Você está na página 1de 11

See

discussions, stats, and author profiles for this publication at: http://www.researchgate.net/publication/51064197

Influenza virosomes as a vaccine adjuvant and


carrier system. Expert Rev Vaccines
ARTICLE in EXPERT REVIEW OF VACCINES APRIL 2011
Impact Factor: 4.21 DOI: 10.1586/erv.11.15 Source: PubMed

CITATIONS

READS

40

51

3 AUTHORS:
Christian Moser

Mario Amacker

Pevion Biotech

Mymetics SA

30 PUBLICATIONS 1,325 CITATIONS

48 PUBLICATIONS 1,736 CITATIONS

SEE PROFILE

SEE PROFILE

Rinaldo Zurbriggen
Lonza
63 PUBLICATIONS 1,711 CITATIONS
SEE PROFILE

Available from: Mario Amacker


Retrieved on: 03 December 2015

Review

SPECIAL FOCUS y Adjuvants


www.expert-reviews.com/toc/erv/10/4
For reprint orders, please contact reprints@expert-reviews.com

Influenza virosomes as a vaccine


adjuvant and carrier system
Expert Rev. Vaccines 10(4), 437446 (2011)

Christian Moser1
Mario Amacker1 and
Rinaldo Zurbriggen2
Pevion Biotech AG,
Worblentalstrasse32, CH-3063 Ittigen,
Switzerland
2
Lonza AG, CH-3930 Visp, Switzerland

Author for correspondence:


Tel.: +41 315 504 424
Fax: +41 315 504 445
christian.moser@pevion.com
1

Influenza virosomes have been used for more than 10 years in commercial vaccines. The
technology has been further developed as a carrier and adjuvant system for subunit vaccines,
in particular for synthetic peptides. The extensive amount of preclinical and clinical data supports
the notion that influenza virosomes represent a platform technology that ensures robust and
long-lasting immune responses against subunit antigens with an excellent safety profile.
Structurally and functionally, virosomes are enveloped virus-like particles, although they are
assembled invitro. This unique feature ensures a tight control of their composition and at the
same time provides the flexibility to adapt the particle to various types of antigens. The mode
of action of virosomes is complex and includes carrier as well as immune-stimulatory functions.
Keywords : adjuvant antigen delivery influenza vaccines virosomes

The need for safe and efficacious vaccines has


promoted the use of subunit antigens in order
to remove antigenic components that are unrelated to protection, potentially harmful or even
immune suppressive. Subunit antigens can be
purified from the respective pathogens, as illustrated by influenza vaccines, but recombinant proteins or synthetic peptides are increasingly being
used. However, the reduction in antigen complexity in combination with a high purity frequently
impacts negatively on the immunogenicity.
The induction of an adaptive immune
response via vaccination is a highly complex process, which requires several distinct cell types to
interact, differentiate and migrate between the
administration site and lymphatic tissue within
a time course of several weeks [1,2] . The goal of
any vaccination is to generate populations of
mature, antigen-specific effector and accessory
cells, including memory cells, for sustained,
long-lasting immunity. Vaccine adjuvants can
come to action at any stage of this process in
order to amplify the desired type of immune
response [1] .
Historically, vaccine adjuvants were identified
empirically by evaluation of their potential to
enhance immune responses against coadministered antigens. Novel insights into the interplay
between innate and adaptive immunity have
not only led to more rational selection of novel
adjuvant candidates, such as specific ligands of
innate danger and tissue damage sensors [35] ,
but have also provided explanations for the
www.expert-reviews.com

10.1586/ERV.11.15

mode of action of established adjuvants [38] .


Antigen-presenting cells (APCs), in particular
dendritic cells (DCs), play a central role in this
process. DCs express a wide range of innate sensor molecules and strongly influence the activation of antigen-specific B and T lymphocytes in
reaction to innate stimuli [1,6,9,10] .
In-depth knowledge of the mode of action
of adjuvants provides a rational basis for risk
assessments, and for the design of conclusive
toxicology studies and clinical trials. It should
be emphasized that safety concerns are more difficult to address in clinical trials than immuno
genicity because severe side effects are not
acceptable even at very low frequencies [11,12] .
Therefore, a thorough safety assessment is also
the key issue in regulatory guidelines for clinical testing and market approval of novel adjuvants [1316] . A wide variety of adjuvants are
capable of enhancing the immunogenicity of
weak antigens but, aside from aluminum salts,
only very few are actually applied in licensed
human vaccines, namely MF59 (Novartis, Basel,
Switzerland), AS03 and AS04 (GSK, London,
UK), and influenza virosomes (Crucell, Leiden,
The Netherlands; Solvay, Brussels, Belgium).
Virus-like particles (VLPs) represent a distinct class of adjuvants. Aside from their individual components, their particle characteristics
contribute to their function as adjuvants, and
thus, they have a complex mode of action [3,17] .
Originally, VLPs were defined as nonreplicating, recombinant virus capsids [18,19] , but it

2011 Expert Reviews Ltd

ISSN 1476-0584

437

Review

Moser, Amacker & Zurbriggen

seems appropriate to expand the definition and to include viruslike structures derived from enveloped viruses [20,21] . Common
to all VLPs is their particulate structure that mimics a virus
combined with their complete inability to replicate. These features ensure a high safety level combined with the advantages
of a VLP structure enhancing the immune stimulation and the
stability of the antigen.
Virus-like particles can be derived from a variety of nonenveloped and enveloped viruses, and are generally manufactured
in recombinant expression systems. VLPs are applied in several licensed vaccines (hepatitis B virus, human papillomavirus,
influenza and hepatitis A virus) and in a considerable number
of prophylactic and therapeutic vaccine candidates in clinical
development in the field of infectious diseases, cancer and allergy.
Originally, VLPs represented just another form of vaccine
antigen, but their unique versatility soon promoted their use as
carrier and adjuvant systems suitable for heterologous antigens,
where they provide a higher, immune-potentiating particle structure [3,2226] . Most VLP-based vaccines are combined with other
adjuvant types such as alum salts and Toll-like receptor (TLR)
agonists, while influenza-derived VLPs, including virosomes, are
generally used as stand-alone products.
Virosome technology

The concept of virosomes and the idea to use them as vaccines


dates back to the 1970s [27,28] . Virosomes are spherical, unilamellar vesicles composed of membrane lipids and integrated viral
envelope proteins. Unlike most VLPs, virosome particles are not
produced by host cells but assembled invitro in a three-step process from the parental virus. First, the purified and inactivated
virus is dissolved in detergent. Second, the envelope fraction
comprising the membrane-associated viral proteins and lipids
is separated from the core complex containing the genetic material and internal proteins. Third, detergent removal triggers the
formation of empty membrane vesicles.
This simple principle of assembling empty envelope particles
invitro is applicable to all enveloped viruses, for the purpose of
functional studies or as vaccine candidates, most recently for HIV
and respiratory syncytial virus [25,29,30] .
Influenza virus was used in the early studies on virosomes[27,28] .
In the following years, the production of influenza virosomes was
further optimized [22,31] and established as an industrial scale
process compliant with good manufacturing practice (GMP)
guidelines [32] . To date, influenza virosomes remain the only
virosome type that is applied in licensed human vaccines[33,34] .
In the past few years, several clinical trials have been performed,
using influenza virosomes as a carrier and adjuvant system for
heterologous subunit antigens [3537] . The dominance of the
influenza virus as the substrate for virosome-based products may
be explained by two aspects, namely, by the biological properties of the viral envelope proteins, in particular of influenza
hemagglutinin (HA), and by the availability of virus material.
GMP-grade influenza virus is produced in embryonated chicken
eggs every year for several hundred million doses of influenza
vaccines. The same material has proven suitable and cost-efficient
438

for the generation of influenza virosomes. Notably, influenza


virus grown in cell culture is equally suited for the production
of virosomes [201] .
The envelope of influenza virus contains predominantly HA
trimers, approximately fivefold less neuraminidase tetramers,
and traces of M2e tetramers [38] . All these proteins are also present in influenza virosomes in similar ratios as in the parental
virus. Retaining the authentic conformation and functionality
of the viral proteins is a key aspect of the virosome formulation
process, as exemplified by the pH-dependent fusion activity of
HA[39,40] . As a consequence, a thorough quality control of virosomal vaccines goes far beyond content analysis and includes
particle characterization, as well as a verification of the fusion
activity [40] .
The addition of phospholipids to the solubilized viral envelope
fraction has proven essential for a robust, scalable particle assembly step. The addition of lipids minimizes the loss of influenza
components during detergent removal and ensures the assembly
of a homogenous particle population that renders further purification steps obsolete [32] . However, it should be noted that the
density of the envelope proteins on the virosome surface is reduced
in comparison to the parental virus particle, since the density is a
direct function of the protein:lipid ratio. The physico-chemical
and biological properties of the resulting virosome particles can
be modulated not only by the amount, but also by the type of
lipids added [4143] . The addition of charged lipids has proven
useful to associate and deliver negatively charged molecules such
as nucleic acids [39,44,45] .
Conventional influenza virosomes have a limited stability
unless stored at 28C, and are particularly sensitive to freezing. The addition of sugars, with or without cationic lipids,
was shown to protect the virosomes from the negative effects
of freezing and to generate temperature-insensitive virosomes
that can be lyophilized. These formulations are stable and retain
their fusion activity in a broad temperature range [46,47] . In
addition, lyophilization can stabilize labile subunit antigens,
and enables a novel approach to efficiently encapsulate small
antigens like peptides, by reconstituting lyophilized empty
virosomes with a peptide solution [46] . Influenza virosomes are
protected by a considerable number of patents, covering multiple aspects of use as a vaccine platform and drug delivery
system, the formulation process including lyophilization and
combination with other adjuvants.
A wide range of antigen types, including peptides, proteins
and carbohydrates, have been combined successfully with influenza virosomes in order to obtain immunogenic vaccine candidates [4856] . In addition, virosomes were shown to function as
delivery system for nucleic acids and drugs [39,44,45,57] .
The physical association of the payload, the antigen of interest (AoI), with the virosome particle is a prerequisite for the full
immune-enhancing function [49,58] . Depending on the preferred
type of immunity, antibody or cytotoxic T lymphocytes (CTL),
the antigen is either formulated to be displayed on the surface or
encapsulated in the lumen, respectively [40,59] . For surface display, the AoI needs to be anchored in the lipid bilayer, either
Expert Rev. Vaccines 10(4), (2011)

Influenza virosomes as a vaccine adjuvant & carrier system

via a hydrophobic protein domain in the AoI, or via a synthetic


phospholipid conjuguated to the AoI. By contrast, efficient encapsulation is achieved by a specifically designed formulation process [46,48] , but does not require integration of the AoI into the
lipid membrane.
Antigens of interest displayed on the outer surface of the virosomes in a repetitive array are optimal to prime B lymphocytes for
antibody production. After uptake by APCs, AoI on the virosome
surface are processed within the late endosome and presented in
the context of MHC class II to generate T-helper cells. For AoI
encapsulated in the lumen, the pH-dependent fusion activity of
HA enables virosomes to act as cytoplasmatic delivery vehicles.
After uptake by APCs, the acidification within the late endosome
triggers HA-mediated fusion between the virosomal and endosomal membrane. As a result, the encapsulated AoI is released into
the cytoplasm, thereby providing access to MHC class I presentation and CTL induction [46,48,6062] . CTL induction critically
depends on the fusion activity of HA [63] .
Mode of action

Virus-like particles including virosomes exert their effects on


APCs and on antigen-specific lymphocytes via their overall particle structure and their individual components [3,17,23,64] . In
addition, pre-existing immunity significantly impacts on the
adjuvant function of virosomes [40,49] . Therefore, virosomes represent an excellent example for a multifunctional carrier and
adjuvant system (Figure1).

Carrier

Review

Adjuvant

Delivery
Antigen protection

Repetitive
antigen display

PAMP signals
Costimulation

VLP
structure
APC targeting

CD4+ help

Pre-existing immunity
Antibodies

T-cell memory

Figure1. Multifunctional mode of action of virosomes.


Schematic representation of the overlapping functions of
virosomes to highlight the central role of the VLP structure. The
carrier functions result predominantly from the particle structure.
The adjuvant functions relate to both the particle structure and
the individual virosome components with stimulatory effects and
synergistic potential. Pre-existing immunity against influenza
affects the carrier as well as the adjuvant functions. Antibodies
interact directly with the particle structure, while cellular immunity
expands the pool of T-helper cells.
APC: Antigen-presenting cell; PAMP: Pathogen-associated
molecular pattern; VLP: Virus-like particle.

Carrier functions

The carrier function is related to the particle structure and plays


a key role in the early events after vaccine administration. The
VLP structure facilitates a rapid uptake by APCs. The embedding of the AoI in the virosome particle can also protect it from
premature degradation by extracellular proteases. Virosomes
are rapidly taken up by cells, both invitro and invivo [58,62,65] .
When administered via intramuscular injection, virosomes can
access the draining lymph node via free lymph drainage, or in
association with migrating cells. The free draining hypothesis
is supported by a recent report, demonstrating that nanoparticles up to 200nm in size and nonenveloped VLPs are detectable in APCs resident in the lymph node, but larger particles
were found only in macrophages originating from the injection
site[66] . Within the lymph node, virosomes are thought to interact with B cells as intact particles, as it has been shown for other
viruses and particulate antigens [67,68] . Therefore, the repetitive display of the AoI on the virosome surface acts as a strong
activation signal for antigen-specific B cells via cross-linking of
immunoglobulin receptors [69,70] .

invitro stimulation with virosomes [62] , although this cell line


is highly responsive to both DNA and RNA. Nucleic acids
are degraded by treatment with b-propiolactone, which is performed to inactivate the influenza virus prior to virosome assembly. Accordingly, no nucleic acids are detectable in virosomes.
However, recombinant influenza HA was reported to activate
both murine and human DCs invitro [7173] . Further investigations are needed to clarify whether virosomes can directly
activate DCs, and if so, by which mechanisms. Virosomes have
been shown to stimulate human peripheral blood mononuclear
cells (PBMCs) invitro, resulting in cytokine secretion (TNFa, GM-CSF, IFN-g and IL-2 but not IL-4) consistent with a
Th1 profile, and proliferation of influenza-specific memory T
cells in PBMCs obtained from adult donors but not from cord
blood [61,74] . Notably, TNF-a and GM-CSF were secreted at
high levels rapidly after exposure to virosomes, suggesting that
these cytokines at least do not originate from the proliferating
memory cells but from a more abundant, so far unidentified,
cell type present in PBMC preparations.

Adjuvant functions

Role of pre-existing immunity

The adjuvant effect of virosomes relates to their ability to


enhance antigen processing and presentation by APCs.
Direct activation of DCs by virosomes was demonstrated in
the murine system [63] , while a human plasmacytoid DC cell
line did not increase expression of activation markers upon

Pre-existing immunity against the carrier proteins can impair


the function of viral vectors and VLPs, and quench the immune
response against the AoI [7577] . By contrast, the immune potentiating effect of influenza virosomes is enhanced by pre-existing immunity against influenza. This feature is of particular

www.expert-reviews.com

439

Review

Moser, Amacker & Zurbriggen

Reactivation of influenza-specific memory cells, as demonstrated in human PBMCs [74] , is considered the main cause
for cell-mediated immune enhancement. B cells specific for
the AoI are activated upon contact with the antigen displayed
on the surface of virosomes. Subsequently, these cells process
and present all virosome components in the context of MHC
classII, including the influenza proteins. Therefore, these cells
can obtain their signal 2 not only from antigen-specific but also
from influenza-specific CD4 + T-helper cells. In a situation of
pre-existing immunity, influenza-specific CD4 + T cells are more
abundant and faster activated than naive CD4 + T cells reactive
to the AoI. Therefore, influenza-specific CD4 + memory cells can
significantly contribute to the differentiation and proliferation
of AoI-specific effector cells.
Multiple infections by constantly drifting virus strains result
in a complex immunity against influenza in the human population. For that reason, no conclusive clinical data are available
on the positive effect of pre-existing immunity on the response
against a heterologous AoI applied in the context of virosomes.
The hemagglutination inhibition (HI) titer against the virus
strain used in virosomes A/Singapore/6/86 (H1N1) was measured in several clinical trials. Consistently, no significant correlation was found between the response against hepatitis A virus
and the baseline HI titer against influenza, thereby excluding at
least a negative effect [33,80] . However, the value of these data is
limited because HI titers account for neither humoral immunity
against heterologous influenza strains nor
for any cellular immunity. No correlation
Anti-virosome A d21
100,000
was detectable between either pre-existing
Anti-malaria d42
cellular immunity against influenza and
Anti-malaria d63
the response against the malaria-derived
10,000
peptides formulated on virosomes [79] .
Without truly naive subjects available as
negative controls, it appears extremely difficult to clinically verify the positive cor1000
relation between baseline anti-influenza
immunity and response to the AoI, as it
has been clearly demonstrated in animal
100
models [49] .
Further preclinical and clinical studies
are necessary to dissect the contributions
10
of the different elements and to determine
None
Influenza A
Influenza B
Virosome A
whether the positive effect directly correlates
Pre-immunization
with the magnitude of pre-existing immunity or rather requires threshold levels of
Figure2. Positive effect of pre-existing immunity against influenza is strain
pre-existing humoral and cellular immunity.
ELISA titer

importance since pre-existing immunity against influenza is


widespread among the human population and can be detected
in nearly every individual [74,78,79] .
Preclinical studies in mice have clearly demonstrated that
pre-existing immunity against influenza enhances the antibody
response against unrelated antigens administered subsequently
in the context of virosomes [49] . The enhancer effect is strain
independent, since immunity against influenza B had the same
positive effect as the homologous influenza A/H1N1 strain used
to assemble the virosomes (Figure2) . By contrast, the magnitude
of CTL responses against virosome-encapsulated peptides was
not significantly increased by pre-existing immunity against
influenza [46,48] .
Both humoral and cellular elements of pre-existing immunity
against influenza are thought to contribute to the immuneenhancing effects. Anti-influenza antibodies are thought to target virosomes to Fc receptors of APCs, thereby accelerating and
enhancing uptake by APCs. For this purpose, the antibodies
need neither virus neutralizing nor hemagglutination inhibiting
capacity. Mere binding to any of the influenza proteins present
on virosomes is sufficient. This hypothesis is consistent with the
observed strain-type independence. A speculative explanation for
the lack of enhanced CTL responses is interference of antibodies
with the HA function necessary for the cytoplasmatic delivery,
which might be sufficient to compensate for the positive effects
of improved APC targeting.

independent. BALB/c mice were pre-immunized at day 0 with either influenza A/H1N1
virus, influenza B virus or virosomes made from homologous influenza A. At day 21 and
day 42, the animals were immunized with a virosomal vaccine composed of a
malariaderived peptide and virosomes assembled from influenza A. Antibody titers were
determined by ELISA: first, against influenza A virosomes at day 21, before the first
immunization with the virosomal peptide vaccine (anti-Virosome A d21) and, second,
against the malaria peptide 3weeks after the first and second immunization with the
virosomal peptide vaccine, respectively (anti-malaria d42 and anti-malaria d43). In the
presence of pre-existing immunity, a single immunization induces antipeptide titers
comparable to those achieved with two immunizations in naive animals.
Data provided by Pevion Biotech AG (Ittigen, Switzerland).

440

Clinical experience with virosomes

Over the last two decades, a considerable


amount of clinical data has been generated with influenza virosomes. Four vaccines based on the virosome technology
are licensed for commercial use, while over
70million doses of Crucells influenza and
hepatitis A vaccines have been distributed
Expert Rev. Vaccines 10(4), (2011)

Influenza virosomes as a vaccine adjuvant & carrier system

so far (Table1) . Since 2005, five novel product candidates have been tested in clinical
trials or are part of ongoing studies, all
of them based on subunit antigens in the
form of synthetic peptides or recombinant
proteins (Table2) .
Hepatitis A vaccines

Review

Table 1. Licensed vaccines based on virosomes.


Product

Vaccine composition

Launched
(year)

Ref.

Epaxal (Crucell,
Leiden,
TheNetherlands)

A(H1N1) virosomes +
inactivated hepatitis A virus

1994

[33,81]

Inflexal V (Crucell)

Virosomes from three influenza strains 1997


A(H1N1), A(H3N2) and B

[34]

The hepatitis A vaccine, Epaxal (Crucell),


was approved for commercial use in 1994 Nasalflu (Berna
[89,90]
Virosomes from three influenza strains 2000,
as the first vaccine on the basis of the influ- Biotech, Bern,
A(H1N1), A(H3N2) and B + heat labile withdrawn 2001
toxin adjuvant
enza virosome technology. The virosomes Switzerland)
in this product are generated from the Invivac (Solvay,
[91,92]
Virosomes from three influenza strains 2004,
influenza strain A/Singapore/6/86 (H1N1) Brussels, Belgium)
A(H1N1), A(H3N2) and B
not on the market
since 2005
and coated after assembly with the inactivated hepatitis A virus as the AoI. The Epaxal Junior (Crucell) A(H1N1) virosomes + inactivated
[33,80]
2008
same virus strain has been used for all subhepatitis A virus
sequent applications of influenza virosomes
as carrier and adjuvant system for heterologous antigens.
influenza strain [32] . Nasalflu (Berna Biotech, Bern, Switzerland)
Epaxal and the follow-up product Epaxal Junior were character- was a virosomal influenza vaccine for intranasal application, which
ized in numerous clinical studies with regard to safety, immunoge- contained heat labile toxin from Escherichia coli as an additional
nicity and efficacy [33,81] . Notably, when compared with alum-adju- mucosal adjuvant [89] . The product was launched in 2000, but
vanted competitor products, the virosomal products demonstrated withdrawn from the market due to an association between heat
in several independent studies a significantly better tolerability[8285] labile toxin adjuvanted vaccination and Bells palsy [90] . In 2004,
and a clear trend towards improved immunogenicity [80,82] .
Solvay launched Invivac, a trivalent seasonal influenza vaccine
for intramuscular injection based on the virosome concept [91,92] .
Influenza vaccines
However, the product was only commercially available during the
When the virosome concept is applied to influenza vaccines, season 2004/2005.
the basic building blocks of virosomes, namely the influenza
proteins themselves, represent the AoI. Compared to conven- Vaccines in clinical development
tional, nonparticulate subunit influenza vaccines, the assembly Malaria
of virosomes provides the antigens a VLP structure and native Based on two synthetic peptides, a bivalent malaria virosomal vacpresentation. Both aspects are expected to improve the immuno cine was developed to induce protective antibodies against sporozological properties [59,86] . By contrast to the application of viro- ite and blood-stage parasites [93,94] . The vaccine has been tested in
somes for heterologous antigens, the positive effects related to the three clinical trials, including a Phase I safety trial, an experimental
presence of influenza proteins, such as stimulatory signals and challenge study and a PhaseI trial in an endemic area (Tanzania).
expanded CD4 + T-cell help, do not come into play in the con- The vaccine proved to be safe and immunogenic, and induced
text of influenza vaccines. Virosomes and
conventional subunit vaccines essentially Table2. Virosome-based vaccines in clinical development.
contain the same components. Therefore,
Ref.
when applied to influenza vaccines, the Target Vaccine composition Status
[36,37,79,95,96]
role of virosome is limited to providing Malaria Two peptides
Three Phase I/II trials with bivalent
+ A(H1N1) virosomes
vaccine completed:
the antigen a particle structure.
safe and immunogenic
Comparative clinical studies with conventional subunit vaccines demonstrated Hepatitis Three peptides
PhaseI completed:
[46,100]
[Pevion Biotech AG,
+ A(H1N1) virosomes
safe but insufficiently
that the virosome formulation did not sig- C virus
Unpublished Data]
immunogenic
nificantly increase immunogenicity, but did
[35]
improve tolerability [34,87] . Whether the lat- Breast
Three peptides
PhaseI completed:
+ A(H1N1) virosomes
safe and immunogenic
ter results from the particle structure or from cancer
the higher purity of the product remains HIV
One peptide
PhaseI initiated 2010
[301]
[Pevion Biotech AG,
unclear [88] . InflexalV (Crucell) is a triva+ A(H1N1) virosomes
Unpublished Data]
lent seasonal influenza vaccine successfully
[99,101,302]
Candida
One
recombinant
PhaseI
initiated
2010
marketed since 1997 [34] . The product is
albicans
protein
+
A(H1N1)
a blend of three different virosome parvirosomes
ticles, each assembled separately from one

www.expert-reviews.com

441

Review

Moser, Amacker & Zurbriggen

long-lasting functional antibody titers [36,37,95] . The challenge study


showed evidence of blood-stage efficacy [37] , illustrated by lower
rates of parasite growth and by appearance of morphologically
abnormal parasites in vaccinated volunteers. The third study demonstrates the safety and immunogenicity results for the lyophilized
vaccine formulation, comparable to the previous studies performed
with the liquid, temperature sensitive form of the vaccine [96] .
Hepatitis C virus

The virosomal hepatitis C virus vaccine was designed as a therapeutic T-cell vaccine to clear chronic hepatitis C virus infection.
The vaccine included two peptides representing CTL epitopes,
encapsulated in virosomes, and a peptide acting as a CD4 epitope
displayed on the virosome surface. Preclinical models demonstrated induction of both peptide-specific CTL and CD4 + lymphocytes[46] . In the Phase I clinical trial the vaccine was well tolerated
and safe but failed to induce satisfactory levels of T-cell responses
[Pevion Biotech, Unpublished Data] . It remains unclear whether this
shortcoming is due to the peptide design, or indicates a limitation
of the virosome technology.
Breast cancer

The B-cell vaccine was designed to induce antibodies against Her2,


a target validated by the established passive immunotherapy with
Trastuzumab [97] . Encouraging preclinical results with a virosome
formulation containing three synthetic peptides warranted a Phase
I clinical trial. Beyond meeting the safety end points, the vaccine
induced antibodies in eight out of ten breast cancer patients [35] ,
recognizing not only the peptides in the vaccine but also the fulllength Her2/neu protein. Notably, this response was achieved with
only 10g of each peptide.
HIV

The concept of the prophylactic HIV vaccine is to induce a protective mucosal immunity that has been shown to efficiently prevent
infection [98] . The candidate vaccine is composed of subunit antigens derived from HIV gp41 displayed by virosomes. The liquid
formulation was designed for intramuscular as well as mucosal
administration, and both routes are currently being tested with a
monovalent peptide vaccine in a Phase I clinical trial for safety and
immunogenicity [301] .
Candida

The Candida vaccine aims to prevent recurrent vulvovaginal candidiasis. The vaccine is based on a recombinant, truncated form
of the Sap2 protein, a virulence factor of Candida albicans [99] .
Lyophilized virosome formulations for intramuscular and intravaginal application were developed with the intention to induce
mucosal immunity via a systemic prime/mucosal boost regime. The
vaccine presentation in a capsule for intravaginal application represents a novelty with advantages regarding delivery and patient compliance. The capsule is designed to dissolve upon administration,
resulting in the reconstitution of the lyophilized vaccine invivo,
directly on the mucosal surface. The vaccine is currently being
tested in a Phase I clinical trial for safety and immunogenicity [302] .
442

Expert commentary

The regulatory hurdles for clinical testing and market licensure of


novel vaccines have become increasingly stringent, with a strong
focus on safety and product definition. Subunit vaccines meet
these requirements, in particular when based on synthetic peptides and recombinant proteins. However, owing to their generally
poor immunogenicity, subunit vaccines frequently need to be
combined with adjuvants, which in turn raise safety issues on their
own. Thus, the mode of action of adjuvants has become a central
theme in vaccinology, driven by increasingly detailed insights into
the underlying biological mechanisms.
The knowledge of the mode of action of influenza virosomes
remains fragmented, but there is sufficient data to declare them
multifunctional. Although difficult to investigate, this complexity might be the key to a balanced, efficient, but well-tolerated
immune stimulation, as it mimics the broad range of stimulatory
signals mediated by a natural infection.
With three products on the market, 15years of clinical experience and 70million doses distributed to date, a solid clinical track
record for virosomes has been established, which emphasizes the
safety and efficacy of the technology [33,34] . Over the past 5years,
early stage clinical studies have demonstrated that the same virosome technology is a safe and potent adjuvant system for synthetic
peptides designed as B-cell vaccines against malaria and breast
cancer [35,36,79,95] . Antibodies were induced even in elderly breast
cancer patients, notably against low dosages of peptides derived
from self-antigen [35] . However, the T-cell vaccine approach for
hepatitis C virus failed to induce robust responses in humans
despite encouraging preclinical data [46] .
Unlike most VLPs, virosomes are assembled invitro in a tightly
controlled process. Modifications of the particle structure or
inclusion of additional compounds can be achieved independently
of a host cell system, which saves time and increases reproducibility. The successful development of a temperature-insensitive,
lyophilized form of virosomes illustrates the flexible application
of the system [46] .
Five-year view

A further validation of the virosome technology can be expected


from ongoing clinical studies with new targets (e.g., Candida
and HIV), thereby expanding the antigen spectrum from synthetic peptides to larger recombinant proteins. In addition, both
approaches include a mucosal route of application and may thus
provide further information on the potential of virosomes as a
mucosal adjuvant system. The clinical proof of concept for virosomal subunit vaccines represents the next level of validation,
specifically the demonstration of protective efficacy in humans.
Novel antigens and targets for the virosome technology are
continuously being evaluated at the preclinical stage and are likely
to enter the clinical development phase in the near future.
The mode of action of adjuvants will remain a key topic in vaccinology, and novel insights can be expected on a regular basis.
Regarding virosomes, further investigations on the mode of action
are needed to better define the carrier and adjuvant functions, and
the impact of pre-existing immunity.
Expert Rev. Vaccines 10(4), (2011)

Influenza virosomes as a vaccine adjuvant & carrier system

Financial & competing interests disclosure

Christian Moser and Mario Amacker are employees of Pevion Biotech AG.
All three authors are shareholders of Pevion Biotech AG. The authors have
no other relevant affiliations or financial involvement with any organization

Review

or entity with a financial interest in or financial conflict with the subject


matter or materials discussed in the manuscript apart from those
disclosed.
No writing assistance was utilized in the production of this manuscript.

Key issues
Virosomes are enveloped virus-like particles assembled invitro, composed of purified or synthetic phospholipids and the envelope
fraction of influenza virus.
Virosomes are a commercially validated vaccine platform with an excellent safety and efficacy track record.
Synthetic peptides displayed on virosomes are safe and immunogenic in humans, even at low doses.
Virosomes are an adjuvant and carrier system for subunit vaccines; owing to their structure and composition they feature a
multifunctional mode of action.
Pre-existing immunity against influenza has no negative effect on the functionality of virosomes as adjuvant and carrier system.
The invitro assembly process is current good manufacturing practice compatible and established at commercial scale. It allows for
flexible particle design and integration of various antigen types. The lyophilized form of virosomes gives access to numerous novel
applications of the technology.

References

Kool M, Petrilli V, De Smedt T etal.


Cutting edge: alum adjuvant stimulates
inflammatory dendritic cells through
activation of the NALP3 inflammasome.
J.Immunol. 181(6), 37553759 (2008).

Describes a novel, specific


immunostimulatory effect of alum.

Tritto E, Mosca F, De Gregorio E.


Mechanism of action of licensed vaccine
adjuvants. Vaccine 27(2526), 33313334
(2009).

Papers of special note have been highlighted as:


of interest
of considerable interest
1

Ribeiro CMS, Schijns VEJC. Immunology


of vaccine adjuvants. In: Vaccine Adjuvants.
Davies G (Ed.). Springer, Berlin, Germany
114 (2010).
Comprehensive review of various classes of
vaccine adjuvants and their mode of action.
Wilson-Welder JH, Torres MP, Kipper MJ,
Mallapragada SK, Wannemuehler MJ,
Narasimhan B. Vaccine adjuvants: current
challenges and future approaches. J.Pharm.
Sci. 98(4), 12781316 (2009).

Review of particulate adjuvants and their


interaction with the immune system.

Ishii KJ, Akira S. Toll or Toll-free adjuvant


path toward the optimal vaccine
development. J.Clin. Immunol. 27(4),
363371 (2007).

10

Bachmann MF, Jennings GT. Vaccine


delivery: a matter of size, geometry, kinetics
and molecular patterns. Nat. Rev. Immunol.
10(11), 787796 (2010).

Overview of signaling pathways activated


by various adjuvant types.
Pashine A, Valiante NM, Ulmer JB.
Targeting the innate immune response with
improved vaccine adjuvants. Nat. Med.
11(Suppl. 4), S63S68 (2005).
De Gregorio E, DOro U, Wack A.
Immunology of TLR-independent vaccine
adjuvants. Curr. Opin. Immunol. 21(3),
339345 (2009).
Emphasis on role of dendritic cells in
adjuvant action and induction of
immune responses.

www.expert-reviews.com

Coffman RL, Sher A, Seder RA. Vaccine


adjuvants: putting innate immunity to
work. Immunity 33(4), 492503 (2010).
Macagno A, Napolitani G, LanzavecchiaA,
Sallusto F. Duration, combination and
timing: the signal integration model of
dendritic cell activation. Trends Immunol.
28(5), 227233 (2007).

clinical testing World Health Organization


organisation Mondiale de la Sante
Fondation Merieux, Annecy, France, 2325
June 2003. Vaccine 22(1718), 20972102
(2004).
16

The European Agency for the Evaluation of


Medicinal Products. Guideline on
adjuvants in vaccines. EMA, London, UK
(2004).

17

Grgacic EV, Anderson DA. Virus-like


particles: passport to immune recognition.
Methods 40(1), 6065 (2006).

18

Jennings GT, Bachmann MF. The coming


of age of virus-like particle vaccines. Biol.
Chem. 389(5), 521536 (2008).

Summarizes the arguments for virus-like


particle vaccines.

19

Jansen KU, Conner ME, Estes MK.


Virus-like particles as vaccines and vaccine
delivery systems. In: New Generation
Vaccines. Levine MM, Dougan G,
GoodMF etal. (Eds). Informa Healthcare,
London, UK, 298305 (2010).

11

Harandi AM, Davies G, Olesen OF.


Vaccine adjuvants: scientific challenges and
strategic initiatives. Expert Rev. Vaccines
8(3), 293298 (2009).

12

Petrovsky N. Freeing vaccine adjuvants


from dangerous immunological dogma.
Expert Rev. Vaccines 7(1), 710 (2008).

20

Haynes JR. Influenza virus-like particle


vaccines. Expert Rev. Vaccines 8(4),
435445 (2009).

13

Sesardic D. Regulatory considerations on


new adjuvants and delivery systems.
Vaccine 24(Suppl. 2), S2S7 (2006).

21

Cox MM. Progress on baculovirus-derived


influenza vaccines. Curr. Opin. Mol. Ther.
10(1), 5661 (2008).

14

Mastelic B, Ahmed S, Egan WM etal.


Mode of action of adjuvants: implications
for vaccine safety and design. Biologicals
38(5), 594601 (2010).

22

How understanding an adjuvants mode of


action can impact on the assessment of
vaccine safety and help to develop
target-specific vaccines.

Gluck R, Mischler R, Brantschen S,


JustM, Althaus B, Cryz SJ Jr.
Immunopotentiating reconstituted
influenza virus virosome vaccine delivery
system for immunization against
hepatitisA. J. Clin. Invest. 90(6),
24912495 (1992).

23

Ludwig C, Wagner R. Virus-like particlesuniversal molecular toolboxes. Curr. Opin.


Biotechnol. 18(6), 537545 (2007).

15

Pink JR, Kieny MP. 4th meeting on novel


adjuvants currently in/close to human

443

Review

Moser, Amacker & Zurbriggen

24

Smith ML, Fitzmaurice WP, Turpen TH,


Palmer KE. Display of peptides on the
surface of tobacco mosaic virus particles.
Curr. Top. Microbiol. Immunol. 332, 1331
(2009).

25

Buonaguro L, Tornesello ML,


BuonaguroFM. Virus-like particles as
particulate vaccines. Curr. HIV Res. 8(4),
299309 (2010).

26

27

Neirynck S, Deroo T, Saelens X,


Vanlandschoot P, Jou WM, Fiers W.
Auniversal influenza A vaccine based on
the extracellular domain of the M2 protein.
Nat. Med. 5(10), 11571163 (1999).
Morein B, Helenius A, Simons K,
Pettersson R, Kaariainen L,
SchirrmacherV. Effective subunit vaccines
against an enveloped animal virus. Nature
276(5689), 715718 (1978).

Original concept of using virus-like


particles and virosomes as vaccines.

28

Almeida JD, Edwards DC, Brand CM,


Heath TD. Formation of virosomes from
influenza subunits and liposomes. Lancet
2(7941), 899901 (1975).

29

Stegmann T, Kamphuis T, Meijerhof T,


Goud E, de Haan A, Wilschut J.
Lipopeptide-adjuvanted respiratory
syncytial virus virosomes: a safe and
immunogenic non-replicating vaccine
formulation. Vaccine 28(34), 55435550
(2010).

30

Datta SA, Rein A. Preparation of


recombinant HIV-1 gag protein and
assembly of virus-like particles in vitro.
Methods Mol. Biol. 485, 197208 (2009).

31

Stegmann T, Morselt HW, Booy FP, van


Breemen JF, Scherphof G, Wilschut J.
Functional reconstitution of influenza virus
envelopes. EMBO J. 6(9), 26512659
(1987).

32

33

Mischler R , Metcalfe IC. Inflexal V a


trivalent virosome subunit influenza
vaccine: production. Vaccine 20(Suppl. 5),
B17B23 (2002).

Herzog C, Hartmann K, Kunzi V etal.


Eleven years of Inflexal V-a virosomal
adjuvanted influenza vaccine. Vaccine
27(33), 43814387 (2009).

Comprehensive review of the clinical data


with a virosomal influenza vaccine.

444

Wiedermann U, Wiltschke C, Jasinska J


etal. A virosomal formulated Her-2/neu
multi-peptide vaccine induces Her-2/
neu-specific immune responses in patients
with metastatic breast cancer: a Phase I
study. Breast Cancer Res. Treat. 119(3),
673683 (2010).

36

Genton B, Pluschke G, Degen L etal.


Arandomized placebo-controlled Phase Ia
malaria vaccine trial of two virosomeformulated synthetic peptides in healthy
adult volunteers. PLoS ONE 2(10), e1018
(2007).

37

Thompson FM, Porter DW, Okitsu SL etal.


Evidence of blood stage efficacy with a
virosomal malaria vaccine in a Phase IIa
clinical trial. PLoS ONE 3(1), e1493 (2008).

38

Lamb RA, Krug RM. Orthomyxoviridae:


the viruses and their replication. In: Fields
Virology. Knipe DM, Howley PM (Eds).
Lippincott Williams & Wilkins,
Philadelphia, PA, USA, 14871531 (2001).

39

Daemen T, de Mare A, Bungener L,


deJonge J, Huckriede A, Wilschut J.
Virosomes for antigen and DNA delivery.
Adv. Drug Deliv. Rev. 57(3), 451463
(2005).

40

Moser C, Amacker M, Kammer AR,


RasiS, Westerfeld N, Zurbriggen R.
Influenza virosomes as a combined vaccine
carrier and adjuvant system for prophylactic
and therapeutic immunizations. Expert Rev.
Vaccines 6(5), 711721 (2007).

41

Felnerova D, Viret JF, Gluck R, Moser C.


Liposomes and virosomes as delivery
systems for antigens, nucleic acids and
drugs. Curr. Opin. Biotechnol. 15(6),
518529 (2004).

42

Khoshnejad M, Young PR, Toth I,


Minchin RF. Modified influenza
virosomes: recent advances and potential in
gene delivery. Curr. Med. Chem. 14(29),
31523156 (2007).

43

Markgraf K, Muller K, Ponimaskin EG,


Rudolph M, Schmidt MF, Herrmann A.
Lipid composition of virosomes modulates
their fusion efficiency with cryopreserved
bull sperm cells. Cloning 3(1), 1121
(2001).

Bovier PA. Epaxal: a virosomal vaccine to


prevent hepatitis A infection. Expert Rev.
Vaccines 7(8), 11411150 (2008).

Comprehensive review of the clinical data


on virosomal hepatitis A vaccines and
comparison to alum-adjuvanted
competitorvaccines.
34

35

44

45

Cusi MG, Terrosi C, Savellini GG,


DiGenova G, Zurbriggen R, Correale P.
Efficient delivery of DNA to dendritic cells
mediated by influenza virosomes. Vaccine
22(56), 735739 (2004).
de Jonge J, Leenhouts JM, Holtrop M etal.
Cellular gene transfer mediated by
influenza virosomes with encapsulated
plasmid DNA. Biochem. J. 405(1), 4149
(2007).

46

Kammer AR, Amacker M, Rasi S etal.


Anew and versatile virosomal antigen
delivery system to induce cellular and
humoral immune responses. Vaccine
25(41), 70657074 (2007).

First report on lyophilizable virosomes.

47

Wilschut J, de Jonge J, Huckriede A,


Amorij JP, Hinrichs WL, Frijlink HW.
Preservation of influenza virosome
structure and function during freezedrying and storage. J.Liposome Res.
17(34), 173182 (2007).

48

Amacker M, Engler O, Kammer AR etal.


Peptide-loaded chimeric influenza
virosomes for efficient in vivo induction of
cytotoxic T cells. Int. Immunol. 17(6),
695704 (2005).

49

Zurbriggen R, Gluck R. Immunogenicity


of IRIV- versus alum-adjuvanted diphtheria
and tetanus toxoid vaccines in influenza
primed mice. Vaccine 17(1112),
13011305 (1999).

50

Westerfeld N, Zurbriggen R.
Peptidesdelivered by immunostimulating
reconstituted influenza virosomes. J.Pept.
Sci. 11(11), 707712 (2005).

51

Okitsu SL, Mueller MS, Amacker M etal.


Preclinical profiling of the immunogenicity
of a two-component subunit malaria
vaccine candidate based on virosome
technology. Hum. Vaccin. 4(2), 106114
(2008).

52

Nallet S, Amacker M, Westerfeld N etal.


Respiratory syncytial virus subunit vaccine
based on a recombinant fusion protein
expressed transiently in mammalian cells.
Vaccine 27(46), 64156419 (2009).

53

Liu X, Siegrist S, Amacker M,


ZurbriggenR, Pluschke G, Seeberger PH.
Enhancement of the immunogenicity of
synthetic carbohydrates by conjugation to
virosomes: a leishmaniasis vaccine
candidate. ACS Chem. Biol. 1(3), 161164
(2006).

54

Zurbriggen R, Amacker M, Kammer AR


etal. Virosome-based active immunization
targets soluble amyloid species rather than
plaques in a transgenic mouse model of
Alzheimers disease. J.Mol. Neurosci. 27(2),
157166 (2005).

55

Correale P, Del Vecchio MT, Renieri T


etal. Anti-angiogenetic effects of immunereconstituted influenza virosomes
assembled with parathyroid hormonerelated protein derived peptide vaccine.
Cancer Lett. 263(2), 291301 (2008).

56

Kopp N, Diaz D, Amacker M etal.


Identification of a synthetic peptide
inducing cross-reactive antibodies binding

Expert Rev. Vaccines 10(4), (2011)

Influenza virosomes as a vaccine adjuvant & carrier system

to Rhipicephalus (Boophilus) decoloratus,


Rhipicephalus (Boophilus) microplus,
Hyalomma anatolicum anatolicum and
Rhipicephalus appendiculatus BM86
homologues. Vaccine 28(1), 261269
(2009).
57

Waelti E, Wegmann N, Schwaninger R


etal. Targeting Her-2/neu with antirat Neu
virosomes for cancer therapy. Cancer Res.
62(2), 43744 (2002).

58

Amacker M, Moese S, Kammer AR,


Helenius A, Zurbriggen R. Influenza
virosomes as delivery systems for antigens.
In: Delivery Technologies for
Biopharmaceuticals: Peptides, Proteins,
Nucleic Acids, and Vaccines. Jorgensen L,
Nielsen HM (Eds). John Wiley & Sons
Ltd., Chichester, UK, 377393 (2009).

59

60

61

62


63

Huckriede A, Bungener L, Stegmann T


etal. The virosome concept for influenza
vaccines. Vaccine 23(Suppl. 1), S26S38
(2005).
Bungener L, Huckriede A, de Mare A,
deVries-Idema J, Wilschut J, Daemen T.
Virosome-mediated delivery of protein
antigens in vivo: efficient induction of class
I MHC-restricted cytotoxic Tlymphocyte
activity. Vaccine 23(10), 12321241
(2005).
Schumacher R, Amacker M, Neuhaus D
etal. Efficient induction of tumoricidal
cytotoxic T lymphocytes by HLA-A0201
restricted, melanoma associated, L(27)
Melan-A/MART-1(2635) peptide
encapsulated into virosomes in vitro.
Vaccine 23(4849), 55725582 (2005).

Invivo tracing of particulate vaccines.

67

Junt T, Moseman EA, Iannacone M etal.


Subcapsular sinus macrophages in lymph
nodes clear lymph-borne viruses and
present them to antiviral B cells. Nature
450(7166), 110114 (2007).

68

Carrasco YR, Batista FD. B cells acquire


particulate antigen in a macrophage-rich
area at the boundary between the follicle
and the subcapsular sinus of the lymph
node. Immunity 27(1), 160171 (2007).

69

Bachmann MF, Speiser DE, Ohashi PS.


Functional management of an antiviral
cytotoxic T-cell response. J.Virol. 71(8),
57645768 (1997).

70

Justewicz DM, Doherty PC, Webster RG.


The B-cell response in lymphoid tissue of
mice immunized with various antigenic
forms of the influenza virus hemagglutinin.
J. Virol. 69(9), 54145421 (1995).

71

Liu WC, Lin SC, Yu YL, Chu CL, Wu SC.


Dendritic cell activation by recombinant
hemagglutinin proteins of H1N1 and
H5N1 influenza A viruses. J. Virol. 84(22),
1201112017 (2010).

72

Song H, Wittman V, Byers A etal. In vitro


stimulation of human influenza-specific
CD8 + T cells by dendritic cells pulsed with
an influenza virus-like particle (VLP)
vaccine. Vaccine 28(34), 55245532 (2010).

73

Barton GM. Viral recognition by Toll-like


receptors. Semin. Immunol. 19(1), 3340
(2007).

74

Angel J, Chaperot L, Molens JP etal.


Virosome-mediated delivery of tumor
antigen to plasmacytoid dendritic cells.
Vaccine 25(19), 39133921 (2007).

Schumacher R, Adamina M, Zurbriggen R


etal. Influenza virosomes enhance class I
restricted CTL induction through CD4 +
Tcell activation. Vaccine 22, 714723
(2004).

Interaction of virosomes with a human


dendritic cell line.

Evidence that virosomes reactivate


influenza-specific memory cells.

75

McCoy K, Tatsis N, Korioth-Schmitz B


etal. Effect of preexisting immunity to
adenovirus human serotype 5 antigens on
the immune responses of nonhuman
primates to vaccine regimens based on
human- or chimpanzee-derived adenovirus
vectors. J.Virol. 81(12), 65946604
(2007).

Bungener L, Serre K, Bijl L etal.


Virosome-mediated delivery of protein
antigens to dendritic cells. Vaccine
20(1718), 22872295 (2002).

64

Noad R, Roy P. Virus-like particles as


immunogens. Trends Microbiol. 11(9),
438444 (2003).

65

Hofer U, Lehmann AD, Waelti E,


AmackerM, Gehr P, Rothen-Rutishauser
B. Virosomes can enter cells by nonphagocytic mechanisms. J.Liposome Res.
19(4), 301309 (2009).

66

Manolova V, Flace A, Bauer M, Schwarz K,


Saudan P, Bachmann MF. Nanoparticles
target distinct dendritic cell populations
according to their size. Eur. J.Immunol.
38(5), 14041413 (2008).

www.expert-reviews.com

76

77

Sharpe S, Polyanskaya N, Dennis M etal.


Induction of simian immunodeficiency
virus (SIV)-specific CTL in rhesus
macaques by vaccination with modified
vaccinia virus Ankara expressing SIV
transgenes: influence of pre-existing
anti-vector immunity. J.Gen. Virol.
82(Pt9), 22152223 (2001).
Jegerlehner A, Wiesel M, Dietmeier K etal.
Carrier induced epitopic suppression of
antibody responses induced by virus-like

Review

particles is a dynamic phenomenon caused


by carrier-specific antibodies. Vaccine
28(33), 55035512 (2010).
78

Brokstad KA, Cox RJ, Eriksson JC,


Olofsson J, Jonsson R, Davidsson A.
Highprevalence of influenza specific
antibody secreting cells in nasal mucosa.
Scand. J.Immunol. 54(12), 243247
(2001).

79

Peduzzi E, Westerfeld N, Zurbriggen R,


Pluschke G, Daubenberger CA.
Contribution of influenza immunity and
virosomal-formulated synthetic peptide to
cellular immune responses in a Phase I
subunit malaria vaccine trial. Clin.
Immunol. 127(2), 188197 (2008).

80

Van der Wielen M, Vertruyen A,


FroesnerG etal. Immunogenicity and
safety of a pediatric dose of a virosomeadjuvanted hepatitis A vaccine: a controlled
trial in children aged 116 years. Pediatr.
Infect. Dis. J. 26(8), 705710 (2007).

81

Bovier PA. Recent advances with a


virosomal hepatitis A vaccine. Expert. Opin.
Biol. Ther. 8(8), 11771185 (2008).

82

Dagan R, Amir J, Livni G etal.


Concomitant administration of a
virosome-adjuvanted hepatitis a vaccine
with routine childhood vaccines at age
twelve to fifteen months: a randomized
controlled trial. Pediatr. Infect. Dis. J.
26(9), 787793 (2007).

83

Holzer BR, Hatz C, Schmidt-Sissolak D,


Gluck R, Althaus B, Egger M.
Immunogenicity and adverse effects of
inactivated virosome versus alum-adsorbed
hepatitis A vaccine: a randomized controlled
trial. Vaccine 14(10), 982986 (1996).

84

Clarke PD, Adams P, Ibanez R, Herzog C.


Rate, intensity, and duration of local
reactions to a virosome-adjuvanted vs. an
aluminium-adsorbed hepatitis A vaccine in
UK travellers. Travel Med. Infect. Dis. 4(6),
313318 (2006).

85

Bovier PA, Farinelli T, Loutan L.


Interchangeability and tolerability of a
virosomal and an aluminum-adsorbed
hepatitis A vaccine. Vaccine 23(19),
24242429 (2005).

86

Wilschut J. Influenza vaccines: the


virosome concept. Immunol. Lett. 122(2),
118121 (2009).

87

Calcagnile S, Zuccotti GV. The virosomal


adjuvanted influenza vaccine. Expert. Opin.
Biol. Ther. 10(2), 191200 (2010).

88

Kursteiner O, Moser C, Lazar H, Durrer P.


Inflexal Vthe influenza vaccine with the
lowest ovalbumin content. Vaccine
24(4446), 66326635 (2006).

445

Review

Moser, Amacker & Zurbriggen

89

Gluck R. Intranasal immunization against


influenza. J.Aerosol Med. 15(2), 221228
(2002).

90

Mutsch M, Zhou W, Rhodes P etal. Use of


the inactivated intranasal influenza vaccine
and the risk of Bells palsy in Switzerland.
N. Engl. J.Med. 350(9), 896903 (2004).

91

Okitsu SL, Silvie O, Westerfeld N etal.


Avirosomal malaria peptide vaccine elicits
a long-lasting sporozoite-inhibitory
antibody response in a Phase 1a clinical
trial. PLoS ONE 2(12), e1278 (2007).

96

Genton B, Aebi T, Abdallah MS et al. A


phase 1b double-blind randomized
controlled age-deescalting trial of two
virosome formulated anti-malaria vaccine
components administered in combination
to healthy semi-immune Tanzanian adults
and children. Presented at: 59th Annual
Meeting of the American Society of Tropical
Medicine and Hygiene. Atlanta, GA, USA,
37 November 2010.

de Bruijn IA, Nauta J, Gerez L,


PalacheAM. The virosomal influenza
vaccine Invivac: immunogenicity and
tolerability compared to an adjuvanted
influenza vaccine (Fluad) in elderly
subjects. Vaccine 24(4446), 66296631
(2006).

92

de Bruijn I, Meyer I, Gerez L, Nauta J,


Giezeman K, Palache B. Antibody
induction by virosomal, MF59-adjuvanted,
or conventional influenza vaccines in the
elderly. Vaccine 26(1), 119127 (2007).

93

Westerfeld N, Pluschke G, Zurbriggen R.


Optimized malaria-antigens delivered by
immunostimulating reconstituted influenza
virosomes. Wien. Klin. Wochenschr.
118(1920 Suppl. 3), 5057 (2006).

94

95

Cavanagh DR, Remarque EJ, Sauerwein


RW, Hermsen CC, Luty AJ. Influenza
virosomes: a flu jab for malaria? Trends
Parasitol. 24(9), 382385 (2008).

446

97

Murphy CG, Fornier M. HER2-positive


breast cancer: beyond trastuzumab.
Oncology (Williston Park) 24(5), 410415
(2010).

98

Tudor D, Derrien M, Diomede L etal.


HIV-1 gp41-specific monoclonal mucosal
IgAs derived from highly exposed but
IgG-seronegative individuals block HIV-1
epithelial transcytosis and neutralize
CD4(+) cell infection: an IgA gene and
functional analysis. Mucosal Immunol. 2(5),
412426 (2009).

99

De Bernardis F, Liu H, OMahony R etal.


Human domain antibodies against
virulence traits of Candida albicans inhibit
fungus adherence to vaginal epithelium
and protect against experimental vaginal
candidiasis. J.Infect. Dis. 195(1), 149157
(2007).

100

Hunziker IP, Zurbriggen R, Glueck R etal.


Perspectives: towards a peptide-based
vaccine against hepatitis C virus. Mol.
Immunol. 38(6), 475484 (2001).

101

De Bernardis F, Boccanera M, Adriani D,


Girolamo A, Cassone A. Intravaginal and
intranasal immunizations are equally
effective in inducing vaginal antibodies and
conferring protection against vaginal
candidiasis. Infect. Immun. 70(5),
27252729 (2002).

Patent
201

Pevion Biotech AG. WO2009/000433 A1


(2008).

Websites
301

Mymetics Corp Homepage


www.mymetics.com

302

Pevion Biotech Homepage


www.pevion.com

Expert Rev. Vaccines 10(4), (2011)

Você também pode gostar