You are on page 1of 8

Iranian Journal of Virology 2017;11(3): 33-40

©2017, Iranian Society of Virology. All rights reserved.

Review Article
Challenges and Perspectives towards the Development of more
Effective Influenza Vaccine
Downloaded from journal.isv.org.ir at 2:33 +0430 on Saturday May 2nd 2020

Ahmadi M, Shahsavandi S*
1. Razi Vaccine & Serum Research Institute, Agricultural Research Education and Extension Organization, Karaj,
Iran.

Abstract
Influenza viruses continue to be a major health threat in human and bird populations. The
improvements in formulation and production level of the current influenza vaccines are not
sufficient to afford complete protection. The continuous antigenic drifts and emergence of
endemic and zoonotic strains make influenza vaccine planning difficult. Concern about the
emergence of new influenza pandemic provides subject for developing a universal influenza
vaccine to be most effective in preventing influenza A either by targeting the HA or other
viral proteins. The recombinant and synthetic antigens used in influenza vaccine research and
development are generally less immunogenic and need to incorporate novel adjuvants with
modified delivery carriers to develop broad-protective vaccines.
Keywords: Influenza virus, vaccine, adjuvant, delivery system.

Introduction* enable the influenza viruses to undergo


antigenic shift and drift (3-5) the two possible
mechanisms for antigenic evolution.

R
genomes.
ecombination in RNA viruses involves
the exchange of genetic information
between two nonsegmented RNA
Since the first detected
recombination in poliovirus in the early 1960s,
Antigenic drifts or small changes in the genes
of viruses are happening continually in the
types of influenza viruses. These gradual and
subtle mutations produce in the two surface
the major evolutionary significance occurs in glycoproteins, HA and NA may lead to a loss
many RNA viruses that plays an important role of immunity, or in vaccine mismatch (6-8).
in viral biology (1). The influenza A virus The antigenic shift is the process by which at
genomes comprise eight negative-sense, least two different strains or different influenza
single-stranded RNA segments, which are A viruses combine to form a new subtype. The
numbered in order of decreasing length. The recombine or reassorted virus, which has a
polymerase subunit 2 or PB2, PB1 (in some mixture of the surface antigens of the two
strains this segment also codes for the original strains, may follow to enter a new
accessory protein PB1-F2), PA, the strongly niche and transmit directly from animal to
immunogenic surface protein HA, NP, NA, M1 human (9-11). The host shifts have resulted in
(the M2 ion channel is also expressed from human pandemics, such the H1N1 “Spanish
segment 7 by RNA splicing), NS1 and flu” (1918-1920) killed an estimated 50 million
NEP/NS2 by mRNA splicing (2). The people or more globally (12). The causative
segmented genome structure and the error- agent was an avian-descended H1N1 virus and
prone RNA-dependent RNA polymerases a direct progenitor of all of the influenza A
viruses circulating in humans today. Along
with the Asian flu (1957) and Hong Kong flu
: (1963) human pandemics, the avian H5N1
*Corresponding author s.shahsavandi@rvsri.ac.ir
Tel: +98 26 34570038, Fax: +98 26 4552194

Iranian Journal of Virology, Volume 11, Number 3, 2017 33


Challenges and perspectives toward development of more effective influenza vaccine

viruses caused devastating outbreaks in 2004 efficacy, limited worldwide vaccine


in domestic poultry worldwide and posed a availability, and lack of cross-reactivity. The
major challenge to human health (13-16). The seasonal vaccine is effective in eliciting
recent 2009 H1N1 pandemic virus with 0.03% protection when the vaccine strains and the
case fatality rate was derived by reassortment circulating viral are closely antigenic matched
between a triple reassortant lineage of North together. Due to frequent antigenic drifts,
American swine and a Eurasian H1N1 swine seasonal influenza vaccines need to be re-
Downloaded from journal.isv.org.ir at 2:33 +0430 on Saturday May 2nd 2020

lineage virus (11, 17). formulated regularly to match the circulating


The influenza researches into the genes of the strains. During each influenza season, Centers
recombined viruses as well as the circulated for Disease Control and Prevention (CDC) has
avian subtypes such as H9N2, H5N1 and estimated the effectiveness of the used vaccine.
H7N9 show they have genes may adapt to Based on the data from children and adults
human (18-22). Thus, the viruses have to be enrolled in the U.S. during 2004-2018
considered a potentially serious pandemic (www.cdc.gov/flu), the vaccine effectiveness
threat. against influenza A and influenza B virus
Traditional influenza virus vaccines infection was 36% (95% confidence interval
and challenges [CI]=27%–44%). Most (69%) influenza
Vaccination against influenza is the most infections were caused by H3N2 viruses. The
effective way to prevent morbidity and vaccine effectiveness was estimated to be 25%
mortality in the target risk groups. Influenza (CI=13% to 36%) against H3N2 virus, 67%
virus vaccines comprise whole inactivated (CI=54%–76%) against H1N1 pdm09 viruses,
virus, split, virosomal, or subunit antigen are and 42% (CI=25%–56%) against influenza B
used in widespread annual influenza viruses. The best overall estimated
vaccination campaigns (23, 24). The vaccine effectiveness was 60% for 2010-2011
production platforms are including influenza seasons. However the vaccine
embryonated eggs licensed worldwide, Madin effectiveness confirm the value of influenza
Darby canine kidney (MDCK) and monkey vaccination as a public health intervention, the
kidney (Vero) cells licensed in EU and USA, results are vary based on study design,
and trivalent vaccine of recombinant HA outcome measured, population and the season
produced in baculovirus licensed in USA and in which the influenza vaccine was studied.
Japan. The egg-based production system has Because the seasonal vaccine may provide
been used for more than 60 years, and it is still limited protection in cases of antigenic
the most extensively used method to generate mismatch, the quadrivalent vaccine consists of
the influenza vaccine. In cell-based production H3N2 and H1N1, and two distinct influenza B
system, viral seeds recommended for the lineages was also licensed for induction a
seasonal or epidemic vaccination are separately better immunogenicity (27, 28).
expanded in each of the mammalian cells and WHO has recommended that quadrivalent
pooled to formulate the vaccine (25). vaccines for use in the 2018-2019 northern
For more than 50 years, World Health hemisphere influenza season contain
Organization (WHO) has been collaborating A/Michigan/45/2015 (H1N1) pdm09-like
with influenza vaccine development to identify virus; A/Singapore/INFIMH-16-0019/2016
the dominant circulating strains during the (H3N2) like virus; B/Colorado/06/2017-like
previous and the following vaccination seasons virus (B/Victoria/2/87 lineage); and
in the northern and southern hemispheres. The B/Phuket/3073/2013-like virus
traditional “trivalent” vaccines are formulated (B/Yamagata/16/88 lineage)
based on an influenza A (H1N1) virus, an (www.who.int/influenza/vaccines).
influenza A (H3N2) virus, and an influenza B The reformulation is only useful for battling
virus (26). Despite the marketing of influenza seasonal influenza epidemics and remind
vaccines since the 1930s, several limitations ineffective in preventing unexpected new
still involve their effectiveness include limited influenza pandemic.

34 Iranian Journal of Virology, Volume 11, Number 3, 2017


Ahmadi M et al

In the last decade, the emergence of H1N1 disruption viral membrane fusion it should be
pandemic and outbreaks of various avian determined whether these antibodies are
influenza viruses have been a threat to the suitable for protection against influenza
effectiveness of the current vaccines and an infection (34-36).
alarm for public health organizations (29, 30). Beside the development of vaccines inducing
On the other hand, zoonotic influenza viruses HA stalk-reactive antibodies, the M2e-targeted
continue to be identified and evolve both vaccines attracted the attention of researchers
Downloaded from journal.isv.org.ir at 2:33 +0430 on Saturday May 2nd 2020

genetically and antigenically, leading to the for developing a universal influenza vaccine or
need to select and develop additional candidate a vaccine that provides robust, long-lasting
vaccine viruses for pandemic preparedness protection. M2e is a potential target for cross-
purposes (18). These events as well as the reactive immune responses among all known
increased demand for effective coverage influenza A viruses that circulated between
against the infection are great challenges to 1918 to now. In contract to the stalk domain of
vaccine production. Most countries without HA, M2e vaccines do not prevent viral
vaccine production facilities will have no infection but efficiently inhibit viral replication
access to the effective vaccines during the first once inside the host. M2e itself has low
pandemic wave. All these challenges call immunogenicity and need fused with a carrier
researchers for the development of the proper vehicle to enhance anti-M2e immune
influenza vaccines capable of conferring broad responses. The impact of various viral and
cross-protection against multiple subtypes of bacterial carriers for increasing the
influenza A viruses (31). The current in immunogenicity of M2e was tested in animal
development researches can be divided into the models (37, 38). The studies on the protective
various categories: elicit antibody responses to efficacies by M2e-based vaccination suggest
structurally conserved regions of the stalk that M2e immunity-mediated cross protection
domain of HA and M2 ectodomains (M2e); is relatively weak and need to become
induce cross-protective T-cell responses sufficiently immunogenic.
against internal proteins like nucleoprotein T cell-inducing influenza vaccines
(NP) and matrix 1 (M1); and improved Cell-mediated immune responses play an
adjuvants and delivery systems. important role in the cross-protective immune
Universal influenza vaccines response against influenza virus. It is well
Upon influenza infection virus-specific T cell documented that individuals who possessed
responses including CD4+ T helper cells and preexisting CTLs cells displayed decreased
CD8+ cytotoxic T cells (CTLs) are induced. morbidity after infection with pandemic H1N1
These cells are primarily targeted to induce influenza, or patients with early CTL responses
immunity to the HA antigen then directed to recovered quickly from H7N9 infection (39).
conserved proteins (32). Among the viral The half-life of circulating T cells specific to
antigen, HA is considered as most (or highly) influenza calculated at least 2 years, so a
immunogenic. The molecule comprises the central memory response can be maintained for
head, an immuno-dominant and the primary many years following recovery from the
target for currently licensed influenza vaccines, infection (40). The induction of influenza-
and stalk regions. The 18 HAs are specific cellular responses might be a great
phylogenetically divided into two groups based addition to current antibody-inducing influenza
on similarities in their stalk regions. Thus, the vaccines. The naturally acquired immune
portion displays cross-reactivity with different responses to influenza include CTLs
subtypes (33). The conserved feature of HA recognizing antibodies to HA as well as
stalk domain makes it an attractive target for conserved internal viral antigens. Most highly
the induction of a cross-reactive humoral conserved T cell epitopes are located on NP
response. However, HA stalk-reactive and M1 proteins, which role in preventing
antibodies prevent infection by inhibiting the infection or reducing disease severity (41). As
virus attachment to host cell membrane or a novel vaccine concept based on the induction

Iranian Journal of Virology, Volume 11, Number 3, 2017 35


Challenges and perspectives toward development of more effective influenza vaccine

of influenza-specific T cells, the efficacy of a boost the immune response to the vaccine. The
recombinant replication-deficient poxvirus components act by diverse mechanisms which
vector was assessed in an effort to boost T-cell include enhancing the delivery of antigen,
responses to these internal antigens. However, improving magnitude of the immune response,
a large expansion in circulating influenza- directing antigen presentation by the major
specific T cells was observed after a single histocompatibility complex or providing
vaccination (42), it seems that several immune stimulatory signals that potentiate the
Downloaded from journal.isv.org.ir at 2:33 +0430 on Saturday May 2nd 2020

formulation strategies should utilize to immune response (46, 47). Generally,


successfully induce specific T cell responses to inactivated vaccines should formulate with
the non immunogenic antigens. This approach adjuvants to augment the potency of the
is valuable both for priming and boosting an antigen and presentation to the immune
immune response. system.
Antigenic peptides-based influenza The oil-in-water emulsions are approved as
vaccines suitable adjuvants for influenza vaccines.
Multiple antigenic peptide constructs which MF59, the first oil-in-water emulsion adjuvant
induce both influenza-specific immune B-cell approved for formulation of influenza
and T cell responses against conserved vaccines, is biodegradable squalene oil
epitopes are used for enhancing the droplets stabilized by non-ionic surfactants.
immunogenicity of peptide antigens. A peptide The mechanism actions attribute to MF59 are
construct consists of nine linear B cell and T enhance regulation of genes for cytokines and
cell epitopes of HA, NP and M1, named chemokines, increase influx of macrophages
Multimeric-001 was able to induce and monocytes to the site of injection,
considerable cellular immune responses when differentiation of monocytes to active dendritic
administered twice in both adults and elderly cells, and antigen transportation to draining
(25). This approach has also been used in M2e- lymph nodes (48, 49). AS03 is another oil-in-
derived antigens and in a prime-boost water emulsion based on squalene droplets and
immunization with seasonal traditional only used in pandemic influenza vaccines (50).
inactivated influenza vaccine (43, 44). A prime Impact of saponin-based adjuvants such as
with Multimeric-001 and subsequently immune stimulating complexes (ISCOMs) and
boosting with seasonal vaccine had Matrix-M has been evaluated in clinical studies
significantly higher HI titers compared to in combination with influenza vaccines. The
individuals who received the inactivated ISCOMs-adjuvanted influenza split vaccines
vaccine at both prime and boost. Despite these revealed increase of influenza-specific in
promising results, further formulation with humoral and CD8+T cell responses in
adjuvants might increase the immunogenicity vaccinated individuals. The addition of Matrix-
of Multimeric-001 vaccine in the future. M to virosomal influenza vaccine resulted in
increased vaccine-induced T cell responses
Adjuvants
(51). The third generation of saponin based
The HA stalk domain, M2e- and T cell-based
adjuvant was used for a H7N9 virus-like
influenza vaccine candidates having completed
particle vaccine and showed significantly
phase II trials and will enter phase III trials in
higher seroconversion rates after vaccination.
the coming years. But only the covalently
The co-administration of bacterial-derived
bound M2e antigen to a carrier protein or
components, flagellin and heat-labile
adjuvant could induce potent cross-protective
enterotoxin are potent adjuvants for influenza
immune responses (45). To overcome limited
vaccines and able to induce protective
efficacy of the current influenza vaccines and
hemagglutination inhibition (HI) titers after
also the future universal vaccine, advanced
immunization with influenza vaccines (52).
formulation with adjuvants both
Peptide antigens generally suffer from poor
immunopotentiator and delivery system are
immunogenicity and need adding adjuvant to
considered. For more than 70 years adjuvants
induce influenza-specific cellular responses.
have been added to vaccine formulations to

36 Iranian Journal of Virology, Volume 11, Number 3, 2017


Ahmadi M et al

NP peptide encapsulated in cationic liposomes within the nanoparticles decreases the risk of
could induce potent T cell responses (53) and degradation of antigen (67, 68). A variety of
HLA-A2.1 and HLA-A24.2 restricted peptides polycationic polymeric nanoparticles and their
conjugated to liposomes were able to induce derivatives explored to stimulate immune
CD8+ memory T cells in influenza-infected system. Among them, chitosan is preferred due
mice (54). The same results were found in to its ability in enhancing antigen uptake by
conjugation of PA-derived peptide to Pam2Cys mucosal lymphoid tissues, and inducing
Downloaded from journal.isv.org.ir at 2:33 +0430 on Saturday May 2nd 2020

(a bacterial lipopeptide) (55), and influenza strongly immune responses against the
peptides conjugated to phosphatidylserine (56). antigens (69-71).
With the advent of novel concepts for Utilization of chitosan nanoparticles facilitate
immunity against influenza, novel types of the delivery of a vaccine to the targeted cells
adjuvants such as cytokines, type I interferon, but some researches focus on the ability of the
the tumor necrosis factor, and bacterial polymer as a novel adjuvant to induce specific
derivatives have been evaluated to induce antibody titer against influenza antigens. For
stronger responses as well as CTL-mediated instance, reduced markedly the influenza
immunity against influenza infection (57, 58) morbidity and also complete protection against
are under development. Several studies challenge mice vaccinated with chitosan
suggested that molecular adjuvants such as nanoparticle encapsulated HA-split vaccine
myxovirus resistance (Mx) protein (35), (72) or enhanced immunogenicity of H9N2
Hemokinin-1 (HK-1) (59, 60), unmethylated influenza nanovaccine in chickens in
bacterial CpG motifs (61), Mycobacterium- comparison to an oil adjuvant (73). There is no
dependent protein-1 (62), HSP 70 (63), and doubt that chitosan nanoparticles can stimulate
ESAT-6 (64) might act better by enhanced early immune response and enhance the
antigen persistence, increased antigen uptake, specific HI titer against influenza antigens, but
and recruitment of antigen presenting cells the boost delivery system role of the
(APCs) compared with the current adjuvants. polysaccharide is also extremely
These experimental approaches support recommended. The highly induction of
improvement of adjuvant may be an important immune system and long-lived response are
route to obtaining better protecting influenza invaluable properties of a vaccine thus adding
vaccines. a number of adjuvants to nanovaccines are
Nano particles delivery system highly suggested to enhance the induction of
The delivery of vaccine antigens to dendritic the immune responses (74, 75). Influenza is
cells is critical to the improvement of a still one of the most wide-reaching and deadly
protective immune response. Most current infectious diseases and development of a more
inactivated vaccines are usually administered effective vaccine imply that novel adjuvants
via intramuscular injection may fail to reach correlate of the delivery system need to be
the APCs and induction of immune responses established.
(65). To overcome the difficulty in targeting
the APCs new approaches are being References
investigated to apply more effective vaccine 1. Dermody T, Parker J, Sherry B.
delivery methods (66). Many antigen and Orthoreoviruses. Fields Virology. 2013; 2:1304-46.
adjuvant delivery carriers have been 2. Simon-Loriere E, Holmes EC. Why do RNA
considered for enhancing the efficiency of the viruses recombine? Nat Rev Microbiol 2011;
application of vaccines. Recent studies have 9:617–626.
3. Landolt GA, Olsen CW. Up to new tricks—A
demonstrated that antigen delivery via review of cross-species transmission of influenza A
nanoparticle formulations can significantly viruses. Anim Health Res Rev. 2007; 8:1–21.
improve immunogenicity of vaccines due to 4. Rabadan R, Robins.H. Evolution of the
either intrinsic immunostimulatory properties influenza A virus: some new advances. Evol
of the components or by co-entrapment of Bioinform Online. 2007; 3:299–307.
molecular adjuvants. Encapsulating the antigen

Iranian Journal of Virology, Volume 11, Number 3, 2017 37


Challenges and perspectives toward development of more effective influenza vaccine

5. Boni MF. Vaccination and antigenic drift in H9N2 virus increases mammalian replication and
influenza. Vaccine. 2008; 26 (Suppl 3): C8–14. pathogenicity. J Virol. 2016; 90:8105-81014.
6. Schweiger B, Bruns L, Meixenberger K. 20. Arai Y, Kawashita N, Daidoji T, Ibrahim MS,
Reassortment between human A (H3N2) viruses is El-Gendy EM, Takagi T, et al. Novel Polymerase
an important evolutionary mechanism. Vaccine. Gene Mutations for Human Adaptation in Clinical
2006; 24:6683–90. Isolates of Avian H5N1 Influenza Viruses. PLoS
7. Gamblin SJ, Skehel JJ. Influenza hemagglutinin Pathog. 2016; 12(4): e1005583.
and neuraminidase membrane glycoproteins. J Biol 21. Crusat M, Liu J, Palma AS, Childs RA, Liu Y,
Downloaded from journal.isv.org.ir at 2:33 +0430 on Saturday May 2nd 2020

Chem. 2010; 285:28403-28409. Wharton SA. Changes in the hemagglutinin of


8. Ren XW, Ju LW, Yang JX, Lv XH, Jiang LF, H5N1 viruses during human infection–influence on
Zhao NQ, Jiang QW. Antigenic and genetic receptor binding. Virology. 2013; 447:326–37.
variation in the hemagglutinins of H1N1 and H3N2 22. Imai M, Watanabe T, Kiso M, Nakajima N, et
human influenza a viruses in the Shanghai area al. A highly pathogenic avian H7N9 influenza virus
from 2005 to 2008. J Med Virol. 2011; 83:1113-20. isolated from a human is lethal in some ferrets
9. Doherty PC, Turner SJ, Webby RG, Thomas infected via respiratory droplets. Cell Host Microl;
PG. Influenza and the challenge for immunology. 22:615–626.
Nature immunology. 2006; 7:449-455. 23. Kreijtz JH1, Fouchier RA, Rimmelzwaan GF.
10. Taubenberger JK, Kash JC. Influenza virus Immune responses to influenza virus infection.
evolution, host adaptation and pandemic formation. Virus Res. 2011; 162(1-2):19-30.
Cell Host Microbe. 2010; 7: 440–451. 24. Wong S-S, Webby RJ. Traditional and new
11. Garten RJ, Davis CT, Russell CA, Shu B, influenza vaccines. Clinical microbiology
Lindstrom S, Balish A, Sessions WM, Xu X, reviews.2013; 26:476-492.
Skepner E, Deyde V, et al. Antigenic and genetic 25. Soema PC, Kompier R, Amorij JP, et al.
characteristics of swine-origin 2009 A (H1N1) Current and next generation influenza vaccines:
influenza viruses circulating in humans. Science. Formulation and production strategies. Eur J
2009; 325:197–201. Pharma Biopharma. 2015, 94:251-263.
12. Johnson NP, Mueller J. Updating the accounts: 26. Palese P, García-Sastre A. Influenza vaccines:
global mortality of the 1918–1920 “Spanish” present and future. J Clin Invest. 2002; 110: 9–13.
influenza pandemic. Bull History Med. 2002; 27. Treanor JT, Albano FR, Sawlwin DC, Graves
76:105–115. Jones A, Airey J, Formica N, Matassa V, Leong J.
13. Kilbourne ED. Influenza pandemics of the 20th Immunogenicity and safety of a quadrivalent
century. Emerg Infect Dis. 2006; 12: 9–14. inactivated influenza vaccine compared with two
14. Morens DM, Taubenberger JK, Fauci AS. The trivalent inactivated influenza vaccines containing
persistent legacy of the 1918 influenza virus. N alternate B strains in adults: A phase 3, randomized
Engl J Med. 2009; 361:225–229. noninferiority study. Vaccine. 2017; 35:1856-1864.
15. Kash JC, Tumpey TM, Proll SC, Carter V, 28. Beran J, Peeters M, Dewé W, Raupachová J,
Perwitasari O, Thomas MJ, Basler CF, Palese P, Hobzová L, Devaster J-M. Immunogenicity and
Taubenberger JK, Garcia-Sastre A, et al. Genomic safety of quadrivalent versus trivalent inactivated
analysis of increased host immune and cell death influenza vaccine: a randomized, controlled trial in
responses induced by 1918 influenza virus. Nature. adults. BMC infectious diseases. 2013; 13:224.
2006; 443:578–581. 29. Beyer WEP, Nauta, JJP, Palache AM,
16. Taubenberger JK, Morens DM. Pandemic Giezeman KM, Osterhaus ADME. Immunogenicity
influenza-including a risk assessment of H5N1. and safety of inactivated influenza vaccines in
Rev Sci Tech. 2009; 28: 187–202. primed populations: A systematic literature review
17. Mak GC, Wong, AH, Ho WY, Lim W. The and meta-analysis. Vaccine. 2011; 29: 5785-5792.
impact of pandemic influenza A (H1N1) 2009 on 30. Castilla J, Navascués A, Alonso MF, et al.
the circulation of respiratory viruses 2009–2011. Effectiveness of subunit influenza vaccination in
Influenza Other Respir Viruses. 2012; 6:6-10. the 2014–2015 season and residual effect of split
18. Shahsavandi S, Salmanian A-H, Ghorashi SA, vaccination in previous seasons. Vaccine. 2016;
Masoudi S, Ebrahimi MM. Evolutionary 34:1350-1357.
characterization of hemagglutinin gene of H9N2 31. de Vries RD, Altenburg AF, Rimmelzwaan GF.
influenza viruses isolated from Asia. Research in Universal influenza vaccines, science fiction or
Veterinary Science. 2012; 93: 234-249. soon reality? Expert Rev Vaccines. 2015; 14:1299-
19. Xu G, Zhang X, Gao W, Wang C , Wang J. 1301.
Prevailing PA mutation K356R in avian influenza

38 Iranian Journal of Virology, Volume 11, Number 3, 2017


Ahmadi M et al

32. Kreijtz JH, Fouchier RA, Rimmelzwaan GF. implications for vaccine design and production.
Immune responses to influenza virus infection. Hum Vaccin Immunother. 2016; 12:763–767.
Virus Res. 2011; 162:19-30. 46. Even-Or O, Samira S, Ellis R, Kedar E,
33. Shahsavandi S, Salmanian AH, Ghorashi SA, Barenholz Y. Adjuvanted influenza vaccines.
Masoudi S, Fotouhi F, Ebrahimi MM. Specific Expert Review of Vaccines. 2013; 12: 1095-1108.
subtyping of influenza A virus using a recombinant 47. Fox CB, Kramer RM, Barnes L, Dowling QM,
hemagglutinin protein expressed in baculovirus. Vedvick TS. Working together: interactions
Mol Boil Rep. 2011; 38:3293–3298. between vaccine antigens and adjutants.
Downloaded from journal.isv.org.ir at 2:33 +0430 on Saturday May 2nd 2020

34. Krammer F, Palese P. Influenza virus Therapeutic Advances in Vaccines. 2013; 1: 7-20.
hemagglutinin stalk-based antibodies and vaccines. 48. Banzhoff A, Haertel S, Praus M. Passive
Curr Opin Virol. 2013; 3:521-530. surveillance of adverse events of an MF59-
35. Soleimani S, Shahsavandi S, Madadgar O. adjuvanted H1N1 vaccine during the pandemic
Improvement influenza HA2 DNA vaccine cellular mass vaccinations. Human vaccines.2011; 7:539-
and humoral immune responses with Mx bio 548.
adjuvant. Biologicals. 2017; 46:6-10. 49. Clark TW, Pareek M, Hoschler K, Dillon H,
36. Krammer F, Pica N, Hai R, Margine I, Palese P. Nicholson KG, Groth N, Stephenson I .Trial of
Chimeric hemagglutinin influenza virus vaccine 2009 influenza A (H1N1) monovalent MF59-
constructs elicit broadly protective stalk-specific adjuvanted vaccine. N Engl J Med. 2009;
antibodies. J Virol. 2013; 87:6542-50. 361:2424-2435.
37. Rappazzo CG et al. Recombinant M2e outer 50. Wilkins AL, Kazmin D, Napolitani G,
membrane vesicle vaccines protect against lethal Clutterbuck EA, Pulendran B, Siergrist C-A,
Influenza A challenge in BALB/c mice. Vaccine. Pollard AJ. AS03 and MF59 adjuvanted influenza
2016; 34:1252-1258. vaccines in children. Frontiers in Immunology.
38. Lee YN, Kim MC, Lee YT, Kim YJ, Kang SM. 2017; 8:1760 10.
Mechanisms of cross-protection by influenza virus 51. Lövgren K, Morein B, Osterhaus AD. ISCOM
M2-based vaccines. Immune Net. 2015; 15:213- technology-based Matrix M™ adjuvant: Success in
221. future vaccines relies on formulation. Expert Rev
39. Gilbert SC. T-cell-inducing vaccines – what's Vaccines. 2011; 10:401-403.
the future. Immunology. 2012; 135: 19–26. 52. Wang B-Z, Xu R, Quan F-S, Kang S-M, Wang
40. Wang Z, Wan Y, Qiu C, Quiñones-Parra S. L, Compans RW. Intranasal Immunization with
Recovery from severe H7N9 disease is associated Influenza VLPs Incorporating Membrane-
with diverse response mechanisms dominated by Anchored Flagellin Induces Strong Heterosubtypic
CD8+ T cells. Nat Commun. 2015; 6: 6833. Protection. PLoS ONE. 2010; 5(11): e13972.
41. Ben-Yedidia T, Arnon R. Epitope-based vaccine 53. Taneichi M, Tanaka Y, Kakiuchi T, Uchida T.
against influenza. Expert Review of Vaccines Liposome-coupled peptides induce long-lived
2007; 6: 939-48. memory CD8 T cells without CD4 T cells. PLoS
42. Antrobus RD, Lillie PJ, Berthoud TK, Spencer One. 2010; 5:e15091.
AJ, McLaren JE, Ladell K, et al. A T Cell-Inducing 54. Matsui M, Kohyama S, Suda T, Yokoyama S,
Influenza Vaccine for the Elderly: Safety and Mori M, Kobayashi A, Taneichi M, Uchida T. A
Immunogenicity of MVA-NP+M1 in Adults Aged CTL-based liposomal vaccine capable of inducing
over 50 Years. PLoS ONE. 2012; 7(10): e48322. protection against heterosubtypic influenza viruses
43. Atsmon J, Caraco Y, Ziv-Sefer S, Shaikevich D, in HLA-A∗0201 transgenic mice. Biochem.
Abramov E, Volokhov I, Bruzil S, Haima KY, Biophys. Res. Commun. 2010; 391:1494-1499.
Gottlieb T, Ben-Yedidia T. Priming by a novel 55. Day EB, Zeng W, Doherty PC, Jackson DC,
universal influenza vaccine (Multimeric-001)-a Kedzierska K, Turner SJ. The context of epitope
gateway for improving immune response in the presentation can influence functional quality of
elderly population. Vaccine. 2014; 32:5816-5823. recalled influenza A virus-specific memory CD8+
44. Atsmon J, Kate-Ilovitz E, Shaikevich D, Singer T cells. J. Immunol.2007; 179:2187-2194.
Y, Volokhov I, Haim KY, Ben-Yedidia T. Safety 56. Ichihashi T, Satoh T, Sugimoto C, Kajino K
and immunogenicity of multimeric-001-a novel .Emulsified phosphatidylserine, simple and
universal influenza vaccine. J Clin Immunol. 2012; effective peptide carrier for induction of potent
32:595-603. epitope-specific T cell responses. PLoS One. 2013;
45. Oyarzún P, Kobe B. Recombinant and epitope- 8:e60068.
based vaccines on the road to the market and

Iranian Journal of Virology, Volume 11, Number 3, 2017 39


Challenges and perspectives toward development of more effective influenza vaccine

57. Singh M, O'hagan DT. Recent advances in 67. Zhao L, Seth A, Wibowo N, et al. Nanoparticle
vaccine adjuvants. Pharmaceutical research. 2002; vaccines. Vaccine. 2014; 32: 327-337.
19:715-728. 68. Storni T, Kündig TM, Senti G, Johansen P.
58. Tovey MG, Lallemand C. Adjuvant activity of Immunity in response to particulate antigen
cytokines. Methods Mol Biol. 2010; 626:287-309. delivery systems. Advanced drug delivery
59. Shahsavandi S, Ebrahimi MM, Mahravani H, reviews.2005; 57:333-355.
Saedghi K. Design of a heterosubtypic epitope- 69. Sanchez MV, Ebensen T, Schulze K,
based peptide vaccine fused with hemokinin-1 Cargnelutti D, Blazejewska P, Scodeller EA,
Downloaded from journal.isv.org.ir at 2:33 +0430 on Saturday May 2nd 2020

against influenza viruses. VirologicaSinica .2015; Guzman CA. Intranasal delivery of influenza rNP
3: 200-207. adjuvanted with c-di-AMP induces strong humoral
60. Sadghi K, Shahsavandi S, Ebrahimi MM, and cellular immune responses and provides
Mahravani H, Fazel, H. Hemokinin-1 molecular protection against virus challenge. PLoS One.
adjuvant: an approach to enhance the efficacy of 2014; 9:e104824.
influenza vaccine. Arak Med Uni J. 2014; 17:62- 70. Dobhal A, Bangde P, Dey A, Dandekar P, Jain
69. R. Chitosan-based nanoparticulate systems:
61. Wu F, Yuan XY, Li J, Chen YH. The co- implication towards therapeutics application. In:
administration of CpG-ODN influenced protective Particulate Technology for Delivery of
activity of influenza M2e vaccine. Vaccine. 2009; Therapeutics. Springer. 2017; 167-225.
27: 4320-4324. 71. Hajizade A, Ebrahimi F, Salmanian A-H,
62. Rasoli M, Omar AR, Aini I, Jalilian B, Syed Arpanae A, Amani J .Nanoparticles in vaccine
Hassan SH, Mohamed M. Fusion of HSP70 gene of development. J Applied Biotech Rep. 2015; 1:125-
Mycobacterium tuberculosis to hemagglutinin (H5) 134.
gene of avian influenza virus in DNA vaccine 72. Sawaengsak C, Mori Y, Yamanishi K, Mitrevej
enhances its potency. Acta Virologica. 2010; 54: A, Sinchaipanid N. Chitosan nanoparticle
33-39. encapsulated hemagglutinin-split influenza virus
63. Jalilian B, Omar A, Bejo M, Alitheen N, Rasoli mucosal vaccine. AAPS PharmSciTech 2014;
M, Matsumoto S: Development of avian influenza 15:317-325.
virus H5 DNA vaccine and MDP-1 gene of 73. Khalili I, Ghadimipour R, Sadigh Eteghad S,
mycobacterium bovis as genetic adjuvant. Genetic Fathi Najafi M, Ebrahimi MM, Godsian N, Sefidi
Vaccines and Therapy. 2010; 8: 4-10. Heris Y, Khalili MT. Evaluation of immune
64. Oveissi S, Omar AR, Yusoff K, Jahanshiri F, response against inactivated avian influenza
Hassan SS. DNA vaccine encoding avian influenza (H9N2) vaccine, by using chitosan nanoparticles.
virus H5 and Esat-6 of mycobacterium tuberculosis Jundishapur J Microbiol 2015; 8(12): e27035.
improved antibody responses against AIV in 74. Dehghan A., Shahsavandi S., Jabalameli L.
chickens. Comp Immunol Microbiol Infect Dis. Improvement efficacy of H9N2 influenza
2010; 33: 491-503. nanovaccine in combination with hemokinin-1
65. Sridhar S, Brokstad KA, Cox RJ. Influenza molecular adjuvant. AJMB. 2018; 10 (in press).
vaccination strategies: comparing inactivated and 75. Sawaengsak C, Mori Y, Yamanishi K, Mitrevej
live attenuated influenza vaccines Vaccines. 2015; A, Sinchaipanid N. Chitosan nanoparticle
3:373-389. encapsulated hemagglutinin-split influenza virus
66. Peek LJ, Middaugh CR, Berkland C. mucosal vaccine. AAPS Pharm Sci Tech. 2014;
Nanotechnology in vaccine delivery. Advanced 15:317-325.
drug delivery reviews. 2008; 60:915-928.

40 Iranian Journal of Virology, Volume 11, Number 3, 2017

You might also like