Invest Clin 62(Suppl. 2): 3 - 17, 2021 https://doi.org/10.22209/IC.v62s2a01
Corresponding author: Alexis Rodríguez-Acosta, Facultad de Farmacia, Universidad Central de Venezuela, Caracas,
República Bolivariana de Venezuela. Tel: +58 491 7243; E-mail: rodriguezacosta1946@yahoo.es
Production of equine sera as a potential
immunotherapy against COVID-19
Mariana V. Cepeda
1
,
Juan C. Jiménez
2
, Flor H. Pujol
3
, Héctor R. Rangel
3
, Carlos Bello
1
,
José Cubillan
3
, María L. Serrano
4
, Tony Chacón
1
, Antonietta Saba
1
, Miguel A. López
1
and
Alexis Rodríguez-Acosta
1,5
1
Biotecfar C.A, Facultad de Farmacia, Universidad Central de Venezuela, Caracas,
República Bolivariana de Venezuela.
2
Instituto de Inmunología “Dr. Nicolás Bianco”, Facultad de Medicina, Universidad
Central de Venezuela, Caracas, República Bolivariana de Venezuela.
3
Laboratorio de Virología Molecular, Centro de Microbiología y Biología Celular,
Instituto Venezolano de Investigaciones Científicas, Miranda, República Bolivariana
de Venezuela.
4
Unidad de Química Medicinal, Facultad de Farmacia, Universidad Central de Venezuela,
Caracas, República Bolivariana de Venezuela.
5
Laboratorio
de
Inmunoquímica y Ultraestructura, Instituto Anatómico “Dr. José
Izquierdo”, Facultad de Medicina, Universidad Central de Venezuela, Caracas, República
Bolivariana de Venezuela.
Key words: Equine antiserum; anti-RBD; immunotherapy; pandemic; SARS-CoV-2;
equine coronavirus.
Abstract. Emerging viruses such as the COVID-19-inducing virus, SARS-
CoV-2, represent a threat to human health, unless effective vaccines, drugs
or alternative treatments, such as passive immunization, become accessible.
Animal-derived immunoglobulins, such as equine immunoglobulins might be
useful as immunoprophylaxis or immunotherapy against this viral disease.
Therapeutic antibodies (Abs) for SARS-CoV-2 were obtained from hyperimmune
equine plasma using the Spike protein receptor binding domain (RBD) as an
immunogen. The presence of anti-RBD antibodies was evaluated by ELISA and
the titres of neutralizing antibodies were determined in viral cell culture. Im-
munized horses generated high-titre of anti-RBD antibodies with antiviral neu-
tralizing activity on Vero-E6 cells of 1/1,000. To minimize potential adverse
effects, the immunoglobulins were digested with pepsin, and purified to obtain
the F(ab’)2 fragments with the protocol standardized by Biotecfar C.A for
the production of snake antivenom. Pre-immune serum displayed an unexpect-
ed anti-RBD reactivity by ELISA (titre up to 1/900) and Western Blot, but no
4 Cepeda et al.
Investigación Clínica 62(Suppl. 2): 2021
Producción de un suero equino como inmunoterapia potencial
contra la COVID-19
Invest Clin 2021; 62 (Suppl. 2): 3-17
Palabras clave: Antisuero equino; anti-RBD; inmunoterapia; pandemia; SARS-CoV-2;
coronavirus equino.
Resumen. Los virus emergentes, como el virus causante de la COVID-19,
el SARS-CoV-2, representan una amenaza para la salud de la humanidad, mien-
tras no estén disponibles vacunas, medicamentos o tratamientos alternativos
eficaces, como la inmunización pasiva. Las inmunoglobulinas de producción
animal, como las de los equinos, pueden ser útiles como inmunoprofilaxis o
inmunoterapia contra esta enfermedad viral. Se produjeron anticuerpos tera-
péuticos (Abs) contra el SARS-CoV-2 a partir de plasma equino hiperinmune in-
munizado con el dominio de unión al receptor (RBD) de la proteína de la espiga
viral. La presencia de anticuerpos contra RBD se evaluó mediante ELISA y de
anticuerpos neutralizantes por inhibición del crecimiento del virus en cultivos
celulares. Los caballos inmunizados generaron títulos elevados de anticuerpos
anti-RBD con actividad neutralizante antiviral en células Vero-E6 de 1/1.000.
El suero preinmune mostró una reactividad anti-RBD por ELISA (título hasta
1/900) y Western Blot pero sin actividad neutralizante. Con el propósito de
disminuir posibles efectos adversos, se realizó la digestión proteica con pepsina
de las inmunoglobulinas y posterior purificación para obtener los fragmentos
F(ab’)2 empleando el protocolo utilizado por Biotecfar C.A para la producción
de los antivenenos de serpientes. El modelado del RBD del coronavirus equino
mostró que algunos de los epítopos conocidos del RBD del SARS-CoV-2 se con-
servaban estructuralmente en la proteína del coronavirus equino. Esto podría
sugerir que parte de la reactividad del suero preinmune al RBD del SARS-CoV-2
podría deberse a una exposición previa al coronavirus equino en el animal.
Received: 28-02-2021 Accepted: 11-06-2021
INTRODUCTION
SARS-CoV-2, the new coronavirus re-
sponsible for the disease COVID-19, belongs
to the family Coronaviridae, genus Betacoro-
navirus, subgenera Sarbecovirus. The virus
genome encodes for four structural proteins
and several non-structural proteins. Among
the structural proteins, the Spike protein
(S) contains two domains: S1 that includes
the receptor-binding domain (RBD), which
interacts with the human receptor angio-
neutralizing activity. Modelling of the RBD of equine coronavirus showed that
some of the known epitopes of SARS-CoV-2 RBD were structurally conserved in
the equine coronavirus protein. This might suggest that some of the reactivity
observed in the pre-immune serum to the SARS-CoV-2 RBD might be due to a
previous exposure to equine coronavirus.
Production of equine sera as immunotherapy against COVID-19 5
Vol. 62(Suppl. 2): 3 - 17, 2021
tensin-converting enzyme 2 (hACE2), and
S2 that includes the fusion domain, which
promotes the fusion of the viral envelope
with the cellular membrane, to promote vi-
ral entry, through an early or late endosomal
pathway (1, 2)
SARS-CoV-2 RBD has been
documented to enclose immune epitopes
proficient of stimulating antibodies able to
neutralise viral invasion and impede viral
entry by competing with hACE2 binding
(3)
or targeting the receptor-binding domain
(RBD) (4). Several authors have found that
SARS-CoV-2 RBD immunogen stimulated
upper titres of neutralizing antibodies (5)
and displaying a strong antibody response
in the immunized mammals and non-human
primates (6).
Even if vaccines are becoming available
to prevent COVID-19 (7), the pandemic is
still highly active, and vaccination of all the
population will not be feasible in a short
term (8). Thus, therapeutic tools will still be
needed for preventing the severe evolution
of the disease.
Equine-derived polyclonal antibodies
against several proteins have shown good ef-
ficacy against infectious diseases caused by
highly pathogenic agents, such as Ebola (9),
West Nile virus (10), H
5
N
1
influenza virus
(11), respiratory syncytial syndrome virus
(12) and Middle East respiratory coronavi-
rus syndrome (13).
Production of equine anti-SARS-CoV-2
antibodies have been reported in China (14-
19) India (20) and Argentina (21).
The aim of this study was the produc-
tion of F(ab’)2 anti-RBD equine antibodies
as a potential passive immunization medica-
tion against COVID-19.
MATERIALS AND METHODS
Virus antigen, plasma pools, animals and
reagents
Protein RBD antigen, expressed in
HEK293 line was purchased from Acro Bio-
system, USA. Specific hyper-immune horse
plasma (SHHP) was obtained of experimen-
tal animals from Biotecfar. C.A. Mice (Mus
musculus) NIH strain (18-20 g) of both sex-
es, purchased from the Instituto Nacional de
Higiene “Rafael Rangel”, Caracas, Venezu-
ela, were used for toxicity assays. Bovine se-
rum albumin (BSA), Tris-base and Tween-20
were from Sigma-Aldrich (USA). Pepsin
(from porcine gastric mucosa) 0.7 FIP-U/
mg) was from Merck (Germany). Goat anti-
horse F(ab’)
2 IgG conjugated with horserad-
ish peroxidase (HRP) was from LSBio (USA).
All other chemicals for buffers and solutions
were from Sigma-Aldrich (USA), unless oth-
erwise stated.
Experimental horses
Three male horses (Equus ferus ca-
ballus) were selected between the ages of 3
and 5 years old from the Purebred Racing
Race. These specimens were examined pre-
vious to starting the trial: general physical
evaluation, equine infectious anaemia test,
cellular and biochemical haematology and
a complete health plan was implemented in
accordance with national regulations. The
specimens were evaluated weekly over and
done with a complete physical examination
and cellular and biochemical haematology
(22). The horses were housed in individual
stalls or boxes, feed with Bermuda grass hay,
commercial concentrate and ad libitum wa-
ter, respecting all national and international
animal welfare conditions.
Ethical approval
This research has been approved by the
Bioethics Committee of Hospital Universita-
rio de Caracas, in Ordinary Meeting via online
N° 06 dated December 22, 2020, following
the norms obtained from the ARRIVE guide-
lines and were carried out in accordance with
the U.K. Animals (Scientific Procedures) Act,
1986 and associated guidelines, EU Directive
2010/63/EU for animal experiments. Trained
staff organised all the experimental methods
relating to the use of live animals.
6 Cepeda et al.
Investigación Clínica 62(Suppl. 2): 2021
Selection of immunogen
In order to guarantee a safe handling
of the material with which the horses were
immunised, it was decided to work with the
SARS-CoV-2 S protein RBD (N354D, D364Y),
which was expressed from human 293 cells
(HEK293) with a molecular mass of 27.0
kDa purchased from (Acro Biosystem, USA).
The protein migrates as 33-35 kDa under re-
ducing condition (SDS-PAGE) due to glyco-
sylation (data not shown).
Horse immunization
The selected horses received a variable
amount (100 to 600 µg/mL) of the RBD anti-
gen in 1 mL of PBS: first injection of 100 µg of
antigen in Complete Freund adjuvant at week
0; injection of 200 µg of antigen in Incomplete
Freund adjuvant at week 1; injection of 300 µg
of antigen in PBS buffer at week 2; injection
of 600 µg of antigen in PBS buffer at week 3;
injection of 600 µg of antigen in PBS buffer at
week 5. The antigen injection was subdermal
performed in the lateral region of the neck.
The sampling was carried out after elapsed
times and disinfection and asepsis of the jugu-
lar vein to recover the plasma was performed.
Preliminary sample collection (negative
control)
The previous sample extraction was car-
ried out from normal horses after disinfec-
tion and asepsis of the jugular vein to recov-
er the blood.
Preparation of equine IgG immunoglobu-
lins and F (ab’)
2
fragments
Plasma samples were taken every 7 days
(from day 14), storing them at 4°C prior to
processing and analysis. Each sample was
placed in a thermal bath at 56°C for one
hour, and at that time were centrifuged at
2000 rpm for 15 min and then pass through
a 0.45 µm filter. After the initial immuniza-
tion, horse’s plasmas were collected on the
days indicated in the previous table, to be
stored at 4°C for further processing and
analysis.
Obtaining purified immunoglobulins
A pool of horse anti-RBD sera from the
three horses was diluted with equivalent vol-
umes of saline solution, and 1/2 volume of a
saturated ammonium sulphate solution was
included at that time.
The mixture was stired together softly
at room temperature for 30 min and after-
ward centrifuged at 5000 rpm for 20 min.
The precipitates were running in saline solu-
tion, previously, 1/3 volume of ammonium
sulphate was slowly added. Subsequently,
incubation of the sample was kept at room
temperature for 30 min. Then, the solution
was centrifuged at 5000 rpm for 20 min. The
precipitates were dissolved in saline and dia-
lysed overnight at 4 °C to remove the ammo-
nium sulphate.
Preparation of F(ab)2 Fragments
For the preparation of F(ab’)2, the pro-
tocol standardised by Biotecfar C.A was
used, Pepsin was activated in order to elimi-
nate the Fc fragment from total IgG. Briefly,
the pH of the horse anti-RBD immunoglob-
ulins was upheld to 3.3 with 1 mol/L HCl.
The total IgG was diluted in a 1: 2 ratios and
then the pepsin was added at a concentra-
tion of 1.25 g/L. The enzymatic digestion
reaction was carried out at 30°C for 30 min.
Then was stopped to adjust the pH and carry
out the first precipitation of contaminating
proteins. It was filtered and the pH of the
filtrate was adjusted for a second precipita-
tion with ammonium sulphate, to recover
the hyper-immune proteins. This prepara-
tion was diafiltered with distilled water, be-
fore evaluation, and the protein was stored
at 4°C until use.
Analysis of antibodies against SARS CoV-2
by SDS-PAGE
The F(ab)
2
purity (5µg) was analysed
by non-reducing sodium dodecyl sulphate
polyacrylamide gel electrophoresis (10%
SDS-PAGE). After running, the SDS-PAGE
were stained with Coomassie brilliant blue.
Gel images after decolouration were cap-
Production of equine sera as immunotherapy against COVID-19 7
Vol. 62(Suppl. 2): 3 - 17, 2021
tured with a ChemiDoc MP imaging system
(Bio-Rad).
Determination of equine antibody titres
by Enzyme Linked Immunosorbent Assay
(ELISA)
RBD was diluted to 1 µg/mL in PBS pH
7.4, and added to 96-well polystyrene plates
(100 µL/well). The microtitre plates were
incubated at 4°C overnight. The plates were
then washed three times with PBS-Tween 20
(0.05%) and blocked with 5% skimmed milk
for 1 h at 37°C. Subsequently, the wells were
washed three times and the samples (serum
or diluted antibodies, 100 µL volume) were
added to each well; as negative controls PBS
or nonspecific antibodies were used. The
plates were placed at 37°C for one hour and
at that point incubated with horseradish per-
oxidase (HRP), and conjugated to anti-horse
IgG antibodies at room temperature for an-
other hour. After washing, 3,3’,5, 5’-tetra-
methyl-benzidine was added. The plates were
placed in the dark room for approximately 15
minutes with the chromogens and the reac-
tion was stopped by adding 50 µL well of 2M
H
2
SO
4
. Finally, the absorbance was measured
at 450 nm with a microplate reader (BioTek,
USA), and values twice times greater than
controls were considered positive.
Immunoblotting assays
The equine IgG immunoglobulins and F
(ab’)
2
fragments reactivity versus the SARS-
CoV-2 RBD protein were estimated using
western immunoblotting (23).
With this purpose, the gel was incu-
bated for 10 min in transfer solution (50
mM Tris-HCl at pH 8.0, containing 380 mM
glycine, 0.1% SDS and 20% methanol). After-
ward, the gel was located in a transfer cham-
ber, permitting the proteins to pass from the
polyacrylamide matrix to a nitrocellulose
membrane. This process was carried out at
180 milliamps (mA) for 2 h.
Next, the transfer, the nitrocellulose
membrane was blocked for 2 h at room tem-
perature with a 0.2 M PBS solution at pH
7.0, with 5% (w/v) skimmed milk and 0.1%
(w/v) Tween 20. Following, three washes were
carried out for 5 min, each with a solution
of 0.05% (w/v) Tween 20 and 0.2 M PBS at
pH 7.0. The membrane was incubated over
at room temperature for another 90 min,
with equine IgG immunoglobulins and F
(ab’)
2
fragments diluted to 1:10 in blocking
solution. Next, after the incubation inter-
val, the membrane was washed three times.
Straightaway after the washes, the secondary
antibody anti-equine IgG (coupled to horse-
radish peroxidase) diluted 1:10 in blocking
solution was added. Then, the membrane
was incubated at room temperature for an-
other 90 min and washed, as specified. The
electrophoretic bands recognised by equine
IgG immunoglobulins and F (ab’)
2
fragments
were visualised and the image was analysed.
Anti-SARS-CoV-2 serum toxicity test
The toxicity assay of anti COVID-19 se-
rum was carried out by intraperitoneal injec-
tion of 0.5 mL of the serum in NIH mice. The
clinical toxicological manifestations were re-
corded during a period of 7 days. None of the
mice showed signs of clinical toxicity after
seven days of anti-SARS-CoV-2 serum injec-
tion. All animals were considered healthy
with a normal behaviour.
Cell line
Vero-E6 cells was maintained in RPMI-
1640 medium (Gibco, NY, USA) supplement-
ed with 8% foetal bovine serum (FBS, Gibco,
NY, USA) were cultured in an incubator at
37 °C with 5% CO
2
.
Neutralization assay
Vero-E6 cells were seeded in 96-well
plates with 2 × 10
4
cells/well overnight in
8% FBS-RPMI medium supplemented with
1600 U/mL of penicillin, 800 µg/mL of
streptomycin and 10 µg/mL of amphotericin
B. Horse sera were diluted in culture medi-
um and incubated with 10 TCID50 (10 times
the tissue culture infective dose, which pro-
duce cytopathic effect in half of the plates)
8 Cepeda et al.
Investigación Clínica 62(Suppl. 2): 2021
of SARS-CoV-2 at 37°C for 1 h. The mixture
was then added to the cells and incubated
at 37 °C for 1 h. The cells were washed with
PBS and incubated in the culture medium.
The cytopathic effect (CPE) was examined
with an inverted microscope after three days
post-infection. The neutralization titre was
defined as the dilution of sample where 50%
of the wells exhibited no CPE (24).
Phylogenetic analysis
Coronavirus RBD were aligned using
DNAman version 5.2.2 (Lynnon Biosoft, Can-
ada) and BLOSUM matrix. Phylogenetic tree
constructed with Poisson correction and 100
bootstrap replicas. RBD alignment was also
analysed for protein conservation using PRA-
LINE sequence alignment (https://www.ibi.
vu.nl/programs).
Protein modelling
The crystal structure of the RBD of
SARS-CoV-2 bound to neutralizing antibody
CR3022 (PDB code 6W41) was retrieved and
selected for the comparative analyses. Homol-
ogy structural model of the RBD of Equine
coronavirus, was generated using the crystal
structures of the RBD domain of the human
coronavirus OC43(PDB code 6nzk) and the
beta-coronavirus HKU1 (PDB code 5kwb) as
templates. The model was obtained with the
Phyre2 modelling server using the intensive
modelling mode (25). The quality of the mod-
els was established via ProSA (26) and PRO-
CHECK programs (27). The Deep-View/Swiss-
PdbViewer 4.01 and Biovia Discovery Studio
Visualizer 17.2.0 were used to superposition
and visualisation of protein structures.
B-cell epitope prediction
EIDB (Immune-Epitope-Database and
Analysis-Resource) were used to predict lin-
ear and discontinuous B-cell epitopes using
Bepipred and ElliPro with default parameter
settings (28). Biovia Discovery Studio Visu-
alizer 17.2.0 software was used to examine
the positions of discontinuous epitopes on
the 3D structures (29).
Protein-protein docking
The crystal structure of the RBD of
SARS-CoV-2 bound to neutralizing antibody
CR3022 (PDB code 6W41) was downloaded
from Protein Data Bank. In addition, the ho-
mology model for the RBD of Equine corona-
virus was assayed. The viral spike RBD and the
structural model of the ECoV RBD were eval-
uated against the CR3022 antibody through
molecular docking. Then, the obtained bind-
ing patterns and affinity estimations were
analysed and compared. This process was
performed through two steps; first, ProABC-2
(30) was used to identify the paratope resi-
dues in CR3022 and the information was
utilized to drive the modelling of antibody–
antigen complexes using HADDOCK2.4 web
server (31) and the best model was further
refined with HADDOCK Refinement inter-
face. Then, the resulting docking data were
processed and analysed by using the tools of
PRODIGY software (32). Finally, results were
evaluated considering binding energies and
main interacting residues in each complex.
RESULTS
Horse immunization
Three male horses were immunized with
the Spike RBD viral recombinant protein.
No major adverse effects or chronic clini-
cal alterations were observed for any of the
horses. The evaluation of immunized horse’s
sera binding to the SARS-CoV-2 RBD as as-
sessed by ELISA, further validated the quali-
ties of RBD antigens tested in this study and
is shown in Fig. 1. Antibody titers were devel-
oped since the first week of immunization,
increasing with the boosts of weeks 1, 2, 3
and 5, reaching a titer of 1/24,300 main-
tained after 50 days. Some antibody reactiv-
ity was observed for the pre-immune equine
serum at 1/100 (not shown in the Fig.1: OD
1.1 in pre-immune serum vs 3.4 in the im-
munised horse), 1/300 and even 1/900 (Fig.
1). This reactivity was not observed when the
ELISA plates were not coated with the RBD
antigen (OD 0.1 in the pre-immune serum).
Production of equine sera as immunotherapy against COVID-19 9
Vol. 62(Suppl. 2): 3 - 17, 2021
Anti-SARS-CoV-2 serum toxicity test
None of the mice showed signs of clini-
cal toxicity after seven days of anti-SARS-
CoV-2 serum injection. All animals were con-
sidered healthy with a normal behaviour.
Determination of equine antibody titres
by Enzyme Linked Immunosorbent Assay
(ELISA)
The evaluation of immunised horse’s
sera binding to the SARS-CoV-2 RBD as mea-
sured by ELISA binding to RBD, further vali-
dated the qualities of RBD antigens tested in
this study is showed in Fig. 1.
Purification and characterisation of the
F(ab’)2 preparation
The purity of the F(ab’)2 anti-RBD S
protein fragments was evaluated by 10%
SDS-PAGE under reduced conditions. Three
conspicuous bands between 90 and 150
kDa bands were observed (data not shown).
Specificity of anti SARS-CoV-2 to RBD S
protein via Western blot
Western Blot analysis of the antibody
reactivity showed the specific recognition of
the RBD antigen by the immune serum, to-
gether with another band of higher molecu-
lar weight, also recognised by other immune
equine sera, probably a contaminant of the
antigen preparation. Some recognition of
the RBD band was observed in the pre-im-
mune serum, together with the other upper
band (Fig. 2, band 5).
Determination of the neutralizing activity
of the F(ab’)2 preparations
Two F(ab’)2 preparations (1 and 2, at
the middle and the end of immunization,
respectively) were evaluated for the pres-
ence of total and neutralizing antibodies
against SARS-CoV-2 RBD (Fig. 3). As ex-
pected, the titre of both anti-RBD antibod-
ies and neutralizing antibodies increased
with time and immunization boosts. The
titre of neutralizing antibodies was ap-
proximatively 1/550 for the first F(ab’)2
preparation and 1/2,300 for the second
one. Pre-immune and F(ab’)2 preparations
against other antigen (anti-venom against
snake or scorpion) did not produce any vi-
ral neutralization, even at 1/30 dilution
(data not shown).
Fig. 1. Evaluation of horse sera binding to the SARS-CoV-2 RBD as measured by ELISA. One (1.0) µg/mL
of RBD protein was coated and 7-fold serially diluted serum was supplemented after blocking. Expe-
riments were carried out in triplicate and the error bars denote ± SE, n = 2. Statistical significance
was defined as * P < 0.05. O.D: optical density.
10 Cepeda et al.
Investigación Clínica 62(Suppl. 2): 2021
Analysis of the possible cross-reactivity
of antibodies against equine coronavirus
RBD and SARS-CoV-2 RBD
Since the equine pre-immune serum
exhibited some degree of reactivity, both
by ELISA and Western Blot, the presence
of epitopes shared between the RBD of the
Equine Coronavirus (ECoV) and SARS-CoV-2
(33) was evaluated. ECoV and SARS-CoV-2
RBD shares only 25% protein homology (Fig.
Fig. 2. Western blotting. (1 and 2) Polyvalent anti-ophidic sera. (3) Anti-scorpion serum. (4) anti-RBD S
protein [oval]. (5) Normal equine serum. 6) Membrane stained with red Ponceau. (7) Molecular mass
markers. RBD band is shown with a circle.
Fig. 3: Anti-RBD and neutralizing titres in F(ab’)2 preparations produced at different immunization
times. F(ab’)2 preparation 1 was obtained after 3 immunizations and preparation 2 after the com-
plete scheme. The neutralization titre is the highest dilution of serum that prevents infection of 50%
of replicate inoculations. Neutralization tests were run in triplicate for Preparation 1 and duplicate
for Preparation 2, with 3 replicas each time.
Production of equine sera as immunotherapy against COVID-19 11
Vol. 62(Suppl. 2): 3 - 17, 2021
4A). Three domains known as main epitope
regions in the SARS-CoV-2 RBD did not ex-
hibit neither more conservation. Only one
of them (epitope region 2) shared 30 % of
sequence identity among them (Fig. 5B).
However, structural modelling of the ECoV
RBD showed that epitope domains 1,2 and 3
share structural homology (Fig. 4B).
ECoV and SARS-CoV-2 RBDs were also
analysed in order to evaluate not only identi-
cal amino acids but also conservative substi-
tutions, particularly in the structurally con-
served regions (Fig. 5).
Additionally, by docking studies, the
binding free energy of a monoclonal anti-
body against SARS-CoV-2 RBD (CR3022), to
the EqCoV RBD was -10.5 Kcal/mol, similar
to the one obtained for SARS-CoV-2 RBD
(-14.9 Kcal/mol). CR3022 interacts with
EqCoV RBD and recognizes a discontinuous
epitope that includes several residues on the
same region (epitope 2) than with SARS-
CoV-2 (Fig. 6). Although the SARSCoV2
and ECoV RBD epitopes do not completely
overlap and different binding modes were
determined, the interaction energy suggests
that CR3022 might show some degree of
ECoV recognition. Interestingly, the epitope
of CR3022 does not overlap with the ACE2-
binding site of the RBD SARS-CoV-2 (34).
Fig. 4. Protein and structural homology between ECov and SARS-CoV-2 RBD. (A) Phylogenetic analysis
of RBD proteins from different coronavirus. Percent identity between ECoV and SARS-CoV-2 RBDs is
shown. The sequences are entitled with their accession or GISAID number. (B) Structural modelling
of EqCoV RBD and comparison with SARS-CoV-2 structure. Epitope domains are shown in colours.
12 Cepeda et al.
Investigación Clínica 62(Suppl. 2): 2021
Fig. 5. Analysis of conservative substitutions between SARS-CoV.2 and ECoV RBD. All epitope domains are
underline in colours.
Fig. 6. A. Interaction of monoclonal antibody CR30221 with Coronavirus RBDs. Binding free energies cal-
culated for the interaction of CR3022 with SARS-CoV-2 RBD or ECoV RBD are shown. B. Aminoacids
of the respective RBDs involved in the interaction with CR30221 are observed.
Production of equine sera as immunotherapy against COVID-19 13
Vol. 62(Suppl. 2): 3 - 17, 2021
DISCUSSION
COVID-19 is one of the most important
highly pathogenic viral disease at present.
The COVID-19 pandemic has keep on for al-
most a year since the beginning of 2021, and
it is expected that this severe viral pathology
will pursue as an endemic disease for many
years (33). The response to the international
health crisis necessitates a fast answer from
the medical organizations in the form of a
prompt treatment, while vaccines become
available for all individuals. Passive immuno-
therapy with equine antibodies may help to
improve the medical treatment of the SARS-
CoV-2.
In this study, we report the prepara-
tion of hyper-immune equine sera to dem-
onstrate their protective efficacy against
SARS-CoV-2 virus, using a virus neutraliza-
tion assay. The production of equine anti-
bodies was obtained by immunising horses
with the recombinant RBD antigen. High
titres of anti-RBD antibodies were obtained
in horses after a five-boost immunization
scheme. Western Blot analysis confirmed
the specific recognition of these antibodies
of the RBD protein. To minimise potential
adverse effects, the immunoglobulins were
digested with pepsin, and purified to obtain
the F(ab’)2 fragments. The F(ab’)2 prepa-
ration unambiguously recognize the RBD
antigen and exhibited anti-SARS-CoV-2
neutralizing activity. Moreover, the ratio of
neutralizing to binding antibodies obtained
in the present work and the seroconversion
rate observed in horses after 49 days of im-
munization indicates the high quality of
the antibodies generated by using the RBD
spike protein as immunogen. Other studies
have described slightly higher titres of an-
tibodies against RBD than the one found in
this study (15,21). These differences might
be due to the immunization schemes used
in each study. In our study, the RBD dose
was lower than the one used in these stud-
ies, but with an additional booster dose.
The titres of neutralizing antibodies found
in the current report were comparable to
the ones previously described (15,21). The
hyper-immune equine sera was purified to
obtain the F(ab’)2 fragments with the pro-
tocol standardised by Biotecfar C.A for
the production of snake and scorpion an-
tivenom, and produces a satisfactory yield
and adequate purity of preparations. The
antivenoms generated using ammonium
sulphate fractionation protocol have prov-
en safe and effective in patients suffering
snakebite and scorpion envenoming.
Pre-immune serum exhibited some de-
gree of cross reactivity with the SARS-CoV-2
RBD by ELISA and Western Blot, but not in
the neutralization test. In contrast to the
F(ab’)2 preparations against ophidic or scor-
pionic venoms, the pre-immune serum of the
horses used in this study exhibit a light spe-
cific recognition of the SARS-CoV-2 RBD by
Western Blot.
ECoV belongs to Betacoronavirus spe-
cies, subgenus Embecovirus and was first
isolated from a diarrheic foal in the USA in
1999 (35). Since then, several cases of ECoV
infections have also been reported in adult
horses from the USA, Europe and Japan.
This coronavirus has been detected in fae-
cal samples from horses with clinical signs
such as anorexia, lethargy, fever and, less
frequently, diarrhoea, colic and neurologic
deficits. The morbidity rate varies from 10%
to 83% during outbreaks, with low mortality
(35, 36).
Seroprevalence of equine coronavirus in
the U.S.A is relatively high: 9.3% (37). Even
if SARS-CoV-2 and ECoV belong to different
subgenera, the cross-reactivity observed in
the pre-immune serum prompt us to evalu-
ate if these two coronaviral RBDs might
share some epitopes. Structural modelling
of ECoV and SARS-CoV-2 RBD S showed that
these two proteins shared the structural mo-
tifs of three of the main epitope domains rec-
ognised in the SARS-CoV-2 RBD. Moreover,
docking analysis showed that one monoclo-
nal antibody produced against SARS-CoV-2
RBD exhibited a similar binding energy to
14 Cepeda et al.
Investigación Clínica 62(Suppl. 2): 2021
the ECoV RBD. It is worth to mention that
the epitope of CR3022 does not overlap with
the ACE2-binding site of the RBD SARS-
CoV-2 (34).
Poly-specific interaction of an antibody
with two epitopes unrelated in terms of se-
quence homology has been described (37).
In this study, no neutralizing activity was
found associated to this pre-immune cross-
reactivity. Nevertheless, (15) observed a
partial neutralizing activity in pre-immune
horse serum at low dilution. This observa-
tion is in agreement with the cross-reactivity
found in this study.
Cross reactivity with other coronavirus-
es in immunoassays, detecting antibodies
against SARS-CoV-2 in humans has also been
suggested (38). It has been also proposed
that previous exposure to other human
coronaviruses might bring some protection
for COVID-19 (39). Moreover, some cross-
neutralization activity against SARS-CoV-2
has been described in some intravenous im-
munoglobulin preparations (40). Ladner et
al. (41) proposed that the SARS-CoV-2 pro-
teome reveals regions of conservation with
endemic human coronaviruses (CoVs), but
it keep unidentified to what degree of these
may be cross-recognized by the antibody re-
sponse. The SARS-CoV-2 response seems to
be modulated by previous CoV exposures and
which have the capacity to increase roughly
neutralizing responses
To conclude, in the current work, we
prepared horse anti-RBD S protein serum by
immunizing equines with the SARS-CoV-2
RBD. In our program, the horses could be
immunized numerous times, and satisfacto-
ry titres of antibodies are predictable to be
achieved from hyper-immune horse’s plasma
for the SARS-CoV-2 therapeutic (42). Work-
ing with a scheme comprising increase im-
munogen injections, high-titre of horse an-
tisera were achieved, and F(ab’)2 fragments
have been mass-produced using good man-
ufacturing practices (GMP), in order to be
used in human clinical studies.
ACKNOWLEDGEMENTS
We wish to thank the helpful commen-
taries from Dr. Guillermo León, Instituto
“Clodomiro Picado” República de Costa Rica
and Dr. Christian Dokmetjian A, Director of
the Instituto Nacional de Producción de Bi-
ológicos de la ANLIS, Secretaria de Gobierno
de Salud de la Nación, República Argentina.
Funding for the research was provided by
Biotecfar C.A and Ministerio del Poder Popu-
lar para Ciencia y Tecnología de la República
Bolivariana de Venezuela.
REFERENCES
1. Ortega JT, Zambrano JL, Jastrzebska B,
Liprandi F, Rangel HR, Pujol FH. Unders-
tanding Severe Acute Respiratory Syndro-
me coronavirus 2 Replication to design
efficient drug combination therapies. Inter-
virology 2020; 1–8.
2. Hoffmann M, Kleine-Weber H, Schroeder
S, Krüger N, Herrler T, Erichsen S. SARS-
CoV-2 cell entry depends on ACE2 and TM-
PRSS2 and is blocked by a clinically proven
protease inhibitor. Cell 2020; 181:271-280.
3. Mittal A, Manjunath K, Ranjan RK, Kaus-
hik S, Kumar S, Verma V. COVID-19 pan-
demic: Insights into structure, function,
and hACE2 receptor recognition by SARS-
CoV-2. PLoS Pathog 2020; 16: e1008762.
4. Tian X, Li C, Huang A, Xia S, Lu S, Shi Z,
Lu L, Jiang S, Yang Z, Wu Y, Ying T. Potent
binding of 2019 novel coronavirus spike
protein by a SARS coronavirus-specific hu-
man monoclonal antibody. Emerg Microbes
Infect 2020; 9:382-385.
5. Ravichandran S, Coyle EM, Klenow L,
Tang J, Grubbs G, Liu S. Antibody signatu-
re induced by SARS-CoV-2 spike protein im-
munogens in rabbits. Sci Transl Med 2020;
12: eabc3539.
6. Yang J, Wang W, Chen Z, Lu S, Yang F, Bi
Z., Bao L, Mo F, Li X, Huang Y, Hong W,
Yang Y, Zhao Y, Ye F, Lin S, Deng W, Chen
H, Lei H, Zhang Z, Luo M, Gao H, Zheng
Y, Gong Y, Jiang X, Xu Y, Lv Q, Li D, Wang
M, Li F, Wang S, Wang G, Yu P, Qu Y, Yang
L, Deng H, Tong A, Li J, Wang Z, Yang J,
Production of equine sera as immunotherapy against COVID-19 15
Vol. 62(Suppl. 2): 3 - 17, 2021
Shen G, Zhao Z, Li Y, Luo J, Liu H, Yu
W, Yang M, Xu J, Wang J, Li H, Wang H,
Kuang D, Lin P, Hu Z, Guo W, Cheng W, He
Y, Song X, Chen C, Xue Z, Yao S, Chen L,
Ma X, Chen S, Gou M, Huang W, Wang Y,
Fan C, Tian Z, Shi M, Wang FS, Dai L, Wu
M, Li G, Wang G, Peng Y, Qian Z, Huang
C, Lau JY, Yang Z, Wei Y, Cen X, Peng X,
Qin C, Zhang K, Lu G, Wei X. A vaccine
targeting the RBD of the S protein of SARS-
CoV-2 induces protective immunity. Natu-
re 2020; 586(7830):572-577.
7. Callaway E, Ledford H, Viglione G, Watson
T, Witze A. COVID and 2020: An extraor-
dinary year for science. Nature 2020;588
(7839): 550-552.
8. Mills MC, Salisbury D. The challenges of
distributing COVID-19 vaccinations. Clini-
cal Medicine 2020; 100674. Online ahead
of print.
9. Zheng X, Wong G, Zhao Y, Wang H, He S,
Bi Y, Chen W, Jin H, Gai W, Chu D, Cao
Z, Wang C, Fan Q, Chi H, Gao Y, Wang T,
Feng N, Yan F, Huang G, Zheng Y, Li N,
Li Y, Qian J, Zou Y, Kobinger G, Gao GF,
Qiu X, Yang S, Xia X. Treatment with hype-
rimmune equine immunoglobulin or immu-
noglobulin fragments completely protects
rodents from Ebola virus infection. Sci Rep
2016; 24179.
10. Nybakken G E, Oliphant T, Johnson S,
Burke S, Diamond M S, Fremont DH.
Structural basis of West Nile virus neutra-
lization by a therapeutic antibody. Nature
2005; 437: 764-769.
11. Knossow M, Gaudier M, Douglas A, Ba-
rrere B, Bizebard T, Barbey C, Gigant B,
Skehel JJ. Mechanism of neutralization of
influenza virus infectivity by antibodies. Vi-
rology 2002; 302: 294-298.
12. Martínez I, Melero JA. Enhanced neutra-
lization of human respiratory syncytial vi-
rus by mixtures of monoclonal antibodies
to the attachment (G) glycoprotein. J Gen
Virol 1998; 79 (Pt 9), 2215-2220.
13. Yang S, Xia X. Passive immunotherapy for
Middle East Respiratory Syndrome corona-
virus infection with equine immunoglobu-
lin or immunoglobulin fragments in a mou-
se model. Antivir Res 2017; 137.
14. Pan X, Wu Y, Wang W. Development of hor-
se neutralizing immunoglobulin and im-
munoglobulin fragments against Junin vi-
rus. Antivir Res 2020; 174: 104666.
15. Pan X, Zhou P, Fan T, Wu Y, Zhang J, Shi
X, Shang W, Fang L, Jiang X, Shi J, Sun Y,
Zhao S, Gong R, Chen Z, Xiao G. Immu-
noglobulin fragment F(ab’) 2 against RBD
potently neutralises SARS-CoV-2 in vitro.
Antiviral Res 2020; 182:104868.
16. Wu C, Liu Y, Yang Y, Zhang P, Zhong W,
Wang Y, Wang Q, Xu Y, Li M, Li X, Zheng
M, Chen L, Li H. Analysis of therapeutic
targets for SARS-CoV-2 and discovery of po-
tential drugs by computational methods.
Acta Pharm Sin B 2020; 10:766-788.
17. Wu Z, McGoogan JM. Characteristics of
and important lessons from the coronavi-
rus disease 2019 (COVID-19) outbreak in
china: summary of a report of 72314 cases
from the Chinese Center for Disease Con-
trol and Prevention. JAMA 2020; 323: 1239-
1242.
18. Hui DS. I Azhar E, Madani TA, Ntoumi F,
Kock R, Dar O, Ippolito G, Mchugh TD,
Memish ZA, Drosten C, Zumla A, Peter-
sen E. The continuing 2019-nCoV epide-
mic threat of novel coronaviruses to global
health – the latest 2019 novel coronavirus
outbreak in Wuhan, China. Intl J Infect Dis
2020; 91: 264-266.
19. Deng SQ, Peng H J. Characteristics of and
public health responses to the coronavirus
disease 2019 outbreak in China. J Clin Med
2020; 9: 575-581.
20. Deshpande GR, Sapkal GN, Tilekar BN,
Yadav PD, Gurav Y, Gaikwad S, Kaushal
H, Deshpande KS, Kaduskar O, Sarkale
P, Baradkar S, Suryawanshi A, Lakra R,
Sugunan AP, Balakrishnan A, Abraham P,
Salve P. Neutralizing antibody responses to
SARS-CoV-2 in COVID-19 patients. Indian J
Med Res 2020; 152: 82-87.
21. Zylberman V, Sanguineti S, Pontoriero AV,
Higa SV, Cerutti ML, Morrone Seijo SM,
Pardo R, Muñoz L, Acuña Intrieri ME, Al-
zogaray VA, Avaro MM, Benedetti E, Ber-
guer PM, Bocanera L, Bukata L, Bustelo
MS, Campos AM, Colonna M, Correa E,
Cragnaz L, Dattero ME, Dellafiore M, Fos-
caldi S, González JV, Guerra LL, Klinke S,
Labanda MS, Lauché C, López JC, Mar-
tínez AM, Otero LH, Peyric EH, Ponziani
PF, Ramondino R, Rinaldi J, Rodríguez S,
16 Cepeda et al.
Investigación Clínica 62(Suppl. 2): 2021
Russo JE, Russo ML, Saavedra SL, Seigel-
chifer M, Sosa S, Vilariño C, López Bisca-
yart P, Corley E, Spatz L, Baumeister EG,
Goldbaum FA. Development of a hyperim-
mune equine serum therapy for COVID-19
in Argentina. Medicina (B Aires) 2020;80
(Suppl 3):1-6.
22. Schaefer E, Harms C, Viner M, Barnum S,
Pusterla N. Investigation of an experimen-
tal infection model of equine coronavirus
in adult horses. J Vet Intern Med 2018; 32:
2099-2104.
23. Towbin H, Stachelin T, Gordon J. Elec-
trophoretic transfer of proteins from po-
lyacrylamide gels to nitrocellulose sheets:
procedure and some applications. Proc Natl
Acad Sci USA 1979; 76: 4350–4354.
24. Kelley LA, Mezulis S, Yates CM, Wass MN,
Sternberg MJ. The Phyre2 web portal for
protein modelling, prediction and analysis.
Nat Protoc 2015; 10: 845–858.
25. Wiederstein M, Sippl MJ. ProSA-web: in-
teractive web service for therecognition of
errors in three-dimensional structures of
proteins. Nucleic Acids Res 2007; W407–
W410.
26. Laskowski RA, MacArthur MW, Moss DS,
Thornton J M. PROCHECK-a program to
check the stereochemical quality of protein
structures. J Appl Crystallogr 1993; 26:
283-291.
27. Ponomarenko J, Bui HH, Li W, Fusseder
N, Bourne PE, Sette A, Peters B. ElliPro: a
new structure-based tool for the prediction
of antibody epitopes. BMC Bioinformatics
2008; 9: 514.
28. Behar A, Dennouni-Medjati N, Harek Y,
Dali-Sahi M, Belhadj M, Meziane FZ. Se-
lenium overexposure induces insulin resis-
tance: In silico study. Diabetes Metab Syndr
2020; 14:1651-1657.
29. Ambrosetti F, Olsen TH, Olimpieri PP,
Jiménez-García B, Milanetti E, Marcatilli
P, Bonvin AMJJ. proABC-2: Prediction of
antibody contacts v2 and its application to
information-driven docking. Bioinformatics
2020;36:5107-5108.
30. van Zundert GCP, Rodrigues JPGLM, Tre-
llet M, Schmitz C, Kastritis PL, Karaca E,
Melquiond ASJ, van Dijk M, de Vries SJ,
Bonvin AMJJ. The HADDOCK2.2 Web Ser-
ver: User-Friendly Integrative Modelling of
Biomolecular Complexes. J Mol Biol 2016;
4:720-725,
31. Xue LC, Rodrigues JP, Kastritis PL, Bon-
vin
Alexandre Mjj, Vangone A. PRODI-
GY: a web server for predicting the binding
affinity of protein-protein complexes. Bioin-
formatics 2016; 32:3676-3678.
32. Yuan M, Wu NC, Zhu X, Lee CCD, So
RTY, Lv H, Mok CKP, Wilson, IA. A highly
conserved cryptic epitope in the receptor
binding domains of SARS-CoV-2 and SARS-
CoV. Science 2020; 6491: 630-633.
33. Tang D, Comish P, Kang R. The hallmarks
of COVID-19 disease. 2020; PLoS Pathog
16: e1008536.
34. Pusterla N. Science-in-brief: Equine coro-
navirus - a decade long journey to investi-
gate an emerging enteric virus of adult hor-
ses. Equine Vet J 2020;52: 651-653.
35. Zhao S, Smits C, Schuurman N, Barnum
S, Pusterla N, Kuppeveld FV, Bosch BJ,
Maanen KV, Egberink H. Development and
validation of a S1 protein-based ELISA for
the specific detection of antibodies against
equine coronavirus. Viruses 2019; 11:1109.
36. Kooijman LJ, James K, Mapes SM, The-
elen MJ, Pusterla N. Seroprevalence and
risk factors for infection with equine coro-
navirus in healthy horses in the USA. Vet J
2017; 220:91-94.
37. Otte L, Knaute T, Schneider-Mergener J,
Kramer A. Molecular basis for the binding
polyspecificity of an anti-cholera toxin pep-
tide 3 monoclonal antibody. J Mol Recognit
2006; 19:49-59.
38. Latiano A, Tavano F, Panza A, Palmieri O,
Niro GA, Andriulli N, Latiano T, Corrito-
re G, Gioffreda D, Gentile A, Fontana R,
Guerra M, Biscaglia G, Bossa F, Carella
M, Miscio G, di Mauro L. False positive
results of IgM/IgG antibodies against anti-
gen of the SARS-CoV-2 in sera stored before
the 2020 Endemia in Italy. Int J Infect Dis
2020: S1201-9712: 32591-32601.
39. Mateus J, Grifoni A, Tarke A, Sidney J,
Ramirez SI, Dan JM, Burger ZC, Rawlings
SA, Smith DM, Phillips E, Mallal S, Lam-
mers M, Rubiro P, Quiambao L, Sutherland
A, Yu ED, da Silva Antunes R, Greenbaum
J, Frazier A, Markmann AJ, Premkumar
L, de Silva A, Peters B, Crotty S, Sette A,
Weiskopf D. Selective and cross-reactive
Production of equine sera as immunotherapy against COVID-19 17
Vol. 62(Suppl. 2): 3 - 17, 2021
SARS-CoV-2 T cell epitopes in unexposed
humans. Science 2020;370(6512):89-94.
40. Díez JM, Romero C, Vergara-Alert J, Be-
lló-Perez M, Rodon J, Honrubia JM, Sega-
lés J, Sola I, Enjuanes L, Gajardo R. Cross-
neutralization activity against SARS-CoV-2
is present in currently available intravenous
immunoglobulins. Immunotherapy 2020;
12:1247-1255.
41. Ladner JT, Henson SN, Boyle AS, Engelbre-
ktson AL, Fink ZW, Rahee F, D’ambrozio J,
Schaecher KE, Stone M, Dong W, Dadwal
S, Yu J, Caligiuri MA, Cieplak P, Bjørås
M, Fenstad MH, Nordbø SA, Kainov DE,
Muranaka N, Chee MS, Shiryaev SA, Altin
JA. Epitope-resolved profiling of the SARS-
CoV-2 antibody response identifies cross-
reactivity with endemic human coronaviru-
ses. Cell Rep Med 2021; 2:100189.
42. Yang S, Xia X. Passive immunotherapy for
Middle East Respiratory Syndrome corona-
virus infection with equine immunoglobu-
lin or immunoglobulin fragments in a mou-
se model. Antivir Res 2017; 137: 125–130.