Overview


History and evolution of influenza control through vaccination: from the first monovalent vaccine

to universal vaccines

I. BARBERIS1, P. MYLES2, S.K. AULT3, N.L. BRAGAZZI1, M. MARTINI4

1 Department of Health Sciences (DISSAL), University of Genoa, Italy; 2 Division of Epidemiology and Public Health, University of Nottingham, UK.; 3 Pan American Health Organization/World Health Organization (retired), Washington, D.C., United States of

America; currently Office of the Dean, School of Public Health, University of Maryland, United States of America; 4 Section of History of Medicine and Ethics, Department of Health Sciences (DISSAL), University of Genoa, Italy


Keywords


Influenza • Vaccination • History of medicine


Summary


Influenza is a highly infectious airborne disease with an impor- tant epidemiological and societal burden; annual epidemics and pandemics have occurred since ancient times, causing tens of mil- lions of deaths. A hundred years after this virus was first isolated, influenza vaccines are an important influenza prevention strategy and the preparations used display good safety and tolerability profiles. Innovative tools, such as recombinant technologies and

intra-dermal devices, are currently being investigated in order to improve the immunological response. The recurring mutations of influenza strains has prompted the recent introduction of a quad- rivalent inactivated vaccine. In the near future, scientific research will strive to produce a long-lasting universal vaccine containing an antigen that will offer protection against all influenza virus strains.


Introduction


Influenza viruses are negative-sense, single-stranded RNA viruses belonging to the Orthomyxoviridae family, together with Isavirus, Thogotovirus and Quaranjavi- rus. Three types of influenza viruses, namely influenza A, B and C, are capable of determining epidemics and pandemics in humans, with influenza A being the most common circulating type and causing significant illness, being most prone to antigenic shifts and the more likely type to lead to a pandemic [1, 2]. Recently, a new genus (termed influenza virus D) has been discovered in pigs and cattle with influenza-like illness syndrome in the United States [3, 4] and in Europe [5].

Influenza is a highly infectious airborne disease that affects a significant percentage of the world’s popula- tion; local annual epidemics and pandemics have oc- curred since ancient times, causing tens of millions of deaths [6].

The aim of this mini-review is to provide a brief over- view of the history and evolution of influenza and influ- enza control using vaccines.


A history of influenza: from the classical period to the nineteenth century


In 412 BC, in the “Book of Epidemics”, Hippocrates de- scribed a putative influenza-like illness syndrome called

“fever of Perinthus” or “cough of Perinthus” [7]. While some scholars claim that this is probably the first histori- cal description of influenza (a winter and a spring epi- demic of an upper respiratory tract infection occurring regularly every year at Perinthus, a port-town in Mar- maraereglisi, a northern part of Greece, now Turkey), others, including the notable 19th-century editor of Hip- pocrates, Émile Littré (1801-1881), think that a diagno- sis of diphtheria would better fit the description of com- plications (pneumonia, fits of coughing and wheezing, angina and paralysis of soft palate and limbs). On the other hand, symptoms such as disturbed vision and night blindness suggest a combination of diseases, including deficiency syndromes (e.g. vitamin A deficiency) [8]. In the years 1173 and 1500, two other influenza outbreaks were described, though in scant detail [9-11]. The name “influenza” originated in the 15th century in Italy, from an epidemic attributed to the “influence of the stars”, which, according to Ginctrac, raged across Europe and perhaps in Asia and Africa [12].

It seems that influenza also reached the Americas. Scholars and historians debate whether influenza was already present in the New World or whether it was car- ried by contaminated pigs transported on ships. Some Aztec texts speak of a “pestilential catarrh” outbreak in 1450-1456 in an area now corresponding to Mexico, but these manuscripts are difficult to interpret correctly and this hypothesis seems controversial [13].

The first reliable documents regarding influenza-like ill-

ness syndrome date from 1510, when the virus spread from Africa to Europe. The first pandemic, or worldwide epi- demic, occurred in 1557, though some scholars deny that it really was an outbreak of influenza. The first pandemic/ worldwide epidemic that undoubtedly fits the description of influenza appeared in 1580, beginning in Asia and Rus- sia and spreading to Europe via Asia Minor and North-West Africa. In Rome, it caused the death of over 8,000 people, while in Spain it decimated the populations of entire cities. Subsequently, it also affected the Americas [14].

Over the centuries, other pandemics were described worldwide. From 1404 to the middle of the 19th century, 31 influenza epidemics were recorded, including eight large-scale pandemics. Subsequently, others appeared, including three in the 20th century [14]. Some of the most notable outbreaks occurred in 1729, in 1781-1782 (a pandemic spreading from China to Russia, Europe and North America), in 1830-1833 (a pandemic which again spread from China to India, the Philippines, Indonesia, Russia, Europe and North America), in 1847-1848, and in 1898-1900 (spreading from Europe to India, Austra- lia, and North and South America) [14].

One of the most devastating was the pandemic of “Span- ish” influenza in 1918–1919, which caused an estimated 21 million deaths worldwide and was defined by Waring as “the greatest medical holocaust in history” [14, 15]. At the end of the 19th century, the etiology of this disease had yet not been well clarified; it was believed that the disease, termed “winter catarrh”, was caused by bacte- ria (the so-called bacterial hypothesis), such as pneu- mococcus, streptococcus or Haemophilus influenzae. This latter was also named Bacillus influenzae or Pfei- ffer’s bacillus, after Richard Pfeiffer (1858-1945), who described it during the 1889-1892 influenza epidemic. This bacillus had already been discovered by the Pol- ish microbiologist Bujwid Odo Feliks Kazimierz (1857- 1942) in biopsy material a year earlier [16].

In the same period, the French microbiologists Charles Nicolle (1866-1936), Charles Lébally and René Dujar- ric de la Rivière (1885-1969) of the Pasteur Institute showed that the flu pathogen could pass through a fine filter. However, despite their brilliant experiments, the viral hypothesis continued to be neglected until the virus was isolated [16, 17].

In 1889, some Spanish doctors believed that influenza was a variant of dengue fever, whilst others attributed influenza outbreaks to a variety of causes including can- non fire on the western front, the building of the Madrid underground, air pollution, sunspots, or the spread of the habit of smoking poor-quality tobacco [18].


The thirties: virus isolation and the first experimental vaccines


During the 1918-1919 pandemic, some scientists began to suspect that bacteria were not the real agent of in- fluenza disease. One of these was the scholar Richard Edwin Shope (1901-1966), who deeply investigated swine flu in 1920. However, it was only in 1932-1933

that the English scientists Wilson Smith (1897-1965), Sir Christopher Andrewes (1896-1988) and Sir Patrick Laidlaw (1881-1940), working at the Medical Research Council at Mill Hill, first isolated the influenza A vi- rus from nasal secretions of infected patients, thereby demonstrating the intranasal human transmission of this virus [19, 20]. A few years later, the American virologist and epidemiologist Thomas Francis Junior (1900-1969) and Smith, in England, were able to transmit the virus to mice [21]. Subsequently, in 1935, Sir Frank Macfarlane Burnet (1899-1985) and Smith separately discovered that the flu virus could be grown on the chorio-allantoid membrane of embryonated hens’ eggs [22], and in 1936 the first neutralized antibodies generated by infection by human influenza virus were isolated [23].

In the next five years, important developments took place: the demonstration that the virus inactivated by formalin was immunogenic in humans, purification of the virus by means of high-speed centrifugation, and the discovery that the influenza virus grew easily in fer- tilized hen eggs, a procedure that is still used today to manufacture most influenza vaccines [23].

The first clinical trials of influenza vaccines were con- ducted in the mid-1930s [24, 25].

A study by Smith, Andrewes and Stuart-Harris was con- ducted among military forces in England in 1937 using a subcutaneous vaccination with an inactivated strain iso- lated from a mouse lung [25].

In 1938, Francis, together with Jonas Edward Salk (1914-1995), managed to protect USA military forces. Salk would subsequently use this successful experience to develop an effective polio vaccine in 1952 [26, 27].


The forties: inactivated influenza vaccines


Influenza vaccination had two main objectives: (i) to protect against disease, and (ii) to achieve a high vacci- nation rate in order to ensure protection in unvaccinated people. The first vaccine was an inactivated, monovalent preparation which only contained a subtype of the influ- enza A virus [26, 27].

In December 1942, large studies were begun to be conduct- ed on the first influenza virus vaccines; these provided the first official proof that inactivated influenza vaccines could yield effective protection against flu epidemics [28].

The efficacy and safety of inactivated vaccines were first studied between 1942 and 1945; in the meantime, a new strain of flu virus was discovered, the influenza virus type B, which is the main cause of seasonal epi- demics, as was the phenomenon of so-called “influenza mismatch”. Influenza mismatch is caused by major and minor mutations of circulating viruses. As a result, the virus contained in the vaccine does not match the circu- lating strain, determining a reduction in the effectiveness of subtype A influenza vaccines.

A new route of influenza immunization was tested in December 1942, with the subcutaneous inactivated bi- valent vaccine containing viruses of type A and type B.

The following years, the first bivalent vaccine was li- censed in the United States and became available for use in the general population [29, 30].


The fifties: influenza mismatch and influenza surveillance


The first system for the surveillance of circulating influenza virus strains in several countries worldwide was created in 1952 by the World Health Organization (WHO) in order to monitor the various virus mismatches reported. This impor- tant innovative tool enabled the composition of seasonal in- fluenza vaccines to be determined on the basis of the epide- miology of influenza in the previous season [31]. In 1946, as a result of viral mutation, a new variant of influenza A (H1N1), A/FM/1/47, appeared in Australia. This gave rise to a new influenza subtype, the H2N2 strain, which caused the pandemic known as Asian flu [32].

The following year, the US Commission on Influenza recommended that a representative of this strain be in- cluded in subsequent vaccines.

The emergence of an HA subtype different from those circulating in previous seasons determined the need for pandemic influenza vaccines [31].


The sixties: split vaccines


New inactivated compounds were tested for safety and ef- ficacy during seasonal epidemics in the 1960s, in particu- lar two new formulations were created: split and subunit vaccines. The 1968 pandemic led to the development of trivalent inactivated vaccines (TIVs) against influenza vi- ruses; moreover the development of new split or subunit vaccines led to a decrease of adverse reactions in chil- dren. These vaccines were split using ether and/or deter- gent, and haemagglutinin and neuraminidase were, in the case of subunit vaccines, purified and enriched [ 33].

In the same period, the first flu vaccines were licensed in Europe, while in the US annual influenza vaccination was recommended for individuals at major risk of influ- enza complications.

In 1968, the new virus strain H3N2 (Hong Kong) ap- peared, completely replacing the previous type A strain (H2N2, or Asian influenza), and led to another global pandemic with high morbidity and mortality [34]. In the same year, a new type of vaccine, the split vaccine, was authorized in the US after several clinical studies had demonstrated that it was less reactogenic than whole vi- rus vaccines, especially in the early years of life [35].


The seventies: genetic reassortment


Split vaccines were widely used during the pandemic swine influenza in 1976 and in 1977, when the H1N1 subtype re-emerged worldwide. However, they were seen to be less immunogenic than whole virus vaccines in “primed” subjects who had never been vaccinated. In-

deed, it was shown that two vaccine doses were needed in order to ensure effective protection [36].

At the beginning of the 1970s, an important innovation was introduced into the production of influenza vac- cines: the genetic reassortment of influenza virus strains; this technique enabled the vaccine strains to grow faster in embryonated hen eggs [37].

The first subunit vaccine was created between 1976 and 1977. This contained only the surface antigens, hemag- glutinin (HA) and neuraminidase (NA), which were iso- lated by means of successive purification steps.

This innovative tool proved to be highly immunogenic and well tolerated in humans, especially in children, al- though two doses were needed to guarantee vaccine ef- fectiveness during epidemics [38].


The eighties: subunit vaccines


In 1980, the first subunit vaccines were licensed in the United Kingdom and are currently available in several countries worldwide.

In 1978, as a result of a major mutation, a new virus strain, H1N1, appeared on the global epidemiological scene. This strain, which was similar to a virus circulat- ing in 1958, emerged in Russia and began to co-circu- late, either simultaneously or alternately, with the previ- ous one [39].

Antigenic drift, caused by frequent changes in the com- position of the virus, determined the need to update the vaccine composition each year. This necessity prompted both the implementation of the first surveillance systems and the production of the first trivalent vaccine, which included three formulation strains (one strain of influ- enza A/ H1N1, an influenza virus A /H3N2 and a type B virus), in order to ensure effective protection during the 1978 pandemic.


Live attenuated influenza vaccines


In the period 1935-1941, the first clinical trials involv- ing live attenuated influenza vaccines were conducted. The efficacy of these seasonal vaccines was guaranteed by the correspondence between the circulating strain and the strain contained in the vaccine and by the virus dose grown in hen egg embryos [34].

In 1944, Stanley described in detail the preparation and properties of an influenza virus vaccine produced in em- bryonated hen eggs; this vaccine was concentrated and purified by means of differential centrifugation and inac- tivated by means of various procedures [23].

In 1949, an important change in vaccine development involved the introduction of the use of cell cultures for virus growth.

In 1997,the so-called “avian flu” pandemic broke out in Hong Kong. This was caused by influenza virus A/ H5N1, a highly pathogenic strain.

In order to contain this pandemic, the techniques of ge- netic rearrangement developed in those years enabled a

huge number of vaccine doses to be produced in a short time by applying recombinant DNA technology to the influenza A/H5N1 virus [34].


Recent years


In recent years, scientific research developed new tech- niques of immunization, which may be more immuno- genic and better tolerated during administration, thereby reducing adverse events. In 2003, for instance, the FDA in the United States authorized the use of an intranasally administered live attenuated vaccine, called FluMist®, in adults [40]. In the 2003-2004 influenza season, an out- break in Asia was caused by an influenza A/H5N1 strain. This was later used to produce a vaccine, which was li- censed in the United States by the FDA in 2007.

More recent years saw the development of adjuvanted vaccines, such as those containing alum adjuvants and the oil in water adjuvant MF-59, which significantly en- hanced antigenicity [6].

Specifically, MF-59-adjuvanted vaccines were used in the elderly and in young children, and proved to elicit a good response even to pandemic strains with which subjects had not been primed by natural influenza infection. Similar re- sponses were obtained through the use of other emulsions, such as stable emulsion (SE) and AS03, which were includ- ed in the 2009 pandemic influenza vaccines [36].

In the most recent pandemic season (2009), the influ- enza virus H1N1, which was transmitted to humans by pigs, was estimated to have caused more than 200,000 deaths in the first 12 months of its circulation [41].

A massive effort to produce vaccine for the new H1N1 strain began shortly after scientists identified the virus. The virus proved to grow slowly during the manufac- turing process, which relies on cultivation of the virus in chicken eggs. Because of manufacturing delay, the vaccine was available in most countries after the second peak of influenza cases at the end of October leaving most people not immunized while influenza H1N1 virus was circulating [42].

In the elderly, the vaccine efficacy normally decreases, be- cause of immunosenescence. For this reason, in 2009 the Advisory Committee on Immunization Practices (ACIP) recommended and authorized the use of high-dose Flu- zone®, a new formulation containing a 4-fold higher HA dose than the traditional trivalent vaccine [43].

In 2011, as a result of developments in research into new vaccine delivery techniques, the FDA first autho- rized the intradermal administration of Fluzone®. This new route of administration involved antigen-presenting cells (APCs) in the dermis; these cells process antigens for subsequent presentation in the lymphoid organs, re- sulting in the stimulation of both innate and adaptive immunity. The intradermal vaccines elicited a better im- munological response than intramuscular vaccines, par- ticularly in the elderly; in healthy adults, it yielded an immune response comparable to that elicited by the tra- ditional vaccines, while saving on the HA dose [44-48]. In 2012, the FDA approved Fluarix®, the first quadri- valent vaccine in the United States. This split vaccine

contained two influenza A strains and two influenza B antigens. The presence of an additional influenza B strain reduced the possibility of a mismatch between the circulating viruses and the vaccine composition, while maintaining the same immunogenicity and safety as standard trivalent vaccines [49].

In 2013, the FDA approved FluBlock®, a recombinant trivalent influenza vaccine, for use in people aged be- tween 18 and 49 years. FluBlock® was licensed in a spray formulation and was the first trivalent influenza vaccine made by using recombinant DNA technology. Derived from Baculovirus, it contained a 3-fold higher HA dose than traditional trivalent vaccines [50, 51]. The scale-up potential of the insect cell/baculovirus vector system may offer advantages in terms of rapid antigen change and response to a pandemic situation [31].

Currently, scientists are exploring the fascinating prospect of developing a universal vaccine by exploiting T-cells and by attempting to elicit broadly neutralizing antibodies. Moreo- ver, efforts are being made to design M2e- or stalk-based vaccines, since these proteins (the type-2 matrix protein and the stalk domain of HA, respectively) are quite well con- served from an evolutionary standpoint [52, 53].


Conclusions


In the hundred years since the influenza virus was iso- lated, influenza vaccine preparations have evolved to ensure effective protection, while maintaining a good safety and tolerability profile.

The recurring mutations of influenza strains prompted the introduction of a quadrivalent inactivated vaccine, the composition of which is determined on the basis of the most frequent strains isolated in the previous season during continuous surveillance by the WHO.

Current research priorities include the development of a universal influenza vaccine that could offer protection against all influenza virus strains, thereby overcoming the challenges faced due to antigenic drift and shift or of co-circulation of different viral strains. Another important priority is to identify sustainable vaccine production plat- forms capable of rapidly meeting the large global demands for influenza vaccine in the face of an influenza pandemic.


Acknowledgments


No funding declared for this overview. The authors thank Dr. Bernard Patrick for revising the manuscript.


Authors’ contribution


MM conceived and designed the overview. IB and PM performed a search of the literature and contributed to the draft of the article. SA and NLB revised critically the manuscript. MM supervised the manuscript. All authors read and approved the final version of the manuscript.

References


[1] Gasparini R, Amicizia D, Lai PL, Bragazzi NL, Panatto D. Compounds with anti-influenza activity: present and future of strategies for the optimal treatment and management of influ- enza. Part I: Influenza life-cycle and currently available drugs. J Prev Med Hyg 2014;55:69-85.

[2] Gasparini R, Amicizia D, Lai PL, Bragazzi NL, Panatto D. Compounds with anti-influenza activity: present and future of strategies for the optimal treatment and management of influ- enza. Part II: Future compounds against influenza virus. J Prev Med Hyg 2014;55:109-29.

[3] Hause BM, Collin EA, Liu R, Huanq B, Shenq Z, Lu W, Wanq D, Nelson EA, Li F. Characterization of a novel influenza virus in cattle and Swine: proposal for a new genus in the Orthomyxo- viridae family. MBio 2014;5:e00031-14.

[4] Collin EA, Sheng Z, Lang Y, Ma W, Hause BM, Li F. Cocircula- tion of two distinct genetic and antigenic lineages of proposed influenza D virus in cattle. J Virol 2015;89:1036-42.

[5] Ducatez MF, Pelletier C, Meyer G. Influenza D virus in cattle, France, 2011-2014. Emerg Infect Dis 2015;21:368-71.

[6] Soema PC, Kompier R, Amorij JP, Kersten GF. Current and next generation influenza vaccines: Formulation and produc- tion strategies. Eur J Pharm Biopharm 2015;94:251-63.

[7] Pappas G, Kiriaze IJ, Falagas ME. Insights into infectious dis- ease in the era of Hippocrates. Int J Infect Dis 2008;12:347-50.

[8] Kohn GC Encyclopedia of Plague and Pestilence: From Ancient Times to the Present. Infobase Publishing 2007.

[9] Beveridge WIB. Influenza: The Last Great Plague. London: Heineman Educational Books 1977.

[10] Wood JM, Influenza. In: Crovari P, Principi N, eds. Le vaccina- zioni. Pisa: Pacini Editore 2000.

[11] Kuszewski K, Brydak. L. The epidemiology and history of influ- enza. Biomed Pharmacoter 2000;54:188-95.

[12] Gintrac H. Grippe. In: Nouveau dictionnaire de médicine et de chirurgie pratiques, directeur de la redaction: le docteur Jaccoud. 1872, Tome 16, page. 728-753.

[13] de Souza CM. The Spanish flu epidemic: a challenge to Bahian medicine. Hist Cienc Saude Manguinhos 2008;15:945-72.

[14] Potter CW. A history of influenza. J Appl Microbiol 2001;91:572-9.

[15] Waring J.I. A History of Medicine in South Carolina 1900-70.

South Carolina Medical Association 1971.

[16] Bazin H. Vaccination: a history. France: Editions John Libbey Eurotext Montrouge 2011.

[17] Renaud F, Hansen W, Freney J. Dictionnaire des precurseurs en bacteriologie, SFM Éditions Eska, 2005, 249 pages.

[18] Craddock S, Giles-Vernick T, Gunn JL. Influenza and Public Health: Learning from Past Pandemics. Earthscan, 2010 - 293 pages.

[19] Smith W, Andrewes CH, Laidlaw PP. A virus obtained from in- fluenza patients. Lancet 1933;2:66-8.

[20] Smith W, Andrewes CH, Stuart Harris CH. The immuniza- tion of human volunteers. Special Rep Ser Med Res Council 1938;228:137-44.

[21] Francis T Jr. Vaccination against influenza. Bull World Health Organ 1953;8:725-41.

[22] Burnett FM. Influenza virus infection of the chick embryo lung.

Br J Exp Pathol 1940;21:147-53.

[23] Stanley WM. The preparation and properties of influenza virus vaccines concentrated and purified by differential centrifuga- tion. J Exp Med 1945;81:193-218.

[24] Davenport FM. The search for the ideal influenza vaccine. Post- graduate Medical Journal 1979;55:78-86.

[25] Francis T. Vaccination against influenza. Bull World Health Org 1953;8:725-41.

[26] Parodi V, de Florentiis D, Martini M, Ansaldi F. Inactivated in- fluenza vaccines: recent progress and implications for the el- derly. Drugs Aging 2011;28:93-106.

[27] Barberis I, Martini M, Iavarone F, Orsi A. Available influenza vaccines: immunization strategies, history and new tools for fighting the disease. J Prev Med Hyg 2016;57:E41-6.

[28] Francis T jr, Salk JE, Pearson HE, Brown PN. Protective ef- fect of vaccination against induced influenza A. J Clin Invest 1945;24:536-46.

[29] Weir JP, Gruber MF. An overview of the regulation of influenza vaccines in the United States. Influenza and Other Respiratory Viruses 2016;10:354-60.

[30] Keitel WA, Neuzil KM, Treanor J. Immunogenicity, efficacy of inactivated/live virus seasonal and pandemic vaccines. Text- book of Influenza. Wiley-Blackwell 2013, pp. 311-326.

[31] WHO Global Influenza Surveillance Network (GISN) http://www. who.int/influenza/gisrs_laboratory/en/ [Accessed 20/9/2016].

[32] Oxford J, Gilbert A, Lambkin-Williams R. Influenza vaccines have a short but illustrious history of dedicated science en- abling the rapid global production of A/swine (H1N1) vaccine in the current pandemic. In: Rappuoli R, Del Giudice G, eds. Influenza vaccines for the future. Springer Verlag, 2011.

[33] Krammer F, Palese P. Advances in the development of influenza virus vaccines. Nat Rev Drug Discov 2015;14:167-82.

[34] Zaman M, Ashraf S, Dreyer NA, Toovey S. Human infection with avian influenza virus, Pakistan, 2007. Emerg Infect Dis 2011;17:1056-9.

[35] Parkman PD, Hopps HE, Rastogi SC, Meyer HM Jr. Summary of clinical trials of influenza virus vaccines for adults. J Infect Dis 1977;136:722-30

[36] Hampson AW. Vaccines for pandemic influenza. The history of our current vaccines, their limitations and the requirements to deal with a pandemic threat. Ann Acad Med Singapore 2008;37:510-7.

[37] Crovari P, Alberti M, Alicino C. History and evolution of influ- enza vaccines. J Prev Med Hyg 2011;52:91-4.

[38] Gianchecchi E, Trombetta C, Piccirella S, Montomoli E. Evalu- ating influenza vaccines: progress and perspectives. Future Vi- rology 2016;11:379-93.

[39] Kendal AP, Maassab HF, Alexandrova GI, Ghendon YZ. Development of cold-adapted recombinant live attenuated influenza vaccines in the U.S.A. and U.S.S.R. Antiviral Res 1982;1:339-65.

[40] Food and Drug Administration (FDA). FluMist. Prior Approval Supplemental BLA. April 2007. Available at: http://www.fda. gov/ohrms/dockets/ac/07/briefing/2007-4292B1-02.pdf.[Ac- cessed 20/9/2016].

[41] Dawood FS, Iuliano AD, Reed C, Meltzer MI, Shay DK, Cheng PY, Bandaranayake D, Breiman RF, Brooks WA, Buchy P, Feikin DR, Fowler KB, Gordon A, Hien NT, Horby P, Huang QS, Katz MA, Krishnan A, Lal R, Montgomery JM, Mølbak K, R Pebody, Presanis AM, Razuri H, Steens A, Tinoco YO, Wallinga J, Yu H, S Vong, Bresee J, Widdowson MA. Esti- mated global mortality associated with the first 12 months of 2009 pandemic influenza A H1N1 virus circulation: a modelling study. Lancet Infect Dis 2012;12:687-95.

[42] Sullivan SJ, Jacobson RM, Dowdle WR, Poland GA. 2009 H1N1 influenza. Mayo Clin Proc 2010;85: 64-76.

[43] Falsey AR, Treanor JJ, Tornieporth N, Capellan J, Gorse GJ. Randomized, double-blind controlled phase 3 trial comparing the immunogenicity of high-dose and standard-dose influenza vaccine in adults 65 years of age and older. J Infect Dis 2009;200:172-80.

[44] Durando P, Iudici R, Alicino C, Alberti M, de Florentiis, Ansaldi F, Icardi G. Adjuvants and alternative routes of administration towards the development of the ideal influenza vaccine. Hum Vaccin 2011;7(Suppl):29-40.

[45] Holland D, Booy R, De Looze F, Eizenberg P, Mc Donald J, Karrasch J, Mc Keirnan M, Salem H, Mills G, Reid J, Weber F, Saville M. Intradermal influenza vaccine administered using a new microinjection system produces superior immunogenic- ity in elderly adults: a randomized controlled trial. J Infect Dis 2008;198:650-8.

[46] Arnou R, Icardi G, De Decker M, Ambrozaitis A, Kazek MP, Weber F, Van Damme P. Intradermal influenza vaccine for old- er adults: a randomized controlled multicenter phase III study. Vaccine 2009;27:7304-12.

[47] Belshe RB, Newman FK, Cannon J, Duane C, Treanor J, Van Hoecke C, Howe BJ, Dubin G. Serum antibody responses af- ter intradermal vaccination against influenza. N Engl J Med 2004;351:2286-94.

[48] Bragazzi NL, Orsi A, Ansaldi F, Gasparini R, Icardi G. Flu- zone® Intra-dermal (Intanza® / Istivac® Intra-dermal): an up- dated overview. Hum Vaccin Immunother 2016, in press.

[49] Tisa V, Barberis I, Faccio V, Paganino C, Trucchi C, Martini M, Ansaldi F. Quadrivalent influenza vaccine: a new op-

portunity to reduce the influenza burden. J Prev Med Hyg 2016;57:E28-33.

[50] Treanor JJ, El Sahly H, King J, Graham I, Izikson R, Kohberger R, Patriarca P, Cox M. Protective efficacy of a trivalent recombi- nant hemagglutinin protein vaccine (FluBlok) against influenza in healthy adults: a randomized, placebo-controlled trial. Vac- cine 2011;29:7733-9.

[51] Baxter R, Patriarca PA, Ensor K, Izikson R, Goldenthal KL, Cox M. Evaluation of the safety, reactogenicity and immunoge- nicity of FluBlok trivalent recombinant baculovirus-expressed hemagglutinin influenza vaccine administered intramuscularly to healthy adults 50-64 years of age. Vaccine 2011;29:2272-8.

[52] Sridhar S. Heterosubtypic T-cell immunity to influenza in hu- mans: challenges for universal T-cell influenza vaccines. Front Immunol 2016;7:195.

[53] Wiersma LC, Rimmelzwaan GF, de Vries RD. Developing uni- versal influenza vaccines: hitting the nail, not just on the head. Vaccines (Basel) 2015;3:239-62.


n Received on July 8, 2016. Accepted on August 25, 2016.


n Correspondence: N.L. Bragazzi, Department of Health Sciences (DISSAL), University of Genoa, via Antonio Pastore 1, 16132 Ge- noa, Italy - E-mail: robertobragazzi@gmail.com