Sie sind auf Seite 1von 10

REVIEW ARTICLE

published: 16 May 2013


doi: 10.3389/fimmu.2013.00114

Mechanisms of action of adjuvants


Sunita Awate 1,2 , Lorne A. Babiuk 3 and George Mutwiri 1,2 *
1
Vaccine and Infectious Disease Organization-International Vaccine Centre, School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada
2
Vaccinology and Immunotherapeutics program, School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada
3
University of Alberta, Edmonton, AB, Canada

Edited by: Adjuvants are used in many vaccines, but their mechanisms of action are not fully under-
Volker Gerdts, Vaccine and Infectious
stood. Studies from the past decade on adjuvant mechanisms are slowly revealing the
Disease Organization-International
Vaccine Centre, Canada secrets of adjuvant activity. In this review, we have summarized the recent progress in
Reviewed by: our understanding of the mechanisms of action of adjuvants. Adjuvants may act by a com-
Sylvie Fournel, Strasbourg University, bination of various mechanisms including formation of depot, induction of cytokines and
France chemokines, recruitment of immune cells, enhancement of antigen uptake and presenta-
Paola Massari, Boston University,
tion, and promoting antigen transport to draining lymph nodes. It appears that adjuvants
USA
activate innate immune responses to create a local immuno-competent environment at
*Correspondence:
George Mutwiri , Vaccine and the injection site. Depending on the type of innate responses activated, adjuvants can alter
Infectious Disease the quality and quantity of adaptive immune responses. Understanding the mechanisms
Organization-International Vaccine of action of adjuvants will provide critical information on how innate immunity influences
Centre, School of Public Health,
the development of adaptive immunity, help in rational design of vaccines against various
University of Saskatchewan, 120
Veterinary Road, Saskatoon, SK S7N diseases, and can inform on adjuvant safety.
5E3, Canada.
Keywords: adjuvants, mechanisms, innate immunity, cell recruitment and activation, inflammasomes, antigen
e-mail: george.mutwiri@usask.ca
presentation, dendritic cells

INTRODUCTION maturation of APC [increased major histocompatibility complex


The goal of vaccination is induction of protective immunity and (MHC) class II and co-stimulatory molecules expression] and
in some vaccines this can be enhanced by addition of adju- migration to the draining lymph nodes, and (6) activation of
vants. Adjuvants (Latin word adjuvare, meaning to help or aid) inflammasomes (Figure 1) (Cox and Coulter, 1997; Hoebe et al.,
were first described by Ramon as substances used in combi- 2004; Fraser et al., 2007). In this review, we will address the pro-
nation with a specific antigen that produced a more robust posed mechanisms of action of vaccine adjuvants with specific
immune response than the antigen alone (Ramon, 1924). Many emphasis on licensed adjuvants (Table 1).
diverse classes of compounds have been assessed as adjuvants
including mineral salts, microbials products, emulsions, saponins, FORMATION OF DEPOT AT THE SITE OF INJECTION
cytokines, polymers, microparticles, and liposomes (Guy, 2007). The formation of a depot at the injection site is perhaps the oldest
Based on their proposed mechanisms of action, vaccine adjuvants and most widely recognized mechanism of action of adjuvants.
have been broadly divided into delivery systems and immuno- Antigen trapping and slow release at the site of injection ensures
stimulatory adjuvants (Singh and OHagan, 2003). In general, constant stimulation of the immune system for production of
delivery systems were previously thought to act by providing a high antibody titers (Siskind and Benacerraf, 1969). Until recently,
depot while immuno-stimulatory adjuvants activate cells of the depot effect was considered a classic mechanism of action of many
innate immune system (Pashine et al., 2005). However, this classi- adjuvants. Glenny et al. (1926) were the first to propose the impor-
fication is no longer appropriate since now there is evidence that tance of depot formation in the adjuvant activity of alum. Antigen
some delivery systems can activate innate immunity. was detected for 23 weeks in alumina gel-induced granulomas
Surprisingly, despite the wide use of vaccine adjuvants in bil- (Osebold, 1982). Antigens are simply adsorbed onto the alum but
lions of doses of human and animal vaccines, the mechanisms of the binding is proposed to be due to strong electrostatic inter-
action by which they potentiate immune responses are not well action between antigen and alum (Burrell et al., 2000), which
characterized. This is well captured in a famous quote by Janeway enhanced antigen uptake and presentation by APCs (Mannhal-
(1989) who observed that adjuvants are the immunologists ter et al., 1985). Various other adjuvants such as water-in-oil
dirty little secret. However, recent advances in immunobiological emulsions [Complete Freunds Adjuvant (CFA)] and biodegrad-
research have revealed several mechanisms by which adjuvants act. able micro-and nano-particles were shown to act by depot effect
Available evidence suggests that adjuvants employ one or more of to generate prolonged and sustained high antibody titers (Herbert,
the following mechanisms to elicit immune responses: (1) sus- 1968; Kreuter, 1988). AS04, an adjuvant combination consist-
tained release of antigen at the site of injection (depot effect), (2) ing of monophosphoryl lipid A (MPL) and alum was shown to
up-regulation of cytokines and chemokines, (3) cellular recruit- induce optimal immune responses only when co-localized with
ment at the site of injection, (4) increase antigen uptake and antigen (Didierlaurent et al., 2009). The presence of alum in
presentation to antigen presenting cells (APC), (5) activation and AS04 is important in stabilizing the MPL and antigen within the

www.frontiersin.org May 2013 | Volume 4 | Article 114 | 1


Awate et al. Adjuvant mechanisms

FIGURE 1 | Proposed mechanisms of action of adjuvants. (1) Some express various PRRs both on the surface (TLRs, CLRs) and intracellularly
adjuvants presumably form a depot at the site of injection, which is (NLRs and RLRs), which are recognized and/or are activated by the adjuvants.
associated with slow release of antigen. (2) Other adjuvants are associated (5) This leads to maturation and activation of recruited APCs. Mature APCs
with transient secretion of cytokines and chemokines. (3) Secreted cytokines up-regulate the expression of MHC and co-stimulatory molecules. (6) They are
and chemokines are involved in recruitment of various immune cells to the also characterized by increased capacity for antigen processing and
injection site. These recruited cells secrete cytokines and chemokines, in turn presentation. (7) Mature APCs then migrate to the draining lymph nodes to
attract other immune cells. All these events lead to formation of a local interact with antigen-specific B or T cell to (8) activate potent antibody
immuno-competent environment at the injection site. (4) The recruited APCs secreting B cells and/or effector CD8+ T cell responses.

Frontiers in Immunology | Immunotherapies and Vaccines May 2013 | Volume 4 | Article 114 | 2
Awate et al. Adjuvant mechanisms

Table 1 | Mechanisms of action of adjuvants licensed for human use.

Adjuvants Proposed mechanisms of Immune response Licensed vaccines Reference


action activated

Alum No depot effect Ab responses Many human vaccines (e.g., DTap, Gavin et al. (2006), Franchi and
Hib, Hepatitis A, Hepatitis B) Nez (2008), Kool et al. (2008a),
McKee et al. (2009), Hutchison
et al. (2012)
NLRP3 activation in vivo?
Independent of TLR signaling
Local cytokines and chemokines Th2 responses
Cell recruitment (eosinophils, Poor Th1 responses
monocytes, macrophages)
Ag presentation

MF59 No depot effect Balanced Th1 and Th2 Licensed for influenza vaccine Dupuis et al. (1999), Mosca et al.
responses (Fluad ), H5N1 pre-pandemic (2008), Calabro et al. (2011),
vaccine (Aflunov ), H1N1 pandemic Ellebedy et al. (2011)
vaccines (Focetria and Celtura )
NLRP3 independent but
ASC-dependent
Independent of TLR signaling but
MyD88-dependent for Ab responses
Local cytokines and chemokines
Cell recruitment (neutrophils,
macrophages, and monocytes)
Ag uptake
Activate muscle cells
Ag-loaded neutrophils and
monocytes in dLNs

AS04 MPL signals through TLR4 to activate Ab responses Licensed for human papilloma virus Didierlaurent et al. (2009)
APCs (HPV) (Cervarix), hepatitis B virus
(Fendrix )
Local cytokines and chemokines
Cell recruitment (DCs and Th1 responses
monocytes)
Ag-loaded DCs and monocytes in
dLNs

AS03 Spatio-temporal co-localization with Ab responses Licensed for pandemic flu vaccine Morel et al. (2011)
Ag (Pandemrix )
Transient cytokines locally and in
dLNs
Cell recruitment (granulocytes and Immune memory
monocytes)
Ag-loaded monocytes in dLNs

Virosomes Ag delivery vehicle Ab responses Licensed for Inflexal V and Glck et al. (1992), Bungener
Invivac influenza vaccine and et al. (2002a,b), Khoshnejad et al.
hepatitis A vaccines (Epaxal ) (2007)
Bind APCs and induce CTL responses
receptor-mediated endocytosis
Escape endosomal degradation
Ag presentation via MHC class II and
MHC class I to CD4+ T cells and
CD8+ T cells respectively
Immunopotentiator

Ab, antibody; Ag, antigen; CTL, cytotoxic T lymphocytes; dLNs, draining lymph nodes.

www.frontiersin.org May 2013 | Volume 4 | Article 114 | 3


Awate et al. Adjuvant mechanisms

vaccine, along with providing a depot effect (Didierlaurent et al., and eosinophil chemotaxin eotaxin (CCL11) in the peritoneal
2009). The cationic adjuvant formulation (CAF) 01, a combina- cavity of mice (Kool et al., 2008b). Similarly, a novel adjuvant,
tion of dimethyldioctadeclammonium/trehalose-6,6-dibehenate poly[di(sodiumcarboxylatoethylphenoxy)phosphazene] (PCEP),
(DDA/TDB), which is currently in phase I clinical trial, is also induced stronger expression of adjuvant core response genes com-
thought to induce long lasting depot effect (Henriksen-Lacey et al., pared to CpG at the site of injection. Locally, PCEP triggered pro-
2010). duction of pro-inflammatory cytokines and chemokines including
There is no definitive evidence that depot effect significantly CCL2 (Awate et al., 2012).
contributes to adjuvant activity (Marrack et al., 2009). In various Alum promotes Th2-type immune responses and differentia-
studies, it has been shown that surgical removal of the antigen- tion of B cells resulting in robust antibody production (Grun and
alum depot 14 days after immunization had no effect on the Maurer, 1989). However, the role of Th2 cytokines in the adjuvant
immune responses (Schijns, 2000). Apparently, the adsorption activity of alum is not clearly defined. In vitro studies indicate
of antigen to alum was not required for alum adjuvant activ- that alum-induced activation of macrophages and up-regulation
ity (Iyer et al., 2003; De Gregorio et al., 2008). It was recently of co-stimulatory molecules did not depend on IL-4 (Rimaniol
reported that removal of the injection site 2 h after antigen and et al., 2004). However, in in vivo studies, alum-induced priming
alum administration had no effect on humoral or cell-mediated of B cells through IL-4 producing Gr1+ cells in mouse spleen,
immunity (Hutchison et al., 2012). Similarly, MF59 was rapidly which is required for proliferation of antigen-specific B cells and
cleared and did not form a depot at the injection site (Ott et al., for optimal antibody production (Jordan et al., 2004). IL-4 pro-
1995). MF59 was distributed and cleared independent of anti- ducing Gr1+ cells were mainly eosinophils, which appeared within
gen with a half-life of 42 h in the muscle tissue (Dupuis et al., 24 h and induced expansion of B cells and enhanced IgM produc-
1999). Likewise, ISCOMs tend to be rapidly transported to the tion (Wang and Weller, 2008). Further, studies with eosinophil-
draining lymph nodes after administration (Morein and Bengts- deficient mice showed that the priming of B cells was abolished
son, 1999). Together, these studies clearly indicate that depot effect after alum injection confirming the central role of eosinophils in
is not required for adjuvant activity of alum, and possibly MF59 alum-induced Th2-type immune responses (Jordan et al., 2004;
or ISCOMs. Wang and Weller, 2008). In addition, a study by Serre et al. (2008)
revealed that the Th2-type immune responses generated by alum
UP-REGULATION OF CYTOKINES AND CHEMOKINES may signal through IL-25/IL-17RB and/or IL-6 pathways.
LEADING TO CELLULAR RECRUITMENT AT THE INJECTION Alum has been shown to activate the complement cascade and
SITE recruit cells from blood to create an inflammatory environment
Recent studies on the mechanisms of adjuvants have focused at the site of injection (Ramanathan et al., 1979; Goto et al.,
on recruitment of innate immune cells at the site of injec- 1997). Similar to alum, MF59 has been shown to recruit CD11b+
tion. Particulate adjuvants have been shown to create a local blood mononuclear cells in the mouse muscle (Mosca et al., 2008).
pro-inflammatory environment to recruit immune cells (Goto Intra-peritoneal injection of alum-induced rapid cell recruitment
and Akama, 1982). Using genome wide microarray analysis, of inflammatory Ly6C+ CD11b+ monocytes. The inflammatory
Mosca et al. (2008) demonstrated that a cluster of genes encod- monocytes take up antigen, differentiate into CD11c+ MHC
ing cytokines, chemokines, innate immune receptors, interferon- class II+ DCs in a myeloid differentiation primary response gene
induced genes, and gene encoding adhesion molecules defined 88 (MyD88)-dependent manner and migrate to draining lymph
as adjuvant core response genes were commonly modulated nodes, where they induced proliferation of antigen-specific T cells
by alum, MF59, and CpG-ODN at the site of injection. Com- (Kool et al., 2008a). In similar studies by McKee et al. (2009), alum-
pared with alum and CpG-ODN (TLR9 agonist), MF59 was a induced rapid recruitment of various polymorphonuclear (PMN)
strong modulator of adjuvant core response genes. Chemokines, cells including eosinophils, monocytes, neutrophils, DCs, natural
which play a critical role in tissue specific migration of immune killer (NK), and NKT cells at the site of vaccination. Interestingly,
cells, were shown to be up-regulated by adjuvants at the injec- in cell depletion studies in mice, alum-mediated humoral and cel-
tion site. MF59 significantly up-regulated the expression of CCR2, lular responses were independent of mast cells, macrophages, and
a receptor for CCL2, which is involved in monocyte infiltra- of eosinophils (McKee et al., 2009).
tion. This was in agreement with previous in vitro results show- MF59-mediated immune cell recruitment to the injection site
ing that MF59 induced release of chemo-attractants like CCL2, has been studied in detail (Calabro et al., 2011). MF59 induced
CCL3, CCL3 and CXCL8 (Seubert et al., 2008). Further, studies recruitment of neutrophils, monocytes, eosinophils, macrophages
in CCR2-deficient mice showed that MF59-induced mononuclear followed by DCs after i.m. injection in mice. The recruited cells
cell recruitment is CCR2 dependent (Dupuis et al., 2001). Another especially neutrophils, monocytes, and B cells take up both antigen
oil-in-water emulsion AS03 co-localizes with antigen to trigger and adjuvant and traffick to draining lymph nodes. Neutrophils
colony-stimulating factor 3 (CSF3) and IL-6, and leukocyte- are the first cells to be recruited at the site of adjuvant injection and
recruiting chemokines CCL2, CCL3, and CCL5 at the site of also one of the highest in numbers. However, depletion of neu-
injection (Morel et al., 2011). Similar cytokine and chemokine trophils had no impact on the antigen-specific immune responses
mRNA expression profiles were up-regulated in the draining induced by MF59 (Calabro et al., 2011). Similar to MF59, admin-
lymph nodes (Morel et al., 2011). Likewise, alum-induced infiltra- istration of AS03 led to enhanced recruitment of neutrophils,
tion of immune cells was accompanied by production of chemo- eosinophils, and monocytes at the site of injection, which take up
attractants like CCL2, the neutrophil chemotaxin KC (CXCL1), antigen and traffick to draining lymph nodes (Morel et al., 2011).

Frontiers in Immunology | Immunotherapies and Vaccines May 2013 | Volume 4 | Article 114 | 4
Awate et al. Adjuvant mechanisms

At the injection site, neutrophils attract other immune cells by delivers the antigen via abortive phagocytosis. In this regard, alum
producing increased amounts of chemokines and transport anti- interacts with membrane lipids on DCs leading to lipid sorting,
gen to the draining lymph nodes (Calabro et al., 2011; Morel et al., recruitment of ITAM containing molecules Syk and PI3 activation.
2011). However, the role of neutrophils in adjuvant activity is not These events eventually lead to uptake of antigen that is adsorbed
completely clear. on alum, DC activation, and MHC class II expression (Flach et al.,
ASO4 induces transient local NFB activity and cytokine pro- 2011).
duction (Didierlaurent et al., 2009). The TLR4 agonist MPL, one The role of adjuvant-induced increased antigen presentation in
of the components of AS04, stimulated increased numbers of development of adaptive immunity has not been clearly evaluated.
DCs and monocytes in the draining lymph nodes. Likewise CpG, Hence, our knowledge is limited regarding the role of this adju-
a TLR9 agonists, signals through activation of MyD88, IRAK, vant mechanism. Recently, Ghimire et al. (2012) investigated the
and TRAF-6, leading to recruitment of transcriptional factors, impact of antigen presentation on alum adjuvanticity. In addition
which in turn up-regulates the pro-inflammatory genes and pro- to confirming the ability of alum to increase the antigen internal-
tein expression (IL-1, IL-6, IL-12, IL-18, and TNF-) within 3 h ization, the study also showed that alum plays an important role in
of injection (Klinman et al., 1996; Klaschik et al., 2009). Genes reducing the rate of degradation of internalized antigen (Ghimire
up-regulated by CpG included cytokines, cell signaling, cell move- et al., 2012). Similarly, MF59 facilitated internalization of gD2 anti-
ment, and DNA damage response genes (Klaschik et al., 2010). gen from type 2 herpes simplex virus (HSV) by recruited APCs at
One of the roles of cationic liposomes is to recruit immune cells the site of injection and increased phagocytosis in human PBMCs
and increase antigen presentation. Intra-peritoneal injection of (Dupuis et al., 1999). Antigen size seems to play an important
cationic liposome (DDA/MPL) increases influx of neutrophils, role in modulating the antigen presentation efficiency. Large lipid
monocytes, macrophages, and activated NK cells in the peritoneal vesicles end up in early endosome/phagosomes and increases anti-
cavity (Korsholm et al., 2010). Another cationic liposome CAF01 gen presentation whereas smaller vesicles rapidly localize to late
induced recruitment of monocytes to the site of injection and lysosomes leading to reduced antigen presentation (Brewer et al.,
increased trafficking of liposomes to the draining lymph nodes 2004).
(Henriksen-Lacey et al., 2010).
Therefore, adjuvants induce recruitment of various immune ACTIVATION AND MATURATION OF DCs
cells to the site of injection, some of which then traffick the Activation of DCs is essential for induction of adaptive immune
antigen to the draining lymph nodes to induce specific immune responses. Increased expression of MHC class II, activation marker
responses. However, the relationship between these recruited cells CD86, and maturation marker CD83 leads to enhanced abil-
and induction of immune responses is not very clear. Depletion ity of APCs to induce T lymphocyte activation and differen-
studies suggest that the role of recruited innate immune cells at the tiation (Coyle and Gutierrez-Ramos, 2001). Freunds complete
injection site is redundant in the generation of adaptive immune adjuvant, lipopolysaccharide (LPS), liposomes, CpG-ODN, MF59,
responses (McKee et al., 2009; Calabro et al., 2011). Interestingly, AS04, and -galactosylceramide (-GAL) have all been shown
these studies were performed by depleting single cell populations. to induce DC maturation to enhance adaptive immunity (De
Identifying the role of a specific cell population in vivo is even Smedt et al., 1996; De Becker et al., 2000; Copland et al., 2003;
more challenging due to complex environment at the injection Fujii et al., 2003; Shah et al., 2003). Intra-peritoneal injection
site. Injection of adjuvants often leads to recruitment of a variety of OVA and alum led to uptake of antigen and maturation of
of cell populations and due to high redundancy in the immune sys- DCs (Kool et al., 2008a). However, in vitro studies on human
tem, other recruited cells may compensate for the depleted single cells have shown that alum and MF59 failed to directly acti-
cell population. In this regard, mice whose specific cell popula- vate DCs but enhanced the surface expression of MHC class
tions have been depleted were shown to produce cytokines and II and co-stimulatory molecules (CD83 and CD86) on mono-
chemokines to recruit innate immune cells and activate T cells cytes, macrophages, and granulocytes that resulted in increased
(Seubert et al., 2008; Calabro et al., 2011). Further studies are T cell proliferation (Sun et al., 2003; Seubert et al., 2008). Fur-
required to investigate the detailed relationship between recruited ther, in vitro activation of DCs by alum has generated conflicting
immune cells and adjuvant activity. results. One study suggested that alum failed to induce maturation
and antigen presentation (Sun et al., 2003) where as another study
ANTIGEN PRESENTATION showed that the activation marker CD86 and antigen presenta-
Efficient antigen presentation by MHCs on APCs is important tion was increased in DCs (Sokolovska et al., 2007). The source
for the induction of adaptive immune response. It was thought of alum may have been a contributing factor in the conflicting
that many adjuvants including alum, oil-based emulsions, and results.
microparticles act by targeting antigens to APCs resulting in AS04 has been shown to induce maturation of DCs (via TLR4),
enhanced antigen presentation by MHC (Gury et al., 1996; Schi- which then trafficks to the draining lymph nodes to activate
jns and Lavelle, 2011). Alum was shown to increase antigen uptake antigen-specific T cells (Didierlaurent et al., 2009). Similarly,
by DCs and alter the magnitude and duration of antigen presen- CpG induced up-regulation of CD40, CD54, CD80, CD86, and
tation (Mannhalter et al., 1985; Morefield et al., 2005). Antigen MHC class II molecules and antigen processing and presenta-
adsorption on alum led to an increase in internalization of anti- tion in plasmacytoid DCs (pDCs) (Krieg, 2002; Kerkmann et al.,
gen (Morefield et al., 2005). Recent studies by Flach et al. (2011) 2003). A novel class of TLR-independent adjuvants, mycobacte-
have shown that alum does not enter DCs directly but rather rial cord factor trehalose-6-6-dimycolate (TDM) and TDB have

www.frontiersin.org May 2013 | Volume 4 | Article 114 | 5


Awate et al. Adjuvant mechanisms

been shown to directly activate DCs through the FcR-Syk-Card9- and a procaspase-1 (Schroder and Tschopp, 2010). Activation of
Bcl10-Malt1 pathways, and up-regulates the expression of co- NLRP3 inflammasome induces caspase-1 activation, which in turn
stimulatory molecules (Werninghaus et al., 2009). Microparticles cleaves proforms of IL-1, IL-18, and IL-33 to their bioactive
such as Poly-lactic-co-glycolic acid (PLGA) did not induce co- forms (Martinon et al., 2009). The NLRP3 inflammasome can
stimulatory molecules expression on bone marrow derived DCs be activated by various stimuli including DAMPs, environmental
(BMDCs) but enhanced antigen presentation efficiency (Sun et al., irritants such as asbestos and silica, metabolic stress, and UVB
2003). DOTAP (1,2-dioleoyl-3-trimethylammonium-propane)- irradiation (Schroder and Tschopp, 2010). Apart from danger sig-
based cationic liposomes have been shown to induce maturation of nals, inflammasomes can be activated by PAMPs such as bacterial
DCs through activation of MAPK (extracellular signal-regulated flagellin through NLRC4 activation (Zhao et al., 2011).
kinase and p38), leading to up-regulation of co-stimulatory Li et al. (2007) reported for the first time that alum-induced
molecules (Yan et al., 2007). Likewise, diC14-amidine (3- secretion of IL-1 and IL-18 was caspase-1 dependent. Subsequent
tetradecylamino-tert-butyl-N-tetradecylpropion-amidine) based in vitro studies by various groups showed that activation of NLRP3
cationic liposomes up-regulates the expression of CD80 and is required for alum-induced IL-1 and IL-18 secretion (Eisen-
CD86 on DCs through specific TLR4/MD2 ligation (Tanaka et al., barth et al., 2008; Franchi and Nez, 2008; Hornung et al., 2008;
2008). Overall, adjuvants stimulate DC maturation and enhance Kool et al., 2008a). However, LPS priming to induce pro-IL-1 in
the expression of MHC and co-stimulatory molecules, which is APCs prior to alum stimulation was a pre-requisite for secretion
required for efficient T cell activation. of IL-1. Contrary to in vitro studies, the role of inflammasomes
in the adjuvant activity of alum in vivo has yielded conflicting
ACTIVATION OF INFLAMMASOMES results. Using NLRP3, ASC and caspase-1 knockout mice, Eisen-
Innate immune cells express various pathogen-recognition recep- barth et al. (2008) showed that the NLRP3 inflammasome is a
tors (PRRs) to recognize infectious agents. In recent years, various crucial component in the adjuvant activity of alum. NLRP3, ASC,
new families of PRRs have been identified including TLRs, C-type and caspase-1 knockout mice immunized with OVA adsorbed
lectin-like receptors (CLRs), nucleotide oligomerization domain on alum, failed to induce antigen-specific antibody responses
(NOD) like receptors (NLRs), and Retinoic acid-inducible gene- (Eisenbarth et al., 2008). Another study by Kool et al. (2008a)
1 (RIG-1) like receptors (RLRs). Many immunological adjuvants showed that alum-induced lower influx of inflammatory cells in
signal via PRRs or act as ligands for innate immune receptors the peritoneal cavity of NLRP3 deficient mice. They also showed
(Table 2). In contrast to TLR agonists, particulate adjuvants are that alum-mediated activation of adaptive immune responses was
not recognized by specific PRRs but they still induce adaptive NLRP3-dependent (Kool et al., 2008a). Similar studies done by
immune responses. The danger hypothesis was first advanced by Li et al. (2008) showed that NLRP3 deficient mice injected with
Matzinger (1994), who proposed that apart from self/non-self dis- alum-adsorbed diphtheria toxoid or OVA vaccine elicited impaired
crimination against infection, danger signals from damaged cells levels of antigen-specific antibody responses. All these studies indi-
can trigger activation of the immune system. Molecules associ- cate that NLRP3 inflammasome is critical in the adjuvant activity
ated with tissue damage such as uric acid, nucleotides, adenosine of alum in vivo. In contrast, Franchi and Nez (2008) clearly
triphosphate (ATP), reactive oxygen intermediates, and cytokines showed that antigen-specific IgG production was not impaired
are released at the injection site due to tissue damage (Shi et al., in NLRP3 deficient mice following intra-peritoneal injection of
2003). These non-infectious damage signals have now been named human serum albumin (HSA) in the presence of alum. However,
damage-associated molecular patterns (DAMPs) to distinguish NLRP3 did affect alum-mediated cellular recruitment suggesting
them from pathogen-associated molecular patterns (PAMPs). that inflammasomes might play an important role in activat-
Particulate adjuvants cause local tissue damage and cell death ing innate immunity, but the contribution of inflammasomes in
at the injection site (Kool et al., 2008a). In addition, many adju- activation of adaptive immunity remains elusive. The conflicting
vants induce release of pro-inflammatory cytokines at the site of results with regard to the role of inflammasomes in adjuvant activ-
injection (Didierlaurent et al., 2009; Calabro et al., 2011; Awate ity of alum have been attributed to the differences in the nature of
et al., 2012). These damage signals trigger non-specific activation alum used in different studies, immunization protocols, and the
of the innate immune system, subsequently stimulating adaptive mouse strains used (De Gregorio et al., 2008; Marrack et al., 2009).
immunity. Recently inflammasomes have been one of the most To date, the ligand for NLRP3 has not been identified. Some
widely investigated topics due to their potential role in adjuvant theories proposed for alum-mediated activation of NLRP3 include
activity. The inflammasome belongs to the NLR family, which phagosomal destabilization and release of cathepsin B, low intra-
also includes various other receptors, such as the NODs (NOD1- cellular potassium (K+) concentrations, and generation of reac-
5), NLRPs (NLRP1-14), NLRP1 (NAIP), NLRC4 (IPAF), and the tive oxygen species (ROS) (Petrilli et al., 2007; Hornung et al.,
major histocompatibility complex II transactivator (CIITA) (Mar- 2008; Kool et al., 2008a). It was proposed that a catabolic product of
tinon et al., 2009). Compared to others, NOD-like receptor family, nucleotides, uric acid, and ATP released at the site of alum injection
pyrin-domain-containing 3 (NLRP3) is the most studied inflam- due to cell damage or necrosis act as danger signals for activation of
masome receptor in regards to adjuvant mechanisms. NLRP3, also NLRP3. Saturation of uric acid due to tissue damage forms mono-
known as cryopyrin or NALP3 (NACHT, LRR, and PYD domains- sodium ureate crystals (MSU). Phagocytosis of crystalline particles
containing protein 3), is an intra-cytoplasmic multi-protein com- such as MSU or alum results in phagosomal destabilization and
plex that consists of three components; a NLRP3 receptor, an lysosomal rupture releasing the protease cathepsin B in the cytosol
apoptosis-associated speck-like protein containing a CARD (ASC) (Hornung et al., 2008). The released cathepsin B led to activation

Frontiers in Immunology | Immunotherapies and Vaccines May 2013 | Volume 4 | Article 114 | 6
Awate et al. Adjuvant mechanisms

Table 2 | Innate immune receptors activated by vaccine adjuvants.

PRRs Adjuvants Type of immune response Reference


induced

TLRs TLR1/2 Triacyl lipopeptides Th1, Th2, CTL responses Deres et al. (1989), Schild et al. (1991)
Synthetic Pam3 Cys
TLR2/6 Diacyl lipopeptides Th1, Th2, CTL responses Moyle and Toth (2008)
Pam2 Cys
TLR2 Pam3 Cys Th1, Th2, CTL responses Deres et al. (1989), Schild et al. (1991)
TLR3 Poly I:C Both Th1 and Th2 Tamura and Sasakawa (1983), Choi et al. (2012)
TLR4 LPS, AS04 (MPL) Th1 Sasaki et al. (1997), Casella and Mitchell (2008)
TLR5 Flagellin Th1 and Th2 Didierlaurent et al. (2004), McCarron and Reen (2009)
TLR 7 Imiquimod Th1, CD8+ T cell, CTL responses Wagner et al. (1999), Stanley (2002)
Resiquimod
TLR8 Resiquimod Th1, CD8+ T cell, CTL responses Wagner et al. (1999), Wu et al. (2004)
TLR9 CpG-ODN Th1, CD8+ T cells, CTL responses Kobayashi et al. (1999)

NLRs NOD1/NLRC1 DAP Th1, Th2, Th17 Chamaillard et al. (2003), Girardin et al. (2003a), Fritz
et al. (2007)
NOD2/NLRC2 MDP Th1, Th17 Girardin et al. (2003b), van Beelen et al. (2007), Shaw
et al. (2009)
NLRP1 Toxoids, MDP Th1 Hsu et al. (2008)
NLRP3 Alum, MDP, ATP Th2 Mariathasan et al. (2006), Li et al. (2007), Eisenbarth
et al. (2008)
IPAF/NLRC4 Flagellin Th1 and Th2 Lightfield et al. (2011), Zhao et al. (2011)
NAIP5 Flagellin Th1 and Th2 Kofoed and Vance (2011)

RLRs RIG-1 DNA vectors Th1, CD8+ T cells Luke et al. (2011)
MDA5 Poly I:C Th1, CD8+ T cells Wang et al. (2010)

CLRs Dectin-1 Flagellin, -glucan/zymosan Th17 LeibundGut-Landmann et al. (2007)


Mincle CAF01 Th1, Th17 CD8+ T cells Gram et al. (2009), Rosenkrands et al. (2011)

Pam3Cys, tri-palmitoyl-S-glyceryl cysteine; LPS, lipopolysaccharide; AS04, adjuvant system 04; MPL, monophosphoryl lipid A; CpG-ODN, cytidine-phosphate-guanosine
oligodeoxynucleotides; Poly I:C, polyinosinic-polycytidylic acid; DAP, diaminopimelic acid; MDP, muramyl dipeptide; CAF01, cationic adjuvant formulation-01; TLR, toll-
like receptor; NLR, NOD-like receptors; RLR, RIG-1 like receptors; CLR, C-type lectins; NOD, nucleotide-binding oligomerization domain; NLRP3, NOD-like receptor
family, pyrin-domain-containing 3; IPAF, IL-1-converting enzyme protease-activating factor; NAIP, neuronal apoptosis inhibitory protein; RIG-1, retinoic acid-inducible
gene-1; MDA5, melanoma differentiation associated gene 5.

of NLRP3 and secretion of pro-inflammatory cytokines IL-1 and showed that MPL failed to induce intra-cytoplasmic assembly of
IL-18. Treatment of mice using uricase, a uric acid degrading NLRP3 inflammasome leading to failure of caspase-1 activation
enzyme, led to reduced cellular recruitment to draining lymph and maturation of pro-inflammatory cytokines IL-1 and IL-18.
nodes in mice injected with alum (Kool et al., 2008a). Similarly,
ATP released by the damaged cells at the injection site has been CONCLUSION
shown to indirectly activate NLRP3. Extracellular ATP triggered The ultimate goal of vaccination is to generate protection against
stimulation of purinergic P2X7 receptor, resulting in activation of disease causing pathogens. Protective immunity against different
cation channel for K+ efflux and opening of pannexin-1 pore for pathogens requires different immune responses that can be gener-
entry of danger signals generated by alum, activate NLRP3 and ated by using appropriate vaccine adjuvants. Therefore, a detailed
subsequently caspase-1 (Solle et al., 2001; Petrilli et al., 2007). Fur- knowledge of the mechanisms of action of adjuvants is very
ther, blocking ROS using chemical scavengers abolished NLRP3 important in the rational design of vaccines. In recent years, con-
activation in response to MSU suggesting a link between NLRP3 siderable advances have been made in understanding the mecha-
activation and ROS generation (Dostert et al., 2008). nisms of action of various adjuvants, particularly the activation of
Recently, the role of the inflammasome in adjuvant activity of innate immunity via various mechanisms (Table 1). The future of
MF59 was evaluated (Ellebedy et al., 2011; Seubert et al., 2011). vaccine adjuvant research is heading toward developing novel
Two independent studies using NLRP3 deficient mice demon- combination adjuvants that consist primarily of PRRs agonists
strated that NLRP3 is not required for the adjuvant activity of and particulate adjuvants. While combining different adjuvants
MF59. However, an adaptor molecule required for the assembly of results in potent formulation that can enhance the quality and
inflammasome, ASC was found to be crucial for MF59 adjuvant quantity of immune response against vaccine antigens, adjuvant
activity (Ellebedy et al., 2011). A recent study by Embry et al. (2011) combinations may also have more complex mechanisms of action.

www.frontiersin.org May 2013 | Volume 4 | Article 114 | 7


Awate et al. Adjuvant mechanisms

Safety is a major concern when it comes to adjuvant approval need to identify and develop good mucosal adjuvants. In the com-
for human use. Detailed understanding of the mechanisms of ing years, we hope to learn more details of the various mechanisms
action of adjuvants will provide some insight into their safety. of action of adjuvants, which will be valuable in rational vaccine
In addition, since all of the adjuvants approved and currently in design and hopefully lead to approval of new adjuvants for use in
clinical trials are in vaccines administered by injection, there is a vaccines for humans.

REFERENCES of antigen to human dendritic cells. MF59 induces macrophage traffick- coadministered protein. J. Exp. Med.
Awate, S., Wilson, H. L., Lai, K., Babiuk, Vaccine 21, 883890. ing and apoptosis. Eur. J. Immunol. 198, 267279.
L. A., and Mutwiri, G. (2012). Activa- Cox, J. C., and Coulter, A. R. 31, 29102918. Gavin, A. L., Hoebe, K., Duong, B.,
tion of adjuvant core response genes (1997). Adjuvants-a classification Dupuis, M., McDonald, D. M., and Ott, Ota, T., Martin, C., Beutler, B., et
by the novel adjuvant PCEP. Mol. and review of their modes of action. G. (1999). Distribution of adjuvant al. (2006). Adjuvant-enhanced anti-
Immunol. 51, 292303. Vaccine 15, 248256. MF59 and antigen gD2 after intra- body responses in the absence of toll-
Brewer, J. M., Pollock, K. G. J., Tetley, Coyle, A. J., and Gutierrez-Ramos, muscular injection in mice. Vaccine like receptor signaling. Science 314,
L., and Russell, D. G. (2004). Vesicle J.-C. (2001). The expanding B7 18, 434439. 19361938.
size influences the trafficking, pro- superfamily: increasing complexity Eisenbarth, S. C., Colegio, O. R., Ghimire, T. R., Benson, R. A., Garside,
cessing, and presentation of antigens in costimulatory signals regulating OConnor, W., Sutterwala, F. S., and P., and Brewer, J. M. (2012). Alum
in lipid vesicles. J. Immunol. 173, T cell function. Nat. Immunol. 2, Flavell, R. A. (2008). Crucial role increases antigen uptake, reduces
61436150. 203209. for the Nalp3 inflammasome in the antigen degradation and sustains
Bungener, L., Idema, J., ter Veer, W., De Becker, G. V., Moulin, V. R., immunostimulatory properties of antigen presentation by DCs in vitro.
Huckriede, A., Daemen, T., and Pajak, B., Bruck, C., Francotte, M., aluminium adjuvants. Nature 453, Immunol. Lett. 147, 5562.
Wilschut, J. (2002a). Virosomes Thiriart, C., et al. (2000). The 11221126. Girardin, S. E., Boneca, I. G., Carneiro,
in vaccine development: induction adjuvant monophosphoryl lipid A Ellebedy, A. H., Lupfer, C., Ghoneim, L. A. M., Antignac, A., Jhanno,
of cytotoxic T lymphocyte activ- increases the function of antigen- H. E., DeBeauchamp, J., Kanneganti, M., Viala, J., et al. (2003a). Nod1
ity with virosome-encapsulated pro- presenting cells. Int. Immunol. 12, T.-D., and Webby, R. J. (2011). detects a unique muropeptide from
tein antigens. J. Liposome Res. 12, 807815. Inflammasome-independent role of gram-negative bacterial peptidogly-
155163. De Gregorio, E., Tritto, E., and Rap- the apoptosis-associated speck-like can. Science 300, 15841587.
Bungener, L., Serre, K., Bijl, L., Leser- puoli, R. (2008). Alum adjuvantic- protein containing CARD (ASC) Girardin, S. E., Boneca, I. G., Viala,
man, L., Wilschut, J., Daemen, T., ity: unraveling a century old mystery. in the adjuvant effect of MF59. J. R. M., Chamaillard, M., Labigne,
et al. (2002b). Virosome-mediated Eur. J. Immunol. 38, 20682071. Proc. Natl. Acad. Sci. U.S.A. 108, A. S., Thomas, G., et al. (2003b).
delivery of protein antigens to den- De Smedt, T., Pajak, B., Muraille, E., 29272932. Nod2 is a general sensor of peptido-
dritic cells. Vaccine 20, 22872295. Lespagnard, L., Heinen, E., De Baet- Embry, C. A., Franchi, L., Nunez, G., and glycan through muramyl dipeptide
Burrell, L. S., White, J. L., and Hem, S. selier, P., et al. (1996). Regulation of Mitchell, T. C. (2011). Mechanism (MDP) detection. J. Biol. Chem. 278,
L. (2000). Stability of aluminium- dendritic cell numbers and matura- of impaired NLRP3 inflammasome 88698872.
containing adjuvants during aging tion by lipopolysaccharide in vivo. J. priming by monophosphoryl lipid Glenny, A. T., Pope, C. G., Waddington,
at room temperature. Vaccine 18, Exp. Med. 184, 14131424. A. Sci. Signal. 4, ra28. H., and Wallace, V. (1926). The anti-
21882192. Deres, K., Schild, H., Wiesmuller, K.- Flach, T. L., Ng, G., Hari, A., Desrosiers, genic value of toxoid precipitated by
Calabro, S., Tortoli, M., Baudner, B. C., H., Jung, G., and Rammensee, H.-G. M. D., Zhang, P., Ward, S. M., et al. potassium-alum. J. Pathol. Bacteriol.
Pacitto, A., Cortese, M., OHagan, (1989). In vivo priming of virus- (2011). Alum interaction with den- 29, 3845.
D. T., et al. (2011). Vaccine adju- specific cytotoxic T lymphocytes dritic cell membrane lipids is essen- Glck, R., Mischler, R., Brantschen, S.,
vants alum and MF59 induce rapid with synthetic lipopeptide vaccine. tial for its adjuvanticity. Nat. Med. Just, M., Althaus, B., and Cryz, S. J.
recruitment of neutrophils and Nature 342, 561564. 17, 479487. (1992). Immunopotentiating recon-
monocytes that participate in anti- Didierlaurent, A., Ferrero, I., Otten, Franchi, L., and Nez, G. (2008). The stituted influenza virus virosome
gen transport to draining lymph L. A., Dubois, B., Reinhardt, M., Nlrp3 inflammasome is critical for vaccine delivery system for immu-
nodes. Vaccine 29, 18121823. Carlsen, H., et al. (2004). Flagellin aluminium hydroxide-mediated IL- nization against hepatitis A. J. Clin.
Casella, C., and Mitchell, T. (2008). promotes myeloid differentiation 1beta secretion but dispensable for Invest. 90, 24912495.
Putting endotoxin to work for us: factor 88-dependent development of adjuvant activity. Eur. J. Immunol. Goto, N., and Akama, K. (1982).
monophosphoryl lipid A as a safe Th2-type response. J. Immunol. 172, 38, 20852089. Histopathological studies of
and effective vaccine adjuvant. Cell. 69226930. Fraser, C. K., Diener, K. R., Brown, M. P., reactions in mice injected with
Mol. Life Sci. 65, 32313240. Didierlaurent, A. M., Morel, S., Lock- and Hayball, J. D. (2007). Improving aluminum-adsorbed tetanus toxoid.
Chamaillard, M., Hashimoto, M., Horie, man, L., Giannini, S. L., Bisteau, M., vaccines by incorporating immuno- Microbiol. Immunol. 26, 11211132.
Y., Masumoto, J., Qiu, S., Saab, Carlsen, H., et al. (2009). AS04, an logical coadjuvants. Expert Rev. Vac- Goto, N., Kato, H., Maeyama, J.-I.,
L., et al. (2003). An essential role aluminum salt- and TLR4 agonist- cines 6, 559578. Shibano, M., Saito, T., Yamaguchi,
for NOD1 in host recognition of based adjuvant system, induces a Fritz, J. H., Le Bourhis, L., Sellge, G., J., et al. (1997). Local tissue irri-
bacterial peptidoglycan containing transient localized innate immune Magalhaes, J. G., Fsihi, H., Kufer, tating effects and adjuvant activ-
diaminopimelic acid. Nat. Immunol. response leading to enhanced adap- T. A., et al. (2007). Nod1-mediated ities of calcium phosphate and
4, 702707. tive immunity. J. Immunol. 183, innate immune recognition of pep- aluminium hydroxide with differ-
Choi, J. P., Kim, Y. S., Kim, O. Y., 61866197. tidoglycan contributes to the onset ent physical properties. Vaccine 15,
Kim, Y. M., Jeon, S. G., Roh, T. Dostert, C., Ptrilli, V., Van Bruggen, of adaptive immunity. Immunity 26, 13641371.
Y., et al. (2012). TNF-alpha is a R., Steele, C., Mossman, B. T., and 445459. Gram, G. J., Karlsson, I., Agger, E.
key mediator in the development Tschopp, J. (2008). Innate immune Fujii, S.-I., Shimizu, K., Smith, C., M., Andersen, P., and Fomsgaard,
of Th2 cell response to inhaled activation through Nalp3 inflamma- Bonifaz, L., and Steinman, R. M. A. (2009). A novel liposome-based
allergens induced by a viral PAMP some sensing of asbestos and silica. (2003). Activation of natural killer adjuvant CAF01 for induction of
double-stranded RNA. Allergy 67, Science 320, 674677. T cells by -Galactosylceramide CD8(+) cytotoxic T-lymphocytes
11381148. Dupuis, M., Denis-Mize, K., LaBar- rapidly induces the full maturation (CTL) to HIV-1 minimal CTL
Copland, M. J., Baird, M. A., Rades, bara, A., Peters, W., Charo, I. F., of dendritic cells in vivo and thereby peptides in HLA-A0201 trans-
T., McKenzie, J. L., Becker, B., Reck, McDonald, D. M., et al. (2001). acts as an adjuvant for combined genic mice. PLoS ONE 4:e6950.
F., et al. (2003). Liposomal delivery Immunization with the adjuvant CD4 and CD8 T cell immunity to a doi:10.1371/journal.pone.0006950

Frontiers in Immunology | Immunotherapies and Vaccines May 2013 | Volume 4 | Article 114 | 8
Awate et al. Adjuvant mechanisms

Grun, J. L., and Maurer, P. H. (1989). alum-induced myeloid cell popula- the site of injection. Immunology McCarron, M., and Reen, D. J. (2009).
Different T helper cell subsets tion. Science 304, 18081810. 129, 7586. Activated human neonatal CD8+
elicited in mice utilizing two dif- Kerkmann, M., Rothenfusser, S., Hor- Kreuter, J. (1988). Possibilities of using T cells are subject to immunomod-
ferent adjuvant vehicles: the role of nung, V., Towarowski, A., Wag- nanoparticles as carriers for drugs ulation by direct TLR2 or TLR5
endogenous interleukin 1 in pro- ner, M., Sarris, A., et al. (2003). and vaccines. J. Microencapsul. 5, stimulation. J. Immunol. 182,
liferative responses. Cell. Immunol. Activation with CpG-A and CpG- 115127. 5562.
121, 134145. B oligonucleotides reveals two dis- Krieg, A. M. (2002). CpG motifs in McKee, A. S., Munks, M. W., MacLeod,
Gury, J. C., Ria, F., and Adorini, L. tinct regulatory pathways of type I bacterial DNA and their immune M. K. L., Fleenor, C. J., Van Rooijen,
(1996). Dendritic cells but not B cells IFN synthesis in human plasmacy- effects. Annu. Rev. Immunol. 20, N., Kappler, J. W., et al. (2009). Alum
present antigenic complexes to class toid dendritic cells. J. Immunol. 170, 709760. induces innate immune responses
II-restricted T cells after administra- 44654474. LeibundGut-Landmann, S., Grosz, O., through macrophage and mast cell
tion of protein in adjuvant. J. Exp. Khoshnejad, M., Young, P. R., Toth, I., Robinson, M. J., Osorio, F., Slack, sensors, but these sensors are not
Med. 183, 751757. and Minchin, R. F. (2007). Mod- E. C., Tsoni, S. V., et al. (2007). required for alum to act as an
Guy, B. (2007). The perfect mix: recent ified influenza virosomes: recent Syk- and CARD9-dependent cou- adjuvant for specific immunity. J.
progress in adjuvant research. Nat. advances and potential in gene pling of innate immunity to the Immunol. 183, 44034414.
Rev. Microbiol. 5, 505517. delivery. Curr. Med. Chem. 14, induction of T helper cells that pro- Morefield, G. L., Sokolovska, A., Jiang,
Henriksen-Lacey, M., Bramwell, V. 31523156. duce interleukin 17. Nat. Immunol. D., HogenEsch, H., Robinson, J.
W., Christensen, D., Agger, E.-M., Klaschik, S., Tross, D., and Klinman, 8, 630638. P., and Hem, S. L. (2005). Role
Andersen, P., and Perrie, Y. (2010). D. M. (2009). Inductive and sup- Li, H., Nookala, S., and Re, F. of aluminum-containing adjuvants
Liposomes based on dimethyldioc- pressive networks regulate TLR9- (2007). Aluminum hydroxide adju- in antigen internalization by den-
tadecylammonium promote a depot dependent gene expression in vivo. vants activate caspase-1 and induce dritic cells in vitro. Vaccine 23,
effect and enhance immunogenicity J. Leukoc. Biol. 85, 788795. IL-1 and IL-18 release. J. Immunol. 15881595.
of soluble antigen. J. Control Release. Klaschik, S., Tross, D., Shirota, H., 178, 52715276. Morein, B., and Bengtsson, K. L.
142, 180186. and Klinman, D. M. (2010). Short- Li, H., Willingham, S. B., Ting, J. P.- V. (1999). Immunomodulation by
Herbert, W. J. (1968). The mode of and long-term changes in gene Y., and Re, F. (2008). Cutting edge: iscoms, immune stimulating com-
action of mineral-oil emulsion adju- expression mediated by the activa- inflammasome activation by alum plexes. Methods 19, 94102.
vants on antibody production in tion of TLR9. Mol. Immunol. 47, and alums adjuvant effect are medi- Morel, S., Didierlaurent, A., Bour-
mice. Immunology 14, 301318. 13171324. ated by NLRP3. J. Immunol. 181, guignon, P., Delhaye, S., Baras, B.,
Hoebe, K., Janssen, E., and Beutler, Klinman, D. M., Yi, A. K., Beaucage, 1721. Jacob, V., et al. (2011). Adjuvant sys-
B. (2004). The interface between S. L., Conover, J., and Krieg, A. M. Lightfield, K. L., Persson, J., Trinidad, tem AS03 containing -tocopherol
innate and adaptive immunity. Nat. (1996). CpG motifs present in bac- N. J., Brubaker, S. W., Kofoed, E. M., modulates innate immune response
Immunol. 5, 971974. teria DNA rapidly induce lympho- Sauer, J.-D., et al. (2011). Differential and leads to improved adaptive
Hornung, V., Bauernfeind, F., Halle, A., cytes to secrete interleukin 6, inter- requirements for NAIP5 in activa- immunity. Vaccine 29, 24612473.
Samstad, E. O., Kono, H., Rock, K. leukin 12, and interferon gamma. tion of the NLRC4 inflammasome. Mosca, F., Tritto, E., Muzzi, A., Monaci,
L., et al. (2008). Silica crystals and Proc. Natl. Acad. Sci. U.S.A. 93, Infect. Immun. 79, 16061614. E., Bagnoli, F., Iavarone, C., et al.
aluminum salts activate the NALP3 28792883. Luke, J. M., Simon, G. G., Soderholm, (2008). Molecular and cellular sig-
inflammasome through phagosomal Kobayashi, H., Horner, A. A., Tak- J., Errett, J. S., August, J. T., Gale, natures of human vaccine adjuvants.
destabilization. Nat. Immunol. 9, abayashi, K., Nguyen, M.-D., Huang, M., et al. (2011). Coexpressed RIG-I Proc. Natl. Acad. Sci. U.S.A. 105,
847856. E., Cinman, N., et al. (1999). agonist enhances humoral immune 1050110506.
Hsu, L.-C., Ali, S. R., McGillivray, Immunostimulatory DNA preprim- response to influenza virus DNA Moyle, P. M., and Toth, I. (2008). Self-
S., Tseng, P.-H., Mariathasan, S., ing: a novel approach for pro- vaccine. J. Virol. 85, 13701383. adjuvanting lipopeptide vaccines.
Humke, E. W., et al. (2008). A longed Th1-biased immunity. Cell. Mannhalter, J. W., Neychev, H. O., Curr. Med. Chem. 15, 506516.
NOD2-NALP1 complex mediates Immunol. 198, 6975. Zlabinger, G. J., Ahmad, R., and Osebold, J. W. (1982). Mechanisms of
caspase-1-dependent IL-1 secre- Kofoed, E. M., and Vance, R. E. (2011). Eibl, M. M. (1985). Modulation of action by immunologic adjuvants. J.
tion in response to Bacillus anthracis Innate immune recognition of bac- the human immune response by the Am. Vet. Med. Assoc. 181, 983987.
infection and muramyl dipeptide. terial ligands by NAIPs determines non-toxic and non-pyrogenic adju- Ott, G., Barchfeld, G. L., Chernoff, D.,
Proc. Natl. Acad. Sci. U.S.A. 105, inflammasome specificity. Nature vant aluminium hydroxide: effect Radhakrishnan, R., van Hoogevest,
78037808. 477, 592595. on antigen uptake and antigen pre- P., and Van Nest, G. (1995). MF59.
Hutchison, S., Benson, R. A., Gibson, V. Kool, M., Ptrilli, V., De Smedt, T., sentation. Clin. Exp. Immunol. 61, Design and evaluation of a safe and
B., Pollock, A. H., Garside, P., and Rolaz, A., Hammad, H., van Nimwe- 143151. potent adjuvant for human vaccines.
Brewer, J. M. (2012). Antigen depot gen, M., et al. (2008a). Cutting edge: Mariathasan, S., Weiss, D. S., Newton, Pharm. Biotechnol. 6, 277296.
is not required for alum adjuvantic- alum adjuvant stimulates inflamma- K., McBride, J., ORourke, K., Roose- Pashine, A., Valiante, N. M., and Ulmer,
ity. FASEB J. 26, 12721279. tory dendritic cells through activa- Girma, M., et al. (2006). Cryopy- J. B. (2005). Targeting the innate
Iyer, S., HogenEsch, H., and Hem, S. tion of the NALP3 inflammasome. J. rin activates the inflammasome in immune response with improved
L. (2003). Relationship between Immunol. 181, 37553759. response to toxins and ATP. Nature vaccine adjuvants. Nat. Med. 11,
the degree of antigen adsorp- Kool, M., Soulli, T., van Nimwegen, M., 440, 228232. 6368.
tion to aluminum hydroxide Willart, M. A. M., Muskens, F., Jung, Marrack, P., McKee, A. S., and Munks, Petrilli, V., Papin, S., Dostert, C., Mayor,
adjuvant in interstitial fluid and S., et al. (2008b). Alum adjuvant M. W. (2009). Towards an under- A., Martinon, F., and Tschopp, J.
antibody production. Vaccine 21, boosts adaptive immunity by induc- standing of the adjuvant action of (2007). Activation of the NALP3
12191223. ing uric acid and activating inflam- aluminium. Nat. Rev. Immunol. 9, inflammasome is triggered by
Janeway, C. A. (1989). Approaching the matory dendritic cells. J. Exp. Med. 287293. low intracellular potassium con-
asymptote? Evolution and revolu- 205, 869882. Martinon, F., Mayor, A., and Tschopp, centration. Cell Death Differ. 14,
tion in immunology. Cold Spring Korsholm, K. S., Petersen, R. V., Agger, J. (2009). The inflammasomes: 15831589.
Harb. Symp. Quant. Biol. 54, 113. E. M., and Andersen, P. (2010). T- guardians of the body. Annu. Rev. Ramanathan, V. D., Badenoch-Jones, P.,
Jordan, M. B., Mills, D. M., Kap- helper 1 and T-helper 2 adjuvants Immunol. 27, 229265. and Turk, J. L. (1979). Complement
pler, J., Marrack, P., and Cam- induce distinct differences in the Matzinger, P. (1994). Tolerance, danger, activation by aluminium and zirco-
bier, J. C. (2004). Promotion of magnitude, quality and kinetics of and the extended family. Annu. Rev. nium compounds. Immunology 37,
B cell immune responses via an the early inflammatory response at Immunol. 12, 9911045. 881888.

www.frontiersin.org May 2013 | Volume 4 | Article 114 | 9


Awate et al. Adjuvant mechanisms

Ramon, G. (1924). Sur la toxine et sur Seubert, A., Calabro, S., Santini, L., Galli, cytokine production in mice lacking Werninghaus, K., Babiak, A., Gro, O.,
Ianatoxine diphtheriques. Ann. Inst. B., Genovese, A., Valentini, S., et al. P27receptors. J. Biol. Chem. 276, Hlscher, C., Dietrich, H., Agger,
Pasteur 38, 110. (2011). Adjuvanticity of the oil-in- 125132. E. M., et al. (2009). Adjuvantic-
Rimaniol, A.-C. C., Gras, G., Verdier, water emulsion MF59 is indepen- Stanley, M. A. (2002). Imiquimod and ity of a synthetic cord factor ana-
F. O., Capel, F., Grigoriev, V. dent of Nlrp3 inflammasome but the imidazoquinolines: mechanism logue for subunit Mycobacterium
B., Porcheray, F., et al. (2004). requires the adaptor protein MyD88. of action and therapeutic potential. tuberculosis vaccination requires
Aluminum hydroxide adjuvant Proc. Natl. Acad. Sci. U.S.A. 108, Clin. Exp. Dermatol. 27, 571577. FcR-Syk-Card9-dependent innate
induces macrophage differentiation 1116911174. Sun, H., Pollock, K. G. J., and Brewer, immune activation. J. Exp. Med. 206,
towards a specialized antigen- Seubert, A., Monaci, E., Pizza, M., J. M. (2003). Analysis of the role 8997.
presenting cell type. Vaccine 22, OHagan, D. T., and Wack, A. (2008). of vaccine adjuvants in modulating Wu, J. J., Huang, D. B., and Tyring, S. K.
31273135. The adjuvants aluminum hydrox- dendritic cell activation and anti- (2004). Resiquimod: a new immune
Rosenkrands, I., Vingsbo-Lundberg, C., ide and MF59 induce monocyte and gen presentation in vitro. Vaccine 21, response modifier with potential as
Bundgaard, T. J., Lindenstrm, T., granulocyte chemoattractants and 849855. a vaccine adjuvant for Th1 immune
Enouf, V., van der Werf, S., et enhance monocyte differentiation Tamura, M., and Sasakawa, S. (1983). responses. Antiviral Res. 64, 7983.
al. (2011). Enhanced humoral and toward dendritic cells. J. Immunol. Induction of human leukocyte inter- Yan, W., Chen, W., and Huang, L. (2007).
cell-mediated immune responses 180, 54025412. feron by heat-treated poly I: poly Mechanism of adjuvant activity of
after immunization with trivalent Shah, J. A., Darrah, P. A., Ambrozak, C. Biochem. Biophys. Res. Commun. cationic liposome: phosphorylation
influenza vaccine adjuvanted with D. R., Turon, T. N., Mendez, S., Kir- 110, 851858. of a MAP kinase, ERK and induc-
cationic liposomes. Vaccine 29, man, J., et al. (2003). Dendritic cells Tanaka, T., Legat, A., Adam, E., Steuve, J., tion of chemokines. Mol. Immunol.
62836291. are responsible for the capacity of Gatot, J.-S., Vandenbranden, M., et 44, 36723681.
Sasaki, S., Tsuji, T., Hamajima, K., CpG oligodeoxynucleotides to act as al. (2008). DiC14-amidine cationic Zhao, Y., Yang, J., Shi, J., Gong, Y.-N., Lu,
Fukushima, J., Ishii, N., Kaneko, an adjuvant for protective vaccine liposomes stimulate myeloid den- Q., Xu, H., et al. (2011). The NLRC4
T., et al. (1997). Monophosphoryl immunity against Leishmania major dritic cells through Toll-like receptor inflammasome receptors for bacte-
lipid A enhances both humoral and in mice. J. Exp. Med. 198, 281291. 4. Eur. J. Immunol. 38, 13511357. rial flagellin and type III secretion
cell-mediated immune responses to Shaw, M. H., Reimer, T., Sanchez- van Beelen, A. J., Zelinkova, Z., apparatus. Nature 477, 596600.
DNA vaccination against human Valdepenas, C., Warner, N., Kim, Taanman-Kueter, E. W., Muller, F.
immunodeficiency virus type 1. Y.-G., Fresno, M., et al. (2009). T J., Hommes, D. W., Zaat, S. A. J., Conflict of Interest Statement: The
Infect. Immun. 65, 35203528. cell-intrinsic role of Nod2 in pro- et al. (2007). Stimulation of the authors declare that the research was
Schijns, V. E. J. C. (2000). Immuno- moting type 1 immunity to Tox- intracellular bacterial sensor NOD2 conducted in the absence of any com-
logical concepts of vaccine adjuvant oplasma gondii. Nat. Immunol. 10, programs dendritic cells to pro- mercial or financial relationships that
activity: commentary. Curr. Opin. 12671274. mote interleukin-17 production in could be construed as a potential con-
Immunol. 12, 456463. Shi, Y., Evans, J. E., and Rock, K. L. human memory T cells. Immunity flict of interest.
Schijns, V. E. J. C., and Lavelle, E. C. (2003). Molecular identification of a 27, 660669.
(2011). Trends in vaccine adjuvants. danger signal that alerts the immune Wagner, T. L., Ahonen, C. L., Couture, A. Received: 20 March 2013; paper pend-
Expert Rev. Vaccines 10, 539550. system to dying cells. Nature 425, M., Gibson, S. J., Miller, R. L., Smith, ing published: 14 April 2013; accepted:
Schild, H., Deres, K., Wiesmller, K.- 516521. R. M., et al. (1999). Modulation of 29 April 2013; published online: 16 May
H., Jung, G., and Rammensee, H.-G. Singh, M., and OHagan, D. T. (2003). TH1 and TH2 cytokine production 2013.
(1991). Efficiency of peptides and Recent advances in veterinary vac- with the immune response modi- Citation: Awate S, Babiuk LA and
lipopeptides for in vivo priming of cine adjuvants. Int. J. Parasitol. 33, fiers, R-848 and imiquimod. Cell. Mutwiri G (2013) Mechanisms of action
virus-specific cytotoxic T cells. Eur. 469478. Immunol. 191, 1019. of adjuvants. Front. Immunol. 4:114. doi:
J. Immunol. 21, 26492654. Siskind, G. W., and Benacerraf, B. Wang, H.-B., and Weller, P. F. (2008). 10.3389/fimmu.2013.00114
Schroder, K., and Tschopp, J. (2010). (1969). Cell selection by antigen Pivotal advance: eosinophils medi- This article was submitted to Frontiers
The inflammasomes. Cell 140, in the immune response. Adv. ate early alum adjuvant-elicited B in Immunotherapies and Vaccines, a spe-
821832. Immunol. 10, 150. cell priming and IgM production. J. cialty of Frontiers in Immunology.
Serre, K., Mohr, E., Toellner, K. M., Sokolovska, A., Hem, S. L., and Leukoc. Biol. 83, 817821. Copyright 2013 Awate, Babiuk and
Cunningham, A. F., Granjeaud, S., HogenEsch, H. (2007). Activation Wang, Y., Cella, M., Gilfillan, S., and Mutwiri. This is an open-access arti-
Bird, R., et al. (2008). Molecular of dendritic cells and induction Colonna, M. (2010). Cutting edge: cle distributed under the terms of the
differences between the divergent of CD4+ T cell differentiation polyinosinic:polycytidylic acid Creative Commons Attribution License,
responses of ovalbumin-specific by aluminum-containing adjuvants. boosts the generation of memory which permits use, distribution and
CD4 T cells to alum-precipitated Vaccine 25, 45754585. CD8 T cells through melanoma reproduction in other forums, provided
ovalbumin compared to ovalbumin Solle, M., Labasi, J., Perregaux, D. G., differentiation-associated protein the original authors and source are cred-
expressed by Salmonella. Mol. Stam, E., Petrushova, N., Koller, 5 expressed in stromal cells. J. ited and subject to any copyright notices
Immunol. 45, 35583566. B. H., et al. (2001). Altered Immunol. 184, 27512755. concerning any third-party graphics etc.

Frontiers in Immunology | Immunotherapies and Vaccines May 2013 | Volume 4 | Article 114 | 10

Das könnte Ihnen auch gefallen