Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Virus Research 297 (2021) 198339

Contents lists available at ScienceDirect

Virus Research
journal homepage: www.elsevier.com/locate/virusres

FMD empty capsids combined with the Immunostant Particle Adjuvant


-ISPA or ISA206 induce protective immunity against foot and mouth
disease virus
J. Bidart a, b, A. Mignaqui c, C. Kornuta a, b, G. Lupi b, d, M. Gammella a, I. Soria a, R. Galarza a,
A. Ferella a, S. Cardillo e, C. Langellotti a, b, V. Quattrocchi a, Y. Durocher f, A. Wigdorovitz a,
I. Marcipar b, d, P. Zamorano a, b, g, *
a
Instituto de Virología e Innovaciones Tecnológicas-IVIT, CICVyA, INTA-CONICET, Hurlingham, Argentina
b
Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
c
Instituto de Investigaciones Forestales y Agropecuarias Bariloche, IFAB, INTA – CONICET, San Carlos de Bariloche, Rio Negro, Argentina
d
Facultad de Bioquímica y Ciencias Biológicas – Universidad Nacional del Litoral, Santa Fe, Argentina
e
Biogenesis Bago SA, Buenos Aires, Argentina
f
Human Health Therapeutics Research Center, National Research Council Canada, Montreal, QC, Canada
g
Universidad del Salvador, Buenos Aires, Argentina

A R T I C L E I N F O A B S T R A C T

Keywords: Foot and Mouth Disease Virus (FMDV) causes economy losses and is controlled by vaccination in many countries.
FMDV1 Vaccine formulations based on empty capsids or Virus-Like Particles (VLPs) have the advantage of avoiding the
Virus-like particles2 biological hazard of using infectious FMDV, albeit are poorly immunogenic. Recently, we have described that
Protection3
ISPA a new Immune Stimulating Complex adjuvant, is useful to improve the response against FMD of vaccines
ISPA4
that use inactivated virus. Now, the adjuvant effects of ISPA and ISA 206 (water/oil/water) on a VLPs-based FMD
Vaccine5
Adjuvant6 vaccine were evaluated.
VLPs (strain A/Argentina/2001) were obtained in mammalian cell cultures and their elicitation of an immune
response against FMDV with and without ISPA or ISA 206 was evaluated in mice as a first approach. Notably,
VLPs-ISPA and VLPs-ISA 206 vaccines induced protection against viral challenge in 100 % of mice, while pro­
tection induced by VLPs alone was of 40 %. Total and neutralizing FMDV antibodies were higher in the VLPs-
ISPA and VLPs-ISA 206 groups compared to the VLPs group. VLPs-ISPA induced significantly higher (p <
0.001) IgG1, IgG2a, IgG2b and IgG3 titers than the VLPs vaccine. Moreover, in comparison with non-adjuvanted
VLPs, VLPs-ISPA and VLPs-ISA 206 elicited an increased virus-specific T response, including higher IFNγ+/CD8
+ lymphocyte production in mice. When these vaccines were tested in calves, antibody titers reached an Ex­
pected Percentage of Protection (EPP) above 90 % in the case of the VLPs-ISPA and VLPs-ISA 206 vaccines,
while, in the VLPs group, EPP reached 25 %.
IFNγ levels secreted by mononuclear cells of VLP-ISPA-vaccinated cattle were significantly higher than in the
VLPs group. Overall, the results demonstrate that VLPs-ISPA or VLPs-ISA 206 are promising formulations for the
development of a novel FMD vaccine.

1. Introduction economic losses (Thompson et al., 2002; OIE, 2018). FMD control re­
quires cost and effort from developing countries, and vaccination is the
Foot-and-Mouth Disease (FMD) is a contagious viral infection of best strategy to prevent it. (Parida, 2009).
cloven-hoofed animals. FMD is endemic in many parts of the world and The etiological agent is Foot-and-Mouth Disease Virus (FMDV), a
is still a major menace to the stockbreeding industries. An outbreak in an member of the Aphthovirus genus within the Picornaviridae family. The
FMD-free country leads to restrictions on livestock trade, causing large viral particle consists of a capsid formed by 60 copies of the VP1-VP4

* Corresponding author at: Instituto de Virología e Innovaciones Tecnológicas-IVIT, CICVyA, INTA-CONICET, Hurlingham, Argentina.
E-mail address: zamorano.patricia@inta.gob.ar (P. Zamorano).

https://doi.org/10.1016/j.virusres.2021.198339
Received 23 December 2020; Received in revised form 3 February 2021; Accepted 6 February 2021
Available online 14 February 2021
0168-1702/© 2021 Published by Elsevier B.V.
J. Bidart et al. Virus Research 297 (2021) 198339

proteins, and a positive RNA genome. VP1-VP3 form the surface of the vaccine doses and/or allow to reach homogenous immune responses
particle and VP4 is internal (Acharya et al., 1989). The capsid proteins (Mohan et al., 2013). It is of great importance to find new adjuvants that
facilitate virus entry and delivery of the genome into the cell cytoplasm. allow reducing the amount of virus or antigens in vaccines and induce
Then, the viral RNA genome is translated and replicated, and new virus protective immune responses. A Th1 response is desirable for FMDV
particles are formed (Jackson et al., 2000; Belsham, 2005; Monaghan clearance in cattle and a Th2 response is preferred for the
et al., 2005). The immune system of virus-infected animals produces antibody-based immunity (Childerstone et al., 1999; Doel, 2003; Patch
neutralizing antibodies (Abs) against the surface-exposed capsid pro­ et al., 2013; Habiela et al., 2014; Quattrocchi et al., 2014; Stenfeldt
teins, which is a key requirement for protection (Doel, 2003). et al., 2017). Other desirable characteristics include low cost and sta­
Seven FMDV serotypes (A, ASIA1, C, O and Southern African Terri­ bility. It has been reported that Immune Stimulating Complexes
tories 1–3) and several strains within each serotype have been described. (ISCOMs) are capable of developing a Th1/Th2 balanced immune
Mattion et al. (2004) demonstrated that in Argentina, vaccination with a response, in addition to increasing cytotoxic responses (Singh, 2006;
FMDV strain A24/Cruzeiro, yielded acceptable levels of protection only Maraskovsky et al., 2009; Sun et al., 2009; Bertona et al., 2017). ISCOMs
after re-vaccination because two serotype A lineages (A/Argentina/00 are spherical particles of approximately 40 nm in diameter, composed of
and A/Argentina/01 prototypes) were identified during 2000–2002 phospholipids, cholesterol and saponin, which can retain the antigen
epidemic. The updating of the antigenic composition of the vaccines through hydrophobic interactions (Morein et al., 1984; Singh, 2006).
used in the emergency was a key issue, since the outbreaks stopped They have been applied to the development of several registered vac­
shortly after the implementation of the vaccination programs. There­ cines for veterinary applications (Sun et al., 2009). Recently, the
fore, the new field strains A/Argentina/00 and A/Argentina/01 were Immunostimulating Particle Adjuvant or ISPA, an empty cage-like par­
incorporated into the vaccine, leading to an effective control of the ticle formulation similar to ISCOMATRIX™, was described. It contains
disease. dipalmitoyl-phosphatidylcholine (DPPC), cholesterol (CHO), stearyl­
The commercial vaccine used is manufactured with binary ethylene- amine (STEA), alpha-tocopherol (TOCO) and Quil A saponin (Bertona
imine (BEI)-inactivated virus. (Doel, 2003; Grubman and Baxt, 2004; et al., 2017; Prochetto et al., 2017; Bidart et al., 2020). This adjuvant
OIE - World Organisation for Animal Health, 2012). Disadvantages of was shown to surpass conventional ones by improving humoral and
the use of this type of vaccine include: the need for high biosafety pro­ cellular CD4+ / CD8+ responses (Bertona et al., 2017). Fontana et al.
duction facilities (BSL-4 OIE o higher), strict protocols of production, (2020) demonstrated that Rabies virus-like particles plus ISPA formu­
constant investments in manufacturing plant up-grades and personnel lation induces neutralizing antibodies in cat, dogs and bovines (Fontana
training, and a strict purification process that guarantees differentiation et al., 2020). Recently, we reported that an inactivated Foot-and-Mouth
of infected from vaccinated animals. Moreover, some serotypes and Virus serotype A vaccine adjuvanted with ISPA was capable of inducing
strains do not grow properly in cell culture (Grubman, 2005; Rodriguez protection against challenge in a murine model and of improving the
and Grubman, 2009). Thus, development of new vaccines is desirable. specific immune responses against FMDV in cattle (Bidart et al., 2020).
Recombinant VLPs may represent a good alternative to the conven­ On the other hand, ISA 206 (Seppis, France) is a mineral oil that contains
tional FMD vaccine since they are non-infectious and can be produced octadecenoic acid esters and anhydromanitol, which formulation is
without the need of high containment facilities, and may also be water-in-oil-in-water (w/o/w) (Barnett et al., 1996). These emulsions
modified to enhance their stability (Ellard et al., 1999; Mateo et al., have a low-viscosity, which facilitates inoculation and gives the ability
2008; Porta et al., 2013; Rincón et al., 2014; Caridi et al., 2015; Kotecha to increase the immune response in the short and long term in cattle,
et al., 2015). The use of suspension-growing mammalian cells, economic pigs and sheep (Barnett et al., 1996, 2002; Patil et al., 2002; Cox et al.,
and efficient transfection reagents and optimized expression vectors has 2003; Rigden et al., 2003; Cox and Barnett, 2009). ISA206 had achieved
allowed transient gene expression (TGE) to become a simple, scalable increased opsonization and phagocytosis in the early protective immune
and powerful technology to generate large amounts of recombinant responses against FMDV in the murine model (Quattrocchi et al., 2011).
VLPs within a short time period (Pham et al., 2006; Baldi et al., 2007; This adjuvant has been used to formulate emergency vaccines against
Mignaqui et al., 2013). FMDV and was tested in cattle and swine showing an increase in pro­
Although some reports have demonstrated the efficacy of FMD vac­ tection (Barnett et al., 2002). ISA 206 is used for formulating FMD
cines based on VLPs (Li et al., 2012; Porta et al., 2013; Xiao et al., 2016), vaccines in many Asian and South American countries.
a lower performance of a VLP based vaccine can be expected when In this report, we demonstrate for first time the effect of ISPA as
compared to a traditional vaccine due to the lack of viral RNA, a adjuvant for a new subunit vaccine using VLPs from FMDV both in a
well-known activator of the innate immune response (Medina et al., murine model and in cattle and alternatively ISA 206 may be used to
2018). Moreover, VLPs vaccines may still be expected to suffer from enhance the immunity induced by VLPs.
some of the same limitations, (e.g. in terms of duration of immunity)
(Gullberg et al., 2016). The use of novel adjuvants can be a promising 2. Materials and methods
tool to improve the performance of these vaccines.
In the present work, FMDV strain A/Argentina/2001 VLPs were 2.1. Animals
produced by transient gene expression in serum-free suspension-
growing mammalian cells, using polyethylenimine (PEI) as transfection All experiments involving the use of animals were carried out ac­
reagent (Mignaqui et al., 2013). Strain A/Argentina/2001 (A2001), cording to National Agricultural Technology Institute (INTA) Ethics
isolated during an outbreak of FMD in Argentina in 2000 (Mattion et al., Manual “Guide for the use and care of experimental animals”, under
2004), was used in the present study as proof of concept. protocol Number: 26/2016.
Our laboratory has ample experience in a murine model that proved Male BALB/c mice, 8–12 weeks old from the animal facilities of the
useful to evaluate the potency of FMDV vaccines (Quattrocchi et al., School of Veterinary Sciences, University of Buenos Aires, Argentina,
2005, 2011, 2013; Batista et al., 2010; Zamorano et al., 2010; Langellotti were used.
et al., 2012, 2015; Romanutti et al., 2013; Bidart et al., 2020; Gnazzo FMDV-seronegative calves, of approximately 8–10 months old, were
et al., 2020). In this model, there is a correlation between the humoral used in the experiment.
and protective immune responses against infective FMDV that take place
in mice and cattle (Gnazzo et al., 2020). 2.2. Virus-like particles (VLPs)
Some vaccine adjuvants improve the immune response elicited
against antigens, direct the immune response to a particular profile, Recombinant VLPs were obtained as previously reported (Mignaqui
increase the number of responding individuals, reduce the amount of et al., 2013). Briefly, suspension-growing 293− 6E cells were grown in

2
J. Bidart et al. Virus Research 297 (2021) 198339

serum-free F17 medium (Gibco) at 37 ◦ C with 5% CO2 and agitation at 2.5. Adjuvants
120 rpm. Cells were transiently transfected with pTT5-P12A3C plasmid
encoding for FMDV strain A/Argentina/2001 VLPs using poly­ ISPA adjuvant is composed of alpha-tocopherol (TOCOP), phospati­
ethylenimine (LPEI-MAX) (Polysciences, Warrington, PA, USA). Cells dylcholine (DPPC), stearylamine (STEA), cholesterol (CHOL) and QuilA
were harvested 48 h post transfection, centrifuged at 4000 g and the saponin. ISPA particles have a cage-like structure of 73.0 ± 1.5 nm in
pellet was resuspended in Tris− HCl, pH 8, and subjected to three size as assessed by dynamic light scattering. First, liposomes were pre­
freeze-thaw cycles at -80 ◦ C / 25 ◦ C. Finally, the lysate was clarified by pared with final proportions of TOCOP: 0.00074 % (0.017 mM), DPPC:
centrifugation and VLPs were quantified by ELISA and characterized by 0.320 % (4.35 mM), STEA: 0.0216 % (0.8 mM) and CHOL: 0.143 %
Western Blot and sucrose gradient, as previously reported (Mignaqui (3.70 mM). Then, the suspension was extruded through a 50 nm pore
et al., 2013). Briefly for ELISA, a polyclonal anti-FMDV serum raised in membrane and a QuilA saponin solution in acetate buffer was added to
rabbit was used for coating microtiter plates (Maxisorp). After washing liposomes (6.5 mg/300 μL per ml of liposomes) and extruded through a
steps, plates were blocked for 30 min at 37 ◦ C with 5% normal equine 50 nm membrane pore (Bertona et al., 2017; Prochetto et al., 2017).
serum in PBS/0.1 % Tween-20. VLPs samples were added to the wells The commercial oil adjuvant (CA) used was ISA206 (Seppic, Paris,
and incubated at 37 ◦ C for 1 h. A standard curve was generated using France). It is a mineral oils containing esters of octadecenoic acid and
serial dilutions of a quantified aliquot of inactivated A2001 FMDV. anhydromannitol for example, readily form double or mixed (water-in-
Plates were then incubated for 1 h with a polyclonal guinea pig oil-in-water) emulsions which are both stable and of low viscosity
anti-FMDV serum, followed by horseradish peroxidase-conjugated goat (Barnett et al., 1996). The formulations were prepared following the
anti-guinea pig IgG (KPL). Then, tetramethylbenzidine was used as manufacturer’s indications in a proportion adjuvant/antigen of 60:40.
substrate and the absorbance at 450 nm was recorded in a microplate
reader (Thermo Scientifics MultiskanFC). For Western Blotting analysis,
cell lysates were separated by SDS-PAGE electrophoresis and then pro­ 2.6. Vaccine formulations and vaccination experiments
teins were transferred onto a membrane. After blocking, membrane was
incubated with anti-FMDV guinea pig serum. After washes, membranes The vaccines to be applied in mice at days: 0 and 21, were formulated
were incubated with horseradish peroxidase-conjugated anti guinea pig with: (i) 0.5 μg VLPs in PBS (VLPs); (ii) 0.5 μg VLPs in PBS mixed with 6
goat serum (KPL) and the reaction was visualized with a chem­ μl ISPA (VLPs-ISPA) or (iii) 0.5 μg VLPs in PBS mixed with ISA 206
iluminescence method. For structural analysis, a 45− 15% sucrose (VLPs-ISA 206), in a final volume of 0.1 mL/dose. BALB/c mice were s.c.
gradient was prepared. Samples were added on top of the gradient and immunized at day 0 and 21 with: (i) VLPs (n = 5); (ii) VLPs-ISPA (n = 5);
tubes were centrifuged in a BeckmanOptima-LP X-100 Ultracentrifuge (iii) VLPs-ISA 206 (n = 5); (iv) Commercial Vaccine (C+) (n = 5); (v) 6 μl
using a SW 55 Ti rotor for 2 h at 45,000 rpm at 4◦C, acceleration: 9, ISPA (n = 2); (vi) (ISA 206) (n = 2) or (vii) PBS (n = 2). Mice were
deceleration: 9. Once the centrifugation was completed, aliquots were challenged at 36 days post first immunization (dpfi) as described in 2.4.
collected and tested for ELISA-specific FMDV protein. The commercial vaccine consisted in a water-in-oil single emulsion
containing O1/Campos/Brazil/58, A24/Cruzeiro/Brazil/55, C3/
2.3. Virus Indaial/Brazil/71 and A/Argentina/2001 iFMDV and was provided by
Biogénesis Bagó S.A.
BEI inactivated A/Argentina/2001(A2001) FMDV strain (iFMDV) The vaccines used in cattle were formulated with (i) 25 μg VLPs in
was provided by Biogenesis Bagó (Buenos Aires, Argentina). iFMDV was PBS (VLPs); (ii) 25 μg VLPs with 1 mL ISPA (VLPs-ISPA); (iii) 25 μg VLPs
purified by sucrose gradient and was used as a positive control for in PBS mixed with ISA 206 (VLPs-ISA 206); or (iv) 25 μg iFMDV in PBS
protein characterization assays, to formulate experimental vaccines, in mixed with ISA 206 (iFMDV-ISA 206), in a final volume of 2 mL/dose.
ELISA and in cellular assays. Infectious virus, obtained from vesicles of We use an antigen mass equivalent to the total amount of iFMDV used in
experimentally infected cattle (provided by the Argentine National previous cattle trials (Bidart et al., 2020). Cattle were s.c. vaccinated
Service of Animal Health, SENASA) with one or two passages in Baby with: (i) VLPs (n = 4); (ii) VLPs-ISPA (n = 4); (iii) VLPs-ISA 206 (n = 4)
Hamster Kidney-21 (BHK-21) cells, was used for viral challenge. All or (iv) iFMDV-ISA 206 (n = 2).
experiments involving infectious virus were performed in the INTA BSL-
4 OIE facilities.
2.7. Measurement of murine FMDV-specific Abs by sandwich ELISA
2.4. Selection of VLPs dose for vaccine formulation in mice
Total Ab titers against FMDV were assessed by ELISA as described
To select the VLPs vaccine dose, dilutions of VLPs in PBS containing: previously (Batista et al., 2010; Zamorano et al., 2010; Quattrocchi
8, 4, 2, 1, 0.5, 0.3, 0.15 or 0 μg in a final volume of 0.1 mL were pre­ et al., 2011). Briefly, Greiner Microlon® plates were coated ON at 4 ◦ C
pared. Groups of mice (n = 5) were subcutaneously (sc) inoculated at with anti-FMDV rabbit serum in carbonate–bicarbonate buffer, pH 9.6.
0 and 21 dpv with these formulations. Mice were challenged with an ip After three washing steps, plates were blocked for 30 min at 37 ◦ C with
injection of 102.5 TCID50/mL infectious FMDV, A2001 strain, at 36 days polyvinylpyrrolidone blocking solution (0.5 M NaCl/ 0.01 M phosphate
post first immunization (dpfi). Twenty-four h later, viremia was evalu­ buffer/ 0.05 % Tween-20/ 1 mM EDTA/ 1% polyvinylpyrrolidone 30–40
ated as described in Bidart et al. (2020). Briefly, heparinized blood K, pH 7.2). An optimal dilution of inactivated A2001 FMDV in blocking
withdrawn at 24 h post infection was spread onto BHK-21 cell mono­ solution was added, followed by incubation at 37◦C for 30 min. Then,
layers grown in 48-well plates and incubated at 37 ◦ C in a 5% CO2 at­ serially diluted mouse sera (1:4) in blocking solution were added. After 1
mosphere. Then, cell monolayers were washed twice with sterile PBS. h 20 min incubation at room temperature, plates were washed and an
Fresh D-MEM supplemented with 2% fetal calf serum (FCS) was added optimal dilution of horse radish peroxidase (HRP)-conjugated
and the cells were incubated for 48 h at 37 ◦ C in 5% CO2. Mice were anti-mouse IgG (H + L) (KPL®). Plates were incubated for 1 h at room
considered infected if the cell monolayer presented cytopathic effects temperature and then washed. Ortho-phenylene-diamine (1,2-benze­
after a blind passage, as established in previous studies (Zamorano et al., nediamine) dihydrochloride (SIGMA®) (OPD)/H2O2 was used as chro­
2010; Quattrocchi et al., 2011, 2014; Gnazzo et al., 2020). Percentages mogenic substrate. Reactions were stopped using 1.25 M H2SO4 and
of protection induced by VLPs were calculated as: 100 x [pro­ A492 was measured in a microplate reader. Positive and negative control
tected/challenged mice]. A dose of 0.5 μg VLPs induced 40 % of pro­ sera were included in every plate. The cut-off was established as the
tection and was thus selected for subsequent experiments, since it allows mean of the values of negative sera (n = 10) plus two standard
a good margin to observe the adjuvant effects. deviations.

3
J. Bidart et al. Virus Research 297 (2021) 198339

2.8. Measurement of bovine FMDV-specific Abs by liquid phase ELISA mM 2-mercaptoethanol, and were subjected to: (i) no stimulation
(mock), (ii) 2.5 μg/mL iFMDV or (iii) 5 μg/mL Concanavalin A (Sigma
An lpELISA test was used according to Hamblin et al. (1986), with Aldrich®, St. Louis, MO) as positive control. Cells were incubated at 37
modifications (Hamblin et al., 1986; OIE - World Organisation for Ani­ ◦
C in 5% CO2 atmosphere for 4 days and then fixed with 0.2 % para­
mal Health, 2012). Briefly, Greiner Microlon® plates were coated formaldehyde. Cell proliferation was analyzed by flow cytometry using
overnight at 4 ◦ C with rabbit anti-FMDV serum diluted to a previously FACSCalibur® (Becton Dickinson, San Jose, CA) and Flowing Software
established optimal concentration in carbonate-bicarbonate buffer, pH (Turku Centre for Biotechnology, Finland). Results were expressed as
9.6. After washing with 0.05 % Tween-20/phosphate buffered saline delta proliferation and were calculated as the difference between the
(PBST), plates were blocked with PBST/1% ovalbumin (blocking buffer) percentage of proliferating cells stimulated with inactivated FMD virus
for 30 min at 37 ◦ C. Bovine sera were serially diluted (1:10) in blocking and the percentage of proliferating cells without stimuli.
buffer in separate tubes and a fixed amount of inactivated A2001 FMDV
was added. After 1 h incubation at 37 ◦ C with shaking, the 2.12. Surface and intracytoplasmic staining for the detection of IFNγ-
virus-antibody mixtures were transferred to the blocked plates, and producing murine splenocytes
incubated for 1 h at 37 ◦ C. An optimal dilution of guinea pig anti-FMDV
serum in PBS/2% normal bovine serum/2% normal rabbit serum was Murine splenocytes were incubated in complete RPMI 1640 medium
added for detection, followed by 1 h incubation at 37 ◦ C. Plates were supplemented with 10 % FCS and 50 μM 2-mercaptoethanol and were
washed and peroxidase-conjugated anti-guinea pig IgG (Jackson subjected to: (i) mock, (ii) 2.5 μg/mL iFMDV or (iii) 5 μg/mL Conca­
ImmunoResearch®) serum diluted in the same buffer was added, fol­ navalin A (Sigma Aldrich®, St. Louis, MO) as positive control. Cells were
lowed by 1 h incubation at 37 ◦ C. OPD/H2O2 was used as chromogen incubated for 18 h in the presence of brefeldin A (BD GolgiPlug™),
substrate as above and A492 was measured in a microplate reader. Strong according to the manufacturer’s recommendations. After washing, cells
positive, weak positive and negative bovine reference sera were were fixed in 0.5 % paraformaldehyde and permeated with saponin (0.1
included in each test for validation. Antibody titers were expressed as % in PBS). Permeated cells were incubated for 20 min at RT with allo­
the reciprocal logarithm of the highest dilution of serum that causes an phycocyanin (APC) anti-mouse INF-γ (clone XMG1.2, BD Pharmingen®)
inhibition of color development higher than 50 % in the average values or isotype-matched control Abs. After 20 min, cells were washed twice
of the control samples. and stained for 30 min at 4 ◦ C with fluorescein isothiocyanate (FITC)
anti-mouse CD4 (clone GK1.5, BDBioscience®); or phycoerythrin (PE)
2.9. Measurement of IgG isotypes against FMDV anti-mouse CD8 (clone 53− 6.7, eBioscience®). Cells were then washed
and fixed with 0.2 % paraformaldehyde. Flow cytometry was performed
Greiner Microlon® plates were coated ON at 4 ◦ C with anti-FMDV as in 2.11.
rabbit serum in carbonate–bicarbonate buffer, pH 9.6. After three
washing steps, plates were blocked for 30 min at 37 ◦ C with poly­ 2.13. IFNγ detection
vinylpyrrolidone blocking solution in the case of mouse sera (0.5 M
NaCl/ 0.01 M phosphate buffer/ 0.05 % Tween-20/ 1 mM EDTA/ 1% Peripheral blood mononuclear cells PBMC were obtained from cattle
polyvinylpyrrolidone 30–40 K, pH 7.2) or with PBS / 10 % fetal calf as described before (Romera et al., 2014). Then, cells were cultured with
serum (FCS) in the case of bovine sera. An optimal dilution of inacti­ 5 μg/mL iFMDV for 5 days. Supernatants were analyzed using ELISA as
vated A2001 FMDV in blocking solution was added, followed by incu­ described previously (Quattrocchi et al., 2014). Briefly, plates were
bation at 37◦C for 30 min. Then, serially diluted mouse sera (1:4) or coated with a monoclonal Ab (mAb) against IFN-γ (kindly donated by
bovine sera (1:5) in blocking solution were added. After 1 h 20 min Dr. L. Babiuk). Samples and recombinant IFNγ standard (Serotec, UK)
incubation at room temperature, plates were washed and an optimal were added, and IFN-γ was detected using rabbit polyclonal anti-IFNγ
dilution of horse radish peroxidase (HRP)-conjugated anti-mouse iso­ Abs. After incubation, biotinylated goat anti-rabbit IgG antibody was
types (Southern Biotech®), or anti-bovine IgG1 or IgG2 (KPL®) were added, followed by HRP-conjugated streptavidin (KPL, USA). Plates
added. Plates were incubated for 1 h at room temperature and then were washed, incubated with (OPD)-H2O2 and A492 was recorded. IFNγ
washed. Ortho-phenylene-diamine (1,2-benzenediamine) dihydro­ concentration was calculated from interpolation of data in a standard
chloride (SIGMA®) (OPD)/H2O2 was used as chromogenic substrate. curve.
Reactions were stopped using 1.25 M H2SO4 and A492 was measured in a
microplate reader. Positive and negative control sera were included in 2.14. Statistical analysis
every plate. The cut-off was established as the mean of the values of
negative sera (n = 10) plus two standard deviations. GraphPad InStat® program (GraphPad, California, USA) was used.
Differences between groups were analyzed by applying the non-
2.10. Neutralizing antibody titers parametric Kruskal–Wallis test, followed by Mann–Whitney U test for
comparisons between two groups. P value <0.05 was considered as an
Serum samples were examined for anti-FMDV neutralizing Abs as indicator of significant differences.
described before (Quattrocchi et al., 2014). Briefly, serial dilutions of
complement-inactivated sera were incubated for 1 h at 37 ◦ C with 100 3. Results
TCID50 of infective FMDV. Then virus-serum mixtures were seeded on
BHK-21 monolayers. After 40 min at 37 ◦ C, fresh DMEM /2% FCS was 3.1. VLPs production
added to the monolayers, which were incubated at 37 ◦ C, under 5% CO2.
Cytopathic effects were observed after 48 h. Titers are expressed as log10 Suspension growing mammalian cells were transiently transfected
of the reciprocal of the serum dilution which neutralizes 50 % of 100 with pTT5-based plasmid encoding P12A polyprotein and 3C protease
TCID50 infective FMDV. from FMDV strain A/Argentina/2001 using polyethylenimine as trans­
fection reagent (Mignaqui et al., 2013). After transfection, cells were
2.11. Lymphoproliferation assay harvest 48 h post transfection and cell lysates were analyzed by western
blot to confirm proper expression and processing of FMDV P12A poly­
Murine splenocytes were labeled with 3 μM CFSE in PBS for 30 min protein by 3C protease into VP0, VP1 and VP3 structural proteins
at 37 ◦ C. Labeled cells were added to 96-well plates (5 × 105 cell/well) (Fig. 1A). During viral infection, one copy of VP0, VP1 and VP3 struc­
in complete RPMI 1640 medium supplemented with 10 % FCS and 50 tural proteins assemble to form the protomer (5S), then five protomers

4
J. Bidart et al. Virus Research 297 (2021) 198339

Fig. 1. (A) Western Blotting of cell lysates 48 h post transfection with pTT5-P12A3C, mock transfected cells and inactivated FMDV (iFMDV). Primary antibody: anti-
FMDV polyclonal serum made in guinea pig (1/500) and secondary antibody: anti-guinea pig peroxidase (1/10,000). (B) 45-15 % sucrose gradient analysis of cell
lysate after 48 h of transfection with pTT5-P12A3C and iFMDV. Aliquots from sucrose gradient were analyzed by ELISA and optical density was read at 450 nm.

form the pentamer (12S) and twelve pentamers assemble into the empty 3.3. VLPs-ISPA and VLPs-ISA 206 vaccines confer total protection
capsid (75S), finally upon encapsidation of genomic RNA into the empty against FMDV in a murine model
capsids, occurs cleavage of VP0 into VP2 and VP4 and the virion (146S)
is form. All these subviral particles produced during viral morphogenesis The protective efficacy of the inclusion of ISPA or ISA 206 as adju­
can be characterized by different sedimentation coefficients in sucrose vants in a VLPs vaccine (VLPs-ISPA and VLPs-ISA206) was tested in
gradients. To demonstrate that the VLPs were produced in mice. We use VLPs 0.5 μg/dose for vaccine formulation.
pTT5-P12A3C transfected cells, lysates were analyzed by sucrose Groups of mice were vaccinated with VLPs, VLPs-ISPA, VLPs-ISA
gradient and most of the structural proteins were assemble into VLPs 206, Commercial vaccine, ISA 206 alone, ISPA alone or PBS (negative
(Fig. 1B). Cell lysates containing VLPs were quantified by ELISA for controls) at 0 and 21 dpv. Then, mice were challenged with 102.5
vaccine formulation. Empty capsids yields achieved were between 3− 7
mg per liter of 293− 6E cell culture.

3.2. Selection of the VLPs dose for vaccine formulation with adjuvants

To analyze the modulatory effect of ISPA on the immune response to


VLPs, a dose of VLPs capable of inducing 40 % protection was first
selected. To this end, BALB/c mice were s.c. vaccinated with different
amounts of VLPs in PBS at 0 and 21 dpv and challenged with 102.5
TCID50/mL infectious FMDV, strain A/Argentina/2001, after 36 days
post first immunization. A dose-dependent decrease in antibody titers
with decreasing amounts of VLPs (Fig. 2A), as well as a concomitant
protective effect were observed (Fig. 2B). Forty percent of mice vacci­
nated with 0.5 μg VLPs were protected upon viral challenge, as
measured by the presence or absence of viremia 24 h after infection. All
animals immunized only with PBS, as negative controls, were not pro­
tected. Thus, 0.5 μg VLPs dose was chosen for vaccine formulations to
detect the action of ISPA or ISA 206 on the immune response against Fig. 3. Protection upon viral challenge elicited by experimental vaccines.
FMDV. Groups of mice (n = 5) were vaccinated with VLPs (0.5 μg), VLPs (0.5 μg)-ISPA,
VLPs (0.5 μg)-ISA 206 or a Control (+) vaccine, and groups of mice (n = 2) were
vaccinated with ISPA, CA or PBS alone at 0 dpv and 21 dpv, and challenged
with infective FMDV at 36 dpv. Protection was calculated as in Fig. 1.

Fig. 2. Selection of VLPs dose for vaccination of


BALB/c mice. Groups of mice (n = 5) were
vaccinated with 8, 4, 2, 1, 0.5, 0.3, 0.15 or 0 μg
VLPs in PBS at 0 and 21 days and challenged
with infective virus at 36 dpv. (A) Abs against
FMDV elicited by vaccination with different
amounts of VLPs measured by sandwich ELISA
at 36 dpv. (B) Percentages of protected animals
upon viral challenge. Animals were considered
protected if viremia was absent at 24 h post
challenge. Protection percentages were calcu­
lated as: 100 x [number of vaccinated animals
without viremia / number of vaccinated
animals].

5
J. Bidart et al. Virus Research 297 (2021) 198339

TCID50/mL infectious FMDV, strain A/Argentina/2001, after 36 dpfi higher than in the VLPs group.
(Fig. 3). Notably, while protection with VLPs alone was achieved in 40 % When the virus neutralization test (VNT) was applied (Table 1),
of mice, inclusion of ISPA in the formulation increased protection levels neutralizing antibody titers at 36 dpfi were significantly higher in
to 100 % as well as with the VLPs-ISA 206 and commercial vaccines groups vaccinated with VLPs-ISPA (1.2 ± 0.06), VLPs-ISA 206 (1.41 ±
(positive control). The use of ISPA or ISA206 in VLPs-based vaccines 0.02) and Commercial vaccine (2.20 ± 0.08), than in the VLPs (0.90)
increases protection in all the vaccinated animals against viral group (p < 0.05, p < 0.05 and p < 0.001 respectively). VNT in the VLPs-
challenge. ISA 206 and Commercial vaccine groups were similar. No neutralizing
Mice inoculated with ISPA, ISA 206 or PBS were not protected, Abs were detected in the ISPA, ISA 206 and PBS groups.
indicating that the viral challenge was conducted properly and indicates The results presented in Fig. 4 and Table 1 show a concordance be­
that protection depends on the inclusion of antigen in the vaccine and tween anti-FMDV Ab response and the protection against the challenge
was not due to innate immune mechanisms elicited by any of the following vaccination in mice.
adjuvants.
3.5. Immunization with VLPs-ISPA and VLPs-ISA 206 induce a specific
3.4. Murine specific anti-FMDV Abs and neutralizing Abs are increased cellular immune response against FMDV in mice
when ISPA or ISA 206 are used as adjuvants
After results about the humoral response induced by formulations,
Generally, vaccines that induce greater protection in mice show high spleen cells were labeled and analyzed by flow cytometry, in order to
Ab levels, while those that do not induce good protection show lower investigate the cellular response. Murine splenocytes were obtained 36
levels of Abs (Borca et al., 1986; Fernandez et al., 1986; López et al., days after first immunization. Then, the cells were incubated with
1990; Dus Santos et al., 2000; Gnazzo et al., 2020). In order to evaluate inactivated A2001 FMDV strain in vivo, and labeled with CFSE or anti-
the performance in mice of the formulations used in this report, humoral CD4, anti-CD8 and anti-IFNγ.
response elicit was measured by sandwich ELISA, isotype profiles and At 36 dpv, when splenocytes were in vitro exposed to inactivated
VNT. virus, FMDV-specific T-cell stimulation levels were significantly higher
Antibody responses elicited by VLPs, VLPs-ISPA, VLPs-ISA 206, in cells derived from mice immunized with VLPs-ISPA and VLPs-ISA 206
Commercial vaccine (C(+)), ISA 206, ISPA and PBS were evaluated at (p < 0.05) or with the Commercial vaccine (p < 0.001) than in those
15, 21 and 36 dpfi (Fig. 4A). At 15 dpfi, total specific anti-FMDV Ab derived from VLPs, ISPA, ISA 206 or PBS-inoculated mice (Fig. 5A).
titers measured by sandwich ELISA were significantly higher in the On the other hand, lymphocytes were stained with anti-murine CD4,
VLPs-ISPA (1.6 ± 0.1) and C (+) (2.9 ± 0.2) groups as compared to the anti-murine CD8, and anti-murine INFγ and then studied by flow
VLPs-ISA206 (1.03 ± 0.05) and VLPs group (1.00 ± 0.01) (p < 0.01 and cytometry. As show in Fig. 5B, a tendency of an increased production of
p < 0.001, respectively). At 21 dpfi, Ab titer in VLPs-ISPA (3.4 ± 0.4), IFNγ was observed in CD4+ cells of the VLPs–ISPA and VLPs-ISA206
VLPs-ISA206 (2.7 ± 0.4) and C (+) (4.6 ± 0.3) groups were significantly group as compared to the VLPs group, although the difference was not
higher than VLPs group (1.4 ± 0.3) (p < 0.01, p < 0.01 and p < 0.001, statistically significant (p = 0.68).
respectively). Finally, at 36 dpfi, Ab titers were significantly higher in
the VLPs-ISPA (5.2 ± 0.2), VLPs-ISA206 (4.9 ± 0.2) and C (+) (5.8 ± Table 1
0.3) groups as compared to the VLPs group (2.7 ± 0.4) (p < 0.001) Virus neutralizing antibody titers (VNT) at 36 days after
Analysis of isotype profiles at 36 dpfi showed that the VLPs-ISPA inoculating mice with experimental vaccines. Titers are
group achieved higher IgG1 titers (5.2 ± 0.1) than VLPs-ISA 206 (4.5 expressed as log10 of the reciprocal of the serum dilu­
± 0.1), Commercial vaccine (4.61 ± 0.03) and VLPs (2.2 ± 0.01) groups tion which neutralizes 50 % of 100 TCID50 infective
(p<0.001). Moreover, IgG1 titers in the VLPs-ISA 206 and Commercial FMDV, using the fixed virus – variable serum method. *
Significant differences against the VLPs group. *(p <
vaccine groups were similar (Fig. 4B). IgG2a titers were also higher in
0.05), *** (p < 0.001).
VLPs-ISPA (3.4 ± 0.2), VLPs-ISA 206 (3.4 ± 0.2) and Commercial vac­
cine (4.2 ± 0.4) groups than in the VLPs (1.5 ± 0.3) group (p<0.001). Vaccine VNT

Interestingly, IgG2b titers were higher for VLPs-ISPA (4.3 ± 0.6) than VLPs 0.90
VLPs-ISA 206 (3.6 ± 0.6), Commercial vaccine (3.71 ± 0.07) and VLPs VLPs-ISPA 1.20 ± 0.06*
VLPs-ISA206 1.41 ± 0.02*
(1.8 ± 0.2) groups (p<0.001). Finally, IgG3 titers in the VLPs-ISPA (3.3
C(+) 2.20 ± 0.08**
± 0.2) group were significantly higher than in the VLPs (1.1 ± 0.1) and ISPA <0.90
VLPs-ISA 206 (2.7 ± 0.2) groups (p < 0.001 and p < 0.01, respectively), ISA206 <0.90
and similar to the titers elicited by the Commercial vaccine (3.5 ± 0.2). PBS <0.90
Moreover, the IgG3 titer in the VLPs-ISA 206 group was significantly

Fig. 4. Abs against FMDV elicited by experimental vaccines in mice. FMDV specific Ab titers were measured by (A) sandwich ELISA at 15, 21 and 36 dpv. Each point
represents the mean (n = 5) Ab titer ± SD in each group. * Significant differences against VLPs group. ** (p < 0.01); ***(p < 0.001). (B) Isotype profile of vaccinated
animals at 36 dpv. Data are expressed as the mean Ab titer ± SD. *** p < 0.001, **(p < 0.01).

6
J. Bidart et al. Virus Research 297 (2021) 198339

Fig. 5. Cellular immune response in mice


splenocytes at 36 days post first immunization.
Animals were vaccinated with VLPs, VLPs-ISPA,
VLPs-ISA 206, control (+) vaccine, ISPA, com­
mercial adjuvant alone or PBS. (A) Lymphocyte
proliferative response after stimulation with
iFMDV measured by decrease in CFSE fluores­
cence levels. Results are expressed as the dif­
ference (Δ%) between the percentage of
proliferating splenocytes stimulated with inac­
tivated virus and the percentage of proliferating
splenocytes without stimuli. *** p < 0.001; *p
< 0.05. Cellular immune response in mice
splenocytes at 36 days post first immunization.
Animals were vaccinated with VLPs, VLPs-ISPA,
VLPs-ISA 206, control (+) vaccine, ISPA, com­
mercial adjuvant alone or PBS. Lymphocyte
IFNγ-producing after stimulation with iFMDV in
vitro for 18 h in presence of brefeldin A. (B)
Percentages of CD4+/IFNγ + T cells and (C)
percentages of CD8+/IFNγ + T cells. ** p <
0.01; *p < 0.05.

At 36 days post first immunization, the VLPs-ISPA and VLPs-ISA FMDV-serologically negative calves were inoculated with VLPs (n =
groups presented increased percentages of CD8+/IFNγ+ secreting 4), VLPs-ISPA (n = 4), VLPs-ISA 206 (n = 4) or iFMDV-ISA 206 (n = 2) at
cells as compared to the VLPs group (p<0.05 and p<0.01 respectively). day 0.
No significant differences were detected between the VLPs alone and the First, we studied the humoral response elicits by the formulations.
commercial vaccine groups (Fig. 5C). There is a general consensus about the correlation that exists between
An increased lymphoproliferative response was evident in mice the presence of anti-viral antibodies and protection in cattle vaccinated
immunized with VLPs-ISPA and VLPs-ISA206. These results suggest that against FMDV (van Bekkum, 1969; Gomes and Astudillo, 1975; Pay
the adjuvants improve the adaptive immune response against FMDV. et al., 1983; Hamblin et al., 1987; Doel, 2003). High Ab titres in serum
Moreover, we could see FMDV-specific T-cell stimulation after vacci­ are considered a guarantee of adequate immunity and protection against
nation with empty capsids. the virus (van Bekkum, 1969; Maradei et al., 2008).
When Ab titers were measured by Lp ELISA, calves vaccinated with
3.6. VLPs plus ISPA or ISA 206 induce increased levels of anti-FMDV Abs VLPs-ISPA and VLPs-ISA 206 displayed a significant increment in the
in cattle elicited anti-FMDV humoral response as compared to calves vaccinated
with VLPs alone (p<0.01) at 15, 30 and 45 dpv (Figs. 6A). At 15 dpv,
After promising results obtained in the murine model, the immune anti-FMDV Ab titers were significantly higher (p < 0.01) in the VLPs-
efficacy of the VLPs-ISPA and VLPs-ISA 206 vaccines was studied in ISPA (2.73 ± 0.31), VLPs-ISA206 (2.78 ± 0.27) and iFMDV-ISA206
cattle, a natural host of the virus. (3.10 ± 0.34) groups than the VLPs group (1.4 ± 0.2). At 21 dpv, Ab

Fig. 6. Humoral response elicited in cattle by experimental vaccines. FMDV-specific antibody titers were measured by Lp ELISA. (A) Kinetics of total anti-FMDV
serum Abs. Each bar represents the mean Ab titer ± SD (n = 4) at 15, 30 and 45 dpv. (B) FMDV-specific serum IgG1 and IgG2 response at 45 dpv. Each point
represents the serum IgG1 or IgG2 anti-FMDV Ab titers (log10) of each animal. Isotype profiles at 45 dpv, expressed as mean Ab titers ± SD. ** p < 0.01, *p < 0.05.

7
J. Bidart et al. Virus Research 297 (2021) 198339

titer in VLPs-ISPA (2.46 ± 0.40), VLPs-ISA206 (2.67 ± 0.51) and iFMDV- vaccinated cattle the level of IFNγ secreted in vitro by mononuclear
ISA206 (2.70 ± 0.10) groups were significantly higher than VLPs group cells of those animals was measured.
(1.31 ± 0.30) (p < 0.01). Finally, at 45 dpv, Ab titers were significantly At 45 dpv, PBMCs from vaccinated bovines were cultured with 5 μg/
higher (p < 0.01) in the VLPs-ISPA (2.90 ± 0.41), VLPs-ISA206 (3.04 ± mL iFMDV for 5 days, then IFNγ levels were measured in the supernatant
0.40) and iFMDV-ISA206 (2.96 ± 0.43) groups as compared to the VLPs of cells cultures. The group immunized with VLPs-ISPA reached signif­
group (1.52 ± 0.31). Moreover, the FMDV humoral response achieved in icantly higher levels of IFNγ than those inoculated with VLPs alone (p <
VLPs-ISPA and VLPs-ISA 206 vaccinated bovines was similar to that 0.05) (Fig. 7).
achieved in iFMDV-ISA 206 vaccinated animals. The Expected Per­ An FMDV-specific PBMCs stimulation was evident in cattle immu­
centage of Protection (EPP) for FMDV A/Argentina/2001 strain ac­ nized with VLPs-ISPA. That results suggest an adaptive immune
cording to SENASA (Resolution N◦ 609/2017) was calculated by response against A2001 FMDV strain induced by the empty capsid plus
comparing antibody lpELISA titres from vaccinated cattle sera with re­ ISPA adjuvant.
sults from Protection against Podal Generalization (PPG) test (Maradei
et al., 2008). In cattle, vaccines that induced an average anti-FMDV Ab 4. Discussion
titre equal to or higher than 2.19 were considered protective (approved),
while vaccines with Abs anti-FMDV titres lower than 2.19 were The use of recombinant VLPs to produce a novel vaccine against
considered rejected as indicated by Argentine Animal Health Service FMDV offers many advantages, among which, avoiding the manipula­
regulations (SENASA, 2017). tion of infectious virus is by far the most important. Furthermore, cir­
Analysis of IgG1 isotype at 45 dpv showed no significant differences cumventing the manipulation of infectious virus avoids the need of
in Ab titers between cattle vaccinated with VLPs, VLPs-ISPA or VLPs-ISA complex and costly biocontainment production facilities. From a sani­
206, However, the group vaccinated with VLPs had a mean IgG1 titer of tary perspective, the use of empty capsids completely prevents the risk of
2.5, while the mean titers in the VLPs-ISPA or VLPs-ISA 206 groups an outbreak due to lack of full-inactivation or virus escape (Mignaqui
were: 3.36 and 3.74 respectively (Fig. 6B). Moreover, the VLPs-ISPA and et al., 2019).
VLPs-ISA 206 vaccines achieved higher IgG2 titers than the VLPs vaccine In this work, we examined the capacity of ISPA and ISA 206 adju­
(p<0.01). vants, to elicit a protective and specific immune response against FMDV
OIE recommends in-vitro viral neutralization as “gold standard” for when used in VLPs-based vaccines. For that purpose, we used a mouse
predicting vaccine-induced protection in-vivo in bovines (OIE- World model developed in our laboratory, in which the humoral and protective
Organisation for Animal Health, 2012). At 45 dpv, VNT results (Table 2)
showed a significant increase (p < 0.001) in mean VNT titers in the
VLPs-ISPA and VLPs-ISA 206 groups (2.1 ± 0.1 and 2.4 ± 0.2 respec­
tively) compared to the VLPs group (1.1 ± 0.1). Calves vaccinated with
iFMDV-ISA 206 also presented high levels of neutralizing Abs (2.45 ±
0.07). These values have been reported to be associated with 90 % Ex­
pected Percentage of Protection (EPP) (Maradei et al., 2008), which
estimates the likelihood that cattle would be protected against a chal­
lenge of 10.000 bovine infective doses after vaccination (OIE - World
Organisation for Animal Health, 2012). Neutralizing Ab induced by
empty capsids with adjuvants were capable of neutralize infectious
A2001 FMDV.
Humoral immunity induced by VLPs with ISPA or ISA 206 were
similar to those induced by formulation containing the complete inac­
tivated FMDV and commercial adjuvant (ISA206).
Fig. 7. IFNγ production by PBMC from calves inoculated with experimental
3.7. Immunization with VLPs plus ISPA or ISA206 increases IFNγ vaccines at 45 dpv. Cells were stimulated in vitro with iFMDV. Supernatants
were tested by ELISA. Each bar represents the IFN-γ levels (pg/mL) secreted by
secreted by bovine PBMCs
PBMCs from each bovine when they were stimulated in vitro with iFMDV, *p
< 0.05.
In order to determine the memory immunity induced in VLPs-

Table 2
Virus neutralizing antibody titers at 15, 30 and 45 days after inoculating cattle with experimental vaccines. Titers are expressed as log10 of the reciprocal of the serum
dilution which neutralizes 50 % of 100 TCID50 infective FMDV, using the fixed virus – variable serum method. * Significant differences against the VLPs group. *** (p
< 0.01).
Vaccine Animal n◦ VNT Mean ± SD VNT Mean ± SD VNT Mean ± SD
15dpv 30dpv 45dpv

2 <1 1 1.20
260 <1 1 1
VLPs
268 <1 1 1
274 <1 <1 1 1.0 ± 0.0 1.20 1.1 ± 0.1
255 1.60 2.00 2.00
257 1.90 1.90 2.00
VLPs-ISPA
263 1.30 2.00 2.20
276 1.60 1.6 ± 0.2 2.00 1.97 ± 0.05*** 2.20 2.1 ± 0.1***
259 1.80 2.61 2.61
264 1.90 2.20 2.51
VLPs-ISA206
273 1.80 2.20 2.40
275 1.90 1.85 ± 0.06 2.20 2.3 ± 0.2*** 2.20 2.4 ± 0.2***
256 1.90 2.20 2.51
iFMDV-ISA206
265 1.80 1.85 ± 0.07 2.20 2.2 ± 0.0 2.40 2.45 ± 0.07

8
J. Bidart et al. Virus Research 297 (2021) 198339

immune responses against FMDV in mice correlate with those elicited in experimental vaccines, both VLPs-ISPA and VLPs-ISA 206 induced an
cattle (Quattrocchi et al., 2005, 2011, 2013; Batista et al., 2010; increased response compared to the VLPs group. These results suggest
Zamorano et al., 2010; Langellotti et al., 2012, 2015; Romanutti et al., that ISPA and the Commercial Adjuvant improve the adaptive immune
2013; Bidart et al., 2020). Then, we studied the immune efficacy of the response against FMDV. ISPA and ISA 206 adjuvant alone has not been
formulations in cattle, a target species of the virus. shown to induce increase in cell proliferation, demonstrating that the
In the murine model, the use of ISPA, a new cage-like particle vaccine-induced proliferation was due to adjuvant and VLPs combina­
adjuvant, in VLPs-based vaccines allowed to achieve protection in all the tion or VLPs alone.
vaccinated animals against viral challenge while the use of VLPs alone The VLPs-ISPA and VLPs-ISA 206 vaccines triggered proliferation
induced a protective response in only 40 % of the mice. VLPs induced and IFNγ production in FMDV-specific CD8 + T lymphocytes (Fig. 5A y
similar protection levels when using ISPA as adjuvant or ISA 206. Mice 5C), as occurred in mice vaccinated with inactivated FMDV (Ostrowski
vaccinated with adjuvant alone were not protected against viral chal­ et al., 2005). Previous studies described that ISCOMs that is similar to
lenge, showing that protection corresponded to an adaptive response ISPA adjuvant improve the dendritic cell cross-presentation (Mar­
against the virus and was not due to innate immune mechanisms elicited askovsky et al., 2009; Wilson et al., 2012, 2014; den Brok et al., 2016).
by the adjuvant. Ab titers against FMDV were significantly elevated in The results described in the present work indicate that the VLPs- ISPA
mice that received VLPs-ISPA as compared to the group vaccinated with formulation generates a strong cellular response in agreement with
VLPs alone and similar to the Ab titers induced by the VLPs-ISA 206 previous reports that used ISPA cage-like particles (Bertona et al., 2017;
vaccine. These results correlate with the protection induced upon Prochetto et al., 2017).
challenge. It is important to mention that the levels of protection ach­ The results of the humoral immune response profiles obtained in the
ieved were the same as those induced by the Commercial Vaccine murine model were further confirmed in calves, as was reported by
(positive control) based on inactivated virus. The ELISA test used here Gnazzo et al. (2020). Numerous studies performed in cattle show a
measures antibodies that recognize inactivated FMDV, thus we could correlation between Ab titers against FMDV elicited by vaccination and
conclude that the formulations containing recombinant VLPs generate in vitro and in vivo protection upon experimental viral challenge. These
antibodies that recognize iFMDV. Neutralizing Ab titers showed a good correlations have allowed to estimate the Expected Percentage of Pro­
correlation with protection levels, substantiating the notion that they tection (EPP) to the homologous infection using titers of systemic
are an in vitro reflection of the immune response that occurs in vivo α-FMDV Ab measured by lpELISA or viral seroneutralization (Maradei
(McCullough et al., 1992; Mattion et al., 2009; OIE - World Organisation et al., 2008; Mattion et al., 2009; OIE - World Organisation for Animal
for Animal Health, 2012). Noteworthy, recombinant VLPs were able to Health, 2012).
induce antibodies that neutralize the infectious FMDV. Also, Ab titers In calves, the VLPs-ISPA formulation elicited total and neutralizing
against FMDV and VNT values induced in mice by VLPs-ISPA and anti-FMDV Ab titers corresponding to an Expected Percentage of Pro­
VLPs-ISA 206 were higher than the response induced by VLPs alone. tection above 90 %, similar to those obtained with the commercial
ISPA has shown its function as adjuvant by achieving the protection adjuvant (inactivated FMDV and ISA 206) and commercial vaccine
induced by VLPs to levels similar to those induced by the commercial (Supplementary Table 1) (Maradei et al., 2008; Robiolo et al., 2010;
vaccine. The latter contains FMDV strains O1/Campos/Brazil/58, Senasa, 2017). An acceptable inactivated vaccine should induce 75 %
A24/Cruzeiro/Brazil/55, C3/Indaial/Brazil/71 and A/Argentina/2001, protection in cattle (OIE - World Organisation for Animal Health, 2012).
all of which bear epitopes that participate in the immune response On the other hand, the Ab titers reached correspond to an EPP of 25.0 %
against FMDV (Mattion et al., 2004). in calves vaccinated with VLPs, 92.8 % in calves vaccinated with
When isotype profiles were characterized, increases in IgG1, IgG2a, VLPs-ISPA, 90.6 % in VLPs-ISA 206 group calves and 92.8 % in VFAi-ISA
IgGb and IgG3 levels were observed in the VLPs-ISPA, VLP-ISA 206 and 206 group calves (Senasa, 2017). EPP reached by formulations con­
Commercial vaccine groups as compared to the group inoculated with taining VLPs and adjuvants would be sufficient to authorize their use in
VLPs alone. Moreover, IgG1 and IgG2b isotypes were detected in mice cattle, according to the current Argentinean regulations.
vaccinated with VLPs-ISPA. These are promising results because Gnazzo IgG2 titers were higher in VLPs plus adjuvant formulations which is
et al. (2020) reported that in the mouse model, IgG1 and IgG2b isotypes related to pathogen opsonization. Bovine macrophages and neutrophils
are important in the protection against FMDV O1 Campos. possess an immunoglobulin receptor to which IgG2 can bind (Tizard,
VLPs-ISPA-vaccinated mice showed higher IgG3 isotype levels than the 1998). On the other hand, we detected a significant increase in IFN-γ
VLPs-ISA 206 group. In mice, is reported that an ISPA-BSA formulation secretion in cattle vaccinated with VLPs-ISPA as compared to cattle
elicited increased IgG1 and IgG2a titers than BSA alone or BSA-CFA or vaccinated with VLPs alone or VLPs-ISA 206. IFN-γ has been reported to
OHAL-BSA or ISA206-BSA (Bertona et al., 2017). In addition, a formu­ display activity against FMDV (Summerfield et al., 2009), by controlling
lation containing ISPA and Tripanosoma cruzi trans-sialidase (mTS) was viral replication and spreading within the host through natural killer cell
shown to induce similar IgG1 and IgG2a titers to those induced by a and macrophage activation (Zhang et al., 2002). Our data of increased
formulation using ISCOMATRIX™-mTS (a commercial ISCOM) (Bertona IFN-γ and increased presence of IgG2 in calves vaccinated with
et al., 2017). It is well described that IgG1 and IgG3-FMDV immune VLPs-ISPA, correlate with what was found by other researchers. Thus, a
complexes are recognized by FcγRI receptors, present in macrophages, positive correlation between IFN-γ response and vaccine-induced pro­
monocytes and dendritic cells, leading to virus clearance (van der Poel tection as well as reduction of long-term persistence of FMDV has been
et al., 2011). Also, IgG2a, IgG2b and IgG3 isotypes play a significant role observed in cattle (Oh et al., 2012). In addition, IFN-γ production is
in FMDV clearance trough complement-mediated phagocytosis associated with a Th1 profile and an increased IgG2 production (Estes
(McCullough et al., 1988; Pérez Filgueira et al., 1995). Moreover, it has and Brown, 2002). The numbers of cattle included in this pilot study
been reported that mice inoculated with inactivated FMDV plus some were as those used in other preliminary studies on vaccine candidates
adjuvants generate a complement-fixing IgG profile that correlates with (Bachmann and Zinkernagel, 1997; Lee et al., 2005), although they were
protection upon FMDV challenge (Pérez Filgueira et al., 1995; Batista insufficient for statistical analysis (Soria et al., 2017). However, the
et al., 2010). The different isotype profiles elicited by inoculation with results obtained serve as proof of concept of the usefulness of VLPs as
these vaccines indicate a high response for all isotypes that guarantee an antigen and ISPA as adjuvant in FMDV vaccines. Although in calves, the
effective immune protection based in a balanced Th1/Th2 response. The iFMDV-ISA 206 group is too small to make any statistical comparisons,
humoral response against VLP antigen was notably taking in mind that it the neutralizing Ab titters induced by the vaccine containing the com­
was measured using FMDV instead of VLP and the values were similar to plete inactivated FMDV and commercial adjuvant were similar to those
commercial vaccine that is formulated with FMDV. induced by VLPs with ISPA or ISA 206.
Regarding the cellular immune response obtained in mice using the The action of ISPA was similar, in terms of levels, kinetics and

9
J. Bidart et al. Virus Research 297 (2021) 198339

profiles of humoral responses, to the commercial oil adjuvant used in Ethics statement
this study. ISPA has also been reported to have a better performance
than ISCOMATRIX™ (Bertona et al., 2017). Also, immunization with a The animal study was reviewed and approved by Comité Institu­
formulation based on Trypanosoma cruzi recombinant transialidase cional para el Cuidado y Uso de Animales de Experimentación (CICUAE)
(mTS) and ISPA elicited high levels of protection upon challenge (Ber­ - Centro de Investigación en Ciencias Veterinarias y Agronómicas del
tona et al., 2017). Moreover, this vaccine favorably modulates the reg­ INTA, under protocol Number: 26/2016.
ulatory arm of the immune system to reach immune protection against
the parasite (Prochetto et al., 2017). Author contributions
Future work will be devoted to examine whether ISPA exerts its
action of enhancement of the immune response and protection levels by JEB: Conceptualization; Methodology; Formal analysis; Investiga­
promoting the virus presentation to the immune effectors. Some authors tion; Writing - Original Draft; Visualization
have reported that ISCOMs induce local recruitment, activation and AM: Conceptualization; Methodology; Formal analysis; Resources;
maturation of immune cells, such as dendritic cells, granulocytes, F4/80 Writing - Original Draft
in. cells, T-, B- and NK-cells (Reed et al., 2009; Sun et al., 2009; Reimer CK: Validation; Investigation
et al., 2012), increasing in this way the chances of the antigen to come in GL: Validation; Investigation, Resources;
contact with immune cells. In addition, Brok et al. (2016) proved that MG: Validation; Investigation
saponin-based adjuvants enhance antigen cross-presentation by den­ IS: Validation; Formal analysis
dritic cells and T-cell activation. RG: Validation; Investigation
VLPs-ISPA and VLPs-ISA 206 elicit high FMDV Ab titers. In cattle, the CS: Resources, Writing Review & Editing
immune response elicited by FMDV (A22 Iraq strain) VLPs produced CL: Methodology; Formal analysis; Investigation; Writing - Original
using the baculovirus technology and oil adjuvant was analyzed in a Draft
study by Porta et al. (2013). Two vaccinations with 12 μg/dose elicited VQ: Methodology; Formal analysis; Investigation; Writing - Original
neutralizing Abs and 50 % protection against challenge. When the Draft
capsids were mutated to increase their stability, the protection levels YD: Conceptualization; Methodology, Resources;
induced were of 75 %. In another study, Li et al. (2012) showed 80 % WA: Conceptualization; Methodology; Writing - Review & Editing
protection with one dose of A/WH/CHA/09 VLPs-ISA206 formulation. IM: Conceptualization; Methodology; Writing - Review & Editing
Our results show a comparable or higher performance of the VLPs/ISPA PZ: Conceptualization; Methodology; Writing - Original Draft;
and VLPs/CA formulations since with a single 25 μg dose, the levels of Writing Review & Editing, Visualization; Supervision; Project Admin­
induced neutralizing Abs correlate with 90 % of protection. istration; Funding acquisition.
The recombinant VLPs used in this study were produced by transient
transfection of suspension-growing mammalian cell cultures in serum Acknowledgments
free medium using a pTT5-based plasmid. VLP yields achieved using this
technology are between 3− 7 mg per liter of cell culture and these levels The authors acknowledge the skilled assistance of Mr. Diego Com­
are comparable to the ones obtained after infection of BHK-21 cells with paired with cell cultures and the technical help of Ms. Pamela Angeletti,
FMDV in vaccine facilities (Mignaqui et al., 2013, 2020; Li et al., 2011). the valuable work of BSc Juan Schammas and Mr. Javier Rosende in the
Moreover, the scalability and reproducibility of this methodology for BSL-4 OIE facilities at CICVyA-INTA, the technical assistance at INTA
VLP production has recently being reported (Mignaqui et al., 2020). bioterium of PhD. Gabriel Pinto and Ms. Sandra Rivarola, the help with
Thus, overall this technology is a safe and cost-effective method of an­ flow cytometer measurements of Ms. Paula Blanco from CIO-FUCA and
tigen production to develop a new non-infectious vaccine for controlling BSc. Julieta Alcain from IMEX-CONICET, and the provision of A/
FMD in vaccination-free countries, as well as in FMD-endemic countries Argentina/2001 BEI-inactivated FMDV by Biogenesis Bago S.A. The
and in the event of an outbreak. research project was fully sponsored by a grant from National Agency for
In conclusion, recombinant VLPs can be used in combination with a Scientific and Technological Promotion, Ministry of Science and Tech­
new cage-particle adjuvant, ISPA, and ISA206 to develop a novel vac­ nology, Argentina, (PID 2013-0022); and a grant from the National
cine against FMD. Results showed ISPA is useful in increasing and Institute of Agricultural Technology (INTA, Project PD-I-105).
modulating the humoral and cellular responses in vaccinated mice and
cattle, yielding enhanced protection against challenge. These results are Appendix A. Supplementary data
similar to induce with ISA 206. Taken together, these results are
promising for the development of a novel FMD vaccine with a recom­ Supplementary material related to this article can be found, in the
binant, non-infectious antigen and a new, cost-effective and innovative online version, at doi:https://doi.org/10.1016/j.virusres.2021.198339.
adjuvant.
References
Declaration of interest statement
Acharya, R., Fry, E., Stuart, D., Fox, G., Rowlands, D., Brown, F., 1989. The three-
The authors declare that the research was conducted in the absence dimensional structure of foot-and-mouth disease virus at 2.9 Å resolution. Nature.
https://doi.org/10.1038/337709a0.
of any commercial or financial relationships that could be construed as a
Bachmann, M.F., Zinkernagel, R.M., 1997. Neutralizing Antiviral B cell responses. Annu.
potential conflict of interest. Rev. Immunol. https://doi.org/10.1146/annurev.immunol.15.1.235.
Cardillo S, employee of Biogénesis Bagó, declares that his judgment Baldi, L., Hacker, D.L., Adam, M., Wurm, F.M., 2007. Recombinant protein production by
large-scale transient gene expression in mammalian cells: state of the art and future
and objectivity was not biased by his contractual condition.
perspectives. Biotechnol. Lett. https://doi.org/10.1007/s10529-006-9297-y.
Barnett, P.V., Pullen, L., Williams, L., Doel, T.R., 1996. International bank for foot-and-
Data availability statement mouth disease vaccine: assessment of Montanide ISA 25 and ISA 206, two
commercially available oil adjuvants. Vaccine 14, 1187–1198. https://doi.org/
10.1016/S0264-410X(96)00055-2.
The datasets generated for this study are available on request to the Barnett, P.V., Cox, S.J., Aggarwal, N., Gerber, H., McCullough, K.C., 2002. Further
corresponding author studies on the early protective responses of pigs following immunisation with high
potency foot and mouth disease vaccine. Vaccine 20, 3197–3208. https://doi.org/
10.1016/s0264-410x(02)00242-6.
Batista, A., Quattrocchi, V., Olivera, V., Langellotti, C., Pappalardo, J.S., Di Giacomo, S.,
et al., 2010. Adjuvant effect of CliptoxTM on the protective immune response

10
J. Bidart et al. Virus Research 297 (2021) 198339

induced by an inactivated vaccine against foot and mouth disease virus in mice. and inactivated virus. Antiviral Res. 94, 62–71. https://doi.org/10.1016/j.
Vaccine 28, 6361–6366. https://doi.org/10.1016/j.vaccine.2010.06.098. antiviral.2012.02.009.
Belsham, G.J., 2005. Translation and replication of FMDV RNA. Curr. Top. Microbiol. Langellotti, C., Cesar, G., Soria, I., Quattrocchi, V., Jancic, C., Zamorano, P., et al., 2015.
Immunol. https://doi.org/10.1007/3-540-27109-0_3. Foot-and-mouth disease virus infection of dendritic cells triggers phosphorylation of
Bertona, D., Pujato, N., Bontempi, I., Gonzalez, V., Cabrera, G., Gugliotta, L., et al., 2017. ERK1/2 inducing class I presentation and apoptosis. Vaccine. https://doi.org/
Development and assessment of a new cage-like particle adjuvant. J. Pharm. 10.1016/j.vaccine.2015.07.038.
Pharmacol. 69, 1293–1303. https://doi.org/10.1111/jphp.12768. Lee, B.O., Rangel-Moreno, J., Moyron-Quiroz, J.E., Hartson, L., Makris, M., Sprague, F.,
Bidart, J., Kornuta, C., Gammella, M., Gnazzo, V., Soria, I., Langellotti, C., et al., 2020. et al., 2005. CD4 t cell-independent antibody response promotes resolution of
A new cage-like particle adjuvant enhances protection of foot-and-Mouth disease primary influenza infection and helps to prevent reinfection. J. Immunol. https://
vaccine. Front. Vet. Sci. 7, 396. https://doi.org/10.3389/fvets.2020.00396. doi.org/10.4049/jimmunol.175.9.5827.
Borca, M.V., Fernández, F.M., Sadir, A.M., Braun, M., Schudel, A.A., 1986. Immune Li, Z., Yin, X., Yi, Y., Li, X., Li, B., et al., 2011. FMD subunit vaccine produced using a
response to foot-and-mouth disease virus in a murine experimental model: effective silkworm-baculovirus expression system: protective efficacy against two type Asia 1
thymus-independent primary and secondary reaction. Immunology 59 (2), 261–267. isolates in cattle. Vet. Microbiol. 149, 99–103. Available:http://www.ncbi.nlm.nih.
http://www.ncbi.nlm.nih.gov/pubmed/3490436. gov/pubmed/21109368Accessed 30 January 2013.
Caridi, F., Vázquez-Calvo, A., Sobrino, F., Martín-Acebes, M.A., 2015. The pH stability of Li, Z., Yi, Y., Yin, X., Zhang, Y., Liu, M., Liu, H., et al., 2012. Development of a foot-and-
foot-and-Mouth disease virus particles is modulated by residues located at the Mouth disease virus serotype a empty capsid subunit vaccine using silkworm
pentameric interface and in the N terminus of VP1. J. Virol. https://doi.org/ (Bombyx mori) pupae. PLoS One 7, e43849. https://doi.org/10.1371/journal.
10.1128/jvi.03358-14. pone.0043849.
Childerstone, A.J., Cedillo-Baron, L., Foster-Cuevas, M., Parkhouse, R.M.E., 1999. López, O.J., Sadir, A.M., Borca, M.V., Fernández, F.M., Braun, M., Schudel, A.A., 1990.
Demonstration of bovine CD8+ T cell responses to foot-and-mouth disease virus. Immune response to foot-and-Mouth disease virus in an experimental murine model
J. Gen. Virol. 80, 663–669. https://doi.org/10.1099/0022-1317-80-3-663. II. Basis of persistent antibody reaction. Vet. Immunol. Immunopathol. 24 (4),
Cox, S.J., Barnett, P.V., 2009. Experimental evaluation of foot-and-mouth disease 313–321. https://doi.org/10.1016/0165-2427(90)90002-A.
vaccines for emergency use in ruminants and pigs: a review. Vet. Res. 40, 13. Maradei, E., La Torre, J., Robiolo, B., Esteves, J., Seki, C., Pedemonte, A., et al., 2008.
https://doi.org/10.1051/vetres:2008051. Updating of the correlation between lpELISA titers and protection from virus
Cox, S.J., Aggarwal, N., Statham, R.J., Barnett, P.V., 2003. Longevity of antibody and challenge for the assessment of the potency of polyvalent aphtovirus vaccines in
cytokine responses following vaccination with high potency emergency FMD Argentina. Vaccine 26, 6577–6586. https://doi.org/10.1016/j.vaccine.2008.09.033.
vaccines. Vaccine. https://doi.org/10.1016/S0264-410X(02)00691-6. Maraskovsky, E., Schnurr, M., Wilson, N.S., Robson, N.C., Boyle, J., Drane, D., 2009.
den Brok, M.H., Büll, C., Wassink, M., de Graaf, A.M., Wagenaars, J.A., Minderman, M., Development of prophylactic and therapeutic vaccines using the ISCOMATRIX
et al., 2016. Saponin-based adjuvants induce cross-presentation in dendritic cells by adjuvant. Immunol. Cell Biol. 87, 371–376. https://doi.org/10.1038/icb.2009.21.
intracellular lipid body formation. Nat. Commun. 7, 13324. https://doi.org/ Mateo, R., Luna, E., Rincón, V., Mateu, M.G., 2008. Engineering viable foot-and-Mouth
10.1038/ncomms13324. disease viruses with increased thermostability as a step in the development of
Doel, T.R., 2003. FMD vaccines. Virus Res. 91, 81–99. https://doi.org/10.1016/S0168- improved vaccines. J. Virol. https://doi.org/10.1128/jvi.01553-08.
1702(02)00261-7. Mattion, N., Konig, G., Seki, C., Smitsaart, E., Maradei, E., Robiolo, B., et al., 2004.
Dus Santos, M.J., Wigdorovitz, A., Maradei, E., Periolo, O., Smitsaart, E., Borca, M.V., Reintroduction of foot-and-mouth disease in Argentina: characterisation of the
Sadir, A.M., 2000. A comparison of methods for measuring the antibody response in isolates and development of tools for the control and eradication of the disease.
mice and cattle following vaccination against foot and mouth disease. Vet. Res. Vaccine 22, 4149–4162. https://doi.org/10.1016/j.vaccine.2004.06.040.
Commun. 24 (4), 261–273. Mattion, N., Goris, N., Willems, T., Robiolo, B., Maradei, E., Beascoechea, C.P., et al.,
Ellard, F.M., Drew, J., Blakemore, W.E., Stuart, D.I., King, A.M.Q., 1999. Evidence for the 2009. Some guidelines for determining foot-and-mouth disease vaccine strain
role of His-142 of protein 1C in the acid-induced disassembly of foot-and-mouth matching by serology. Vaccine. https://doi.org/10.1016/j.vaccine.2008.11.026.
disease virus capsids. J. Gen. Virol. https://doi.org/10.1099/0022-1317-80-8-1911. McCullough, K.C., Parkinson, D., Crowther, J.R., 1988. Opsonization-enhanced
Estes, D.M., Brown, W.C., 2002. Type 1 and type 2 responses in regulation of Ig isotype phagocytosis of foot-and-mouth disease virus. Immunology 65, 187–191.
expression in cattle. Vet. Immunol. Immunopathol. 90, 1–10. https://doi.org/ McCullough, K.C., Bruckner, L., Schaffner, R., Fraefel, W., Muller, H.K., Kihm, U., 1992.
10.1016/S0165-2427(02)00201-5. Relationship between the anti-FMD virus antibody reaction as measured by different
Fernandez, F.M., Borca, M.V., Sadir, A.M., Fondevila, N., Mayo, J., Schudel, A.A., 1986. assays, and protection in vivo against challenge infection. Vet. Microbiol. 30,
Foot-and-mouth disease virus (FMDV) experimental infection: susceptibility and 99–112.
immune response of adult mice. Vet. Microbiol. 12 (June (1)), 15–24. Medina, G.N., Segundo, F.D.-S., Stenfeldt, C., Arzt, J., de los Santos, T., 2018. The
Fontana, D., Marsili, F., Etcheverrigaray, M., Kratje, R., Prieto, C., 2020. Rabies VLPs different tactics of foot-and-Mouth disease virus to evade innate immunity. Front.
adjuvanted with saponin-based liposomes induce enhanced immunogenicity Microbiol. 9, 1–22. https://doi.org/10.3389/fmicb.2018.02644.
mediated by neutralizing antibodies in cattle, dogs and cats. J. Virol. Methods 286, Mignaqui, A.C., Ruiz, V., Perret, S., St-Laurent, G., Singh Chahal, P., Transfiguracion, J.,
113966. https://doi.org/10.1016/j.jviromet.2020.113966. et al., 2013. Transient gene expression in serum-free suspension-growing
Gnazzo, V., Quattrocchi, V., Soria, I., Pereyra, E., Langellotti, C., Pedemonte, A., et al., mammalian cells for the production of foot-and-Mouth disease virus empty capsids.
2020. Mouse model as an efficacy test for foot-and-mouth disease vaccines. PLoS One 8, e72800. https://doi.org/10.1371/journal.pone.0072800. Available at:
Transbound. Emerg. Dis. https://doi.org/10.1111/tbed.13591 tbed.13591. Mignaqui, A.C., Ruiz, V., Durocher, Y., Wigdorovitz, A., 2019. Advances in novel
Gomes, I., Astudillo, V., 1975. Foot and mouth disease: evaluation of mouse protection vaccines for foot and mouth disease: focus on recombinant empty capsids. Crit. Rev.
test results in relation to cattle immunity. Boletin Centro Panamericano Aftosa 17, Biotechnol. 39, 306–320. https://doi.org/10.1080/07388551.2018.1554619.
9–16. Mignaqui, A.C., Ferella, A., Cass, B., Mukankurayija, L., L’Abbé, D., Bisson, L.,
Grubman, M.J., 2005. Development of novel strategies to control foot-and-mouth Sánchez, C., Scian, R., Cardillo, S.B., Durocher, Y., Wigdorovitz, A., 2020. Foot-and-
disease: marker vaccines and antivirals. Biologicals. https://doi.org/10.1016/j. Mouth disease: optimization, reproducibility, and scalability of high-yield
biologicals.2005.08.009. production of virus-like particles for a next-generation vaccine. Front. Vet. Sci. 7
Grubman, M.J., Baxt, B., 2004. Foot-and-Mouth disease. Clin. Microbiol. Rev. 17, (September), 1–9. https://doi.org/10.3389/fvets.2020.00601.
465–493. https://doi.org/10.1128/CMR.17.2.465. Mohan, T., Verma, P., Rao, D.N., 2013. Novel adjuvants & delivery vehicles for vaccines
Gullberg, M., Lohse, L., Bøtner, A., McInerney, G.M., Burman, A., Jackson, T., et al., development: a road ahead. Indian J. Med. Res. 138, 779–795. Available at: http
2016. A prime-boost vaccination strategy in cattle to prevent foot-and-Mouth disease ://www.ncbi.nlm.nih.gov/pubmed/24434331.
using a “Single-Cycle” alphavirus vector and empty capsid particles. PLoS One 11, Monaghan, P., Gold, S., Simpson, J., Zhang, Z., Weinreb, P.H., Violette, S.M., et al., 2005.
e0157435. https://doi.org/10.1371/journal.pone.0157435. The αvβ6 integrin receptor for Foot-and-mouth disease virus is expressed
Habiela, M., Seago, J., Perez-Martin, E., Waters, R., Windsor, M., Salguero, F.J., et al., constitutively on the epithelial cells targeted in cattle. J. Gen. Virol. https://doi.org/
2014. Laboratory animal models to study foot-and-mouth disease: a review with 10.1099/vir.0.81172-0.
emphasis on natural and vaccine-induced immunity. J. Gen. Virol. 95, 2329–2345. Morein, B., Sundquist, B., Höglund, S., Dalsgaard, K., Osterhaus, A., 1984. Iscom, a novel
https://doi.org/10.1099/vir.0.068270-0. structure for antigenic presentation of membrane proteins from enveloped viruses.
Hamblin, C., Barnett, I.T.R., Hedger, R.S., 1986. A new enzyme-linked immunosorbent Nature. https://doi.org/10.1038/308457a0.
assay (ELISA) for the detection of antibodies against foot-and-mouth disease virus I. Oh, Y., Fleming, L., Statham, B., Hamblin, P., Barnett, P., Paton, D.J., et al., 2012.
Development and method of ELISA. J. Immunol. Methods 93, 115–121. https://doi. Interferon-γ induced by in vitro Re-Stimulation of CD4+ T-Cells correlates with in
org/10.1016/0022-1759(86)90441-2. vivo FMD vaccine induced protection of cattle against disease and persistent
Hamblin, C., Kitching, R.P., Donaldson, A.I., Crowther, J.R., Barnett, I.T.R., 1987. A new infection. PLoS One. https://doi.org/10.1371/journal.pone.0044365.
enzyme-linked immunosorbent assay (ELISA) for the detection of antibodies against OIE, 2018. OIE - World Organisation for Animal Health Act N◦ 22. Available at: htt
foot-and-mouth disease virus III. Evaluation of antibodies after infection and p://www.oie.int/fileadmin/Home/esp/Animal_Health_in_the_World/docs/pdf/Re
vaccination. Epidemiol. Infect. 99, 733–744. solutions/2018/E22_RESO_2018_FMD.pdf.
Jackson, T., Sheppard, D., Denyer, M., Blakemore, W., King, A.M.Q., 2000. The epithelial OIE - World Organisation for Animal Health, 2012. Foot and mouth disease (infection
integrin αvβ6 is a receptor for foot-and-Mouth disease virus. J. Virol. https://doi. with FMDV). OIE Terrestrial Manual. OIE 2012 (Paris: OIE 2012), pp. 1–32.
org/10.1128/jvi.74.11.4949-4956.2000. Available at: https://books.google.com.ar/books?id=l5b5AAAACAAJ.
Kotecha, A., Seago, J., Scott, K., Burman, A., Loureiro, S., Ren, J., et al., 2015. Structure- Ostrowski, M., Vermeulen, M., Zabal, O., Geffner, J.R., Sadir, A.M., Lopez, O.J., 2005.
based energetics of protein interfaces guides foot-and-mouth disease virus vaccine Impairment of thymus-dependent responses by murine dendritic cells infected with
design. Nat. Struct. Mol. Biol. 22, 788–794. https://doi.org/10.1038/nsmb.3096. foot-and-Mouth disease virus. J. Immunol. https://doi.org/10.4049/
Langellotti, C., Quattrocchi, V., Alvarez, C., Ostrowski, M., Gnazzo, V., Zamorano, P., jimmunol.175.6.3971.
et al., 2012. Foot-and-mouth disease virus causes a decrease in spleen dendritic cells Parida, S., 2009. Vaccination against foot-and-mouth disease virus: strategies and
and the early release of IFN-α in the plasma of mice. Differences between infectious effectiveness. Expert Rev. Vaccines. https://doi.org/10.1586/14760584.8.3.347.

11
J. Bidart et al. Virus Research 297 (2021) 198339

Patch, J.R., Kenney, M., Pacheco, J.M., Grubman, M.J., Golde, W.T., 2013. vaccine matching carried out by liquid phase ELISA and virus neutralization tests.
Characterization of cytotoxic T lymphocyte function after foot-and-mouth disease Vaccine 28, 6235–6241. https://doi.org/10.1016/j.vaccine.2010.07.012.
virus infection and vaccination. Viral Immunol. 26, 239–249. https://doi.org/ Rodriguez, L.L., Grubman, M.J., 2009. Foot and mouth disease virus vaccines. Vaccine.
10.1089/vim.2013.0011. https://doi.org/10.1016/j.vaccine.2009.08.039.
Patil, P.K., Bayry, J., Ramakrishna, C., Hugar, B., Misra, L.D., Prabhudas, K., et al., 2002. Romanutti, C., D’Antuono, A., Palacios, C., Quattrocchi, V., Zamorano, P., La Torre, J.,
Immune responses of sheep to quadrivalent double emulsion foot-and-mouth disease et al., 2013. Evaluation of the immune response elicited by vaccination with viral
vaccines: rate of development of immunity and variations among other ruminants. vectors encoding FMDV capsid proteins and boosted with inactivated virus. Vet.
J. Clin. Microbiol. https://doi.org/10.1128/JCM.40.11.4367-4371.2002. Microbiol. 165, 333–340. https://doi.org/10.1016/j.vetmic.2013.04.017.
Pay, T.W.F., Hingley, P.H., Radlett, P.J., Black, L., O’Reilly, K.J., 1983. The correlation Romera, S., Puntel, M., Quattrocchi, V., Zajac, P..Del, Zamorano, P., Blanco Viera, J.,
between the 146S antigen dose with the serum neutralising antibody response and et al., 2014. Protection induced by a glycoprotein E-deleted bovine herpesvirus type
with protection from challenge induced by FMD vaccines. In: Report of the Meeting 1 marker strain used either as an inactivated or live attenuated vaccine in cattle.
of the Research Group of the Standing Technical Committee of the European BMC Vet. Res. 10, 8. https://doi.org/10.1186/1746-6148-10-8.
Commission for the Control of Foot-and-Mouth Disease. (Lelystad, Netherlands), Senasa, 2017. Servicio Nacional de Sanidad y Calidad Agroalimentaria Res 609/2017.
(FAO, Rome). CABA, Argentina.
Pérez Filgueira, D.M., Berinstein, A., Smitsaart, E., Borca, M.V., Sadir, A.M., 1995. Singh, M., 2006. Vaccine Adjuvants and Delivery Systems. https://doi.org/10.1002/
Isotype profiles induced in Balb/c mice during foot and mouth disease (FMD) virus 9780470134931.
infection or immunization with different FMD vaccine formulations. Vaccine 13, Soria, I., Quattrocchi, V., Langellotti, C., Gammella, M., Digiacomo, S., Garcia de la
953–960. https://doi.org/10.1016/0264-410X(95)00078-F. Torre, B., et al., 2017. Dendrimeric peptides can confer protection against foot-and-
Pham, P.L., Kamen, A., Durocher, Y., 2006. Large-scale transfection of mammalian cells mouth disease virus in cattle. PLoS One 12. https://doi.org/10.1371/journal.
for the fast production of recombinant protein. Mol. Biotechnol. https://doi.org/ pone.0185184.
10.1385/MB:34:2:225. Stenfeldt, C., Eschbaumer, M., Smoliga, G.R., Rodriguez, L.L., Zhu, J., Arzt, J., 2017.
Porta, C., Kotecha, A., Burman, A., Jackson, T., Ren, J., Loureiro, S., et al., 2013. Rational Clearance of a persistent picornavirus infection is associated with enhanced pro-
engineering of recombinant picornavirus capsids to produce safe, protective vaccine apoptotic and cellular immune responses. Sci. Rep. 7, 1–15. https://doi.org/
antigen. PLoS Pathog. 9, e1003255. https://doi.org/10.1371/journal.ppat.1003255. 10.1038/s41598-017-18112-4.
Prochetto, E., Roldán, C., Bontempi, I.A., Bertona, D., Peverengo, L., Vicco, M.H., et al., Summerfield, A., Guzylack-Piriou, L., Harwood, L., McCullough, K.C., 2009. Innate
2017. Trans-sialidase-based vaccine candidate protects against Trypanosoma cruzi immune responses against foot-and-mouth disease virus: current understanding and
infection, not only inducing an effector immune response but also affecting cells with future directions. Vet. Immunol. Immunopathol. https://doi.org/10.1016/j.
regulatory/suppressor phenotype. Oncotarget 8, 58003–58020. https://doi.org/ vetimm.2008.10.296.
10.18632/oncotarget.18217. Sun, H.-X., Xie, Y., Ye, Y.-P., 2009. ISCOMs and ISCOMATRIX. Vaccine 27, 4388–4401.
Quattrocchi, V., Bianco, V., Fondevila, N., Pappalardo, S., Sadir, A., Zamorano, P., 2005. https://doi.org/10.1016/j.vaccine.2009.05.032.
Use of new adjuvants in an emergency vaccine against foot-and-mouth disease virus: Thompson, D., Muriel, P., Russell, D., Osborne, P., Bromley, A., Rowland, M., et al.,
evaluation of conferred immunity. Dev. Biol. (Basel) 481–497. 2002. Economic costs of the foot and mouth disease outbreak in the United Kingdom
Quattrocchi, V., Langellotti, C., Pappalardo, J.S., Olivera, V., Di Giacomo, S., van in 2001. OIE Rev. Sci. Tech. https://doi.org/10.20506/rst.21.3.1353.
Rooijen, N., et al., 2011. Role of macrophages in early protective immune responses Tizard, I., 1998. Inmunología Veterinaria, 5e ed. McGraw-HillInteramericana Mexico:
induced by two vaccines against foot and mouth disease. Antiviral Res. 92, 262–270. McGraw-Hill Interamericana Mexico.
https://doi.org/10.1016/j.antiviral.2011.08.007. van Bekkum, J.G., 1969. Correlation between serum antibody level and protection
Quattrocchi, V., Molinari, P., Langellotti, C., Gnazzo, V., Taboga, O., Zamorano, P., 2013. against challenge with FMD-virus. In: Sess. Res. Group Stand. Tech. Comm. Eur.
Co-inoculation of baculovirus and FMDV vaccine in mice, elicits very early Commission Cont. FMD. Brescia, Italy. Rome: FAO, 38 pp.
protection against foot and mouth disease virus without interfering with long lasting van der Poel, C.E., Spaapen, R.M., van de Winkel, J.G.J., Leusen, J.H.W., 2011.
immunity. Vaccine 31, 2713–2718. https://doi.org/10.1016/j.vaccine.2013.03.067. Functional characteristics of the high affinity IgG receptor, FcγRI. J. Immunol.
Quattrocchi, V., Pappalardo, J.S., Langellotti, C., Smitsaart, E., Fondevila, N., https://doi.org/10.4049/jimmunol.1003526.
Zamorano, P., 2014. Early protection against foot-and-mouth disease virus in cattle Wilson, N.S., Yang, B., Morelli, A.B., Koernig, S., Yang, A., Loeser, S., et al., 2012.
using an inactivated vaccine formulated with Montanide ESSAI IMS D 12802 VG PR ISCOMATRIX vaccines mediate CD8 + T-cell cross-priming by a MyD88-dependent
adjuvant. Vaccine 32, 2167–2172. https://doi.org/10.1016/j.vaccine.2014.02.061. signaling pathway. Immunol. Cell Biol. 90, 540–552. https://doi.org/10.1038/
Reed, S.G., Bertholet, S., Coler, R.N., Friede, M., 2009. New horizons in adjuvants for icb.2011.71.
vaccine development. Trends Immunol. 30, 23–32. https://doi.org/10.1016/j. Wilson, N.S., Duewell, P., Yang, B., Li, Y., Marsters, S., Koernig, S., et al., 2014.
it.2008.09.006. Inflammasome-dependent and -Independent IL-18 production mediates immunity to
Reimer, J.M., Karlsson, K.H., Lövgren-Bengtsson, K., Magnusson, S.E., Fuentes, A., the ISCOMATRIX adjuvant. J. Immunol. 192, 3259–3268. https://doi.org/10.4049/
Stertman, L., 2012. Matrix-mTM adjuvant induces local recruitment, activation and jimmunol.1302011.
maturation of central immune cells in absence of antigen. PLoS One. https://doi.org/ Xiao, Y., Chen, H.-Y., Wang, Y., Yin, B., Lv, C., Mo, X., et al., 2016. Large-scale
10.1371/journal.pone.0041451. production of foot-and-mouth disease virus (serotype Asia1) VLP vaccine in
Rigden, R.C., Carrasco, C.P., Barnett, P.V., Summerfield, A., McCullough, K.C., 2003. Escherichia coli and protection potency evaluation in cattle. BMC Biotechnol. 16, 56.
Innate immune responses following emergency vaccination against foot-and-mouth https://doi.org/10.1186/s12896-016-0285-6.
disease virus in pigs. Vaccine. https://doi.org/10.1016/S0264-410X(02)00663-1. Zamorano, P., Decheneux, C., Quattrocchi, V., Olivera, V., Langellotti, C., DiGiacomo, S.,
Rincón, V., Rodríguez-Huete, A., López-Argüello, S., Ibarra-Molero, B., Sanchez-Ruiz, J. et al., 2010. Vaccination against foot-and-mouth disease, association between
M., Harmsen, M.M., et al., 2014. Identification of the structural basis of thermal humoral immune response in cattle and mice. Practical Alternatives to Reduce
lability of a virus provides a rationale for improved vaccines. Structure. https://doi. Animal Testing in Quality Control of Veterinary Biologicals in the Americas. OIE-
org/10.1016/j.str.2014.08.019. IABS (Ciudad Autonoma de Buenos Aires: OIE-IABS), pp. 100–111.
Robiolo, B., La Torre, J., Maradei, E., Beascoechea, C.P., Perez, A., Seki, C., et al., 2010. Zhang, Z.D., Hutching, G., Kitching, P., Alexandersen, S., 2002. The effects of gamma
Confidence in indirect assessment of foot-and-mouth disease vaccine potency and interferon on replication of foot-and-mouth disease virus in persistently infected
bovine cells. Arch. Virol. https://doi.org/10.1007/s00705-002-0867-6.

12

You might also like