sergio Benjamín Socias2, Dar Heinze5, Silvia Adriana Navarro2, Conrado Llapur1, Dardo Costas4,

Isolina Flores4, Alexis Edelstein6, Shreyas Kowdle7, Claudia Perandones6, Benhur Lee7, Gabriela

r
Apfelbaum8, Raúl Mostoslavsky9, Gustavo Mostoslavsky5, Gabriela Perdigón3*, Rosana Nieves

Chehín2*

Affiliations:
er
pe
1Tucumán Public Healthcare Administration (SIPROSA). Tucumán, Argentina.
2Instituto de Medicina Molecular y Celular Aplicada - IMMCA (UNT-CONICET-SIPROSA).
Tucumán, Argentina.
3Centro de Referencia para Lactobacilos – CERELA (CONICET). Tucumán, Argentina.
ot

4Néstor Kirchner Hospital, Central Public Health laboratory (LSP) (SIPROSA). Tucumán,
Argentina.
tn

5Section of Gastroenterology, Department of Medicine, Center for Regenerative Medicine


(CReM), Boston University School of Medicine. Boston, MA, USA.
6National Administration of Laboratories and Health Institutes of Argentina (ANLIS), Dr. Carlos
rin

G. Malbrán, Buenos Aires, Argentina.


7Department of Microbiology at the Icahn School of Medicine at Mount Sinai, New York, NY,
USA.
ep

8School of Medicine – Universidad Nacional de Tucumán. Tucumán, Argentina.


9The Massachusetts General Hospital Cancer Center, Harvard Medical School. Boston, MA,
USA.
Pr

†These authors contributed equally to this work

* Corresponding authors. Email: rosanachehin@gmail.com; perdigon@cerela.org.ar

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
ABSTRACT

ed
Background: Gam-COVID-Vac, also known as SPUTNIK V, is the first COVID-19 vaccine

registered, has emergency use authorization in 70 nations, and has been administered to millions

iew
worldwide. However, there are very few peer-reviewed studies describing its effects. As

independent reports begin to be published, a more accurate picture regarding safety and

effectiveness could accelerate approval by the WHO.

ev
Methods: An ELISA that detects anti-SARS-CoV-2-spike-RBD IgG was used, in both transversal

and longitudinal studies, to analyze humoral immune responses in 602 healthcare workers who

received SPUTNIK V between December 2020 and July 2021.

r
Findings: Seroconversion was detected in 97% of individuals after 28 days post-vaccination

er
(dpv). Anti-RBD titers began to decrease after 60 dpv, but remained detectable in 94% of

volunteers at 90 dpv. At 180 dpv, anti-RDB titers persisted in 31% of volunteers. Previous
pe
SARS-CoV-2 infection triggered increased immune response to the first dose, and increased

neutralization activity against different variants of concern. The second dose in previously

infected individuals further increased anti-RBD titers, even after 90 dpv. However, not all

individuals with previous infection responded equally, as only those with basal titers above a
ot

certain threshold showed robust responses. Time elapsed between COVID-19 diagnosis and

vaccination did not influence titers elicited.


tn

Interpretation: Data presented herein provides essential knowledge regarding the kinetics of

antibodies induced by SPUTNIK V up to six months after immunization, and suggests that when
rin

considering one-dose vaccination policies for individuals with previous SARS-CoV-2 infection,

serological studies to determine basal titers may be important, independent of when diagnosis

occurred.
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
1. INTRODUCTION

ed
The need to control the transmissibility and mortality associated with SARS-CoV-2 has

transformed vaccines into a critical resource to mitigate the devastating effects of the pandemic.1

iew
The appearance of new virus variants reinforces the need to accelerate vaccination rates in all

countries.2,3 However, despite the unprecedented speed of research, development and application

of different vaccines around the world, their availability remains limited in developing nations.

In March 2020, Argentina reported its first case of SARS-CoV-2 infection.4 Eight months later, it

ev
was the first of the Americas, and the 3rd in the world, to grant emergency use authorization for

the heterologous prime-boost recombinant adenoviral-based SPUTNIK V (Gam-COVID-Vac)

r
vaccine.5–7 As of August 2021, a total of 70 countries, mostly in Latin America, Asia and Africa,

er
have granted similar approvals. There is limited peer-reviewed published information on

SPUTNIK V, which is unique in that each dose is based on the combination two different human
pe
adenoviral vectors (rAd26 and rAd5).6 Determining how local populations respond to

government-led vaccination programs is essential for an effective public healthcare response to

the pandemic.
ot

The northwestern province of Tucuman, located in the Andean foothills and the second most

densely populated, has been one of the cities hardest-hit by the pandemic, with more than 143,000
tn

infections and 2,400 COVID-deaths in a population of 1.5 million. Here, we examined in 602
public healthcare-personnel (HCP) volunteers, with or without previous SARS-CoV-2 infection,

humoral immune responses elicited after with one or two doses, at different time-points and up to
rin

6 months post-vaccination with SPUTNIK V.


ep

3. RESULTS

Humoral immune responses against the receptor-binding domain (RBD) of the SARS-CoV-2

spike glycoprotein were assayed in 602 healthcare-personnel (HCP) volunteers vaccinated with
Pr

SPUTNIK V in Tucumán, Argentina. The basic demographics of the volunteer population studied

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
are shown in Table 1, and comprised of individuals whose ages ranged from 18-60 years old.

ed
Immune responses were analyzed using an enzyme-linked immunosorbent assay (ELISA) that

detects IgG antibodies targeting the SARS-CoV-2 spike glycoprotein receptor-binding domain

iew
(RBD) 8–10, which have demonstrated excellent correlation with virus neutralizing activity.11–15

The SPUTNIK V vaccination scheme and the days post-vaccination (dpv) in which serum

samples were collected from volunteers are depicted in Fig 1a. In a cross-sectional analysis of the

602 volunteers, basal anti-RBD IgG antibody titers were found in 44% of individuals before

ev
vaccination (dark and light purple), of which more than half (66%) lacked previous COVID

diagnosis (either by RT-PCR or rapid antigen tests) (light purple), implying they underwent

asymptomatic or unrecognized infections (Fig. 1b). Fourteen days after the first dose (14 dpv),

r
anti-RBD IgG antibody titers were detected in only 48% of volunteers (Fig. 1c), and displayed a

er
median antibody titer of 230. However, one week after completing the vaccination scheme (28

dpv), antibody titers were present in 97.6% of volunteers, in accordance with previous reports
6,16,17, and the median titer value rose to 1200 (Fig. 1c). Two months after initiating
pe
immunizations (60 dpv), median anti-RBD levels began to decrease, reaching a median titer of

500, and remained without significant variation at 90 dpv (Fig. 1c). Importantly anti-RBD titers

were still detectable in 31% of volunteers six months after vaccination (Fig. 1c). Titers elicited by
ot

309 convalescent individuals (previous positive RT-PCR), measured by our group with the same

platform but for a different study, are also displayed for comparison and showed a median titer of
tn

350 (Fig. 1c).10

For mRNA vaccine platforms, it has been reported that first doses may act as boosters in
rin

individuals previously exposed to SARS-CoV-2.18–20 In order to determine if this is also the case

in our cohort, a subset of 107 individuals were followed in a longitudinal study. Interestingly, the

group with previous SARS-CoV-2 infection showed a median anti-RBD titer 4.1-fold higher after
ep

the first dose (14 dpv) in comparison to individuals in the unexposed cohort (625 vs 150) (Fig.

2a). Nevertheless, the second SPUTNIK V dose further increased the median anti-RBD titer in

previously infected individuals compared to the control group (1350 vs 800) at 28 dpv. This
Pr

suggests that the first dose of SPUTNIK V in individuals pre-exposed to SARS-CoV-2 elicited a

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
secondary immune response. Due to the fact that high dropout rates were observed at different

ed
stages of the study, a full longitudinal analysis with this exact cohort for all time points was not

possible. Therefore, we evaluated in separate 60, 90, and 180 dpv longitudinal analyses the effect

iew
of previous SARS-CoV-2 infection on the levels of anti-RBD elicited by SPUTNIK V over time.

Upon examining antibody levels at 60 dpv (longitudinal study, N=75), the median titer elicited in

the group with previous infection was still significantly higher than the control group (795 vs

400) (Fig. 2b). This difference, while still significant, was slightly reduced after 90 dpv

ev
(longitudinal study, N=75) (Fig. 2c). However, 6 months after vaccination, anti-RBD titers were

diminished and no significant difference was observed between median titers elicited by both

groups (longitudinal study, N=35) (Fig 2d).

r
Despite the decrease in anti-RBD titers over the 6-month period, the anamnestic anti-RBD

er
binding response elicited by the SPUTNIK V in previously infected individuals resulted in

qualitatively different neutralizing responses compared with that from naïve vaccinees. This can
pe
be seen when we compared pooled sera from the 28 dpv group of uninfected volunteers (‘Group

1’, basal anti-RBD IgG negative) with the 14 dpv group of previously infected individuals

(‘Group 2’, basal anti-RBD IgG positive) (Fig. 3). Due to wide dispersion of anti-RBD titers in
ot

both groups, we pooled samples with the highest titers (>1,200) in both groups and assessed their

neutralizing activity against recombinant VSV engineered to express wild-type, B.1.1.7 (alpha),

B.1.351 (beta), or E484K mutant SARS-CoV-2 spike.21 Group 1 sera effectively neutralized all
tn

SARS-CoV-2 spikes except B.1.351 (Fig. 3a-c) as we and others have previously observed.22,23

Remarkably, Group 2 sera not only inhibited wild-type, B.1.1.7 and E484K viruses 5-20 fold
rin

better than Group 1 sera, they also potently neutralized B.1.351 (Fig. 3d-e). This marked increase

in neutralization potency and breadth can be seen clearly in the aggregated comparison of IC50s

across all the viruses examined (Fig. 3f).


ep

Within the population of volunteers that presented detectable anti-RBD antibodies before

vaccination, we noted a wide range of antibody responses to the first and second doses of
Pr

SPUTNIK V. Therefore, we analyzed if an antibody titer threshold could be determined above

which the vaccination elicited a robust and homogenous response (where median and mean titers

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
would be equal). We found that individuals with basal titers between 100-399 elicited a broad

ed
array of responses, although mostly weak, while volunteers with titers 400 or above induced a

stronger and uniform response to both the first (14 dpv) and second (28 dpv) doses of SPUTNIK

iew
V (Fig. 4a). Notably, in individuals previously infected with SARS-CoV-2, the time elapsed

between diagnosis (RT-PCR or rapid antigen test) and vaccination with the first dose (at least up

to 120 days) had no influence on antibody titers elicited either 14 or 28 dpv (Fig. 4b). In

conclusion, specific basal antibody titers had more influence on anti-RBD responses to

ev
vaccination than the time passed between diagnosis and vaccination.

r
4. DISCUSSION

er
Vaccination against SARS-CoV-2 with different platforms has accelerated worldwide. One of

such vaccines is SPUTNIK V, developed and produced by the Gamaleya National Research

Center of Epidemiology and Microbiology in Moscow, Russia.6,7 Despite the fact that more than
pe
70 countries, mainly in South America, Africa and Asia, have currently issued emergency

approval for this vaccine, very few reports have been published describing its effects and

efficiency outside of Russia.17 Studies, available through pre-print servers and official reports by
ot

ministries of health of different nations, have confirmed in large cohorts the safety and efficacy of

SPUTNIK V originally reported in Phase I/II and III studies.17,24 Nevertheless, SPUTNIK V has
tn

yet to be approved for emergency use by the World Health Organization and the European

Medicines Agency, the former of which is critical for this vaccine to be provided to lower-income

nations through the COVID-19 Global Access (COVAX) initiative.17 This lack of peer-reviewed
rin

published data is in stark contrast to the plethora of studies and wealth of information regarding

other vaccines like BNT162b2 (Pfizer/BioNTech), mRNA-1273 (Moderna/NIAID), AZD1222

(AstraZeneca/University of Oxford), and others.17,25-27 Therefore, it is essential that more peer


ep

reviewed data shed light on this vaccine, widely approved for emergency use.

The fact that Argentina was the second country beyond Russia to grant emergency approval for
Pr

the use of SPUTNIK V provided us with the opportunity to independently assess antibody titers

elicited, in a different population and up to six months post-vaccination. Although only 20% of

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
antibodies induced by the spike glycoprotein are directed against the RBD domain, 60% of these

ed
possess neutralizing activity, in comparison to the 80% of non-RBD antibodies, of which only 5%

show neutralize the virus.28 To our knowledge, this is the first report outside of Russia that

iew
determines the persistence and titers of these specific and highly neutralizing antibodies elicited

by SPUTNIK V.

In Argentina, as in most countries, HCP were among the first to receive COVID-19 vaccines due

to their increased contact with the virus and the strategic tasks they perform during a

ev
pandemic.29,30 For this reason, our study focused on HCP volunteers from the public health care

system, which were between 18 and 60 years of age and comprised of mostly female volunteers

r
(68%) (Table 1). Surprisingly, a great percentage (66%) of HCP that presented basal anti-RBD

titers lacked a previous COVID diagnosis, suggesting they underwent asymptomatic or

er
unrecognized infections, and highlighting the high level of viral exposure in this specific

population. Our study shows that only one week after receiving the second dose of SPUTNIK V,
pe
anti-RBD IgG antibodies were elicited in 97.3% of volunteers; a percentage of seroconversion

that is in accordance with the original phase I/II study, available preprints, and official non-peer-

reviewed national health ministry reports.6,16 Likewise, we also found no major safety issues
ot

related with vaccination, as only mild side-effects were reported in 21.9% of participants (Table

1), consistent with what has been reported previously.6,7,24 Although the phase I/II studies of

SPUTNIK V report higher anti-RBD titers measured by ELISA at 28 dpv than what we have
tn

found, this could be due to the fact that the anti-RBD ELISA platforms used in both studies were

not the same, as individuals who recovered from SARS-CoV-2 infection also showed lower anti-
rin

RBD titers with our platform.6,10 Importantly, we found that although anti-RBD titers begin to

drop at 2 months, antibodies are still present in 94.6% of volunteers after 3 months of vaccination

(Fig. 1c).
ep

A major question upon vaccination with different COVID-19 vaccine platforms around the globe

has been what type of humoral immune response would individuals who recovered from previous
Pr

SARS-CoV-2 infection have, compared to individuals without prior contact.31 For mRNA

vaccines, it has been shown that in previously infected individuals, the first doses essentially act

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
as boosters.18–20 This has also been reported for adenoviral-based platforms.16,32,33 In a context of

ed
high viral circulation, with millions of patients recovered from severe, mild or asymptomatic

COVID-19, this information could be important for designing future health policies, especially in

iew
areas of vaccine shortages such as developing nations. Here, we confirm that SPUTNIK V also

behaves similarly, in those individuals that underwent previous SARS-CoV-2 infection

(detectable basal anti-SARS-CoV-2 antibodies or positive RT-PCR/rapid antigen test) elicited

higher antibody titers than those without previous infection, inducing a secondary immune

ev
response to the first dose (Fig. 2). Recently, the neutralizing activity of post-Sputnik V

vaccination sera against alpha (B.1.1.7) and beta (B.1.351) variants of concern has been

characterized by the use of de novo generated replication-competent vesicular stomatitis virus

r
expressing various SARS-CoV-2 spike mutations.22 We now show that antibodies induced by the

er
first dose of SPUTNIK V in previously infected individuals (14 dpv) were above 10-fold more

potent in neutralization assays against these same variants compared to sera from uninfected

volunteers who received the complete vaccination scheme (28 dpv) (Fig. 3).
pe
Although previously infected individuals elicited higher anti-RBD titers post-vaccination with

SPUTNIK V, not all responded equally. We found that only volunteers that possessed basal anti-
ot

RBD IgG levels above 400 consistently reached high titers after the first dose (median of 2000),

which further increased after the second dose (median of 3600). Therefore, basal titer levels, not

only seropositivity or negativity, could be crucial when considering previous SARS-CoV-2


tn

infection status as a parameter to guide vaccination strategies or assess potential benefits, or not,

of administering second doses. Importantly, we also show that the time elapsed between previous
rin

COVID diagnosis and vaccination, at least up to 120 days, did not affect antibody titers (Fig. 4b).

This could also be important for implementing vaccination guidelines for previously infected

individuals, as there appeared to be no difference, at least between the timelines studied, in


ep

antibody responses to either the first or second dose.

For many infections, humoral immune kinetics show that antibody levels peak and then begin to
Pr

drop after a certain number of weeks.34 Following infection with SARS-CoV-2, antibodies begin

to wane after 3 to 8 months.35,36 Although the half-life of antibodies elicited by COVID-19

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
vaccines is still currently being analyzed, it is suggested that these can last as long as 6-8

ed
months.37–40 We have studied the humoral immune kinetics of anti-RBD antibodies elicited by

SPUTNIK V up to 180 days post-vaccination in both a transversal and longitudinal manner. To

iew
our knowledge, this is the first study reporting the effect of this vaccine at these time-points.

From our transversal study, we found that although anti-RBD titers begin to drop 60 days after

vaccination, antibodies are still present in 94.6% of volunteers at 90 dpv (Fig. 1c). Participant

dropout for the 60 and 90-day time-points, due to the effects of the second wave of COVID-19 in

ev
Argentina with subsequent restrictions and lockdowns, imposed challenges to our longitudinal

study. Nevertheless, we report in separate 60, 90, and 180-day longitudinal analyses with 98, 75,

and 35 volunteers, respectively, the levels of anti-RBD IgGs. We find that even 3 months after

r
vaccination (90 dpv), individuals with previous SARS-CoV-2 infection still had higher anti-RBD

er
titers than uninfected individuals, although the difference was less significant than earlier time-

points (Fig 2).


pe
Of note, 7 of the 602 (1.2%) fully vaccinated HCP volunteers, who are constantly exposed to high

viral loads and new variants of concern, contracted SARS-CoV-2 within 6 months post-

vaccination and corresponded to mild COVID-19 cases without need of hospitalization (Table 1).
ot

Our results, although they have been obtained in a smaller cohort, are in agreement and

comparable to the recently-published paper by Moriah Bergwerk and colleagues.41 In this study,

among 1497 fully-vaccinated health-care workers for whom RT-PCR data were available, 39
tn

SARS-CoV-2 (2.6 %) breakthrough infections were documented. Neutralizing antibody titers in

case patients during the peri-infection period were lower than those in matched uninfected
rin

controls. Higher peri-infection neutralizing antibody titers were associated with lower infectivity.

Most breakthrough cases were mild or asymptomatic, although 19% had persistent symptoms (>6

weeks). These findings allow to suggest that peri-infection neutralizing antibody titers correlated
ep

with the viral load and thus with the infectivity of breakthrough cases. Clearly, vaccine-induced

immunity has been shown to be greatly protective against clinical disease but somewhat less

protective against both infection and infectivity. Both studies confirm that, while both the Sputnik
Pr

V vaccine and the BNT162b2 vaccine are extremely effective, rare breakthrough infections carry

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
an infectious potential and create a special challenge, since such infections are often

ed
asymptomatic and may pose a risk to vulnerable populations.

Many countries are considering postponing the second dose of two-dose vaccination regimens in

iew
order to vaccinate more people.42 Our results with SPUTNIK V highlight the importance of

evaluating specific basal antibody titers in previously exposed individuals in order to better assess
optimization of vaccination strategies. In a context where combining different vaccine platforms

is under evaluation in order to counteract shortages or increase efficiency, here we provide the

ev
longest-term study describing anti-RBD IgG levels elicited after vaccination with SPUTNIK V,

which consists of 2 different adenoviral vector platforms and has been granted emergency

r
approval in more than 70 countries worldwide.43,44

2. METHODS

2.1 Population studied


er
pe
Healthcare personnel (HCP) from Tucumán corresponding to 12 different vaccination nodes, who

received SPUTNIK V between December 2020 and February 2021, were invited to participate in
ot

the study. The 602 enrolled volunteers were properly informed regarding their choice to
participate in the study. Protocols were approved by the regional ethics research boards (Ex. N°

3929-410-P-2020), following the Declaration of Helsinki. Personal data from all volunteers were
tn

encrypted. Eligibility criteria were age between 18 to 60 years without any COVID-19 symptoms

at the time of vaccination. Individuals provided a signed informed consent to be included in the
rin

database of the present study.

2.2 Anti-RBD IgG antibody ELISA assay


ep

Anti-RBD IgG titers were determined by an “In-House” ELISA developed and validated with

more than 758 samples in our laboratory10, following a protocol modified from Stadlbauer and
Pr

colleagues.45 which presents high sensitivity (92.2%) and specificity (100%). Briefly,

recombinant RBD from SARS-CoV-2 was obtained from HEK293 cells, which were previously

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
transduced with a pHAGE2 lentivirus (pHAGE2-RBD-His).46 The transgenic cell line generated

ed
constitutively secretes a his-tagged RBD. This protein domain subsequently purified from the

conditioning media by affinity chromatography and purity confirmed by SDS-PAGE. Purified

iew
RBD was immobilized in each well (0.1 µg) of 96-well flat polystyrene bottom plates (High

Binding, Half-Area, Greiner #675061). The antibody titer value representing the highest accuracy

(Sensitivity + Specificity) was calculated using receiver operating curve (ROC) to establish the

cutoff value. Titers were calculated as the dilution in which the optical density (OD450nm)

ev
obtained was equal to the cutoff. Blood samples for IgG anti-RBD detection were taken: a) the

same day before the first dose application (0 day post vaccine-dpv), b) 14 ± 1 days after first dose

(14 dpv), c) 28 ± 2 days after first dose (28 dpv) (which corresponds to 7 days after the second

r
dose). After peripheral blood extraction (5 ml), serum was obtained by spontaneous coagulation

er
within two hours of collection. The samples were stored at -20 oC until use.
pe
2.3 Virus neutralization assays

Virus neutralization assays were performed using recombinant VSV expressing the indicated

wild-type, B.1.1.7, B.1.351, or E484K mutant SARS-CoV-2 spikes on 293T-ACE2-TMPRSS2


ot

(F8-2 clone) cells exactly as described previously.22


tn

2.4 Statistical analysis

Statistical analyses were performed using non-parametric tests in Prism 8.0 software (GraphPad,
rin

San Diego, CA). The post-tests applied in each trial are indicated in the figure legends.
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
ed
FIGURES AND LEGENDS

Table 1

iew
Basic demographics of the volunteer HCP population studied.

r ev
er
pe
ot
tn
rin
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
ed
iew
r ev
er
pe
ot

Fig. 1. Humoral immune response to the SPUTNIK V vaccine. (a) Diagram depicting

SPUTNIK V vaccination scheme and the days when serum samples were extracted for antibody
tn

determination. (b) Distribution of the vaccinated population (n=602) according to the presence of

basal IgG anti-RBD and documented previous SARS-CoV-2 infection. (c) A cross-sectional
rin

study of 602 HCP volunteer anti-RBD titers, as measured by ELISA at 0, 14, 28, 60, 90 and 180

days post-vaccination (dpv). Statistical analyses were performed with Mann-Withney test.
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
ed
iew
r ev
er
pe
ot
tn

Fig. 2. Anti-RBD titers elicited after SPUTNIK V in individuals with or without previous
rin

SARS-CoV-2 infection measured up to 180 dpv. Anti-RBD titers in four separate longitudinal

case-controlled studies, in which HCP volunteers began the vaccination scheme with or without
ep

previous SARS-CoV-2 infection. Anti-RBD titers were measured at (a) 0, 14 and 28 dpv

(N=107), (b) 60 dpv (N=98), (c) 90 dpv (N=75) and (d) 180 dpv (N=35). Statistical analyses

were performed with Mann-Withney test.


Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
a b c

ed
150 wild type 150 wild type 150 wild type
B.1.1.7 B.1.1.7 B.1.1.7
% of no serum

% of no serum

% of no serum
B.1.351 B.1.351 B.1.351
E484K E484K E484K
100 100 100

50 50 50

iew
0 0 0
No 105 104 103 102 101 100 No 105 104 103 102 101 100 No 105 104 103 102 101 100
Serum Serum Serum
1/Serum Dilution 1/Serum Dilution 1/Serum Dilution

d e f
150 wild type 150 wild type 104 WT
% of no serum

% of no serum
B.1.1.7 B.1.1.7

ev
B.1.1.7
B.1.351 B.1.351 103
E484K B.1.351
100 100 E484K
E484K

1/IC50
102

101
50 50
100
0 0 10-1
No 105 104 103 102 101 100 No 105 104 103 102 101 100

r
7

51
T

4K
.
Serum Serum

.1

.3

8
.1

E4
.1
B

B
1/Serum Dilution 1/Serum Dilution

er
Fig. 3. Neutralization activity of antibody responses elicited by the Sputnik V vaccine in
pe
naïve versus previously infected individuals. Neutralizing activity of pooled serum samples

taken at 28 dpv (a-c) or 14 dpv (d-e) from naïve (Group 1) versus previously infected (Group 2)

individuals, respectively. Each pooled serum sample comprises of three serum samples from each
ot

group (see Fig. 1a) with anti-RBD IgG titers >1200. Nine and six serum samples from Group 1

and 2 were tested as three (a-c) and two (d-e) pooled samples, respectively. Neutralization was
tn

performed against recombinant VSV engineered to express wild-type, B.1.1.7 (alpha), B.1.351

(beta), or E484K mutant SARS-CoV-2 spike. Neutralization curves were generated using a serial
rin

four-fold dilution of serum (1:10 – 1:40,960) and normalized against the no-serum control set at

100% using nonlinear regression (PRISM v9.12). Data points represent mean +/- s.d. with each

neutralization curve performed in triplicates. The dotted line represents the normalized 50%
ep

infection mark. (f) The reciprocal IC50 (1/IC50) values from the nonlinear regression curves shown

in (a-e) are plotted for comparison. The colored symbols correspond to the viruses indicated. The
Pr

open and filled symbols represent the reciprocal IC50 values from Group 1 and Group 2 pooled

sera, respectively.

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
ed
iew
ev
Fig. 4 Influence of basal titer and time elapsed after SARS-CoV-2 detection on SPUTNIK V

r
humoral immune response. (a) Distribution of IgG anti-RBD antibody titers at 0, 14 and 28 dpv

er
in individuals previously infected with SARS-CoV-2, segregated according to basal titers of 100-

399 or ≥400. (b) Anti-RBD titers triggered by the first (14 dpv) and second (28 dpv) doses when
pe
administered less than 90, between 90-120, and more than 120 days post-diagnosis of SARS-

CoV-2 infection. Statistical analyses were performed with Kruskal-Wallis test. *** p<0.0001, **

p<0.005, and ns= not-significant.


ot
tn
rin
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
DATA AVAILABILITY

ed
Raw data acquired during this study is available from the corresponding author upon reasonable

request.

iew
ACKNOWLEDGMENTS

We thank Mr. Claude Burgio (SkyBio LLC), Ing. Luis Rocha, Ing. Marina Gandur, Dr. Christian

ev
Jaroszewski, Lic. Griselda Figueroa, Dr. María de los Angeles Peral de Bruno and Lic. Lorena

Naidicz. We thank Jared Feldman and Aaron Schmidt (Harvard University) for providing
valuable reagents. This research was supported by the Ministry of Public Health of Tucumán

r
(Argentina), Argentinean Research Council-CONICET (PIP 722 and 806), Argentinean Research

er
Agency-MINCYT grants (PICT-2018-3379 and PICT2018-02989), National University of

Tucuman (PIUNT-UNT D644/1 and D624), and Florencio Fiorini Foundation.


pe
AUTHOR CONTRIBUTIONS

Conceptualization: REC, RNC, GP, GA, CLL; methodology: RHTG, SIC, CMG, DP, EVP, DH,
ot

CLA, SN; data acquisition: PA, MEA, EMdMV, AS, CLA, SBS, CMG; data analysis and

visualization: RHTG, DP, SIC, GA, RNC, GP, GM, RM; funding acquisition: REC, RNC, DP,
tn

SBS, CA; project administration: REC, MAL; supervision: RNC, SC, EVP, DC, IF; writing: DP,

RNC, GP, RHTG, SC; writing – review & editing: DP, RHTG, SC, EVP, GM, RM, GA, RNC.
rin

COMPETING INTERESTS

B. L. is a named inventor on a patent filed by the Icahn School of Medicine which includes the
ep

293T-ACE2-TMPRSS2 (F8-2) cells used for the virus neutralization assay.


Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
REFERENCES

ed
1. Pollard AJ, Bijker EM. A guide to vaccinology: from basic principles to new developments. Nat
Rev Immunol 2021; 21(2): 83-100.
2. Boehm E, Kronig I, Neher RA, et al. Novel SARS-CoV-2 variants: the pandemics within the

iew
pandemic. Clin Microbiol Infect 2021.
3. Fontanet A, Autran B, Lina B, Kieny MP, Karim SSA, Sridhar D. SARS-CoV-2 variants and
ending the COVID-19 pandemic. Lancet 2021; 397(10278): 952-4.
4. Rabinovich GA, Geffner J. Facing up to the COVID-19 pandemic in Argentina. Nat Immunol
2021; 22(3): 264-5.

ev
5. Ministerio de Salud de la Nación. República Argentina. Informe de la ANMAT sobre la vacuna
Sputnik V. 23 December 2020. Available from: https://www.argentina.gob.ar/noticias/informe-de-la-
anmat-sobre-la-vacuna-sputnik-v.
6. Logunov DY, Dolzhikova IV, Zubkova OV, et al. Safety and immunogenicity of an rAd26 and

r
rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-
randomised phase 1/2 studies from Russia. Lancet 2020; 396(10255): 887-97.
7.

er
Logunov DY, Dolzhikova IV, Shcheblyakov DV, et al. Safety and efficacy of an rAd26 and rAd5
vector-based heterologous prime-boost COVID-19 vaccine: an interim analysis of a randomised controlled
phase 3 trial in Russia. Lancet 2021; 397(10275): 671-81.
8. Rowntree LC, Chua BY, Nicholson S, et al. Robust correlations across six SARS-CoV-2 serology
pe
assays detecting distinct antibody features. Clin Transl Immunology 2021; 10(3): e1258.
9. Amanat F, Stadlbauer D, Strohmeier S, et al. A serological assay to detect SARS-CoV-2
seroconversion in humans. Nat Med 2020; 26(7): 1033-6.
10. Tomas Grau RH, Ploper D, Ávila CL, et al. An “In-House” ELISA for SARS-CoV-2 RBD
uncovers elevated immune response at higher altitudes. medRxiv 2021; published online March 12.
ot

https://doi.org/10.1101/2021.03.10.21252711 (preprint).
11. Premkumar L, Segovia-Chumbez B, Jadi R, et al. The receptor binding domain of the viral spike
protein is an immunodominant and highly specific target of antibodies in SARS-CoV-2 patients. Sci
Immunol 2020; 5(48).
tn

12. Röltgen K, Powell AE, Wirz OF, et al. Defining the features and duration of antibody responses to
SARS-CoV-2 infection associated with disease severity and outcome. Sci Immunol 2020; 5(54).
13. Wajnberg A, Amanat F, Firpo A, et al. Robust neutralizing antibodies to SARS-CoV-2 infection
persist for months. Science 2020; 370(6521): 1227-30.
rin

14. Garcia-Beltran WF, Lam EC, Astudillo MG, et al. COVID-19-neutralizing antibodies predict
disease severity and survival. Cell 2021; 184(2): 476-88.e11.
15. Ju B, Zhang Q, Ge J, et al. Human neutralizing antibodies elicited by SARS-CoV-2 infection.
Nature 2020; 584(7819): 115-9.
ep

16. Rossi AH, Ojeda DS, Varese A, et al. Sputnik V Vaccine Elicits Seroconversion and Neutralizing
Capacity to SARS CoV-2 after a Single Dose. Cell Reports Medicine 2021;100359.
17. Nogrady B. Mounting evidence suggests Sputnik COVID vaccine is safe and effective. Nature
Pr

2021; 595(7867): 339-40.


18. Krammer F, Srivastava K, Alshammary H, et al. Antibody Responses in Seropositive Persons after
a Single Dose of SARS-CoV-2 mRNA Vaccine. N Engl J Med 2021; 384(14): 1372-4.

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
19. Prendecki M, Clarke C, Brown J, et al. Effect of previous SARS-CoV-2 infection on humoral and

ed
T-cell responses to single-dose BNT162b2 vaccine. Lancet 2021; 397(10280): 1178-81.
20. Saadat S, Rikhtegaran Tehrani Z, Logue J, et al. Binding and Neutralization Antibody Titers After
a Single Vaccine Dose in Health Care Workers Previously Infected With SARS-CoV-2. JAMA 2021;
325(14): 1467-9.

iew
21. Cho A, Muecksch F, Schaefer-Babajew D, et al. Antibody Evolution after SARS-CoV-2 mRNA
Vaccination. bioRxiv 2021; published online July 29. https://doi.org/10.1101/2021.07.29.454333 (preprint).
22. Ikegame S, Siddiquey MNA, Hung C-T, et al. Neutralizing activity of Sputnik V vaccine sera
against SARS-CoV-2 variants. Nat Commun 2021; 12: 4598.
23. Gushchin VA, Dolzhikova IV, Shchetinin AM, et al. Neutralizing Activity of Sera from Sputnik
V-Vaccinated People against Variants of Concern (VOC: B.1.1.7, B.1.351, P.1, B.1.617.2, B.1.617.3) and

ev
Moscow Endemic SARS-CoV-2 Variants. Vaccines 2021; 9: 779.
24. Pagotto V FA, Soriano MM, Diaz M, Braguisnky Golde M, Gonzalez MI, Asprea V, Staneloni I,
Vidal G, Silveira M, Zingoni P, Aliperti V, Michelangelo H, Figar S. Active surveillance of the Sputnik V
vaccine in health workers. medRxiv 2021; published online Februay 5.
https://doi.org/10.1101/2021.02.03.21251071 (preprint).

r
25. Lopez Bernal J, Andrews N, Gower C, et al. Effectiveness of the Pfizer-BioNTech and Oxford-
AstraZeneca vaccines on covid-19 related symptoms, hospital admissions, and mortality in older adults in

26.
er
England: test negative case-control study. BMJ 2021; 373: n1088.
Voysey M, Clemens SAC, Madhi SA, et al. Safety and efficacy of the ChAdOx1 nCoV-19
vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in
pe
Brazil, South Africa, and the UK. Lancet 2021; 397(10269): 99-111.
27. Baden LR, El Sahly HM, Essink B, et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2
Vaccine. N Engl J Med 2021; 384(5): 403-16.
28. Dejnirattisai W, Zhou D, Ginn HM, et al. The antigenic anatomy of SARS-CoV-2 receptor
binding domain. Cell 2021; 184(8): 2183-200.e22.
ot

29. Bandyopadhyay S, Baticulon RE, Kadhum M, et al. Infection and mortality of healthcare workers
worldwide from COVID-19: a systematic review. BMJ Glob Health 2020; 5(12).
30. Khunti K, Kamal A, Pareek M, Griffiths A. Should vaccination for healthcare workers be
tn

mandatory? J R Soc Med 2021; 114(5): 235-6.


31. Dolgin E. Is one vaccine dose enough if you've had COVID? What the science says. Nature 2021;
595(7866): 161-2.
32. Eyre DW, Lumley SF, Wei J, et al. Quantitative SARS-CoV-2 anti-spike responses to Pfizer-
rin

BioNTech and Oxford-AstraZeneca vaccines by previous infection status. Clin Microbiol Infect 2021;
https://doi.org/10.1016/j.cmi.2021.05.041.
33. Claro F, Silva D, Rodriguez M, Rangel R, de Waard JH. IgG Antibody response to the Sputnik V
vaccine: previous SARS-CoV-2 seropositive individuals might need just one vaccine dose. Int J Infect Dis
2021; https://doi.org/10.1016/j.ijid.2021.07.070.
ep

34. Sebina I, Pepper M. Humoral immune responses to infection: common mechanisms and unique
strategies to combat pathogen immune evasion tactics. Curr Opin Immunol 2018; 51: 46-54.
35. Chia WN, Zhu F, Ong SWX, et al. Dynamics of SARS-CoV-2 neutralising antibody responses
and duration of immunity: a longitudinal study. Lancet Microbe 2021; 2(6): e240-e9.
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468
36. Seow J, Graham C, Merrick B, et al. Longitudinal observation and decline of neutralizing

ed
antibody responses in the three months following SARS-CoV-2 infection in humans. Nat Microbiol 2020;
5(12): 1598-607.
37. Salvagno GL, Henry BM, Pighi L, De Nitto S, Gianfilippi GL, Lippi G. Three-month analysis of
total humoral response to Pfizer BNT162b2 mRNA COVID-19 vaccination in healthcare workers. J Infect
2021. https://doi.org/10.1016/j.jinf.2021.06.024.

iew
38. Barouch DH, Stephenson KE, Sadoff J, et al. Durable Humoral and Cellular Immune Responses 8
Months after Ad26.COV2.S Vaccination. N Engl J Med 2021. https://doi.org/10.1056/NEJMc2108829.
39. Doria-Rose N, Suthar MS, Makowski M, et al. Antibody Persistence through 6 Months after the
Second Dose of mRNA-1273 Vaccine for Covid-19. N Engl J Med 2021; 384(23): 2259-61.
40. Lippi G, Henry BM, Plebani M. Anti-SARS-CoV-2 Antibodies Testing in Recipients of COVID-

ev
19 Vaccination: Why, When, and How? Diagnostics (Basel) 2021; 11(6).
41. Bergwerk M, Gonen T, Lustig Y, et al. Covid-19 Breakthrough Infections in Vaccinated Health
Care Workers. N Engl J Med 2021. https://doi.org/10.1056/NEJMoa2109072.
42. Robertson JFR, Sewell HF, Stewart M. Delayed second dose of the BNT162b2 vaccine:

r
innovation or misguided conjecture? Lancet 2021; 397(10277): 879-80.
43. Shaw RH, Stuart A, Greenland M, et al. Heterologous prime-boost COVID-19 vaccination: initial

er
reactogenicity data. Lancet 2021; 397(10289): 2043-6.
44. Borobia AM, Carcas AJ, Pérez-Olmeda M, et al. Immunogenicity and reactogenicity of
BNT162b2 booster in ChAdOx1-S-primed participants (CombiVacS): a multicentre, open-label,
randomised, controlled, phase 2 trial. Lancet 2021; 398(10295): 121-30.
pe
45. Stadlbauer D, Amanat F, Chromikova V, et al. SARS-CoV-2 Seroconversion in Humans: A
Detailed Protocol for a Serological Assay, Antigen Production, and Test Setup. Curr Protoc Microbiol
2020; 57(1): e100.
46. Mostoslavsky G, Fabian AJ, Rooney S, Alt FW, Mulligan RC. Complete correction of murine
Artemis immunodeficiency by lentiviral vector-mediated gene transfer. Proc Natl Acad Sci U S A 2006;
ot

103(44): 16406-11.
tn
rin
ep
Pr

This preprint research paper has not been peer reviewed. Electronic copy available at: https://ssrn.com/abstract=3902468

You might also like