ArticlePDF Available

Heterosubtypic protective immunity against influenza A virus induced by fusion peptide of the hemagglutinin in comparison to ectodomain of M2 protein

Authors:

Abstract and Figures

Several types of influenza vaccines are available, but due to the highly unpredictable variability of influenza virus surface antigens (hemagglutinin (HA) and neuraminidase) current vaccines are not sufficiently effective against broad spectrum of the influenza viruses. An innovative approach to extend the vaccine efficacy is based on the selection of conserved influenza proteins with a potential to induce inter-subtype protection against the influenza A viruses. A promising new candidate for the preparation of broadly protective vaccine may be a highly conserved N-terminal part of HA2 glycopolypeptide (HA2 gp) called fusion peptide. To study its capacity to induce a protective immune response, we immunized mice with the fusion peptide (aa 1-38 of HA2 gp). The protective ability of fusion peptide was compared with the ectodomain aa 2-23 of M2 protein (eM2) that is antigenically conserved and its immunogenic properties have already been well documented. Corresponding peptides (both derived from A/Mississippi/1/85 (H3N2) virus) were synthesized and conjugated to the keyhole limpet hemocyanin (KLH) and used for the immunization of mice. Both antigens induced a significant level of specific antibodies. Immunized mice were challenged with the lethal dose of homologous (H3N2) or heterologous A/PR/8/34 (H1N1) influenza A viruses. Immunization with the fusion peptide led to the 100% survival of mice infected with 1 LD50 of homologous as well as heterologous virus. Survival rate decreased when infectious dose was raised to 2 LD50. The immunization with eM2 induced effective cross-protection of mice infected even with 3 LD50 of both challenge viruses. The lower, but still effective protection induced by the fusion peptide of HA2 gp suggested that besides ectodomain of M2, fusion peptide could also be considered as a part of cross-protective influenza vaccine. To our knowledge, this is the first report demonstrating that active immunization with the conjugated fusion peptide of HA2 gp provided the effective production of antibodies, what contributed to the cross-protection against influenza infection.
Content may be subject to copyright.
Acta virologica 55: 261 – 265, 2011 doi:10.4149/av_2011_03_261
Evaluation of anti-inuenza eciency of polyclonal IgG antibodies specic to
the ectodomain of M2 protein of inuenza A virus by passive
immunizationof mice
J. KIRÁLY, E. VAREČKOVÁ, V. MUCHA, F. KOSTOLANSKÝ*
Department of Orthomyxoviruses, Institute of Virology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava,
Slovak Republic
Received May 24, 2011; accepted June 27, 2011
Summary. – We attempted to quantify the protective potential of polyclonal IgG antibodies specic to the
ectodomain of M2 protein (eM2) of inuenza A virus (IAV) against lethal inuenza infection of mice. For
this purpose, eM2 conjugated with keyhole limpet hemocyanin (KLH) or KLH alone were administered with
Freund’s adjuvant intraperitoneally (i.p.) to BALB/c mice. IgG antibodies specic to the KLH-eM2 conjugate
(anti-KLH-eM2 IgGs) and KLH (anti-KLH IgGs), respectively, were puried from ascitic uids. Analysis of the
preparation of anti-KLH-eM2 IgGs by ELISA revealed that it contained about 25% of anti-eM2 IgGs and 75%
of anti-KLH IgGs. Taking into account this nding mice were passively immunized by intravenous route with
320, 160, 80, and 40 µg of anti-eM2 IgGs per mouse, respectively, while 320 µg of anti-KLH IgGs were used
in control. Following subsequent infection with 3 LD50 IAV the survival of mice was determined. An absolute
protection (100% survival) was obtained with 320 µg of anti-eM2 IgGs, and a relatively strong signicant
protection (~80% survival, p = 0.024) with 160 µg. e amount 160 µg of IgGs represents approx. 100 µg IgGs
per 1 ml of blood.
Keywords: inuenza; M2 protein; eM2-specic IgG concentration; protection
*Corresponding author. E-mail: viruos@savba.sk; fax: +421-2-
54114284.
Abbreviations: IAV = inuenza A virus; eM2 = ectodomain of M2
protein; HA = hemagglutinin; anti-KLH-eM2 IgGs = IgG antibodies
to KLH-eM2; anti-KLH IgGs = IgG antibodies to KLH; i.n. = in-
tranasally; i.p. = intraperitoneally; i.v. = intravenously; KLH = key-
hole limpet hemocyanin; MAb(s) = monoclonal antibody(ies);
NA = neuraminidase
Introduction
ere are current eorts to avoid every year vaccination
against inuenza with a vaccine containing seasonally actual-
ized hemaglutinin (HA) and neuraminidase (NA) antigens
by use of a vaccine based on a conserved antigen(s). Such
a vaccine should evoke a long-lasting immune response
eciently suppressing the infection with various antigenic
variants or even subtypes. e main candidate molecule is
the M2 protein of IAV, particularly its 23 aa-long ectodomain
(eM2) that is characteristic by an outstanding antigenic con-
servativity and is abundantly expressed in the membrane of
infected cells (Neirynck et al., 1999; Palese and Garcia-Sastre,
2002; Lamb, 1985; Zebedee and Lamb, 1988). Nevertheless,
its immunogenicity during the natural inuenza infection is
very weak and short-term (Feng et al., 2006). ese incon-
venient properties of eM2 represent the main issues to be
solved. A number of various approaches for preparation of
eM2-based vaccine focused on increasing its immunogenicity
have been published (Slepuskin et al., 1995; Neirynck et al.,
1999; Wynne et al., 1999; Okuda et al., 2001; Liu et al., 2004;
Ben-Yedidia and Arnon, 2005; Huleatt et al., 2008; DeFilette
et al., 2008). e mechanism of the eM2-associated biological
action was described as the antibody-dependent cell-mediated
cytotoxicity (ADCC), in which NK cells with their low-anity
Fc gamma receptors bind IgG antibodies already bound to
the M2 protein expressed on the surface of infected cells and
mediate their lysis (Jagerlehner et al., 2004). is hypothesis
explains why anti-eM2 IgGs, when present in suciently high
262 SHORT COMMUNICATIONS
concentration, only attenuate the infection while neutralizing
antibodies to HA that directly inhibit the binding of virus to
the cell receptor provide a very eective instant protection.
Despite the lower eciency of eM2-specic antibodies, the
idea of utilization of eM2 in future inuenza vaccine remains
actual and promising as this molecule has a potential to elicit
a a broad cross-protective response (Sui et al., 2010; Stanekova
et al., 2011). In addition to the eM2 immunogenicity, the con-
centration of eM2-specic antibodies remains an important
characteristic to be followed that can also serve as an indicator
of eectivity of particular eM2-based vaccine preparation.
In this study, we atttempted to quantify the protective po-
tential of polyclonal IgG antibodies specic to eM2 IAV against
lethal inuenza infection of mice by determining the depend-
ence of survival of infected mice on actual concentration of these
antbodies in their blood following passive immunization.
Materials and Methods
Virus. A/Mississippi/1/85 (H3N2) was propagated in allantoic
uid of 10-day chicken embryos and stored at -70°C.
Mice. Six-week-old female BALB/c mice purchased from the
Faculty of Medicine, Masaryk University, Brno, Czech Republic,
were used. e animals were treated according to the European
Union standards and fundamental ethical principles including
animal welfare requirements.
eM2 peptide and KLH-eM2 conjugate. A 23-aa-long synthetic
eM2 peptide of IAV (H3 subtype) of the sequence SLLTEVET-
PIRNEWGSRSNDSSD, Mr of 2,592.74 and 93.94% purity was
purchased from ProImmune (USA). e peptide contained substi-
tutions C17S and C19S to avoid formation of disulphide bonds in
the peptide. e conjugation of eM2 with KLH (Sigma) was done
using glutaraldehyde as described by Staneková et al. (2011).
Immunization of mice. BALB/c mice were immunized i.p. with
three doses of KLH-eM2 (30 µg of eM2 per mouse) or KLH (30 µg
pre mouse), respectively, supplemented with Freund’s adjuvant, in
14 day intervals Staneková et al. (2011).
Polyclonal antibodies were puried from ascitic uids by anity
chromatography on Protein A-Sepharose columns (Ey et al., 1978).
Passive immunization of mice. BALB/c mice – 5 animals per
group – were administered i.v. anti-eM2 IgGs in 200 µl doses of 320,
160, 80, and 40 µg per mouse, respectively, while control mice ob-
tained 320 µg of anti-KLH IgGs and 200 µl of PBS, respectively.
Infection of mice. Two hrs aer passive immunization the mice
were intranasally (i.n.) inoculated with 3 LD50 of A/Mississippi/1/85
(H3N2) in 40 μl. Survival of mice was recorded daily for 14 days.
Statistical signicance of survival was evaluated by Fisher exact
test.
ELISA. eM2- or KLH-specic antibodies were assayed by a bind-
ing test according to Varečková et al. (2003a). Wells of 96-well plates
were coated overnight with 100 ng of eM2 or KLH as antigens in
100 μl at 4°C. e antibody titer was calculated as the reciprocal
of sample dilution at the point where the regression line drawn
through the titration curve crossed the cut-o line. e latter value
was the mean from 5 negative control samples plus 3 SD.
Results and Discussion
Antibody response in mice to immunization with eM2
In this work, we used a simple model of the eM2-KLH
conjugate as immunogen supplemented with the Fre-
Fig. 1
Antibody response in mice to immunization with KLH-eM2
Mice were i.p. immunized with 3 doses of KLH-eM2 and their sera were assayed for eM2-specic IgG antibodies by ELISA.
SHORT COMMUNICATIONS 263
und’s adjuvant to induce an antibody response in mice.
Control mice were given KLH alone. Following three im-
munizations the majority of mice developed ascites due to
administration of the adjuvant. e antibody response to
eM2 gradually increased, corresponding to serum titers of
528, 10,800, and 28,800, respectively (Fig. 1). Ascitic uids
obtained aer the last immunization served for purication
of polyclonal IgG antibodies.
Proportions of eM2- and KLH-specic antibodies in IgGs
puried from ascitic uid from mice immunized with KLH-
eM2
Proportions of IgG antibodies specic to eM2 and KLH,
present in IgGs puried from mice immunized with KLH-
eM2, were assayed by ELISA (Fig. 2). e distance of titra-
tion curves at A492 of 1.5 was estimated at 1.49 log2 units and
corresponding titers of anti-eM2 and anti-KLH IgGs were
1120 and 3149, respectively. is means that provided equal
numbers of accessible eM2 epitopes specic to anti-eM2 IgGs
and KLH epitopes specic to anti-KLH IgGs adsorbed onto
respective wells, the ratio of anti-eM2 and anti-KLH IgGs
was approximately 1:3.
Eective anti-eM2 IgG concentration required for the
protection to inuenza infection
Taking into account the anti-eM2 IgGs content of the
anti-KLH-eM2 IgGs puricate, groups of mice were given
i.v. 320, 160, 80, and 40 µg of anti-eM2 IgGs per animal.
Control mice were administered 320 µg of anti-KLH IgGs
and 200 µl of PBS, respectively. Two hrs later the mice were
infected with 3 LD50 IAV and observed for survival. An ab-
solute protection (100% survival) was obtained with 320 µg,
a relatively strong signicant protection (~80% survival) with
160 µg (p = 0.024), and a weak protection (~20% survival)
with 80 and 40 µg of anti-eM2 IgGs (Fig. 3). Control mice
scored a 100% mortality.
Estimating total blood volume in mouse at 1.5 ml, the
applied doses 320 µg and 160 µg of anti-eM2 IgGs resulting
in signicant protection corresponded to the concentra-
tions of 213 and 107 µg/ml anti-eM2 IgGs, respectively, in
the blood.
ese results roughly agree with those of Fu et al. (2009),
who also evaluated the anti-eM2 protective response, how-
ever, by use of MAbs. ey found that two of four tested
MAbs at doses of 0.2–2.0 mg per mouse ensured a high sur-
vival, while 20 µg resulted in a low survival. Another group
of authors (Beerli et al., 2009) found that a most eective
anti-eM2 MAb exhibited a fair protection against infection
with 4 LD50 of virus at a dose of 20 µg per mouse, but only
a weak one at a dose of 6 µg. Such a high eciency of this
MAb as compared with our observations as well as those
of Fu et al. (2009) can be most probably ascribed to a high
anity (Kd = 4 nmol/l) of that particular MAb.
In conclusion, we assume that results of this study con-
tribute to a recently accepted presumption that the eM2
molecule can be an eective and cross-protective immuno-
gen. Its immunogenicity can be enhanced by applying it in
appropriate form and/or with an optimally selected adjuvant.
Its cross-reactivity can be ensured mainly by polyclonal
character of the resulting antibody response. Moreover, the
Fig. 2
Proportions of eM2- and KLH-specic antibodies in IgGs puried from ascitic uid of mice immunized with KLH-eM2
Titration curves of anti-eM2 and anti-KLH IgGs (a) and titers of anti-eM2 and anti-KLH IgGs at A492 of 1.5 (b) in ELISA.
264 SHORT COMMUNICATIONS
latter may contribute to the prevention of the appearance
of antibody-escape mutants during natural infection (Zha-
rikova et al., 2005).
Acknowledgements. e authors thank Mmes M. Némethová and
M. Mišovičová for excellent technical assistance. is work was
supported by the VEGA grants Nos. 2/0101/10 and 2/0154/09 from
the Scientic Grant Agency of Ministry of Education of Slovak Re-
public and Slovak Academy of Sciences and the Slovak Research and
Development Agency under the contract No. APVV-0250-10.
References
Beerli RR, Bauer M, Schmitz N, Buser RB, Gwerder M, Muntwiler
S, Renner WA, Saudan P, Bachmann MF (2009): Prophy-
lactic and therapeutic activity of fully human monoclonal
antibodies directed against inuenza A M2 protein. Virol.
J. 6, 224. doi.org/10.1186/1743–422X–6–224
Ben-Yedidia T, Arnon R (2006): Flagella as a Platform for Epitope–
Based Vaccines. Isr. Med. Assoc. J. 8, 316–318 8
De Filette M, Martens W, Roose K, Deroo T, Vervalle F, Bentahir
M, Vandekerckhove J, Fiers W, Saelens X (2008): An
inuenza A vaccine based on tetrameric ectodomain of
matrix protein 2. J. Biol. Chem. 283, 11382–11387. doi.
org/10.1074/jbc.M800650200
Ey PL, Prowse SJ, Jenkin CR (1978): Isolation of pure IgG1, IgG2a
and IgG2b immunoglobulins from mouse serum using
Protein A-Sepharose. Immunochemistry 15, 429–436.
doi.org/10.1016/0161–5890(78)90070–6
Feng J, Zhang M, Mozdzanowska K, Zharikova D, Ho H, Wun-
ner W, Couch RB, Gerhard W (2006): Inuanza A virus
infection engenders a poor antibody response against
the ectodomain of matrix protein 2. Virol. J. 3, 102. doi.
org/10.1186/1743–422X–3–102
Fu TM, Freed DC, Horton MS, Fan J, Citron MP, Joyce JG, Garsky
VM, Casimiro DR, Zhao Q, Shiver JW, Liang X (2009):
Characterizations of four monoclonal antibodies against
M2 protein ectodomain of inuenza A virus. Virology
385, 218–226. doi.org/10.1016/j.virol.2008.11.035
Huleatt JW, Nakaar V, Desai P, Huang Y, Hewitt D, Jacobs A, Tang
J, Mcdonald W, Song L, Evans RK, Umlauf S, Tussey L,
Powell TJ (2008): Potent immunogenicity and ecacy
of a universal inuenza vaccine candidate comprising
a recombinant fusion protein linking influenza M2e
to the TLR5 ligand agellin. Vaccine 26, 201–214. doi.
org/10.1016/j.vaccine.2007.10.062
Jegerlehner A, Schmitz N, Storni T, Bachmann MF (2004): Inu-
enza A vaccine based on the extracellular domain of M2:
weak protection mediated via antibodydependent NK cell
activity. J. Immunol. 172, 5598–5605.
Lamb RA, Zebedee SL, Richardson CD (1985): Inuenza virus
M2 protein is an integral membrane protein expressed
on the infected-cell surface. Cell 40, 627–633. doi.
org/10.1016/0092–8674(85)90211–9
Liu W, Peng Z, Liu Z, Lu Y, Ding J, Chen YH (2004): High epitope
density in a single recombinant protein molecule of the
extracellular domain of inuenza A virus M2 protein
signicantly enhances protective immunity. Vaccine 23,
366–371. doi.org/10.1016/j.vaccine.2004.05.028
Neirynck S, Deroo T, Saelens X, Vanlandschoot P, Jou WM, Fiers
W (1999): A universal inuenza A vaccine based on the
extracellular domain of the M2 protein. Nat. Med. 5,
1157–1163. doi.org/10.1038/13484
Okuda K, Ihata A, Watabe S, Okada E, Yamakawa T, Hamajima K,
Yang J, Ishii N, Nakazawa M, Okuda K, Ohnari K, Naka-
jima K, Xin KQ (2001): Protective immunity against inu-
Fig. 3
Survival of mice passively immunized with puried anti-KLH-eM2 IgGs and infected with IAV
In immunization, the puried anti-KLH-eM2 IgGs were applied in amounts corresponding to 320, 160, 80, and 40 µg of anti-eM2 IgGs. Control mice
obtained 320 μg of puried anti-KLH IgGs and PBS, respectively.
SHORT COMMUNICATIONS 265
enza A virus induced by immunization with DNA plasmid
containing inuenza M gene. Vaccine 19, 3681–3691. doi.
org/10.1016/S0264–410X(01)00078–0
Palese P, Garcia-Sastre A (2002): Inuenza vaccines: present and
future. J. Clin. Invest. 110, 9–13.
Slepushkin VA, Katz JM, Black RA, Gamble WC, Rota PA, Cox
NJ (1995): Protection of mice against inuenza A virus
challenge by vaccination with baculovirusexpressed M2
protein. Vaccine 13, 1399–1402. doi.org/10.1016/0264–
410X(95)92777–Y
Staneková Z, Király J, Stropkovská A, Mikušková T, Mucha V,
Kostolanský F, Varečková E (2011): Heterosubtypic
protective immunity against inuenza A virus induced
by fusion peptide of the hemagglutinin in comparison
to ectodomain of M2 protein. Acta Virol. 55, 61–67. doi.
org/10.4149/av-2011-01-61
Sui Z, Chen Q, Wu R, Zhang H, Zheng M, Wang H, Chen Z (2010):
Cross-protection against inuenza virus infection by
intranasal administration of M2-based vaccine with
chitosan as an adjuvant. Arch. Virol. 155, 535–544. doi.
org/10.1007/s00705–010–0621–4
Varečková E, Mucha V, Wharton SA, Kostolanský F (2003a): Inhibi-
tion of fusion activity of inuenza A hemagglutinin medi-
ated by HA2–specic monoclonal antibodies. Arch. Virol.
148, 469–486. doi.org/10.1007/s00705–002–0932–1
Wynne SA, Crowther RA, Leslie AG (1999): e crystal structure of
the human hepatitis B virus capsid. Mol. Cell. 3, 771–780.
doi.org/10.1016/S1097–2765(01)80009–5
Zebedee SL, Lamb RA (1988): Inuenza A virus M2 protein: mono-
clonal antibody restriction of virus growth and detection
of M2 in virions. J. Virol. 62, 2762–2772.
Zharikova D, Mozdzanowska K, Feng J, Zhang M, Gerhard W
(2005): Inuenza type A virus escape mutants emerge in
vivo in the presence of antibodies to the ectodomain of
matrix protein 2. J. Virol. 79, 6644–6654.
... The first attempts to enhance the immunogenicity of HA2 gp utilized the carriers of different origins. As carries were frequently used Keyhole limpet hemocyanin (KLH) Staneková et al., 2011;Janulíková et al., 2012;, flagellin from the Salmonella typhimurium vaccine strain (Arnon, 2006;Ben-Yedida and Arnon, 2007;Stepanova et al., 2018), nanoparticles (Kanekiyo et al., 2013;Yassine et al., 2015), or virus-like particles (VLP) (Kang et al., 2012;Chen et al., 2015). These vectors enable to present antigen in many copies, resulting in increased robustness of specific immune response. ...
... A simple comparison of in vivo immunization with KLH-conjugated HA2 fusion peptide 1-38 and M2e was performed (Staneková et al., 2011). Repeatedly immunized mice were challenged with the lethal dose of homologous A/Mississippi/1/85(H3N2) or heterologous A/PR/8/34(H1N1) influenza A viruses. ...
... Even though the protection induced by the HA2 fusion peptide was lower, it was still effective. Results of this study suggested that apart from the ectodomain of M2, HA2 fusion peptide could also be considered as a part of cross-protective influenza vaccine (Staneková et al., 2011). ...
Article
Full-text available
Aquatic birds are the main reservoir of influenza A viruses (IAVs). These viruses can infect humans repeatedly and cause acute respiratory disease with potential of spread in the form of epidemics. In addition, avian influenza viruses that overcome the interspecies barrier and adapt to humans can cause a world-wide pandemic with severe consequences to human health. Therefore, scientists are focused on the development of a “universal” vaccine with a broad protective efficacy, i.e. against different subtypes of influenza A viruses and not only against the currently co-circulating human epidemic strains. Nowadays, several new vaccine design strategies have been described. Most of them utilize the conserved stem part of influenza surface glycoprotein hemagglutinin (HA) or the ectodomain of M2 (M2e) protein with proton-channel activity. A comparison of the efficacy of novel vaccines and their protective mechanisms against influenza infection is discussed in this review and should be considered for the construction of the most effective broadly protective vaccine with minimal side effects. This is the essential goal in influenza virus research today, especially when the infection with new human coronavirus SARS-CoV-2 can interfere with the course of influenza virus infection.
... The analysis of paired acute and convalescent sera of human patients with confirmed influenza infection showed that HA2 antigenic sites II and IV, localized in the region aa 125-175, are the most immunogenic. These two antigenic sites, together with antigenic site I could be important for the hetero-protective immunity induced during the influenza infection of humans, as antibodies recognizing these sites inhibited the fusion activity of HA as well as the replication of virus (Varečková et al., 2003b;Stropkovská et al., 2009;Staneková et al., 2011Staneková et al., , 2013Janulíková et al., 2012). ...
... Other approach utilized non-infectious non-replicating Escherichia coli-derived plasmids as DNA vaccines (Katz et al., 2006), or detoxified bacterial toxin from Bordetella pertussis (Staneková et al., 2013). KLH was used as the carrier for different peptides of HA2, for example aa sequence of highly conserved long α-helix recognized by cross-reactive Ab 12D1 (Wang et al., 2010), aa sequence of fusion peptide (Staneková et al., 2011) or the ectodomain of HA2 (Janulíková et al., 2012). Immunization of mice with two or three immunization doses of mentioned immunogens induced significant antibody response with cross-protective potential, resulting in improved survival and morbidity of mice challenged with homologous or heterologous virus. ...
... However, due to the conserved character of HA2 gp, the subsequent infection by several antigenically different IAVs, or immunization by selected epitopes of HA2 led to a stronger HA2-specific antibody response (Kostolanský, 2002;Gocník et al., 2008;Sui et al. 2009). Significantly increased level of anti-HA2 antibodies positively contributes to the efficient protection against lethal infection with homologous or heterologous virus in mice (Gocník et al., 2008;Staneková et al., 2011Staneková et al., , 2013Janulíková et al., 2012). The presence of these antibodies influences the infection, which becomes milder and the recovery from the disease is faster. ...
Article
Full-text available
Influenza A viruses (IAVs) cause yearly repeating infections in humans. The current vaccination approach is based on the production of virus-neutralizing antibodies. Virus-neutralizing antibodies, however, are closely strain-specific due to the IAV variability. Therefore, antibodies produced during the previous influenza season do not provide sufficient protection against new infection, and, hence, annual revaccination is needed. The utilization of the influenza conserved stem domain of hemagglutinin (HA), the HA2 gp, led to a new vaccine design based on cross-reactive cellular and especially humoral immune responses represented by HA2-specific antibodies. The HA2-specific antibodies exhibit cross-reactivity with HA2 gp within one subtype or even among subtypes and play a role in protective immunity against influenza infection. There are several elimination mechanisms of viral replication mediated by HA2-specific antibodies. After recognition of the epitope, they prevent the conformational rearrangement of HA or the insertion of the fusion protein into the endosomal membrane and, consequently, the fusion pore formation. In this case, no release of viral genetic information into the target cell is enabled and virus cannot replicate. The HA2-specific antibodies are involved in the elimination of pathogen via the Fc fragment by activation of the cytotoxic mechanisms of innate immunity as are the antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent phagocytosis (ADP), or complement-dependent cytotoxicity (CDC), resulting in virus elimination and earlier recovery of the host from the infection. Though the protective effect of HA2-specific antibodies on the course of IAV infection was shown, few cases of worsening of IAV infection mediated by HA2-specific antibodies have been described. The identification of antigenic epitopes on HA2 gp that induce antibodies with such deteriorating effect on influenza infection can help to eliminate the unsuitable epitopes of HA2 gp as immunogens during the design of heteroprotective vaccine against influenza and can remove the side effects linked with the observations mentioned above. Keywords: influenza A virus; HA2 stem domain of hemagglutinin; immunization strategies; HA2-specific antibodies.
... M2e-based peptide vaccines were shown to provide heterogenetic immunity against IAV in mice, but were not as effective in swine models [10,14,15]. Similarly, the fusion peptide located in the HA2 protein is highly conserved among the different influenza virus strains [13,16,17] and provided broad protection in mice, but has not been tested in swine [18,19]. A third epitope, ATGLRNIPSIQSRLE, located in the HA1 region, was selected for its protective potential against H1 subtype viruses [20]. ...
... Several approaches such as linking epitopes with immunogenic peptides or proteins, using multiple copies of epitopes, creating virus-like particles or using amphiphilic polymers [3][4][5][6]8,18,25] have been previously used to improve the weak immunogenicity of peptide antigens [16,44]. Amphiphilic polymers have several advantages; they can form micelles and micellar assemblies that can load antigen in a controllable manner, can be used for controlled antigen release and are generally immunologically safe [57]. ...
Article
Influenza A viruses (IAVs) are a group of genetically diverse and economically important zoonotic pathogens. Despite decades of research, effective and broadly protective vaccines are yet to be developed. Recent breakthroughs in epitope-based immunization for influenza viruses identify certain conserved regions of the HA2 and M2e proteins as capable of inducing broad protection against multiple influenza strains. The M2e and HA2 peptides have been evaluated in mice but not as a combination in pigs, which play an important role in the transmission and evolution of IAV. Peptides are inherently weak immunogens; and effective delivery of peptide antigens is challenging. To enhance the delivery and immunogenicity of peptide-based vaccines, the conserved M2e and HA2 and a strain-specific HA1 epitope of Influenza A (H1N1) pdm09 were expressed as a chain in a bacterial expression system and entrapped in a novel amphiphilic invertible polymer made from polyethyelene glycol (PEG, molecular weight 600 g/mol) and polytetrahydrofuran (PTHF, molecular weight 650 g/mol), PEG600PTHF650. Piglets vaccinated with polymeric peptide vaccine mounted significantly stronger antibody responses against the peptide construct when compared to piglets immunized with the multi-epitope peptide alone. When vaccinated pigs were challenged with Influenza A (H1N1) pdm09, viral shedding in nasal secretions and lung lesion scores were significantly reduced when compared to the unvaccinated controls and pigs vaccinated with the peptide alone at six days post-challenge. Thus, the combination of the PEG600PTHF650 polymer and trimeric peptide construct enhanced delivery of the peptide antigen, acted as an adjuvant in stimulating strong antibody responses, reduced the effects of viral infection in vaccinated pigs.
... The subtype A(H1N1) provided the highest rate of variation in both countries. The negative Pearson's coefficients of correlation between the A(H3N2) and the A(H1N1) suggest a dynamic influence between both subtypes, indicator of their recognized competitive interference during the sequence epidemics, related to an existing heterosubtypic immunity that reduces the probability of reinfections [34]. ...
... The proposed dynamic and the lack of correlation of the type B with both A subtypes and the differences in the frequencies of the local extremes in the simple linear chart of these viruses show the different rhythms of the antigenic changes and the predominance of one or another type/subtype. However, little is known about the genomic scale evolutionary dynamics of the pathogen and its relation with these changes [4,[34][35][36]. ...
... However, the stability of different peptides varies in different species; the same peptide which works in mice may not always work the same in primates [11,82]. So proper experimentation and improvisation of peptide-based therapeutics allowed the development of many peptide-based drugs that are in use for the treatment of various neurodegenerative diseases [83], influenza [84], various types of cancer [85], diabetes [86], etc. Moreover, immune therapeutics, mostly antibody-drug conjugates (ADCs), has already been established clinically for cancer therapeutics [87][88][89][90][91]. Peptide drug conjugates (PDCs) have wildly succeeded in oncology. ...
Article
Aims Malaria deaths occur primarily due to complicated malaria associated with the sequestration of Plasmodium falciparum-infected erythrocyte (PfIE) in the capillary microvasculature. This study aims to design peptide ligand conjugates (PLCs) for treating complicated malaria using various in silico techniques. The PLC includes a natural ligand for the Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1): expressed explicitly on the surface of PfIE, and a highly immunogenic peptide derived from the commonly used peptide vaccines in malaria-endemic countries. The ligand is predicted to prevent the sequestration of PfIE, and the peptide is predicted to eliminate PfIE from circulation by the pre-existing vaccine-induced immunity. Main methods The epitope identification from the vaccines and the analysis of physicochemical properties, antigenicity, allergenicity, and toxicity were performed using SVMTriP, ProtParam, VaxiJen, AllerTop, and ToxinPred servers, respectively. The high throughput virtual screening (HTVS) and drug-like properties analysis of natural compound ligands were carried out by Schrodinger-2021 software. The molecular dynamics simulations were performed through the WebGro server. Key findings HTVS revealed three bioactive natural ligands for PfEMP1 from (NPASS) database. Three super immunogenic peptides were identified from malaria-endemic countries' commonly used peptide vaccines. Finally, Nine PLCs were designed with different combinations of peptides and ligands with the suitable non-cleavable triazole linker. Significance Antimalarials have been losing efficacy in a time when malaria deaths in 2020 significantly increased than in 2019. In this scenario, further research on the designed PLCs may offer some innovative immune therapeutics for complicated malaria with minimum possibilities of drug resistance.
... Promising results for peptide vaccine development have been obtained with the following viruses: Influenza virus [56], hepatitis B virus [57], respiratory syncytial virus [58,59], bovine leukemia virus [60,61], feline immunodeficiency virus [62,63] and hepatitis C virus [64]. Lately, many predictions of peptide vaccine candidates for RNA viruses have been made due to progress in bioinformatics, for e.g.: corona virus [65][66][67][68], rotavirus [69][70][71], foot-and-mouth disease [72,73], Hepatitis C [74][75][76], influenza [77,78]. ...
Article
Full-text available
RNA viruses have high rate of replication and mutation that help them adapt and change according to their environmental conditions. Many viral mutants are the cause of various severe and lethal diseases. Vaccines, on the other hand have the capacity to protect us from infectious diseases by eliciting antibody or cell-mediated immune responses that are pathogen-specific. While there are a few reviews pertaining to the use of artificial intelligence (AI) for SARS-COV-2 vaccine development, none focus on peptide vaccination for RNA viruses and the important role played by AI in it. Peptide vaccine which is slowly coming to be recognized as a safe and effective vaccination strategy has the capacity to overcome the mutant escape problem which is also being currently faced by SARS-COV-2 vaccines in circulation.Here we review the present scenario of peptide vaccines which are developed using mathematical and computational statistics methods to prevent the spread of disease caused by RNA viruses. We also focus on the importance and current stage of AI and mathematical evolutionary modeling using machine learning tools in the establishment of these new peptide vaccines for the control of viral disease.
... On the contrary, because of the small size peptide vaccine is weakly immunogenic and require a carrier molecule for adjuvanting and chemical stability [19]. Several peptide vaccines are under process of development for different viral diseases, such as hepatitis C virus (HCV) [23], human papilloma virus (HPV) [24], human immunodeficiency virus (HIV) [25], and influenza [26] etc. A multi-epitope vaccine contains a series of overlapping epitopes that can elicit effective cytotoxic T cells, T helper cells and B cells responses against viral pathogen [27]. ...
Article
Full-text available
Middle East respiratory syndrome coronavirus (MERS-CoV) is an emerging health concern due to its high mortality rate of 35%. At present, no vaccine is available to protect against MERS-CoV infections. Therefore, an in silico search for potential antigenic epitopes in the non-redundant proteome of MERS-CoV was performed herein. First, a subtractive proteome-based approach was employed to look for the surface exposed and host non-homologous proteins. Following, immunoinformatics analysis was performed to predict antigenic B and T cell epitopes that were used in the design of a multi-epitopes peptide. Molecular docking study was carried out to predict vaccine construct affinity of binding to Toll-like receptor 3 (TLR3) and understand its binding conformation to extract ideas about its processing by the host immune system. We identified membrane protein, envelope small membrane protein, non-structural protein ORF3, non-structural protein ORF5, and spike glycoprotein as potential candidates for subunit vaccine designing. The designed multi-epitope peptide then linked to β-defensin adjuvant is showing high antigenicity. Further, the sequence of the designed vaccine construct is optimized for maximum expression in the Escherichia coli expression system. A rich pattern of hydrogen and hydrophobic interactions of the construct was observed with the TLR3 allowing stable binding of the construct at the docked site as predicted by the molecular dynamics simulation and MM-PBSA binding energies. We expect that the panel of subunit vaccine candidates and the designed vaccine construct could be highly effective in immunizing populations from infections caused by MERS-CoV and could possible applied on the current pandemic COVID-19.
... Multi-epitope vaccine was introduced in 1985 for the first time to develop a vaccine against cholera and E. coli heat-labile toxin [75]. Recently, many multi-epitope vaccines have been suggested as a new effective approach against some infectious diseases or cancers, i.e. human immunodeficiency virus (HIV), hepatitis C virus (HCV), human papilloma virus (HPV), Neisseria meningitidis, malaria, tuberculosis, swine fever, influenza, foot and mouth disease, anthrax, and melanoma [54,[57][58][59][60][61][62][63][64][65][66][67][68][69][70][71][72][73][74]. Identification of T-cell epitopes in a desired protein had been the key point in this kind of studies [75][76][77][78]. ...
Article
Presentation of many unwanted epitopes within tetanus toxoid vaccine to lymphocyte clones may lead to production of many unwanted antibodies. Moreover an ideal vaccine must cover all individuals in a population that is dependent to the kinds of human leukocyte antigen alleles. Concerning these issues, our study was aimed to in silico design of a multi-epitope tetanus vaccine (METV) in order to improve population coverage and protectivity of tetanus vaccine as well as reduction of complications. Concerning these issues, a novel rational filtration was implemented to design a novel METV using immunoinformatics and surface epitope mapping approaches. Prediction of epitopes for tetanus toxin was performed in the candidate country in which the frequency had been gathered from almost all geographical distributions. The most strong binder epitopes for major histocompatibility complex class II were selected and among them the surface epitopes of native toxin were selected. The population coverage of the selected epitopes was estimated. The final candidate epitopes had highly population coverage. Molecular docking was performed to prediction of binding affinity of our candidate epitopes to the HLA-DRB1 alleles. At first, 680 strong binder epitopes were predicted. Among them 11 epitopes were selected. Finally, 4 epitopes had the most population coverage and suggested as a tetra-epitope tetanus vaccine. 99.41% of inessential strong binders were deleted using our tree steps filtration. HLA-DP had the most roles in epitope presentation. Molecular docking analysis proved the strong binding affinity of candidate epitopes to the HLA-DRB1 alleles. In conclusion, we theoretically reduced 99.41% of unwanted antibodies using our novel filtration strategies. Our tetra-epitope tetanus vaccine showed 100% population coverage in the candidate country. Furthermore, it was demonstrated that HLA-DP and HLA-DQ had more potential in epitope presentation in comparison to HLA-DRB1.
... Therefore epitopes/peptide based vaccines can be an alternative strategy for an efficient vaccine production, to induce positive humoral and cell mediated immune responses (Sesardic, 1993;Li et al., 2014). There are many peptide based vaccines which are under development to combat several disease, such for anthrax (Oscherwitz et al., 2010), hepatitis C virus (HCV) (Kolesanova et al., 2013), malaria (Epstein et al., 2007)], foot and mouth disease (FMD) (Volpina et al., 1993), swine fever (Tarradas et al., 2011), influenza (Stanekova et al., 2011), human immunodeficiency virus (HIV) (Liu et al., 2007), human papilloma virus (HPV) (Solares et al., 2011). ...
Thesis
Recombinant adenoviruses (Ad) have recently been employed for a wide range of vaccination strategies. Unfortunately, highly prevalent pre-existing neutralizing antibodies (Abs), reduce their ability to trigger transgene expression. To avoid the step of gene transfer a new vaccination strategy has been proposed based on the use of Ad displaying epitopes inserted into their capsid proteins. Using an ovalbumin-derived B cell epitope, our group demonstrated that vaccination efficiency depends on both the site of peptide insertion and the host immune status towards Ad (Lanzi et al; 2011). The present work aims at (1) evaluating the potency of Ad displaying T-cell epitopes from ovalbumin to elicit cellular responses and (2) understanding the molecular bases controlling the efficacy of this vaccination strategy. 1) Ad displaying T-cell epitopes from ovalbumin were constructed, produced and characterized in vitro. First in vivo experiments in naive mice showed induction of cellular responses, assessed with techniques like ELISPOT, tetramer staining and in vitro splenocyte restimulation. Subsequent experiments showed that pre-exisitng anti-vector immunity is hampering the potent induction of anti-epitope cellular responses. Current work is aiming at confirming the obtained results as well as at evaluating the kinetics of cellular responses induced upon "epitope display" vaccination. 2)First, the influence of interactions of Ad (displaying OVA peptide) with their natural receptors was investigated. Different detargeted Ads were produced and characterized in vitro. Upon mice immunization these vectors led to unmodified anti-epitope humoral responses, suggesting that their efficacy does not depend on the ability to transduce cells. In parallel we sought to evaluate the impact of innate immunity on the outcome of anti-epitope adaptive immune responses. Upon immunization of WT and MyD88-/- mice with Ad displaying OVA epitope we observed that cellular responses induced in MyD88-/- mice are significantly diminished while humoral responses were not altered. These results remain to be confirmed but question the role of other innate immunity sensors in the immunogenicity of Ad-based vaccines. Altogether, our work is expected to provide the foundations for the development of Ad-based vaccines with minimized side effects and unaltered adjuvant properties.
Article
Full-text available
Influenza virus infection is a prevalent disease in humans. Antibodies against hemagglutinin have been shown to prevent infection and hence hemagglutinin is the major constituent of current vaccines. Antibodies directed against the highly conserved extracellular domain of M2 have also been shown to mediate protection against Influenza A infection in various animal models. Active vaccination is generally considered the best approach to combat viral diseases. However, passive immunization is an attractive alternative, particularly in acutely exposed or immune compromized individuals, young children and the elderly. We recently described a novel method for the rapid isolation of natural human antibodies by mammalian cell display. Here we used this approach to isolate human monoclonal antibodies directed against the highly conserved extracellular domain of the Influenza A M2 protein. The identified antibodies bound M2 peptide with high affinities, recognized native cell-surface expressed M2 and protected mice from a lethal influenza virus challenge. Moreover, therapeutic treatment up to 2 days after infection was effective, suggesting that M2-specific monoclonals have a great potential as immunotherapeutic agents against Influenza infection.
Article
Full-text available
Fusion of the influenza virus envelope with the endosomal membrane of host cells is mediated by the hemagglutinin glycoprotein (HA). The most conserved region of HA is at the N-terminus of the HA2 subunit, a relatively hydrophobic sequence of amino acids referred to as the fusion peptide. This domain is critical both for setting the trigger for fusion and for destabilizing target membranes during the fusion process. The "trigger" is set by cleavage of the HA precursor polypeptide, when the newly-generated HA2 N-terminal fusion peptide positions itself into the trimer interior and makes contacts with ionizable residues to generate a fusion competent neutral pH structure. This essentially "primes" the HA such that subsequent acidification of the endosomal environment can induce the irreversible conformational changes that result in membrane fusion. A key component of these acid-induced structural rearrangements involves the extrusion of the fusion peptide from its buried position and its relocation to interact with the target membrane. The role of the fusion peptide for both priming the neutral pH structure and interacting with cellular membranes during the fusion process is discussed.
Article
Full-text available
Since its identification in April 2009, an A(H1N1) virus containing a unique combination of gene segments from both North American and Eurasian swine lineages has continued to circulate in humans. The lack of similarity between the 2009 A(H1N1) virus and its nearest relatives indicates that its gene segments have been circulating undetected for an extended period. Its low genetic diversity suggests that the introduction into humans was a single event or multiple events of similar viruses. Molecular markers predictive of adaptation to humans are not currently present in 2009 A(H1N1) viruses, suggesting that previously unrecognized molecular determinants could be responsible for the transmission among humans. Antigenically the viruses are homogeneous and similar to North American swine A(H1N1) viruses but distinct from seasonal human A(H1N1).
Article
Full-text available
The reactivity of monoclonal antibodies (MAbs) prepared to the HA2 glycopeptide (gp) of A/Dunedin/4/73 (H3N2) hemagglutinin was tested against influenza A viruses of H3, H4, and H7 subtypes. Only one (CF2) out of six MAbs reacted with influenza A viruses of all three subtypes (H3, H4 and H7). The inter-subtype reactivity of this MAb (CF2) is in accord with the highly conservative sequence in the previously defined MAb-binding site I, i.e. the aa 1-38 of N-terminus of HA2 gp. MAb CF2 as well as inter-subtype cross-reactive MAb IIF4, recognizing the binding site II of HA2 gp, were tested for their effect on replication of influenza A viruses. Both these MAbs reduced the number of plaques of viruses of homologous (H3) as well as heterologous (H4) virus subtypes, the latter less efficiently. The potential of these MAbs to influence in vivo replication of influenza A viruses of various subtypes is discussed.
Article
Full-text available
Influenza virus remains a serious health threat, owing to its ability to evade immune surveillance through rapid genetic drift and reassortment. Here we used a human non-immune antibody phage-display library and the H5 hemagglutinin ectodomain to select ten neutralizing antibodies (nAbs) that were effective against all group 1 influenza viruses tested, including H5N1 'bird flu' and the H1N1 'Spanish flu'. The crystal structure of one such nAb bound to H5 shows that it blocks infection by inserting its heavy chain into a conserved pocket in the stem region, thus preventing membrane fusion. Nine of the nAbs employ the germline gene VH1-69, and all seem to use the same neutralizing mechanism. Our data further suggest that this region is recalcitrant to neutralization escape and that nAb-based immunotherapy is a promising strategy for broad-spectrum protection against seasonal and pandemic influenza viruses.
Article
Full-text available
Multiorgan spread and pathogenesis of influenza infection with three human influenza A viruses was studied in mice. Mouse-adapted viruses A/Dunedin/4/73(H3N2), A/Mississippi/1/85(H3N2), and A/PR/8/34(H1N1) differed considerably in virulence (p.f.u./LD(50)): 79,000 p.f.u. for Dunedin, 5,000 p.f.u. for Mississippi, and 65 p.f.u. for PR/8, which qualified Dunedin as low virulent, Mississippi as intermediate, and PR/8 as highly virulent. All three viruses were detected in lungs, heart, and thymus by cultivation and RT-PCR. Moreover, vRNA of all viruses was found in liver and spleen, of Dunedin and PR/8 also in kidneys and that of Dunedin and Mississippi in blood. Only vRNA of Dunedin was demonstrated in brain. Lung damage accompanied by histopathological changes and thymus reduction were most extensive after infection with the highly virulent virus PR/8. We assume that the ability to spread to multiple organs may be a more common property of influenza viruses in mammalian hosts than previously believed.
Article
The fusion peptide of influenza viral hemagglutinin plays a critical role in virus entry by facilitating membrane fusion between the virus and target cells. As the fusion peptide is the only universally conserved epitope in all influenza A and B viruses, it could be an attractive target for vaccine-induced immune responses. We previously reported that antibodies targeting the first 14 amino acids of the N-terminus of the fusion peptide could bind to virtually all influenza virus strains and quantify hemagglutinins in vaccines produced in embryonated eggs. Here we demonstrate that these universal antibodies bind to the viral hemagglutinins in native conformation presented in infected mammalian cell cultures and neutralize multiple subtypes of virus by inhibiting the pH-dependant fusion of viral and cellular membranes. These results suggest that this unique, highly-conserved linear sequence in viral hemagglutinin is exposed sufficiently to be attacked by the antibodies during the course of infection and merits further investigation because of potential importance in the protection against diverse strains of influenza viruses.
Article
Influenza vaccines based on conserved antigens could provide cross-protection against infections by multiple subtypes of influenza A virus. Influenza matrix protein 2 (M2) is highly conserved in all influenza A strains. In this study, we deleted the transmembrane domain of the M2 of the avian influenza virus (AIV) A/Chicken/Jiangsu/7/2002 (H9N2) strain to create an M2 without a transmembrane domain, named sM2, which was efficiently expressed in Escherichia coli. The sM2 protein was administered intranasally to mice in combination with chitosan adjuvant three times at an interval of 3 weeks. Three weeks after the last immunization, the mice were challenged with a lethal dose (5 x LD(50)) of A/Chicken/Jiangsu/7/2002 (H9N2) virus, PR8 (H1N1) virus and A/Chicken/Henan/12/2004 (H5N1) virus. The protective immunity of the vaccine was evaluated by determining the survival rates, residual lung virus titers, body weight, and the serum antibody titers of the mice. Nasal administration of 15 microg sM2 in combination with chitosan completely protected mice against the homologous virus and protected 90 and 30% of the mice against the heterologous H1N1 and H5N1 viruses, respectively. The study indicated that the sM2 protein was a candidate antigen for a broad-spectrum influenza virus vaccine and that the adjuvant chitosan improved the efficacy of the sM2 vaccine.
Article
Human influenza causes substantial morbidity and mortality. Currently, licensed influenza vaccines offer satisfactory protection if they match the infecting strain, but they come with significant drawbacks. These vaccines are derived from prototype viruses, containing the hemagglutinin of influenza viruses that are likely to cause the next epidemic. Their usefulness against a future pandemic, however, remains problematic. A vaccine based on the ectodomain of influenza matrix protein 2 (M2e) could overcome these drawbacks. M2e is highly conserved in both human and avian influenza A viruses. The low immunogenicity against natural M2e can be overcome by fusing M2e to an appropriate carrier such as Hepatitis B virus-derived virus-like particles. Such chimeric particles can be produced in a simple and safe bacterial expression system, requiring minimal biocontainment, and can be obtained in a pure form. Experiments in animal models have demonstrated that M2e-based vaccines induce protection against a lethal challenge with various influenza A virus subtypes. Furthermore, the production and use of an effective M2e-vaccine could be implemented at any time regardless of seasonality, both in an epidemic as well as in a pandemic preparedness program. In animal models, M2e-vaccines administered parenterally or intranasally protect against disease and mortality following challenge with various influenza A strains. Adjuvants suitable for human use improve protection, which correlates with higher anti-M2e antibody responses of defined subtypes. Recently, Phase I clinical studies with M2e-vaccines have been completed, indicating their safety and immunogenicity. Further clinical development of this universal influenza A vaccine candidate is being pursued in order to validate its protective efficacy in humans.