Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

Advanced Drug Delivery Reviews 57 (2005) 333 – 355

www.elsevier.com/locate/addr

Immunity in response to particulate antigen-delivery systems


Tazio Storni, Thomas M. Kqndig, Gabriela Senti, P3l Johansen*
Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland
Received 29 March 2004; accepted 1 September 2004
Available online 12 October 2004

Abstract

Adjuvants and antigen-delivery systems are essential in inducing and modifying immune responses, and despite the variety
of materials available for such use, mechanisms by which they support immunity appear to be little known. A common
denominator for most antigen-delivery systems is their particulate nature. Together with a certain depot effect, it is the
particulate nature that primarily decides whether the antigen-delivery system will be successful in inducing an immune
response. If this first requirement is fulfilled, the chemical composition of the vaccine decides which type of immune response
will develop, e.g. which isotype of antibodies the B cells will produce, and which cytokines the T cells will secrete, and can be
controlled by combining the antigen with immunomodulatory or co-stimulatory molecules. It is our goal to provide an overview
of the cellular and molecular factors involved in the induction of immunity and how such factors may influence the potency of
an adjuvant or a vaccine. Such factors should then be implemented in the design of new vaccines or in tuning the properties of
existing vaccines in order to reach the properties that are necessary for successful vaccination.
D 2004 Elsevier B.V. All rights reserved.

Keywords: Innate immunity; Adaptive immunity; Viruses; Bacteria; Particles; Vaccine design

Contents

1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 334
2. Immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 335
2.1. Innate immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 335
2.2. Adaptive immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 338
2.2.1. Antigen presentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 338
2.2.2. T cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 338
2.2.3. B cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 339
2.3. Immunological memory . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 340
2.4. The mucosal immune system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341
3. Infectious pathogens as models for vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342

* Corresponding author. Tel.: +41 1 255 1111; fax: +41 1 255 4418.
E-mail address: pal.johansen@usz.ch (P. Johansen).

0169-409X/$ - see front matter D 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2004.09.008
334 T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355

3.1. Viruses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342


3.2. Bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343
4. Adjuvants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 344
5. Designing pharmaceutical antigen-delivery systems. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 345
5.1. Targeting professional antigen-presenting cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 345
5.2. Concomitant delivery of antigen and co-stimulatory signals. . . . . . . . . . . . . . . . . . . . . . . . . 345
5.3. Immunomodulating activities of synthetic polymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 347
5.4. Antigen dose and structure affect the type of immunity induced . . . . . . . . . . . . . . . . . . . . . . 347
6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 348
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 348

1. Introduction tion or delivery system varies considerably between


vaccines. DNA and some protein vaccines can be
Although we encounter microorganisms on a administered in solution without adjuvant enhance-
daily basis, this causes disease only occasionally. ment of the immune response. However, most protein
Most organisms are detected and destroyed within and peptide vaccines show only low immunological
hours by defence mechanisms, which are not activity when administered in an aqueous solution and
antigen-specific and which do not require any need the help of adjuvants to induce the desired
prolonged period of induction. These are the immunological response.
mechanisms of innate immunity. Only when the In the first pioneering works of Jenner (small pox)
infectious agent is capable to break this early line of and Pasteur (cholera), less virulent forms of the whole
defence, an adaptive immune response will develop, pathogen were used as a vaccine. Today, still many of
including generation of antigen-specific effector cells the most potent vaccines are given as a live attenuated
that specifically target the pathogen, secretion of or killed form of the particulate microorganism. A
antibodies (B cells), through direct cytotoxic activity unique property, especially of live vaccines, is that
(T cells), or secretion of immunological mediators they often induce strong T-cell responses, a property
and effector molecules such as cytokines and very much missed by protein or peptide vaccines
chemokines. Although most of these effector cells administered with those adjuvants that are in general
will die within 10–14 days after infection, some cells use today, e.g. aluminium salts. A drawback of live
will survive as highly reactive plasma cells (B cells) vaccines or inactivated virus or bacteria is potential
or memory cells (B and T cells) and prevent adverse effects, as reported for whole pertussis
subsequent infection by the same microorganism. vaccine [5], the Sabin polio vaccine [6] and measles
Along with long-lasting antibodies against a specific vaccine [7].
pathogen, the induction of memory cells is the final During the last 25–30 years, research and develop-
goal of preventive vaccination. ment of alternative particulate adjuvants and vaccines
Immunity against an infectious agent by vaccina- has increased [8]. This has been stirred partly by the
tion can be achieved in various ways. Firstly, the fact that all pathogens are particulate and that particles
antigen itself can be a synthetically produced peptide can target vaccine delivery to antigen-presenting cells,
representing an epitope of a pathogen protein. It can and provide persistent antigen due to slow degradation
also be the full-length protein carrying several or clearance of the carrier particle. This has the
epitopes that are recognisable by B and T cells. Such potential of obviating the requirement for multiple
full-length proteins can be secreted from the pathogen vaccinations to boost the antibody response up to a
(DT, TT, PT) or produced synthetically or recombi- protective level. Moreover, many of the particulate
nantly. During the last decade, vaccine development formulations tested were shown to have the potential
has also focussed on experimental vaccines where the of inducing helper- and cytotoxic T-cell responses
gene encoding a particular protein is fused into a [9,10], which were previously only seen induced with
DNA or RNA plasmid [1–4]. Secondly, the formula- attenuated or killed microorganisms and hardly seen
T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355 335

at all with aluminium-adjuvanted antigens. The poorly taken up by antigen-presenting cells, whereas
development of particulate vaccines has also been peptides, which can bind directly onto MHC class-I
motivated by safety issues, i.e., to avoid the infectious molecules on the cell surface, have the limitation to be
risks with live attenuated vaccines and to avoid IgE- presented not only by professional antigen-presenting
mediated allergic side effects frequently caused by cells, but also by any other types of cells, which do
aluminium salts [11]. Such a T-helper type 2 (Th2) not express co-stimulatory receptors or cytokines and,
immunity is undesired in immunotherapy of cancer thus, may induce only sub-optimal T-cell activation or
and allergies and in vaccination against intracellular even tolerance. To avoid these drawbacks, dendritic
pathogens. Other major disadvantages of aluminium cells have been generated from the bone marrow or
are unpredictable extent of adsorption of certain from peripheral blood monocytes and pulsed in vitro
proteins and low stability of some adsorbates [12]. with peptide or protein [22] together with agents to
Nevertheless, aluminium salts are the benchmark to mature antigen-presenting cells [23]. Although the
which other adjuvants must compare [13]. process of dendritic-cell isolation, peptide loading and
The success of preventive vaccination is evident vaccine administration is complex and requires well-
from the history of immunology, the classical example trained personnel, this technique represents a good
being the eradication of small pox. The use of model for particulate antigen-delivery systems with
therapeutic vaccines to boost immunity against respect to efficient immunological recognition, anti-
already established infections, tumours or allergies is gen presentation and lymphocyte activation.
much less successful than preventive disease vacci- In the following, we will review the immunological
nation [3,14,15]. Although antibodies are able to events and requirements in the generation of adaptive
neutralise pathogens with high efficacy, they do not immunity, including how immunity is induced by the
eliminate and kill cells of infected organs in a bnature’s own particulate antigen-delivery systemsQ,
comparably efficient way. Moreover, the immune e.g. bacteria and viruses. Further, possible mecha-
system faces additional hurdles during chronic dis- nisms in which detecting and processing of particles
eases, e.g. viruses and tumours possess various escape by the immune system can be improved are discussed
mechanisms like high antigenic mutation or down- in light of developing vaccine carriers or delivery
regulation of MHC molecule expression in infected systems.
cells, which in some instances can cause tolerance or
immune suppression [16–18]. Therefore, immuno-
therapy of chronic diseases must target the cellular 2. Immunity
branch of the immune system, especially T-helper
cells, which are capable to modulate immune The early phase of the host response to an infection
responses including cytotoxic T cells (CTLs); CTLs depends on the innate part of the immune system
can kill altered self-cells by perforin, granzymes or including a variety of resistance mechanisms that
other messengers of death, and secrete cytokines such recognise and respond to the presence of a pathogen.
as IFN-g and TNF-a. Innate immunity is followed by adaptive immunity,
Many efforts have been made to target antigens to which includes proliferation of pathogen-specific B or
professional antigen-presenting cells and in particular T lymphocytes and the induction of B- and T-cell
to dendritic cells, which are known to be the most memory that provides a long-term protection from
prominent T-cell activators [19,20]. Although peptides disease. Although the innate immune system lacks
and recombinant proteins are easily manufactured, many features and the specificity of adaptive system,
their immunogenicity is relatively low [21], mainly it can distinguish non-self from self, and is crucial for
because of the lack of activation of antigen-presenting the stimulation of adaptive immunity.
cells and the short half-life in vivo. Therefore,
proteins and peptides are generally administered in 2.1. Innate immunity
combination with adjuvants. Classical adjuvants,
however, induce only poor maturation of antigen- The first line of defence against intruding micro-
presenting cells. Furthermore, soluble proteins are organisms includes mechanisms such as fever,
336 T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355

mucosal secretions, chemical mediators and phago- Table 1


cytic cells, which fight pathogens in an unspecific and Description of Toll-like receptor (TLR) ligands
non-instructive way. In particular, macrophages and TLR-L Ligand Description (Source)
number
dendritic cells are active in eliminating the intruders
by phagocytosis, a mechanism that is followed by 1 Pam3Cys Synthetic lipoprotein
2 LTA Lipoteichoic acid
antigen presentation to generate adaptive immunity.
(St. aureus)
The uptake of a pathogen or antigen normally takes PGN Peptidoglycan
either of three pathways. Firstly, antigen-presenting (St. aureus, B. subtilis,
cells are capable to engulf particles or microorganisms E. coli)
non-specifically [24]. Secondly, phagocytes are equip- HKML Heat Killed L.
monocytogenes (Gram+)
ped with several cell-surface receptors that recognise
LPS Lipopolysaccharide
pathogen surfaces for receptor-mediated endocytosis. (E. coli)
Among such receptors are the mannan-binding lectin, 3 dsRNA Oligonucleotide
which initiates complement activation, and the macro- PolyI:C Synthetic analog of
phage mannose receptor, where cell-bound lectin dsRNA
4 LPS Lipopolysaccharide
binds certain sugar molecules found on the surface (E. coli)
of many bacteria and some viruses, including the 5 Flagellin S. thyphimurium
human immunodeficiency virus [25]. Also Fcg or Fcq 6 PGN Peptidoglycan
receptors complement secondary phagocytosis by (St. aureus, B. subtilis,
opsonisation. Another set of phagocytic receptors, E. coli)
7 ssRNA Oligonuleotide with
called scavenger receptors, recognises certain anionic
repeating G and U
polymers [26]. Thirdly, phagocytic cells can take up motives
soluble substances in pinocytic vacuoles by so-called Loxoribine Guanosine analogue
macropinocytosis [25,27]. While phagocytosis is a Imiquimod and Small synthetic antiviral
common feature of macrophages and various mono- Resiquimod imidazoqionolines
8 Imiquimod and Small synthetic antiviral
cytes, macro-pinocytosis seems to be a unique
Resiquimod imidazoqionolines
property of dendritic cells. 9 dsDNA Oligodeoxynuleotide
Unlike the receptors of adaptive immunity, the with repeating C and G
receptors of the innate immune system are not motives (CpG)
clonally distributed; a given set of receptors will be 10 Unknown Unknown
11 Unknown Unknown
present on all cells of the same type. The surfaces of
microorganisms bear pathogen-associated molecular
patterns (PAMPs), and the binding of PAMPs by important mediators of innate immunity, e.g. IL-6, IL-
receptors of the innate immune system gives rise to 12, IL-18, and IFN-a and IFN-g. Concurrently, TLR
very rapid responses, which are put into effect without signalling induces co-stimulatory molecules essential
the delay that is necessary to allow for clonal for the induction of adaptive immune responses, i.e.,
expansion of cells needed in the adaptive immune B7.1 (CD80) and B7.2 (CD86), which are cell-surface
response [28]. Recognition and signalling via recep- proteins expressed by antigen-presenting cells (Fig. 1,
tors for PAMPs was originally discovered in the fruit- inset). It is the presence of these molecules on antigen-
fly Drosophila melanogaster [29], but is now known presenting cells along with presentation of microbial
to play a key role in the defence against infection in antigens that activates the CD4 T cells required to
plants, insects, and vertebrates [30–32]. The receptors initiate most adaptive immune responses. Lack of co-
mediating these functions have been named the Toll- stimulation normally leads to tolerance [34–36].
like receptors (TLRs) and their ligation activates the Substances that induce co-stimulatory activity have
cells to respond to pathogens [33]. So far, 11 been mixed with protein antigens for many years in
mammalian TLRs have been identified (Table 1), order to enhance immunogenicity and are known as
and their stimulation activates the transcription factors adjuvants, the best being those containing microbial
NFnB and MAP kinase leading to the production of components. TLR2 and TLR6 have been shown to
T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355 337

Fig. 1. Antigen uptake and presentation. Schematic overview of the major events and signalling during uptake of antigen by antigen-presenting
cells (APCs) and subsequent presentation of the digested epitopes on MHC molecules. Immunity is primarily activated by endogenous (e.g.
heat-shock proteins, DNA and RNA damage, IL-1b, CD40L, hyaluron metabolites) or exogenous (e.g. pathogen or adjuvant) danger signals
(Signal 0). These signals are typically mediated by Toll-like receptors, enable defence by the innate immunity and are succeeded by activation of
the adaptive immunity through antigen presentation (Signal 1) and concurrent co-stimulation (Signal 2) of CD4-positive T-helper (Th) cells.
Finally, the activation of Th cells result in their secretion of cytokines and chemokines, which can directly affect viability and replication of live
pathogens, or the Th cells can further activate CD8-positive cytotoxic T cells or antibody-producing B cells; the secondary activation of other T
and B cells is further supported by co-stimulatimulatory CD40 on Th cells. In the absence of concurrent co-stimulation of CD28 on T cells by B7
molecules (CD80 and CD86) on the APCs, stimulation of T cells through the T-cell receptor complex does not induce immunity but tolerance.

bind to various microbial patterns like peptidoglycans TLR8 can bind to synthetic low molecular weight
from Gram-positive bacteria, Gram-negative lipoly- compounds like imiquimod and resiquimod, which
saccharide, lipoproteins, and lipopeptides, porins and then possess the capacity to induce cytokines like IL-
zymosan from yeast cell walls [37,38]. TLR3 is 12 and IFN-g and have therefore potent anti-viral and
involved in the recognition of dsRNA, suggesting that anti-tumour properties [45,46]. TLR9 is responsible
this TLR is involved in the initiation of immune for the innate immune responses against bacterial and
responses mainly against viruses, which induce viral DNA, which are rich in non-methylated CpG-
dsRNA production during replication in infected cells containing consensus sequences [47]. TLR10 and
[39]. Lipopolysaccharide also binds to the serum TLR11 were recently discovered, but their exact
protein MD-2, which interacts with CD14. This function and ligands and still to be described,
enables the whole protein–lipopolysaccharide com- although its function seems to be linked with other
plex to bind to TLR4 [40,41]. TLR5 is expressed on TLRs [48].
the inner part of intestinal epithelia and has been Important differences have been observed for
shown to induce pro-inflammatory factors. The ligand TLR2/4 and TLR3/9 in terms of cellular localisation.
for TLR5 is the monomeric protein flagellin, a TLR2 and 4 require the interaction of MD-2 and
component in the flagella of motile bacteria [42]. CD14 and, therefore, are expressed on the cell surface
The natural ligand of TLR7 and TLR8 was recently [41]. TLR3 and 9 are principally expressed intra-
found to be ssRNA [43,44]. In addition, TLR7 and cellularly [49,50]. In addition, recent studies sug-
338 T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355

gested that the various TLRs induce different immune microbes and particulate antigens to prevent them from
responses implying different signalling pathways [51– reaching the blood circulation. B cells, which concen-
54]. The exact profile of cytokines produced varies trate in lymphoid follicles, are particularly efficient in
according to the receptors involved and this will in taking up soluble antigens such as bacterial toxins by
turn influence the functional character of the adaptive specific binding of antigen to surface immunoglobu-
immune response that develops in their presence. In lins. B cells are, however, very inefficient at initiating
this way, the ability of the innate immune system to adaptive immune responses.
discriminate between different types of pathogens is Following uptake in any antigen-presenting cell,
used to ensure an appropriate type of adaptive antigens are fragmented by proteases in endosomes,
immune response. which also contain high concentrations of MHC
molecules. Antigenic peptides are then loaded onto
2.2. Adaptive immunity MHC class-II molecules before exported to the cell
surface to bind T-cell receptors of specific CD4 T-
Adaptive immunity is mediated and maintained by helper cells (Fig. 1) [58]. In contrast, activation of
B and T lymphocytes. B lymphocytes develop, cytotoxic CD8 T cells is driven by recognition of
independent of antigen in the bone marrow, but peptide on MHC class-I molecules. Here, a consistent
emerge in the periphery when the immature B cells part of intracellularly synthesised proteins unfolds and
express immunoglobulin (Ig)M. Here, B cells further is degraded by cytosolic proteasomes [59]. The protein
differentiate into naRve B cells, which also express fragments are then transported into the endoplasmatic
IgD and circulate in secondary lymphoid tissues, i.e., reticulum by the transporter-associated protein (TAP)
lymph nodes, spleen and mucosa-associated lymphoid complex for subsequent loading onto MHC class-I
tissues (MALT), where they may encounter foreign molecules followed by cell-surface expression [60].
antigen. T lymphocytes develop from a common Alternatively, exogenous antigens from microorgan-
lymphoid progenitor in the bone marrow that also isms or infected apoptotic and necrotic cells can be
gives raise to B lymphocytes, but those cells destined taken up and processed by antigen-presenting cells for
to give rise to T cells leave the bone marrow and cross-presentation on MHC class-I molecules in a TAP-
migrate to the thymus for development into CD4- or dependent or non-dependent way [61–65]. In the TAP-
CD8-positive T cells. dependent endosome-to-cytosol pathway, the captured
antigen leaks from the endosomes into the cytosol and
2.2.1. Antigen presentation is processed by the proteasomes. In the TAP-independ-
Immature dendritic cells take up soluble or solid ent direct endosomal loading mechanism, the antigen is
antigens at peripheral tissues and are activated to degraded by endosomal proteases and exchanged with
migrate into local lymphoid tissue. Here they mature peptides bound on MHC class I, a process favoured by
into cells that express high levels of co-stimulatory the low pH values in endosomal vesicles [66]. While
molecules and adhesion molecules that mediate dendritic cells can operate both cross-presentation
interactions with the naRve T cells, which are contin- mechanisms, macrophages seem to be restricted to
uously re-circulating through these tissues [55]. the direct endosomal-loading pathway [67].
Although questioned by some recent observations
[56,57], it is generally accepted that by the time 2.2.2. T cells
dendritic cells arrive in the lymph nodes, they have lost In addition to stimulation of the T-cell receptor and
their ability to capture new antigen. They are, however, its co-receptor CD3 by the foreign peptide bound to a
now capable to present the antigens ingested at the site self MHC molecule (Signal 1; Fig. 1), antigen-specific
of infection. Macrophages, which are phagocytic cells, clonal expansion of naRve T cells requires a so-called
and B cells, which bind pathogen components, may be second or co-stimulatory signal (Signal 2; Fig. 1),
similarly induced through non-specific receptors to which must be delivered by the same antigen-
express co-stimulatory molecules and act as antigen- presenting cell on which the T cell recognises its
presenting cells. Macrophages are found in many areas antigen. The most potent activators of naRve T cells are
of the lymph node where they can actively ingest mature dendritic cells, with one single dendritic cell is
T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355 339

able to activate about 100–1000 T cells [68,69]. The right activation stimulus. Daughter cells divide and
best-characterised co-stimulatory molecules belong to differentiate into effector CTLs without further stimuli
the B7-familiy of glycoproteins CD80 and CD86. in a pre-programmed fashion. On the other side,
Ligation of CD28 on T cells by B7 molecules proliferation can sometimes be uncoupled from
enhances production of IL-2, the major T-cell growth effector function [77]. This indicates that CD8 T-cell
hormone, thereby driving clonal expansion of naRve T activation and in particular differentiation may also
cells. In contrast, inhibited binding of B7 to CD28 depend on cell extrinsic factors. CTL responses
inhibits T-cell responses [35]. Various other co- against certain viruses and tissue grafts seem to
stimulatory molecules such as 4-1BB, OX40 and require the presence of CD4 T cells during priming
CD27, and secreted cytokines such as IL-12, IL-1h, of naRve CD8 T cells. In these responses, both the
IFN-a/h/g, as well as the density of surface-expressed naRve CD8 T cell and the CD4 T cell must recognise
MHC molecules also influence the strength of signal related antigens on the surface of the same antigen-
transduction [70–74]. Once a naRve T cell is activated, presenting cells [78–80], hence, a potent CTL vaccine
it expresses a number of proteins that contribute to should contain both CD4 and CD8 epitopes. It is
sustaining or modifying the co-stimulatory signal that thought that the actions of the CD4 T cell are needed
drives expansion and differentiation. One such protein to compensate for inadequate co-stimulation of naRve
is CD40 ligand (CD40L or CD154), which binds to CD8 T cells by the antigen-presenting cell [81,82],
CD40 on antigen-presenting cells, thereby transmit- and to guarantee optimal fitness of the induced CTLs
ting activation signals to T or B cells. CD40 also [83]. This compensatory effect is currently thought to
activates the antigen-presenting cells to express B7 occur by the recruitment of effector CD4 T cell that
molecules, thus stimulating further T-cell activation. through CD40 up-regulate co-stimulatory activity of
NaRve CD4 T cells can differentiate upon activation the antigen-presenting cell [84].
into either Th1 or Th2 cells, which differ in the type of
cytokines they produce. The role of effector Th cells is 2.2.3. B cells
to control whole immune responses by secreting a B cells are responsible for the recognition of
multitude of cytokines and by expressing surface pathogen-derived protein in its native form. They
molecules, such as CD40L for activation of B cells, can directly interact with viruses and bacteria through
antigen-presenting cells, CTLs and several other cells immunoglobulin receptors with an almost infinite
of the immune system. Th1 cells are associated with number of unique specificities. Mature B cells express
IFN-g, IL-12 and TNF-a whereas Th2 cells typically surface IgM and IgD as well as various accessory
produce IL-4, IL-5, IL-10 and IL-13. CD4 T-cell molecules, e.g. CD19, CD21, CD23 and CD37. The
polarisation depends both on the duration of antigenic surface immunoglobulins that serve as the antigen
stimulation and the cytokine environment to which the receptor play two roles in B-cell activation. First, they
cells are exposed [74,75]. Hence, the two CD4 T-cell transmit signals directly to the cell’s interior upon
subsets can regulate each other. Once one subset binding antigen (Signal 1). Second, they deliver the
becomes dominant, it is hard to shift the response to antigen to intracellular sites where it is digested and
the other subset. IL-10 can inhibit the development of returned to the B-cell surface as peptides bound to
Th1 cells by acting on the antigen-presenting cell, MHC class II molecules. The peptide-MHC class-II
whereas IFN-g can prevent the activation of Th2 cells. complex can be recognised by antigen-specific CD4
NaRve CD8 T cells are predestined to become helper T cells, stimulating them to make proteins such
cytotoxic. Proliferating CD8 T cells acquire effector as CD40 ligand (Signal 2), which in turn cause B cells
function and migrate to the peripheral site of inflamed to proliferate and differentiate into antibody-secreting
tissue to eliminate virus-infected cells and release cells. The more an antigen is ordered and repetitive,
cytokines capable to slow down viral replication. the more the IgM on the B cell surface become cross-
Observations indicated that CD8 T cells undergo a so- linked, thereby increasing Signal 1 strength [85]. The
called programmed differentiation upon antigenic formation of clusters of cross-linked immunoglobulin
encounter by precursor CTLs [76]. This model receptors generates delivery of a strong Signal 1 that
implies that only naRve T cells need to receive the is potent enough by itself to induce the production of
340 T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355

IgM antibodies of activated B cells. Less ordered and When activated B cells receive signals from CD4
repetitive antigens require additional stimuli like Th cells, they form so-called germinal centers, which
lipopolysaccharide or other TLR ligands and the help purpose is to enhance the later part of the primary
of CD4 T cells. T-helper cells are also crucial for immune response [86,87]. Some germinal center cells
somatic hypermutation and immunoglobulin isotype (centroblasts) first differentiate into centrocytes and
switching [86]. then into plasma cells. The centrocytes are rescued by
The linked recognition of antigen by B cells and binding to follicular dendritic cells or Th cells, which
CD4 T-helper cells, presumably mediated by an possess antigen–antibody complexes or express
antigen-presenting dendritic cell, is followed by CD40L, respectively [87]. This process is thought to
somatic hypermutation and specific antibody produc- be necessary for the affinity maturation of the B cells
tion [87]. However, B and T-helper cells do not need [86]. Low antigen-affinity B cells undergo apoptosis.
to recognise the same epitopes on the antigen. For The centroblasts continue to divide rapidly, but
complex antigens or pathogens, the two cells might successively begin to secrete antibodies at a high
not even recognise the same protein. Nevertheless, it rate. These cells are destined to become non-dividing,
is crucial that the peptide recognised by the T cell is a terminally differentiated plasma cells and represent an
physical part of the antigen recognised by the B cell, intermediate stage of differentiation. Upon migration
which can thus produce the appropriate peptide after to the bone marrow, a subset of them survive for a
internalisation of the antigen bound to its B-cell long period of time providing a source of high-affinity
receptors. Apart from its importance in the regulation antibody. Other germinal center cells differentiate into
and manipulation of the humoral immune response, long-lived memory B cells. It is still a matter of debate
e.g. in ensuring self-tolerance, linked recognition is if memory B cells require antigen to survive,
important in the design of vaccines. Haemophilus proliferate and develop into plasma cells or not
influenzae type b is a bacterial pathogen that can [91,92]. Early studies showed that follicular dendritic
cause meningitis. Antibodies against its capsular cells are capable to display antibody-captured antigen
polysaccharide mediate protective immunity. While in its native form for years and therefore may become
adults generate thymus-independent antibody an important source of antigen, indicating a role for
responses to these polysaccharide antigens very Ag presence in the maintenance of memory B cell
efficiently, such responses are weak in the immature responses [93,94]. Also, mice lacking mature follic-
immune system of the infant. In vaccines, therefore, ular dendritic cells and B-cell follicles, adoptively
the polysaccharide of Hemophilus influenzae type b is transferred with memory B cells could not maintain
chemically linked to tetanus or diphtheria toxoid. B elevated titers of specific antibodies, indicating a role
cells that bind the polysaccharide component of the for follicular dendritic cells presenting antigen-anti-
vaccine can be activated by helper T cells specific for body complexes in the development of plasma cells
peptides of the linked toxoid. from memory B cells and in pathogen antibody-
Re-recognition of peptide-MHC class-II complexes mediated protection [95,96]. More recently, memory
on B cells triggers Th2 cells to synthesise molecules B cells were shown to survive for long periods
that can further activate B cells. One important T-cell without the presence of antigen [92].
effector molecule is CD40L, which binds CD40 on B
cells and is involved in all phases of the B-cell 2.3. Immunological memory
response [88,89]. CD40L and IL-4 are thought to be
synergistic in driving the clonal expansion that Immunological memory, perhaps the most impor-
precedes antibody production [90]. IL-4 is secreted tant consequence of adaptive immunity, is the capacity
in a polar fashion by Th2 cells and acts selectively on of the immune system to respond more rapidly and
antigen-specific B cells. The B cells start proliferating, effectively to pathogens that have been encountered
and after several rounds of proliferation, aided by the previously and reflects the pre-existence of a clonally
two additional T-cell-derived cytokines IL-5 and IL-6, expanded population of antigen-specific lymphocytes.
B cells further differentiate into antibody-secreting Memory responses also differ qualitatively from
plasma cells. primary responses. The characteristics of antibodies
T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355 341

produced in secondary and subsequent responses are node-homing phenotype (CD62L and CCR7 posi-
distinct from those produced in the primary response tive). They are long-lived in the absence of antigen
to the same antigen. Other differences are their and are able to face systemic pathogenic infections. In
responsiveness to a secondary contact with pathogens, contrast, effector memory T cells may be dependent
with respect to cytokine secretion profile, expression on the presence of antigen, home to peripheral tissues,
of other effector proteins, e.g. granzyme B, perforin, produce IFN-g and perforin to protect against
and Fas ligand (CD95L), proliferation rate and peripheral infection and are relatively short lived.
distribution. Specific memory is maintained by dis- Effector memory T cells probably develop from
tinct populations of long-lived memory cells, and effector T cells [103]. The decreasing-potential
there has been a lengthy discussion on whether the hypothesis states that early effector T cells have the
survival and function of memory cells are dependent highest potential to become effector memory cells,
on residual antigen or not [97–99]. whereas T cells that have encountered antigen later
A memory B cell is one that has undergone during the immune response have the tendency to
immunoglobulin isotype switching and somatic become anergic or die by apoptosis [104–106].
hypermutation [89]. This differentiation process starts Hence, the definition of memory T cells is not nearly
late in the primary immune response and takes place as clear-cut as with B cells. Nonetheless, in immune
in the germinal centers. CD4 T-helper cells trigger the individuals, secondary and subsequent responses are
differentiation of naRve B lymphocytes into memory mediated solely by memory lymphocytes and not by
cells through CD40 ligation. The process of differ- naRve lymphocytes [107,108]. Therefore, it would be
entiation induced by this signal is irreversible, but important to know the capacity of vaccines to induce
necessary and sufficient to induce B-cell memory memory responses and to analyse the frequency and
[100]. The generation of memory B cells is pro- phenotype of the generated effector and central
gressive, starting late in the expansion phase of the memory lymphocytes.
primary immune response, well before the antigen is
eliminated; in contrast to T cells, which are considered 2.4. The mucosal immune system
effectors until all antigen is gone, the moment when
antigen is eliminated is not taken into account when The immune system can be divided into a series of
defining B-cell memory. Memory B cells divide very functional anatomical compartments, of which the two
slowly if at all and do not secrete antibody. However, most important ones are the peripheral lymphoid
upon re-encountering the same pathogen (or antigen system, e.g. spleen and lymph nodes, and the mucosal
in a booster vaccine), the memory cells start dividing lymphoid system. Specific homing mechanisms for
at a high rate and differentiate into antibody-secreting lymphocytes to each of these compartments maintain
plasma cells. a separate population of lymphocytes in each com-
T-cell activation and division may issue from partment. The mucosa-associated lymphoid tissues
different types of stimulation and lead to different (MALT) lining the gut are known as gut-associated
functional outcomes. Memory T cells are increased in lymphoid tissue (GALT) and consist of the tonsils and
frequency and have distinct activation requirements the adenoids, the Peyer’s patches of the small
and cell-surface proteins that distinguish them from intestine, the appendix, and solitary lymphoid follicles
effector T cells. After immunisation, the number of T of the large intestine and rectum. The overlying layer
cells reactive to a given antigen increases markedly as of the Peyer’s patches contains specialised epithelial
effector T cells are produced, and then falls back to cells. These microfold or M cells take up molecules
persist at a level significantly (100- to 1000-fold) and particles from the gut lumen by endocytosis or
above the initial frequency. These cells carry cell- phagocytosis. Some intestinal pathogens infect enter-
surface proteins more characteristic of effector cells ocytes, the absorptive cells that line much of the
than of naRve T cells. Two subsets of memory T cells intestine. Enterocytes do not act as passive victims of
may be distinguished, the so-called central memory T infection, but signal infection by releasing cytokines
cells and the effector memory T lymphocytes and chemokines such as IL-8, MCP-1, MIP-1a and h,
[101,102]. Central memory T cells display a lymph and RANTES that are chemo-attractants for neutro-
342 T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355

phils, monocytes, eosinophils and T lymphocytes. In of the high damage the virus inflicts on the host. For
this way, the onset of infection triggers an influx of this reason, the infection usually has an acute phase
inflammatory cells and lymphocytes, leading to the and resolves in either immunity or in death of the
induction of an immune response to the antigens of host. In contrast, non-cytopathic viruses (e.g. LCMV
the infectious agent. For a more comprehensive and cytomegalovirus) are best cleared by CTLs
review on the mucosal immunity and vaccines, refer capable to migrate to the peripheral tissues to
the two papers in this issue by Hirst et al. and by eliminate virus-infected cells [109]. A drawback of
Alpar et al. this defence mechanism is that CTLs themselves can
cause severe damage to the host. This highlights
another important aspect of viral infection, which is
3. Infectious pathogens as models for vaccines the kinetics of virus replication. If the virus is rapidly
controlled by CTLs, the self-inflicted damage is
The mammalian body is susceptible to infection by marginal. In contrast, if the virus replicates fast and
many pathogens, which must first get in contact with can infect a large number of cells, the tissue damage
the host and then establish a focus of infection in may become more severe [110]. Interestingly, during
order to cause infectious disease. To establish an generalised and prolonged non-cytopathic viral infec-
infection, the pathogen must first colonise the skin or tions, e.g. LCMV or hepatitis C virus, a detrimental
the internal mucosal surfaces of the respiratory, outcome may be observed [16,17]. In fact, when the
gastrointestinal, or urogenital tracts and then over- host is confronted with systemically distributed and
come or bypass the innate immune defences mecha- high amounts of antigen, virtually all antigen-specific
nisms associated with the epithelia and underlying T lymphocytes become simultaneously activated and
tissues. If it succeeds in doing this, it will provoke an die off rapidly. If the viral infection persists, the
adaptive immune response that will take effect after antigen may spread into the thymus and induce
several days and will usually clear the infection. negative selection of antigen-specific maturing T
Pathogens differ greatly in their means of patho- cells, a phenomenon that leads to a hole in the T-
genesis, requiring an equally diverse set of defensive lymphocyte repertoire. In this case, CTLs can no
responses from the host immune system. In the longer harm virus-infected cells and the host can
following, we will discuss how particular pathogens survive as chronic virus carrier.
induce immunity and how this knowledge can be used Most anti-viral vaccines that are currently in use
in the design and development of vaccines and consist of killed or live attenuated viruses. Inactivated
vaccine delivery systems, which should adopt some or killed viral vaccines consist of viruses typically
of the characteristics of pathogens in order to improve fixed with aldehydes or h-propiolactone so that they
potency. are unable to replicate, e.g. rabies, polio and influenza
vaccines. The chemical or physical inactivation of
3.1. Viruses pathogens in some instances may have the drawback
that the used agents change the protein structure or
The immunological mechanisms in anti-viral even denature the viral epitopes [111]. As a con-
defence are diverse. Antibody titers, their avidity sequence, the immune system may not recognise the
and affinity, CTL phenotypes, their activity and original virus structure. Live-attenuated viral vaccines
frequency, as well as anti-viral cytokines typically are generally far more potent, also because they elicit
secreted by T-helper cells and CTLs may all a greater number of relevant effector mechanisms,
contribute to immunity at some point. Cytopathic including T cells. Killed viruses cannot produce
viruses, e.g. polio, measles, vaccinia and smallpox, proteins in the cytosol, so peptides from the viral
are best controlled by B cells and neutralising antigens are normally not presented by MHC class-I
antibodies, as well as by cytokines that induce molecules, and thus, CTLs are typically not generated
metabolic changes in neighbouring cells, rendering by these vaccines. Attenuated viral vaccines are now
them less prone to viral replication [109]. Cytopathic in use against polio, measles, mumps, rubella, and
virus infections have to be rapidly controlled because varicella.
T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355 343

Attenuation is typically achieved by growing the ing for Listeria monocytogenes antigens was engi-
virus in non-human cell cultures. In the course of such neered into an auxotrophic mutant of Salmonella
selection, the viruses become less capable to grow in typhimurium, i.e., the mutant was dependent on an
human cells. Such attenuated strains replicate poorly external supply of an essential nutrient that the wild-
in human hosts. Ideally, replication is just enough to type would be capable of biosynthesising [117,118].
induce immunity, but not disease. Although attenuated After oral administration, the live attenuated bacteria
virus strains contain multiple mutations in genes then carried the plasmid to the M cells that cover the
encoding several of their proteins, it might be possible Peyer’s Patches of the gut (the M cells are the natural
for a pathogenic virus strain to re-emerge by a further target cells of S. thyphimurium). Invasion by this route
series of mutations, e.g. the Sabin 3 polio vaccine, delivers salmonella bacteria straight to the lymphoid
especially in immuno-deficient recipients, in whom system of the host. From the M cells, the bacteria
they often behave as virulent opportunistic infections enter macrophages and dendritic cells, where they die
[112]. However, novel recombinant DNA technolo- because of their mutation, thereby liberating multiple
gies are now used to replace wild-type genes in the copies of the DNA vaccine right where it is needed,
virus genome. The advantage of this approach is that i.e., inside the antigen-presenting cell. By this means,
mutations can be engineered so that reversion to wild the antigen is more efficiently targeted to the immune
type is virtually impossible. system and it curtails the need of administering large
After the first reports on the efficacy of DNA doses of plasmid. Attenuated strains of Salmonella
vaccines in animal models [2], it soon became evident have also been used as carriers of heterologous genes
that the efficiency had to be improved if technology encoding tetanus toxoid and antigens from other
should have any chance surviving the transfer into a organisms such as Bacillus anthracis, Yersinia pestis,
clinical setting, as very large amounts of DNA and Schistosoma mansoni. Each of these has been
plasmid would be needed for human vaccinations. used as an oral vaccine to protect mice against
One strategy to resolve this problem was to grow the experimental challenge with the respective pathogens
DNA plasmid in a mutant microorganism, which [119–121].
would not grow in vivo upon administration. This S. thyphimurium as well as attenuated strains of L.
could replace the labour intensive process of growing monocytogenes and Bacille Calmette-Guerin (BCG)
the DNA vaccine as a plasmid in E. coli and represent effective vehicles for delivery of heterolo-
subsequently extracting and purifying the plasmid gous antigens due to their preferred intracellular
for direct administration. Hence, viral vectors were replication in professional antigen-presenting cells
engineered to carry heterologous peptides or proteins [24,122–124]. The sub-cellular localisation within
from other microorganisms. Adenoviruses, alfavi- antigen-presenting cells differs among these bacteria.
ruses, fowlpox, canarypox and vaccinia viruses have L. monocytogenes resides within the cytoplasmic
all demonstrated their potential to serve as an non- compartment of antigen-presenting cells, whereas S.
pathogenic carrier of heterologous antigens [113– thyphimurium and BCG persist in the phagosomal
116]. Genes encoding protective antigens from several vacuole. The preferred intracellular localisation for
different organisms could be placed in a single these vector strains determines the trafficking of
vaccine strain. This approach makes it possible to bacterial antigens through different MHC-processing
immunise individuals against several pathogens at pathways, i.e., S. thyphimurium and BCG to the MHC
once, but such a vaccine could not be used twice class-II pathway with the subsequent stimulation of
because the vector used for vaccination itself gen- CD4 T cells, especially Th1 cells [125], and L.
erates long-lasting immunity that would neutralise its monocytogenes to the MHC class-I pathway for
effectiveness during a second administration. stimulation of CD8 T cells. Although this is the way
the named bacteria theoretically would induce
3.2. Bacteria immunity, experimental data suggest that recombinant
variants of the bacteria that express foreign proteins
Similar approaches are being used for anti-bacterial can induce both CD4- and CD8-type immune
vaccine development. For example, a plasmid encod- responses [126,127].
344 T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355

Vaccine efficacy of such antigen-delivery systems non-viable category is the safest one, but because of
is corroborated by their ability to induce strong Th1- poor or no immunogenicity, it often requires adjuvants
like cytokine responses. By using bacterial and viral to elicit an adequate immune response. In the absence
vectors, the antigens are not only targeted to the of adjuvant, a lack of responsiveness may occur, and
appropriate pathways of MHC, but also stimulate the naRve antigen-specific T cells may recognise the
innate immune system to provide the adequate antigen but are not sufficiently activated or they even
cytokine milieu and appropriate expression of co- become tolerised. Adjuvants are defined as a group of
stimulatory molecules [128]. Non-proteinaceous structurally heterogeneous compounds, used to evoke
immunogenic components of these antigen carrier or increase an immune response to an antigen [131].
systems, e.g. phospholigands for recognition by gy T Classically recognised examples include oil emul-
cells or glycolipids for stimulating T cells via CD1 sions, saponins, aluminium or calcium salts, non-ionic
binding, not only provide potential target antigens, but block polymer surfactants, derivatives of lipopolysac-
also represent moieties with additional immuno- charide (LPS), mycobacteria and many others
stimulating properties. Such components could theo- [8,132,133]. Theoretically, each molecule or sub-
retically be used as adjuvants in other pharmaceutical stance that is capable to favour or amplify a particular
antigen-delivery systems. step in the cascade of immunological events, ulti-
mately leading to a better immunological response,
can be defined as an adjuvant. Obviously, the first step
4. Adjuvants is very important. However, the in vivo molecular and
cellular mechanisms required for the generation of an
The design of vaccines primarily requires the effective immune response, which depends critically
identification of immunological correlates of protec- on co-injection of adjuvant, are poorly understood.
tion, i.e., the immune effector mechanism(s) respon- The structural requirements of adjuvants are similarly
sible for protection against disease, and the poorly understood. Adjuvants have therefore been
subsequent selection of an antigen that is capable to surrounded by obscurity and called bthe immunolo-
elicit the desired adaptive response. Once this gist’s dirty little secretQ [28].
appropriate antigen has been identified, it is essential Schijns classified adjuvants functionally according
to deliver it effectively to the host’s immune system to five recently proposed concepts of immunogenicity
[129,130]. [134] (Table 2): firstly, the geographical concept of
Traditionally, vaccines come in several forms: live- immune reactivity and, secondly, the theory of depot
attenuated, replicating pathogens and non-replicating, effect, both emphasizing the importance of antigen
inactivated pathogens or their subunits. The latter, localisation for a period of time after immunisation

Table 2
Classification of adjuvants based on their functional properties
Mode of action Examples Key event(s)
Facilitation of antigen uptake, transport Liposome, VLPs, aluminium salts, Antigen localisation in the lymph node
and presentation by APCs polymeric nano- and microparticles,
ISCOMs, QuilA
Depot effect Oil emulsions, gels, polymeric nano- and Prolonged antigen presentation
microparticles,
Signal 0 Cytokines, TLR ligands (e.g. LPS, CpG) Signalling through PAMP receptors on
innate immune cells
Signal 2 Cytokines, co-stimulatory molecules APC polarisation, T- and B-cell help
(e.g. B7 analogues, anti-CD28
antibodies)
Danger signal (the Matzinger theory) Oil-emulsions, surfactants, aluminium Tissue destruction/stress, DNA and RNA
salts, cytokines, interferons, CD40L damage, hyaluron metabolites, IL-1b,
analogues, heat shock proteins heat shock proteins, CD40L
T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355 345

[135]; thirdly, the paradigm that adjuvants act as gocytic cells. The chemical and physical composition
Signal 0, which precedes the induction of the epitope of the vaccine will partly define the quality and type
Signal 1 and co-stimulatory Signal 2 and, which is of cells attracted to the site of the vaccine entry.
characterised by pathogen recognition through
PAMPs [28]; fourthly, the role of Signal 2 molecules 5.1. Targeting professional antigen-presenting cells
as natural adjuvants in the activation of naive T-helper
cells, the master coordinators of subsequent T-cell- Among the cells picking up and transporting
dependent immune responses; finally, the hypothesis antigens to the lymph nodes, the dendritic cells that
that immunity is activated by exogenous and even- are resident in all tissues are by far the most potent
tually endogenous danger signals [136,137]. Accor- activators of T cells. They migrate to secondary
ding to the danger model, immune activation occurs lymphoid tissues and present the antigens they
upon recognition of host-derived danger signals from originally ingested at the peripheral site of infec-
stressed cells in damaged tissues—not from an tion/injection. Hence, in ideal priming, vaccines
invading pathogen. These signals have yet to be should preferentially be directed to the dendritic
defined molecularly, they may include cell debris cells. This can either be achieved by administration
from necrotic cells, heat shock proteins, nucleotides, of the vaccine to a tissue which contains a relative
reactive oxygen intermediates, cytokines, etc. high frequency of such cells, e.g. Langerhans cells
in the epidermis of the skin, or by guiding the
vaccine to dendritic cells using targeting molecules
5. Designing pharmaceutical antigen-delivery such as the myeloid marker CD11c, the adhesion
systems molecule DC-SIGN [145], the putative antigen-
uptake receptor DEC-205 [146,147], or the T-cell
Pharmaceutical vaccine formulations presently analogues of ICAM-1, ICAM-2 and ICAM-3. By
being tested in various experimental and clinical such means, it was also shown that CTLs can bee
models have one basic common denominator: they primed after a single injection, bypassing require-
are of particulate nature. In contrast to most soluble ment for adjuvant, CD4 T cell help and CD40
antigens, which are ignored by the immune system, signalling [148].
particles are recognised as foreign and as danger.
Hence, the starting-point when designing a new 5.2. Concomitant delivery of antigen and co-stimula-
vaccine delivery system is often that a particulate tory signals
structure is required. This is easily obtained by
aggregation or cross-linking of antigen [1,138,139] The binding of PAMPs by TLRs on antigen-
or, conventionally, by adsorption or precipitation of presenting cells initiates the adaptive immune response
and antigen on aluminium salts. Alternatively, the with maturation of antigen-presenting cells, up-regu-
antigen can be chemically attached to a pre-formed lation of co-stimulatory molecules (e.g. of the B7
particular carrier [140], or it is chemically or physi- family) and migration to the draining lymph nodes.
cally distributed in or on particles in a more (in The ligation of TLRs through PAMPs or their
liposomes and virus-like particles) or less (in immunostimulatory analogues in vaccines (Table 1)
polymeric microspheres) organised way [140–144]. further induces the production of several important
Any type of particle typically facilitates the recog- mediators of innate immunity, such as cytokines and
nition of the vaccine by professional antigen-present- chemokines, including co-stimulatory molecules
ing cells as well as the total or partial uptake of the essential for the induction of adaptive immune
vaccine into these cells. As a result of activation, the responses. For instance, by combining antigen-loaded
antigen-presenting cells normally start their migration virus-like particles with CpG, a vaccine possessing the
from their peripheral location towards the nearest most important immunological features of viruses was
draining lymphatic organ. However, prior to the engineered, with the immune system perceiving the
uptake into antigen-presenting cells, the formulation virus-like particles as dangerous pathogens against
itself influences the property of the recruited pha- which a strong immune response should be mounted
346 T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355

[77]. By this way, professional antigen-presenting cells and a co-stimulatory molecule is not effective if the
were shown to take up particles for antigen processing molecule dissociates prior to encountering antigen-
and presentation to specific T cells for several days. presenting cells. Furthermore, to encounter a CD4 T
Efficient maturation of the antigen-presenting cells cell, the antigen-presenting cell must migrate to a
was guaranteed by the packaged CpGs, which trigger nearby lymph node, and this migration is stimulated by
endosomal TLR-9. This vaccine formulation induced cytokines such as TNF-a [161,162].
high frequencies of specific CD8 T cells protective Once a naRve T cell is activated, it expresses
against cytopathic and non-cytopathic viruses as well proteins that contribute to clonal expansion and
as an established fibrosarcoma tumour [149]. The data differentiation. One such protein is CD40 ligand,
imply that effective stimulation of the innate immune which binds to CD40 on antigen-presenting cells. This
system is a key factor for the induction of strong and binding transmits activating signals to the T cell and
protective T-cell immune responses. Having received also activates the antigen-presenting cells to express
both the antigen and the co-stimulatory signal, T cells B7 molecules, thus stimulating further T-cell prolifer-
can initiate adaptive immune responses. Whereas most ation, and CD40 ligation is a prerequisite for the
PAMPs bind to receptors on the surface of antigen- generation of B cell memory [100]. There are several
presenting cells, some viruses are thought to be reports on experimental and clinical vaccines where
recognised trough the TLR3 and TLR9 inside the CD40 antibodies or CD40L are used to promote the
cells [49,50,150] as a consequence of the production of clonal expansion and differentiation of lymphocytes
double-stranded RNA or DNA, respectively, in the [116,163,164]. When a T cell has differentiated into
course of their replication. Hence, when using TLR3 an effector cell, encounter with its specific antigen
and TLR9 ligands in particulate vaccines, one must results in immune attack without the need for co-
assure that the same antigen-presenting cell takes up stimulation. This applies to all classes of effector T
both antigen and TLR ligand. Viral infections also cells, and its importance is easy to understand in the
induce the infected cells to produce IFN-a, which case of cytotoxic CD8 T cells, which must be able to
subsequently can activate dendritic cells to express co- act on any cell infected with a virus, whether or not
stimulatory molecules. Antigen-delivery systems that the infected cell can express co-stimulatory mole-
are capable of doing the same will also have the cules. Hence, for a boosting vaccine, it is not
general potential to stimulate adaptive immunity, necessarily required to provide co-stimulation. This
especially CTLs. is also illustrated by the ambiguity in adjuvant
Because activation of naRve T cells requires the requirements for the boosting of immune responses
simultaneous delivery of the antigen-specific and the against tetanus and diphtheria [11]; in contrast to the
co-stimulatory signals, only activated professional primary administration of soluble antigen, which can
antigen-presenting cells can usually initiate T-cell cause tolerance, once an immune response is primed,
responses. If the vaccine by itself is not capable to subsequent administration of soluble antigen does not
stimulate expression of molecules necessary for T-cell cause tolerisation, but boosts the already established
activation, a combined delivery of antigen and co- immune response. The boosting doses of antigen are
stimulation within the same pharmaceutical formula- rarely presented to new naRve T cells. In the
tion could improve the priming of lymphocytes. Here, competition for antigen, memory cells and specific
DNA vaccines offer an opportunity as they permit the antibodies are superior to naRve cells and will capture
concomitant delivery of pathogen-derived genes and neutralise boosting antigens before they get to
together with genes encoding for stimulatory cyto- prime naRve lymphocytes [107,108]. Such a principle
kines, e.g. GM-CSF, IL-2, IFN-g, and IL-12 [3,151– could be applied in a prime-boost vaccine were the
154]. The feasibility of this concept has been demon- priming vaccine contains the antigen plus all neces-
strated with recombinant viral vectors and liposomes, sary co-stimulatory additives, whereas the boost
which provided co-stimulatory molecules such as vaccine could consist of a pharmaceutical formula-
those of the B7 family [155–158], 4-1BB ligand tion, which provided long-term delivery of the antigen
[159], and NF-nB (RANK/RANK) ligand [116,160]. only. Similarly, it is conceivable that a vaccine-
However, a particulate vaccine combining an antigen delivery system may be combined with factors that
T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355 347

drive the differentiation of CD4 T cells into Th1 (e.g. polymers or excipients (e.g. chitosan, ethylene imine,
in HIV, malaria or tuberculosis vaccines) or Th2 (e.g. collagen, poly-phosphates, some substituted lignins
in polio and tetanus vaccines). Th1-polarising cyto- and dextrans, as well as anionic lipids and surfactants)
kines have been used in particulate antigen-delivery with proton scavenger properties may prevent antigen
systems to stimulate cell-mediated immunity presentation by the MHC class-II pathway. Indeed,
[165,166]. Especially, IL-12 may find application in chitosan has been shown to promote Th1 cytokine
therapeutic vaccination [3,167]. reponses and MHC class-I antigen presentation [172].

5.3. Immunomodulating activities of synthetic 5.4. Antigen dose and structure affect the type of
polymers immunity induced

The introduction of a synthetic material into the The amount and sequence of the antigen that
body affects different body systems, including the initiates the response also influence the differentiation
defence system. Synthetic polymers have only a of CD4 T cells into distinct effector subsets, with high
limited capacity to elicit antibody formation or to and low density of peptide on the surface of antigen-
induce a cellular immune response against them. presenting cells stimulating Th1 or Th2 cell responses,
However, synthetic polymers sometimes exhibit sig- respectively [173,174]. Hence, when stability of the
nificant immuno-modulating activity [168–170], antigen is compromised, as may occur in poly(lactide-
mainly concerning the activation/suppression of nat- co-glycolide) microspheres [175,176], this might also
ural killer cells (NK cells), so called lymphokine- have consequences for the Th1/Th2 skewing of the
activated killer cells (LAK cells) and macrophages immune response, since the relative accessibility of
[171]. Some polymers can be used as effective protein different epitopes might change. Moreover, peptides
carriers, and the development of biodegradable and that interact strongly with the T-cell receptor tend to
biocompatible vaccine delivery systems based on stimulate Th1-like responses, whereas peptides that
liposomes, microspheres, nanoparticles or water-solu- bind weakly tend to stimulate Th2-like responses
ble synthetic polymers has received considerable [174,177]. This difference could be very important in
attention, as they can be tailored to meet the specific several circumstances. Allergy is caused by the
physical, chemical, and immunogenic requirements of production of IgE antibody, which requires high
a particular antigen [171]. As mentioned above, the levels of IL-4, but does not occur in the presence of
loading of peptides on MHC class-II molecules occurs IFN-g [178]. Antigens that elicit IgE-mediated allergy
subsequent to the acidification of the antigen-contain- are generally delivered in minute doses, and they elicit
ing endosomes upon fusion with lysosomes. By Th2 cells that make IL-4 and no IFN-g. It is also
preventing acidification of these vacuoles, antigen relevant that allergens do not elicit any of the known
presentation can be blocked. Drugs, such as chlor- innate immune responses, which produce cytokines
oquine, raise the pH of vacuoles and inhibit the that tend to bias CD4 T-cell differentiation toward Th1
presentation of antigens that enter the cells by cells, probably because allergen should not represent a
endocytosis. This is also why chloroquine is a bdangerQ. Allergens are also delivered to humans in
therapeutic principle in malaria. However, antigen- minute doses across a thin mucosa, such as that of the
delivery systems, which assist the acidification of lung. Something about this route of sensitisation
endosomes, also promote the MHC class-II presenta- allows even potent generators of Th1 responses like
tion of antigen they are carrying. This might be one Leishmania major to induce Th2 responses.
mechanism by which nano- and microparticles of Ordered and repetitive structures permit to elicit
poly(lactide-co-glycolide) work. The acidic moieties significantly more potent B-cell responses [85], and
of lactic or glycolic acid monomers or soluble this principle has been exploited in virus-like par-
oligomers, as well as the proton liberated into the ticles, which are composed by spontaneous self-
endosomes during erosion of the polymers will trigger assembling viral proteins in a quasi-crystalline struc-
MHC class-II presentation [9]. On the other hand, ture [140,179]. B-cell receptor cross-linking by the
pharmaceutical formulations, which contain basic repetitively displayed epitopes on virus-like particles
348 T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355

activates B cells, which in turn will be able to induce participants of the innate immune system, their action
activation of Th cells. Secondary, this permits the is not yet clear and probably rely on different
development of IgG secreting plasma cells and B- mechanisms. The adjuvants must therefore be chosen
memory cells. Hepatitis B s antigen (HBsAg) virus- according to several criteria, like the target species,
like particles are a successful example of an effective the antigens, the type of desired immune response, the
vaccination approach against hepatitis B [180,181]. route of administration, or the duration of immunity.
Moreover, responses against chosen antigenic targets Many papers reports on the mechanism by which a
can be obtained by genetically fusing the B-cell pharmaceutical antigen-delivery system actually
epitopes to exposed regions of various types of virus- works. The usual take-home message is that it is a
like particles, or alternatively, by chemically cross- particle and that this is the target function for antigen
linking antigenic peptides or even whole proteins to presentation. bNanoparticles because size mattersQ,
the virus-like particles [140,182]. This latter method is bThe smaller the betterQ or bAPC targetingQ are
currently preferred because it allows the display of frequently used as catchphrases, but do not really
proteins in their native form, which is important for represent thorough studies of the uptake and antigen-
the generation of antibodies able to bind to pathogenic presentation mechanism for the actual pharmaceutical
antigens in a functional conformation. Similarly, formulation. Although the most important feature of
influenza hemaglutinin and neuraminidase proteins adjuvants or antigen-delivery systems is likely to be
put into a highly repetitive order on the surface of the particulate nature, there is a lot more to be done in
phospholipid liposomes (so-called virosomes) have basic research on how such systems can be engineered
proven powerful in inducing immunity [143,183]. to trigger a certain strength and type of immune
Hemaglutinin, which possess a sialic acid binding response. With today’s knowledge in basic immunol-
region, binds to surface receptors of host antigen- ogy, the opportunities for a rational design and testing
presenting cells and promotes up-take into the cell. By of antigen-delivery systems should be available. The
cross-linking or integrating proteins or B-cell epitopes success is perhaps only limited by the will and
to the virosomal structures, one can induce B-cell opportunity to break with conventional topical board-
responses against selected antigens [184]. This pro- ers in natural and medical science. Furthermore,
cedure proved particularly effective for the develop- although historically the best medical developments
ment of vaccines against influenza [185] and hepatitis were achieved by empirical approaches, the highly
A [186]. For an extended review on the properties of specific medical and regulatory requirements of
liposomes and virosomes, we refer to the paper of modern vaccines are probably better met by rational
Wilschut and co-workers in this issue. design than by trial and error.

6. Conclusion
References
Vaccination techniques do not always stimulate [1] I. Hoerr, R. Obst, H.G. Rammensee, G. Jung, In vivo
immunity because of the inappropriate elicitation of application of RNA leads to induction of specific cytotoxic
immune responses. Such limitations have spurred the T lymphocytes and antibodies, Eur. J. Immunol. 30 (2000)
development of new adjuvants and antigen-delivery 1 – 7.
[2] J.J. Donnelly, J.B. Ulmer, J.W. Shiver, M.A. Liu, DNA
systems. Adjuvants play an important role in enhanc-
vaccines, Annu. Rev. Immunol. 15 (1997) 617 – 648.
ing the efficacy of vaccines that consist of antigens [3] D.B. Lowrie, R.E. Tascon, V.L. Bonato, V.M. Lima, L.H.
becoming more and more purified. Indeed, recombi- Faccioli, E. Stavropoulos, M.J. Colston, R.G. Hewinson, K.
nant proteins or synthetic peptides are safer than crude Moelling, C.L. Silva, Therapy of tuberculosis in mice by
inactivated microorganism, but less immunogenic. DNA vaccination, Nature 400 (1999) 269 – 271.
This can be balanced by specific adjuvants. But there [4] J.Y. Scheerlinck, Genetic adjuvants for DNA vaccines,
Vaccine 19 (2001) 2647 – 2656.
are no universally active adjuvants and, apart from [5] S. Donnelly, C.E. Loscher, M.A. Lynch, K.H. Mills, Whole-
often being particulate and thereby attract the attention cell but not acellular pertussis vaccines induce convulsive
of professional antigen-presenting cells and other activity in mice: evidence of a role for toxin-induced
T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355 349

interleukin-1beta in a new murine model for analysis of [22] J.I. Mayordomo, T. Zorina, W.J. Storkus, L. Zitvogel, C.
neuronal side effects of vaccination, Infect. Immun. 69 Celluzzi, L.D. Falo, C.J. Melief, S.T. Ildstad, W.M. Kast,
(2001) 4217 – 4223. A.B. Deleo, et al., Bone marrow-derived dendritic cells
[6] E.A. Cherkasova, E.A. Korotkova, M.L. Yakovenko, O.E. pulsed with synthetic tumour peptides elicit protective and
Ivanova, T.P. Eremeeva, K.M. Chumakov, V.I. Agol, Long- therapeutic antitumour immunity, Nat. Med. 1 (1995)
term circulation of vaccine-derived poliovirus that causes 1297 – 1302.
paralytic disease, J. Virol. 76 (2002) 6791 – 6799. [23] A. Reddy, M. Sapp, M. Feldman, M. Subklewe, N.
[7] S. Piyasirisilp, T. Hemachudha, Neurological adverse events Bhardwaj, A monocyte conditioned medium is more
associated with vaccination, Curr. Opin. Neurol. 15 (2002) effective than defined cytokines in mediating the terminal
333 – 338. maturation of human dendritic cells, Blood 90 (1997)
[8] D.T. O’Hagan, N.M. Valiante, Recent advances in the 3640 – 3646.
discovery and delivery of vaccine adjuvants, Nat. Rev., Drug [24] K. Inaba, M. Inaba, M. Naito, R.M. Steinman, Dendritic cell
Discov. 2 (2003) 727 – 735. progenitors phagocytose particulates, including bacillus
[9] Y. Men, H. Tamber, R. Audran, B. Gander, G. Corradin, Calmette-Guerin organisms, and sensitize mice to mycobac-
Induction of a cytotoxic T lymphocyte response by immu- terial antigens in vivo, J. Exp. Med. 178 (1993) 479 – 488.
nization with a malaria specific CTL peptide entrapped in [25] F. Sallusto, M. Cella, C. Danieli, A. Lanzavecchia, Dendritic
biodegradable polymer microspheres, Vaccine 15 (1997) cells use macropinocytosis and the mannose receptor to
1405 – 1412. concentrate macromolecules in the major histocompatibility
[10] K. Peter, Y. Men, G. Pantaleo, B. Gander, G. Corradin, complex class II compartment: downregulation by cytokines
Induction of a cytotoxic T-cell response to HIV-1 proteins and bacterial products, J. Exp. Med. 182 (1995) 389 – 400.
with short synthetic peptides and human compatible adju- [26] S. Acton, D. Resnick, M. Freeman, Y. Ekkel, J. Ashkenas, M.
vants, Vaccine 19 (2001) 4121 – 4129. Krieger, The collagenous domains of macrophage scavenger
[11] R.K. Gupta, Aluminum compounds as vaccine adjuvants, receptors and complement component C1q mediate their
Adv. Drug Deliv. Rev. 32 (1998) 155 – 172. similar, but not identical, binding specificities for polyanionic
[12] W. Matheis, A. Zott, M. Schwanig, The role of the adsorption ligands, J. Biol. Chem. 268 (1993) 3530 – 3537.
process for production and control combined adsorbed [27] F. Sallusto, A. Lanzavecchia, Efficient presentation of soluble
vaccines, Vaccine 20 (2001) 67 – 73. antigen by cultured human dendritic cells is maintained by
[13] B.P. Mahon, A. Moore, P.A. Johnson, K.H. Mills, Ap- granulocyte/macrophage colony-stimulating factor plus inter-
proaches to new vaccines, Crit. Rev. Biotechnol. 18 (1998) leukin 4 and downregulated by tumor necrosis factor alpha, J.
257 – 282. Exp. Med. 179 (1994) 1109 – 1118.
[14] R.M. Zinkernagel, Immunology taught by viruses, Science [28] C.H. Janeway, Approaching the asymptote? Evolution and
271 (1996) 173 – 178. revolution in immunology, Cold Spring Harbor Symp. Quant.
[15] D.M. Pardoll, Spinning molecular immunology into success- Biol. 54 (1989) 1 – 13.
ful immunotherapy, Nat. Rev., Immunol. 2 (2002) 227 – 238. [29] C. Hashimoto, K.L. Hudson, K.V. Anderson, The Toll gene
[16] F. Lechner, D.K. Wong, P.R. Dunbar, R. Chapman, R.T. of Drosophila, required for dorsal–ventral embryonic polar-
Chung, P. Dohrenwend, G. Robbins, R. Phillips, P. Klener- ity, appears to encode a transmembrane protein, Cell 52
man, B.D. Walker, Analysis of successful immune responses (1988) 269 – 279.
in persons infected with hepatitis C virus, J. Exp. Med. 191 [30] R. Medzhitov, C.A. Janeway, Self-defense: the fruit fly style,
(2000) 1499 – 1512. Proc. Natl. Acad. Sci. U. S. A. 95 (1998) 429 – 430.
[17] A.J. Zajac, J.N. Blattman, K. Murali-Krishna, D.J. Sourdive, [31] K. Takeda, S. Akira, Toll receptors and pathogen resistance,
M. Suresh, J.D. Altman, R. Ahmed, Viral immune evasion Cell. Microbiol. 5 (2003) 143 – 153.
due to persistence of activated T cells without effector [32] S. Janssens, R. Beyaert, Role of Toll-like receptors in
function, J. Exp. Med. 188 (1998) 2205 – 2213. pathogen recognition, Clin. Microbiol. Rev. 16 (2003)
[18] V. Appay, D.F. Nixon, S.M. Donahoe, G.M. Gillespie, T. 637 – 646.
Dong, A. King, G.S. Ogg, H.M. Spiegel, C. Conlon, C.A. [33] S. Akira, Mammalian Toll-like receptors, Curr. Opin.
Spina, D.V. Havlir, D.D. Richman, A. Waters, P. Easterbrook, Immunol. 15 (2003) 5 – 11.
A.J. McMichael, S.L. Rowland-Jones, HIV-specific CD8(+) [34] M.F. Bachmann, R.M. Zinkernagel, A. Oxenius, Immune
T cells produce antiviral cytokines but are impaired in responses in the absence of costimulation: viruses know the
cytolytic function, J. Exp. Med. 192 (2000) 63 – 75. trick, J. Immunol. 161 (1998) 5791 – 5794.
[19] C. Ardavin, Origin, precursors and differentiation of mouse [35] V.L. Perez, L. Van Parijs, A. Biuckians, X.X. Zheng, T.B.
dendritic cells, Nat. Rev., Immunol. 3 (2003) 582 – 590. Strom, A.K. Abbas, Induction of peripheral T cell tolerance
[20] J. Banchereau, R.M. Steinman, Dendritic cells and the in vivo requires CTLA-4 engagement, Immunity 6 (1997)
control of immunity, Nature 392 (1998) 245 – 252. 411 – 417.
[21] P. Aichele, K. Brduscha-Riem, R.M. Zinkernagel, H. [36] R.H. Schwartz, T cell anergy, Annu. Rev. Immunol. 21
Hengartner, H. Pircher, T cell priming versus T cell (2003) 305 – 334.
tolerance induced by synthetic peptides, J. Exp. Med. 182 [37] O. Takeuchi, T. Kawai, P.F. Muhlradt, M. Morr, J.D. Radolf,
(1995) 261 – 266. A. Zychlinsky, K. Takeda, S. Akira, Discrimination of
350 T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355

bacterial lipoproteins by Toll-like receptor 6, Int. Immunol. [51] N. Suzuki, S. Suzuki, G.S. Duncan, D.G. Millar, T. Wada,
13 (2001) 933 – 940. C. Mirtsos, H. Takada, A. Wakeham, A. Itie, S. Li, J.M.
[38] C. Werts, R.I. Tapping, J.C. Mathison, T.H. Chuang, V. Penninger, H. Wesche, P.S. Ohashi, T.W. Mak, W.C. Yeh,
Kravchenko, I. Saint Girons, D.A. Haake, P.J. Godowski, F. Severe impairment of interleukin-1 and Toll-like receptor
Hayashi, A. Ozinsky, D.M. Underhill, C.J. Kirschning, H. signalling in mice lacking IRAK-4, Nature 416 (2002)
Wagner, A. Aderem, P.S. Tobias, R.J. Ulevitch, Leptospiral 750 – 756.
lipopolysaccharide activates cells through a TLR2-dependent [52] T. Kawai, O. Adachi, T. Ogawa, K. Takeda, S. Akira,
mechanism, Nat. Immunol. 2 (2001) 346 – 352. Unresponsiveness of MyD88-deficient mice to endotoxin,
[39] L. Alexopoulou, A.C. Holt, R. Medzhitov, R.A. Flavell, Immunity 11 (1999) 115 – 122.
Recognition of double-stranded RNA and activation of NF- [53] T. Kawai, O. Takeuchi, T. Fujita, J. Inoue, P.F. Muhlradt, S.
kappaB by Toll-like receptor 3, Nature 413 (2001) 732 – 738. Sato, K. Hoshino, S. Akira, Lipopolysaccharide stimulates
[40] A. Poltorak, X. He, I. Smirnova, M.Y. Liu, C. Van Huffel, X. the MyD88-independent pathway and results in activation of
Du, D. Birdwell, E. Alejos, M. Silva, C. Galanos, M. IFN-regulatory factor 3 and the expression of a subset of
Freudenberg, P. Ricciardi-Castagnoli, B. Layton, B. Beutler, lipopolysaccharide-inducible genes, J. Immunol. 167 (2001)
Defective LPS signaling in C3H/HeJ and C57BL/10ScCr 5887 – 5894.
mice: mutations in Tlr4 gene, Science 282 (1998) 2085 – 2088. [54] M. Yamamoto, S. Sato, H. Hemmi, H. Sanjo, S. Uematsu, T.
[41] R. Shimazu, S. Akashi, H. Ogata, Y. Nagai, K. Fukudome, K. Kaisho, K. Hoshino, O. Takeuchi, M. Kobayashi, T. Fujita,
Miyake, M. Kimoto, MD-2, a molecule that confers lip- K. Takeda, S. Akira, Essential role for TIRAP in activation of
opolysaccharide responsiveness on Toll-like receptor 4, J. the signalling cascade shared by TLR2 and TLR4, Nature
Exp. Med. 189 (1999) 1777 – 1782. 420 (2002) 324 – 329.
[42] F. Hayashi, K.D. Smith, A. Ozinsky, T.R. Hawn, E.C. Yi, D.R. [55] E. Ingulli, A. Mondino, A. Khoruts, M.K. Jenkins, In vivo
Goodlett, J.K. Eng, S. Akira, D.M. Underhill, A. Aderem, The detection of dendritic cell antigen presentation to CD4(+) T
innate immune response to bacterial flagellin is mediated by cells, J. Exp. Med. 185 (1997) 2133 – 2141.
Toll-like receptor 5, Nature 410 (2001) 1099 – 1103. [56] K.J. Maloy, I. Erdmann, V. Basch, S. Sierro, T.A. Kramps,
[43] F. Heil, H. Hemmi, H. Hochrein, F. Ampenberger, C. R.M. Zinkernagel, S. Oehen, T.M. Kundig, Intralymphatic
Kirschning, S. Akira, G. Lipford, H. Wagner, S. Bauer, immunization enhances DNA vaccination, Proc. Natl. Acad.
Species-specific recognition of single-stranded RNA via Toll- Sci. U. S. A. 98 (2001) 3299 – 3303.
like receptor 7 and 8, Science 303 (2004) 1526 – 1529. [57] C. Ruedl, P. Koebel, K. Karjalainen, In vivo-matured
[44] S.S. Diebold, T. Kaisho, H. Hemmi, S. Akira, E.S.C. Reis, Langerhans cells continue to take up and process native
Innate antiviral responses by means of TLR7-mediated proteins unlike in vitro-matured counterparts, J. Immunol.
recognition of single-stranded RNA, Science 303 (2004) 166 (2001) 7178 – 7182.
1529 – 1531. [58] P. Pierre, S.J. Turley, E. Gatti, M. Hull, J. Meltzer, A. Mirza,
[45] M. Urosevic, T. Maier, B. Benninghoff, H. Slade, G. Burg, R. K. Inaba, R.M. Steinman, I. Mellman, Developmental
Dummer, Mechanisms underlying imiquimod-induced regulation of MHC class II transport in mouse dendritic
regression of basal cell carcinoma in vivo, Arch. Dermatol. cells, Nature 388 (1997) 787 – 792.
139 (2003) 1325 – 1332. [59] U. Schubert, L.C. Anton, J. Gibbs, C.C. Norbury, J.W.
[46] H. Hemmi, T. Kaisho, O. Takeuchi, S. Sato, H. Sanjo, K. Yewdell, J.R. Bennink, Rapid degradation of a large fraction
Hoshino, T. Horiuchi, H. Tomizawa, K. Takeda, S. Akira, of newly synthesized proteins by proteasomes, Nature 404
Small anti-viral compounds activate immune cells via the (2000) 770 – 774.
TLR7 MyD88-dependent signaling pathway, Nat. Immunol. [60] T.J. Braciale, L.A. Morrison, M.T. Sweetser, J. Sambrook,
3 (2002) 196 – 200. M.J. Gething, V.L. Braciale, Antigen presentation pathways
[47] S. Bauer, C.J. Kirschning, H. Hacker, V. Redecke, S. to class I and class II MHC-restricted T lymphocytes,
Hausmann, S. Akira, H. Wagner, G.B. Lipford, Human Immunol. Rev. 98 (1987) 95 – 114.
TLR9 confers responsiveness to bacterial DNA via species- [61] M.F. Bachmann, A. Oxenius, H. Pircher, H. Hengartner,
specific CpG motif recognition, Proc. Natl. Acad. Sci. U. S. A. P.A. Ashton-Richardt, S. Tonegawa, R.M. Zinkernagel,
98 (2001) 9237 – 9242. TAP1-independent loading of class I molecules by
[48] T. Chuang, R.J. Ulevitch, Identification of hTLR10: a novel exogenous viral proteins, Eur. J. Immunol. 25 (1995)
human Toll-like receptor preferentially expressed in immune 1739 – 1743.
cells, Biochim. Biophys. Acta 1518 (2001) 157 – 161. [62] M. Kovacsovics-Bankowski, K.L. Rock, A phagosome-to-
[49] M. Matsumoto, K. Funami, M. Tanabe, H. Oshiumi, M. cytosol pathway for exogenous antigens presented on MHC
Shingai, Y. Seto, A. Yamamoto, T. Seya, Subcellular locali- class I molecules, Science 267 (1995) 243 – 246.
zation of Toll-like receptor 3 in human dendritic cells, J. [63] W.R. Heath, F.R. Carbone, Cross-presentation, dendritic
Immunol. 171 (2003) 3154 – 3162. cells, tolerance and immunity, Annu. Rev. Immunol. 19
[50] P. Ahmad-Nejad, H. Hacker, M. Rutz, S. Bauer, R.M. (2001) 47 – 64.
Vabulas, H. Wagner, Bacterial CpG-DNA and lipopolysac- [64] J.M. den Haan, M.J. Bevan, Antigen presentation to CD8+ T
charides activate Toll-like receptors at distinct cellular cells: cross-priming in infectious diseases, Curr. Opin.
compartments, Eur. J. Immunol. 32 (2002) 1958 – 1968. Immunol. 13 (2001) 437 – 441.
T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355 351

[65] C. Kurts, F.R. Carbone, M. Barnden, E. Blanas, J. Allison, mediated by CD40–CD40L interactions, Nature 393 (1998)
W.R. Heath, J.F. Miller, CD4+ T cell help impairs CD8+ T cell 480 – 483.
deletion induced by cross-presentation of self-antigens and [80] S.R. Bennett, F.R. Carbone, F. Karamalis, R.A. Flavell, J.F.
favors autoimmunity, J. Exp. Med. 186 (1997) 2057 – 2062. Miller, W.R. Heath, Help for cytotoxic-T-cell responses is
[66] P.J. Chefalo, C.V. Harding, Processing of exogenous antigens mediated by CD40 signalling, Nature 393 (1998) 478 – 480.
for presentation by class I MHC molecules involves post- [81] D.J. Shedlock, H. Shen, Requirement for CD4 T cell help in
Golgi peptide exchange influenced by peptide-MHC complex generating functional CD8 T cell memory, Science 300
stability and acidic pH, J. Immunol. 167 (2001) 1274 – 1282. (2003) 337 – 339.
[67] C. Ruedl, T. Storni, F. Lechner, T. Bachi, M.F. Bachmann, [82] C. Bourgeois, H. Veiga-Fernandes, A.M. Joret, B. Rocha, C.
Cross-presentation of virus-like particles by skin-derived Tanchot, CD8 lethargy in the absence of CD4 help, Eur. J.
CD8( ) dendritic cells: a dispensable role for TAP, Eur. J. Immunol. 32 (2002) 2199 – 2207.
Immunol. 32 (2002) 818 – 825. [83] P. Johansen, P. Stamou, R.E. Tascon, D.B. Lowrie, B.
[68] L. Timares, A. Takashima, S.A. Johnston, Quantitative Stockinger, CD4 T cells guarantee optimal competitive
analysis of the immunopotency of genetically transfected fitness of CD8 memory T cells, Eur. J. Immunol. 34 (2004)
dendritic cells, Proc. Natl. Acad. Sci. U. S. A. 95 (1998) 91 – 97.
13147 – 13152. [84] C. Bourgeois, B. Rocha, C. Tanchot, A role for CD40
[69] O. Akbari, N. Panjwani, S. Garcia, R. Tascon, D. Lowrie, B. expression on CD8+ T cells in the generation of CD8+ T cell
Stockinger, DNA vaccination: transfection and activation of memory, Science 297 (2002) 2060 – 2063.
dendritic cells as key events for immunity, J. Exp. Med. 189 [85] M.F. Bachmann, U.H. Rohrer, T.M. Kundig, K. Burki, H.
(1999) 169 – 178. Hengartner, R.M. Zinkernagel, The influence of antigen
[70] M. Kopf, C. Ruedl, N. Schmitz, A. Gallimore, K. Lefrang, B. organization on B cell responsiveness, Science 262 (1993)
Ecabert, B. Odermatt, M.F. Bachmann, OX40-deficient mice 1448 – 1451.
are defective in Th cell proliferation but are competent in [86] I.C. MacLennan, Germinal centers, Annu. Rev. Immunol. 12
generating B cell and CTL responses after virus infection, (1994) 117 – 139.
Immunity 11 (1999) 699 – 708. [87] Y.J. Liu, D.E. Joshua, G.T. Williams, C.A. Smith, J. Gordon,
[71] L.A. Gravestein, J.D. Nieland, A.M. Kruisbeek, J. Borst, I.C. MacLennan, Mechanism of antigen-driven selection in
Novel mAbs reveal potent co-stimulatory activity of murine germinal centres, Nature 342 (1989) 929 – 931.
CD27, Int. Immunol. 7 (1995) 551 – 557. [88] B.O. Lee, J. Moyron-Quiroz, J. Rangel-Moreno, K.L. Kusser,
[72] A. Shahinian, K. Pfeffer, K.P. Lee, T.M. Kundig, K. L. Hartson, F. Sprague, F.E. Lund, T.D. Randall, CD40, but not
Kishihara, A. Wakeham, K. Kawai, P.S. Ohashi, C.B. CD154, expression on B cells is necessary for optimal primary
Thompson, T.W. Mak, Differential T cell costimulatory B cell responses, J. Immunol. 171 (2003) 5707 – 5717.
requirements in CD28-deficient mice, Science 261 (1993) [89] D. Gray, K. Siepmann, G. Wohlleben, CD40 ligation in B cell
609 – 612. activation, isotype switching and memory development,
[73] D. Vremec, K. Shortman, Dendritic cell subtypes in mouse Semin. Immunol. 6 (1994) 303 – 310.
lymphoid organs: cross-correlation of surface markers, [90] J. Banchereau, P. de Paoli, A. Valle, E. Garcia, F. Rousset,
changes with incubation, and differences among thymus, Long-term human B cell lines dependent on interleukin-4 and
spleen, and lymph nodes, J. Immunol. 159 (1997) 565 – 573. antibody to CD40, Science 251 (1991) 70 – 72.
[74] A. Langenkamp, M. Messi, A. Lanzavecchia, F. Sallusto, [91] M.H. Kosco-Vilbois, Are follicular dendritic cells really good
Kinetics of dendritic cell activation: impact on priming of for nothing? Nat. Rev., Immunol. 3 (2003) 764 – 769.
TH1, TH2 and nonpolarized T cells, Nat. Immunol. 1 (2000) [92] M. Maruyama, K.P. Lam, K. Rajewsky, Memory B-cell
311 – 316. persistence is independent of persisting immunizing antigen,
[75] J.J. Bird, D.R. Brown, A.C. Mullen, N.H. Moskowitz, M.A. Nature 407 (2000) 636 – 642.
Mahowald, J.R. Sider, T.F. Gajewski, C.R. Wang, S.L. [93] T.E. Mandel, R.P. Phipps, A. Abbot, J.G. Tew, The follicular
Reiner, Helper T cell differentiation is controlled by the cell dendritic cell: long term antigen retention during immunity,
cycle, Immunity 9 (1998) 229 – 237. Immunol. Rev. 53 (1980) 29 – 59.
[76] S.M. Kaech, R. Ahmed, Memory CD8+ T cell differentiation: [94] J.G. Tew, R.P. Phipps, T.E. Mandel, The maintenance and
initial antigen encounter triggers a developmental program in regulation of the humoral immune response: persisting
naive cells, Nat. Immunol. 2 (2001) 415 – 422. antigen and the role of follicular antigen-binding dendritic
[77] T. Storni, C. Ruedl, W.A. Renner, M.F. Bachmann, Innate cells as accessory cells, Immunol. Rev. 53 (1980) 175 – 201.
immunity together with duration of antigen persistence [95] U. Karrer, C. Lopez-Macias, A. Oxenius, B. Odermatt, M.F.
regulate effector T cell induction, J. Immunol. 171 (2003) Bachmann, U. Kalinke, H. Bluethmann, H. Hengartner, R.M.
795 – 801. Zinkernagel, Antiviral B cell memory in the absence of
[78] J.P. Ridge, F. Di Rosa, P. Matzinger, A conditioned dendritic mature follicular dendritic cell networks and classical
cell can be a temporal bridge between a CD4+ T-helper and a germinal centers in TNFR1 / mice, J. Immunol. 164
T-killer cell, Nature 393 (1998) 474 – 478. (2000) 768 – 778.
[79] S.P. Schoenberger, R.E. Toes, E.I. van der Voort, R. Offringa, [96] M. Le Hir, H. Bluethmann, M.H. Kosco-Vilbois, M. Muller,
C.J. Melief, T-cell help for cytotoxic T lymphocytes is F. di Padova, M. Moore, B. Ryffel, H.P. Eugster, Tumor
352 T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355

necrosis factor receptor-1 signaling is required for differ- [111] R.W. Ellis, The new generation of recombinant viral subunit
entiation of follicular dendritic cells, germinal center vaccines, Curr. Opin. Biotechnol. 7 (1996) 646 – 652.
formation, and full antibody responses, J. Inflam. 47 [112] G. Ada, Vaccines and vaccination, N. Engl. J. Med. 345
(1995) 76 – 80. (2001) 1042 – 1053.
[97] T.M. Kundig, M.F. Bachmann, S. Oehen, U.W. Hoffmann, [113] G.L. Smith, B.R. Murphy, B. Moss, Construction and
J.J. Simard, C.P. Kalberer, H. Pircher, P.S. Ohashi, H. characterization of an infectious vaccinia virus recombinant
Hengartner, R.M. Zinkernagel, On the role of antigen in that expresses the influenza hemagglutinin gene and induces
maintaining cytotoxic T-cell memory, Proc. Natl. Acad. Sci. resistance to influenza virus infection in hamsters, Proc. Natl.
U. S. A. 93 (1996) 9716 – 9723. Acad. Sci. U. S. A. 80 (1983) 7155 – 7159.
[98] R. Ahmed, D. Gray, Immunological memory and protective [114] B. Moss, Genetically engineered poxviruses for recombinant
immunity: understanding their relation, Science 272 (1996) gene expression, vaccination, and safety, Proc. Natl. Acad.
54 – 60. Sci. U. S. A. 93 (1996) 11341 – 11348.
[99] R.M. Zinkernagel, On differences between immunity and [115] M.W. Carroll, W.W. Overwijk, R.S. Chamberlain, S.A.
immunological memory, Curr. Opin. Immunol. 14 (2002) Rosenberg, B. Moss, N.P. Restifo, Highly attenuated
523 – 536. modified vaccinia virus Ankara (MVA) as an effective
[100] D. Gray, P. Dullforce, S. Jainandunsing, Memory B cell recombinant vector: a murine tumor model, Vaccine 15
development but not germinal center formation is impaired (1997) 387 – 394.
by in vivo blockade of CD40–CD40 ligand interaction, J. [116] C. Wiethe, K. Dittmar, T. Doan, W. Lindenmaier, R. Tindle,
Exp. Med. 180 (1994) 141 – 155. Enhanced effector and memory CTL responses generated by
[101] M.F. Bachmann, T.M. Kundig, H. Hengartner, R.M. Zinker- incorporation of receptor activator of NF-kappa B (RANK)/
nagel, Protection against immunopathological consequences RANK ligand costimulatory molecules into dendritic cell
of a viral infection by activated but not resting cytotoxic T immunogens expressing a human tumor-specific antigen, J.
cells: T cell memory without bmemory T cellsQ? Proc. Natl. Immunol. 171 (2003) 4121 – 4130.
Acad. Sci. U. S. A. 94 (1997) 640 – 645. [117] A. Darji, S. zur Lage, A.I. Garbe, T. Chakraborty, S. Weiss,
[102] F. Sallusto, D. Lenig, R. Forster, M. Lipp, A. Lanzavecchia, Oral delivery of DNA vaccines using attenuated Salmonella
Two subsets of memory T lymphocytes with distinct typhimurium as carrier, FEMS Immunol. Med. Microbiol. 27
homing potentials and effector functions, Nature 401 (2000) 341 – 349.
(1999) 708 – 712. [118] A. Darji, C.A. Guzman, B. Gerstel, P. Wachholz, K.N.
[103] J.T. Opferman, B.T. Ober, P.G. Ashton-Rickardt, Linear Timmis, J. Wehland, T. Chakraborty, S. Weiss, Oral somatic
differentiation of cytotoxic effectors into memory T lympho- transgene vaccination using attenuated S. typhimurium, Cell
cytes, Science 283 (1999) 1745 – 1748. 91 (1997) 765 – 775.
[104] H. Unsoeld, S. Krautwald, D. Voehringer, U. Kunzendorf, H. [119] C.M. Khan, B. Villarreal-Ramos, R.J. Pierce, G. Riveau, R.
Pircher, Cutting edge: CCR7+ and CCR7 memory T cells Demarco de Hormaeche, H. McNeill, T. Ali, N. Fair-
do not differ in immediate effector cell function, J. Immunol. weather, S. Chatfield, A. Capron, et al., Construction,
169 (2002) 638 – 641. expression, and immunogenicity of the Schistosoma man-
[105] D.L. Barber, E.J. Wherry, R. Ahmed, Cutting edge: rapid in soni P28 glutathione S-transferase as a genetic fusion to
vivo killing by memory CD8 T cells, J. Immunol. 171 (2003) tetanus toxin fragment C in a live Aro attenuated vaccine
27 – 31. strain of Salmonella, Proc. Natl. Acad. Sci. U. S. A. 91
[106] E.J. Wherry, V. Teichgraber, T.C. Becker, D. Masopust, S.M. (1994) 11261 – 11265.
Kaech, R. Antia, U.H. von Andrian, R. Ahmed, Lineage [120] E.M. Mauriello, H. Duc le, R. Isticato, G. Cangiano,
relationship and protective immunity of memory CD8 T cell H.A. Hong, M. De Felice, E. Ricca, S.M. Cutting,
subsets, Nat. Immunol. 4 (2003) 225 – 234. Display of heterologous antigens on the Bacillus subtilis
[107] T. Pang, R.V. Blanden, Regulation of the T-cell response to spore coat using CotC as a fusion partner, Vaccine 22
ectromelia virus infection: I. Feedback suppression by (2004) 1177 – 1187.
effector T cells, J. Exp. Med. 143 (1976) 469 – 481. [121] H.S. Garmory, K.F. Griffin, K.A. Brown, R.W. Titball, Oral
[108] P.G. Stevenson, G.T. Belz, M.R. Castrucci, J.D. Altman, P.C. immunisation with live aroA attenuated Salmonella enterica
Doherty, A gamma-herpesvirus sneaks through a CD8(+) T serovar Typhimurium expressing the Yersinia pestis V
cell response primed to a lytic-phase epitope, Proc. Natl. antigen protects mice against plague, Vaccine 21 (2003)
Acad. Sci. U. S. A. 96 (1999) 9281 – 9286. 3051 – 3057.
[109] P. Seiler, M.A. Brundler, C. Zimmermann, D. Weibel, M. [122] I. Guleria, R. Teitelbaum, R.A. McAdam, G. Kalpana, W.R.
Bruns, H. Hengartner, R.M. Zinkernagel, Induction of Jacobs Jr., B.R. Bloom, Auxotrophic vaccines for tuber-
protective cytotoxic T cell responses in the presence of high culosis, Nat. Med. 2 (1996) 334 – 337.
titers of virus-neutralizing antibodies: implications for [123] G.R. Stewart, V.A. Snewin, G. Walzl, T. Hussell, P. Tormay,
passive and active immunization, J. Exp. Med. 187 (1998) P. O’Gaora, M. Goyal, J. Betts, I.N. Brown, D.B. Young,
649 – 654. Overexpression of heat-shock proteins reduces survival of
[110] S. Oehen, H. Hengartner, R.M. Zinkernagel, Vaccination for Mycobacterium tuberculosis in the chronic phase of infec-
disease, Science 251 (1991) 195 – 198. tion, Nat. Med. 7 (2001) 732 – 737.
T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355 353

[124] J. Hess, U. Schaible, B. Raupach, S.H. Kaufmann, Exploiting [141] P. Johansen, Y. Men, H.P. Merkle, B. Gander, Revisiting
the immune system: toward new vaccines against intra- PLA/PLGA microspheres: an analysis of their potential in
cellular bacteria, Adv. Immunol. 75 (2000) 1 – 88. parenteral vaccination, Eur. J. Pharm. Biopharm. 50 (2000)
[125] N. Dhar, V. Rao, A.K. Tyagi, Skewing of the Th1/Th2 129 – 146.
responses in mice due to variation in the level of expression [142] T. Uchida, STX-liposome conjugates as candidate vaccines,
of an antigen in a recombinant BCG system, Immunol. Lett. Drugs Today (Barc.) 39 (2003) 673 – 693.
88 (2003) 175 – 184. [143] R. Zurbriggen, Immunostimulating reconstituted influenza
[126] N.J. Leung, A. Aldovini, R. Young, M.A. Jarvis, J.M. Smith, virosomes, Vaccine 21 (2003) 921 – 924.
D. Meyer, D.E. Anderson, M.P. Carlos, M.B. Gardner, J.V. [144] G.F. Kersten, D.J. Crommelin, Liposomes and ISCOMs,
Torres, The kinetics of specific immune responses in rhesus Vaccine 21 (2003) 915 – 920.
monkeys inoculated with live recombinant BCG expressing [145] A. Engering, T.B. Geijtenbeek, S.J. van Vliet, M. Wijers, E.
SIV Gag, Pol, Env, and Nef proteins, Virology 268 (2000) van Liempt, N. Demaurex, A. Lanzavecchia, J. Fransen, C.G.
94 – 103. Figdor, V. Piguet, Y. van Kooyk, The dendritic cell-specific
[127] I.A. Jabbar, G.J. Fernando, N. Saunders, A. Aldovini, R. adhesion receptor DC-SIGN internalizes antigen for presen-
Young, K. Malcolm, I.H. Frazer, Immune responses induced tation to T cells, J. Immunol. 168 (2002) 2118 – 2126.
by BCG recombinant for human papillomavirus L1 and E7 [146] K. Mahnke, M. Guo, S. Lee, H. Sepulveda, S.L. Swain, M.
proteins, Vaccine 18 (2000) 2444 – 2453. Nussenzweig, R.M. Steinman, The dendritic cell receptor for
[128] J.A. Hoffmann, F.C. Kafatos, C.A. Janeway, R.A. Ezekowitz, endocytosis, DEC-205, can recycle and enhance antigen
Phylogenetic perspectives in innate immunity, Science 284 presentation via major histocompatibility complex class II-
(1999) 1313 – 1318. positive lysosomal compartments, J. Cell Biol. 151 (2000)
[129] W.G. Degen, T. Jansen, V.E. Schijns, Vaccine adjuvant 673 – 684.
technology: from mechanistic concepts to practical applica- [147] L. Bonifaz, D. Bonnyay, K. Mahnke, M. Rivera, M.C.
tions, Expert Rev. Vaccines 2 (2003) 327 – 335. Nussenzweig, R.M. Steinman, Efficient targeting of protein
[130] V.E. Schijns, Mechanisms of vaccine adjuvant activity: antigen to the dendritic cell receptor DEC-205 in the steady
initiation and regulation of immune responses by vaccine state leads to antigen presentation on major histocompati-
adjuvants, Vaccine 21 (2003) 829 – 831. bility complex class I products and peripheral CD8+ T cell
[131] R.K. Gupta, E.H. Relyveld, E.B. Lindblad, B. Bizzini, S. tolerance, J. Exp. Med. 196 (2002) 1627 – 1638.
Ben-Efraim, G. C.K., Adjuvants—a balance between toxicity [148] P. Guermonprez, C. Fayolle, M.J. Rojas, M. Rescigno, D.
and adjuvanticity, Vaccine 11 (1993) 293 – 306. Ladant, C. Leclerc, In vivo receptor-mediated delivery of a
[132] D.J. Marciani, Vaccine adjuvants: role and mechanisms of recombinant invasive bacterial toxoid to CD11c+ CD8 alpha
action in vaccine immunogenicity, Drug Discov. Today 8 CD11b high dendritic cells, Eur. J. Immunol. 32 (2002)
(2003) 934 – 943. 3071 – 3081.
[133] F. Villinger, Cytokines as clinical adjuvants: how far are we? [149] T. Storni, C. Ruedl, K. Schwarz, R.A. Schwendener, W.A.
Expert Rev. Vaccines 2 (2003) 317 – 326. Renner, M.F. Bachmann, Nonmethylated CG motifs pack-
[134] V.E. Schijns, Immunological concepts of vaccine adjuvant aged into virus-like particles induce protective cytotoxic T
activity, Curr. Opin. Immunol. 12 (2000) 456 – 463. cell responses in the absence of systemic side effects, J.
[135] R.M. Zinkernagel, S. Ehl, P. Aichele, S. Oehen, T. Kundig, Immunol. 172 (2004) 1777 – 1785.
H. Hengartner, Antigen localisation regulates immune [150] A.M. Krieg, CpG motifs in bacterial DNA and their immune
responses in a dose- and time-dependent fashion: a geo- effects, Annu. Rev. Immunol. 20 (2002) 709 – 760.
graphical view of immune reactivity, Immunol. Rev. 156 [151] B.D. Livingston, M. Newman, C. Crimi, D. McKinney, R.
(1997) 199 – 209. Chesnut, A. Sette, Optimization of epitope processing
[136] P. Matzinger, The danger model: a renewed sense of self, enhances immunogenicity of multiepitope DNA vaccines,
Science 296 (2002) 301 – 305. Vaccine 19 (2001) 4652 – 4660.
[137] P. Matzinger, Tolerance, danger, and the extended family, [152] S. Oehen, T. Junt, C. Lopez-Macias, T.A. Kramps, Antiviral
Annu. Rev. Immunol. 12 (1994) 991 – 1045. protection after DNA vaccination is short lived and not
[138] M. Watanabe, M. Nagai, K. Funaishi, M. Endoh, Efficacy of enhanced by CpG DNA, Immunology 99 (2000) 163 – 169.
chemically cross-linked antigens for acellular pertussis [153] J.J. Kim, M.L. Bagarazzi, N. Trivedi, Y. Hu, K. Kazahaya,
vaccine, Vaccine 19 (2000) 1199 – 1203. D.M. Wilson, R. Ciccarelli, M.A. Chattergoon, K. Dang, S.
[139] W.T. Phillips, R. Klipper, B. Goins, Novel method of greatly Mahalingam, A.A. Chalian, M.G. Agadjanyan, J.D. Boyer,
enhanced delivery of liposomes to lymph nodes, J. Pharma- B. Wang, D.B. Weiner, Engineering of in vivo immune
col. Exp. Ther. 295 (2000) 309 – 313. responses to DNA immunization via codelivery of cos-
[140] A. Jegerlehner, A. Tissot, F. Lechner, P. Sebbel, I. Erdmann, timulatory molecule genes, Nat. Biotechnol. 15 (1997)
T. Kundig, T. Bachi, T. Storni, G. Jennings, P. Pumpens, 641 – 646.
W.A. Renner, M.F. Bachmann, A molecular assembly [154] C.F. Hung, W.F. Cheng, K.F. Hsu, C.Y. Chai, L. He, M. Ling,
system that renders antigens of choice highly repetitive T.C. Wu, Cancer immunotherapy using a DNA vaccine
for induction of protective B cell responses, Vaccine 20 encoding the translocation domain of a bacterial toxin linked
(2002) 3104 – 3112. to a tumor antigen, Cancer Res. 61 (2001) 3698 – 3703.
354 T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355

[155] P. Zajac, A. Schutz, D. Oertli, C. Noppen, C. Schaefer, [166] S. Ozbas-Turan, J. Akbuga, C. Aral, Controlled release of
M. Heberer, G.C. Spagnoli, W.R. Marti, Enhanced interleukin-2 from chitosan microspheres, J. Pharm. Sci. 91
generation of cytotoxic T lymphocytes using recombinant (2002) 1245 – 1251.
vaccinia virus expressing human tumor-associated antigens [167] U. Palendira, A.T. Kamath, C.G. Feng, E. Martin, P.J.
and B7 costimulatory molecules, Cancer Res. 58 (1998) Chaplin, J.A. Triccas, W.J. Britton, Coexpression of inter-
4567 – 4571. leukin-12 chains by a self-splicing vector increases the
[156] S. Santra, D.H. Barouch, A.H. Sharpe, N.L. Letvin, B7 co- protective cellular immune response of DNA and Mycobac-
stimulatory requirements differ for induction of immune terium bovis BCG vaccines against Mycobacterium tuber-
responses by DNA, protein and recombinant pox virus culosis, Infect. Immun. 70 (2002) 1949 – 1956.
vaccination, Eur. J. Immunol. 30 (2000) 2650 – 2659. [168] Y. Shibata, I. Honda, J.P. Justice, M.R. Van Scott, R.M.
[157] S. Santra, D.H. Barouch, S.S. Jackson, M.J. Kuroda, J.E. Nakamura, Q.N. Myrvik, Th1 adjuvant N-acetyl-D-glucos-
Schmitz, M.A. Lifton, A.H. Sharpe, N.L. Letvin, Functional amine polymer up-regulates Th1 immunity but down-
equivalency of B7-1 and B7-2 for costimulating plasmid regulates Th2 immunity against a mycobacterial protein
DNA vaccine-elicited CTL responses, J. Immunol. 165 (MPB-59) in interleukin-10-knockout and wild-type mice,
(2000) 6791 – 6795. Infect. Immun. 69 (2001) 6123 – 6130.
[158] T. Tsuji, K. Hamajima, N. Ishii, I. Aoki, J. Fukushima, K.Q. [169] K. Sorimachi, K. Akimoto, Y. Hattori, T. Ieiri, A. Niwa,
Xin, S. Kawamoto, S. Sasaki, K. Matsunaga, Y. Ishigatsubo, Secretion of TNF-alpha, IL-8 and nitric oxide by macro-
K. Tani, T. Okubo, K. Okuda, Immunomodulatory effects of phages activated with polyanions, and involvement of
a plasmid expressing B7-2 on human immunodeficiency interferon-gamma in the regulation of cytokine secretion,
virus-1-specific cell-mediated immunity induced by a plas- Cytokine 11 (1999) 571 – 578.
mid encoding the viral antigen, Eur. J. Immunol. 27 (1997) [170] T. Ueno, K. Ohtawa, Y. Kimoto, K. Sakurai, Y. Kodera, M.
782 – 787. Hiroto, A. Matsushima, H. Nishimura, Y. Inada, Polyethylene
[159] C. Wiethe, K. Dittmar, T. Doan, W. Lindenmaier, R. Tindle, glycol-modified concanavalin A as an effective agent to
Provision of 4-1BB ligand enhances effector and memory stimulate anti-tumor cytotoxicity, Cancer Detec. Prev. 24
CTL responses generated by immunization with dendritic (2000) 100 – 106.
cells expressing a human tumor-associated antigen, J. [171] B. Rihova, Immunomodulating activities of soluble synthetic
Immunol. 170 (2003) 2912 – 2922. polymer-bound drugs, Adv. Drug Deliv. Rev. 54 (2002)
[160] Y. Miyahira, H. Akiba, M. Katae, K. Kubota, S. Kobayashi, 653 – 674.
T. Takeuchi, A. Garcia-Sastre, Y. Fukuchi, K. Okumura, H. [172] P. Strong, H. Clark, K. Reid, Intranasal application of chitin
Yagita, T. Aoki, Cutting edge: a potent adjuvant effect of microparticles down-regulates symptoms of allergic hyper-
ligand to receptor activator of NF-kappaB gene for inducing sensitivity to Dermatophagoides pteronyssinus and Asper-
antigen-specific CD8+ T cell response by DNA and viral gillus fumigatus in murine models of allergy, Clin. Exp.
vector vaccination, J. Immunol. 171 (2003) 6344 – 6348. Allergy 32 (2002) 1794 – 1800.
[161] C.H. Poehlein, H.M. Hu, J. Yamada, I. Assmann, W.G. [173] C. Ruedl, M.F. Bachmann, M. Kopf, The antigen dose
Alvord, W.J. Urba, B.A. Fox, TNF plays an essential role in determines T helper subset development by regulation of
tumor regression after adoptive transfer of perforin/IFN- CD40 ligand, Eur. J. Immunol. 30 (2000) 2056 – 2064.
gamma double knockout effector T cells, J. Immunol. 170 [174] P.R. Rogers, M. Croft, Peptide dose, affinity, and time of
(2003) 2004 – 2013. differentiation can contribute to the Th1/Th2 cytokine
[162] I. Bank, A. Achiron, G. Levie, A. Koltakov, M. Mandel, balance, J. Immunol. 163 (1999) 1205 – 1213.
Interaction of disease-related antigen-reactive T-cell lines [175] P. Johansen, H.P. Merkle, B. Gander, Physico-chemical and
from multiple sclerosis patients with type IV collagen: role of antigenic properties of tetanus and diphtheria toxoids and
integrin VLA-1 and effects of irradiation, J. Clin. Immunol. steps towards improved stability, Biochim. Biophys. Acta
22 (2002) 153 – 163. 1425 (1998) 425 – 436.
[163] H.I. Huang, P.Y. Wu, C.Y. Teo, M.N. Chen, Y.C. Chen, D. [176] P. Johansen, B. Gander, H.P. Merkle, D. Sesardic,
Silin, M.H. Tao, Improved immunogenicity of a self tumor Ambiguities in the preclinical quality assessment of
antigen by covalent linkage to CD40 ligand, Int. J. Cancer microparticulate vaccines, Trends Biotechnol. 18 (2000)
108 (2004) 696 – 703. 203 – 211.
[164] K.J. Liu, L.F. Lu, H.T. Cheng, Y.M. Hung, S.R. Shiou, J. [177] R.J. Boyton, D.M. Altmann, Is selection for TCR affinity a
Whang-Peng, S.H. Juang, Concurrent delivery of tumor factor in cytokine polarization? Trends Immunol. 23 (2002)
antigens and activation signals to dendritic cells by irradiated 526 – 529.
CD40 ligand-transfected tumor cells resulted in efficient [178] K. Blaser, Allergen dose dependent cytokine production
activation of specific CD8+ T cells, Cancer Gene Ther. 11 regulates specific IgE and IgG antibody production, Adv.
(2004) 135 – 147. Exp. Med. Biol. 409 (1996) 295 – 303.
[165] J.W. Koten, M.J. Van Luyn, J.A. Cadee, L. Brouwer, W.E. [179] A. Jegerlehner, T. Storni, G. Lipowsky, M. Schmid, P.
Hennink, C. Bijleveld, W. Den Otter, IL-2 loaded dextran Pumpens, M.F. Bachmann, Regulation of IgG antibody
microspheres with attractive histocompatibility properties for responses by epitope density and CD21-mediated costimula-
local IL-2 cancer therapy, Cytokine 24 (2003) 57 – 66. tion, Eur. J. Immunol. 32 (2002) 3305 – 3314.
T. Storni et al. / Advanced Drug Delivery Reviews 57 (2005) 333–355 355

[180] J. Wild, M.J. Grusby, R. Schirmbeck, J. Reimann, Priming towards a peptide-based vaccine against hepatitis C virus,
MHC-I-restricted cytotoxic T lymphocyte responses to Mol. Immunol. 38 (2001) 475 – 484.
exogenous hepatitis B surface antigen is CD4+ T cell [184] R. Zurbriggen, I. Novak-Hofer, A. Seelig, R. Gluck, IRIV-
dependent, J. Immunol. 163 (1999) 1880 – 1887. adjuvanted hepatitis A vaccine: in vivo absorption and
[181] R. Schirmbeck, W. Bohm, K. Melber, J. Reimann, Processing biophysical characterization, Prog. Lipid Res. 39 (2000)
of exogenous heat-aggregated (denatured) and particulate 3 – 18.
(native) hepatitis B surface antigen for class I-restricted [185] R. Glueck, Review of intranasal influenza vaccine, Adv.
epitope presentation, J. Immunol. 155 (1995) 4676 – 4684. Drug Deliv. Rev. 51 (2001) 203 – 211.
[182] P. Pumpens, E. Grens, Hepatitis B core particles as a [186] B.R. Holzer, C. Hatz, D. Schmidt-Sissolak, R. Gluck, B.
universal display model: a structure–function basis for Althaus, M. Egger, Immunogenicity and adverse effects of
development, FEBS Lett. 442 (1999) 1 – 6. inactivated virosome versus alum-adsorbed hepatitis A
[183] I.P. Hunziker, R. Zurbriggen, R. Glueck, O.B. Engler, J. vaccine: a randomized controlled trial, Vaccine 14 (1996)
Reichen, W.J. Dai, W.J. Pichler, A. Cerny, Perspectives: 982 – 986.

You might also like