G Model

ARTICLE IN PRESS

JVAC-16280; No. of Pages 11

Vaccine xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Vaccine journal homepage: www.elsevier.com/locate/vaccine

Review

Vaccines against influenza A viruses in poultry and swine: Status and future developments J. Rahn, D. Hoffmann, T.C. Harder, M. Beer ∗ Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany

a r t i c l e

i n f o

Article history: Received 17 December 2014 Received in revised form 1 March 2015 Accepted 18 March 2015 Available online xxx Keywords: Influenza virus vaccines Swine Chicken Novel developments

a b s t r a c t Influenza A viruses are important pathogens with a very broad host spectrum including domestic poultry and swine. For preventing clinical disease and controlling the spread, vaccination is one of the most efficient tools. Classical influenza vaccines for domestic poultry and swine are conventional inactivated preparations. However, a very broad range of novel vaccine types ranging from (i) nucleic acid-based vaccines, (ii) replicon particles, (iii) subunits and virus-like particles, (iv) vectored vaccines, or (v) liveattenuated vaccines has been described, and some of them are now also used in the field. The different novel approaches for vaccines against avian and swine influenza virus infections are reviewed, and additional features like universal vaccines, novel application approaches and the “differentiating infected from vaccinated animals” (DIVA)-strategy are summarized. © 2015 Elsevier Ltd. All rights reserved.

1. Introduction Avian influenza (AI) in poultry and swine influenza (SI) in pigs are economically important diseases, which are caused by influenza A viruses (IAV) [1–3]. Certain phenotypes of IAV subtypes H5 and H7, termed highly pathogenic (HP) AI viruses, cause devastating mortality in gallinaceous poultry, as well as affect water fowl in a more strain dependent manner [4]. Furthermore, AI viruses (AIV) of low pathogenicity (LP) may cause economically tangible losses in gallinaceous poultry due to a drop in egg production in layer flocks and reduced daily weight gains in fattening poultry. In addition, substantial mortality in galliforme poultry may be noticed also in association with LP AIV infection in concert with bacterial or viral co-infections [5]. Influenza virus infections of swine are characterized by an acute febrile respiratory disease of usually short duration that often is complicated by opportunistic bacterial infections and then may lead to reduction in daily weight gains of fattening pigs; also, fertility disorders such as fever-induced abortion in sows have been associated with SI virus infections (reviewed by [6]). Influenza virus infected poultry and swine are also of pivotal importance for public health as they may play a role as sources of parental influenza virus from which, by reassortment and adaptation processes, yield virus strains with zoonotic and even pandemic potential that by spill-over transmission, may spread to human populations [7–9].

∗ Corresponding author. Tel.: +49 38351 7 1200; fax: +49 38351 7 1275. E-mail address: martin.beer@fli.bund.de (M. Beer).

Education of stakeholders, strict biosecurity regimes combined with rapid diagnosis and surveillance programs and rigorous eradication measures (“stamping out”) comprise the recommended primary line of defense against AI in especially poultry industrial holdings [10–13]. Such regimes, however, require comprehensive investments in stable facilities and farm management, veterinary administration and laboratory capacities, and are out of scope for production units in developing countries, smallholders and backyard rearing communities [14–16]. Preventive vaccination against AI in poultry is generally considered as a secondary line of defense and its pro’s and con’s should be weighted with respect to the specific epidemiologic situation including, in particular, (i) extent and impact of AI related disease, (ii) incursions into areas with a high density of poultry populations, (iii) establishment of endemic infections, and (iv) increased risks of spill-over infections of zoonotic IAV to humans [17–21]. In case of outbreaks of notifiable AI, i.e., caused by subtype H5 or H7 viruses, further caution is recommended and the use of vaccination might require explicit legal permissions by the competent authorities [22]. Protection against clinical disease caused by SI and its economical impact are of primary interest in case of vaccination against SI; SI viruses, some of them with zoonotic potential, are endemic to the worlds swine population and there are no mandatory eradication programs (at least in Europe). Vaccination against AI in poultry and SI in pigs follows two primary intentions: (i) Successful vaccination reduces clinical signs of disease upon infection and, hence, prevents economic losses, (ii) vaccinated animals excrete significantly less virus following infection with field virus whereby reducing risks of further spread of

http://dx.doi.org/10.1016/j.vaccine.2015.03.052 0264-410X/© 2015 Elsevier Ltd. All rights reserved.

Please cite this article in press as: Rahn J, et al. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.03.052

G Model JVAC-16280; No. of Pages 11

ARTICLE IN PRESS J. Rahn et al. / Vaccine xxx (2015) xxx–xxx

2

field virus. A reduction of the amount of circulating virus is of particular interest if outbreaks are caused by zoonotic influenza viruses that may cause spill-over infection in humans, e.g., in the course of the on-going hemiglobal epizootic of Asian origin H5N1 HPAI virus or the emerging H7N9 virus in China [18].

[13]. Reaching every individual animal by needle and syringe for prime-and-boost vaccination rounds becomes practically near impossible [13]. The available licensed vaccines and approved vaccine strategies provide no universal solution to these problems and intensive research, as reviewed here, is required to provide new solutions to vaccination against IAV infections in pigs and poultry.

1.1. Classical vaccines: inactivated influenza virus Historically, influenza vaccination is based on culture-grown IAVs. Usually, embryonated chicken eggs from specific pathogen free (SPF) flocks are used for vaccine virus propagation, mainly because of their unsurpassed yield of virus compared to cell culture systems. For use in pigs or poultry, culture-derived virions in crude allantoic fluids are chemically inactivated and then formulated into mineral oil emulsion vaccines whereby avoiding, in contrast to vaccine formulations for human use, sophisticated and costly purification steps for the enrichment of the immunodominant envelope glycoproteins hemagglutinin (HA) and neuraminidase (NA) [22]. Depending on the host species, primary and booster vaccinations by the subcutaneous or intramuscular routes are required to induce protective levels of systemic hemagglutinationinhibiting (HI) antibodies, and at least annual re-vaccinations are required to ensure that such antibody levels are maintained [23]. Success in prevention largely depends on a maximized antigenic match between the virus strain used for vaccine preparation and the virus(es) circulating in the field [13]. Epidemiological data supported by model calculations indicated that achieving and maintaining a broad and resilient herd immunity is another prerequisite for successful influenza vaccination in pigs and poultry [24]. Vaccine-induced immunity that imprecisely fits the antigenic make-up of the circulating viruses and patchy herd immunity are major drivers of antigenic drift and silent virus spread [25]. Viral antigenic drift by continuing accidental point mutations in the hemagglutinin gene and consecutive selection of mutants that escape vaccine-induced immunity will lead to the emergence of virus escape mutants that gradually replace older circulating virus strains (see e.g., [26,27] and references therein). In such case, spread of circulating field viruses under the cover of a vaccination campaign, i.e., “silent spread”, ensues: clinically the animals may still be protected to an extent that incursions of virulent virus into these flocks would easily be missed by syndrome surveillance [28]. This situation prompts an update of the vaccine to re-attain efficacy by using the escape mutant strain for production [29]. Escape mutants have repeatedly emerged at different geographic localizations (China, Indonesia, Vietnam, Egypt) in the course of the HPAI H5N1 epizootic [26,27] and there is evidence for antigenic drift also in SIV [2,30,31]. By the use of reverse genetics the antigenic makeup of vaccine virus strains is adjustable to circulating viruses within a relative short time period: recombinant viruses with the HA and NA antigenic determinants specifically replaced have been successfully applied against HPAI H5N1 [13]. Furthermore the use of inactivated influenza vaccines and serology techniques based on detection of antibodies directed against NS1 protein permits the establishment of the “differentiating infected from vaccinated animals” concept (DIVA) (see Section 2.8) and are therefore an option for eradication programs [32]. The ideal vaccine, in an infectological sense, should induce sterilizing immunity that blocks infection with circulating field viruses completely after a single immunization. Protection should be generated against changing field strains, i.e., a broader cross protection, at best even across different subtypes, is needed. Maternally derived immunity should be circumvented to enable protection as early as possible in life. High numbers of animals in industrial settings, sequestration of backyard poultry in difficult-to-reach locations and rapid turnover rates of swine and poultry populations are mounting further practical problems of vaccine delivery

2. Novel approaches in influenza virus vaccination in swine and chickens 2.1. Nucleic acid based vaccines Nucleic acid-based vaccines combine the advantages of (i) a molecularly defined antigen, (ii) the induction of both humoral and cellular immune response associated with MHC class I and class II molecules, (iii) a fast and cheap development without the need for embryonated eggs and even without handling the potentially hazardous pathogen when synthetic nucleic acids are used, as well as (iv) the reduced need of an adjuvant, as the nuclide acid in it itself represents a potent target for the immune system. 2.1.1. Messenger RNA-based vaccines The proof-of-principle of mRNA vaccines has first been described in the mouse model 20 years ago with an mRNA of the influenza virus nucleoprotein (NP) [33], and due to its simple and safe production and its tailored immunogenicity, this strategy has been further improved in the recent years. These vaccines typically comprise of a simple vector carrying the information for the antigen of interest, as well as 5 and 3 untranslated regions (UTR), the CAP structure, and the poly-A-tail for an efficient translation and the stability of the vector within the eukaryotic cell [34]. Optimizing these cis-acting structures is one goal of the recent improvements of the mRNA vectors [35–39]. The mRNA vaccines are produced by an enzymatic in vitro transcription from the tailored DNA template downstream of a suitable promoter, followed by purification steps. Thus, only sequence information of the nucleic acid of the antigen of interest is needed [34]. This process can be easily adapted to new antigens in emergency scenarios, allowing cost-effective manufacturing processes in classical clean-room settings. Recent improvements have been made regarding the enzymatic reactions [40–42], purification [43], delivering [37,44] and complexing with stabilizing proteins such as protamine [45]. In contrast to DNA, exogenous RNA only has to cross the cell membrane to facilitate protein expression in the cytoplasm and several cell types tend to take up mRNAs by receptor-mediated endocytosis [46–49]. The physical delivery, improved uptake, increased protein expression and induction of the immune system are also under development [45,50–53]. In the host, the RNA is recognized by pattern recognition receptors (PRRs) like toll-like receptors (TLRs), retinoic acid inducible gene I (RIG-I), or protein kinase R (PKR) [54–56], leading to the activation of CD8+ T cells (by delivering the antigen to the MHC-I processing pathway) and B cells directly or via the induction of interferon expression [49,57,58]. Dendritic cells (DCs) represent the best target as mRNA-mediated antigen expression in these cells or in secondary lymphoid tissue induces cellular immunity [36,59], but it was suggested that also other cell types, that take up the mRNA after intramuscular or intradermal application, are able to transfer it to DCs and other immune cells and induce the immunity via cross-priming [48,60–62]. In addition, antigen expression activates the signaling pathway for B cells [63]. As most of the studies are done in the mouse model, further studies are needed to adapt these principles to pigs and/or poultry species. Recently, Petsch et al. showed that vaccination with the mRNAs of HA, NA, and NP of H1N1, H3N2, and H5N1 viruses, which have previously been optimized regarding GC content and the UTR sequence composition,

Please cite this article in press as: Rahn J, et al. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.03.052

G Model JVAC-16280; No. of Pages 11

ARTICLE IN PRESS J. Rahn et al. / Vaccine xxx (2015) xxx–xxx

induced immunological correlates of protection in pigs as well as in mice and ferrets, and protected them against clinical signs and reduced viral shedding in challenge experiments [37]. 2.1.2. DNA-based vaccines DNA-based vaccines are based on bacterial plasmids that are constructed to express an encoded protein, e.g. an antigen, and are used for the transfection of cells, resulting in the induction of the cellular and humoral immune response [64]. The first description of a DNA vaccine was made by Wolff et al. in the mouse model [60], and since then improvements of this technique have been made, especially regarding the stability, expression, delivery, and immunogenicity [65–71]. Numerous studies showed the induction of a strong cytotoxic T lymphocyte (CTL) response with DNA vaccines expressing HA, NP, or M1 in various animal models [72–76]. A strong antibody response mediated by DNA vaccination of pigs followed by viral challenge [77] was observed in the study of Larsen et al., in which pigs were immunized with a priming dose of a HA DNA vaccine followed by a boost immunization with a conventional, inactivated whole-virus vaccine, resulting in an enhanced immune response and protection from challenge infection [78]. A further promising approach was made by Gorres et al., who designed a DNA vaccine comprising of trivalent or monovalent HA-genes in a backbone with the cytomegalovirus enhancer/promoter and the human T-cell leukemia virus type 1 R region [79]. The trivalent vaccine induced both humoral and IFN-␥ responses, protected pigs against viral shedding and lung disease after H1N1 challenge, and reduced viral shedding after H3N2 challenge, regardless of the application route (i.m. or needle-free s.c.). Macklin et al. used DNA-coated gold particles to deliver a HA-based DNA vaccine to the epidermis or the tongue of pigs with a gene gun and thereby induced an immune response that confers protection against homologous challenge [80]. In recent studies, Lim and colleagues examined the immunogenicity of different plasmid DNAs encoding the HA, NA and NP genes from an AIV combined with chicken IL-15 and IL-18 [81,82]. Prime vaccinated and boostered chickens (both i.m.) developed high antibody titers against N1 and an increase in CD8+ T cells or CD4+ T cells for the IL-15 or IL-18 adjuvanted groups, respectively, indicating that IL-15 enhances the immunogenicity of the DNA vaccine in chickens and, in accordance with other studies, showing that the NP alone is less immunogenic, but nevertheless induces cellmediated immune response leading to the survival of challenged chicken [83,84]. Jiang et al. reported DNA vaccines based on the codon-optimized HA-genes of an H5N1 or an H7N1 virus that after i.m. injection induced high antibody titers and a solid protection of chickens against lethal H5N1 or H7N1 challenge and of quails against lethal H5N1 challenge, respectively [85–87]. A significant advantage of DNA vaccines is their ability to encode multiple genes of interest [88], but in contrast to mRNA vaccines, are poorly transported into target cells [77,89,90]. Therefore, different modes of application have been studied (see Section 2.7). Nevertheless, big concerns for the use of DNA vaccines are the danger of genomic integration into the host cell and the presence of selective markers, such as antibiotic resistance genes [91]. 2.2. RNA replicon particle-based vaccines RNA replicon vaccines are ((+)ss, (−)ss or ds) RNA viruses from which at least one essential structural protein has been deleted and that possess an engineered virus genome to express foreign antigens [92,93]. They undergo autonomous replication and transcription of the genome, thereby stimulating both humoral and cellular immune response, but are unable to produce infectious progeny. Replicon particles are mainly based on alphaviruses, such as the Venezuelan equine encephalitis virus (VEEV), that are

3

produced in mammalian cell lines after transfection of a set of in vitro transcribed RNAs (replicon RNA with viral non-structural proteins and heterologous antigen plus helper RNA that encode the viral structural proteins) [94]. Others have used the vesicular stomatitis virus (VSV) that lacks the envelope glycoprotein G (VSVG) [95,96]. An alphavirus replicon RNA for the expression of the HA from swine influenza viruses or the pH1N1 has been developed containing the alphavirus ORF1 (with deleted non-structural proteins rendering the replicon replication-defective) and the heterologous genes in ORF2 [93,97,98]. The corresponding genes responsible for replication of the viral RNA have to be provided in trans together with the replicon (during electroporation). The HA protein was expressed in cells and after combination with an adjuvant induced high HA-specific antibody titers after vaccination of pigs, resulting in reduced lung lesions and viral shedding after homologous challenge. The tropism of the constructs for DCs promotes a fast immune response. Also an attenuated variant of the VEEV was used as a replicon vaccine expressing the HA-gene of a human H3N2 virus or the pH1N1 strain and induced high HI antibody titers in vaccinated pigs, and also protected them from homologous challenge infection [99,100]. The use of an alphavirus replicon system for the expression of the HA and NP genes of a pH1N1 and a H3N2 virus, respectively, eliminated viral shedding and reduced pulmonary damage in vaccinated and pH1N1-challenged pigs [101]. Avian species are naturally not infected by VEEV and VSV making these viruses perfect vectors for replicon-based vaccines [92]. In a study by Schultz-Cherry et al., immunization with a VEEV expressing the HA of an H5N1 virus protected chickens against a lethal challenge with the homologous virus to differing extends, depending on the age of the bird [102], whereas Sylte et al. used a similar approach for the expression of the NA of an H5N2 or H3N2 virus, that could only mediate a partial protection in vaccinated and challenged chickens [103]. Kalharo et al. used the VSVG approach to express the HA or NP of an HPAIV H7N1 strain, that was able to protect vaccinated chickens against a challenge with a heterologous H7N1 strain [104]. More recently, Halbherr et al. used a VSVG replicon expressing the HA of an H5N1 virus [105]. Chickens receiving one vaccination were completely protected from challenge and chickens boostered with the same vaccine were even protected from viral shedding. After establishment of the vaccine vector, the development of a new and multivalent vaccine is possible within a short time period. Replicon-based vaccines are safe, as they are not spread to the environment and cannot integrate into the host genome [92,100]. Furthermore, the host immunity of birds to the alphavirus is described to be reduced, allowing several replicon-based vaccinations of the same animal [88]. 2.3. Synthetic peptides, subunit vaccines, and virus-like particles 2.3.1. Synthetic peptides The potential of synthetic peptides as a vaccine candidate has recently been shown by Ma et al., who designed a tetra-branched multiple antigenic peptide based vaccine constructed by fusing four copies of M2e to one copy of a foreign T-helper cell epitope, which was able to induce a strong immune response and protected mice from otherwise lethal challenge [106]. Several studies reported synthetic peptide vaccines that, when administered to mice induced antibodies directed against the HA-stalk, which have a broad neutralizing activity due to the conserved sequence among different influenza virus subtypes [107–109]. Vergara-Alert et al. developed a broadly protective influenza vaccine based on a theoretical prediction of highly conserved peptide sequences from either H5 or H1 viruses [110]. After (i.m.) immunization with the synthetic peptides pigs developed humoral responses and the

Please cite this article in press as: Rahn J, et al. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.03.052

G Model JVAC-16280; No. of Pages 11 4

ARTICLE IN PRESS J. Rahn et al. / Vaccine xxx (2015) xxx–xxx

induced antibodies were able to recognize heterologous influenza viruses in vitro (pH1N1, SwH1N1, SwH3N2, H5N1 HPAIV). However, more research has to be done to evaluate the potential of these vaccines to elicit broader immune responses in pigs and poultry species than conventional vaccines.

against a lethal challenge with the heterologous strain [131]. Thus, these approaches may also be considered as vaccine candidates for swine and poultry [88].

2.3.2. Subunit vaccines A subunit vaccine is an immunizing agent that contains viral proteins, but no viral nucleic acid and that therefore induces a focused immune response [111]. It comprises of at least one recombinant Influenza virus protein, which in most cases is the HA, as it induces hemagglutination inhibiting antibodies [112,113]. Liu et al. used a baculovirus system for the expression of the HA of an H5N1 HPAIV and were able to induce a strong antibody response and fully protected chicken (and mice) from lethal challenge [114]. Crawford et al. designed baculoviruses, that express the HA of an H5 or an H7 virus [115]. These purified proteins completely protected (s.c.) vaccinated SPF-chickens against challenge with the homologous virus and abolished or reduced viral shedding. Wu et al. used a baculovirus pseudotyped with VSV glycoprotein as a vector to express the hemagglutinin (HA) protein of an H5N1 HPAIV that offered complete protection of chickens from homologous challenge [116]. Sylte et al. found that the recombinant baculovirus-expressed N2 induced high levels of NA inhibiting activity and protected 88% of the vaccinated chicken from challenge with a H5N2 virus with the homologous NA [103]. Kalthoff et al. developed a viral vector for the expression of a fulllength HA in plants, which was able to reduce viral shedding and to protect chickens from otherwise lethal challenge with a heterologous H5N1 virus [117]. In other studies, an ‘immuno-stimulating complex’ (ISCOM) obtained from n-octyl-beta-d-glucopyranosidetreated H5N2 viruses was used to induce high antibody titers and a cellular immune response in turkeys, and were able to protect them from both homologous and heterologous challenge infection as shown by reduced viral titers in the lung and trachea [118,119].

Vector vaccines represent modified live-attenuated viruses obtained with reverse genetics technology, that contain genomic material that can be expressed in the cell after entry via infection by the vector used, leading to endogenous antigen processing and MHC class I restricted presentation [88]. Thus, the vector has to have a wide host range and the host species should not have preexisting antibodies against the vector. Several vectors such as paramyxoviruses, herpesviruses, adenoviruses, or poxviruses have been studied for the use in poultry and swine. The Human adenovirus 5 with a deleted transcription region, rendering the vector replication-defective, was used to express the HA and NP genes of an H3N2 virus, and was shown to protect (i.m.) vaccinated pigs from viral shedding and the development of lung lesions after challenge with the homologous virus [136,137]. This vaccine candidate can only be used as a prime vaccination, as pigs develop antibodies against the adenovirus vector, and these antibodies could reduce the boost reaction [138]. The pseudorabies virus (PrV) represents a further potential vector, as several non-essential genes of the virus can be replaced by other genes, leading to a reduced virulence of the vector virus [139]. Such a virus was used to express the HA of an H3N2 strain and tested successfully in the mouse model [140]. Klingbeil et al. inserted the codon-optimized HA of a pH1N1 virus into the PrV genome and immunized pigs (i.n.) with the respective vector vaccine [141]. The pigs developed HA-specific antibodies, showed only minimal respiratory symptoms and less nasal shedding after challenge infection with a related pH1N1 virus. As the host develops antibodies against the vector, multiple vaccinations of the same animal are also not possible and may in addition hamper PrV surveillance [142]. For AI, other vaccine types have been licensed as well. In general, these were built on viral vectors, which expressed at least the HA as the major target of a protective humoral immune response. In particular, fowl pox virus recombinant vaccines expressing the hemagglutinin gene of AIV subtypes H5 or H7, and, more recently, further vector vaccines based on Newcastle Disease virus, herpesvirus of turkey or duck enteritis virus have been licensed for use in the field. However, it has been estimated that these vaccines account for less than 5% of vaccine doses used for immunization of poultry in the field in Central America (rFPV), Southeast Asia and Egypt (rNDV) [13,143]. In a recent study, Said et al. employed an equine herpesvirus (EHV-1) as a vector for the expression of the HA-gene of a pH1N1 virus and demonstrated that the vaccine induces influenza virusspecific antibody responses in pigs and was able to protect at least partially against challenge infection as evidenced by decreased nasal virus shedding and faster virus clearance [144]. Furthermore, a Modified Vaccinia Ankara (MVA) virus was used to express the HA and NP genes of a classical H1N1 Swine influenza strain and vaccination of pigs (i.n. or i.m.) resulted in shorter duration and lower titers of viral shedding after homologous challenge as well as in the prevention of the lung lesion development [145]. As an addition poxvirus vector, the highly attenuated vaccinia virus NYVAC strain [146] was used for the expression of a H5N1 HPAIV HA to induce potent neutralizing antibody responses in swine, which were protected against viral replication after challenge infection with a low-pathogenic H5N2 strain [147]. In several studies, recombinant Newcastle Disease viruses (rNDV) were tested as a vaccine vector for HPAIV-challenge infection [148–156]. Swayne et al. described an NDV with an insertion of the HA-gene from an H7N2 strain that partially protected chicken

2.3.3. Virus-like particles The expression and self-assembling of viral structural proteins into virus-like particles (VLPs) represents another promising type of vaccine [120]. They resemble infectious virus particles in structure and morphology, induces a sufficient immune response but do not contain any viral genetic material and are therefore not infectious. Several systems for the production of influenza VLPs have been described, including baculovirus [121–125], transient plasmid expression [126–128], stable cell-line transformation [129] and expression in plants [130], but mostly mice or ferrets were used for vaccine studies. Chang et al. generated a baculovirus-based VLP which incorporated the H7 HA alone and showed that it was immunogenic in chickens [131], however, its protective efficacy has not been tested. Using also a baculovirus expression system, Lee et al. developed VLPs with HA and M1 from a H9N2 virus [132]. Chickens immunized with the adjuvanted vaccine (i.m.) elicited high levels of antibody responses and lessened viral shedding. In similar approaches, other groups generated VLPs comprising HA, NA and/or M1 from a H5N1 HPAIV and showed, that immunized chickens were protected against a lethal homologous challenge, showing no clinical signs of infection [133,134]. Pyo et al. generated VLPs consisting of HA, NA and M1 from pandemic H1N1/2009 virus that elicited robust levels of humoral and mucosal immune responses in immunized pigs which after challenge infection displayed reduced lung lesions and inhibition of virus replication in the lung [135]. A plant-based transient expression of the HA proteins of an H5N1 and an H1N1 virus revealed that HA was predominantly assembled into high-molecular-weight structures, triggered a strong immune response against the homologous virus in immunized mice and conferred complete protection

2.4. Vectored vaccines

Please cite this article in press as: Rahn J, et al. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.03.052

G Model JVAC-16280; No. of Pages 11

ARTICLE IN PRESS J. Rahn et al. / Vaccine xxx (2015) xxx–xxx

from homologous HPAIV challenge [153]. Veits et al. inserted the HA-gene from an H5N2 HPAIV and demonstrated that immunization of chickens with that recombinant vector induced NDV- and AIV H5-specific antibodies and protected chickens against clinical disease and shedding of AIV after challenge infection with a lethal dose of velogenic NDV or highly pathogenic AIV, respectively [157]. Nayak et al. generated recombinant rNDVs expressing the HA, NA, or M2 protein of an H5N1 HPAIV, and showed that the immunization of chickens with HA alone or in combination with NA induced a complete protection against HPAIV infection [155]. In the study of Lardois et al., it could be shown that an NDV expressing the HA of an H5N1 HPAIV induced an H5 serologic response and afforded a high degree of clinical protection against challenge infection with a related H5N1 virus, independent of the inoculation route (oculonasal versus drinking water), but dose-dependent [156]. In order to overcome pre-existing NDV antibodies within the host, Steglich et al., constructed a NDV vector which carries the fusion (F) and hemagglutinin-neuraminidase (HN) proteins of the avian paramyxovirus type 8 instead of the corresponding NDV proteins, and inserted a gene expressing the HA of an H5 HPAIV between the F and HN genes [158]. This chimeric virus induced full protection against lethal HPAIV-infection in chickens without as well as with maternally derived NDV-specific antibodies. Today, rNDVs are approved as bivalent vaccines in poultry against NDV and AIV, and are widely used in China [159,160]. The Gallid Herpesvirus (infectious laryngotracheitis virus (ILTV)) was used as a vector for the insertion of the HA-gene of an H5 or H7 virus at the non-essential UL50 gene locus [157,161]. Immunized chickens produced specific antibodies against ILTV and AIV HA, and were protected against challenge infections with either virulent ILTV, or two different highly pathogenic AIV strains, but developed minimal clinical signs. Therefore, Pavlova et al. designed an attenuated ILTV variant for the insertion of the H5 (or N1) gene, which proved to offer protection of chickens, when vaccinated with the H5-expressing vector or the combination of the H5 and N1 expressing ILTVs, without causing clinical signs [162]. A genetically engineered Marek’s Disease vaccine of serotype 3 (turkey Herpesvirus, HVT) expressing the H5-protein is commercialized in Egypt and a recent study in layer chicken breed flocks attested a considerable protection after a one shot immunization strategy in day-old-chicks [163]. Toro et al. have shown that a non-replicating human adenovirus vector encoding the HA of an H5N9 avian influenza virus induced a protective immunity against avian influenza virus in chickens by a single-dose in ovo vaccination [164]. In another study, Gao et al. demonstrated that an adenovirus-based vaccine expressing different portions of the HA of a H5N1 virus completely protected chickens after a single subcutaneous immunization from homologous challenge [165]. Wu et al. designed a pseudotyped baculovirus, carrying the G protein of VSV and the HA-gene of a H5N1 HPAIV [116]. Immunized chickens (and mice) elicited strong serum antibody responses and protected from H5N1 challenge. Finally, also the fowlpox virus provides a suitable candidate for the development of a species-specific recombinant viral vector as it has a natural host range limited to avian species. Already in the late 80s, Taylor et al. reported a recombinant fowlpox virus expressing the HA from a HPAIV, that provided protection of immunized chickens and turkeys against a lethal challenge with either the homologous or a heterologous influenza virus strain [166]. Since then, several recombinant fowlpox viruses expressing the HA of H5 viruses have been described [167–173] and successfully used in an HPAIV outbreak [174]. Currently, at least ten poxvirus-vectored vaccines have been licensed for veterinary use [175], including an MVA virus expressing the HA which is licensed as a vectored influenza vaccine in poultry and equines [147].

5

2.5. Live attenuated influenza vaccines In 2003 a live attenuated influenza vaccine (LAIV) was approved for humans by the US Food and Drug Administration (FDA). While since 2012 a quadrivalent LAIV is licensed by the FDA for immunization of humans, such vaccines did yet not enter the market of swine/poultry production. Several proof-of-principle-studies demonstrated extraordinary protection induced by LAIV in swine and poultry ([176–187]. Attenuation of the vaccine virus may be achieved by truncation of the Non-structural protein (NS1), which in turn attenuates the virus´ı ability to antagonize the effects of the host’s type I interferon antiviral response [176,181,182,184,188]) by genetic mutations within the HA leading to an altered protease dependency and impaired replication [177–180] or by engineering mutations into the polymerase genes, impairing polymerase activity and restricting virus replication at elevated temperatures [185]. Masic et al. generated an eight-segment SIV harboring two different SIV HA (H1 and H3) by fusing the H3 HA ectodomain to the cytoplasmic tail, transmembrane domain and stalk region of the NA [189]. The resulting LAIV was attenuated in pigs, and even after intranasal application conferred reduction of fever and other clinical signs as well as the development of gross lesions in the lungs after challenge infection with both H1 and H3 SIV [186]. The use of LAIV in poultry remains prohibited to date and their use is not recommended by relevant international organizations such as OIE and FAO since adaptation to gallinaceous birds and/or reassortment events may potentially lead to the generation of HPAI viruses. However, Röhrs and colleagues demonstrated exceptional protective propensity of a live neuraminidase deleted H5-virus, which induced full protection in chickens seven days after a single shot immunization [187]. The most efficient and beneficial applications for LAIV may be toward emerging vaccination campaigns rather than for routine mass vaccination. In addition Hai and colleagues attempted to generate a reassortment-incompetent virus by manipulating an influenza B virus to express different HA proteins originating from influenza A virus strains. Although the authors did not perform a vaccine/challenge experiment, they demonstrated seroconversion of chicken after intravenous inoculation [190]. 2.6. “Universal” vaccines The optimal universal vaccine should cover all relevant subtypes of influenza A viruses, however, it is more realistic to develop a vaccine that is more broadly cross-protective than the currently licensed vaccines, and that induces a longer lasting, sterile immunity. Most neutralizing antibodies are directed against the globular head of the HA and are, therefore, in most cases very strain specific. Additionally, both HA and NA glycoproteins are highly variable and are drifting under immune pressure [191]. Regions of the HA that carry out functions essential for infection and replication present sites of vulnerability for recognition by more cross-reactive and broadly neutralizing antibodies [192]. Thus, the receptor binding site on the HA1-subunit and the fusion machinery of the HA2subunit are prime targets for antibody intervention. Several groups described (human or murine) antibodies directed against the HA-stem that neutralize several group 1 viruses (H2, H5, H6, H9) and/or some group 2 viruses (H3, H7, H10), respectively, and proofed their therapeutic potential in mice [193–197]. Other groups identified antibodies, that neutralize both group 1 and group 2 viruses, and that are directed against a conserved epitope near the receptor binding site [198–200]. However, recent publications from swine experiments verified the contribution of non-neutralizing, but cross-reactive antibodies against the HA2 domain to induction of vaccine-associated enhanced

Please cite this article in press as: Rahn J, et al. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.03.052

G Model JVAC-16280; No. of Pages 11

ARTICLE IN PRESS J. Rahn et al. / Vaccine xxx (2015) xxx–xxx

6

respiratory disease after infection with a heterologous virus [201,202]. While this phenomenon is also a problem of whole virus inactivated vaccines (primarily used today within the swine industry) it impacts the idea of a universal vaccine negatively. Antibodies raised against regions with high amino acid conservation within the NA-protein were shown to be cross-reactive by Western Blot to representative NAs from nine subtypes [203], yet the in vivo efficacy has not been investigated. Unfortunately, literature lacks studies performed in more relevant host species than mice. In contrast to the HA, the internal viral proteins including NP, PA, M1 and M2 are highly conserved across the subtypes and induce cellular immune response, mainly CTL response against processed peptides [204], thus, represent promising targets for a universal vaccine. Boyd et al. developed recombinant MVA and Adenoviruses expressing a fusion construct of NP and M1 and, as a proof of concept, prime-boost vaccinated chickens showed an increased T-cell response, and cloacal shedding was at least reduced after challenge infection with a low pathogenic H7N7 virus [205]. As the extracellular domain of the M2-protein (M2e) is conserved among the influenza subtypes [206], this domain is thought to mediate heterosubtypic immunity, although the peptide itself is relatively low immunogenic [207]. Several studies analyzed different approaches using the M2e-epitope in mice, revealing its potential [125,207–210]. Kim et al. produced VLPs expressing tandem repeats of M2e peptides containing two human, two avian and one swine origin M2e peptide sequence fused with HA transmembrane and cytoplasmic domains [211,212]. Sera from immunized mice reacted with a range of Influenza A viruses, including H1N1, H3N2, and H5N1 viruses. Heinen et al. developed a DNA construct by the fusion of M2e to the Hepatitis B core protein or the influenza NP and vaccinated pigs with the resulting DNA vaccines [213]. Unfortunately, although vaccinated pigs developed antibodies against M2e, and even virus-specific lymphoproliferation in case of the M2e-NP vaccine, clinical signs and mortality were exacerbated after challenge infection, indicating that further research is needed in terms of using M2e as immunogenic protein. 2.7. Novel application approaches Needle-mediated injections (i.m., s.c., i.d.) are the most common application techniques, as upon pathogen contact these tissues rapidly deploy innate defenses to contain the infection and engender protective adaptive immunity [214,215]. Needlefree/non-invasive application is associated with less pain, an easier application, improved vaccine acceptance, reduced costs, reduced safety risks, and no need for trained personnel [216]. Intranasal administration of IAV vaccines has been attempted as an alternative method to protect pigs and induce local immune responses [217]. Furthermore, for DNA-based vaccines the delivery via “gene gun” to the skin or the tongue has been shown to enhance the transfection efficiency and thereby the induction of antibodies after immunization [77,78,218,219]. Chen et al. have shown that mice immunized via electroporation (i.m. administration followed by electrical stimulation) with a DNA-plasmid containing a consensus sequence of the HA of an H5N1 virus – optimized for an improved protein expression – elicited neutralizing antibodies against viruses from different H5N1 clades, and were completely or significantly protected against challenge viruses [220]. Also linear DNA-cassettes have been successfully tested in mice [221]. Finally, by the use of a conventional whole virus inactivated influenza vaccine (without adjuvant) applied as a dry powder aerosol vaccine at the syrinx of chickens, Peeters and co-workers obtained significantly reduced replication of the challenge virus [222]. The authors also tested vaccination by passive inhalation as

this method might be suitable for mass application, however by this technique only partial protection could be achieved [222]. 2.8. DIVA vaccines The acronym DIVA stands for ‘differentiating infected from vaccinated animals’. In order to fulfill the criteria of the DIVA-principle, a marker vaccine and its corresponding companion serological assay have to be available. While international organizations like OIE recommend DIVA vaccines for pigs against pseudorabies and classical swine fever [223], swine influenza vaccines are applied without any DIVA strategy until now. However, vaccines from any of the former presented types are described to fit in principal to the DIVA concept: A) For inactivated whole virus vaccine: several DIVA-vaccines based on a heterologous NA-concept were developed for avian influenza (reviewed in [32]). The ultimate heterologous NArecombinant virus was generated by Peeters et al. containing the (modified low-pathogenic) HA-gene of a contemporary H5N1 HPAIV strain in combination with the NA gene of a human type B influenza virus (which only occur in humans and not in birds), that, as inactivated vaccine, protected chickens against clinical disease and completely prevented virus shedding after H5N1 HPAIV challenge [224]. As inactivated virus preparations do not induce antibodies against non-structural proteins such vaccines and the sensitive detection of NS1 antibodies do meet the criteria of the DIVA principle. The advantage versus the heterologous NA-vaccine is that the NS1 DIVA is working irrespective of the antigenic subtype of vaccine strain and circulating virus strain. Although the general suitability was demonstrated for several avian species, vaccinated and infected animals maybe missed due to reduced seroconversion rates (reviewed in [32]). B) For nucleic acid-based vaccines, RNA replicon particle-based vaccines, synthetic peptides, subunit vaccines, virus-like particles and vector vaccines induce an antibody response only against the influenza proteins that are specifically encoded by the relevant vaccine, which is in most cases the HA alone or in combination with the NA protein. Therefore, sera from vaccinated animals consist of antibodies against the HA (and the NA), but not against the internal proteins like the nucleoprotein NP. NPELISAs can therefore be used as a possible accompanying marker assay. C) There are live attenuated influenza vaccines that meet the criteria of the DIVA concept. The NA-deleted H5-vaccine virus from Röhrs and co-workers [187], and the chimeric influenza B and influenza A construct from Hai and colleagues [190] both induce an antibody response, which is distinct from wildtype infection induced antibodies and NA-specific ELISA systems can be used as marker assays. Finally, these universal vaccine approaches that work with the expression of the M2-protein are likewise appropriate DIVA vaccines. However, while serological reactions against the M2 protein were demonstrated in general, more work is needed to show their suitability [32]. 3. Conclusions In conclusion, a whole range of novel influenza vaccine approaches is available for both poultry and swine. In order to overcome the obstacles associated with the conventional vaccines, several different inactivated and attenuated vaccine types are in the focus of influenza vaccine research. An overview of the most promising examples is given in Table 1.

Please cite this article in press as: Rahn J, et al. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.03.052

G Model JVAC-16280; No. of Pages 11

ARTICLE IN PRESS J. Rahn et al. / Vaccine xxx (2015) xxx–xxx

7

Table 1 Overview of vaccine types and their features. Vaccine type

Species

Main application routes

DIVA

Universal

Selected literature references

Inactivated homologous whole virus vaccines Inactivated heterologous whole virus vaccines Live attenuated vaccines mRNA vaccines DNA vaccines Replicon vaccines Synthetic peptides Subunit vaccines Virus-like particles Vectored vaccines

Swine, chicken Chicken Chicken, swine Swine Swine, chicken Swine, chicken Swine Chicken, swine Chicken, swine Swine, chicken

i.m.a i.m. i.m., i.n.b i.m., i.d.c i.m., i.d., s.c.d , n.f.e i.m, i.n., s.c. i.m., s.c. i.m., s.c. i.m., s.c. i.m., s.c., n.f., i.n., o.n.f

− + − + + + + + + +

− − − − − − + − + +

[172] [227] [180,187,190] [34] [64,228] [92,93,98] [106,110] [114,116] [125,131] [229]

a b c d e f

Intramuscularly. Intranasally. Intradermally. Subcutaneously. Needle free. Oronasally.

Modified live vaccines seem to be the most efficient vaccines with a very early onset of immunity and the potential of mucosal immunization, and cold-adapted MLV (achieved by inducing combined mutations in PB1 and PB2 genes) are registered for humans and horses [225,226]; but there are still safety issues including the risk of reassortment and reversion to virulence [179]. In addition, not all approaches allow a DIVA-strategy, and licensing of genetically modified organisms could be an additional hurdle. In contrast, the very safe subunit preparations enable different DIVA concepts, but still need two applications, have to be injected, and reports are mainly available from the immunization of chickens and turkeys. Therefore, vector-based vaccines seem to be an alternative combining the subunit idea with a replicating viral vector. However, vector-specific immunity is a major problem for the continuous use of the same vector and is difficult to overcome. Beside the classic approaches, promising results were also obtained from studies with mRNA-based vaccines, inducing a good protection level in several mammalian species including pigs, and such vaccines should therefore also be tested in poultry. Furthermore, these vaccines are considered to be safe, since, in contrast to DNA-based vaccines, integration into the host genome is not possible. Nevertheless, those vaccine types have to be injected and a needle-free application should be considered in future studies. One of the major future goals in influenza vaccine research is the development of a “universal” vaccine, providing protection against a broad range of influenza subtypes. Therefore, further research targeting the relevant livestock species is required. Finally, vaccine application by injection of individual animals is nowadays still obligatory for nearly all of the registered and most of the novel vaccine types. However, mass application is only feasible e.g. by mucosal immunization especially in industrial settings. Hence, research has to focus on mucosal/needle-free immunization of poultry and pigs also. These obstacles pose significant challenges of vaccine development for avian/swine influenza. The ongoing influenza A endemic/epidemic or even pandemic situations clearly indicate that further efforts are needed to control Influenza A virus infections in livestock animals and, to the authors´ı opinion, vaccines are a very promising tool. In summary, promising novel approaches have been reported in recent years, and there continues to be progress in the development of novel influenza virus vaccines for livestock animals. However, the “ideal influenza vaccine” for poultry and pigs is still missing. A safe and efficacious vaccine with a very broad reactivity against several subtypes, with DIVA-features and which allows mass application e.g. by oral immunization, would be the perfect tool in the future, and further efforts are necessary to reach this goal.

References [1] McLeod A, Guerne-Bleich E. Social, economic and policy issues in the longterm control of HPAI. Dev Biol (Basel) 2006;124:171–6. [2] Vincent AL, Ma W, Lager KM, Janke BH, Richt JA. Swine influenza viruses a North American perspective. Adv Virus Res 2008;72:127–54. [3] Mastin A, Alarcon P, Pfeiffer D, Wood J, Williamson S, Brown I, et al. Prevalence and risk factors for swine influenza virus infection in the English pig population. PLoS Curr 2011;3:RRN1209. [4] Pantin-Jackwood MJ, Swayne DE. Pathogenesis and pathobiology of avian influenza virus infection in birds. Rev Sci Tech 2009;28:113–36. [5] Alexander DJ. A review of avian influenza in different bird species. Vet Microbiol 2000;74:3–13. [6] Olsen CWBIH, Easterday BC, Van Reeth K. Swine influenza. In: Straw BEZJJ, dÁllaire S, Taylor DJ, editors. Disease of Swine. Oxford, UK: Blackwell Publishing; 2006. p. 469–82. [7] Kuiken T, Holmes EC, McCauley J, Rimmelzwaan GF, Williams CS, Grenfell BT. Host species barriers to influenza virus infections. Science 2006;312:394–7. [8] Van Reeth K. Avian and swine influenza viruses: our current understanding of the zoonotic risk. Vet Res 2007;38:243–60. [9] Ma W, Kahn RE, Richt JA. The pig as a mixing vessel for influenza viruses: human and veterinary implications. J Mol Genet Med 2008;3:158–66. [10] Werner OHTC. Control and eradication strategies for classical fowl plague in Germany and the European Union. Berl Munch Tierarztl Wochenschr 2006;9. [11] Bunn D, Beltran-Alcrudo D, Cardona C. Integrating surveillance and biosecurity activities to achieve efficiencies in national avian influenza programs. Prev Vet Med 2011;98:292–4. [12] Pavade G, Awada L, Hamilton K, Swayne DE. The influence of economic indicators, poultry density and the performance of veterinary services on the control of high-pathogenicity avian influenza in poultry. Rev Sci Tech 2011;30:661–71. [13] Swayne DE. Impact of vaccines and vaccination on global control of avian influenza. Avian Dis 2012;56:818–28. [14] Conan A, Goutard FL, Sorn S, Vong S. Biosecurity measures for backyard poultry in developing countries: a systematic review. BMC Vet Res 2012;8:240. [15] Fasina FO, Ali AM, Yilma JM, Thieme O, Ankers P. The cost-benefit of biosecurity measures on infectious diseases in the Egyptian household poultry. Prev Vet Med 2012;103:178–91. [16] Negro-Calduch E, Elfadaly S, Tibbo M, Ankers P, Bailey E. Assessment of biosecurity practices of small-scale broiler producers in central Egypt. Prev Vet Med 2013;110:253–62. [17] Marangon S, Cecchinato M, Capua I. Use of vaccination in avian influenza control and eradication. Zoonoses Public Health 2008;55:65–72. [18] Sims LD. Intervention strategies to reduce the risk of zoonotic infection with avian influenza viruses: scientific basis, challenges and knowledge gaps. Influenza Other Respir Viruses 2013;7(Suppl. 2):15–25. [19] Pongcharoensuk P, Adisasmito W, Sat le M, Silkavute P, Muchlisoh L, Cong Hoat P, et al. Avian and pandemic human influenza policy in South-East Asia: the interface between economic and public health imperatives. Health Policy Plan 2012;27:374–83. [20] Swayne DE, Pavade G, Hamilton K, Vallat B, Miyagishima K. Assessment of national strategies for control of high-pathogenicity avian influenza and lowpathogenicity notifiable avian influenza in poultry, with emphasis on vaccines and vaccination. Rev Sci Tech 2011;30:839–70. [21] Domenech J, Dauphin G, Rushton J, McGrane J, Lubroth J, Tripodi A, et al. Experiences with vaccination in countries endemically infected with highly pathogenic avian influenza: the Food and Agriculture Organization perspective. Rev Sci Tech 2009;28:293–305. [22] Bruschke CJ, Pittman M, Laddomada A. International regulations and standards for avian influenza, including the vaccine standards of the World Organisation for Animal Health. Rev Sci Tech 2009;28:379–89.

Please cite this article in press as: Rahn J, et al. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.03.052

G Model JVAC-16280; No. of Pages 11 8

ARTICLE IN PRESS J. Rahn et al. / Vaccine xxx (2015) xxx–xxx

[23] Rudolf M, Poppel M, Frohlich A, Breithaupt A, Teifke J, Blohm U, et al. Longitudinal 2 years field study of conventional vaccination against highly pathogenic avian influenza H5N1 in layer hens. Vaccine 2010;28:6832–40. [24] Longworth N, Mourits MC, Saatkamp HW. Economic analysis of HPAI control in the Netherlands I: epidemiological modelling to support economic analysis. Transbound Emerg Dis 2014;61:199–216. [25] Savill NJ, St Rose SG, Keeling MJ, Woolhouse ME. Silent spread of H5N1 in vaccinated poultry. Nature 2006;442:757. [26] Grund C, Abdelwhab el SM, Arafa AS, Ziller M, Hassan MK, Aly MM, et al. Highly pathogenic avian influenza virus H5N1 from Egypt escapes vaccine-induced immunity but confers clinical protection against a heterologous clade 2.2.1 Egyptian isolate. Vaccine 2011;29:5567–73. [27] Connie Leung YH, Luk G, Sia SF, Wu YO, Ho CK, Chow KC, et al. Experimental challenge of chicken vaccinated with commercially available H5 vaccines reveals loss of protection to some highly pathogenic avian influenza H5N1 strains circulating in Hong Kong/China. Vaccine 2013;31:3536–42. [28] Iwami S, Suzuki T, Takeuchi Y. Paradox of vaccination: is vaccination really effective against avian flu epidemics? PLoS One 2009;4:e4915. [29] Hsu SM, Chen TH, Wang CH. Efficacy of avian influenza vaccine in poultry: a meta-analysis. Avian Dis 2010;54:1197–209. [30] Vijaykrishna D, Smith GJ, Pybus OG, Zhu H, Bhatt S, Poon LL, et al. Longterm evolution and transmission dynamics of swine influenza A virus. Nature 2011;473:519–22. [31] Harder TC, Grosse Beilage E, Lange E, Meiners C, Dohring S, Pesch S, et al. Expanded cocirculation of stable subtypes, emerging lineages, and new sporadic reassortants of porcine influenza viruses in swine populations in Northwest Germany. J Virol 2013;87:10460–76. [32] Suarez DL. DIVA vaccination strategies for avian influenza virus. Avian Dis 2012;56:836–44. [33] Martinon F, Krishnan S, Lenzen G, Magne R, Gomard E, Guillet JG, et al. Induction of virus-specific cytotoxic T lymphocytes in vivo by liposome-entrapped mRNA. Eur J Immunol 1993;23:1719–22. [34] Kramps T, Probst J. Messenger RNA-based vaccines: progress, challenges, applications. Wiley Interdiscip Rev RNA 2013;4:737–49. [35] van der Velden AW, Voorma HO, Thomas AA. Vector design for optimal protein expression. Biotechniques 2001;31:572 [574, 576–580, passim]. [36] Kreiter S, Selmi A, Diken M, Koslowski M, Britten CM, Huber C, et al. Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity. Cancer Res 2010;70:9031–40. [37] Petsch B, Schnee M, Vogel AB, Lange E, Hoffmann B, Voss D, et al. Protective efficacy of in vitro synthesized, specific mRNA vaccines against influenza A virus infection. Nat Biotechnol 2012;30:1210–6. [38] Grudzien-Nogalska E, Kowalska J, Su W, Kuhn AN, Slepenkov SV, Darzynkiewicz E, et al. Synthetic mRNAs with superior translation and stability properties. Methods Mol Biol 2013;969:55–72. [39] Rabinovich PM, Weissman SM. Cell engineering with synthetic messenger RNA. Methods Mol Biol 2013;969:3–28. [40] Moon SL, Wilusz J. In vitro transcription of modified RNAs. Methods Mol Biol 2012;941:171–80. [41] Padilla R, Sousa R. A Y639F/H784A T7 RNA polymerase double mutant displays superior properties for synthesizing RNAs with non-canonical NTPs. Nucleic Acids Res 2002;30:e138. [42] Paul S, Stang A, Lennartz K, Tenbusch M, Uberla K. Selection of a T7 promoter mutant with enhanced in vitro activity by a novel multi-copy bead display approach for in vitro evolution. Nucleic Acids Res 2013;41:e29. [43] Kariko K, Muramatsu H, Ludwig J, Weissman D. Generating the optimal mRNA for therapy: HPLC purification eliminates immune activation and improves translation of nucleoside-modified, protein-encoding mRNA. Nucleic Acids Res 2011;39:e142. [44] Jones KL, Drane D, Gowans EJ. Long-term storage of DNA-free RNA for use in vaccine studies. Biotechniques 2007;43:675–81. [45] Hoerr I, Obst R, Rammensee HG, Jung G. In vivo application of RNA leads to induction of specific cytotoxic T lymphocytes and antibodies. Eur J Immunol 2000;30:1–7. [46] Diken M, Kreiter S, Selmi A, Britten CM, Huber C, Tureci O, et al. Selective uptake of naked vaccine RNA by dendritic cells is driven by macropinocytosis and abrogated upon DC maturation. Gene Ther 2011;18:702–8. [47] Lorenz C, Fotin-Mleczek M, Roth G, Becker C, Dam TC, Verdurmen WP, et al. Protein expression from exogenous mRNA: uptake by receptormediated endocytosis and trafficking via the lysosomal pathway. RNA Biol 2011;8:627–36. [48] Probst J, Weide B, Scheel B, Pichler BJ, Hoerr I, Rammensee HG, et al. Spontaneous cellular uptake of exogenous messenger RNA in vivo is nucleic acid-specific, saturable and ion dependent. Gene Ther 2007;14:1175–80. [49] DeWitte-Orr SJ, Collins SE, Bauer CM, Bowdish DM, Mossman KL. An accessory to the ‘Trinity’: SR-As are essential pathogen sensors of extracellular dsRNA, mediating entry and leading to subsequent type I IFN responses. PLoS Pathog 2010;6:e1000829. [50] Scheel B, Teufel R, Probst J, Carralot JP, Geginat J, Radsak M, et al. Tolllike receptor-dependent activation of several human blood cell types by protamine-condensed mRNA. Eur J Immunol 2005;35:1557–66. [51] Fotin-Mleczek M, Duchardt KM, Lorenz C, Pfeiffer R, Ojkic-Zrna S, Probst J, et al. Messenger RNA-based vaccines with dual activity induce balanced TLR7 dependent adaptive immune responses and provide antitumor activity. J Immunother 2011;34:1–15.

[52] Rettig L, Haen SP, Bittermann AG, von Boehmer L, Curioni A, Kramer SD, et al. Particle size and activation threshold: a new dimension of danger signaling. Blood 2010;115:4533–41. [53] Honda K, Ohba Y, Yanai H, Negishi H, Mizutani T, Takaoaka A, et al. Spatiotemporal regulation of MyD88-IRF-7 signalling for robust type-I interferon induction. Nature 2005;434:1035–40. [54] Desmet CJ, Ishii KJ. Nucleic acid sensing at the interface between innate and adaptive immunity in vaccination. Nat Rev Immunol 2012;12:479–91. [55] Barbalat R, Ewald SE, Mouchess ML, Barton GM. Nucleic acid recognition by the innate immune system. Annu Rev Immunol 2011;29: 185–214. [56] Kariko K, Ni H, Capodici J, Lamphier M, Weissman D. mRNA is an endogenous ligand for Toll-like receptor 3. J Biol Chem 2004;279:12542–50. [57] Kratky W, Reis e Sousa C, Oxenius A, Sporri R. Direct activation of antigenpresenting cells is required for CD8+ T-cell priming and tumor vaccination. Proc Natl Acad Sci U S A 2011;108:17414–9. [58] Hou B, Saudan P, Ott G, Wheeler ML, Ji M, Kuzmich L, et al. Selective utilization of Toll-like receptor and MyD88 signaling in B cells for enhancement of the antiviral germinal center response. Immunity 2011;34:375–84. [59] Boczkowski D, Nair SK, Snyder D, Gilboa E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J Exp Med 1996;184:465–72. [60] Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, et al. Direct gene transfer into mouse muscle in vivo. Science 1990;247:1465–8. [61] Granstein RD, Ding W, Ozawa H. Induction of anti-tumor immunity with epidermal cells pulsed with tumor-derived RNA or intradermal administration of RNA. J Invest Dermatol 2000;114:632–6. [62] Liard C, Munier S, Joulin-Giet A, Bonduelle O, Hadam S, Duffy D, et al. Intradermal immunization triggers epidermal Langerhans cell mobilization required for CD8 T-cell immune responses. J Invest Dermatol 2012;132:615–25. [63] Hangartner L, Zinkernagel RM, Hengartner H. Antiviral antibody responses: the two extremes of a wide spectrum. Nat Rev Immunol 2006;6:231–43. [64] Liu MA. DNA vaccines: an historical perspective and view to the future. Immunol Rev 2011;239:62–84. [65] Little SR, Lynn DM, Puram SV, Langer R. Formulation and characterization of poly (beta amino ester) microparticles for genetic vaccine delivery. J Control Release 2005;107:449–62. [66] Singh M, Briones M, Ott G, O’Hagan D. Cationic microparticles: a potent delivery system for DNA vaccines. Proc Natl Acad Sci U S A 2000;97:811–6. [67] Howard KA, Li XW, Somavarapu S, Singh J, Green N, Atuah KN, et al. Formulation of a microparticle carrier for oral polyplex-based DNA vaccines. Biochim Biophys Acta 2004;1674:149–57. [68] Ozaki T, Yauchi M, Xin KQ, Hirahara F, Okuda K. Cross-reactive protection against influenza A virus by a topically applied DNA vaccine encoding M gene with adjuvant. Viral Immunol 2005;18:373–80. [69] Tang DC, DeVit M, Johnston SA. Genetic immunization is a simple method for eliciting an immune response. Nature 1992;356:152–4. [70] Suarez DL, Schultz-Cherry S. The effect of eukaryotic expression vectors and adjuvants on DNA vaccines in chickens using an avian influenza model. Avian Dis 2000;44:861–8. [71] Montgomery DL, Shiver JW, Leander KR, Perry HC, Friedman A, Martinez D, et al. Heterologous and homologous protection against influenza A by DNA vaccination: optimization of DNA vectors. DNA Cell Biol 1993;12:777–83. [72] Ulmer JB, Donnelly JJ, Parker SE, Rhodes GH, Felgner PL, Dwarki VJ, et al. Heterologous protection against influenza by injection of DNA encoding a viral protein. Science 1993;259:1745–9. [73] Tao P, Luo M, Pan R, Ling D, Zhou S, Tien P, et al. Enhanced protective immunity against H5N1 influenza virus challenge by vaccination with DNA expressing a chimeric hemagglutinin in combination with an MHC class I-restricted epitope of nucleoprotein in mice. Antiviral Res 2009;81:253–60. [74] Donnelly JJ, Friedman A, Ulmer JB, Liu MA. Further protection against antigenic drift of influenza virus in a ferret model by DNA vaccination. Vaccine 1997;15:865–8. [75] Ulmer JB, Fu TM, Deck RR, Friedman A, Guan L, DeWitt C, et al. Protective CD4+ and CD8+ T cells against influenza virus induced by vaccination with nucleoprotein DNA. J Virol 1998;72:5648–53. [76] Saha S, Yoshida S, Ohba K, Matsui K, Matsuda T, Takeshita F, et al. A fused gene of nucleoprotein (NP) and herpes simplex virus genes (VP22) induces highly protective immunity against different subtypes of influenza virus. Virology 2006;354:48–57. [77] Olsen CW. DNA vaccination against influenza viruses: a review with emphasis on equine and swine influenza. Vet Microbiol 2000;74:149–64. [78] Larsen DL, Karasin A, Olsen CW. Immunization of pigs against influenza virus infection by DNA vaccine priming followed by killed-virus vaccine boosting. Vaccine 2001;19:2842–53. [79] Gorres JP, Lager KM, Kong WP, Royals M, Todd JP, Vincent AL, et al. DNA vaccination elicits protective immune responses against pandemic and classic swine influenza viruses in pigs. Clin Vaccine Immunol 2011;18:1987–95. [80] Macklin MD, McCabe D, McGregor MW, Neumann V, Meyer T, Callan R, et al. Immunization of pigs with a particle-mediated DNA vaccine to influenza A virus protects against challenge with homologous virus. J Virol 1998;72:1491–6. [81] Lim KL, Jazayeri SD, Yeap SK, Mohamed Alitheen NB, Bejo MH, Ideris A, et al. Antibody and T cell responses induced in chickens immunized with avian influenza virus N1 and NP DNA vaccine with chicken IL-15 and IL-18. Res Vet Sci 2013;95:1224–34.

Please cite this article in press as: Rahn J, et al. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.03.052

G Model JVAC-16280; No. of Pages 11

ARTICLE IN PRESS J. Rahn et al. / Vaccine xxx (2015) xxx–xxx

[82] Lim KL, Jazayeri SD, Yeap SK, Alitheen NB, Bejo MH, Ideris A, et al. Coadministration of avian influenza virus H5 plasmid DNA with chicken IL-15 and IL-18 enhanced chickens immune responses. BMC Vet Res 2012;8:132. [83] Kodihalli S, Kobasa DL, Webster RG. Strategies for inducing protection against avian influenza A virus subtypes with DNA vaccines. Vaccine 2000;18:2592–9. [84] Pertmer TM, Oran AE, Moser JM, Madorin CA, Robinson HL. DNA vaccines for influenza virus: differential effects of maternal antibody on immune responses to hemagglutinin and nucleoprotein. J Virol 2000;74:7787–93. [85] Jiang Y, Yu K, Zhang H, Zhang P, Li C, Tian G, et al. Enhanced protective efficacy of H5 subtype avian influenza DNA vaccine with codon optimized HA gene in a pCAGGS plasmid vector. Antiviral Res 2007;75:234–41. [86] Jiang Y, Zhang H, Wang G, Zhang P, Tian G, Bu Z, et al. Protective efficacy of H7 subtype avian influenza DNA vaccine. Avian Dis 2010;54:290–3. [87] Li J, Jiang Y, Zhao S, Chang X, Liu J, Zeng X, et al. Protective efficacy of an H5N1 DNA vaccine against challenge with a lethal H5N1 virus in quail. Avian Dis 2012;56:937–9. [88] Chen Q, Madson D, Miller CL, Harris DL. Vaccine development for protecting swine against influenza virus. Anim Health Res Rev 2012;13:181–95. [89] Pertmer TM, Eisenbraun MD, McCabe D, Prayaga SK, Fuller DH, Haynes JR. Gene gun-based nucleic acid immunization: elicitation of humoral and cytotoxic T lymphocyte responses following epidermal delivery of nanogram quantities of DNA. Vaccine 1995;13:1427–30. [90] Dhama K, Mahendran M, Gupta PK, Rai A. DNA vaccines and their applications in veterinary practice: current perspectives. Vet Res Commun 2008;32:341–56. [91] Wang Z, Troilo PJ, Wang X, Griffiths TG, Pacchione SJ, Barnum AB, et al. Detection of integration of plasmid DNA into host genomic DNA following intramuscular injection and electroporation. Gene Ther 2004;11:711–21. [92] Zimmer G. RNA replicons – a new approach for influenza virus immunoprophylaxis. Viruses 2010;2:413–34. [93] Rayner JO, Dryga SA, Kamrud KI. Alphavirus vectors and vaccination. Rev Med Virol 2002;12:279–96. [94] Bredenbeek PJ, Frolov I, Rice CM, Schlesinger S. Sindbis virus expression vectors: packaging of RNA replicons by using defective helper RNAs. J Virol 1993;67:6439–46. [95] Schwartz JA, Buonocore L, Roberts A, Suguitan Jr A, Kobasa D, Kobinger G, et al. Vesicular stomatitis virus vectors expressing avian influenza H5 HA induce cross-neutralizing antibodies and long-term protection. Virology 2007;366:166–73. [96] Roberts A, Buonocore L, Price R, Forman J, Rose JK. Attenuated vesicular stomatitis viruses as vaccine vectors. J Virol 1999;73:3723–32. [97] Vander Veen R, Kamrud K, Mogler M, Loynachan AT, McVicker J, Berglund P, et al. Rapid development of an efficacious swine vaccine for novel H1N1. PLoS Curr 2009;1:RRN1123. [98] Vander Veen RL, Harris DL, Kamrud KI. Alphavirus replicon vaccines. Anim Health Res Rev 2012;13:1–9. [99] Erdman MM, Kamrud KI, Harris DL, Smith J. Alphavirus replicon particle vaccines developed for use in humans induce high levels of antibodies to influenza virus hemagglutinin in swine: proof of concept. Vaccine 2010;28:594–6. [100] Vander Veen RL, Loynachan AT, Mogler MA, Russell BJ, Harris DL, Kamrud KI. Safety, immunogenicity, and efficacy of an alphavirus replicon-based swine influenza virus hemagglutinin vaccine. Vaccine 2012;30:1944–50. [101] Vander Veen RL, Mogler MA, Russell BJ, Loynachan AT, Harris, K.I. Kamrud DL. Haemagglutinin and nucleoprotein replicon particle vaccination of swine protects against the pandemic H1N1 2009 virus. Vet Rec 2013;173:344. [102] Schultz-Cherry S, Dybing JK, Davis NL, Williamson C, Suarez DL, Johnston R, et al. Influenza virus (A/HK/156/97) hemagglutinin expressed by an alphavirus replicon system protects chickens against lethal infection with Hong Kong-origin H5N1 viruses. Virology 2000;278:55–9. [103] Sylte MJ, Hubby B, Suarez DL. Influenza neuraminidase antibodies provide partial protection for chickens against high pathogenic avian influenza infection. Vaccine 2007;25:3763–72. [104] Kalhoro NH, Veits J, Rautenschlein S, Zimmer G. A recombinant vesicular stomatitis virus replicon vaccine protects chickens from highly pathogenic avian influenza virus (H7N1). Vaccine 2009;27:1174–83. [105] Halbherr SJ, Brostoff T, Tippenhauer M, Locher S, Berger Rentsch M, Zimmer G. Vaccination with recombinant RNA replicon particles protects chickens from H5N1 highly pathogenic avian influenza virus. PLOS ONE 2013;8:e66059. [106] Ma JH, Yang FR, Yu H, Zhou YJ, Li GX, Huang M, et al. An M2e-based synthetic peptide vaccine for influenza A virus confers heterosubtypic protection from lethal virus challenge. Virol J 2013;10:227. [107] Wang TT, Tan GS, Hai R, Pica N, Ngai L, Ekiert DC, et al. Vaccination with a synthetic peptide from the influenza virus hemagglutinin provides protection against distinct viral subtypes. Proc Natl Acad Sci U S A 2010;107:18979–84. [108] Margine I, Krammer F, Hai R, Heaton NS, Tan GS, Andrews SA, et al. Hemagglutinin stalk-based universal vaccine constructs protect against group 2 influenza A viruses. J Virol 2013;87:10435–46. [109] Krammer F, Pica N, Hai R, Tan GS, Palese P. Hemagglutinin stalk-reactive antibodies are boosted following sequential infection with seasonal and pandemic H1N1 influenza virus in mice. J Virol 2012;86:10302–7. [110] Vergara-Alert J, Argilaguet JM, Busquets N, Ballester M, Martin-Valls GE, Rivas R, et al. Conserved synthetic peptides from the hemagglutinin of influenza viruses induce broad humoral and T-cell responses in a pig model. PLOS ONE 2012;7:e40524.

9

[111] Myers T. Subunit vaccine. In: Myers T, editor. Mosby’s Midical dictionary. Saint Lois, MO: Academic Press; 2010. p. 1778. [112] Cox MM, Hollister JR. FluBlok, a next generation influenza vaccine manufactured in insect cells. Biologicals 2009;37:182–9. [113] Shoji Y, Chichester JA, Jones M, Manceva SD, Damon E, Mett V, et al. Plantbased rapid production of recombinant subunit hemagglutinin vaccines targeting H1N1 and H5N1 influenza. Hum Vaccines 2011;7(Suppl.):41–50. [114] Liu G, Zhang F, Shi J, Tian G, Chen H, Yu K, et al. A subunit vaccine candidate derived from a classic H5N1 avian influenza virus in China protects fowls and BALB/c mice from lethal challenge. Vaccine 2013;31:5398–404. [115] Crawford J, Wilkinson B, Vosnesensky A, Smith G, Garcia M, Stone H, et al. Baculovirus-derived hemagglutinin vaccines protect against lethal influenza infections by avian H5 and H7 subtypes. Vaccine 1999;17:2265–74. [116] Wu Q, Fang L, Wu X, Li B, Luo R, Yu Z, et al. A pseudotype baculovirus-mediated vaccine confers protective immunity against lethal challenge with H5N1 avian influenza virus in mice and chickens. Mol Immunol 2009;46:2210–7. [117] Kalthoff D, Giritch A, Geisler K, Bettmann U, Klimyuk V, Hehnen HR, et al. Immunization with plant-expressed hemagglutinin protects chickens from lethal highly pathogenic avian influenza virus H5N1 challenge infection. J Virol 2010;84:12002–10. [118] Morein B, Sundquist B, Hoglund S, Dalsgaard K, Osterhaus A. Iscom, a novel structure for antigenic presentation of membrane proteins from enveloped viruses. Nature 1984;308:457–60. [119] Kodihalli S, Sivanandan V, Nagaraja KV, Shaw D, Halvorson DA. A type-specific avian influenza virus subunit vaccine for turkeys: induction of protective immunity to challenge infection. Vaccine 1994;12:1467–72. [120] Haynes JR. Influenza virus-like particle vaccines. Expert Rev Vaccines 2009;8:435–45. [121] Latham T, Galarza JM. Formation of wild-type and chimeric influenza viruslike particles following simultaneous expression of only four structural proteins. J Virol 2001;75:6154–65. [122] Pushko P, Tumpey TM, Bu F, Knell J, Robinson R, Smith G. Influenza virus-like particles comprised of the HA, NA, and M1 proteins of H9N2 influenza virus induce protective immune responses in BALB/c mice. Vaccine 2005;23:5751–9. [123] Quan FS, Huang C, Compans RW, Kang SM. Virus-like particle vaccine induces protective immunity against homologous and heterologous strains of influenza virus. J Virol 2007;81:3514–24. [124] Bright RA, Carter DM, Daniluk S, Toapanta FR, Ahmad A, Gavrilov V, et al. Influenza virus-like particles elicit broader immune responses than whole virion inactivated influenza virus or recombinant hemagglutinin. Vaccine 2007;25:3871–8. [125] Song JM, Van Rooijen N, Bozja J, Compans RW, Kang SM. Vaccination inducing broad and improved cross protection against multiple subtypes of influenza A virus. Proc Natl Acad Sci U S A 2011;108:757–61. [126] Chen BJ, Leser GP, Morita E, Lamb RA. Influenza virus hemagglutinin and neuraminidase, but not the matrix protein, are required for assembly and budding of plasmid-derived virus-like particles. J Virol 2007;81:7111–23. [127] Mena I, Vivo A, Perez E, Portela A. Rescue of a synthetic chloramphenicol acetyltransferase RNA into influenza virus-like particles obtained from recombinant plasmids. J Virol 1996;70:5016–24. [128] Neumann G, Watanabe T, Kawaoka Y. Plasmid-driven formation of influenza virus-like particles. J Virol 2000;74:547–51. [129] Wu CY, Yeh YC, Yang YC, Chou C, Liu MT, Wu HS, et al. Mammalian expression of virus-like particles for advanced mimicry of authentic influenza virus. PLoS ONE 2010;5:e9784. [130] Chang GRL, Lai SY, Chang PC, Wang MY. Production of immunogenic onecomponent avian H7-subtype influenza virus-like particles. Process Biochem 2011;46:1292–8. [131] D’Aoust MA, Lavoie PO, Couture MM, Trepanier S, Guay JM, Dargis M, et al. Influenza virus-like particles produced by transient expression in Nicotiana benthamiana induce a protective immune response against a lethal viral challenge in mice. Plant Biotechnol J 2008;6:930–40. [132] Lee DH, Park JK, Lee YN, Song JM, Kang SM, Lee JB, et al. H9N2 avian influenza virus-like particle vaccine provides protective immunity and a strategy for the differentiation of infected from vaccinated animals. Vaccine 2011;29:4003–7. [133] Choi JG, Kim MC, Kang HM, Kim KI, Lee KJ, Park CK, et al. Protective efficacy of baculovirus-derived influenza virus-like particles bearing H5 HA alone or in combination with M1 in chickens. Vet Microbiol 2013;162:623–30. [134] Park JK, Lee DH, Youn HN, Kim MS, Lee YN, Yuk SS, et al. Protective efficacy of crude virus-like particle vaccine against HPAI H5N1 in chickens and its application on DIVA strategy. Influenza Other Respir Viruses 2013;7:340–8. [135] Pyo HM, Masic A, Woldeab N, Embury-Hyatt C, Lin L, Shin YK, et al. Pandemic H1N1 influenza virus-like particles are immunogenic and provide protective immunity to pigs. Vaccine 2012;30:1297–304. [136] Wesley RD, Lager KM. Overcoming maternal antibody interference by vaccination with human adenovirus 5 recombinant viruses expressing the hemagglutinin and the nucleoprotein of swine influenza virus. Vet Microbiol 2006;118:67–75. [137] Wesley RD, Tang M, Lager KM. Protection of weaned pigs by vaccination with human adenovirus 5 recombinant viruses expressing the hemagglutinin and the nucleoprotein of H3N2 swine influenza virus. Vaccine 2004;22:3427–34. [138] Pandey A, Singh N, Vemula SV, Couetil L, Katz JM, Donis R, et al. Impact of preexisting adenovirus vector immunity on immunogenicity and protection conferred with an adenovirus-based H5N1 influenza vaccine. PLOS ONE 2012;7:e33428.

Please cite this article in press as: Rahn J, et al. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.03.052

G Model JVAC-16280; No. of Pages 11 10

ARTICLE IN PRESS J. Rahn et al. / Vaccine xxx (2015) xxx–xxx

[139] Tian ZJ, Zhou GH, Zheng BL, Qiu HJ, Ni JQ, Yang HL, et al. A recombinant pseudorabies virus encoding the HA gene from H3N2 subtype swine influenza virus protects mice from virulent challenge. Vet Immunol Immunopathol 2006;111:211–8. [140] Li GX, Zhou YJ, Yu H, Tian ZJ, Yan LP, Zhang Q, et al. Prime-boost immunization with HA/C3d DNA followed by a recombinant pseudorabies virus boost enhanced protective immunity against H3N2 swine influenza virus in mice. Res Vet Sci 2010;88:345–51. [141] Klingbeil K, Lange E, Teifke JP, Mettenleiter TC, Fuchs W. Immunization of pigs with an attenuated pseudorabies virus recombinant expressing the haemagglutinin of pandemic swine origin H1N1 influenza A virus. J Gen Virol 2014;95:948–59. [142] Ma W, Richt JA. Swine influenza vaccines: current status and future perspectives. Anim Health Res Rev 2010;11:81–96. [143] Swayne DE, Spackman E, Pantin-Jackwood M. Success factors for avian influenza vaccine use in poultry and potential impact at the wild birdagricultural interface. Ecohealth 2014;11:94–108. [144] Said A, Lange E, Beer M, Damiani A, Osterrieder N. Recombinant equine herpesvirus 1 (EHV-1) vaccine protects pigs against challenge with influenza A(H1N1)pmd09. Virus Res 2013;173:371–6. [145] Foley PL. Purified DNA. Google Patents; 2001. [146] Najera JL, Gomez CE, Domingo-Gil E, Gherardi MM, Esteban M. Cellular and biochemical differences between two attenuated poxvirus vaccine candidates (MVA and NYVAC) and role of the C7L gene. J Virol 2006;80:6033–47. [147] Kyriakis CS, De Vleeschauwer A, Barbe F, Bublot M, Van Reeth K. Safety, immunogenicity and efficacy of poxvirus-based vector vaccines expressing the haemagglutinin gene of a highly pathogenic H5N1 avian influenza virus in pigs. Vaccine 2009;27:2258–64. [148] Park MS, Steel J, Garcia-Sastre A, Swayne D, Palese P. Engineered viral vaccine constructs with dual specificity: avian influenza and Newcastle disease. Proc Natl Acad Sci U S A 2006;103:8203–8. [149] Ge J, Deng G, Wen Z, Tian G, Wang Y, Shi J, et al. Newcastle disease virus-based live attenuated vaccine completely protects chickens and mice from lethal challenge of homologous and heterologous H5N1 avian influenza viruses. J Virol 2007;81:150–8. [150] Nayak B, Rout SN, Kumar S, Khalil MS, Fouda MM, Ahmed LE, et al. Immunization of chickens with Newcastle disease virus expressing H5 hemagglutinin protects against highly pathogenic H5N1 avian influenza viruses. PLoS One 2009;4:e6509. [151] Ramp K, Veits J, Deckers D, Rudolf M, Grund C, Mettenleiter TC, et al. Coexpression of avian influenza virus H5 and N1 by recombinant Newcastle disease virus and the impact on immune response in chickens. Avian Dis 2011;55:413–21. [152] Cornelissen LA, de Leeuw OS, Tacken MG, Klos HC, de Vries RP, de Boer-Luijtze EA, et al. Protective efficacy of Newcastle disease virus expressing soluble trimeric hemagglutinin against highly pathogenic H5N1 influenza in chickens and mice. PLOS ONE 2012;7:e44447. [153] Swayne DE, Suarez DL, Schultz-Cherry S, Tumpey TM, King DJ, Nakaya T, et al. Recombinant paramyxovirus type 1-avian influenza-H7 virus as a vaccine for protection of chickens against influenza and Newcastle disease. Avian Dis 2003;47:1047–50. [154] Veits J, Wiesner D, Fuchs W, Hoffmann B, Granzow H, Starick E, et al. Newcastle disease virus expressing H5 hemagglutinin gene protects chickens against Newcastle disease and avian influenza. Proc Natl Acad Sci U S A 2006;103: 8197–202. [155] Nayak B, Kumar S, DiNapoli JM, Paldurai A, Perez DR, Collins PL, et al. Contributions of the avian influenza virus HA, NA, and M2 surface proteins to the induction of neutralizing antibodies and protective immunity. J Virol 2010;84:2408–20. [156] Lardinois A, Steensels M, Lambrecht B, Desloges N, Rahaus M, Rebeski D, et al. Potency of a recombinant NDV-H5 vaccine against various HPAI H5N1 virus challenges in SPF chickens. Avian Dis 2012;56:928–36. [157] Veits J, Luschow D, Kindermann K, Werner O, Teifke JP, Mettenleiter TC, et al. Deletion of the non-essential UL0 gene of infectious laryngotracheitis (ILT) virus leads to attenuation in chickens, and UL0 mutants expressing influenza virus haemagglutinin (H7) protect against ILT and fowl plague. J Gen Virol 2003;84:3343–52. [158] Steglich C, Grund C, Ramp K, Breithaupt A, Hoper D, Keil G, et al. Chimeric newcastle disease virus protects chickens against avian influenza in the presence of maternally derived NDV immunity. PLOS ONE 2013;8:e72530. [159] Nakaya T, Cros J, Park MS, Nakaya Y, Zheng H, Sagrera A, et al. Recombinant newcastle disease virus as a vaccine vector. J Virol 2001;75:11868–73. [160] Chen H, Bu Z. Development and application of avian influenza vaccines in China. Curr Top Microbiol Immunol 2009;333:153–62. [161] Luschow D, Werner O, Mettenleiter TC, Fuchs W. Protection of chickens from lethal avian influenza A virus infection by live-virus vaccination with infectious laryngotracheitis virus recombinants expressing the hemagglutinin (H5) gene. Vaccine 2001;19:4249–59. [162] Pavlova SP, Veits J, Keil GM, Mettenleiter TC, Fuchs W. Protection of chickens against H5N1 highly pathogenic avian influenza virus infection by live vaccination with infectious laryngotracheitis virus recombinants expressing H5 hemagglutinin and N1 neuraminidase. Vaccine 2009;27:773–85. [163] Kilany W, Dauphin G, Selim A, Tripodi A, Samy M, Sobhy H, et al. Protection conferred by recombinant HVT avian influenza (rHVT-H5) vaccine in rearing period in two commercial layer chicken breeds in Egypt. Avian Pathol 2014:1–38.

[164] Toro H, Tang DC, Suarez DL, Sylte MJ, Pfeiffer J, van Kampen KR. Protective avian influenza in ovo vaccination with non-replicating human adenovirus vector. Vaccine 2007;25:2886–91. [165] Gao W, Soloff AC, Lu X, Montecalvo A, Nguyen DC, Matsuoka Y, et al. Protection of mice and poultry from lethal H5N1 avian influenza virus through adenovirus-based immunization. J Virol 2006;80:1959–64. [166] Taylor J, Weinberg R, Kawaoka Y, Webster RG, Paoletti E. Protective immunity against avian influenza induced by a fowlpox virus recombinant. Vaccine 1988;6:504–8. [167] Beard CW, Schnitzlein WM, Tripathy DN. Effect of route of administration on the efficacy of a recombinant fowlpox virus against H5N2 avian influenza. Avian Dis 1992;36:1052–5. [168] Beard CW, Schnitzlein WM, Tripathy DN. Protection of chickens against highly pathogenic avian influenza virus (H5N2) by recombinant fowlpox viruses. Avian Dis 1991;35:356–9. [169] Swayne DE, Beck JR, Mickle TR. Efficacy of recombinant fowl poxvirus vaccine in protecting chickens against a highly pathogenic Mexican-origin H5N2 avian influenza virus. Avian Dis 1997;41:910–22. [170] Swayne DE, Garcia M, Beck JR, Kinney N, Suarez DL. Protection against diverse highly pathogenic H5 avian influenza viruses in chickens immunized with a recombinant fowlpox vaccine containing an H5 avian influenza hemagglutinin gene insert. Vaccine 2000;18:1088–95. [171] Webster RG, Taylor J, Pearson J, Rivera E, Paoletti E. Immunity to Mexican H5N2 avian influenza viruses induced by a fowl pox-H5 recombinant. Avian Dis 1996;40:461–5. [172] Swayne DE, Suarez DL. Highly pathogenic avian influenza. Rev Sci Tech 2000;19:463–82. [173] Bublot M, Pritchard N, Swayne DE, Selleck P, Karaca K, Suarez DL, et al. Development and use of fowlpox vectored vaccines for avian influenza. Ann N Y Acad Sci 2006;1081:193–201. [174] Villarreal-Chavez C, Rivera-Cruz E. An update on avian influenza in Mexico. Avian Dis 2003;47:1002–5. [175] Meeusen ENT, Walker J, Peters A, Pastoret PP, Jungersen G. Current status of veterinary vaccines. Clin Microbiol Rev 2007;20:489–510. [176] Richt JA, Lekcharoensuk P, Lager KM, Vincent AL, Loiacono CM, Janke BH, et al. Vaccination of pigs against swine influenza viruses by using an NS1-truncated modified live-virus vaccine. J Virol 2006;80:11009–18. [177] Masic A, Booth JS, Mutwiri GK, Babiuk LA, Zhou Y. Elastase-dependent live attenuated swine influenza A viruses are immunogenic and confer protection against swine influenza A virus infection in pigs. J Virol 2009;83:10198–210. [178] Masic A, Babiuk LA, Zhou Y. Reverse genetics-generated elastase-dependent swine influenza viruses are attenuated in pigs. J Gen Virol 2009;90:375–85. [179] Babiuk S, Masic A, Graham J, Neufeld J, van der Loop M, Copps J, et al. An elastase-dependent attenuated heterologous swine influenza virus protects against pandemic H1N1 2009 influenza challenge in swine. Vaccine 2011;29:3118–23. [180] Masic A, Lu X, Li J, Mutwiri GK, Babiuk LA, Brown EG, et al. Immunogenicity and protective efficacy of an elastase-dependent live attenuated swine influenza virus vaccine administered intranasally in pigs. Vaccine 2010;28:7098–108. [181] Kappes MA, Sandbulte MR, Platt R, Wang C, Lager KM, Henningson JN, et al. Vaccination with NS1-truncated H3N2 swine influenza virus primes T cells and confers cross-protection against an H1N1 heterosubtypic challenge in pigs. Vaccine 2012;30:280–8. [182] Vincent AL, Ma W, Lager KM, Richt JA, Janke BH, Sandbulte MR, et al. Live attenuated influenza vaccine provides superior protection from heterologous infection in pigs with maternal antibodies without inducing vaccine-associated enhanced respiratory disease. J Virol 2012;86:10597–605. [183] Solorzano A, Ye J, Perez DR. Alternative live-attenuated influenza vaccines based on modifications in the polymerase genes protect against epidemic and pandemic flu. J Virol 2010;84:4587–96. [184] Vincent AL, Ma W, Lager KM, Janke BH, Webby RJ, Garcia-Sastre A, et al. Efficacy of intranasal administration of a truncated NS1 modified live influenza virus vaccine in swine. Vaccine 2007;25:7999–8009. [185] Pena L, Vincent AL, Ye J, Ciacci-Zanella JR, Angel M, Lorusso A, et al. Modifications in the polymerase genes of a swine-like triple-reassortant influenza virus to generate live attenuated vaccines against 2009 pandemic H1N1 viruses. J Virol 2011;85:456–69. [186] Pyo HM, Zhou Y. Protective efficacy of intranasally administered bivalent live influenza vaccine and immunological mechanisms underlying the protection. Vaccine 2014;32:3835–42. [187] Rohrs S, Kalthoff D, Beer M. A model for early onset of protection against lethal challenge with highly pathogenic H5N1 influenza virus. Vaccine 2014;32:2631–6. [188] Solorzano A, Webby RJ, Lager KM, Janke BH, Garcia-Sastre A, Richt JA. Mutations in the NS1 protein of swine influenza virus impair anti-interferon activity and confer attenuation in pigs. J Virol 2005;79:7535–43. [189] Masic A, Pyo HM, Babiuk S, Zhou Y. An eight-segment swine influenza virus harboring H1 and H3 hemagglutinins is attenuated and protective against H1N1 and H3N2 subtypes in pigs. J Virol 2013;87:10114–25. [190] Hai R, Garcia-Sastre A, Swayne DE, Palese P. A reassortment-incompetent live attenuated influenza virus vaccine for protection against pandemic virus strains. J Virol 2011;85:6832–43. [191] Subbarao K, Matsuoka Y. The prospects and challenges of universal vaccines for influenza. Trends Microbiol 2013;21:350–8. [192] Ekiert DC, Wilson IA. Broadly neutralizing antibodies against influenza virus and prospects for universal therapies. Curr Opin Virol 2012;2:134–41.

Please cite this article in press as: Rahn J, et al. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.03.052

G Model JVAC-16280; No. of Pages 11

ARTICLE IN PRESS J. Rahn et al. / Vaccine xxx (2015) xxx–xxx

[193] Ekiert DC, Bhabha G, Elsliger MA, Friesen RH, Jongeneelen M, Thorsby M, et al. Antibody recognition of a highly conserved influenza virus epitope. Science 2009;324:246–51. [194] Ekiert DC, Kashyap AK, Steel J, Rubrum A, Bhabha G, Khayat R, et al. Crossneutralization of influenza A viruses mediated by a single antibody loop. Nature 2012;489:526–32. [195] Corti D, Voss J, Gamblin SJ, Codoni G, Macagno A, Jarrossay D, et al. A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza A hemagglutinins. Science 2011;333:850–6. [196] Sui J, Hwang WC, Perez S, Wei G, Aird D, Chen LM, et al. Structural and functional bases for broad-spectrum neutralization of avian and human influenza A viruses. Nat Struct Mol Biol 2009;16:265–73. [197] Friesen RH, Lee PS, Stoop EJ, Hoffman RM, Ekiert DC, Bhabha G, et al. A common solution to group 2 influenza virus neutralization. Proc Natl Acad Sci U S A 2014;111:445–50. [198] Yoshida R, Igarashi M, Ozaki H, Kishida N, Tomabechi D, Kida H, et al. Cross-protective potential of a novel monoclonal antibody directed against antigenic site B of the hemagglutinin of influenza A viruses. PLoS Pathog 2009;5:e1000350. [199] Ohshima N, Iba Y, Kubota-Koketsu R, Asano Y, Okuno Y, Kurosawa Y. Naturally occurring antibodies in humans can neutralize a variety of influenza virus strains, including H3, H1, H2, and H5. J Virol 2011;85: 11048–57. [200] Ekiert DC, Friesen RH, Bhabha G, Kwaks T, Jongeneelen M, Yu W, et al. A highly conserved neutralizing epitope on group 2 influenza A viruses. Science 2011;333:843–50. [201] Khurana S, Loving CL, Manischewitz J, King LR, Gauger PC, Henningson J, et al. Vaccine-induced anti-HA2 antibodies promote virus fusion and enhance influenza virus respiratory disease. Sci Transl Med 2013;5:200ra114. [202] Rajao DS, Loving CL, Gauger PC, Kitikoon P, Vincent AL. Influenza A virus hemagglutinin protein subunit vaccine elicits vaccine-associated enhanced respiratory disease in pigs. Vaccine 2014;32:5170–6. [203] Gravel C, Li C, Wang J, Hashem AM, Jaentschke B, van Domselaar G, et al. Quantitative analyses of all influenza type A viral hemagglutinins and neuraminidases using universal antibodies in simple slot blot assays. J Vis Exp 2011. [204] Hillaire ML, Osterhaus AD, Rimmelzwaan GF. Induction of virus-specific cytotoxic T lymphocytes as a basis for the development of broadly protective influenza vaccines. J Biomed Biotechnol 2011;2011:939860. [205] Boyd AC, Ruiz-Hernandez R, Peroval MY, Carson C, Balkissoon D, Staines K, et al. Towards a universal vaccine for avian influenza: protective efficacy of modified Vaccinia virus Ankara and Adenovirus vaccines expressing conserved influenza antigens in chickens challenged with low pathogenic avian influenza virus. Vaccine 2013;31:670–5. [206] Liu W, Zou P, Ding J, Lu Y, Chen YH. Sequence comparison between the extracellular domain of M2 protein human and avian influenza A virus provides new information for bivalent influenza vaccine design. Microbes Infect 2005;7:171–7. [207] Neirynck S, Deroo T, Saelens X, Vanlandschoot P, Jou WM, Fiers W. A universal influenza A vaccine based on the extracellular domain of the M2 protein. Nat Med 1999;5:1157–63. [208] Schotsaert M, Ysenbaert T, Neyt K, Ibanez LI, Bogaert P, Schepens B, et al. Natural and long-lasting cellular immune responses against influenza in the M2e-immune host. Mucosal Immunol 2013;6:276–87. [209] Treanor JJ, Tierney EL, Zebedee SL, Lamb RA, Murphy BR. Passively transferred monoclonal antibody to the M2 protein inhibits influenza A virus replication in mice. J Virol 1990;64:1375–7.

11

[210] Price GE, Soboleski MR, Lo CY, Misplon JA, Quirion MR, Houser KV, et al. Single-dose mucosal immunization with a candidate universal influenza vaccine provides rapid protection from virulent H5N1, H3N2 and H1N1 viruses. PLoS ONE 2010;5:e13162. [211] Kim MC, Song JMOE, Kwon YM, Lee YJ, Kang SM. Virus-like particles containing multiple M2 extracellular domains confer improved cross-protection against various subtypes of influenza virus. Mol Ther 2013;21:485–92. [212] Kim MC, Lee JS, Kwon YMOE, Lee YJ, Wang BZ, et al. Multiple heterologous M2 extracellular domains presented on virus-like particles confer broader and stronger M2 immunity than live influenza A virus infection. Antiviral Res 2013;99:328–35. [213] Heinen PP, Rijsewijk FA, de Boer-Luijtze EA, Bianchi AT. Vaccination of pigs with a DNA construct expressing an influenza virus M2-nucleoprotein fusion protein exacerbates disease after challenge with influenza A virus. J Gen Virol 2002;83:1851–9. [214] Marino M, Scuderi F, Provenzano C, Bartoccioni E. Skeletal muscle cells: from local inflammatory response to active immunity. Gene Ther 2011;18:109–16. [215] Nestle FO, Di Meglio P, Qin JZ, Nickoloff BJ. Skin immune sentinels in health and disease. Nat Rev Immunol 2009;9:679–91. [216] Amorij JP, Hinrichs W, Frijlink HW, Wilschut JC, Huckriede A. Needle-free influenza vaccination. Lancet Infect Dis 2010;10:699–711. [217] Lim YK, Takada A, Tanizaki T, Ozaki H, Okazakiand K, Kida H. Mucosal vaccination against influenza: protection of pigs immunized with inactivated virus and ether-split vaccine. Jpn J Vet Res 2001;48:197–203. [218] Fynan EF, Webster RG, Fuller DH, Haynes JR, Santoroand JC, Robinson HL. DNA vaccines: protective immunizations by parenteral, mucosal, and gene-gun inoculations. Proc Natl Acad Sci U S A 1993;90:11478–82. [219] Larsen DL, Olsen CW. Effects of DNA dose, route of vaccination, and coadministration of porcine interleukin-6 DNA on results of DNA vaccination against influenza virus infection in pigs. Am J Vet Res 2002;63:653–9. [220] Chen MW, Cheng TJ, Huang Y, Jan JT, Ma SH, Yu AL, et al. A consensushemagglutinin-based DNA vaccine that protects mice against divergent H5N1 influenza viruses. Proc Natl Acad Sci U S A 2008;105:13538–43. [221] Vilalta A, Jimenez G, Rusalov D, Planchon R, Lalor P, Carner K, et al. Vaccination with polymerase chain reaction-generated linear expression cassettes protects mice against lethal influenza A challenge. Hum Gene Ther 2007;18: 763–71. [222] Peeters B, Tonnis WF, Murugappan S, Rottier P, Koch G, Frijlink HW, et al. Pulmonary immunization of chickens using non-adjuvanted spray-freeze dried whole inactivated virus vaccine completely protects against highly pathogenic H5N1 avian influenza virus. Vaccine 2014. [223] OIE. Manual of Diagnostic Tests and Vaccines for Terrestrial Animals. 7th ed. Paris, France: International Office of Epizootics; 2012. [224] Peeters B, de Boer SM, Tjeerdsma G, Moormann R, Koch G. New DIVA vaccine for the protection of poultry against H5 highly pathogenic avian influenza viruses irrespective of the N-subtype. Vaccine 2012;30:7078–83. [225] Belshe R, Lee MS, Walker RE, Stoddard J, Mendelman PM. Safety, immunogenicity and efficacy of intranasal, live attenuated influenza vaccine. Expert Rev Vaccines 2004;3:643–54. [226] Paillot R, Hannant D, Kydd JH, Daly JM. Vaccination against equine influenza: quid novi? Vaccine 2006;24:4047–61. [227] Capua I, Marangon S, Dalla Pozza M, Santucci U. Vaccination for avian influenza in Italy. Vet Rec 2000;147:751. [228] Laddy DJ, Weiner DB. From plasmids to protection: a review of DNA vaccines against infectious diseases. Int Rev Immunol 2006;25:99–123. [229] Liniger M, Zuniga A, Naim HY. Use of viral vectors for the development of vaccines. Expert Rev Vaccines 2007;6:255–66.

Please cite this article in press as: Rahn J, et al. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine (2015), http://dx.doi.org/10.1016/j.vaccine.2015.03.052

Vaccines against influenza A viruses in poultry and swine: Status and future developments.

Influenza A viruses are important pathogens with a very broad host spectrum including domestic poultry and swine. For preventing clinical disease and ...
2MB Sizes 0 Downloads 6 Views