794
Views
11
CrossRef citations to date
0
Altmetric
Mini Review

Development of a universal CTL-based vaccine for influenza

, , &
Pages 374-378 | Received 11 Oct 2012, Accepted 10 Jan 2013, Published online: 21 Jan 2013

Abstract

In pursuit of better influenza vaccines, many strategies are being studied worldwide. An attractive alternative is the generation of a broadly cross-reactive vaccine based on the induction of cytotoxic T-lymphocytes (CTL) directed against conserved internal antigens of influenza A virus. The feasibility of this approach using recombinant viral vectors has recently been demonstrated in mice and humans by several research groups. However, similar results might also be achieved through immunization with viral proteins expressed in a prokaryotic system formulated with the appropriate adjuvants and delivery systems. This approach would be much simpler and less expensive. Recent results from several laboratories seem to confirm this is as a valid option to be considered.

Control of Seasonal and Pandemic Influenza

Seasonal influenza is an acute respiratory illness caused by influenza virus. This disease has a strong impact on public health worldwide causing, annually, 3 to 5 million cases of severe illness and between 250,000 and 500,000 deaths, mainly among children, elderly and immune-suppressed individuals (http://www.who.int/mediacentre/factsheets/fs211/en/). The best way to fight the impact of this disease is to vaccinate the population. Available vaccines are mostly inactivated ones, with a smaller proportion of live attenuated vaccines. Inactivated vaccines are produced mainly in embryonated chicken eggs and to a lesser extent in cell culture.Citation1,Citation2 Influenza vaccines induce protection in immunized individuals through the generation of neutralizing antibodies, mainly directed against the viral envelope glycoprotein hemagglutinin (HA). Virus antigenic variants arise constantly due to the high variability of the gene encoding the HA. Given the high rate of antigenic variation of the HA, antibodies that neutralize a subtype are often ineffective to neutralize other subtypes, and consequently the strains included in seasonal vaccines must be constantly updated. Vaccine efficiency severely diminishes when new strains emerge with antigenic changes in the virus envelope proteins, and situations where the antigenic matching between vaccine strains and the new circulating ones is sub-optimal are not exceptional. On rare occasions, completely new pandemic influenza variants arise, to which most of the human population has not been exposed. Under these circumstances, population immunity is low or null, allowing for an accelerated transmission of the new strain worldwide, which can have devastating consequences in terms of human lives. A recent analysis of the effectiveness of influenza vaccines holds that the immunity generated during certain seasons is at best moderate, when not significantly low or absent. In the ideal situation when antigenic matching between vaccines and circulating strains is optimal, average effectiveness was 69%.Citation3 During an outbreak of a pandemic strain there is a risk that the development of a vaccine for the emerging strain be too slow and, when available, come too late.

Improving the Performance of Current Vaccines

Much effort has been devoted to the improvement of current vaccines using different strategies, such as exploring new routes of vaccine administration, like oral,Citation4 intranasal,Citation5 or intradermal immunization,Citation6 new delivery systems like micro-needlesCitation7 or addition of adjuvants which might allow for an increase in the humoral and cellular responses,Citation8,Citation9 and a significant decrease in the dose of protective antigen needed.Citation10 It has also been reported that variations in the method of inactivation can significantly improve vaccine efficacy.Citation11,Citation12

Development of New Generation Vaccines

The main line of work in this field is focused on generating totally new vaccines using genetic engineering techniques. Conspicuous examples of this are: expression of the viral HA by means of recombinant vectors,Citation13-Citation15 production of virus-like particles (VLP) of influenza containing the influenza proteins HA and Matrix1 (M1),Citation16 production of recombinant HA subunit vaccines in insect cells through the baculovirus system,Citation17 and production of recombinant HA or its fusion with flagellin in Escherichia coli.Citation18,Citation19 DNA vaccines and peptide based vaccines have already been assayed in humans with promising results.Citation20,Citation21 While many of these strategies proved to be very efficient and in some cases induced significant increases in cross-reactive immune responses, they did not completely solve the problems derived from the high antigenic variability of influenza virus.

A Universal Vaccine for Influenza

This line of work is pursuing a “universal” type of vaccine, that is, a vaccine that can protect against almost all known subtypes of influenza, including pandemic strains.Citation22 In this regard three main strategies are being pursued:

Induction of neutralizing antibodies against highly conserved regions of the HA

The dogma that the influenza virus neutralization is mediated only by antibodies that bind to the globular head of the HA protein has been recently challenged. Several laboratories worldwide have generated broad spectrum human monoclonal antibodies capable of neutralizing the virus by binding to a highly conserved region of the HA (the stem or stalk domain.Citation23 Some of these monoclonal antibodies are capable of reacting with all the known specificities of HA, and it has been demonstrated that passive transfer of this kind of antibodies to mice and ferrets may protect against a challenge with heterologous strains.Citation24 However, it is difficult to find the appropriate antigen capable of efficiently inducing such antibodies in vivo after immunization.Citation25

An influenza A vaccine based on the ectodomain of the Matrix2 protein

The M2 protein (encoded in the same gene as the M1 protein) is a tetramer, functioning as an ion channel, and is present in very low amounts on the viral particle surface. The N-terminus of this protein (known as ectodomain or M2e) is highly conserved among strains of almost any origin. Based on this, M2e has been postulated as a very good candidate for the development of a universal vaccine. This assumption has been thoroughly demonstrated in pre-clinical models using various strategies.Citation26,Citation27 It was demonstrated that alveolar macrophages and Fc-receptors are fundamental for anti-M2e IgGs-mediated protection to occur.Citation28 Currently, the potential of an M2e-based universal vaccine is also being analyzed in human clinical trials.Citation29,Citation30 Although a vaccine based on the induction of anti-M2e antibodies is very promising, it will probably need to be combined with other conserved influenza antigens, able to elicit an adequate cellular response for a fully protective immunity.

A T-cell vaccine for influenza

The goal of a T-cell vaccine is to induce a strong response of specific CD4+ and CD8+ lymphocytes which may contribute to pathogen clearance by recognition and elimination of infected cells. For several important human infectious diseases, the efficacy of T-cells to induce therapeutic or prophylactic vaccines based on the use of replication-deficient viral vectors has already been established.Citation31 In the particular case of influenza, the generation of a non-sterilizing cellular based immunity is being sought; an immunity which would substantially decrease morbidity and mortality induced by the infection. Cytotoxic T- lymphocytes (CTL) are very effective in killing target cells by different mechanisms, thus eliminating the viruses from the infected organisms. In the case of influenza, it has been demonstrated that this type of immunity can protect mice from a lethal challenge with influenza A virus.Citation32

Candidate Proteins for a T-Cell Vaccine for Influenza

Unlike surface glycoproteins, proteins located within the influenza virion such as the NP and M1 are highly conserved due to their functional role during the viral replication cycle. It is well known that while they cannot induce neutralizing antibodies in infected or immunized animals, they are capable of inducing strong cellular immunity. For many years it has been known that mice recovering from infection with a certain subtype of influenza A virus have some protection against lethal challenge with a heterologous strain, and that the immunological basis of this phenomenon is mediated mainly by anti-influenza specific CTLs.Citation33,Citation34 These CTLs recognize highly conserved amino acid sequences of certain viral proteins, mainly proteins within the virus particle, exposed by the MHC class I pathway on the surface of infected cell.Citation35-Citation38 Although it has not been clearly established yet, there is some evidence of correlation between CTLs and protection, in humans.Citation39,Citation40

The cross-reactivity of polyclonal virus-specific CD8+ T-cell populations (obtained from european subjects), which target cells pulsed with H5N1-derived peptides or NP gene-transfected cells of the same avian influenza virus, demonstrates that human CTL response displays a high degree of cross-reactivity for very diverse influenza virus subtypes.Citation41

The concept that a vaccine formulated with a recombinant antigen unable to induce neutralizing antibodies can protect immunized mice from a lethal viral challenge was validated in the early 90s. Earlier experiences with a NP-based T-cell vaccine using purified recombinant NP (rNP) as a vaccine antigen indicated the validity of this concept,Citation42 which was further confirmed with a genetic vaccine containing a NP gene-carrying plasmid, which was able to protect mice against lethal challenges with an heterologous influenza virus.Citation43

Recombinant Vector Based T-Cell Vaccine for Influenza

Very recently, the use of recombinant adenovirus vectors expressing viral proteins NP and M2 (full length or M2e), showed that a T-cell vaccine against influenza could be extremely effective in mice.Citation44,Citation45 Furthermore, in a similar approach in a Phase I clinical trial, a modified Vaccinia virus Ankara (MVA) vector, encoding NP and M1, generated potent T-cell CD8+ specific immunity in immunized humans.Citation46 A Phase II clinical trial, conducted in healthy volunteers showed the efficacy of this vaccine to protect against flu symptoms after a challenge with live virus.Citation47

Development of a T-Cell Vaccine Based on Adjuvanted rNP Produced in a Prokaryotic System

Although influenza vaccine candidates based on recombinant vectors are very promising, it should be noted that safety trials for this strategy will take a long time before they are approved and massively available in the market. Furthermore, its production involves the handling of sophisticated and expensive technology not available in many developing countries. Therefore, it would be important to develop vaccines able to produce the same results but using simpler and less expensive production systems. Recently published results suggest that a NP based T-cell vaccine against influenza A could also be achieved using the recombinant protein produced in E. coli, which would be a far more simple and inexpensive system. In our laboratory, we have confirmed other group’s results, that is that the NP protein can be easily produced and purified in large quantities at low cost in E. coliCitation48-Citation50. The same appears true for other influenza proteins which are also candidates for a T-cell vaccine, such as M1 and M2.Citation50,Citation51

The main limitation of this approach is the difficulty to induce a strong CTL response in animals immunized with an exogenous protein.Citation31 However, exogenous proteins may induce a CTLs response by the phenomenon known as cross presentation,Citation52 and it is known that cross-presentation of an exogenous protein can be greatly stimulated with certain adjuvants which favor this process.Citation53 Recently, several reports confirmed the effectiveness of vaccines formulated with adjuvanted rNP to protect vaccinated animals against a lethal challenge of homologous or heterologous virus. Intranasal administration of cholera toxin combined with rNP protected against multiple viral subtypes.Citation49 The combination of recombinant NP and M2 formulated as liposomes stimulated a marked increase of specific CTLs and protected the vaccinated mice from a lethal challenge with the H5N1 avian strain.Citation50 A vaccine formulated with rNP and a TLR3 ligand, induced specific CTLs and protection against lethal challenge from influenza virus.Citation54 Fortunately, the knowledge on the possibilities of increasing the CTL responses to a recombinant protein is constantly increasing. A priori, there are multiple potential formulations that could lead to the optimization of a T-cell based rNP vaccine for influenza. In the pursuit of such a strategy, our laboratory has recently begun a systematic search of adjuvants able to promote a strong CTL response in animals vaccinated with rNP.

Iscomatrix Adjuvanted Influenza Vaccines

Iscomatrix (IMX) adjuvant is an immunostimulating system which also optimizes the process of antigen delivery, and is very efficient in obtaining a strong CTL response after immunization with exogenous protein.Citation55,Citation56 IMX consists of nanoparticles of about 40–50 nm in diameter with a strong negative charge, generated by the self assembly of phospholipids, cholesterol and saponins.Citation55 The negative charge of these particles favors their interaction with basic proteins such as the NP.Citation57 After immunization, the nanoparticles containing the antigen migrate to the draining lymph nodes, where they are captured and internalized by lymphoid resident dendritic cells (DC). IMX favors the process of extracellular antigen translocation from the endosome to the cytosol for proteasomal degradation. The processed peptides can then enter the major histocompatibility complex class I (MHC I) pathway, favoring the mechanism of cross presentation and the generation of CD8+ lymphocytes. The DCs that have taken up the particles are also activated, releasing many cytokines and lymphokines which stimulate the magnitude of the immune response. On the other hand, this system also has a strong stimulating activity on the humoral arm of the immune response.Citation56 This system has been successful when combined with inactivated influenza vaccines. It has proven very effective in stimulating mucosal immunity by the intranasal route.Citation58 It has also been effective in decreasing the minimum antigen dose required to obtain protection after pulmonary delivery of an influenza vaccine.Citation59 In mice, it was shown that IMX greatly improves the efficiency of a commercial vaccine increasing hetero-subtypic protection and CTL response.Citation60 In humans, a trivalent inactivated influenza vaccine formulated with IMX, elicited a sharp increase in the CTL response compared with those individuals that received the unadjuvanted vaccine.Citation61,Citation62 Rimmelzwaan et al.Citation62 found that IMX significantly increases the anti-HA CTL response, but not an anti-NP CTL response. This is different from what happens in vivo, where after infection with influenza virus, the anti NP CTL response is dominant. In our experience, analysis of the values of IgG subtypes and interleukins in the sera of mice that had been immunized with IMX-formulated rNP clearly indicated that the response obtained had a strong Th1 profile.Citation63 These experiments also showed the generation of high titers of IgG anti-NP. This should be also taken into account, since it has been demonstrated that specific high titers obtained after immunization of mice with rNP contributed to a rapid antibody-dependent elimination of the year 2009 H1N1 pandemic virus strain and that high anti NP titers correlated with an increase in the CD8+ response.Citation64,Citation65 These results strongly suggest that antibodies induced by immunization with rNP-IMX could also contribute, significantly, to a T-cell vaccine.

Are Adjuvanted Split Virus Vaccines Able to Induce a T-Cell Response?

As mentioned previously, Rimmelzwaan et al.Citation62 found that, contrary to what happens in infected individuals, the formulation of a split trivalent inactivated vaccine with IMX promotes the generation of CTL specific to HA but not NP. Lamere et al.Citation64 found that the lack of immunological reactivity of the endogenous NP contained in the split trivalent inactivated vaccine in mice, may be slightly improved by adding lipopolysaccharides to the vaccine formulation. In humans, preceding titers of specific anti NP IgG can be boosted in only few cases in subjects immunized with conventional trivalent vaccines.Citation64 In a similar way, Savard et al.Citation66 found that NP and M1 proteins present in split virion seasonal flu vaccine, are not immunogenic in immunized mice. However they showed that immunization of mice and ferrets with the same vaccine adjuvanted with papaya mosaic virus nanoparticles triggered a cell-mediated immune response to NP and M1, and long-lasting protection in animals challenged with a heterosubtypic influenza strain. Based on the above mentioned facts, there is some evidence that the endogenous NP contained in split influenza vaccines can be stimulated to produce a CTL response. However, the current processes of vaccine manufacturing are not validated to assess the content of NP in each batch, nor is it certain that the NP complexed with the genomic RNA in whole inactivated virus will be the most suitable antigen. On this basis it is tempting to hypothesize that for the purpose of generating an influenza specific CTL response, it would be more convenient to use recombinant NP, combined with a seasonal subunit vaccine.

Conclusions

In recent years the trend in the field of recombinant vaccines development has been the use of viral vectors when looking for a strong cellular responseCitation31 and purified proteins when looking for a humoral response.Citation67 The use of purified recombinant proteins for the development of vaccines against infectious agents that require a strong cellular response has been virtually neglected. However, our results and those of other laboratories confirm that it is possible to induce cell-mediated immune responses with purified proteins formulated with the appropriate adjuvants. Such formulations may even be improved by using particulate delivery systems. This type of methodology has been developed extensively in recent years and has also proved to be a very powerful method of inducing cellular immunity.Citation68-Citation70 In our laboratory we are currently developing delivery systems which include rNP in adjuvant loaded nanoparticles. In our work we have used rNP alone, however it is clear that the inclusion of other proteins with similar properties such as M1 and M2 is desirable. The production of vaccines based on this technology would be inexpensive due to their simplicity, and the technology could be certainly implemented in developing countries, which are now almost completely dependent on external sources of production. A vaccine of this type should be combined with the seasonal vaccine to elicit both robust influenza-specific antibody and CTL responses for maximal protection.

Disclosure of Potential Conflicts of Interest

The authors declare no conflict of interest.

References

  • Osterhaus A, Fouchier R, Rimmelzwaan G. Towards universal influenza vaccines?. Philos Trans R Soc Lond B Biol Sci 2011; 366:2766 - 73; http://dx.doi.org/10.1098/rstb.2011.0102; PMID: 21893539
  • Dormitzer PR, Tsai TF, Del Giudice G. New technologies for influenza vaccines. Hum Vaccin Immunother 2012; 8:45 - 58; PMID: 22251994
  • Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect Dis 2012; 12:36 - 44; http://dx.doi.org/10.1016/S1473-3099(11)70295-X; PMID: 22032844
  • Quan FS, Compans RW, Kang SM. Oral vaccination with inactivated influenza vaccine induces cross-protective immunity. Vaccine 2012; 30:180 - 8; http://dx.doi.org/10.1016/j.vaccine.2011.11.028; PMID: 22107852
  • Okamoto S, Matsuoka S, Takenaka N, Haredy AM, Tanimoto T, Gomi Y, et al. Intranasal immunization with a formalin-inactivated human influenza A virus whole-virion vaccine alone and intranasal immunization with a split-virion vaccine with mucosal adjuvants show similar levels of cross-protection. Clin Vaccine Immunol 2012; 19:979 - 90; http://dx.doi.org/10.1128/CVI.00016-12; PMID: 22552600
  • Dhont PA, Albert A, Brenders P, Podwapinska A, Pollet A, Scheveneels D, et al. Acceptability of Intanza® 15 μg intradermal influenza vaccine in Belgium during the 2010-2011 influenza season. Adv Ther 2012; 29:562 - 77; http://dx.doi.org/10.1007/s12325-012-0025-9; PMID: 22678831
  • Kang SM, Song JM, Kim YC. Microneedle and mucosal delivery of influenza vaccines. Expert Rev Vaccines 2012; 11:547 - 60; http://dx.doi.org/10.1586/erv.12.25; PMID: 22697052
  • O’Hagan DT, Ott GS, De Gregorio E, Seubert A. The mechanism of action of MF59 - an innately attractive adjuvant formulation. Vaccine 2012; 30:4341 - 8; http://dx.doi.org/10.1016/j.vaccine.2011.09.061; PMID: 22682289
  • Scheifele DW, Ward BJ, Dionne M, Vanderkooi OG, Loeb M, Coleman BL, et al, PHAC/CIHR Influenza Research Network (PCIRN). Compatibility of ASO3-adjuvanted H1N1pdm09 and seasonal trivalent influenza vaccines in adults: results of a randomized, controlled trial. Vaccine 2012; 30:4728 - 32; http://dx.doi.org/10.1016/j.vaccine.2012.05.029; PMID: 22652402
  • Cox RJ, Pedersen G, Madhun AS, Svindland S, Sævik M, Breakwell L, et al. Evaluation of a virosomal H5N1 vaccine formulated with Matrix M™ adjuvant in a phase I clinical trial. Vaccine 2011; 29:8049 - 59; http://dx.doi.org/10.1016/j.vaccine.2011.08.042; PMID: 21864624
  • Budimir N, Huckriede A, Meijerhof T, Boon L, Gostick E, Price DA, et al. Induction of heterosubtypic cross-protection against influenza by a whole inactivated virus vaccine: the role of viral membrane fusion activity. PLoS One 2012; 7:e30898; http://dx.doi.org/10.1371/journal.pone.0030898; PMID: 22303469
  • Furuya Y, Regner M, Lobigs M, Koskinen A, Müllbacher A, Alsharifi M. Effect of inactivation method on the cross-protective immunity induced by whole ‘killed’ influenza A viruses and commercial vaccine preparations. J Gen Virol 2010; 91:1450 - 60; http://dx.doi.org/10.1099/vir.0.018168-0; PMID: 20147516
  • Hessel A, Schwendinger M, Holzer GW, Orlinger KK, Coulibaly S, Savidis-Dacho H, et al. Vectors based on modified vaccinia Ankara expressing influenza H5N1 hemagglutinin induce substantial cross-clade protective immunity. PLoS One 2011; 6:e16247; http://dx.doi.org/10.1371/journal.pone.0016247; PMID: 21283631
  • Schwartz JA, Buonocore L, Suguitan A Jr., Hunter M, Marx PA, Subbarao K, et al. Vesicular stomatitis virus-based H5N1 avian influenza vaccines induce potent cross-clade neutralizing antibodies in rhesus macaques. J Virol 2011; 85:4602 - 5; http://dx.doi.org/10.1128/JVI.02491-10; PMID: 21325423
  • Steitz J, Barlow PG, Hossain J, Kim E, Okada K, Kenniston T, et al. A candidate H1N1 pandemic influenza vaccine elicits protective immunity in mice. PLoS One 2010; 5:e10492; http://dx.doi.org/10.1371/journal.pone.0010492; PMID: 20463955
  • Hossain MJ, Bourgeois M, Quan FS, Lipatov AS, Song JM, Chen LM, et al. Virus-like particle vaccine containing hemagglutinin confers protection against 2009 H1N1 pandemic influenza. Clin Vaccine Immunol 2011; 18:2010 - 7; http://dx.doi.org/10.1128/CVI.05206-11; PMID: 22030367
  • Treanor JJ, El Sahly H, King J, Graham I, Izikson R, Kohberger R, et al. Protective efficacy of a trivalent recombinant hemagglutinin protein vaccine (FluBlok®) against influenza in healthy adults: a randomized, placebo-controlled trial. Vaccine 2011; 29:7733 - 9; http://dx.doi.org/10.1016/j.vaccine.2011.07.128; PMID: 21835220
  • Khurana S, Verma S, Verma N, Crevar CJ, Carter DM, Manischewitz J, et al. Bacterial HA1 vaccine against pandemic H5N1 influenza virus: evidence of oligomerization, hemagglutination, and cross-protective immunity in ferrets. J Virol 2011; 85:1246 - 56; http://dx.doi.org/10.1128/JVI.02107-10; PMID: 21084473
  • Taylor DN, Treanor JJ, Sheldon EA, Johnson C, Umlauf S, Song L, et al. Development of VAX128, a recombinant hemagglutinin (HA) influenza-flagellin fusion vaccine with improved safety and immune response. Vaccine 2012; 30:5761 - 9; http://dx.doi.org/10.1016/j.vaccine.2012.06.086; PMID: 22796139
  • Ledgerwood JE, Wei CJ, Hu Z, Gordon IJ, Enama ME, Hendel CS, et al, VRC 306 Study Team. DNA priming and influenza vaccine immunogenicity: two phase 1 open label randomised clinical trials. Lancet Infect Dis 2011; 11:916 - 24; http://dx.doi.org/10.1016/S1473-3099(11)70240-7; PMID: 21975270
  • Atsmon J, Kate-Ilovitz E, Shaikevich D, Singer Y, Volokhov I, Haim KY, et al. Safety and immunogenicity of multimeric-001--a novel universal influenza vaccine. J Clin Immunol 2012; 32:595 - 603; http://dx.doi.org/10.1007/s10875-011-9632-5; PMID: 22318394
  • Roose K, Fiers W, Saelens X. Pandemic preparedness: toward a universal influenza vaccine. Drug News Perspect 2009; 22:80 - 92; http://dx.doi.org/10.1358/dnp.2009.22.2.1334451; PMID: 19330167
  • Yewdell JW. Viva la revolución: rethinking influenza a virus antigenic drift. Curr Opin Virol 2011; 1:177 - 83; http://dx.doi.org/10.1016/j.coviro.2011.05.005; PMID: 22034587
  • 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; http://dx.doi.org/10.1126/science.1205669; PMID: 21798894
  • 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; http://dx.doi.org/10.1073/pnas.1013387107; PMID: 20956293
  • Schotsaert M, De Filette M, Fiers W, Saelens X. Universal M2 ectodomain-based influenza A vaccines: preclinical and clinical developments. Expert Rev Vaccines 2009; 8:499 - 508; http://dx.doi.org/10.1586/erv.09.6; PMID: 19348565
  • Shim BS, Choi YK, Yun CH, Lee EG, Jeon YS, Park SM, et al. Sublingual immunization with M2-based vaccine induces broad protective immunity against influenza. PLoS One 2011; 6:e27953; http://dx.doi.org/10.1371/journal.pone.0027953; PMID: 22140491
  • El Bakkouri K, Descamps F, De Filette M, Smet A, Festjens E, Birkett A, et al. Universal vaccine based on ectodomain of matrix protein 2 of influenza A: Fc receptors and alveolar macrophages mediate protection. J Immunol 2011; 186:1022 - 31; http://dx.doi.org/10.4049/jimmunol.0902147; PMID: 21169548
  • Talbot HK, Rock MT, Johnson C, Tussey L, Kavita U, Shanker A, et al. Immunopotentiation of trivalent influenza vaccine when given with VAX102, a recombinant influenza M2e vaccine fused to the TLR5 ligand flagellin. PLoS One 2010; 5:e14442; http://dx.doi.org/10.1371/journal.pone.0014442; PMID: 21203437
  • Turley CB, Rupp RE, Johnson C, Taylor DN, Wolfson J, Tussey L, et al. Safety and immunogenicity of a recombinant M2e-flagellin influenza vaccine (STF2.4xM2e) in healthy adults. Vaccine 2011; 29:5145 - 52; http://dx.doi.org/10.1016/j.vaccine.2011.05.041; PMID: 21624416
  • Gilbert SC. T-cell-inducing vaccines - what’s the future. Immunology 2012; 135:19 - 26; http://dx.doi.org/10.1111/j.1365-2567.2011.03517.x; PMID: 22044118
  • Mbawuike IN, Zhang Y, Couch RB. Control of mucosal virus infection by influenza nucleoprotein-specific CD8+ cytotoxic T lymphocytes. Respir Res 2007; 8:44; http://dx.doi.org/10.1186/1465-9921-8-44; PMID: 17597533
  • Effros RB, Doherty PC, Gerhard W, Bennink J. Generation of both cross-reactive and virus-specific T-cell populations after immunization with serologically distinct influenza A viruses. J Exp Med 1977; 145:557 - 68; http://dx.doi.org/10.1084/jem.145.3.557; PMID: 233901
  • Yap KL, Ada GL, McKenzie IF. Transfer of specific cytotoxic T lymphocytes protects mice inoculated with influenza virus. Nature 1978; 273:238 - 9; http://dx.doi.org/10.1038/273238a0; PMID: 306072
  • Wraith DC, Askonas BA. Induction of influenza A virus cross-reactive cytotoxic T cells by a nucleoprotein/haemagglutinin preparation. J Gen Virol 1985; 66:1327 - 31; http://dx.doi.org/10.1099/0022-1317-66-6-1327; PMID: 3874261
  • Hurwitz JL, Hackett CJ, McAndrew EC, Gerhard W. Murine TH response to influenza virus: recognition of hemagglutinin, neuraminidase, matrix, and nucleoproteins. J Immunol 1985; 134:1994 - 8; PMID: 3155776
  • Yewdell JW, Bennink JR, Smith GL, Moss B. Influenza A virus nucleoprotein is a major target antigen for cross-reactive anti-influenza A virus cytotoxic T lymphocytes. Proc Natl Acad Sci U S A 1985; 82:1785 - 9; http://dx.doi.org/10.1073/pnas.82.6.1785; PMID: 3872457
  • Taylor PM, Askonas BA. Influenza nucleoprotein-specific cytotoxic T-cell clones are protective in vivo. Immunology 1986; 58:417 - 20; PMID: 2426185
  • McMichael AJ, Gotch FM, Noble GR, Beare PA. Cytotoxic T-cell immunity to influenza. N Engl J Med 1983; 309:13 - 7; http://dx.doi.org/10.1056/NEJM198307073090103; PMID: 6602294
  • Epstein SL. Prior H1N1 influenza infection and susceptibility of Cleveland Family Study participants during the H2N2 pandemic of 1957: an experiment of nature. J Infect Dis 2006; 193:49 - 53; http://dx.doi.org/10.1086/498980; PMID: 16323131
  • Kreijtz JH, de Mutsert G, van Baalen CA, Fouchier RA, Osterhaus AD, Rimmelzwaan GF. Cross-recognition of avian H5N1 influenza virus by human cytotoxic T-lymphocyte populations directed to human influenza A virus. J Virol 2008; 82:5161 - 6; http://dx.doi.org/10.1128/JVI.02694-07; PMID: 18353950
  • Tite JP, Hughes-Jenkins C, O’Callaghan D, Dougan G, Russell SM, Gao XM, et al. Anti-viral immunity induced by recombinant nucleoprotein of influenza A virus. II. Protection from influenza infection and mechanism of protection. Immunology 1990; 71:202 - 7; PMID: 2172156
  • 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; http://dx.doi.org/10.1126/science.8456302; PMID: 8456302
  • 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; http://dx.doi.org/10.1371/journal.pone.0013162; PMID: 20976273
  • Zhou D, Wu TL, Lasaro MO, Latimer BP, Parzych EM, Bian A, et al. A universal influenza A vaccine based on adenovirus expressing matrix-2 ectodomain and nucleoprotein protects mice from lethal challenge. Mol Ther 2010; 18:2182 - 9; http://dx.doi.org/10.1038/mt.2010.202; PMID: 20877342
  • Berthoud TK, Hamill M, Lillie PJ, Hwenda L, Collins KA, Ewer KJ, et al. Potent CD8+ T-cell immunogenicity in humans of a novel heterosubtypic influenza A vaccine, MVA-NP+M1. Clin Infect Dis 2011; 52:1 - 7; http://dx.doi.org/10.1093/cid/ciq015; PMID: 21148512
  • Lillie PJ, Berthoud TK, Powell TJ, Lambe T, Mullarkey C, Spencer AJ, et al. Preliminary assessment of the efficacy of a T-cell-based influenza vaccine, MVA-NP+M1, in humans. Clin Infect Dis 2012; 55:19 - 25; http://dx.doi.org/10.1093/cid/cis327; PMID: 22441650
  • Ye Q, Krug RM, Tao YJ. The mechanism by which influenza A virus nucleoprotein forms oligomers and binds RNA. Nature 2006; 444:1078 - 82; http://dx.doi.org/10.1038/nature05379; PMID: 17151603
  • Guo L, Zheng M, Ding Y, Li D, Yang Z, Wang H, et al. Protection against multiple influenza A virus subtypes by intranasal administration of recombinant nucleoprotein. Arch Virol 2010; 155:1765 - 75; http://dx.doi.org/10.1007/s00705-010-0756-3; PMID: 20652335
  • Thueng-in K, Maneewatch S, Srimanote P, Songserm T, Tapchaisri P, Sookrung N, et al. Heterosubtypic immunity to influenza mediated by liposome adjuvanted H5N1 recombinant protein vaccines. Vaccine 2010; 28:6765 - 77; http://dx.doi.org/10.1016/j.vaccine.2010.07.065; PMID: 20688037
  • Ebrahimi SM, Tebianian M, Aghaiypour K, Nili H, Mirjalili A. Prokaryotic expression and characterization of avian influenza A virus M2 gene as a candidate for universal recombinant vaccine against influenza A subtypes; specially H5N1 and H9N2. Mol Biol Rep 2010; 37:2909 - 14; http://dx.doi.org/10.1007/s11033-009-9851-5; PMID: 19809890
  • Neefjes J, Sadaka C. Into the intracellular logistics of cross-presentation. Front Immunol 2012; 3:31; http://dx.doi.org/10.3389/fimmu.2012.00031; PMID: 22566915
  • Dresch C, Leverrier Y, Marvel J, Shortman K. Development of antigen cross-presentation capacity in dendritic cells. Trends Immunol 2012; 33:381 - 8; http://dx.doi.org/10.1016/j.it.2012.04.009; PMID: 22677187
  • Jelinek I, Leonard JN, Price GE, Brown KN, Meyer-Manlapat A, Goldsmith PK, et al. TLR3-specific double-stranded RNA oligonucleotide adjuvants induce dendritic cell cross-presentation, CTL responses, and antiviral protection. J Immunol 2011; 186:2422 - 9; http://dx.doi.org/10.4049/jimmunol.1002845; PMID: 21242525
  • Wilson NS, Yang B, Morelli AB, Koernig S, Yang A, Loeser S, et al. ISCOMATRIX vaccines mediate CD8+ T-cell cross-priming by a MyD88-dependent signaling pathway. Immunol Cell Biol 2012; 90:540 - 52; http://dx.doi.org/10.1038/icb.2011.71; PMID: 21894173
  • Morelli AB, Becher D, Koernig S, Silva A, Drane D, Maraskovsky E. ISCOMATRIX: a novel adjuvant for use in prophylactic and therapeutic vaccines against infectious diseases. J Med Microbiol 2012; 61:935 - 43; http://dx.doi.org/10.1099/jmm.0.040857-0; PMID: 22442293
  • McBurney WT, Lendemans DG, Myschik J, Hennessy T, Rades T, Hook S. In vivo activity of cationic immune stimulating complexes (PLUSCOMs). Vaccine 2008; 26:4549 - 56; http://dx.doi.org/10.1016/j.vaccine.2008.06.024; PMID: 18585421
  • Coulter A, Harris R, Davis R, Drane D, Cox J, Ryan D, et al. Intranasal vaccination with ISCOMATRIX adjuvanted influenza vaccine. Vaccine 2003; 21:946 - 9; http://dx.doi.org/10.1016/S0264-410X(02)00545-5; PMID: 12547607
  • Wee JL, Scheerlinck JP, Snibson KJ, Edwards S, Pearse M, Quinn C, et al. Pulmonary delivery of ISCOMATRIX influenza vaccine induces both systemic and mucosal immunity with antigen dose sparing. Mucosal Immunol 2008; 1:489 - 96; http://dx.doi.org/10.1038/mi.2008.59; PMID: 19079216
  • Sambhara S, Kurichh A, Miranda R, Tumpey T, Rowe T, Renshaw M, et al. Heterosubtypic immunity against human influenza A viruses, including recently emerged avian H5 and H9 viruses, induced by FLU-ISCOM vaccine in mice requires both cytotoxic T-lymphocyte and macrophage function. Cell Immunol 2001; 211:143 - 53; http://dx.doi.org/10.1006/cimm.2001.1835; PMID: 11591118
  • Ennis FA, Cruz J, Jameson J, Klein M, Burt D, Thipphawong J. Augmentation of human influenza A virus-specific cytotoxic T lymphocyte memory by influenza vaccine and adjuvanted carriers (ISCOMS). Virology 1999; 259:256 - 61; http://dx.doi.org/10.1006/viro.1999.9765; PMID: 10388649
  • Rimmelzwaan GF, Nieuwkoop N, Brandenburg A, Sutter G, Beyer WE, Maher D, et al. A randomized, double blind study in young healthy adults comparing cell mediated and humoral immune responses induced by influenza ISCOM vaccines and conventional vaccines. Vaccine 2000; 19:1180 - 7; http://dx.doi.org/10.1016/S0264-410X(00)00310-8; PMID: 11137255
  • Cargnelutti DE, Sanchez MV, Alvarez P, Boado L, Glikmann G, Mattion N, et al. Improved immune response to recombinant influenza nucleoprotein formulated with ISCOMATRIX. J Microbiol Biotechnol 2012; 22:416 - 21; http://dx.doi.org/10.4014/jmb.1106.06021; PMID: 22450799
  • Lamere MW, Moquin A, Lee FE, Misra RS, Blair PJ, Haynes L, et al. Regulation of antinucleoprotein IgG by systemic vaccination and its effect on influenza virus clearance. J Virol 2011; 85:5027 - 35; http://dx.doi.org/10.1128/JVI.00150-11; PMID: 21367900
  • LaMere MW, Lam HT, Moquin A, Haynes L, Lund FE, Randall TD, et al. Contributions of antinucleoprotein IgG to heterosubtypic immunity against influenza virus. J Immunol 2011; 186:4331 - 9; http://dx.doi.org/10.4049/jimmunol.1003057; PMID: 21357542
  • Savard C, Guérin A, Drouin K, Bolduc M, Laliberté-Gagné ME, Dumas MC, et al. Improvement of the trivalent inactivated flu vaccine using PapMV nanoparticles. PLoS One 2011; 6:e21522; http://dx.doi.org/10.1371/journal.pone.0021522; PMID: 21747909
  • Duthie MS, Raman VS, Piazza FM, Reed SG. The development and clinical evaluation of second-generation leishmaniasis vaccines. Vaccine 2012; 30:134 - 41; http://dx.doi.org/10.1016/j.vaccine.2011.11.005; PMID: 22085553
  • Tacken PJ, Zeelenberg IS, Cruz LJ, van Hout-Kuijer MA, van de Glind G, Fokkink RG, et al. Targeted delivery of TLR ligands to human and mouse dendritic cells strongly enhances adjuvanticity. Blood 2011; 118:6836 - 44; http://dx.doi.org/10.1182/blood-2011-07-367615; PMID: 21967977
  • Nembrini C, Stano A, Dane KY, Ballester M, van der Vlies AJ, Marsland BJ, et al. Nanoparticle conjugation of antigen enhances cytotoxic T-cell responses in pulmonary vaccination. Proc Natl Acad Sci U S A 2011; 108:E989 - 97; http://dx.doi.org/10.1073/pnas.1104264108; PMID: 21969597
  • Foged C, Hansen J, Agger EM. License to kill: Formulation requirements for optimal priming of CD8(+) CTL responses with particulate vaccine delivery systems. Eur J Pharm Sci 2012; 45:482 - 91; http://dx.doi.org/10.1016/j.ejps.2011.08.016; PMID: 21888971

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.