new technologies for new influenza vaccines

7
Vaccine 30 (2012) 4927–4933 Contents lists available at SciVerse ScienceDirect Vaccine jou rn al h om epa ge: www.elsevier.com/locate/vaccine Review New technologies for new influenza vaccines Alan Shaw VaxInnate, 3 Cedarbrook Drive, Cranbury, NJ 08512, United States a r t i c l e i n f o Article history: Received 28 January 2012 Received in revised form 24 April 2012 Accepted 26 April 2012 Available online 9 May 2012 Keywords: Influenza Vaccine Virus-like particles Attenuated Cold-adapted NS1 deletion Toll-Like Receptors a b s t r a c t The currently available influenza vaccines were developed in the 1930s through the 1960s using tech- nologies that were state-of-the art for the times. Decades of advancement in virology and immunology have provided the tools for making better vaccines against influenza. We now have the means to make vaccines that address some of the shortcomings of the original products, in particular performance in the elderly. © 2012 Elsevier Ltd. All rights reserved. Influenza vaccines have been a feature of the routine immuniza- tion portfolio since the 1960s. These vaccines, developed with the best technology available at the time, have served us well. Over the past several years there has been a growing appreciation of the shortcomings of the original egg based vaccines’ immunogenicity, especially in the elderly, where 40 years of clinical studies have failed to come up with a satisfactory solution. A recent review of this topic from CIDRAP highlights the need to develop a new gen- eration of influenza vaccines that take advantage of the last 60 years of advancement in immunology and molecular biology [1]. Three major events have driven a renewed interest in influenza vaccines. In the late 1990s there were widely publicized outbreaks of avian influenza in Asian bird markets that led to lethal infec- tions in humans. Fortunately, human-to-human transmission of avian H5 virus is very rare, if it happens at all. The specter of such transmission, if it became efficient, was the impetus for a wave of new interest and investment in flu vaccine technology. The second major event was a realization over time that the standard trivalent influenza (TIV) vaccines are relatively poor immunogens in peo- ple over 65 years of age; this is the fastest growing segment of our population. Beginning in the 1960s, a series of clinical studies of TIV were carried out in an effort to find a more immunogenic dose and regimen. Most of these studies focused on increasing the dose from the standard 15 g per component up to as much as 405 g of HA, with the best results showing a doubling of the HAI titer. Recently, Tel.: +1 215 208 9036. E-mail address: [email protected] a “4×dose vaccine with 60 g of each component was approved for people over 65. The third event was the pandemic of 2009. We expected an avian virus pandemic, and we got a swine-human virus instead. The influenza manufacturing community applied the best possible effort, but the vaccine became available far too late and went largely to waste. Together, these three elements now drive a new interest in influenza vaccines. This article will provide a very brief history of the development of the original egg-based vaccines and the technical refinements that have been applied to them. We will also cover the evolution of live nasally delivered vaccines and their commercial development. We will devote the majority of our attention to the avalanche of new approaches triggered by a renewed interest in flu following the avian influenza outbreaks, our experience with the 2009 pandemic, and the impact of funding for influenza vaccine development pro- vided by the US government. Since this is now a fast-moving field, much of the core data, especially clinical results, are not published in a final form that is suitable for citation. Where appropriate, sum- maries of public presentations will be included. The goal here is to provide an overview of where the influenza vaccine field is headed and why rather than to compile a complete list of projects. 1. A short history Influenza virus was first isolated in the early 1930s by British, American and Australian investigators working at the forefront of what we now know as clinical and veterinary virology. The virus was detected initially due to its ability to cause disease in ferrets. Ironically, the use of the ferret for influenza work is at least partly 0264-410X/$ see front matter © 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.vaccine.2012.04.095

Upload: alan-shaw

Post on 30-Oct-2016

218 views

Category:

Documents


2 download

TRANSCRIPT

Page 1: New technologies for new influenza vaccines

R

N

AV

a

ARRAA

KIVVACNT

tbtsefteyTvotatnmippwrtw

0h

Vaccine 30 (2012) 4927– 4933

Contents lists available at SciVerse ScienceDirect

Vaccine

jou rn al h om epa ge: www.elsev ier .com/ locate /vacc ine

eview

ew technologies for new influenza vaccines

lan Shaw ∗

axInnate, 3 Cedarbrook Drive, Cranbury, NJ 08512, United States

r t i c l e i n f o

rticle history:eceived 28 January 2012eceived in revised form 24 April 2012ccepted 26 April 2012vailable online 9 May 2012

eywords:

a b s t r a c t

The currently available influenza vaccines were developed in the 1930s through the 1960s using tech-nologies that were state-of-the art for the times. Decades of advancement in virology and immunologyhave provided the tools for making better vaccines against influenza. We now have the means to makevaccines that address some of the shortcomings of the original products, in particular performance in theelderly.

© 2012 Elsevier Ltd. All rights reserved.

nfluenzaaccineirus-like particlesttenuatedold-adaptedS1 deletion

oll-Like Receptors

Influenza vaccines have been a feature of the routine immuniza-ion portfolio since the 1960s. These vaccines, developed with theest technology available at the time, have served us well. Overhe past several years there has been a growing appreciation of thehortcomings of the original egg based vaccines’ immunogenicity,specially in the elderly, where 40 years of clinical studies haveailed to come up with a satisfactory solution. A recent review ofhis topic from CIDRAP highlights the need to develop a new gen-ration of influenza vaccines that take advantage of the last 60ears of advancement in immunology and molecular biology [1].hree major events have driven a renewed interest in influenzaaccines. In the late 1990s there were widely publicized outbreaksf avian influenza in Asian bird markets that led to lethal infec-ions in humans. Fortunately, human-to-human transmission ofvian H5 virus is very rare, if it happens at all. The specter of suchransmission, if it became efficient, was the impetus for a wave ofew interest and investment in flu vaccine technology. The secondajor event was a realization over time that the standard trivalent

nfluenza (TIV) vaccines are relatively poor immunogens in peo-le over 65 years of age; this is the fastest growing segment of ouropulation. Beginning in the 1960s, a series of clinical studies of TIVere carried out in an effort to find a more immunogenic dose and

egimen. Most of these studies focused on increasing the dose fromhe standard 15 �g per component up to as much as 405 �g of HA,ith the best results showing a doubling of the HAI titer. Recently,

∗ Tel.: +1 215 208 9036.E-mail address: [email protected]

264-410X/$ – see front matter © 2012 Elsevier Ltd. All rights reserved.ttp://dx.doi.org/10.1016/j.vaccine.2012.04.095

a “4×” dose vaccine with 60 �g of each component was approvedfor people over 65. The third event was the pandemic of 2009. Weexpected an avian virus pandemic, and we got a swine-human virusinstead. The influenza manufacturing community applied the bestpossible effort, but the vaccine became available far too late andwent largely to waste. Together, these three elements now drive anew interest in influenza vaccines.

This article will provide a very brief history of the developmentof the original egg-based vaccines and the technical refinementsthat have been applied to them. We will also cover the evolution oflive nasally delivered vaccines and their commercial development.

We will devote the majority of our attention to the avalanche ofnew approaches triggered by a renewed interest in flu following theavian influenza outbreaks, our experience with the 2009 pandemic,and the impact of funding for influenza vaccine development pro-vided by the US government. Since this is now a fast-moving field,much of the core data, especially clinical results, are not publishedin a final form that is suitable for citation. Where appropriate, sum-maries of public presentations will be included. The goal here is toprovide an overview of where the influenza vaccine field is headedand why rather than to compile a complete list of projects.

1. A short history

Influenza virus was first isolated in the early 1930s by British,

American and Australian investigators working at the forefront ofwhat we now know as clinical and veterinary virology. The viruswas detected initially due to its ability to cause disease in ferrets.Ironically, the use of the ferret for influenza work is at least partly
Page 2: New technologies for new influenza vaccines

4 30 (2

cdi

2

othaaapA2(

ieoigls

mtotwvtospstsRm

3

llsvtn[a

paael

wn4ut

928 A. Shaw / Vaccine

oincidence, as the UK lab’s initial mission was to work on canineistemper virus which was affecting a substantial fraction of dogs

n the UK at the time.

. Egg-based vaccines

In relatively short order, this group worked out the culturef influenza virus in minced chick embryo, in eggs, and then inhe allantoic cavity of eggs. Shortly thereafter in the early 1940s,emagglutination was recognized both as a means of titrating viruss well as antibody against the virus. The single radial hemolysisssay [2] was developed in the mid-70s in response to a need for

better means of dosing HA. These basic tools continue to sup-ort the production and analysis of influenza vaccines to this day.

good account of these events can be read in David Tyrell’s Chapter in The Textbook of Influenza edited by Nicholson, Webster and HayBlackwell).

The first useful influenza vaccines can be traced to formalin-nactivated virus preparations made in mouse lungs or chickmbryos from 1937 through the early 1940s. The US Army carriedut a series of studies that demonstrated protective efficacy in mil-tary personnel beginning in 1942 using formalin-inactivated virusrown in eggs and concentrated by centrifugation. The first vaccineicensed by the “FDA” of the time was made by Parke-Davis, and theecond was made by Merck.

In the 1960s, zonal centrifugation was implemented as a betterethod for enrichment of the virus-containing fraction of allan-

oic fluid, and “split” virus vaccines were introduced. Treatmentf the enriched virus with either ether or deoxycholate disruptedhe virion with the effect of reducing adverse reactions associatedith the earlier whole virion preparations. Whole inactivated virus

accine, at a lower dose, has been reintroduced recently by Bax-er. The next advance in the late 1960s was the implementationf “high growth reassortants” [3] where the HA and NA genes areelectively crossed into the PR8 strain that grows well in eggs. Thisrovided a more reliable means of propagating influenza virus atcale and greatly improved the match of the vaccine. At this point,he inactivated “flu vaccine” became a routine product made bytandard methods by numerous commercial vaccine producers.ecently, reverse genetics has become the preferred method foraking production strains.

. Cold adapted live attenuated vaccines

In parallel, beginning in the early [4] 1960s Smorodintsev’saboratory in Leningrad [5,6] began the development of live andive-attenuated vaccines based on a cold-adapted and temperatureensitive virus derived from the 1957 Leningrad isolate. At the Uni-ersity of Michigan, Maassab initiated a similar program based onhe Ann Arbor A and B strains [7]. The Ann Arbor-based vaccine,ow known as FluMist®, was licensed for use in the US in 20038]. The Leningrad-based vaccine is widely used in Russia and isvailable for license via the WHO.

Vaccines are public health tools, but they are also manufacturedroducts made, for the most part, by companies that have to make

profit. This mix of business, public health and public policy in highly regulated environment makes an exciting and rewardingxistence for those engaged. However, the path from concept toicensed vaccine is not always linear.

Two good examples of tortuous business pathways can be seenith the live attenuated vaccines. The live-attenuated, cold adapted

asally delivered vaccines from Leningrad and Michigan took over0 years of development. The Michigan/Maasaab vaccine began as aniversity-NIH project, which was licensed to Wyeth and returnedwice, then licensed to Aviron, a California Biotech Company who

012) 4927– 4933

licensed the vaccine to Wyeth (again) in 1998–1999. Wyeth soldthe project to MedImmune (who acquired Aviron as part of thedeal) where development was completed. FluMist® was approvedfor marketing in the US in 2003. The Leningrad/57 vaccine also hasa complicated commercial history. Development began at the Insti-tute for Experimental Medicine around 1960 with numerous smallclinical studies over decades and approval for use in Russia. In 2002Merck licensed the Leningrad/57 strains and spent three years char-acterizing the genetics of the vaccine and developing processes formanufacturing [9–11]. Ultimately, Merck returned the rights to theLeningrad/57 strains to the Institute. Subsequently, the Leningradstrains were licensed to Nobilon, a division of Schering-Plough. In2010, Merck merged with Schering-Plough, and as a consequence,the Leningrad strains are now back at Merck! The point here is thathaving a developed vaccine is not sufficient for success. A consis-tent business plan is also a necessary part of the equation. This isan element of all of the new vaccine programs on the horizon.

4. New initiatives in influenza: expanding capacity

The standard trivalent inactivated influenza vaccines (TIV)became routine products made by standard methods from the1960s to the present day. US sales were in the range of about 20million doses per year up to the 1990s when a focus on influenzamorbidity and mortality raised the general awareness. The out-breaks of influenza in the poultry markets of Asia in 1997–1998spurred an effort by the major flu vaccine producers to explore thepossibility of expanding their capacity beyond the current marketdemand under the anti-trust provisions of the IFPMA in Geneva.This was not a simple task, and it was a business risk and bur-den. The vaccine producers made the argument that in order tomaintain the capacity to deal with a pandemic, there should be abetter uptake of seasonal vaccine. However, having a significantover-capacity can lead to lower prices that in turn cause produc-ers to exit the business. This is what happened in the US duringthe 1970s and 1980s and led to the US having a single domestic fluvaccine source, sanofi-pasteur. Having excess idle capacity is notgood business. At a recent WHO Influenza vaccine supply meeting(12–14 July, 2011) the International Federation of PharmaceuticalManufacturers Association estimated that current credible demandfor TIV is 553 million doses per year, while annual capacity is 1383million doses. The limit is currently not volume, but in the faceof a pandemic as in 2009, the crucial element is time to last dosedelivered.

Expanding capacity has provided an opportunity to increase thenumber of virus types included in the seasonal vaccine. The cur-rent vaccines comprise an H1, an H3 and a single B-strain. TheB-strains are typically difficult to grow, consuming a disproportion-ate fraction of the manufacturing effort. Expanded capacity allowsthe inclusion of a second B-strain, something that has been dis-cussed for years but has not been implemented. Several tetravalentvaccine development projects are now underway.

5. Year-round egg supply

Up until around 2001, the chicken flocks supporting vaccine pro-duction were expanded to meet seasonal vaccine manufacturingfrom February to August, and were then culled, only to be expandedagain for the next season. The first “novelty” for the flu vaccine busi-ness was an agreement in 2002 by the Secretary of US Health andHuman Services (USHHS) to fund the maintenance of chicken flocks

year-round so as to facilitate emergency ramp-up of vaccine pro-duction. Note that a hen must be at least 20 weeks old before layingeggs. This arrangement for year-round flock maintenance remainsin force and will help with the timing of initiation of manufacturing.
Page 3: New technologies for new influenza vaccines

30 (2

6

mvospimfiorb

7

ivrtaEcb4st

8

iidrMvtAa

iimmTytvfstHco

wSnmm[d

A. Shaw / Vaccine

. Mammalian cell culture

The next advance, also funded in part by USHHS, was the imple-entation in the US of mammalian cell culture-based influenza

accine production. Cell culture is attractive as it does not relyn the maintenance of huge flocks of chickens, and it may belightly faster. Contracts were awarded to three producers (sanofi-asteur, Novartis and Solvay) for US-based mammalian cell culture

nfluenza vaccine production facilities and development. Ulti-ately, Solvay and sanofi-pasteur terminated their programs for

nancial and feasibility reasons. The Novartis facility in North Car-lina is now complete and ready to begin production. The mainemaining hurdle is process economics. Novartis has a cell-cultureased vaccine licensed in the EU, as do Baxter and Solvay.

. Adjuvanted vaccines

Influenza vaccines comprising an adjuvant have been availablen Europe for several years. The primary target population for theseaccines has been those over 65 years of age, in whom the immuneesponse is less robust and who suffer greater morbidity and mor-ality from influenza. During the course of the 2009 pandemic,djuvanted influenza vaccines were used widely in Canada and inurope, but not in the US. We anticipate that these adjuvanted vac-ines will be approved in the US at some point. Addition of oil-waterased emulsion adjuvants to standard egg-based vaccines allows a-fold dose sparing effect. At a recent 2012 conference, Novartishowed that Fluad, their MF59-adjuvanted vaccine provided twicehe efficacy of standard TIV in children aged 6–72 months.

. Recombinant DNA based vaccines made in insect cells

One of the biggest variables in influenza vaccine productions the ability of a given seasonal virus production strain to grown eggs. This was one of the main reasons that the 2009 pan-emic vaccine was delayed. Production strains grew poorly andequired sub-selection and refinement that took precious time.odern rDNA methods can circumvent the need to propagate the

irus. The major immunological target on the virus is hemagglu-inin (HA) which carries the vast majority of neutralizing epitopes.

variety of methods for making influenza HA have been developednd tested in the clinic. In order of chronological development:

Protein Sciences produces influenza hemagglutinin, FluBlok, innsect cells (Army Worm, Spodoptera frugiperda) infected with annsect cell virus vector (Baculovirus). This is now a well-accepted

ethod for making vaccines; one of the licensed human papillo-avirus vaccines, Cervarix® is made using this general system.

he Protein Sciences vaccine comprises the whole HA sequence,ielding a mix of HA monomers and HA rosettes connected via therans-membrane sequence [12–14]. Clinical trials have shown thisaccine to be well tolerated and immunogenic at a dose of 45 �gor each of the three TIV components [14–16]. FluBlok has beenubmitted to the USFDA for approval. Early in the development ofhis vaccine, a candidate pandemic vaccine was made based on theong Kong156/97 strain. This vaccine, like many H5 vaccines thatame afterwards, was suitably immunogenic only after two dosesf 90 �g [17].

Novavax produces virus-like particles in insect cells infectedith a baculovirus vector, analogous to the process used by Protein

ciences, above. The virus-like particles contain hemagglutinin,euraminidase, and the influenza matrix protein. Inclusion of the

atrix protein allows HA and neuraminidase to associate withatrix so as to form a particle that resembles an influenza virion

18]. The advantage of the particulate presentation is that den-ritic cells have a particle-recognizing mechanism that takes up

012) 4927– 4933 4929

virus-sized particles [19]. This in turn activates the dendritic cell.The currently licensed hepatitis B and human papillomavirus vac-cines rely on this type of particulate presentation. These influenzavaccines are immunogenic across doses of 5–45 �g per HA species[20], and they elicit cross-clade protection in animals and inhumans vaccinated with H5 avian HA-containing VLPs [21,22].Addition of a liposomal adjuvant may improve immunogenicity[23].

9. Recombinant DNA based vaccines made in plants

Modern molecular biology provides the ability to expressrecombinant proteins in a variety of cell types, including plant cells.Two groups have successfully made HA in tobacco plants.

Fraunhofer Institute [24] uses a tobacco mosaic virus vector toexpress HA in tobacco plants. The virus vector DNA in solutionis instilled into the plants by inverting them and submerging theleaves. A vacuum is applied, opening the stoma on the bottom ofthe leaves, and drawing the DNA inside the plant. HA is expressed,and the antigen is extracted from the leaves. This vaccine has beenshown to be immunogenic in a phase 1 clinical study VIDADI.

Medicago [25] uses a similar approach in tobacco plants,using an Agrobacter vector introduced by vacuum instillation. Theexpressed HA sequence in the Medicago system creates a virus-like particle. Doses of an H5 Indonesia vaccine ranging from 0.7 to11 �g of VLPs absorbed onto alhydrogel elicited robust HAI titersin a clinical trial [26].

Yields and purity of both plant-based influenza vaccines are con-sistent with the requirements of an industrial process that shouldsatisfy regulatory agencies.

While the major focus on plants has been centered on tobacco,there are many other options. Enough is now known about thegenetics and regulation of gene expression in organisms other thanmammals, yeast and bacteria, to expand the repertoire of hostsfor flu vaccine production. Two examples presented at a recent“New Cells for New Vaccines” meeting included the common but-ton mushroom and brine shrimp.

10. HA-based influenza vaccines made in bacteria

Since the early days of recombinant DNA influenza has been avaccine target of interest. Numerous groups have taken up the chal-lenge. One of the pioneers in this area is the Arnon lab in Israel[27–30]. Since the hemagglutinin molecule is a complex foldedstructure, the Arnon lab created as series of peptide epitope basedvaccines comprising conserved structures of HA as well as nucleo-protein. These vaccines, when adjuvanted, perform well in animalmodels. This effort, which continues aggressively at BiondVax, hasbeen the subject of a recent review [31].

For decades, the influenza vaccine dogma held that the hemag-glutinin molecule had to be produced in a vertebrate cell substratethat could fold, glycosylate and secrete the protein. It turns outthat none of this is true. First, a group at the CDC showed, usinga DNA vaccine, that knocking out the various N-linked glycosyla-tion sites on HA had no measureable impact on immunogenicity inmice [32]. Subsequently, two groups have produced influenza HAin Escherichia coli. At the USFDA/CBER, Hana Golding and her teamhave produced a properly-folded H5 VietNam HA molecule that isimmunogenic in animals. In addition, this group has identified apotential new oligomerization sequence that may be useful [33].

VaxInnate has developed a method for making HA-based

influenza vaccines based on a genetic fusion of the sequence for theHA globular head with the sequence for flagellin, the ligand for Toll-Like Receptor 5 (TLR5) [34–36]. Vaccines of this design have beentested in the clinic and showed an optimal dose of 1–2 �g in normal
Page 4: New technologies for new influenza vaccines

4 30 (2

hambi

Ticwf

1

iaptvuco

1

as[evtan

ha

oatcttnT[r

gaha

tcvt

cppa

930 A. Shaw / Vaccine

ealthy adults and about 5 �g in healthy subjects over 65 years ofge. These vaccines were generally well tolerated. They seem to beore immunogenic in the elderly compared to the standard egg-

ased product [36]. The flagellin-fusion approach also seems to helpn the aged mouse model [37].

The other advantage of the E. coli system is speed and capacity.hese bacteria double every thirty minutes or less, grow to high titern chemically defined medium, and typically make 20% of their totalell protein as your product. New HA constructs can be made in twoeeks. In the face of a serious pandemic, or for a low-cost vaccine

or large populations, this type of approach becomes attractive.

1. Universal vaccines based on conserved protiens

A universal flu vaccine has been a long-standing desire in thenfluenza community. The notion of a flu vaccine that could bedministered once with a booster every 5–10 years, maintainingrotection against mild to severe disease, has attracted the atten-ion of the public, biotech investors, public health officials, andaccine manufacturers over the past decade. Why cannot we comep with something better to replace this 60+ year old vaccine? It isertainly not a question of lack of effort. Several conserved aspectsf the virus have been explored as vaccine candidates.

1.1. M2 ectodomain (M2e)

The conserved ion channel, M2, was discovered back in 1980s a curious second open reading frame on the M protein geneegment [38]. It was conserved [39], it had ion channel activity40] a necessary function, and antibody raised against the surface-xposed ectodomain, M2e, could restrict the growth of influenzairus [41]. This collection of properties makes M2e and attractivearget. There is one major problem. Natural infection rarely elicitsnti-M2e immunity [42], therefore extraordinary measures may beecessary to make this little antigen immunogenic.

A great deal of effort has been invested by several groups thatave produced candidate M2e vaccines since this seems to be suchn obvious target.

Walter Fiers’ group in Ghent made a particulate vaccine basedn the hepatitis B core antigen which can be expressed in bacteriand spontaneously self-assembles. Appending the M2e sequenceo the core antigen monomer yields a small particle with multipleopies of the M2e sequence on the surface [43]. This vaccine wasested in the clinic and was found to be immunogenic, althoughhe data have not been published outside of a CIDRAP commu-ication (sciencedaily.com/releases/2007/07/070718002008.htm).his project has taken up by sanofi [44] for further development44]. The mode of action of anti-M2e antibody depends on Fc-eceptor bearing macrophages.

DynaVax has made a vaccine comprising 8 tandem copies of M2eenetically appended to influenza nucleoprotein, a T-cell antigen,nd conjugated to a CpG-containing oligonucleotide. This vaccineas been the subject of a clinical trial, and both components, M2end NP, were immunogenic.

CYTOS Biotechnology made a HepBcAG-M2e vaccine [45]hat afforded some protection in murine models. This vac-ine was tested in a clinical trial (www.who.int/entity/accine research/diseases/.../Bachmann.pdf) and was showno be immunogenic.

Merck made a candidate vaccine based on the M2e peptide

hemically conjugated to their proprietary outer membrane com-lex (OMPC) carrier [46]. OMPC is a TLR2 agonist as well as being aarticle. The M2e vaccine was tested in a clinical trial as an alum-dsorbed particle at two different dose levels, and in combination

012) 4927– 4933

with an ISCOM adjuvant in a two dose regimen. Best anti-M2e titerswere obtained with the highest dose plus ISCOM.

VaxInnate made an M2e vaccine based on four tandem copies ofthe M2e sequence fused genetically to the C-terminus of Salmonellaflagellin. This vaccine also provided solid protection against a lethalchallenge in mice [47]. This vaccine was tested in a clinical trial [48]in a two-dose regimen. Robust anti-M2e titers were elicited with1 �g or less total protein per dose. In a follow-up study, the M2evaccine was co-delivered with traditional TIV [49]. The immuneresponse against M2e was unaffected by the co-delivery of TIV, andthe immune response against the two type A components was aug-mented by about 50%. This suggests that an M2e vaccine may be auseful adjunct for seasonal vaccination.

12. Other conserved antigens

The stalk of HA is relatively conserved, compared to the glob-ular head. Until recently, little attention has been paid to the HAstalk. That has now changed in the wake of discoveries of broadlyneutralizing monoclonal antibodies that bind to the stalk and inter-fere with infection [51–53]. The stalk structure that binds to theseantibodies is involved in fusion with the endosomal membrane andappears to be a pH sensitive intermediate. If one could stabilize thisstructure and use it as a vaccine, a broadly cross-reactive vaccinemight be feasible. One group comprising scientists from the NehruInstitute in India, Merck and Crucell [54] has made a candidate vac-cine, using a battery of molecular modeling techniques, that comesclose to this goal.

Merck also made a vaccine targeting the stalk cleavage site,a poly-basic sequence on the stalk that must be cleaved by ahost protease in order for the stalk to re-orient and penetratethe cell. Synthetic peptide was chemically conjugated to OMPC, asabove. The OMPC–peptide conjugate underwent extensive preclin-ical testing [50] in mice and non-human primates, but this vaccinewas not taken into clinical trials.

Two other, simpler, stalk antigens have been proposed, one ofwhich dates back to 1982 [55]. Palese and colleagues removedthe globular head from virus particles by proteolytic cleavage. Theremaining HA2 stalk revealed cross-reactive HA2 eptiopes. Thisidea was revisited recently [56] using the straight alpha helix,aa76–130, as an immunogen, affording modest protection in amurine challenge. Alternatively, the same lab engineered a “head-less HA” stalk that afforded good protection in a mouse challengemodel [57] when presented as a particle based on an HIV gag core,delivered as electroporated DNA. The stalk with its more conservedstructure remains a seductive target.

13. Back to live viruses

The original live attenuated influenza viruses were attenuatedby multiple serial passages in eggs at low temperature, around30 ◦C, selecting for variants that can grow in the cool nasopharynxbut cannot grow in the lung. We now know enough about the virusand have the tools to manipulate its genetics in a more directedmanner. The non-structural protein NS1 gene segment encodes twoproteins that serve to highjack the host cell’s synthetic machinery[58] by regulating splicing and export of mRNA. Recently, a his-tone like function has been described as further interfering with thehost anti-viral response [76]. Deleting or truncating the NS1 gene[59] has multiple anti-interferon effects that ultimately result in anattenuated virus with potential as a vaccine. Similar NS1 deletions

have been developed by AVIR-Greenhills in Vienna [60]. Furtherengineering at the Mount Sinai-Palese-Garcia-Sastre lab has gen-erated flu viruses that cannot reassert, a feature that should beuseful in the face of a pandemic [61]. This group has also created
Page 5: New technologies for new influenza vaccines

30 (2

aspfl

1

masHtibhamlacsdi

1

whltboao2ph

1

othhvhtmwltfscv

rii

[

[

[

[

[

[

[

A. Shaw / Vaccine

n influenza virus that can carry two HA species [62] using a nine-egment genome. This type of approach, taking advantage of thelasticity of the flu genome, should change the way we think aboutu vaccines.

4. Vaccines intended to elicit T-cell responses

For decades, influenza vaccines have been characterized pri-arily by their ability to elicit hemagglutination-inhibiting (HAI)

ntibody in serum. In preclinical studies we and many others haveeen protection in challenge models in the absence of detectableAI or neutralizing antibody. We assume that a cellular response is

he protective element. Given the repetitive exposure to influenzan the population, infants excluded, the flu vaccines are partlyoosters of preexisting immunity. Adjuvanted trivalent vaccinesave been shown to raise significant Th1 T-cell responses as wells antibody responses [4]. The literature is replete with studieseasuring antibody fold-rise over background and finding that the

ower the background, the higher the fold-rise. The same thingpplies to T-cell responses [63]. Vaccines intended to induce a T-ell response fall into two general categories; vaccines comprisingelected conserved peptides, often as concatamers, and vectoredelivery of antigens. Some labs have combined techniques deliver-

ng multiple selected epitopes via a viral vector or as DNA.

4.1. Peptide based vaccines

An excellent review of T-cell epitope-based vaccine approachesas published just after the 2009 pandemic [64]. We will highlightere specific examples of the various technologies. Ruth Arnon’s

ab has, as mentioned earlier, a concatamer of peptides approachargeting conserved epitopes from NP, M, and HA produced inacteria. Goodman has developed a comprehensive epitope basedn peptides from NP, M1, NS1, PB1, PA, HA and NA expressed in

vaccinia vector [65]. Enhanced CD4 and CD8+ responses werebtained in mice. Alexander has produced a DNA vaccine encoding0 influenza epitopes derived from an array of past and potentialandemic viruses [66] and has demonstrated protection in HLA-DRumanized mice.

4.2. Vectored delivery of antigens

For years, investigators have been using viral vectors (aden-virus, vaccinia variants, alphaviruses, etc.), as a means of attackinghe Big Three pathogens, HIV, TB and malaria [67]. Expression of HAas been achieved in alphavirus vectors [68,69]. Vaccinia vectorsave been used to express the HAs of equine and human influenzairuses [70,71]. Adenovirus vectors expressing influenza antigensave been tested in the clinic [72]. Perhaps the most advanced ofhese approaches is an MVA-based vaccine delivering the NP and

atrix proteins led by Gilbert at Oxford. An initial clinical studyas designed to explore intramuscular and intradermal routes at

ow and high doses [73]. The low dose IM and ID were similar inerms of CD8+ response, while the higher IM dose response was 3-old higher. In a follow-on human challenge study [74], vaccineeshowed a reduced incidence of influenza illness and virus sheddingompared to controls. A similar approach has been used to make aaccine against H5 viruses in the Osterhaus lab [75].

For the field of vectored delivery, one issue that will only beesolved by time and clinical trials is anti-vector immunity. Willt be feasible to administer these vaccines multiple times withoutnterference due to prior exposure to the vector.

[

012) 4927– 4933 4931

15. Where do we go from here?

Clearly, we have a broad range of methods for making influenzavaccines. With an established 60 year old vaccine as the incum-bent, developing a successor, or multiple successors, will take timeand significant effort. Moving away from growing influenza virus ineggs or cell culture will be the first goal. What comes next dependson how well the candidates discussed here fare in the developmentprocess, especially in the clinic and even more so in clinical stud-ies in the elderly where the need is greatest. A critical element forthe success of any of the above programs will be new thinking onthe part of the regulatory agencies regarding the metrics of successin clinical trials of these vaccine candidates. The present guidanceis rooted in the experience with inactivated split virus vaccines,where HAI titers are accepted as correlates of protection for vaccinesof this type. The newer vaccines may require something differentand yet to be defined. Finally, the successful development candi-dates must be easy to manufacture and test for batch release. Thisis not something that is often considered in the early research anddevelopment effort. It is possible to make a wonderful vaccine thatis so complex and costly to produce that it cannot be implemented.

References

[1] Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efficacy and effectiveness ofinfluenza vaccines: a systematic review and meta-analysis. The Lancet Infec-tious Diseases 2012;12(January (1)):36–44 [Meta-Analysis, Research Support,Non-U.S. Gov’t, Review].

[2] Schild GC, Pereira MS, Chakraverty P. Single-radial-hemolysis: a new methodfor the assay of antibody to influenza haemagglutinin. Applications for diag-nosis and seroepidemiologic surveillance of influenza. Bulletin of the WorldHealth Organization 1975;52(1):43–50.

[3] Kilbourne ED. Future influenza vaccines and the use of genetic recombinants.Bulletin of the World Health Organization 1969;41(3):643–5.

[4] Wack A, Baudner BC, Hilbert AK, Manini I, Nuti S, Tavarini S, et al. Combina-tion adjuvants for the induction of potent, long-lasting antibody and T-cellresponses to influenza vaccine in mice. Vaccine 2008;26(January (4)):552–61.

[5] Smorodintsev AA, Zhdanov VM. Results and immediate tasks of the study oflive influenza vaccine. Voprosy Virusologii 1957;2(March–April (2)):67–72.

[6] Smorodintsev AA. Basic aspects of the study of live influenza vaccine. Klinich-eskaia Meditsina (Mosk) 1962;40(November):33–9.

[7] Maassab HF. Adaptation and growth characteristics of influenza virus at 25degrees C. Nature 1967;213(February (5076)):612–4.

[8] Jin H, Lu B, Zhou H, Ma C, Zhao J, Yang CF, et al. Multiple amino acid residues con-fer temperature sensitivity to human influenza virus vaccine strains (FluMist)derived from cold-adapted A/Ann Arbor/6/60. Virology 2003;306(February(1)):18–24.

[9] Youil R, Su Q, Toner TJ, Szymkowiak C, Kwan WS, Rubin B, et al. Comparativestudy of influenza virus replication in Vero and MDCK cell lines. Journal ofVirological Methods 2004;120(September (1)):23–31 [Comparative Study].

10] Kiseleva I, Su Q, Toner TJ, Szymkowiak C, Kwan WS, Rudenko L, et al. Cell-basedassay for the determination of temperature sensitive and cold adapted phe-notypes of influenza viruses. Journal of Virological Methods 2004;116(March(1)):71–8 [Comparative Study].

11] Palker T, Kiseleva I, Johnston K, Su Q, Toner T, Szymkowiak C, et al. Protectiveefficacy of intranasal cold-adapted influenza A/New Caledonia/20/99 (H1N1)vaccines comprised of egg- or cell culture-derived reassortants. Virus Research2004;105(October (2)):183–94.

12] Cox MM, Karl Anderson D. Production of a novel influenza vaccine using insectcells: protection against drifted strains. Influenza and Other Respiratory Viruses2007;1(January (1)):35–40 [Review].

13] Cox MM, Patriarca PA, Treanor J. FluBlok, a recombinant hemagglutinininfluenza vaccine. Influenza and Other Respiratory Viruses 2008;2(November(6)):211–9 [Multicenter Study, Randomized Controlled Trial, Review].

14] Cox MM, Hollister JR. FluBlok, a next generation influenza vaccine manufac-tured in insect cells. Biologicals 2009;37(June (3)):182–9 [Review].

15] Treanor JJ, Schiff GM, Hayden FG, Brady RC, Hay CM, Meyer AL, et al. Safetyand immunogenicity of a baculovirus-expressed hemagglutinin influenza vac-cine: a randomized controlled trial. The Journal of the American MedicalAssociation 2007;297(April (14)):1577–82 [Multicenter Study, RandomizedControlled Trial, Research Support, Non-U.S. Gov’t].

16] Treanor JJ, Betts RF, Smith GE, Anderson EL, Hackett CS, Wilkinson BE, et al.Evaluation of a recombinant hemagglutinin expressed in insect cells as an

influenza vaccine in young and elderly adults. The Journal of Infectious Dis-eases 1996;173(June (6)):1467–70 [Clinical Trial, Randomized Controlled Trial,Research Support, U.S. Gov’t, P.H.S.].

17] Treanor JJ, Wilkinson BE, Masseoud F, Hu-Primmer J, Battaglia R, O’Brien D,et al. Safety and immunogenicity of a recombinant hemagglutinin vaccine for

Page 6: New technologies for new influenza vaccines

4 30 (2

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

932 A. Shaw / Vaccine

H5 influenza in humans. Vaccine 2001;19(February (13–14)):1732–7 [ClinicalTrial, Controlled Clinical Trial, Research Support, U.S. Gov’t, P.H.S.].

18] Pushko P, Tumpey TM, Bu F, Knell J, Robinson R, Smith G. Influenzavirus-like particles comprised of the HA, NA, and M1 proteins of H9N2influenza virus induce protective immune responses in BALB/c mice. Vaccine2005;23(December (50)):5751–9 [Research Support, N.I.H., Extramural].

19] Song H, Wittman V, Byers A, Tapia T, Zhou B, Warren W, et al. In vitro stim-ulation of human influenza-specific CD8+ T cells by dendritic cells pulsedwith an influenza virus-like particle (VLP) vaccine. Vaccine 2010;28(August(34)):5524–32.

20] Pushko P, Kort T, Nathan M, Pearce MB, Smith G, Tumpey TM. RecombinantH1N1 virus-like particle vaccine elicits protective immunity in ferrets againstthe 2009 pandemic H1N1 influenza virus. Vaccine 2010;28(July (30)):4771–6.

21] Bright RA, Carter DM, Crevar CJ, Toapanta FR, Steckbeck JD, Cole KS, et al. Cross-clade protective immune responses to influenza viruses with H5N1 HA and NAelicited by an influenza virus-like particle. PLoS One 2008;3(1):e1501 [ResearchSupport, Non-U.S. Gov’t].

22] Khurana S, Wu J, Verma N, Verma S, Raghunandan R, Manischewitz J, et al.H5N1 virus-like particle vaccine elicits cross-reactive neutralizing antibodiesthat preferentially bind to the oligomeric form of influenza virus hemagglutininin humans. Journal of Virology 2011;85(November (21)):10945–54.

23] Pushko P, Tumpey TM, Van Hoeven N, Belser JA, Robinson R, Nathan M, et al.Evaluation of influenza virus-like particles and Novasome adjuvant as candi-date vaccine for avian influenza. Vaccine 2007;25(May (21)):4283–90.

24] Shoji Y, Farrance CE, Bautista J, Bi H, Musiychuk K, Horsey A, et al. A plant-basedsystem for rapid production of influenza vaccine antigens. Influenza and OtherRespiratory Viruses 2011;6(October):204–10.

25] Landry N, Ward BJ, Trepanier S, Montomoli E, Dargis M, Lapini G, et al. Preclin-ical and clinical development of plant-made virus-like particle vaccine againstavian H5N1 influenza. PLoS One 2010;5(12):e15559 [Clinical Trial, Phase I,Randomized Controlled Trial, Research Support, Non-U.S. Gov’t].

26] D’Aoust MA, Couture MM, Charland N, Trepanier S, Landry N, Ors F, et al. Theproduction of hemagglutinin-based virus-like particles in plants: a rapid, effi-cient and safe response to pandemic influenza. Plant Biotechnology Journal2010;8(June (5)):607–19 [Review].

27] Shapira M, Jolivet M, Arnon R. A synthetic vaccine against influenzawith built-in adjuvanticity. International Journal of Immunopharmacology1985;7(5):719–23 [Comparative Study].

28] McEwen J, Levi R, Horwitz RJ, Arnon R. Synthetic recombinant vaccine express-ing influenza haemagglutinin epitope in Salmonella flagellin leads to partialprotection in mice. Vaccine 1992;10(6):405–11.

29] Arnon R, Ben-Yedidia T. Preclinical efficacy of a virus-like particle-based vaccineagainst avian influenza H5N1. Future Microbiology 2009;4(June (5)):503–5[Comment].

30] Ben-Yedidia T, Arnon R. Towards an epitope-based human vaccine forinfluenza. Human Vaccines 2005;1(May–June (3)):95–101 [Review].

31] Rudolph W, Ben Yedidia T. A universal influenza vaccine: where are we inthe pursuit of this “Holy Grail”? Human Vaccines 2011;7(January (1)):10–1[Review].

32] Bright RA, Ross TM, Subbarao K, Robinson HL, Katz JM. Impact of glycosyla-tion on the immunogenicity of a DNA-based influenza H5 HA vaccine. Virology2003;308(April (2)):270–8 [Research Support, Non-U.S. Gov’t, Research Sup-port, U.S. Gov’t, P.H.S.].

33] Khurana S, Verma S, Verma N, Crevar CJ, Carter DM, Manischewitz J, et al.Bacterial HA1 vaccine against pandemic H5N1 influenza virus: evidence ofoligomerization, hemagglutination, and cross-protective immunity in ferrets.Journal of Virology 2011;85(February (3)):1246–56 [Research Support, Amer-ican Recovery and Reinvestment Act, Research Support, N.I.H., Extramural].

34] Treanor JJ, Taylor DN, Tussey L, Hay C, Nolan C, Fitzgerald T, et al. Safetyand immunogenicity of a recombinant hemagglutinin influenza-flagellinfusion vaccine (VAX125) in healthy young adults. Vaccine 2010;28(December(52)):8268–74 [Randomized Controlled Trial].

35] Liu G, Tarbet B, Song L, Reiserova L, Weaver B, Chen Y, et al. Immunogenicity andefficacy of flagellin-fused vaccine candidates targeting 2009 pandemic H1N1influenza in mice. PLoS One 2011;6(6):e20928 [Research Support, Non-U.S.Gov’t].

36] Taylor DN, Treanor JJ, Strout C, Johnson C, Fitzgerald T, Kavita U, et al. Inductionof a potent immune response in the elderly using the TLR-5 agonist, flagellin,with a recombinant hemagglutinin influenza-flagellin fusion vaccine (VAX125,STF2.HA1 SI). Vaccine 2011;29(July (31)):4897–902.

37] Leng J, Stout-Delgado HW, Kavita U, Jacobs A, Tang J, Du W, et al. Efficacy ofa vaccine that links viral epitopes to flagellin in protecting aged mice frominfluenza viral infection. Vaccine 2011;29(October (45)):8147–55.

38] Lamb RA, Choppin PW. Identification of a second protein (M2) encoded by RNAsegment 7 of influenza virus. Virology 1981;112(July (2)):729–37 [ResearchSupport, Non-U.S. Gov’t, Research Support, U.S. Gov’t, Non-P.H.S., ResearchSupport, U.S. Gov’t, P.H.S.].

39] Lamb RA, Lai CJ. Conservation of the influenza virus membrane protein (M1)amino acid sequence and an open reading frame of RNA segment 7 encod-ing a second protein (M2) in H1N1 and H3N2 strains. Virology 1981;112(July(2)):746–51 [Comparative Study, Research Support, Non-U.S. Gov’t, Research

Support, U.S. Gov’t, P.H.S.].

40] Pinto LH, Holsinger LJ, Lamb RA. Influenza virus M2 protein has ion chan-nel activity. Cell 1992;69(May (3)):517–28 [Research Support, Non-U.S. Gov’t,Research Support, U.S. Gov’t, P.H.S.].

[

012) 4927– 4933

41] Zebedee SL, Lamb RA. Influenza A virus M2 protein: monoclonal antibodyrestriction of virus growth and detection of M2 in virions. Journal of Virol-ogy 1988;62(August (8)):2762–72 [Research Support, Non-U.S. Gov’t, ResearchSupport, U.S. Gov’t, P.H.S.].

42] Gerhard W, Mozdzanowska K, Zharikova D. Prospects for universal influenzavirus vaccine. Emerging Infectious Diseases 2006;12(April (4)):569–74.

43] Fiers W, De Filette M, El Bakkouri K, Schepens B, Roose K, Schotsaert M,et al. M2e-based universal influenza A vaccine. Vaccine 2009;27(October(45)):6280–3 [Research Support, N.I.H., Extramural, Research Support, Non-U.S. Gov’t, Review].

44] 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: Fcreceptors and alveolar macrophages mediate protection. Journal of Immunol-ogy 2011;186(January (2)):1022–31 [Research Support, N.I.H., Extramural,Research Support, Non-U.S. Gov’t].

45] Jegerlehner A, Schmitz N, Storni T, Bachmann MF. Influenza A vaccinebased on the extracellular domain of M2: weak protection mediated viaantibody-dependent NK cell activity. Journal of Immunology 2004;172(May(9)):5598–605.

46] Fan J, Liang X, Horton MS, Perry HC, Citron MP, Heidecker GJ, et al. Preclinicalstudy of influenza virus A M2 peptide conjugate vaccines in mice, ferrets, andrhesus monkeys. Vaccine 2004;22(August (23–24)):2993–3003 [ComparativeStudy].

47] Huleatt JW, Nakaar V, Desai P, Huang Y, Hewitt D, Jacobs A, et al. Potentimmunogenicity and efficacy of a universal influenza vaccine candidate com-prising a recombinant fusion protein linking influenza M2e to the TLR5 ligandflagellin. Vaccine 2008;26(January (2)):201–14 [Comparative Study].

48] 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(July (32)):5145–52 [ResearchSupport, N.I.H., Extramural, Research Support, Non-U.S. Gov’t].

49] Talbot HK, Rock MT, Johnson C, Tussey L, Kavita U, Shanker A, et al.Immunopotentiation of trivalent influenza vaccine when given with VAX102, arecombinant influenza M2e vaccine fused to the TLR5 ligand flagellin. PLoS One2010;5(12):e14442 [Multicenter Study, Randomized Controlled Trial, ResearchSupport, Non-U.S. Gov’t].

50] Bianchi E, Liang X, Ingallinella P, Finotto M, Chastain MA, Fan J, et al. Universalinfluenza B vaccine based on the maturational cleavage site of the hemagglu-tinin precursor. Journal of Virology 2005;79(June (12)):7380–8.

51] Ekiert DC, Bhabha G, Elsliger MA, Friesen RH, Jongeneelen M, Throsby M,et al. Antibody recognition of a highly conserved influenza virus epitope. Sci-ence 2009;324(April (5924)):246–51 [Research Support, N.I.H., Extramural,Research Support, Non-U.S. Gov’t, Research Support, U.S. Gov’t, Non-P.H.S.].

52] Throsby M, van den Brink E, Jongeneelen M, Poon LL, Alard P, CornelissenL, et al. Heterosubtypic neutralizing monoclonal antibodies cross-protectiveagainst H5N1 and H1N1 recovered from human IgM+ memory B cells. PLoSOne 2008;3(12):e3942 [Research Support, Non-U.S. Gov’t].

53] Sui J, Hwang WC, Perez S, Wei G, Aird D, Chen LM, et al. Structural and func-tional bases for broad-spectrum neutralization of avian and human influenzaA viruses. Nature Structural & Molecular Biology 2009;16(March (3)):265–73[Research Support, N.I.H., Extramural].

54] Bommakanti G, Citron MP, Hepler RW, Callahan C, Heidecker GJ, Najar TA, et al.Design of an HA2-based Escherichia coli expressed influenza immunogen thatprotects mice from pathogenic challenge. Proceedings of the National Academyof Sciences of the United States of America 2010;107(August (31)):13701–6[Research Support, Non-U.S. Gov’t].

55] Graves PN, Schulman JL, Young JF, Palese P. Preparation of influenza virussubviral particles lacking the HA1 subunit of hemagglutinin: unmasking ofcross-reactive HA2 determinants. Virology 1983;126(April (1)):106–16 [Com-parative Study, Research Support, Non-U.S. Gov’t, Research Support, U.S. Gov’t,P.H.S.].

56] Wang TT, Tan GS, Hai R, Pica N, Ngai L, Ekiert DC, et al. Vaccination with asynthetic peptide from the influenza virus hemagglutinin provides protectionagainst distinct viral subtypes. Proceedings of the National Academy of Sciencesof the United States of America 2010;107(November (44)):18979–84 [ResearchSupport, N.I.H., Extramural, Research Support, Non-U.S. Gov’t].

57] Steel J, Lowen AC, Wang TT, Yondola M, Gao Q, Haye K, et al. Influenza virusvaccine based on the conserved hemagglutinin stalk domain. MBio 2010;1,10E000 18–10.

58] Garcia-Sastre A, Egorov A, Matassov D, Brandt S, Levy DE, Durbin JE, et al.Influenza A virus lacking the NS1 gene replicates in interferon-deficient sys-tems. Virology 1998;252(December (2)):324–30 [Research Support, Non-U.S.Gov’t, Research Support, U.S. Gov’t, P.H.S.].

59] Kochs G, Garcia-Sastre A, Martinez-Sobrido L. Multiple anti-interferon actionsof the influenza A virus NS1 protein. Journal of Virology 2007;81(July(13)):7011–21 [Research Support, N.I.H., Extramural, Research Support, Non-U.S. Gov’t].

60] Wacheck V, Egorov A, Groiss F, Pfeiffer A, Fuereder T, Hoeflmayer D, et al. A noveltype of influenza vaccine: safety and immunogenicity of replication-deficientinfluenza virus created by deletion of the interferon antagonist NS1. The Journalof Infectious Diseases 2010;201(February (3)):354–62 [Randomized Controlled

Trial, Research Support, Non-U.S. Gov’t].

61] Gao Q, Palese P. Rewiring the RNAs of influenza virus to prevent reassortment.Proceedings of the National Academy of Sciences of the United States of America2009;106(September (37)):15891–6 [Research Support, N.I.H., Extramural].

Page 7: New technologies for new influenza vaccines

30 (2

[

[

[

[

[

[

[

[

[

[

[

[

[

[

A. Shaw / Vaccine

62] Gao Q, Lowen AC, Wang TT, Palese P. A nine-segment influenza A virus car-rying subtype H1 and H3 hemagglutinins. Journal of Virology 2010;84(August(16)):8062–71 [Research Support, N.I.H., Extramural].

63] Co MD, Orphin L, Cruz J, Pazoles P, Rothman AL, Ennis FA, et al. Discordancebetween antibody and T cell responses in recipients of trivalent inactivatedinfluenza vaccine. Vaccine 2008;26(April (16)):1990–8 [Comparative Study,Research Support, N.I.H., Extramural].

64] La Gruta N, Kelso A, Brown LE, Chen W, Jackson DC, Turner SJ. Role of CD8(+)T-cell immunity in influenza infection: potential use in future vaccine devel-opment. Expert Review of Respiratory Medicine 2009;3(October (5)):523–37.

65] Goodman AG, Heinen PP, Guerra S, Vijayan A, Sorzano CO, Gomez CE, et al. Ahuman multi-epitope recombinant vaccinia virus as a universal T cell vaccinecandidate against influenza virus. PLoS One 2011;6(10):e25938.

66] Alexander J, Bilsel P, del Guercio MF, Stewart S, Marinkovic-Petrovic A, South-wood S, et al. Universal influenza DNA vaccine encoding conserved CD4+ Tcell epitopes protects against lethal viral challenge in HLA-DR transgenic mice.Vaccine 2010;28(January (3)):664–72 [Research Support, N.I.H., Extramural].

67] McMichael AJ. From influenza to HIV—and back? Nature Immunology2007;8(November (11)):1149–51 [Biography, Historical Article].

68] Schultz-Cherry S, Dybing JK, Davis NL, Williamson C, Suarez DL, Johnston R, et al.Influenza virus (A/HK/156/97) hemagglutinin expressed by an alphavirus repli-con system protects chickens against lethal infection with Hong Kong-originH5N1 viruses. Virology 2000;278(December (1)):55–9 [Research Support, Non-U.S. Gov’t, Research Support, U.S. Gov’t, Non-P.H.S., Research Support, U.S.

Gov’t, P.H.S.].

69] Hubby B, Talarico T, Maughan M, Reap EA, Berglund P, Kamrud KI, et al. Develop-ment and preclinical evaluation of an alphavirus replicon vaccine for influenza.Vaccine 2007;25(November (48)):8180–9 [Research Support, N.I.H., Extramu-ral].

[

012) 4927– 4933 4933

70] Breathnach CC, Clark HJ, Clark RC, Olsen CW, Townsend HG, Lunn DP.Immunization with recombinant modified vaccinia Ankara (rMVA) constructsencoding the HA or NP gene protects ponies from equine influenza virus chal-lenge. Vaccine 2006;24(February (8)):1180–90 [Research Support, Non-U.S.Gov’t].

71] Ben-Yehuda A, Ehleiter D, Hu AR, Weksler ME. Recombinant vaccinia virusexpressing the PR/8 influenza hemagglutinin gene overcomes the impairedimmune response and increased susceptibility of old mice to influenza infec-tion. The Journal of Infectious Diseases 1993;168(August (2)):352–7 [ResearchSupport, U.S. Gov’t, P.H.S.].

72] Van Kampen KR, Shi Z, Gao P, Zhang J, Foster KW, Chen DT, et al. Safety andimmunogenicity of adenovirus-vectored nasal and epicutaneous influenza vac-cines in humans. Vaccine 2005;23(January (8)):1029–36 [Clinical Trial, PhaseI, Comparative Study, Research Support, Non-U.S. Gov’t, Research Support, U.S.Gov’t, Non-P.H.S., Research Support, U.S. Gov’t, P.H.S.].

73] Berthoud TK, Hamill M, Lillie PJ, Hwenda L, Collins KA, Ewer KJ, et al. PotentCD8+ T-cell immunogenicity in humans of a novel heterosubtypic influenza Avaccine, MVA-NP + M1. Clinical Infectious Diseases 2011;52(January (1)):1–7[Clinical Trial, Phase I, Research Support, Non-U.S. Gov’t].

74] Lillie PJ, Berthoud TK, Powell TJ, Lambe T, Mullarkey C, Spencer AJ, et al. Apreliminary assessment of the efficacy of a T cell-based influenza vaccine, MVA-NP + M1, in humans. Clinical Infectious Diseases 2012;(March).

75] Kreijtz JH, Suezer Y, de Mutsert G, van den Brand JM, van AmerongenG, Schnierle BS, et al. Preclinical evaluation of a modified vaccinia virus

Ankara (MVA)-based vaccine against influenza A/H5N1 viruses. Vaccine2009;27(October (45)):6296–9 [Research Support, Non-U.S. Gov’t].

76] Marazzi I, Ho JS, Kim J, Manicassamy B, Dewell S, Albrecht RA, et al. Sup-pression of the antiviral response by an influenza histone mimic. Nature2012;483(March (7390)):428–33.