53
Designing and building the next generation of improved vaccine adjuvants Luis A. Brito, Derek T. O’Hagan PII: S0168-3659(14)00426-X DOI: doi: 10.1016/j.jconrel.2014.06.027 Reference: COREL 7261 To appear in: Journal of Controlled Release Received date: 11 March 2014 Revised date: 17 June 2014 Accepted date: 18 June 2014 Please cite this article as: Luis A. Brito, Derek T. O’Hagan, Designing and building the next generation of improved vaccine adjuvants, Journal of Controlled Release (2014), doi: 10.1016/j.jconrel.2014.06.027 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Designing and building the next generation of improved vaccine adjuvants

  • Upload
    derek-t

  • View
    218

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Designing and building the next generation of improved vaccine adjuvants

�������� ����� ��

Designing and building the next generation of improved vaccine adjuvants

Luis A. Brito, Derek T. O’Hagan

PII: S0168-3659(14)00426-XDOI: doi: 10.1016/j.jconrel.2014.06.027Reference: COREL 7261

To appear in: Journal of Controlled Release

Received date: 11 March 2014Revised date: 17 June 2014Accepted date: 18 June 2014

Please cite this article as: Luis A. Brito, Derek T. O’Hagan, Designing and building thenext generation of improved vaccine adjuvants, Journal of Controlled Release (2014), doi:10.1016/j.jconrel.2014.06.027

This is a PDF file of an unedited manuscript that has been accepted for publication.As a service to our customers we are providing this early version of the manuscript.The manuscript will undergo copyediting, typesetting, and review of the resulting proofbefore it is published in its final form. Please note that during the production processerrors may be discovered which could affect the content, and all legal disclaimers thatapply to the journal pertain.

Page 2: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

Designing and building the next generation of improved vaccine adjuvants

Luis A. Brito1 and Derek T. O’Hagan1, 2

1Novartis Vaccines and Diagnostics, Cambridge, MA USA

2Corresponding author:

Derek T. O’Hagan

350 Massachusetts Ave.

Cambridge, MA 02139

[email protected]

Running title: Next generation of improved vaccine adjuvants

Page 3: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

Abstract

Vaccine adjuvants interact with the immune system, to increase the potency of vaccine antigens. Many

of the adjuvants currently available were developed with little understanding of how they worked.

Highly pure recombinant antigens are typically very poorly immunogenic due to a lack of exogenous

immune activating components such as nucleic acids, lipids, and cell membrane components. In this

review we discuss the role of adjuvants and their role as ‘delivery systems’ or ‘immune potentiators’.

We also highlight the need for appropriate delivery of immune potentiators with several ‘delivery

system’ adjuvants such as alum, emulsions, liposomes, and polymeric particles. The challenges faced by

vaccinologists to create the next generation of vaccines can be solved in-part by developing a greater

understanding of the impact of delivery, and an appreciation of the key role of pharmaceutical sciences.

Page 4: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

Introduction – what are vaccine adjuvants?

Vaccine adjuvants are used to enhance the potency of vaccine antigens. Although most of the adjuvants

currently available were developed empirically, without a real understanding of how they worked, this is

no longer an acceptable approach. Increasingly, modern vaccines are being developed based on

rationally designed recombinant highly purified antigens through structure based design, epitope

focusing or genomic based screening [1-4]. However, the inherent immunogenicity of these antigens is

often low in comparison to the more traditional vaccines based on live attenuated or inactivated

pathogens. Hence, there is an increasing need for potent and safe vaccine adjuvants to ensure that

modern vaccines can succeed. Moreover, there is also an increasing awareness that it is necessary to

better understand exactly how adjuvants work, how do they enhance immune responses? Nevertheless,

live attenuated or inactivated vaccines will likely continue to work effectively in the absence of

exogenously added adjuvants, since they already contain several adjuvant active components, including

non-mammalian origin nucleic acids, lipids and cell membrane components. The very practical and

pragmatic reasons why adjuvants are included in vaccines are highlighted in Table 1.

Vaccine adjuvants are defined by what they do, not what they are. The basic definition accepted by

most is that a vaccine adjuvant is something that is added to an antigen to induce a more potent

immune response. Consequently, many diverse substances have been used as adjuvants, with a very

broad range of compositions, and obtained from many sources. Nevertheless, despite the extensive

diversity of materials used, most of the established adjuvants often have more similarities than

differences. Various classifications have been proposed for adjuvants in an attempt to better understand

them, for example they are often classified into two broad groups, as ‘delivery systems’ or ‘immune

potentiators’. However recent insights into how they work, has shown that these simplistic

classifications are incorrect. In the past, we chose to define adjuvants as different ‘generations’, loosely

based on when they were first introduced and the number of components involved [5]. We believe that

this was a helpful interpretation that allowed adjuvants to be considered as more typical pharmaceutical

components of vaccines [6]. The 1st generation adjuvants are particulates of various compositions, but

similar size, including the original and the most commonly used vaccine adjuvant, insoluble aluminium

salts (alum). Most of the other established 1st generation adjuvants are formulations more familiar to

pharmaceutical scientists, since they include emulsions, liposomes and micro or nanoparticles, which

are often used for other purposes in drug delivery. For example, the first novel adjuvant to be approved

Page 5: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

after alum was an o/w emulsion formulation called MF59, which was followed by the introduction of a

new influenza vaccine based on liposomes [7] . Hence, the 1st generation of adjuvants were often called

‘antigen delivery systems’, since they were thought to function mainly by promoting the uptake of

antigens into immune cells. In this review we will discuss the ‘delivery’ aspects of 1st generation

adjuvants, highlighting key ‘pharmaceutical’ considerations and discuss how they might be improved.

There have been many technical innovations in drug delivery in recent years, including targeted systems

and controlled release. We will consider if some of these innovations might contribute to the next

generation of improved vaccine adjuvants? Although vaccine adjuvants will be increasingly required for

different types of vaccines, particularly for therapeutic vaccines, we will focus here on the use of

adjuvants in standard vaccines to protect against infectious disease. Other reviews in this journal will

consider the development of potential therapeutic vaccines and the technologies that might be

necessary to accomplish this significant challenge. Table 2 summarizes a subset of the adjuvants that

have been evaluated in man in clinical studies, which is only a very small subset of all the different

adjuvant technologies that have been described in the literature. Virus like particles (VLP’s) will not be

covered in this review, since they are not a vaccine adjuvant, they are ordered arrays of antigens that

are either expressed from cells or self-assemble into particles. It has been demonstrated repeatedly

that VLP’s induce stronger immune responses than soluble antigens alone, and this is one of the reasons

that commonly used vaccine adjuvants tend to be particulate structures. For example, a novel influenza

vaccine containing a HA fusion with ferritin that self-assembled into nanoparticles was recently

described [8]. The HA particles were found to induce broadly effective neutralizing antibodies to distant

influenza virus strains. However, even though the antigens were displayed in a particulate form as a VLP,

they still required the addition of an oil-in-water adjuvant. This finding is consistent with evaluations of

other purified recombinant antigens which also need added adjuvants, since they typically do not

contain the immunostimulatory molecules that are present in attenuated or inactivated vaccines. The

need for an added adjuvant is necessary for many different VLPs, and even the commercial vaccines that

contain VLPs, also contain an adjuvant, including Hepatitis B virus (HBV) and human papilloma virus

(HPV) vaccines. Interestingly, one of the commercially available HPV vaccines contains a 2nd generation

adjuvant comprising both alum + MPL, also known as AS04, from Glaxo Smith kline (GSK) [9].

Not surprisingly, the most prominent 2nd generation adjuvant approaches have tended to build

on the success of the 1st generation, since they typically comprise a first generation delivery system (e.g.

alum, emulsion, liposome or microparticle), in conjunction with one or more ‘immune potentiators’.

Tables 3a and b illustrate how potentially important delivery of both the antigen and

Page 6: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

immunopotentiator is for generating potent immune responses. The mammalian immune system

comprises both an innate and an adaptive component. It is the adaptive immune system with which

most people are familiar, which is responsible for long-lasting antibody and T cell responses and

immunological memory. Most current vaccines confer protection mainly through the induction of

humoral immunity, which is mediated by antibodies [10]. The adaptive immune system is the target of

vaccines and is a necessarily diverse system, since it based on an almost countless number of potential

recognition molecules (antibodies). Because of its large repertoire of recognition molecules, the

adaptive immune system is able to discriminate between and respond to an almost infinite number of

antigens. However, the successful induction of adaptive immunity not only depends on direct antigen

recognition, but also relies on essential signals that are delivered by the innate immune system [11]. The

innate immune system is our first line of host defense against pathogens and is mediated by leukocytes

including macrophages and dendritic cells (DCs) and by the complement system. It responds

immediately following infection using a limited number of germ-line-encoded pattern-recognition

receptors (PRRs) that recognize invariant pathogen-associated molecular patterns (PAMPs) that are

present on or within the organisms that infect us. PRRs function as sensors that alert the immune

system of ‘danger’, since the host (you or I) has been infected. Innate responses are typically more rapid

than adaptive responses, with responses occurring within minutes to hours, rather than days to weeks.

However, innate responses also wane rapidly, to limit the tissue damage that can result from these

potent, nonspecific effector mechanisms. Hence, innate and adaptive immunity work together in

tandem to offer maximal protection against pathogen invasion, with the ‘nonspecific’ innate system

being activated immediately on infection, while the more complex adaptive system responds more

slowly by triggering specific antibody and T cell responses. The best known PRRs , and also the ones

most commonly exploited by adjuvants, which often operate as ‘agonists’ for the PRRS, are the Toll-like

receptors (TLRs) [12]. There are at least 10 TLRs in man, which can detect infecting organisms. Typically

phagocytic cells like macrophages and DCs display TLR’s and trigger innate immunity, but they can also

function as Antigen-Presenting Cells (APCs), to trigger adaptive immunity. The most effective APC are

DCs (typically called ‘professional APC’), which digest and process antigens for presentation on the cell

surface in conjunction with MHC molecules. APCs also transport antigens to local lymphoid nodes,

where they can be presented to naive T-cells to trigger the adaptive response. Within the cytoplasm,

there are at least two additional families of PRRs: the retinoic acid-inducible gene (RIG)-like helicases

(RLHs) and the nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), which are also

being exploited as targets for some new generation adjuvants.

Page 7: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

Traditionally, vaccinologists have focused primarily on the adaptive immune response, since it

mediates protective immunity for most vaccines. In contrast, innate immunity was considered rather

less interesting until the last 15 years or so, which has seen a huge expansion in basic knowledge. This

came as a consequence of the gradual recognition that innate immunity was the key system necessary

to control and enhance adaptive immunity. The expansion of knowledge in recent years has focused on

the recognition of the different receptor systems involved in innate immunity (TLR, RIG, NOD etc), their

activation pathways and the immune mediators released. Following the initial discovery of TLRs, it

became clear that many of the established vaccine adjuvants, including the MPL included in the licensed

AS04 adjuvant were in fact agonists for PRRs. This was a huge in advance in basic science that was

recognized by the award of a Nobel Prize in medicine to one of the key scientists involved [13, 14].

However, these insights also gave rise to an unrealistic optimism that many new adjuvants would now

be easy to discover. If an adjuvant simply needs to activate innate immunity and the receptors involved

are already known, then the tools of drug discovery could be employed and high throughput screens

could be established to identify many new compounds potentially able to function as adjuvants [15].

Nevertheless, as was already established, generation 2 adjuvants tended to be generation 1 carriers in

conjunction with an ‘immune potentiator’. Perhaps the key role of the carrier was under appreciated at

this time and the contribution it would continue to make was underappreciated. Even if it was finally

clear what the immune potentiator did in 2nd generation adjuvants, which was to activate innate

immunity, there was still a key need to ‘marry’ the immune potentiator to an appropriate and effective

‘delivery system’. This review will focus on which are the delivery systems available, what are their

relative advantages and limitations, and how best can they be exploited with new generation immune

potentiators to design the next generation of adjuvants. In addition, we will consider which advances

from the field of drug delivery might be best applied to vaccine adjuvants. Can we use new concepts to

make better vaccines and which approach is most likely to succeed in the long run?

What is ‘alum’ adjuvant?

The most commonly used vaccine adjuvants in human vaccines are insoluble salts of aluminium

(alum), which have been included in many commercial products over many decades, and have been

shown to be safe and well tolerated [16]. Aluminum adjuvants are currently used in vaccines against

diphtheria, tetanus, pertussis, hepatitis B, anthrax, Hemophilus influenzae and human papilloma virus

[16, 17]. In addition, they are also used in man for immunotherapy for allergic diseases and are used in

Page 8: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

several commercially available animal vaccines. Although alum is the ‘archetypical’ adjuvant, it differs

significantly both structurally and compositionally from the other generation 1 adjuvants and has not

found an alternative pharmaceutical use as an injectable product. Although aluminium salts were

originally used orally as human medicines and are also used topically. The use of alum as an adjuvant is

almost a historical ‘quirk’, rather than the outcome of a considered evaluation of which material was

‘right’ for the job. Alum adjuvant was originally discovered because it was a good adsorbent on which to

purify antigens. Alum is also used orally as an adsorbent for phosphates, to prevent excessive absorption

from the intestines. Although it was not necessarily ‘built for the job’, given the established history of

safety and efficacy for alum, it is clear that it will remain a key component of vaccines in the future. In

fact, as highlighted earlier, alum has already been successfully adapted to develop a 2nd generation

adjuvant that is included in a licensed vaccine product, AS04 in HPV [18]. In marked contrast to alum,

some of the other delivery systems used as adjuvants were first used for alternative purposes, before

being adapted as adjuvants, including emulsions, liposomes and microparticles (Table 4). However,

some of the more recently described 2nd generation adjuvant concepts were designed specifically for the

purpose, e.g. Iscoms, IC31, CAF etc.

The use of aluminium based adjuvants originated in 1926 with the observations of Glenny and Pope,

who showed that alum precipitated diphtheria toxoid was more potent than the antigen alone [19]. This

observation also provided the lasting ‘definition’ of an adjuvant, something that is added to a vaccine to

enhance the immune response to an antigen. Subsequent studies also showed that alum-precipitated

diphtheria and tetanus toxoids enhanced immune responses in humans [20, 21]. However, the original

approach of ‘alum-precipitated’ vaccines has now been largely replaced for commercial products by the

preparation of pre-formed alum gels, to which the antigens can be adsorbed. This approach ensures

greater uniformity and reproducibility for the alum adjuvant. Aluminium based adjuvants are typically

but incorrectly called ‘alum’, as they will be here for brevity, but we acknowledge that this is a

simplification of the complex salts actually used. The two main types of alum used in vaccines are

typically aluminium hydroxide (AH) and aluminium phosphate (AP), which each have very different

physical and chemical compositions, and very different adsorptive behavior with different antigens.

Chemically, AH is aluminium oxyhydroxide, which has a crystalline structure and exists as small (<10nm)

needle like ‘nanoparticles’, which tend to aggregate into structures a few microns in size. The

nanometer dimensions of the primary particles result in a very large surface area for adsorption of

approximately 500 m2/g [22]. In contrast, AP is aluminium hydroxyphosphate, which has an amorphous

structure by X-ray diffraction analysis and tends to exist as ‘plates’ (50nm), which form loose aggregates

Page 9: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

of several microns in size. The point of zero charge (PZC) for AH is 11.4, giving the surface a positive

charge at neutral pH, while the PZC for AP varies at around 5, depending on the manufacturing

conditions, so it has a negative charge at neutral pH. In addition to the well-established and widely

available AH and AP, Merck Aluminum Adjuvant is a proprietary aluminum hydroxyphosphate sulfate

formulation (AAHS) that is structurally related to AH, as it forms an amorphous mesh-like structure and

bears a nearly zero surface charge at neutral pH [23]. In contrast to AH, AP and AAHS, Imject alum (50%

aluminum hydroxide / 50% magnesium hydroxide) has not been included in licensed vaccines, but is

commercially available for use in experimental animals. When compared with the others, Imject alum is

chemically very different, comprising crystalline magnesium hydroxide and amorphous aluminum

hydroxide, so not surprisingly, it’s performance has been shown to be significantly different. Therefore,

Imject alum should not be used in any studies that are designed to address the mechanism of action of

adjuvants [24]. Particularly since the alum actually used in a number of licensed vaccines is available for

purchase from commercial suppliers, including Brentag (http://www.brenntag-biosector.com/).

How are alum adjuvants used?

Alum adjuvants are generally used as an ‘adsorbents’ for vaccine antigens, so they generally

operate as an ‘antigen delivery system’, to promote uptake into immune cells. Although in reality, the

mechanism is much more complex. In most vaccines, antigens are adsorbed to the surface of alum

through a variety of different mechanisms to include ligand exchange, electrostatic attraction, and

hydrophobic or van der Waals forces [25]. Since proteins are complex biomolecules, often more than

one mechanism of interaction operates, and the interactions can change over time on storage, a

concept known as vaccine ‘aging’. The strongest attractive force is ligand exchange, in which phosphates

on antigens will displace hydroxyls on the AH surface [25]. So heavily phosphorylated antigens will be

strongly bound and less readily released in vivo, while antigens bound to alum by electrostatic and other

forces are more easily released. However, the degree of interaction for heavily phosphorylated antigens

can be minimized by pre-treatment of AH with phosphate buffers [25]. It is generally agreed that

antigens need to be adsorbed to alum, since low levels of adsorption has been shown to adversely

impact the immune response. However, the overall data is somewhat inconsistent, and there are

examples in which adsorption has been shown to be unnecessary [25]. Alternatively, there are examples

when too ‘tight’ a binding has been shown to negatively impact the vaccine performance [17, 26].

Nevertheless, high levels of antigen adsorption are preferred for formulation consistency, and for most

Page 10: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

antigens improved adsorption results in an improved immune response [26]. The need for adsorption

likely diminishes when high molecular weight antigens are used, since these are less likely to diffuse

away from the injection site. Moreover, although for many antigens stability can be improved by

adsorption to alum, there are also many examples in the literature of antigens which have been

destabilized through adsorption to alum [17, 27]. It is known that the pH of the microenvironment of the

alum surface can be different from the bulk formulation pH, due to attraction of ions, which can

contribute to protein instability. Hence defining the optimal way to use alum as an adjuvant is a multi-

factorial process that remains somewhat empirical. Extensive formulation studies are required with the

specific vaccine antigens if the maximal response is to be obtained with this adjuvant. Fortunately, we

and others have published extensively on how best to optimize the use of alum adjuvants, so extensive

information is available in the literature [6, 25, 28]. However, it is fair to say that many gaps exist in the

published information and often this area of vaccine development is considered more of an art than a

science. Nevertheless, new techniques have emerged in recent years to allow antigens to be directly

quantified on Alum and to allow quantitative and qualitative assessments in situ [21, 29-31].

Interestingly, alum adjuvant is also an effective adjuvant for DNA vaccines, but only if the DNA is

unadsorbed, AH is ineffective since the negatively charged DNA adsorbs [32].

How do alum adjuvants work?

How alum works or what is the ’mechanism of action’ is a question that has occasionally

interested, but has consistently confounded scientists for many decades since the adjuvant was first

introduced. In recent years there has been a significant expansion of work in this area, as awareness

grew of the crucial role of innate immunity in triggering and controlling the adaptive response. This

expansion in knowledge was also accompanied by the development of many new technological tools to

allow the question to be addressed in different ways. Nevertheless, although there have been one or

two apparent ‘eureka’ moments in recent years, with high profile publications resulting, in reality the

mechanism of action remains somewhat opaque. This is probably because there is no single

overwhelming contributor; the mechanism of action is complex and multifactorial, with many factors

operating simultaneously. Nevertheless, there have been several key insights in recent years which have

moved us toward a much improved understanding of how alum works. Several reviews have discussed

this work in detail, particularly from an immunological and innate signaling perspective, but full

discussion of this is beyond the scope of this review [33-35]. Here we will adopt a different approach

Page 11: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

and try to discuss alum from more of a pharmaceutical perspective, since a key component of the

mechanism relates to the size, shape and composition of the particles. Hopefully, our observations some

insight might contribute to the debate on how we might build a better next generation adjuvant. We will

also adopt a more pharmaceutical approach than is usually applied to compare and contrast the other

established adjuvants, and ask if they employ similar or different mechanisms of action.

Controlled or delayed release of antigen (‘Depot effect’)

Originally alum adjuvants were thought to work mainly through retention of antigen at the

injection site, allowing slow release of the antigen and an extended opportunity for interaction with the

recruited antigen presenting cells. This mechanism was proposed by Glenny et. al. and became known

as the ‘depot’ effect, but could also perhaps be more accurately termed as ‘delayed release of antigen’

[36]. The depot mechanism was initially supported by studies that showed that the tissue granuloma

induced by alum adsorbed vaccine could be removed several weeks after administration and following

maceration, be used to immunize additional animals [37]. This suggested that antigen was indeed

present at the injection site for extended time periods. However, although the depot mechanism was

accepted for many decades, it was subsequently downplayed and the importance of alternative

mechanisms was highlighted. The depot effect was first challenged by Holt, who removed the injected

antigen/alum from tissues 14 days after administration and concluded that since this had no effect on

the immune response, a long term depot was not needed [38]. Nevertheless, work conducted at the

same time showed that the use of alum adsorbed vaccines created local inflammation and ensured that

B cell blasts appeared at the injection site and the draining lymph nodes, which were present for several

weeks [39]. In contrast, more recent studies highlighted that 90% of radio-labelled antigen adsorbed to

AP was cleared from the injection site after 24 hours, although the amount of antigen that stayed local

was higher for AH adsorbed than for soluble antigen [40]. Consequently, a recent paper went so far as to

conclude that the depot is not required for alum adjuvanticity [41]. Nevertheless, as newer adjuvants

have emerged, the depot concept appears to have re-emerged, and has been assigned an important

role in the mechanism of action of CAF01 and IC31 adjuvants (discussed further below). Perhaps if we

re-define the depot effect as simply a ‘delayed release of antigen’, this allows an opportunity to

reconcile some of the apparent contradictory claims. Although long term retention of alum adsorbed

antigen at the injection site is not likely, since we know that antigen is displaced in vivo, it has been

confirmed that the alum itself is retained at the injection site, particularly the more crystalline AH [25,

Page 12: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

42]. In addition, other studies have also confirmed that antigen is retained at the injection site for longer

time periods if adsorbed to alum and the duration of retention is probably dependent on the

mechanisms of adsorption [25, 43, 44]. However, given the rapid cellular recruitment that results from

injection with alum, retention at the injection site for only a relatively short time frame would likely be

sufficient to ensure more extensive interaction of the antigens with APC’s. Although the recent work of

Hutchison et. al. appears to counter this suggestion, this work was done using injection into the mouse

ear, not at the usual site of vaccine administration, the muscle [41] . Moreover, if we conclude that

delayed release of antigen is attractive and a key component of the mechanism of action of alum, an

interesting question to address relates to what are the potential benefits of a slower ‘controlled release’

of antigen. Is there a preferred rate of antigen release and can it be attained in vivo?

The key role of cellular recruitment (‘reverse targeting’)

One of the key characteristic outcomes following injection of alum adsorbed vaccines is a

significant cellular infiltration into the injection site, which has been noted since the earliest studies and

was also highlighted more recently [45]. The cellular infiltration is triggered regardless of the injection

site and comprises a diverse range of immunocompetent cells to include neutrophils, eosinophils,

monocytes and dendritic cells [46-48]. In addition, older studies had shown that alum adsorption

promotes antigen uptake into monocytes and also results in activation of these cells [49-51]. The ability

of antigen rendered particulate through alum adsorption to be more effectively taken up and processed

by immune cells has been highlighted as a key role of particulate adjuvants [52]. Alum adsorption has

also been shown to increase the uptake of antigen into DC [53]. In recent studies, we confirmed in vivo

that adjuvants, including alum, are able to recruit a diverse range of immune cells into the injection site

and that these cells were able to take up antigen and deliver it to the local lymph node [48]. Hence a key

role of alum is to trigger a local inflammatory response in the muscle, which results in the recruitment of

immune cells into the injection site, which are able to process and present the antigen. The creation of a

local inflammatory response was also recently highlighted by microarray analysis and confirmed by local

cytokine production [54, 55]. We propose that this cellular infiltration could be termed ‘reverse

targeting’, since the antigen delivery system (alum and others) recruits immune cells to the site of

administration of the vaccine (figure 1). This contrasts with more established ‘targeting’ concepts in drug

delivery, in which a delivery system typically has a ligand attached which helps it to ‘find’ it’s preferred

cellular target following administration. A question that has been raised many times in the literature

Page 13: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

concerns the potential value of ‘targeting’ in vaccine delivery. However, we are skeptical of the potential

value of this approach. Unlike drug delivery systems which are often administered directly into the

blood stream, vaccine adjuvants are typically held within the tissue mass following IM injection, largely

based on their physical dimensions and they tend not to be ‘mobile’. This is true certainly for alum based

adjuvants, although some alternative carriers with a much smaller particle size may have greater

potential to move away from the injection site following administration. Moreover, since many immune

cells are recruited quickly to the injection site that will take up the particulates in a passive process, it is

not clear how a more ‘actively’ targeted system could offer benefits. Cellular uptake is likely controlled

by physical dimensions and surface characteristics, including size, charge, rigidity, so it is difficult to see

how much control over the uptake process could be exerted in these challenging conditions by any

targeting agent. The vaccine is present in an inflamed local tissue environment with many macrophages

and other phagocytic cells present, which have been recruited to take up the particulates. It remains to

be proven if a targeted delivery system can be any more effective than one that is passively taken up by

the many recruited cells. Moreover, it is not clear if movement away from the injection site is beneficial

for vaccine adjuvants. Perhaps smaller particles could move away from the injection site through the

lymphatics and reach the local lymph node directly, which has been claimed to be potentially

advantageous [56]. A potentially targeted vaccine delivery system could offer advantages if

administered by alternative routes, rather than the usual intramuscular route used for standard

prophylactic vaccines. For example, an innovative targeting approach from the field of cancer

diagnostics (Liu et. al.) was recently used to show that peptides adjuvanted by CpG with a lipophilic

albumin-binding domain as a targeting agent were able to effectively traffic to the lymph node and

improve immune responses after subcutaneous administration in comparison to non-targeted

formulations [57]. However, since it is the immune cells that trigger and control the immune response

in the local lymph node, perhaps it is better if antigens are delivered to the lymph node within the

recruited immune cells, which also have the ability to directly deliver the necessaryco-stimulatory

signals ? Since existing adjuvants are already very effective at recruiting immune cells to the injection

site in the muscle, which take up antigen and deliver it to the lymph node, perhaps this is already the

most efficient and effective antigen delivery approach [48]. Nevertheless, we do not exclude the

possibility that direct trafficking of an adjuvant to the local lymph node with an associated antigen could

offer potential benefits, but this needs to be demonstrated. Moreover, antigen targeting may be more

attractive when using alternative routes of administration, to include skin or subcutaneous tissue, in

which the level of cellular infiltration triggered may be significantly less than following traditional

Page 14: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

intramuscular injection. Hence, if we are to employ a ‘targeting’ concept for adjuvants, maybe it is

better to try to further exploit ‘reverse targeting’, in which we could promote the recruitment of a

preferred cellular population into the injection site? The recruitment factors could be co administered

with the vaccine, or could be formulation components of the vaccine which trigger the release of

recruitment factors. We have shown that a key component of the mechanism of action of the o/w

emulsion adjuvant MF59 is to trigger the release of chemokines from immune cells, which are

responsible for the recruitment of immune cells [58]. However, we are unaware of any efforts to alter

the cellular recruitment pattern triggered by a particulate adjuvant system, although this could be a

productive area of research.

Mediators that control the cellular recruitment

If we accept that cellular recruitment is a key component of the mechanism of alum, an

important question to address is what are the key mediators of this recruitment? As highlighted above,

we examined the impact of alum and other adjuvants on human immune cells in vitro and found that

they induced the secretion of chemokines (including CCL2, CCL3, CCL4 and CXCL8), which are all involved

in cell recruitment from blood into peripheral tissues [58]. Alum appeared to act mainly on macrophages

and monocytes, and triggered enhanced endocytosis and an apparent differentiation towards a

dendritic cell phenotype. These observations were consistent with other groups who also highlighted

rapid recruitment of neutrophils into the injection site, soon after alum administration [46, 55]. It was

also suggested that Mast cells can sense alum directly, and along with other cells are responsible for the

secretion of cytokines and chemokines that engender the cellular recruitment. Much earlier studies had

also highlighted that the majority of leukocytes accumulating at the injection site at early stages (<72hrs)

following alum administration were neutrophils, followed by increasing numbers of macrophages by 1

week [59]. The recruited macrophages can contribute to granuloma formation, which can persist long

after vaccination with alum [42]. Hence it is clear that alum triggers the release of various cellular

mediators following administration that result in the rapid recruitment of many diverse immune cells

into the injection site. Clearly these cells are then able to take up antigen, and to be further activated by

the presence of the alum and by other mediators released locally from the recruited cells.

Alum triggers the release of additional pro-inflammatory mediators.

Page 15: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

It has long been established that alum causes the release of a range of factors contributing to

local inflammation, but recent publications have outlined the mechanisms that alum uses to trigger the

release of various inflammatory substances. Early studies had suggested that alum could trigger

eosinophilia [60] and could activate complement [61]. Alum has more recently been shown to induce

the secretion of IL-1β and IL-18 by dendritic cells [62, 63]. Since this occurred in MyD88-deficient

dendritic cells, this was consistent with the observation that alum adjuvant effects in vivo were

independent of TLR signaling [64]. Alum adjuvants also cause the secretion of IL-1α by dendritic cells,

which has overlapping functions with IL-1β [65]. Of particular interest and debate in the recent past has

been the intracellular signaling mechanism by which alum triggers the release of these various

mediators. Controversy has arisen on the role and necessity of the involvement of the NLRP3

inflammasome, with consistent in-vitro data being somewhat undermined by inconsistent in vivo data.

This story has been reviewed extensively by several experts and is beyond the scope of this review [17,

33, 34, 66]. As highlighted by De Gregorio et al., perhaps the single biggest cause of the conflicting data

that has emerged is the use of non-standardized models and materials to assess the mechanism of

action of alum. The different published studies have differed significantly in their sources of alum, dose

used, routes of administration, mouse strains, antigens and regimens etc. Several recent studies have

also suggested that a variety of danger-associated molecular patterns (DAMPs) are involved in the

mechanism of action of alum, since uric acid, DNA, ATP, and HSP70 have been shown to be released by

local tissues following administration of alum [46, 67-69]. Another possible mechanism of cellular

activation by alum involves the triggering of cell surface re-assortment of lipids and the aggregation of

lipid rafts, which has been claimed to result in activation of the syk kinase and phosphoinositide-3 kinase

(PI3K) pathways [70].

A unified theory of how alum works?

As highlighted earlier, one of the most commonly asked basic research questions in vaccinology

in the last decade has been ‘how does alum work ?’ Fortunately, many new tools and techniques have

become available recently to better address this question. However, it appears increasingly unlikely that

any new ‘eureka’ insight will emerge. The mechanism of action is clearly multi factorial, with many

different components contributing collectively to the overall effect. Alum has both ‘physical’ effects due

to size, mass and composition, since it is an insoluble aggregate that triggers the influx of immune cells

that try to engulf it. But alum also has clear ‘immunological’ effects, triggering local inflammation

Page 16: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

through various innate signaling pathways that help to create a local ‘immune competent’ environment

in the muscle. However, it is impossible to separate the physical from the immunological effects, with

likely both operating simultaneously and acting in synergy. As already highlighted above, in addition to

immune activation by PRRs, the innate and adaptive immune systems can also be activated by

endogenous signals that originate from stressed, injured, or necrotic cells. The ‘danger theory’ of

immune activation was first postulated by Matzinger in 1994 as a key part of a new model of immunity

that suggested that immune responses are triggered by substances that cause damage, rather than by

those that are simply foreign [71]. Hence, endogenous danger signals released from damaged cells

which trigger the inflammatory response have been termed ‘alarmins’ or danger-associated molecular

patterns (DAMPs) [72]. Alarmins are thought to have three properties: they are rapidly released in

response to infection or tissue injury; they have chemotactic and activating effects on APCs, particularly

DCs; and they have potent adjuvant effects in vivo. Hence, the concept of alarmins as a danger signal

may link the physical and immunological effects of alum and better explain how these contribute to the

induction of a successful immune response. Alum acts on the tissue by generating a localized site of

inflammation, through upregulation of innate immune regulators such as cytokines and chemokines [73,

74]. Most likely alarmins may also act synergistically with microbial PRR to enhance the inflammatory

reaction following vaccine administration for 2nd generation adjuvants.

Perhaps the key lesson to learn from all the work on the mechanism of action of alum is that

many factors contribute simultaneously, including the depot effect, without any single component

dominating. This may also partially explain why so many different substances can be adjuvants, because

many different pathways can be exploited, which can converge to achieve a common effect. In addition,

perhaps we can conclude that the most successful adjuvants will likely simultaneously exploit several

alternative mechanisms, so that they can continue to work effectively in a diverse population of

individuals with a range of different antigens.

Building second generation adjuvants on alum.

Since the available vaccines are typically used in young children who are usually healthy, there is

understandably little acceptance for safety issues, or for significant adverse events. Hence the key

challenge for the development of new generation adjuvants for inclusion in vaccine products has always

been and will remain ‘safety’ [5]. Since alum is the most widely used first generation adjuvant and has

Page 17: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

an excellent track record of safety and tolerability, it has logically been used as a basis on which to build

second generation adjuvants. As discussed earlier, second generation adjuvants typically comprise a

delivery system (1st generation) with an added component designed to activate innate immunity (an

immune potentiator). Indeed, the first vaccine (Cervarix - offering protection against HPV infection)

containing a 2nd generation adjuvant that was approved in the US comprises a recombinant virus like

particle (VLP) antigen, which was co-adsorbed to alum alongside a TLR4 agonist, called monophosphoryl

Lipid A (MPL) [18]. More than three decades ago, the pioneering work from Ribi [75] resulted in the

development of less toxic preparations of LPS, which were subsequently further detoxified to create the

MPL, which was adsorbed to alum to create the AS04 adjuvant used in Cervarix (GSK). Studies on the

mechanism of action of AS04 have focused on the key impact of the MPL, while rather playing down the

role of the alum, which is claimed only to magnify the effect of the MPL [76]. Alum has also been

extensively evaluated in the clinic as an adsorption and co-delivery platform for other TLR agonists,

particularly the TLR9 agonist, CpG oligonucleotides [77]. However, it is important to be aware that the

adsorption of TLR agonists, particularly CpG oligonucleotides, to alum adjuvants can also negatively

impact the adsorption of antigens [78]. Hence it is necessary to co-optimize the formulation conditions

to ensure effective delivery of both the TLR agonists and the antigens in these complex combination

adjuvants, particularly if more than one antigen is present.

Overall, although it might be reasonably argued that alum was not ‘built for the purpose’ in relation to

its use in second generation adjuvants, it has many attractive attributes beyond the established safety

profile. Alum provides a solid and robust platform to which a variety of materials can be successfully

adsorbed for co delivery with antigens [25]. Since it has been demonstrated on a number of occasions

that TLR agonists require co-delivery with the antigen for optimal effect [79], it is important that alum

can be an effective delivery system for a range of components. The immune system is readily able to

induce potent adaptive responses to microbial antigens delivered to APCs in association with PRR

ligands, which is how pathogens appear to the immune system. Moreover, alum has already contributed

to the development of many stable vaccine products, so it can clearly enable the development of liquid

single vial products based on second generation adjuvants.

Several years ago, a number of small molecular weight synthetic compounds, which were

originally developed as type I IFN inducers, including imidazoquinolines (Imiquimod and Resiquimod)

and guanosine and adenosine analogs, were shown to activate TLR7 and TLR8 [80], which opened up the

possibility to discover small molecule TLR agonists. We have recently developed an encouraging second

Page 18: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

generation adjuvant technology in which small molecular weight immune potentiators (SMIPs) have

used novel chemistries to enable their adsorption and co delivery on alum [81]. We believe that the new

SMIP adjuvants have some potential advantages over the more ‘traditional’ large molecular weight TLR

agonists that have been used so far as adjuvants, eg. MPL. As we highlighted earlier, MPL is a detoxified

natural product with significant heterogeneity and a complex structure activity profile, which can be

difficult to formulate due to poor solubility, and shows a significant tendency to aggregate. In contrast,

SMIPs are traditional drug like small molecules which can be easily manipulated chemically to control

solubility and other solution behaviours, and are more compatible with a range of adjuvant approaches.

For example, we have modified SMIPs by the addition of a phosphonate group so that they can be

adsorbed onto the surface of alum exploiting the ligand exchange process by which many proteins

routinely adsorb [81]. The simplicity of this approach to enable alum adsorption contrasts with an

alternative formulation approach for SMIPs that was also recently described, involving chemical

conjugation of fatty acids [82]. Conjugation of fatty acids creates complex molecules that will likely

inevitably result in the preparation of heterogeneous formulations. Moreover, linking a fatty acid rather

undermines some of the most attractive features of SMIPs, since they are rendered insoluble and prone

to aggregation, like the larger molecular weight adjuvants already available. Since there are now many

different classes of SMIPs already available, and likely many more still to be discovered [15], identifying

the best approach to formulate these materials into second generation adjuvants has become a key

challenge (NIH - Blue riband Adjuvant panel report). Clearly pharmaceutical scientists are in the best

position to rise effectively to this challenge and the field of vaccinology would benefit from more

contributions from teams with diverse experience in similar drug delivery challenges.

While we believe that the discovery of SMIPs offer many advantages and are a step towards perhaps an

idealized third generation adjuvant based on molecularly defined components, an interesting question

to raise is, what should the delivery component look like ? If exploiting alum is a logical first step to

introduce SMIPs, where should we take them next? What else can we do to move towards an optimal

system, which is the best ‘delivery’, should it be a targeted system, should we control the rate of release

of the antigens. We will try to address some of these questions as we discuss the alternative approaches

to first generation ‘delivery’ based adjuvants and assess which advantages they might bring for the third

generation approach .

Emulsion adjuvants.

Page 19: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

Emulsion adjuvants have a long history of use in vaccine formulations, at least dating back to the studies

with ‘Freunds’ adjuvants in the 1940’s. However, although these adjuvants were very effective, they

were poorly tolerated and were not acceptable for widespread use, mainly due to the inclusion of non-

degradable mineral oils. Extensive work in the 20th century focused on a search for better tolerated

emulsion components as adjuvants. The first adjuvant emulsion comprising a fully degradable oil was

developed in the 1960s by Hilleman using peanut oil and was called adjuvant No. 65 [83]. Although this

was a step in the right direction, it took a further 30 years for the first emulsion adjuvant to be included

in a vaccine licensed for human use. MF59, which is an oil in water (o/w) emulsion comprising a low

content of the biodegradable and biocompatible oil, squalene, was first included in a human flu vaccine

in 1997 [7]. Several additional groups followed a similar path to adjuvant development and squalene is

currently the oil component of several o/w emulsion adjuvanted influenza vaccines, including Fluad®

(MF59 adjuvanted seasonal influenza), Aflunov® (MF59 adjuvanted pandemic influenza), Focetria®

(MF59 adjuvanted pandemic influenza), Prepandrix® (AS03 adjuvanted prepandemic influenza), and

Pandremix® (AS03 adjuvanted pandemic influenza). In addition to influenza vaccines, the MF59

adjuvant has also been evaluated in a number of clinical trials for a wide range of alternative vaccines,

including HSV, HIV, HBV, HCV and CMV [7, 66]. Moreover, MF59 has been extensively evaluated in a

wide range of populations of different ages, including young children, and has been shown to be safe

and well tolerated in patients as young as 6 months of age [7]. It has also been shown to allow the

development of an improved seasonal influenza vaccine in young children with significantly enhanced

vaccine efficacy [84]. Alternative emulsion adjuvants to MF59 were also approved for the first time

during the H1N1 influenza pandemic of 2009, including GSK’s AS03 and Sanofi’s AF03. However, recently

published studies have unfortunately highlighted an apparent association between narcolepsy and the

use of an A/H1N1 2009 AS03 adjuvanted influenza vaccine [85]. In contrast, despite intensive

investigations, there has been no evidence of an increased incidence of narcolepsy following the use of

any vaccines, including A/H1N1 2009 influenza vaccine, adjuvanted with MF59 [86]. It is important to

note that although the emulsion adjuvants AS03 and MF59 both contain squalene oil, they have very

different compositions, since AS03 contains the immunostimulatory adjuvant alpha-tocopherol and a

different type of influenza antigen (split virus vs. subunit) [87]. Pre-clinical studies on the mechanism of

action for AS03 showed that the presence of alpha-tocopherol induced a non-specific activation of the

immune system in the local lymph nodes of mice, in marked contrast to squalene only containing

emulsions, like MF59, which showed specific immune activation only in the muscle where the adjuvant

was injected [87, 88].

Page 20: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

Over the years, we have extensively investigated the mechanism of action of MF59 and have

identified key insights into how it works, which we have described in detail in a number of publications

[48, 58]. We have shown that normal tissue-resident monocytes, macrophages and dendritic cells are

activated by MF59 in the muscle and respond by inducing a mixture of cytokines and chemokines, which

results in the migration of immune cells into the injection site. The recruited cells, including monocytes

and granulocytes, produce the same factors following contact with MF59 and further amplify the

building chemokine gradient in the tissues. The outcome is a dramatically localized signal amplification

loop, which results in a significant influx of phagocytic cells into the injection site in the muscle. The high

number of cells recruited likely results in more efficient transport of antigen to the lymph nodes.

However, in contrast to alum, MF59 does not create any kind of depot and a local granuloma in the

muscle is not induced. Older studies conducted with radiolabeled MF59 showed that the adjuvant is

quickly cleared from the injection site, and that only ∼10% remains 6 h after intramuscular

administration [89]. Moreover, it was also shown that MF59 does not influence the distribution or the

half-life of the co administered vaccine antigen [90]. In addition to promoting antigen uptake by APC’s

[90], MF59 may also enhance and accelerate the differentiation of cells toward DCs and alters their

phenotype. We recently reviewed the mechanism of action of MF59 and compared it to what we know

about how alum works [88]. We noted that there are many similarities, but MF59 tends to trigger more

potent local signals in the muscle, particularly for cell recruitment, and tends to operate on different cell

populations. There are also some additional notable differences, eg. MF59 has recently been shown to

induce the release of the alarmin ATP, while alum does not [91]. We believe that ATP is a crucial

contributor to the enhanced cell recruitment and other events key to the mechanism of MF59, including

the trafficking of antigen to the local lymph node. Interestingly, we have also shown that none of the

individual components of MF59 have an adjuvant effect when used alone, it is necessary to prepare the

MF59 emulsion to have an adjuvant effect [92]. Hence, in contrast to AS03, MF59 does not have any

individual immune stimulating components, although the surfactant Span 85, does appear to contribute

to the innate signaling [92].

o/w Emulsions have also been used to develop second generation adjuvants, through the addition of an

immune potentiator. Most notably, IDRI have used a squalene based emulsion called SE (stable

emulsion), as a delivery system for a synthetic TLR4 agonist called GLA (glucopyranosyl lipid adjuvant)[93,

94]. The emulsion formulation containing GLA is called GLA-SE, its composition has varied in the

literature but primarily consists of 4% squalene (v/v), 0.72% glycerol (v/v), 0.76% phosphatydylcholine

(w/v), 0.4% GLA (w/v), and either 0.036% Pluronic F68 or 0.21% Tween 80 (w/v) in an ammonium

Page 21: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

phosphate buffer [95, 96]. Recent studies highlighted that SE-GLA prepared with squalene induced the

most potent Th1 responses in mice in comparison to alternative delivery technologies including, alum-

GLA, SE-GLA prepared with Miglyol, or grape seed oil, or liposomes [97]. SE-GLA has been evaluated as

an adjuvant for vaccines against a number of diseases, including leishmaniasis, malaria, influenza,

tuberculosis and HIV [93, 96-100]. While this is looking like an encouraging vaccine approach,

particularly to treat and prevent leprosy, our experiences with MF59 make us skeptical about the

suitability of emulsions as the best platform for second generation adjuvants [101]. In our hands,

although combinations of TLR agonists including TLR9 and TLR4 with MF59 looked encouraging pre

clinically [102, 103], no benefit was obtained in the clinic, at least in evaluation for flu vaccines [7].

Although adults are already seropositive to flu, so it is more difficult for additional adjuvants to have an

impact in this situation, rather than in subjects who are seronegative to the pathogen/vaccine.

Moreover, for flu vaccines it is really only the antibody response that matters, inducing a more potent T

cell response or a more focused Th1 response is not really important, although it can be for other

infectious diseases, including leprosy. Attention must be paid to the fundamental formulation science if

a robust combination adjuvant is to result, SE/GLA is prepared by dispersing, not dissolving the

components in the oil phase through sonication and heat [93] . In our hands, we have found that many

compounds, although correctly classified as lipophilic have very poor solubility in squalene (unpublished

results). Consequently, when a formulation comprising an immune potentiator in an emulsion is

prepared, the resulting emulsions can have poor stability, or heterogeneous mixtures can result, in

which the immune potentiator is not evenly distributed. Overall, although there is good evidence for

alum that co-delivery of all the vaccine components is important, the story for emulsions is less clear.

We have highlighted earlier that the antigen can be simply co-administered within the emulsion, but it

may be preferable to ensure that any immune potentiator remains associated with the oil droplets.

However, this remains an area requiring more critical evaluation on a case by case basis. An interesting

story emerged very recently that highlighted that the ‘tolerability’ (pyrogenicity) of a TLR4 agonist could

be improved by ensuring co-delivery in the oil droplet of an o/w emulsion adjuvant (AF03 – J. Haensler,

Sanofi, unpublished observations presented at IMV 2014, Albufeira). As highlighted earlier,

combinations of TLR agonists with MF59 were not well tolerated in the clinic, so we have subsequently

chosen to mostly avoid such combinations [7].

In an effort to expand the utility of emulsions for vaccine delivery, we also created and used

cationic emulsions based on squalene as a delivery system for DNA vaccines [104]and TLR9 agonist

oligonucleotides [105],which were adsorbed to the droplet surface. A similar approach was also

Page 22: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

exploited recently for the delivery of a new generation nucleic acid vaccine based on RNA [106]. In

addition to an adjuvant effect, the emulsion also serves to protect the RNA against degradation, and

promotes cellular uptake and sub-cellular delivery. Interestingly, the need for nucleic acids to be

adsorbed to emulsions for a positive impact contrasts sharply with the performance of alum, which was

an effective adjuvant for DNA only if the nucleic acid was unabsorbed [32].

A key characteristic for o/w emulsions in their performance as vaccine adjuvants is droplet size,

since MF59, AS03, SE and AF03 all have particle sizes between 100 and 160nm[6]. Recent advances in

particle analysis technology such as nanoparticle tracking analysis (NTA) has allowed researchers to

better understand the size distributions of mixtures [107]. The application of cryo-electron microscopy

has also allowed for direct visualization of emulsions. Since the samples are snap frozen prior to analysis

without the formation of ice, the expectation is that the morphology of the emulsion remains intact

[108]. Detailed characterization and analysis of the source of components used in emulsion preparation

also need to be monitored, since small changes can readily impact both stability and potency.

Liposomes as adjuvants.

The first report of the use of liposomes as a vaccine adjuvant was in 1974, when it was shown that

negatively charged liposomes induced an enhanced antibody response to diphtheria toxoid [109].

Liposomes were originally established as drug delivery systems and there are several products on the

market, showing that scale up and development of this approach is eminently feasible. Liposomes were

also adapted as adjuvants, but tend to be relatively lacking in potency in comparison to alternatives.

More often, liposomes have been used as a component of 2nd generation adjuvants, including AS01 [110]

and CAF01 [111]. In fact shortly after the first reported use of liposomes as vaccine adjuvants, Banerji

and Alving reported the use of liposomes for delivery of lipid A [112].

GSK has published clinical comparisons between AS01 (liposomes with QS21 and MPL) and AS02 (o/w

emulsion with QS21 and MPL) as adjuvants for malaria vaccines [113]. In initial clinical studies AS02 was

chosen as the adjuvant for the vaccine candidate after being compared against AS03 (o/w emulsion) and

AS04 (alum / MPL) [114]. However, subsequent trials comparing AS01 and AS02 illustrated that the

malaria vaccine adjuvanted with AS01 elicited stronger anti-CSP antibody responses and multifunctional

CD4+ T-cells than adjuvant AS02, which is similar to the observations in a rhesus model [113]. These

results are an excellent illustration of how delivery of immunepotentiators in different formulations can

Page 23: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

impact immunogenicity performance, since both formulations contained the same amount of QS21 and

MPL per dose. AS01 comprises dioleyl phosphatidylcholine (DOPC), cholesterol, and MPL, co-

formulated with QS21 in a 20 : 5 : 1 : 1 (w/w) ratio [115], whereas AS02 is composed of squalene, DL-α-

tocopherol and tween 80, which is the emulsion adjuvant AS03 [116], with the addition of QS21 and

MPL. For the malaria Phase III trial AS01 and RTS,S are simply combined as a ‘bedside’ mix [117].

Although there are no reports in the literature that we are aware of on how exactly AS01 is prepared,

patents indicate that the liposomes with MPL are prepared and added separately to a suspension of

QS21 [115]. Since it is well established that saponins such as QS21 interact with cholesterol, it would be

expected that there is an interaction between these 2 separate components in the AS01 formulation

[115, 118]. To our knowledge, similar to AS01 the details of how AS02 is prepared has not been

disclosed publicly, although the formulation appears to be a simple mixture of the preformed AS03

emulsion, with QS21 and MPL [119]. We could speculate that differences in the location in the

formulation of the immune potentiators may be responsible for the differences in potency between

these 2 adjuvants formulations (AS01 and AS02) which include the same immune potentiators at the

same dose level, although the contribution of alpha tocopherol in AS02 cannot be ignored.

Liposomes have been shown to have little to no adjuvant effect when used alone, without the

addition of an immune potentiator [120]. Although when administered at high doses IV, liposomes have

been shown to induce immune mediated hypersensitivity via the complement system [121]. Likely the

selection and optimization of the liposome composition, and dose delivered will play a role in activating

the immune system, or in avoiding activation if preferred. Liposome charge was found to impact the

ability of liposomes to induce hypersensitivity, with more negatively charged liposomes generating a

greater number of adverse events [121]. Similarly for vaccine delivery purposes, it was found that

anionic liposomes were more potent than neutral or cationic liposomes in generating Th1 responses [97].

Overall, it appears that perhaps liposomes are entering a new ‘renaissance’ in vaccine delivery, with the

field beginning to understand what impacts their potency as adjuvants, and how to use them most

effectively. In pre clinical studies, basic physicochemical parameters such as particle charge and size,

lipid membrane fluidity, membrane glass transition temperature, and the location of antigen (e.g

entrapped, surface bound, or free) have all been shown to impact potency as adjuvants [122]. If antigen

co-delivery is desired, antigens can be delivered with liposomes through a number of different

approaches, including entrapment, electrostatic binding to the surface, covalent attachment, or

membrane attachment [122-124]. Recently researchers have developed a new method to entrap

antigens and to define the location (outer membrane vs. inner membrane) of immune potentiators

Page 24: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

through the crosslinking of lipid bilayers that rapidly degrade in the presence of lipase [125].

Remarkably, this approach appears to induce similar levels of T-cell responses to those than can be

induced with viral vectors or nucleic acid vaccines in small animal models [125]. Further evaluation is

needed with a range of antigens and in larger species to determine if this approach can be applied

broadly, but early data are certainly encouraging. Additionally, scale up and manufacturing for this

technology remains to be established.

If in theory one can dampen the inherent signaling of liposomes by choosing the right formulation

components, does this potentially make liposomes the ‘best’ carrier for the future ? Since then we can

perhaps fully control the immune activation signal by adding the preferred activation molecules. From a

formulation design perspective this appears attractive, but in reality it may prove very difficult to

implement. Unfortunately, formulation optimization is likely to be antigen dependent, since different

antigens may interact very differently with liposomes, making it more difficult for liposomes to perform

like a broadly applicable adjuvant platform. Liposomes have been around as vaccine adjuvants for 40

years, yet their use still remains limited and so far, alum or emulsions have beeen preferred. Is this

likely to change ? Even if a generic ‘immunosilent liposome’ can be identified, which can carry immune

activation signals, questions will remain on how best to deliver many different antigens, unless it

becomes clear that antigens can be simply co-administered with liposomes and there is no need for

association. Nevertheless, liposomes like emulsions still bring the complication that it is difficult to

envisage how a single vial aqueous formulation can be defined in which all components are stable. As

discussed earlier the AS01 liposomal formulation that is in Phase III trials for malaria involves

reconstitution of the lyophilized antigen with the liposomal formulation of immune potentiators, so two

vials are necessary. While a possible solution, this is not preferred in the field, although ultimately

perhaps the liposome and the antigen can be lyophilized together, this would still require a second vial

of liquid formulation for reconstitution.

There are many examples of components that have been added to liposomes to modulate the

immune responses, including cationic lipids, TLR agonists, saponins, c-type lectin receptor agonists, and

lipids extracted from a type of single cell organisms called archaeons [122]. Each of these added lipid

immune potentiators have been shown to impact how liposomes activate the immune system. Immune

potentiators such as saponins and archaeon lipid extracts are heterogeneous mixtures, since they are

extracted from natural sources. To our knowledge archaeon lipid extracts have not been extensively

studied to determine what are the full range of active components contained. Overall, we feel that

Page 25: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

introducing ill-defined components into a formulation is perhaps a retrogressive step and is potentially

moving the field of adjuvants in the wrong direction. As discussed earlier, we are now entering the time

when molecularly defined adjuvants are available and we believe that these should constitute the

vaccine adjuvants of the future, rather than the ill-defined natural products of the past.

Virosomes are generally described as a liposomal adjuvant, but to our interpretation this

appears to be an inaccurate description, since they are really a different process for making a flu vaccine.

Hence the fundamental definition of an adjuvant cannot be applied, we have not added a component to

an antigen to make it more potent. In reality, following disruption of the viral membranes with

detergents, phospholipids are added to the influenza antigens and both are included in the final

product, which is a liposome. However, since the lipids are included in the final product to allow it’s

formation, it is not possible to test the vaccine antigens alone without the added adjuvant to

demonstrate an adjuvant effect. What has been created is in fact an alternative flu vaccine, with added

phospholipids replacing the natural lipids used in the viral membrane. Although the Virosomal vaccines

contain flu HA in their membranes, the same antigen is a vaccine when used alone as an inactivated

virus, and does not actually need an adjuvant to be effective in most people [126, 127]. Hence, the

description of virosomes as a liposome adjuvanted vaccine appears to be an anomaly and what we really

have is an alternative presentation of a flu vaccine which includes liposomes. Nevertheless, virosomes

are certainly as effective as alternative flu vaccines, but they appear to be lacking in potency relative to a

flu vaccine adjuvanted with an emulsion, such as MF59 [128]. Another product that uses HA virosomes is

Epaxal, a vaccine against hepatitis A virus, in which the virosomes are combined with formalin-

inactivated and purified hepatitis A virions. The HA virosomes and hepatitis A interact with one another

through electrostatic interactions. And the vaccine is stored as a liquid in a pre-filled syringe without any

additional preservatives [129]. The mechanism of improved immune responses is not clear, but there

appears to be improved T cell help and maybe innate activation from the virosome and it’s components,

to which the population has been primed due to previous exposure or immunization with flu virus [130].

Micro/nanoparticles as adjuvants

The use of PLG microparticles for vaccine delivery was first described in the early 1990’s, when

they were proposed by several groups independently as potential controlled release vaccines that could

obviate the need for booster injections [52]. However, the initial observation that polymeric

Page 26: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

microparticles could operate as a synthetic adjuvants was much earlier [131] and PLG microparticles

were actually first used as an oral antigen delivery systems [132]. Novel polymers had also been

proposed for controlled release of vaccine, before the more well established PLG was adopted [133].

PLG had already achieved success for drug delivery with approved products on the market [134].

However, despite extensive effort over several decades, polymeric microparticles have not yet been

successfully developed as a vaccine product, although scale up, processing and manufacturing are

already in place for this technology [135]. The potential use of PLG microparticles with encapsulated

proteins to provide sustained release was widely considered to have failed in the mid 1990’s, mainly

because the harsh process of encapsulation was not suitable for protein antigens [52]. Although, it is

conceivable to use excipients to stabilize and protect the antigen during processing, this approach has

rightly been judged a ‘mission impossible’ by most researchers, although some efforts continue [136].

Moreover, the microencapsulation approach to vaccines would be expensive relative to alternative

adjuvants, due to the need for aseptic manufacturing. Since the cost of vaccines in comparison to

therapeutic products, is usually low, the economics may not be justified for microencapsulated vaccines.

Nevertheless, a novel approach involving adsorption of antigen onto the surface of pre-formed charged

PLG microparticles was developed to overcome the limitations of microencapsulation [137]. Adsorbing

the antigen to the surface overcame the antigen instability issues, and recombinant soluble antigens

were shown to retain structural integrity, which was lost if the antigen was entrapped [138]. In addition,

immune potentiators could also be encapsulated within the PLG particles, since these are often much

more stable than protein antigens [139]. Charged PLG with adsorbed TLR9 oligo’s can also be used as a

combined adjuvant, and were shown to enhance the protective efficacy of a licensed anthrax vaccine in

an established pre-clinical challenge model [140]. An additional step forward to the potential

development of PLG for vaccine delivery involved sterilization by gamma irradiation of the pre-formed

microparticles, prior to antigen adsorption, which eliminated the need for aseptic manufacturing [141].

In contrast to Alum, PLG microparticles are comprised of biodegradable polymers which leave no tissue

residue. In addition, their safety in clinical use has been established at least for drug delivery purposes.

However, a major disadvantage of the PLG polymer is that the particles need to be lyophilized, which

adds significant expense to the product. This is in marked contrast to Alum, which typically allows the

development of stable single vial liquid formulations. In a number of pre-clinical studies in small animal

models, we have shown that PLG perhaps represents an alternative to alum for a range of established

and new generation vaccines [135]. However, although in mice PLG could trigger cross priming and

induce a CD8+ cytotoxic T cell response [142],this was not replicated in studies in non-human primates

Page 27: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

[143]. In addition, PLG were not more potent than the traditional alum adjuvant for the induction of

antibody responses to recombinant protein antigens in non-human primates (unpublished data). Hence,

in the long run, perhaps the biggest potential for PLG is as a co delivery system for antigen and one or

more immune potentiators, including SMIPs, which can be entrapped within the particle to which the

antigen is adsorbed [139]. Alternatively more than one TLR agonists can be co-entrapped within the PLG

with the antigen, if we agree that synergy of different TLR agonists is possible [144]. However, if this

approach is adopted, the antigen will be subjected to instability during microencapsulation and

subsequent release, as highlighted earlier [40, 135]. Moreover, this approach would require the

expensive encapsulation process and efficiencies might be low when trying to simultaneously entrap

antigens and more than one TLR agonists, with potentially very different physicochemical characteristics,

including different solubility profiles. The technical challenges for this interesting concept are quite

significant, so the advantages of encapsulation would need to be very clear and compelling. It has been

highlighted that nanoparticles have a number of potential advantages as adjuvants [56]. We and others

have extensively evaluated PLG and other nanoparticles, but the advantages in terms of potency are not

yet apparent. We described a practical process whereby nanoparticle vaccines could be created and

evaluated for vaccine delivery [145], a step that has been missing from many of the studies in this area,

but we did not see any advantages in terms of potency, at least for antibody induction, for the

nanoparticles versus the microparticles [146]. Recently an alternative way to make PLG particles that

can control the size and shape of the particles was described [147]. Using this technology rod shaped

cationic PLG particles (80nm x 320nm) were added to flu antigen [148]. Immunogenicity was

significantly improved, though no alternative adjuvants were evaluated for comparison, so it’s difficult

to get a real appreciation of the potential of the technology. We understand that this technology was

not more potent than the established flu vaccine when evaluated in a recent clinical trial (unpublished

data). Although it may be shown that the potency of PLG particles is dependent on size, we do not

believe that this has yet been clearly demonstrated. We would be excited to see high quality studies in

this area, particularly focusing on the T cell response, and perhaps using nanoparticles as delivery

systems for TLR agonists.

There are many alternative materials that have been evaluated as particulate delivery systems for

vaccine adjuvants including but not limited to polyorthoesters, polyphosphazenes, chitosan, inulin and

alginates [149-153]. The possible advantages of these systems are not immediately clear, perhaps one

can argue that a fully water soluble system could prove to be an advantage (e.g alginates), in terms of

antigen stability during formulation and antigen entrapment, but as highlighted, antigens do not need to

Page 28: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

be entrapped to ensure co-delivery. One aspect that has been missing from this work is a direct head to

head comparison with established adjuvants. Thus far there appears to be no clear advantages to these

systems, as other more advanced adjuvants have been able to elicit similar responses

Iscom adjuvants.

ISCOMS were originally described by Bror Morein in 1984 as a particulate carrier of Quil adjuvants and

cholesterol with membrane bound viral antigens incorporated [154], but the original technology was not

sufficiently well tolerated in man to allow commercial vaccine development. Nevertheless, extensive

modification of the technology was subsequently undertaken to mitigate the toxicity profile of Quil A,

including the concept of ISCOMATRIX (CSL), which was a pre-formed particulate with a different

composition of Quil A, to which the antigen was simply added [155]. This adjuvant concept and various

modifications with different and sometimes more highly purified Quil fractions continues to be

evaluated as an adjuvant in several settings, but mainly as therapeutic vaccines. Since the antigen is no

longer incorporated into the particulate, many more antigens can be evaluated with Iscoms, including

soluble recombinant antigens. However, the stability and perhaps the localization of the co-delivered

antigens is likely key for this adjuvant concept to be a success. The performance of Iscoms appears to be

partially dependent on antigen association, which is predominantly by electrostatic attraction forces

[117]. The newer concept of a preformed Iscomatrix with highly purified Quils at a fixed ratio was much

more attractive from a manufacturing perspective, but there may be a loss in potency when antigens

are no longer incorporated. However, newer versions of Iscoms are now available which use charged

based interactions to promote antigen association (cationic Iscoms). Mechanistically, ISCOMs appear to

destabilize endosomal membranes, which allows greater cytoplasmic access for co-delivered antigens

compared to other forms of antigen delivery [156]. Therefore, in pre-clinical studies in small animal

models, Iscoms became the gold standard adjuvant for the induction of CTL responses. They were also

able to induce CTL responses in non-human primates [157], which was a very encouraging observation,

but this could not be repeated in human clinical trials.

Rather than the emulsion, liposome and microparticle approaches discussed earlier, Iscoms are

an adjuvant technology that was really built ‘for the purpose’, they have only been used as adjuvants

and not for drug delivery purposes. However, this means that they were not able to capitalize on

process developments elsewhere in the pharmaceutical arena, any developments occurred exclusively

Page 29: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

in the vaccine field. GMP grade ISCOMS are prepared by solubilizing cholesterol and phospholipid in a

detergent that is subsequently mixed with saponin’s in solution, after an equilibration step, the resulting

material undergoes buffer exchange and detergent removal via dialysis or ultrafiltration; upon removal

of the detergent, the ISCOM particles spontaneously form [158]. Other preparation methods familiar

to pharmaceutical scientists such as film hydration have been evaluated, but the formulations created

have not been tested clinically [159]. Once prepared the resulting particles are stored at 4°C. This

complicates the storage of a single vial vaccine, as the antigen would also have to be stable for liquid

storage at 4°C. Since a highly purified fraction of Quil A is now included in a liposomal vaccine adjuvant

formulation (AS01) that is undergoing Phase III evaluation as a malaria vaccine, perhaps it is fair to ask

the question, if you want a Quil as a vaccine adjuvant, why not use the established liposomal approach

rather than Iscoms ? In both approaches, the Quils are bound into particulate structures through their

avid binding to cholesterol, which is a key formulation component. In both adjuvant technologies,

binding to cholesterol is a key approach to minimize the toxicity profile of Quils, a concept than has

been called Quil ‘quenching’ by GSK scientists.

Recently described adjuvant approaches – IC31 and CAF (The depot is back!).

As discussed earlier, alum based adjuvants were once thought to act mainly through the

creation of a depot at the site of injection, leading to a slow release of the antigen, but the depot effect

is in reality only part of a multi-faceted mechanism. Unlike emulsions which do not form a depot at the

site of injection [88], there have been detailed reports recently on the mechanism of action of CAF01

and IC31, two very different adjuvants that appear to require a depot effect for an adjuvant response.

Interestingly both adjuvants are able to elicit potent Th1 biased immune responses in preclinical models.

CAF01 is a cationic liposome composed of Dimethyldioctadecylammonium bromide (DDA), and

the glycolipid trehalose 6,6’-dibehenate (TDB) in a 5:1 (w/w) ratio prepared by film hydration. The

adjuvant has been tested in vaccines to protect against a wide range of disease indications including TB,

HIV, chlamydia, flu, and malaria [160-164]. The adjuvant effect of DDA has been well established since

the 1960’s, yet this molecule was never advanced as a vaccine adjuvant due to inherent instability of the

molecule [111, 165]. However, Davidsen et al. discovered that when formulated with TDB, the stability

of the DDA liposomes improved, and were more potent than DDA liposomes alone [111]. Despite the

increased liposome stability, when mixed with antigens the shelf-life of the vaccine is relatively short,

Page 30: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

requiring the antigen to be combined with the liposomes immediately prior to administration. Once

combined, the antigens adsorb to the surface of the liposomes to form adjuvant / antigen complexes

[166]. It has been shown that changes to the liposome composition change the biodistribution of the

adjuvant and the subsequent immune responses induced. Not surprisingly, increasing rigidity of the

liposomal adjuvant membrane by addition of cholesterol was found to impact the biodistribution of

CAF01. As cholesterol was added into the bilayer, the particles were retained at the site of injection for

an extended time, leading to an increase in the residence time of the antigen, greater activation within

the draining lymph node and an increased T-cell response [167]. Similarly PEGylation of the adjuvant

was found to inhibit the formation of a depot, negatively impacting the immune responses [168]. In fact

many changes to the composition and the biodistribution of CAF01 impacted the adjuvant response

including cationic lipid type, degree of lipid saturation, particle size and charge of the liposome, which is

consistent with what we highlighted above regarding previous liposomal adjuvants [169, 170] . To

summarize, extended muscle retention and slow release of liposome / antigen from the site of injection

favors a stronger Th1 response. However, similar to alum, we believe that this is only a part of the story,

and that this is an area that well thought out studies probing how the immune system interacts with the

liposomes could prove useful to the field.

IC31 is another adjuvant that has recently been shown to form a depot at the site of injection for

optimal immune responses. Originally designed to build on previous work on delivery of TLR9 agonists,

IC31 contains a lysine / leucine rich antimicrobial peptide KLK (KLKLLLLLKLK) derived from sapecin B, that

has a net positive charge that interacts with the negative phosphates of ODN1a [171]. The adjuvant

effect of IC31 appears to be TLR9 dependent through the induction of IFN-gamma [172]. KLK was found

to stimulate uptake and subsequent cellular internalization of ODN1a by endocytosis, similar to other

charged peptides bound with oligonucleotides [173]. Interestingly, it was shown that KLK remains on

the surface of the cell, promoting de-complexation and resulting in endosomal uptake of only the ODN,

contrary to what is observed within the field of non-viral gene therapy, where oligonucleotides are

typically shuttled into cells with the carrier [174]. Work done in-vitro with pDCs and monocyte derived

dendritic cells treated with IC31 showed a greater interaction with monocytes as opposed to MHC class

II cells [175]. IC31 is thought to work on vesicular targeting of TLR ligands by KLK and TLR9 by ODN1a in

moDCs. Spatial distribution of IC31 may be important due to increased endosomal accumulation after

repeated exposure [175]. When injected intramuscularly IC31 was found to form a depot at the site of

injection that was present for as long as 58 days after injection. [172] As one would expect, the KLK

peptide needed to be present to form the depot, and the depot was longest lived when all components

Page 31: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

(antigen, ODN, and KLK) were present [171, 172]. It is unclear what the advantage is to retaining an

antigen at the site of injection for 58 days, when there are other adjuvants that allow for faster

clearance. Additionally, is an extended depot necessary for the adjuvant response of IC31, as it has

been shown that activation of a soluble TLR9 agonist in the presence of a soluble antigen leads to

improved immune responses, though co-delivery of antigen and adjuvant appears to further improve

responses [176]. Some well-designed experiments to determine how critical the depot effect is for the

subsequent immune response would be highly beneficial to the field.

How do we design the next best generation of adjuvants ?

In this section we would like to take the opportunity to highlight a number of questions that we

believe are important and worthy of further consideration, if we are to get closer to defining the next

best adjuvant technology. By asking the questions, we obviously do not believe that an answer is yet

available, but we will attempt to offer our own views of the most likely answers based on the currently

available knowledge. The main purpose for raising these questions is to highlight areas that might be

benefit from some well-designed and appropriately controlled basic research studies.

Do we need targeted adjuvant systems, would they perform better than those that we currently

have? If we believe that the answer is yes, which are the right targets and where do they reside, which

are the receptor systems that can be exploited? As discussed earlier, we are skeptical of the value of

targeting for adjuvants which will be injected into the muscle, since they already benefit significantly

from ‘reverse targeting’, the adjuvant triggered influx of immune cells. So it’s difficult to imagine how a

targeted system could offer benefits when most of the particles are taken up anyway. However, in

theory we could promote particle interaction with a specific cell population that is preferentially

recruited to the injection site and this could potentially bring benefits. Moreover, this is a concept that

can be tested in pre-clinical studies, since targeting agents for different sub populations of immune cells

are readily available. Nevertheless, perhaps a targeted adjuvant system would likely be more beneficial

if delivered into an alternative site, such as the skin, which has many immune competent cells already

present, unlike the muscle where they need to be recruited? Again, this is a testable hypothesis worthy

of investigation in pre-clinical models.

Another good question to address is whether or not there is a new generation of ‘carrier’ that

can replace what is currently available? Many adjuvants have been around for quite some time and have

Page 32: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

been successfully scaled up and developed commercially (Alum, emulsions and liposomes), while some

newer adjuvants have only emerged in recent years (CAF01, IC31 etc.), and are supposedly ‘purpose

built’. Are the newer ones more potent than the ones we had already? We think this remains an open

question, but one that can only be addressed with carefully controlled human studies; fortunately such

studies are already underway. Nevertheless, it should be clear that for adjuvants, potency is certainly

not the only question, safety is actually far more important [5]. In addition, there are many other key

questions to address for the new approaches beyond safety and tolerability, including their ability to be

successfully scaled up, cost of goods, stability with different antigens, suitability for a wide range of

antigens etc. If we are clear that we want to accomplish co-delivery of antigen and immune potentiator

to the same cells, is a formulation approach preferred, or could this be better accomplished by

chemically conjugating the immune potentiator to the antigen [177]. What are the respective pro and

cons of the conjugation approach versus the more established formulation based approaches? As our

understanding of the immune system improves, along with a broader appreciation of its key triggers and

controls, the potential to harness it more effectively, increases not only for preventing infectious

diseases, but possibly also for ‘therapy’. However, it is clear that there will be a need for much more

potent adjuvants to enable the development of therapeutic vaccines, particularly if it is necessary to

overcome ‘tolerance’ to enable cancer vaccines. In addition, the acceptable tolerability and safety

profile of adjuvants will be very different for therapeutic vaccines to be used for people already sick,

rather than standard prophylactic vaccines, which are administered to healthy people. Therefore, a

much broader range of adjuvant technologies than those highlighted here could have a key role in

therapeutic vaccines, particularly if a range of immune potentiators is included. Nevertheless, even the

most potent adjuvant technologies may not be effective, without combination with alternative

approaches such as live vectors or nucleic acids, which will be much more potent for the induction of

CD8+ responses, which likely will be necessary. Hence in many areas, the potential value of new

adjuvants in a prime/boost setting needs to be addressed, in which a potent immune response is already

triggered by a viral vector or a nucleic acid. It is not yet clear which are the best adjuvants to use in this

setting, or if the most potent adjuvants to prime a response in an antigen naive animal will still be the

best to boost that response when the animal, or human, is already ‘heavily’ primed by a vector or

nucleic acid ?

Additional questions worthy of consideration for which we believe answers are not yet available

include the following; which is the best available delivery system to accomplish co delivery of antigen

and TLR agonists? Should we combine TLR agonists, which ones, or should they be combined with

Page 33: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

alternative compounds that activate different elements of innate immunity? Do we need controlled

release, for the antigen, or for the TLR agonist? One key lesson from studies of live attenuated vaccines

is that activation of multiple innate receptors simultaneously is more effective than activation of a single

pathway [178]. Therefore, an optimal vaccine candidate may require the delivery of several TLR agonists

within the same delivery system, in conjunction with the antigens. Alternatively, we may need to deliver

selected TLR agonists in conjunction with additional agonists which activate different pathways of innate

immunity. If we want controlled release, do the antigen and the TLR agonist need to be released at the

same time, or differently? Given the likely different physicochemical characteristics of antigens and TLR

agonists, is there a delivery system with enough flexibility to accommodate co delivery of both agents

simultaneously? Do we know which controlled release profile is preferred, for antigen and TLR agonists,

and how do we get it? Will any preferred release profile that is identified be antigen and adjuvant

dependent, and not universally applicable? If vaccines need mucosal immunity do we have to design an

effective mucosal delivery system with all the associated challenges, or can we induce mucosal

immunity by adding excipients such as retinoic acid to a standard injectable formulation [179]?

Alternatively could metabolites of vitamin B be the most effective way to induce mucosal immunity, but

how should we administer them [180]?

We believe that many of these questions will be best addressed by basic researchers probably

based in academia or the public sector, but this work will need to be collaborative. Formulation

scientists can potentially build a broad range of delivery systems, comprising targeted systems and those

with controlled release characteristics, but they need to collaborate extensively with immunologists and

other scientists to determine what works best. Unfortunately, vaccinology remains an inexact science,

with many basic concepts still not well understood. Even ‘immunogenicity’ is a poorly defined

phenomena, since we often have a limited understanding of why certain antigenic components are

‘immune dominant’, while others are more silent? However, we would contend that pharmaceutical

scientists have a key and distinct role to play in designing the next generation of adjuvants. We hope

that with this discourse on the many ‘unknowns’ we have helped to begin to define a framework of

what is currently known, which is often not nearly enough, and we have encouraged more basic

researchers to help to seek solutions to these complex problems.

Conclusions

Page 34: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

For too long vaccine adjuvants have been a somewhat ill-defined and poorly understood area in

science, particularly mechanistically, but we are at a point in vaccine development when they need to

become as well defined as other pharmaceutical formulations. Although there have been significant

improvements in the relatively recent past, with new fully characterized adjuvant formulations included

in licensed vaccines, there are still too many examples of poorly characterized adjuvants, some of which

are still progressing into clinical evaluation. Even if these adjuvants perform well in the clinic, they risk

repeating the failures of the past and not progressing beyond initial evaluations. We believe that

vaccine adjuvants need to be considered like any other pharmaceutical formulation, and the

fundamental considerations should be unchanged. If they are to succeed and to be eventually included

in licensed products, adjuvants need to be robust, scaleable and reproducible. In addition, they need to

display these characteristics in conjunction with the vaccine antigens with which they will be co-

formulated, to create a vaccine product. Hence it should be clear that there is a key role to play for

pharmaceutical scientists in contributing to the next generation of rationally designed adjuvants. In our

opinion, the role of pharmaceutical scientists in the discovery and development of vaccine adjuvants has

been traditionally under-appreciated, which may partially explain the many failures in this area. The

identification of adjuvant active compounds has been over emphasized, while the need to create an

appropriate formulation with the compounds has been less well understood. We would like to take this

opportunity to encourage all vaccinologists to reach out to pharmaceutical scientists to help to solve the

complex formulation problems of vaccine adjuvants. In addition, we would like to encourage our fellow

pharmaceutical scientists to be more receptive to the challenges and demands of vaccine adjuvant

discovery and development. We believe that pharmaceutical scientists are uniquely positioned to solve

some fundamental problems in this area and can become a key part of the vaccine research and

development enterprise. The authors believe that formulation science is still under represented in

vaccinology, and there are many areas in which we could help. Pharmaceutical scientists could use

existing expertise to lead efforts in screening for new small molecules as adjuvants, or modify the

compounds appropriately to ensure that they have the right features for retention within the

formulation and effective co-delivery with the antigens. They could also lead efforts to design the next

delivery system for antigens and immune potentiators, or work on ensuring antigen stability and optimal

immunogenicity, or even facilitate vaccination by a preferred needle free route of administration. They

could also use existing expertise to determine which is the optimal release profile for immune

potentiators, or to target them to a preferred population of cells, to determine if this is indeed

advantageous. Since many of the challenges in vaccine delivery/formulation are similar to those for a

Page 35: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

variety of ‘drug delivery’ problems, particularly therapeutic proteins, there is already an enormous

wealth of experience waiting to be applied to solve the problems of vaccine delivery. We would like to

take this opportunity to encourage our colleagues both in the pharmaceutical sciences and in vaccines

R&D to engage more fully with one another, to use their respective experiences to help to design the

next generation of vaccines.

Although at this point, it is not yet clear which is the ‘best’ approach in the long term for vaccine

delivery, we would suggest that if we try to use the most established systems (liposomes, emulsions,

microparticles etc), we will get access to all the advances that have already been made for these

technologies for drug delivery. In addition, we will also get access to any future improvements, which is

an area of considerable investment in basic and applied science. Importantly, by focusing on these

established technologies, we potentially also get access to many of the talented minds who have already

worked on the key challenges, including scale up, manufacturing and the development of aseptic

processes etc. The recent success of AS04 adjuvant, including inclusion in a licensed vaccine in the US, is

perhaps a very good example of the advantages of ‘building on success’. This approach utilized a well-

established vaccine adjuvant platform (alum) and adsorbed the TLR4 agonist, to ensure that the

development risks were lowered. We believe that a similar approach can also be applied for other TLR

agonists and will bring many benefits to vaccine development. However, we also believe that there are a

number of alternative drug delivery systems which potentially have inherent advantages over alum and

may be adapted for vaccine delivery. We look forward to hopefully welcoming the endeavor of many

more skilled pharmaceutical scientists to this undertaking in the future.

Figure and table legends:

Tables 3a and b. Synergy between an particulate antigen delivery systems and immune potentiators

(TLR agonists)

We used a soluble recombinant antigen from Neisseria meningitides serotype B (Men B) to illustrate the

value of rationally combining different adjuvants to ensure effective co-delivery of an immune

potentiator (TLR agonist) with the vaccine antigen. In Table 3a, we show in a mouse study that the

soluble recombinant antigen is a typically poor immunogen alone, which was not significantly improved

either by the addition of a TLR9 agonist (CpG) or by conjugation to a protein based TLR5 agonist

(bacterial Flagellae). All of these approaches induced low level of binding antibodies and did not elicit

functional antibodies which can kill the bacteria, bactericidal antibodies. However, if the antigen is

adsorbed to a PLG microparticle, it becomes an effective immunogen, which can induce bactericidal

Page 36: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

antibodies, which are the correlate of protective immunity against this pathogen. Moreover, the titer

can be significantly enhanced if an immune potentiator (CpG) is then added to the formulation. In Table

3b, we take the co-delivery concept to the next level, showing combined delivery of the antigen and a

different TLR agonist, MPL (TLR4) with PLG. Like CpG, MPL can be added to the PLG microparticle with

adsorbed antigen to enhance the level of response, but the response is further enhanced if the PLG

microparticle also simultaneously delivers the antigen and the TLR agonist. In this situation, this was

best accomplished by adsorbing the antigen to the PLG, which already had entrapped MPL. Similar data

has been obtained with alternative delivery systems, including the better established Alum.

Figure 1:

Schematic representations of the concept of ‘reverse targeting’, a key concept in the mechanism of

action of MF59 o/w emulsion adjuvant. Antigen and MF59 emulsion adjuvant are administered into

muscle that does not contain resident immune cells (a). Adjuvant creates local immune competent

environment by interacting with cells to trigger release of recruitment factors (e.g. chemokines) (b).

Recruitment factors attract immune cells to the site of injection, immune cells take up antigen (c).

Adjuvant acts on recruited cells to enhance cellular recruitment (d). Additional cells infiltrate muscle,

take up additional antigen, and traffic to lymph node (e).

References:

[1] B.E. Correia, J.T. Bates, R.J. Loomis, G. Baneyx, C. Carrico, J.G. Jardine, P. Rupert, C. Correnti, O. Kalyuzhniy, V. Vittal, M.J. Connell, E. Stevens, A. Schroeter, M. Chen, S. Macpherson, A.M. Serra, Y. Adachi, M.A. Holmes, Y. Li, R.E. Klevit, B.S. Graham, R.T. Wyatt, D. Baker, R.K. Strong, J.E. Crowe, Jr., P.R. Johnson, W.R. Schief, Proof of principle for epitope-focused vaccine design, Nature, 507 (2014) 201-206. [2] J.S. McLellan, M. Chen, M.G. Joyce, M. Sastry, G.B. Stewart-Jones, Y. Yang, B. Zhang, L. Chen, S. Srivatsan, A. Zheng, T. Zhou, K.W. Graepel, A. Kumar, S. Moin, J.C. Boyington, G.Y. Chuang, C. Soto, U. Baxa, A.Q. Bakker, H. Spits, T. Beaumont, Z. Zheng, N. Xia, S.Y. Ko, J.P. Todd, S. Rao, B.S. Graham, P.D. Kwong, Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus, Science, 342 (2013) 592-598. [3] D. Maione, I. Margarit, C.D. Rinaudo, V. Masignani, M. Mora, M. Scarselli, H. Tettelin, C. Brettoni, E.T. Iacobini, R. Rosini, N. D'Agostino, L. Miorin, S. Buccato, M. Mariani, G. Galli, R. Nogarotto, V. Nardi-Dei, F. Vegni, C. Fraser, G. Mancuso, G. Teti, L.C. Madoff, L.C. Paoletti, R. Rappuoli, D.L. Kasper, J.L. Telford, G. Grandi, Identification of a universal Group B streptococcus vaccine by multiple genome screen, Science, 309 (2005) 148-150. [4] M. Pizza, V. Scarlato, V. Masignani, M.M. Giuliani, B. Arico, M. Comanducci, G.T. Jennings, L. Baldi, E. Bartolini, B. Capecchi, C.L. Galeotti, E. Luzzi, R. Manetti, E. Marchetti, M. Mora, S. Nuti, G. Ratti, L. Santini, S. Savino, M. Scarselli, E. Storni, P. Zuo, M. Broeker, E. Hundt, B. Knapp, E. Blair, T. Mason, H. Tettelin, D.W. Hood, A.C. Jeffries, N.J. Saunders, D.M. Granoff, J.C. Venter, E.R. Moxon, G. Grandi, R. Rappuoli, Identification of vaccine candidates against serogroup B meningococcus by whole-genome sequencing, Science, 287 (2000) 1816-1820.

Page 37: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

[5] D.T. O'Hagan, E. De Gregorio, The path to a successful vaccine adjuvant--'the long and winding road', Drug discovery today, 14 (2009) 541-551. [6] L.A. Brito, P. Malyala, D.T. O'Hagan, Vaccine adjuvant formulations: a pharmaceutical perspective, Seminars in immunology, 25 (2013) 130-145. [7] D.T. O'Hagan, G.S. Ott, G.V. Nest, R. Rappuoli, G.D. Giudice, The history of MF59((R)) adjuvant: a phoenix that arose from the ashes, Expert review of vaccines, 12 (2013) 13-30. [8] M. Kanekiyo, C.J. Wei, H.M. Yassine, P.M. McTamney, J.C. Boyington, J.R. Whittle, S.S. Rao, W.P. Kong, L. Wang, G.J. Nabel, Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies, Nature, 499 (2013) 102-106. [9] A. Roldao, M.C. Mellado, L.R. Castilho, M.J. Carrondo, P.M. Alves, Virus-like particles in vaccine development, Expert review of vaccines, 9 (2010) 1149-1176. [10] S.A. Plotkin, Correlates of protection induced by vaccination, Clinical and vaccine immunology : CVI, 17 (2010) 1055-1065. [11] D. Schenten, R. Medzhitov, The control of adaptive immune responses by the innate immune system, Advances in immunology, 109 (2011) 87-124. [12] E. Meylan, J. Tschopp, M. Karin, Intracellular pattern recognition receptors in the host response, Nature, 442 (2006) 39-44. [13] E.M. Moresco, D. LaVine, B. Beutler, Toll-like receptors, Current biology : CB, 21 (2011) R488-493. [14] B. Beutler, A.J. Hoffman, M. Steinman, The Nobel Prize in Physiology or Medicine 2011, in, 2011. [15] D.R. Flower, Systematic identification of small molecule adjuvants, Expert opinion on drug discovery, 7 (2012) 807-817. [16] N.W. Baylor, W. Egan, P. Richman, Aluminum salts in vaccines--US perspective, Vaccine, 20 Suppl 3 (2002) S18-23. [17] H. Hogenesch, Mechanism of immunopotentiation and safety of aluminum adjuvants, Frontiers in immunology, 3 (2012) 406. [18] N. Garcon, S. Morel, A. Didierlaurent, D. Descamps, M. Wettendorff, M. Van Mechelen, Development of an AS04-adjuvanted HPV vaccine with the adjuvant system approach, BioDrugs : clinical immunotherapeutics, biopharmaceuticals and gene therapy, 25 (2011) 217-226. [19] A.T. Glenny, C.G. Pope, The antigenic effect of intravenous injection of diphtheria toxin, J. Pathol. Bacteriol., 28 (1925) 273-278. [20] V.K. Volk, W.E. Bunney, Diphtheria Immunization with Fluid Toxoid and Alum-Precipitated Toxoid, American journal of public health and the nation's health, 32 (1942) 690-699. [21] F.G. Jones, J.H. Moss, The Antitoxic Titer of Human Subjects Following Immunization with Tetanus Toxoid and Tetanus Alum Precipitated Toxoid, J Immonol, 30 (1936) 115-125. [22] C.T. Johnston, S.L. Wang, S.L. Hem, Measuring the surface area of aluminum hydroxide adjuvant, Journal of pharmaceutical sciences, 91 (2002) 1702-1706. [23] M.J. Caulfield, L. Shi, S. Wang, B. Wang, T.W. Tobery, H. Mach, P.L. Ahl, J.L. Cannon, J.C. Cook, J.H. Heinrichs, R.D. Sitrin, Effect of alternative aluminum adjuvants on the absorption and immunogenicity of HPV16 L1 VLPs in mice, Human vaccines, 3 (2007) 139-145. [24] D.W. Cain, S.E. Sanders, M.M. Cunningham, G. Kelsoe, Disparate adjuvant properties among three formulations of "alum", Vaccine, 31 (2013) 653-660. [25] S.L. Hem, H. Hogenesch, Relationship between physical and chemical properties of aluminum-containing adjuvants and immunopotentiation, Expert review of vaccines, 6 (2007) 685-698. [26] B. Hansen, M. Belfast, G. Soung, L. Song, P.M. Egan, R. Capen, H. Hogenesch, R. Mancinelli, S.L. Hem, Effect of the strength of adsorption of hepatitis B surface antigen to aluminum hydroxide adjuvant on the immune response, Vaccine, 27 (2009) 888-892.

Page 38: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

[27] M. Regnier, B. Metz, W. Tilstra, C. Hendriksen, W. Jiskoot, W. Norde, G. Kersten, Structural perturbation of diphtheria toxoid upon adsorption to aluminium hydroxide adjuvant, Vaccine, 30 (2012) 6783-6788. [28] S. Vecchi, S. Bufali, D.A. Skibinski, D.T. O'Hagan, M. Singh, Aluminum adjuvant dose guidelines in vaccine formulation for preclinical evaluations, Journal of pharmaceutical sciences, 101 (2012) 17-20. [29] M. Ugozzoli, D. Laera, S. Nuti, D.A. Skibinski, S. Bufali, C. Sammicheli, S. Tavarini, M. Singh, D.T. O'Hagan, Flow cytometry: an alternative method for direct quantification of antigens adsorbed to aluminum hydroxide adjuvant, Analytical biochemistry, 418 (2011) 224-230. [30] L.J. Peek, T.T. Martin, C. Elk Nation, S.A. Pegram, C.R. Middaugh, Effects of stabilizers on the destabilization of proteins upon adsorption to aluminum salt adjuvants, Journal of pharmaceutical sciences, 96 (2007) 547-557. [31] S.F. Ausar, J. Chan, W. Hoque, O. James, K. Jayasundara, K. Harper, Application of extrinsic fluorescence spectroscopy for the high throughput formulation screening of aluminum-adjuvanted vaccines, Journal of pharmaceutical sciences, 100 (2011) 431-440. [32] J.B. Ulmer, C.M. DeWitt, M. Chastain, A. Friedman, J.J. Donnelly, W.L. McClements, M.J. Caulfield, K.E. Bohannon, D.B. Volkin, R.K. Evans, Enhancement of DNA vaccine potency using conventional aluminum adjuvants, Vaccine, 18 (1999) 18-28. [33] P. Marrack, A.S. McKee, M.W. Munks, Towards an understanding of the adjuvant action of aluminium, Nature reviews. Immunology, 9 (2009) 287-293. [34] M. Kool, K. Fierens, B.N. Lambrecht, Alum adjuvant: some of the tricks of the oldest adjuvant, Journal of medical microbiology, 61 (2012) 927-934. [35] E. De Gregorio, E. Caproni, J.B. Ulmer, Vaccine adjuvants: mode of action, Frontiers in immunology, 4 (2013) 214. [36] A.T. Glenny, G.A.H. Buttle, M.F. Stevens) Rate of disappearance of diphtheria toxoid injected into rabbits and guinea-pigs: toxoid precipitated with alum, J. Pathol. Bacteriol., 34 (1931) 267-275. [37] W.T. Harrison, Some Observations on the Use of Alum Precipitated Diphtheria Toxoid, American journal of public health and the nation's health, 25 (1935) 298-300. [38] L.B. Holt, Quantitative studies in diphtheria prophylaxis: the second response, British journal of experimental pathology, 31 (1950) 233-241. [39] R.G. White, A.H. Coons, J.M. Connolly, Studies on antibody production. III. The alum granuloma, The Journal of experimental medicine, 102 (1955) 73-82. [40] R.K. Gupta, A.C. Chang, P. Griffin, R. Rivera, G.R. Siber, In vivo distribution of radioactivity in mice after injection of biodegradable polymer microspheres containing 14C-labeled tetanus toxoid, Vaccine, 14 (1996) 1412-1416. [41] S. Hutchison, R. Benson, V. Gibson, A. Pollock, P. Garside, Brewer.J., , Antigen depot is not required for alum adjuvanticity, FASEB, 26 (2012) 1272-1279 [42] F. Verdier, R. Burnett, C. Michelet-Habchi, P. Moretto, F. Fievet-Groyne, E. Sauzeat, Aluminium assay and evaluation of the local reaction at several time points after intramuscular administration of aluminium containing vaccines in the Cynomolgus monkey, Vaccine, 23 (2005) 1359-1367. [43] J.L. Leeling, I.A. Muni, R.J. Helms, N. Johnson, Jr., Rates of release of subcutaneously injected antigens in the rat. Comparison of an aqueous preparation with two alum-precipated preparations, Allergy, 34 (1979) 339-344. [44] S.M. Noe, M.A. Green, H. HogenEsch, S.L. Hem, Mechanism of immunopotentiation by aluminum-containing adjuvants elucidated by the relationship between antigen retention at the inoculation site and the immune response, Vaccine, 28 (2010) 3588-3594. [45] N. Goto, H. Kato, J. Maeyama, M. Shibano, T. Saito, J. Yamaguchi, S. Yoshihara, Local tissue irritating effects and adjuvant activities of calcium phosphate and aluminium hydroxide with different physical properties, Vaccine, 15 (1997) 1364-1371.

Page 39: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

[46] M. Kool, T. Soullie, M. van Nimwegen, M.A. Willart, F. Muskens, S. Jung, H.C. Hoogsteden, H. Hammad, B.N. Lambrecht, Alum adjuvant boosts adaptive immunity by inducing uric acid and activating inflammatory dendritic cells, The Journal of experimental medicine, 205 (2008) 869-882. [47] A.S. McKee, M. MacLeod, J. White, F. Crawford, J.W. Kappler, P. Marrack, Gr1+IL-4-producing innate cells are induced in response to Th2 stimuli and suppress Th1-dependent antibody responses, International immunology, 20 (2008) 659-669. [48] S. Calabro, M. Tortoli, B.C. Baudner, A. Pacitto, M. Cortese, D.T. O'Hagan, E. De Gregorio, A. Seubert, A. Wack, Vaccine adjuvants alum and MF59 induce rapid recruitment of neutrophils and monocytes that participate in antigen transport to draining lymph nodes, Vaccine, 29 (2011) 1812-1823. [49] J.W. Mannhalter, H.O. Neychev, G.J. Zlabinger, R. Ahmad, M.M. Eibl, Modulation of the human immune response by the non-toxic and non-pyrogenic adjuvant aluminium hydroxide: effect on antigen uptake and antigen presentation, Clinical and experimental immunology, 61 (1985) 143-151. [50] M. Ulanova, A. Tarkowski, M. Hahn-Zoric, L.A. Hanson, The Common vaccine adjuvant aluminum hydroxide up-regulates accessory properties of human monocytes via an interleukin-4-dependent mechanism, Infection and immunity, 69 (2001) 1151-1159. [51] A.C. Rimaniol, G. Gras, F. Verdier, F. Capel, V.B. Grigoriev, F. Porcheray, E. Sauzeat, J.G. Fournier, P. Clayette, C.A. Siegrist, D. Dormont, Aluminum hydroxide adjuvant induces macrophage differentiation towards a specialized antigen-presenting cell type, Vaccine, 22 (2004) 3127-3135. [52] R.K. Gupta, Aluminum compounds as vaccine adjuvants, Advanced drug delivery reviews, 32 (1998) 155-172. [53] G.L. Morefield, A. Sokolovska, D. Jiang, H. HogenEsch, J.P. Robinson, S.L. Hem, Role of aluminum-containing adjuvants in antigen internalization by dendritic cells in vitro, Vaccine, 23 (2005) 1588-1595. [54] F. Mosca, E. Tritto, A. Muzzi, E. Monaci, F. Bagnoli, C. Iavarone, D. O'Hagan, R. Rappuoli, E. De Gregorio, Molecular and cellular signatures of human vaccine adjuvants, Proceedings of the National Academy of Sciences of the United States of America, 105 (2008) 10501-10506. [55] A.S. McKee, M.W. Munks, M.K. MacLeod, C.J. Fleenor, N. Van Rooijen, J.W. Kappler, P. Marrack, Alum induces innate immune responses through macrophage and mast cell sensors, but these sensors are not required for alum to act as an adjuvant for specific immunity, J Immunol, 183 (2009) 4403-4414. [56] M.F. Bachmann, G.T. Jennings, Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns, Nature reviews. Immunology, 10 (2010) 787-796. [57] H. Liu, K.D. Moynihan, Y. Zheng, G.L. Szeto, A.V. Li, B. Huang, D.S. Van Egeren, C. Park, D.J. Irvine, Structure-based programming of lymph-node targeting in molecular vaccines, Nature, 507 (2014) 519-522. [58] A. Seubert, E. Monaci, M. Pizza, D.T. O'Hagan, A. Wack, The adjuvants aluminum hydroxide and MF59 induce monocyte and granulocyte chemoattractants and enhance monocyte differentiation toward dendritic cells, J Immunol, 180 (2008) 5402-5412. [59] N. Goto, K. Akama, Histopathological studies of reactions in mice injected with aluminum-adsorbed tetanus toxoid, Microbiology and immunology, 26 (1982) 1121-1132. [60] R.S. Walls, Eosinophil response to alum adjuvants: involvement of T cells in non-antigen-dependent mechanisms, Proc Soc Exp Biol Med, 156 (1977) 431-435. [61] V.D. Ramanathan, P. Badenoch-Jones, J.L. Turk, Complement activation by aluminium and zirconium compounds, Immunology, 37 (1979) 881-888. [62] H. Li, S. Nookala, F. Re, Aluminum hydroxide adjuvants activate caspase-1 and induce IL-1beta and IL-18 release, J Immunol, 178 (2007) 5271-5276. [63] A. Sokolovska, S.L. Hem, H. HogenEsch, Activation of dendritic cells and induction of CD4(+) T cell differentiation by aluminum-containing adjuvants, Vaccine, 25 (2007) 4575-4585. [64] A.L. Gavin, K. Hoebe, B. Duong, T. Ota, C. Martin, B. Beutler, D. Nemazee, Adjuvant-enhanced antibody responses in the absence of toll-like receptor signaling, Science, 314 (2006) 1936-1938.

Page 40: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

[65] F.A. Sharp, D. Ruane, B. Claass, E. Creagh, J. Harris, P. Malyala, M. Singh, D.T. O'Hagan, V. Petrilli, J. Tschopp, L.A. O'Neill, E.C. Lavelle, Uptake of particulate vaccine adjuvants by dendritic cells activates the NALP3 inflammasome, Proceedings of the National Academy of Sciences of the United States of America, 106 (2009) 870-875. [66] E. De Gregorio, E. Tritto, R. Rappuoli, Alum adjuvanticity: unraveling a century old mystery, European journal of immunology, 38 (2008) 2068-2071. [67] T. Marichal, K. Ohata, D. Bedoret, C. Mesnil, C. Sabatel, K. Kobiyama, P. Lekeux, C. Coban, S. Akira, K.J. Ishii, F. Bureau, C.J. Desmet, DNA released from dying host cells mediates aluminum adjuvant activity, Nature medicine, 17 (2011) 996-1002. [68] N. Riteau, L. Baron, B. Villeret, N. Guillou, F. Savigny, B. Ryffel, F. Rassendren, M. Le Bert, A. Gombault, I. Couillin, ATP release and purinergic signaling: a common pathway for particle-mediated inflammasome activation, Cell death & disease, 3 (2012) e403. [69] Y. Wang, D. Rahman, T. Lehner, A comparative study of stress-mediated immunological functions with the adjuvanticity of alum, The Journal of biological chemistry, 287 (2012) 17152-17160. [70] T.L. Flach, G. Ng, A. Hari, M.D. Desrosiers, P. Zhang, S.M. Ward, M.E. Seamone, A. Vilaysane, A.D. Mucsi, Y. Fong, E. Prenner, C.C. Ling, J. Tschopp, D.A. Muruve, M.W. Amrein, Y. Shi, Alum interaction with dendritic cell membrane lipids is essential for its adjuvanticity, Nature medicine, 17 (2011) 479-487. [71] P. Matzinger, Tolerance, Danger, and the Extended Family, Annu Rev Immunol, 12 (1994) 991-1045. [72] M.E. Bianchi, DAMPs, PAMPs and alarmins: all we need to know about danger, Journal of leukocyte biology, 81 (2007) 1-5. [73] N. Goto, H. Kato, J. Maeyama, M. Shibano, T. Saito, J. Yamaguchi, S. Yoshihara, Local tissue irritating effects and adjuvant activities of calcium phosphate and aluminium hydroxide with different physical properties, Vaccine, 15 (1997) 1364-1371. [74] V. Aimanianda, J. Haensler, S. Lacroix-Desmazes, S.V. Kaveri, J. Bayry, Novel cellular and molecular mechanisms of induction of immune responses by aluminum adjuvants, Trends Pharmacol Sci, 30 (2009) 287-295. [75] N. Qureshi, K. Takayama, E. Ribi, Purification and structural determination of nontoxic lipid A obtained from the lipopolysaccharide of Salmonella typhimurium, The Journal of biological chemistry, 257 (1982) 11808-11815. [76] A.M. Didierlaurent, S. Morel, L. Lockman, S.L. Giannini, M. Bisteau, H. Carlsen, A. Kielland, O. Vosters, N. Vanderheyde, F. Schiavetti, D. Larocque, M. Van Mechelen, N. Garcon, AS04, an aluminum salt- and TLR4 agonist-based adjuvant system, induces a transient localized innate immune response leading to enhanced adaptive immunity, J Immunol, 183 (2009) 6186-6197. [77] G.E. Mullen, R.D. Ellis, K. Miura, E. Malkin, C. Nolan, M. Hay, M.P. Fay, A. Saul, D. Zhu, K. Rausch, S. Moretz, H. Zhou, C.A. Long, L.H. Miller, J. Treanor, Phase 1 trial of AMA1-C1/Alhydrogel plus CPG 7909: an asexual blood-stage vaccine for Plasmodium falciparum malaria, PloS one, 3 (2008) e2940. [78] J.A. Aebig, G.E. Mullen, G. Dobrescu, K. Rausch, L. Lambert, O. Ajose-Popoola, C.A. Long, A. Saul, A.P. Miles, Formulation of vaccines containing CpG oligonucleotides and alum, Journal of immunological methods, 323 (2007) 139-146. [79] R.L. Coffman, A. Sher, R.A. Seder, Vaccine adjuvants: putting innate immunity to work, Immunity, 33 (2010) 492-503. [80] H. Hemmi, T. Kaisho, O. Takeuchi, S. Sato, H. Sanjo, K. Hoshino, T. Horiuchi, H. Tomizawa, K. Takeda, S. Akira, Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway, Nature immunology, 3 (2002) 196-200. [81] N.M. Valiante, Designing small molecules as optimal vaccine adjuvants, In preparation, (2014). [82] J.P. Vasilakos, M.A. Tomai, The use of Toll-like receptor 7/8 agonists as vaccine adjuvants, Expert review of vaccines, 12 (2013) 809-819.

Page 41: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

[83] M.R. Hilleman, Critical appraisal of emulsified oil adjuvants applied to viral vaccines, Progress in medical virology. Fortschritte der medizinischen Virusforschung. Progres en virologie medicale, 8 (1966) 131-182. [84] T. Vesikari, M. Knuf, P. Wutzler, A. Karvonen, D. Kieninger-Baum, H.J. Schmitt, F. Baehner, A. Borkowski, T.F. Tsai, R. Clemens, Oil-in-water emulsion adjuvant with influenza vaccine in young children, The New England journal of medicine, 365 (2011) 1406-1416. [85] H. Nohynek, J. Jokinen, M. Partinen, O. Vaarala, T. Kirjavainen, J. Sundman, S.L. Himanen, C. Hublin, I. Julkunen, P. Olsen, O. Saarenpaa-Heikkila, T. Kilpi, AS03 adjuvanted AH1N1 vaccine associated with an abrupt increase in the incidence of childhood narcolepsy in Finland, PloS one, 7 (2012) e33536. [86] T.F. Tsai, A. Crucitti, P. Nacci, U. Nicolay, G. Della Cioppa, J. Ferguson, R. Clemens, Explorations of clinical trials and pharmacovigilance databases of MF59(R)-adjuvanted influenza vaccines for associated cases of narcolepsy, Scandinavian journal of infectious diseases, 43 (2011) 702-706. [87] S. Morel, A. Didierlaurent, P. Bourguignon, S. Delhaye, B. Baras, V. Jacob, C. Planty, A. Elouahabi, P. Harvengt, H. Carlsen, A. Kielland, P. Chomez, N. Garcon, M. Van Mechelen, Adjuvant System AS03 containing alpha-tocopherol modulates innate immune response and leads to improved adaptive immunity, Vaccine, 29 (2011) 2461-2473. [88] D.T. O'Hagan, G.S. Ott, E. De Gregorio, A. Seubert, The mechanism of action of MF59 - an innately attractive adjuvant formulation, Vaccine, 30 (2012) 4341-4348. [89] G. Ott, G.L. Barchfeld, D. Chernoff, R. Radhakrishnan, P. van Hoogevest, G. Van Nest, MF59. Design and evaluation of a safe and potent adjuvant for human vaccines, Pharmaceutical biotechnology, 6 (1995) 277-296. [90] M. Dupuis, D.M. McDonald, G. Ott, Distribution of adjuvant MF59 and antigen gD2 after intramuscular injection in mice, Vaccine, 18 (1999) 434-439. [91] M. Vono, M. Taccone, P. Caccin, M. Gallotta, G. Donvito, S. Falzoni, E. Palmieri, M. Pallaoro, R. Rappuoli, F. Di Virgilio, E. De Gregorio, C. Montecucco, A. Seubert, The adjuvant MF59 induces ATP release from muscle that potentiates response to vaccination, Proceedings of the National Academy of Sciences of the United States of America, 110 (2013) 21095-21100. [92] S. Calabro, E. Tritto, A. Pezzotti, M. Taccone, A. Muzzi, S. Bertholet, E. De Gregorio, D.T. O'Hagan, B. Baudner, A. Seubert, The adjuvant effect of MF59 is due to the oil-in-water emulsion formulation, none of the individual components induce a comparable adjuvant effect, Vaccine, 31 (2013) 3363-3369. [93] R.C. Anderson, C.B. Fox, T.S. Dutill, N. Shaverdian, T.L. Evers, G.R. Poshusta, J. Chesko, R.N. Coler, M. Friede, S.G. Reed, T.S. Vedvick, Physicochemical characterization and biological activity of synthetic TLR4 agonist formulations, Colloids and surfaces. B, Biointerfaces, 75 (2010) 123-132. [94] R.N. Coler, S. Bertholet, M. Moutaftsi, J.A. Guderian, H.P. Windish, S.L. Baldwin, E.M. Laughlin, M.S. Duthie, C.B. Fox, D. Carter, M. Friede, T.S. Vedvick, S.G. Reed, Development and characterization of synthetic glucopyranosyl lipid adjuvant system as a vaccine adjuvant, PloS one, 6 (2011) e16333. [95] M.A. Arias, G.A. Van Roey, J.S. Tregoning, M. Moutaftsi, R.N. Coler, H.P. Windish, S.G. Reed, D. Carter, R.J. Shattock, Glucopyranosyl Lipid Adjuvant (GLA), a Synthetic TLR4 agonist, promotes potent systemic and mucosal responses to intranasal immunization with HIVgp140, PloS one, 7 (2012) e41144. [96] C.H. Clegg, R. Roque, N. Van Hoeven, L. Perrone, S.L. Baldwin, J.A. Rininger, R.A. Bowen, S.G. Reed, Adjuvant solution for pandemic influenza vaccine production, Proceedings of the National Academy of Sciences of the United States of America, 109 (2012) 17585-17590. [97] M.T. Orr, C.B. Fox, S.L. Baldwin, S.J. Sivananthan, E. Lucas, S. Lin, T. Phan, J.J. Moon, T.S. Vedvick, S.G. Reed, R.N. Coler, Adjuvant formulation structure and composition are critical for the development of an effective vaccine against tuberculosis, Journal of controlled release : official journal of the Controlled Release Society, 172 (2013) 190-200. [98] C.B. Fox, S.L. Baldwin, M.S. Duthie, S.G. Reed, T.S. Vedvick, Immunomodulatory and physical effects of oil composition in vaccine adjuvant emulsions, Vaccine, 29 (2011) 9563-9572.

Page 42: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

[99] S. Lousada-Dietrich, P.S. Jogdand, S. Jepsen, V.V. Pinto, S.B. Ditlev, M. Christiansen, S.O. Larsen, C.B. Fox, V.S. Raman, R.F. Howard, T.S. Vedvick, G. Ireton, D. Carter, S.G. Reed, M. Theisen, A synthetic TLR4 agonist formulated in an emulsion enhances humoral and Type 1 cellular immune responses against GMZ2--a GLURP-MSP3 fusion protein malaria vaccine candidate, Vaccine, 29 (2011) 3284-3292. [100] S.L. Baldwin, N. Shaverdian, Y. Goto, M.S. Duthie, V.S. Raman, T. Evers, F. Mompoint, T.S. Vedvick, S. Bertholet, R.N. Coler, S.G. Reed, Enhanced humoral and Type 1 cellular immune responses with Fluzone adjuvanted with a synthetic TLR4 agonist formulated in an emulsion, Vaccine, 27 (2009) 5956-5963. [101] M.S. Duthie, L.H. Sampaio, R.M. Oliveira, V.S. Raman, J. O'Donnell, H.R. Bailor, G.C. Ireton, A.L. Sousa, M.M. Stefani, S.G. Reed, Development and pre-clinical assessment of a 73 kD chimeric fusion protein as a defined sub-unit vaccine for leprosy, Vaccine, 31 (2013) 813-819. [102] A. Wack, B.C. Baudner, A.K. Hilbert, I. Manini, S. Nuti, S. Tavarini, H. Scheffczik, M. Ugozzoli, M. Singh, J. Kazzaz, E. Montomoli, G. Del Giudice, R. Rappuoli, D.T. O'Hagan, Combination adjuvants for the induction of potent, long-lasting antibody and T-cell responses to influenza vaccine in mice, Vaccine, 26 (2008) 552-561. [103] B.C. Baudner, V. Ronconi, D. Casini, M. Tortoli, J. Kazzaz, M. Singh, L.D. Hawkins, A. Wack, D.T. O'Hagan, MF59 emulsion is an effective delivery system for a synthetic TLR4 agonist (E6020), Pharmaceutical research, 26 (2009) 1477-1485. [104] G. Ott, M. Singh, J. Kazzaz, M. Briones, E. Soenawan, M. Ugozzoli, D.T. O'Hagan, A cationic sub-micron emulsion (MF59/DOTAP) is an effective delivery system for DNA vaccines, Journal of controlled release : official journal of the Controlled Release Society, 79 (2002) 1-5. [105] D.T. O'Hagan, M. Singh, J. Kazzaz, M. Ugozzoli, M. Briones, J. Donnelly, G. Ott, Synergistic adjuvant activity of immunostimulatory DNA and oil/water emulsions for immunization with HIV p55 gag antigen, Vaccine, 20 (2002) 3389-3398. [106] L. Brito, M. Chan, C. Shaw, A. Hekele, T. Carsillo, M. Schafer, J. Archer, A. Seubert, G. Otten, C. Beard, A. Dey, A.E. Lilja, N.M. Valiante, P. Mason, C.W. Mandl, S. Barnett, P.R. Dormitzer, J.B. Ulmer, M. Singh, D. O'Hagan, A. Geall, A cationic nanoemulsion for the delivery of next generation RNA vaccines Under revision, (2014). [107] V. Filipe, A. Hawe, W. Jiskoot, Critical evaluation of Nanoparticle Tracking Analysis (NTA) by NanoSight for the measurement of nanoparticles and protein aggregates, Pharmaceutical research, 27 (2010) 796-810. [108] V. Klang, C. Valenta, N.B. Matsko, Analytical electron microscopy for characterization of fluid or semi-solid multiphase systems containing nanoparticulate material, Pharmaceutics, 5 (2013) 115-126. [109] A.G. Allison, G. Gregoriadis, Liposomes as immunological adjuvants, Nature, 252 (1974) 252. [110] S. Pichyangkul, M. Gettayacamin, R.S. Miller, J.A. Lyon, E. Angov, P. Tongtawe, D.L. Ruble, D.G. Heppner, Jr., K.E. Kester, W.R. Ballou, C.L. Diggs, G. Voss, J.D. Cohen, D.S. Walsh, Pre-clinical evaluation of the malaria vaccine candidate P. falciparum MSP1(42) formulated with novel adjuvants or with alum, Vaccine, 22 (2004) 3831-3840. [111] J. Davidsen, I. Rosenkrands, D. Christensen, A. Vangala, D. Kirby, Y. Perrie, E.M. Agger, P. Andersen, Characterization of cationic liposomes based on dimethyldioctadecylammonium and synthetic cord factor from M. tuberculosis (trehalose 6,6'-dibehenate)-a novel adjuvant inducing both strong CMI and antibody responses, Biochimica et biophysica acta, 1718 (2005) 22-31. [112] B. Banerji, C.R. Alving, Lipid A from endotoxin: antigenic activities of purified fractions in liposomes, J Immunol, 123 (1979) 2558-2562. [113] K.E. Kester, J.F. Cummings, O. Ofori-Anyinam, C.F. Ockenhouse, U. Krzych, P. Moris, R. Schwenk, R.A. Nielsen, Z. Debebe, E. Pinelis, L. Juompan, J. Williams, M. Dowler, V.A. Stewart, R.A. Wirtz, M.C. Dubois, M. Lievens, J. Cohen, W.R. Ballou, D.G. Heppner, Jr., Randomized, double-blind, phase 2a trial of

Page 43: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

falciparum malaria vaccines RTS,S/AS01B and RTS,S/AS02A in malaria-naive adults: safety, efficacy, and immunologic associates of protection, The Journal of infectious diseases, 200 (2009) 337-346. [114] J.A. Stoute, M. Slaoui, D.G. Heppner, P. Momin, K.E. Kester, P. Desmons, B.T. Wellde, N. Garcon, U. Krzych, M. Marchand, A preliminary evaluation of a recombinant circumsporozoite protein vaccine against Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation Group, The New England journal of medicine, 336 (1997) 86-91. [115] P. Vandepapeliere, Vaccine compositions comprising a saponin adjuvant., in: 13/020 (Ed.) US patent application No. 2011 Feb 3; Publ No. US 20/0206758, 25 Aug 2011, US patent application, 2011. [116] J.A. Regules, J.F. Cummings, C.F. Ockenhouse, The RTS,S vaccine candidate for malaria, Expert review of vaccines, 10 (2011) 589-599. [117] A.J. Birkett, PATH Malaria Vaccine Initiative (MVI) Perspectives on the status of malaria vaccine development, Human vaccines, 6 (2010) 139-145. [118] C.R. Kensil, R. Kammer, QS-21: a water-soluble triterpene glycoside adjuvant, Expert opinion on investigational drugs, 7 (1998) 1475-1482. [119] P.M. Momin, N. Garcon, Vaccines, in: 08/663 (Ed.) US patent application No. 20 Dec 1994, 1994. [120] C.B. Fox, M. Moutaftsi, J. Vergara, A.L. Desbien, G.I. Nana, T.S. Vedvick, R.N. Coler, S.G. Reed, TLR4 ligand formulation causes distinct effects on antigen-specific cell-mediated and humoral immune responses, Vaccine, 31 (2013) 5848-5855. [121] J. Szebeni, P. Bedocs, Z. Rozsnyay, Z. Weiszhar, R. Urbanics, L. Rosivall, R. Cohen, O. Garbuzenko, G. Bathori, M. Toth, R. Bunger, Y. Barenholz, Liposome-induced complement activation and related cardiopulmonary distress in pigs: factors promoting reactogenicity of Doxil and AmBisome, Nanomedicine : nanotechnology, biology, and medicine, 8 (2012) 176-184. [122] D.S. Watson, A.N. Endsley, L. Huang, Design considerations for liposomal vaccines: influence of formulation parameters on antibody and cell-mediated immune responses to liposome associated antigens, Vaccine, 30 (2012) 2256-2272. [123] E. Shahum, H.M. Therien, Immunopotentiation of the humoral response by liposomes: encapsulation versus covalent linkage, Immunology, 65 (1988) 315-317. [124] D. Christensen, E.M. Agger, L.V. Andreasen, D. Kirby, P. Andersen, Y. Perrie, Liposome-based cationic adjuvant formulations (CAF): past, present, and future, Journal of liposome research, 19 (2009) 2-11. [125] J.J. Moon, H. Suh, A. Bershteyn, M.T. Stephan, H. Liu, B. Huang, M. Sohail, S. Luo, S.H. Um, H. Khant, J.T. Goodwin, J. Ramos, W. Chiu, D.J. Irvine, Interbilayer-crosslinked multilamellar vesicles as synthetic vaccines for potent humoral and cellular immune responses, Nature materials, 10 (2011) 243-251. [126] V. D'Acremont, C. Herzog, B. Genton, Immunogenicity and safety of a virosomal hepatitis A vaccine (Epaxal) in the elderly, Journal of travel medicine, 13 (2006) 78-83. [127] R. Gluck, R. Mischler, S. Brantschen, M. Just, B. Althaus, S.J. Cryz, Jr., Immunopotentiating reconstituted influenza virus virosome vaccine delivery system for immunization against hepatitis A, The Journal of clinical investigation, 90 (1992) 2491-2495. [128] V. Baldo, T. Baldovin, M. Pellegrini, G. Angiolelli, S. Majori, A. Floreani, M.C. Busana, C. Bertoncello, R. Trivello, Immunogenicity of three different influenza vaccines against homologous and heterologous strains in nursing home elderly residents, Clinical & developmental immunology, 2010 (2010) 517198. [129] P.A. Bovier, Epaxal: a virosomal vaccine to prevent hepatitis A infection, Expert review of vaccines, 7 (2008) 1141-1150. [130] L. Bungener, K. Serre, L. Bijl, L. Leserman, J. Wilschut, T. Daemen, P. Machy, Virosome-mediated delivery of protein antigens to dendritic cells, Vaccine, 20 (2002) 2287-2295. [131] S.D. Litwin, J.M. Singer, The adjuvant action of latex particulate carriers, J Immunol, 95 (1965) 1147-1152.

Page 44: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

[132] S.J. Challacombe, D. Rahman, H. Jeffery, S.S. Davis, D.T. O'Hagan, Enhanced secretory IgA and systemic IgG antibody responses after oral immunization with biodegradable microparticles containing antigen, Immunology, 76 (1992) 164-168. [133] I. Preis, R.S. Langer, A single-step immunization by sustained antigen release, Journal of immunological methods, 28 (1979) 193-197. [134] A.S. Hoffman, The origins and evolution of "controlled" drug delivery systems, Journal of controlled release : official journal of the Controlled Release Society, 132 (2008) 153-163. [135] S. Jain, D.T. O'Hagan, M. Singh, The long-term potential of biodegradable poly(lactide-co-glycolide) microparticles as the next-generation vaccine adjuvant, Expert review of vaccines, 10 (2011) 1731-1742. [136] V. Milacic, A.B. Bailey, D. O'Hagan, P. Schwendeman, Injectable PLGA Systems for Delivery of Vaccine in: Long Acting Injections and Implants., Advances in Delivery Science and Technology, 2012, pp. 429. [137] M. Singh, J. Kazzaz, J. Chesko, E. Soenawan, M. Ugozzoli, M. Giuliani, M. Pizza, R. Rappouli, D.T. O'Hagan, Anionic microparticles are a potent delivery system for recombinant antigens from Neisseria meningitidis serotype B, Journal of pharmaceutical sciences, 93 (2004a) 273-282. [138] M. Singh, J. Chesko, J. Kazzaz, M. Ugozzoli, E. Kan, I. Srivastava, D.T. O'Hagan, Adsorption of a novel recombinant glycoprotein from HIV (Env gp120dV2 SF162) to anionic PLG microparticles retains the structural integrity of the protein, whereas encapsulation in PLG microparticles does not, Pharmaceutical research, 21 (2004b) 2148-2152. [139] J. Kazzaz, M. Singh, M. Ugozzoli, J. Chesko, E. Soenawan, D.T. O'Hagan, Encapsulation of the immune potentiators MPL and RC529 in PLG microparticles enhances their potency, Journal of controlled release : official journal of the Controlled Release Society, 110 (2006) 566-573. [140] H. Xie, I. Gursel, B.E. Ivins, M. Singh, D.T. O'Hagan, J.B. Ulmer, D.M. Klinman, CpG oligodeoxynucleotides adsorbed onto polylactide-co-glycolide microparticles improve the immunogenicity and protective activity of the licensed anthrax vaccine, Infection and immunity, 73 (2005) 828-833. [141] S. Jain, P. Malyala, M. Pallaoro, M. Giuliani, H. Petersen, D.T. O'Hagan, M. Singh, A two-stage strategy for sterilization of poly(lactide-co-glycolide) particles by gamma-irradiation does not impair their potency for vaccine delivery, Journal of pharmaceutical sciences, 100 (2011) 646-654. [142] A. Moore, P. McGuirk, S. Adams, W.C. Jones, J.P. McGee, D.T. O'Hagan, K.H. Mills, Immunization with a soluble recombinant HIV protein entrapped in biodegradable microparticles induces HIV-specific CD8+ cytotoxic T lymphocytes and CD4+ Th1 cells, Vaccine, 13 (1995) 1741-1749. [143] G.R. Otten, M. Schaefer, B. Doe, H. Liu, I. Srivastava, J. Megede, J. Kazzaz, Y. Lian, M. Singh, M. Ugozzoli, D. Montefiori, M. Lewis, D.A. Driver, T. Dubensky, J.M. Polo, J. Donnelly, D.T. O'Hagan, S. Barnett, J.B. Ulmer, Enhanced potency of plasmid DNA microparticle human immunodeficiency virus vaccines in rhesus macaques by using a priming-boosting regimen with recombinant proteins, Journal of virology, 79 (2005) 8189-8200. [144] S.P. Kasturi, I. Skountzou, R.A. Albrecht, D. Koutsonanos, T. Hua, H.I. Nakaya, R. Ravindran, S. Stewart, M. Alam, M. Kwissa, F. Villinger, N. Murthy, J. Steel, J. Jacob, R.J. Hogan, A. Garcia-Sastre, R. Compans, B. Pulendran, Programming the magnitude and persistence of antibody responses with innate immunity, Nature, 470 (2011) 543-547. [145] J. Wendorf, M. Singh, J. Chesko, J. Kazzaz, E. Soewanan, M. Ugozzoli, D. O'Hagan, A practical approach to the use of nanoparticles for vaccine delivery, Journal of pharmaceutical sciences, 95 (2006) 2738-2750. [146] J. Wendorf, J. Chesko, J. Kazzaz, M. Ugozzoli, M. Vajdy, D. O'Hagan, M. Singh, A comparison of anionic nanoparticles and microparticles as vaccine delivery systems, Human vaccines, 4 (2008) 44-49.

Page 45: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

[147] J.P. Rolland, B.W. Maynor, L.E. Euliss, A.E. Exner, G.M. Denison, J.M. DeSimone, Direct fabrication and harvesting of monodisperse, shape-specific nanobiomaterials, Journal of the American Chemical Society, 127 (2005) 10096-10100. [148] A.L. Galloway, A. Murphy, J.M. DeSimone, J. Di, J.P. Herrmann, M.E. Hunter, J.P. Kindig, F.J. Malinoski, M.A. Rumley, D.M. Stoltz, T.S. Templeman, B. Hubby, Development of a nanoparticle-based influenza vaccine using the PRINT technology, Nanomedicine : nanotechnology, biology, and medicine, 9 (2013) 523-531. [149] N.F. Eng, S. Garlapati, V. Gerdts, A. Potter, L.A. Babiuk, G.K. Mutwiri, The potential of polyphosphazenes for delivery of vaccine antigens and immunotherapeutic agents, Current drug delivery, 7 (2010) 13-20. [150] Y. Honda-Okubo, F. Saade, N. Petrovsky, Advax, a polysaccharide adjuvant derived from delta inulin, provides improved influenza vaccine protection through broad-based enhancement of adaptive immune responses, Vaccine, 30 (2012) 5373-5381. [151] H.C. Arca, M. Gunbeyaz, S. Senel, Chitosan-based systems for the delivery of vaccine antigens, Expert review of vaccines, 8 (2009) 937-953. [152] X. Li, X. Kong, S. Shi, X. Zheng, G. Guo, Y. Wei, Z. Qian, Preparation of alginate coated chitosan microparticles for vaccine delivery, BMC biotechnology, 8 (2008) 89. [153] C. Wang, Q. Ge, D. Ting, D. Nguyen, H.R. Shen, J. Chen, H.N. Eisen, J. Heller, R. Langer, D. Putnam, Molecularly engineered poly(ortho ester) microspheres for enhanced delivery of DNA vaccines, Nature materials, 3 (2004) 190-196. [154] B. Morein, B. Sundquist, S. Hoglund, K. Dalsgaard, A. Osterhaus, Iscom, a novel structure for antigenic presentation of membrane proteins from enveloped viruses, Nature, 308 (1984) 457-460. [155] D. Drane, C. Gittleson, J. Boyle, E. Maraskovsky, ISCOMATRIX adjuvant for prophylactic and therapeutic vaccines, Expert review of vaccines, 6 (2007) 761-772. [156] M. Schnurr, M. Orban, N.C. Robson, A. Shin, H. Braley, D. Airey, J. Cebon, E. Maraskovsky, S. Endres, ISCOMATRIX adjuvant induces efficient cross-presentation of tumor antigen by dendritic cells via rapid cytosolic antigen delivery and processing via tripeptidyl peptidase II, J Immunol, 182 (2009) 1253-1259. [157] N.K. Polakos, D. Drane, J. Cox, P. Ng, M.J. Selby, D. Chien, D.T. O'Hagan, M. Houghton, X. Paliard, Characterization of hepatitis C virus core-specific immune responses primed in rhesus macaques by a nonclassical ISCOM vaccine, J Immunol, 166 (2001) 3589-3598. [158] R.G. Windon, P.J. Chaplin, P. McWaters, M. Tavarnesi, M. Tzatzaris, W.G. Kimpton, R.N. Cahill, L. Beezum, A. Coulter, D. Drane, A. Sjolander, M. Pearse, J.P. Scheerlinck, J.M. Tennent, Local immune responses to influenza antigen are synergistically enhanced by the adjuvant ISCOMATRIX, Vaccine, 20 (2001) 490-497. [159] M.J. Copland, T. Rades, N.M. Davies, Hydration of lipid films with an aqueous solution of Quil A: a simple method for the preparation of immune-stimulating complexes, International journal of pharmaceutics, 196 (2000) 135-139. [160] V.V. Pinto, A. Salanti, L.M. Joergensen, M. Dahlback, M. Resende, S.B. Ditlev, E.M. Agger, D.E. Arnot, T.G. Theander, M.A. Nielsen, The effect of adjuvants on the immune response induced by a DBL4varepsilon-ID4 VAR2CSA based Plasmodium falciparum vaccine against placental malaria, Vaccine, 30 (2012) 572-579. [161] A. Fomsgaard, I. Karlsson, G. Gram, C. Schou, S. Tang, P. Bang, I. Kromann, P. Andersen, L.V. Andreasen, Development and preclinical safety evaluation of a new therapeutic HIV-1 vaccine based on 18 T-cell minimal epitope peptides applying a novel cationic adjuvant CAF01, Vaccine, 29 (2011) 7067-7074. [162] A.W. Olsen, P. Andersen, F. Follmann, Characterization of protective immune responses promoted by human antigen targets in a urogenital Chlamydia trachomatis mouse model, Vaccine, (2013).

Page 46: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

[163] I. Rosenkrands, C. Vingsbo-Lundberg, T.J. Bundgaard, T. Lindenstrom, V. Enouf, S. van der Werf, P. Andersen, E.M. Agger, Enhanced humoral and cell-mediated immune responses after immunization with trivalent influenza vaccine adjuvanted with cationic liposomes, Vaccine, 29 (2011) 6283-6291. [164] D. Christensen, T. Lindenstrom, G. van de Wijdeven, P. Andersen, E.M. Agger, Syringe free vaccination with CAF01 Adjuvated Ag85B-ESAT-6 in Bioneedles provides strong and prolonged protection against tuberculosis, PloS one, 5 (2010) e15043. [165] D. Gall, The adjuvant activity of aliphatic nitrogenous bases, Immunology, 11 (1966) 369-386. [166] M. Hamborg, R. Kramer, C.E. Schante, E.M. Agger, D. Christensen, L. Jorgensen, C. Foged, C.R. Middaugh, The physical stability of the recombinant tuberculosis fusion antigens h1 and h56, Journal of pharmaceutical sciences, 102 (2013) 3567-3578. [167] D. Christensen, M. Henriksen-Lacey, A.T. Kamath, T. Lindenstrom, K.S. Korsholm, J.P. Christensen, A.F. Rochat, P.H. Lambert, P. Andersen, C.A. Siegrist, Y. Perrie, E.M. Agger, A cationic vaccine adjuvant based on a saturated quaternary ammonium lipid have different in vivo distribution kinetics and display a distinct CD4 T cell-inducing capacity compared to its unsaturated analog, Journal of controlled release : official journal of the Controlled Release Society, 160 (2012) 468-476. [168] R. Kaur, V.W. Bramwell, D.J. Kirby, Y. Perrie, Pegylation of DDA:TDB liposomal adjuvants reduces the vaccine depot effect and alters the Th1/Th2 immune responses, Journal of controlled release : official journal of the Controlled Release Society, 158 (2012) 72-77. [169] M. Henriksen-Lacey, D. Christensen, V.W. Bramwell, T. Lindenstrom, E.M. Agger, P. Andersen, Y. Perrie, Comparison of the depot effect and immunogenicity of liposomes based on dimethyldioctadecylammonium (DDA), 3beta-[N-(N',N'-Dimethylaminoethane)carbomyl] cholesterol (DC-Chol), and 1,2-Dioleoyl-3-trimethylammonium propane (DOTAP): prolonged liposome retention mediates stronger Th1 responses, Molecular pharmaceutics, 8 (2011) 153-161. [170] M. Henriksen-Lacey, A. Devitt, Y. Perrie, The vesicle size of DDA:TDB liposomal adjuvants plays a role in the cell-mediated immune response but has no significant effect on antibody production, Journal of controlled release : official journal of the Controlled Release Society, 154 (2011) 131-137. [171] J.H. Fritz, S. Brunner, M.L. Birnstiel, M. Buschle, A. Gabain, F. Mattner, W. Zauner, The artificial antimicrobial peptide KLKLLLLLKLK induces predominantly a TH2-type immune response to co-injected antigens, Vaccine, 22 (2004) 3274-3284. [172] C. Schellack, K. Prinz, A. Egyed, J.H. Fritz, B. Wittmann, M. Ginzler, G. Swatosch, W. Zauner, C. Kast, S. Akira, A. von Gabain, M. Buschle, K. Lingnau, IC31, a novel adjuvant signaling via TLR9, induces potent cellular and humoral immune responses, Vaccine, 24 (2006) 5461-5472. [173] M.C. Aichinger, M. Ginzler, J. Weghuber, L. Zimmermann, K. Riedl, G. Schutz, E. Nagy, A. von Gabain, R. Schweyen, T. Henics, Adjuvating the adjuvant: facilitated delivery of an immunomodulatory oligonucleotide to TLR9 by a cationic antimicrobial peptide in dendritic cells, Vaccine, 29 (2011) 426-436. [174] L.L. Farrell, J. Pepin, C. Kucharski, X. Lin, Z. Xu, H. Uludag, A comparison of the effectiveness of cationic polymers poly-L-lysine (PLL) and polyethylenimine (PEI) for non-viral delivery of plasmid DNA to bone marrow stromal cells (BMSC), European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V, 65 (2007) 388-397. [175] A. Szabo, P. Gogolak, K. Pazmandi, K. Kis-Toth, K. Riedl, B. Wizel, K. Lingnau, A. Bacsi, B. Rethi, E. Rajnavolgyi, The two-component adjuvant IC31(R) boosts type i interferon production of human monocyte-derived dendritic cells via ligation of endosomal TLRs, PloS one, 8 (2013) e55264. [176] C. Bode, G. Zhao, F. Steinhagen, T. Kinjo, D.M. Klinman, CpG DNA as a vaccine adjuvant, Expert review of vaccines, 10 (2011) 499-511. [177] S. Vecchi, S. Bufali, T. Uno, T. Wu, L. Arcidiacono, S. Filippini, F. Rigat, D. O'Hagan, Conjugation of a TLR7 agonist and antigen enhances protection in the S. pneumoniae murine infection model, European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V, (2014).

Page 47: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

[178] T. Querec, S. Bennouna, S. Alkan, Y. Laouar, K. Gorden, R. Flavell, S. Akira, R. Ahmed, B. Pulendran, Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to stimulate polyvalent immunity, The Journal of experimental medicine, 203 (2006) 413-424. [179] W.I. Lencer, U.H. von Andrian, Eliciting mucosal immunity, The New England journal of medicine, 365 (2011) 1151-1153. [180] L. Kjer-Nielsen, O. Patel, A.J. Corbett, J. Le Nours, B. Meehan, L. Liu, M. Bhati, Z. Chen, L. Kostenko, R. Reantragoon, N.A. Williamson, A.W. Purcell, N.L. Dudek, M.J. McConville, R.A. O'Hair, G.N. Khairallah, D.I. Godfrey, D.P. Fairlie, J. Rossjohn, J. McCluskey, MR1 presents microbial vitamin B metabolites to MAIT cells, Nature, 491 (2012) 717-723. [181] N. Garcon, D.W. Vaughn, A.M. Didierlaurent, Development and evaluation of AS03, an Adjuvant System containing alpha-tocopherol and squalene in an oil-in-water emulsion, Expert review of vaccines, 11 (2012) 349-366. [182] V.R. Roman, K.J. Jensen, S.S. Jensen, C. Leo-Hansen, S. Jespersen, D. da Silva Te, C.M. Rodrigues, C.M. Janitzek, L. Vinner, T.L. Katzenstein, P. Andersen, I. Kromann, L.V. Andreasen, I. Karlsson, A. Fomsgaard, Therapeutic vaccination using cationic liposome-adjuvanted HIV type 1 peptides representing HLA-supertype-restricted subdominant T cell epitopes: safety, immunogenicity, and feasibility in Guinea-Bissau, AIDS research and human retroviruses, 29 (2013) 1504-1512. [183] A.S. Levine, H.B. Levy, Phase I-II trials of poly IC stabilized with poly-L-lysine, Cancer treatment reports, 62 (1978) 1907-1912. [184] S. Daayana, E. Elkord, U. Winters, M. Pawlita, R. Roden, P.L. Stern, H.C. Kitchener, Phase II trial of imiquimod and HPV therapeutic vaccination in patients with vulval intraepithelial neoplasia, British journal of cancer, 102 (2010) 1129-1136. [185] H. Behzad, A.L. Huckriede, L. Haynes, B. Gentleman, K. Coyle, J.C. Wilschut, T.R. Kollmann, S.G. Reed, J.E. McElhaney, GLA-SE, a synthetic toll-like receptor 4 agonist, enhances T-cell responses to influenza vaccine in older adults, The Journal of infectious diseases, 205 (2012) 466-473. [186] A. McKenzie, M. Watt, C. Gittleson, ISCOMATRIX() vaccines: Safety in human clinical studies, Human vaccines, 6 (2010). [187] R. Hutchison, Is Petroleum Emulsion of any Nutritive Value?, British medical journal, 1 (1899) 724. [188] F. Cabot, Report of Experimental Work on the Dilution Method of Immunization from Rabies, The Journal of experimental medicine, 4 (1899) 181-188. [189] D.F. Driscoll, Lipid injectable emulsions: 2006, Nutrition in clinical practice : official publication of the American Society for Parenteral and Enteral Nutrition, 21 (2006) 381-386. [190] J.M. Mirtallo, J.F. Dasta, K.C. Kleinschmidt, J. Varon, State of the art review: Intravenous fat emulsions: Current applications, safety profile, and clinical implications, The Annals of pharmacotherapy, 44 (2010) 688-700. [191] A.D. Bangham, R.W. Horne, Negative Staining of Phospholipids and Their Structural Modification by Surface-Active Agents as Observed in the Electron Microscope, Journal of molecular biology, 8 (1964) 660-668. [192] R. Mischler, I.C. Metcalfe, Inflexal V a trivalent virosome subunit influenza vaccine: production, Vaccine, 20 Suppl 5 (2002) B17-23.

Page 48: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

Table 1.

Why adjuvants?

Increased antibody titers

Induce responses more rapidly

Allow antigen dose reduction or fewer doses

Increased breadth of response to overcome pathogen diversity

Induce long-lasting immune memory responses

Overcome poor immune responses in elderly and young children

Table. 2. Which adjuvants do we have ?

Adjuvant Description Clinical

status

References

Aluminum salts Insoluble aluminum salts eg.

phosphates and hydroxides. Have an

extensive safety record and are being

used as a platform for second

generation adjuvants. Antigens are

typically adsorbed to the surface.

Licensed in

US and EU

[16]

MF59 Squalene oil in water emulsion

adjuvant that has been part of a

licensed flu vaccine since 1997.

Antigens are not associated with

emulsion droplets. extensive safety

record.

Licensed in

EU

[88]

AS03 Squalene oil in water emulsion

adjuvant with the added immune

potentiator alpha tocopherol, used in

flu vaccines during 2009 pandemic,

but associated with narcolepsy.

Licensed in

EU and US

[85] [87]

AS04 Combination of aluminum adjuvant Licensed in [181].

Page 49: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

with the TLR 4 agonist

monophosphoryl Lipid A (MPL) co-

adsorbed. Approved as a licensed HPV

vaccine (Cervarix)

EU and US

Virosomes Influenza virus envelopes

reconstituted in phosphatidylcholine

bilayers (virosomes).

Licensed in

EU

[134]

AS01 Liposomes composed of dioleyl

phosphatidylcholine (DOPC),

cholesterol, MPL and QS21 in a 20 : 5 :

1 : 1 (w/w) ratio.

Phase III

Malaria

[115]

CAF01 Cationic liposome composed of

Dimethyldioctadecylammonium

bromide (DDA), and the glycolipid

trehalose 6,6’-dibehenate (TDB) in a 5:1

(w/w) ratio prepared by film hydration.

Phase I

[182]

Poly I:C Synthetic double stranded

oligonucleotide containing repeating

units of inosine and cytosine that

signals through TLR3.

Phase I [183]

IC31 Antimicrobial peptide KLK bound to

ODN1a, signals through TLR9.

Phase I [171]

AS02 Co-mixture of AS03 emulsion adjuvant

with QS21 and MPL.

Phase II [116, 119]

Imiquimod Small molecules immune potentiators

that signal through TLR7 / 8.

Phase II [184]

CpG

oligonucleotides

(ISS 1018)

TLR9 agonists based on bacterial DNA. Phase III [176]

SE / SE-GLA Squalene based emulsion that is a

stand-alone emulsion adjuvant, or

combined with the TLR4 agonist GLA.

Phase I [155, 185]

Page 50: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

ISCOMS and

ISCOMATRIX

Small (40nm) lipid based adjuvants

consisting of phospholipids,

cholesterol and saponins.

Phase I [186]

Table 3a

Groups Serum IgG Bactericidal titer

Soluble Men B antigen 423 <16

Men B / TLR5 fusion 11 <16 Men B + CpG 511 <16

PLG adsorbed Men B 23,171 64 PLG adsorbed Men B + CpG 137,747 1,024

Table 3b

Groups Serum IgG Bactericidal titer

PLG adsorbed Men B 11,367 512 PLG adsorbed Men B + MPL 18,074 2048 PLG entrapped MPL and adsorbed Men B 66,493 8192

Page 51: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

Table 4.

Delivery

system

First

described in

literature

First medical

uses

First approved for

use as drug delivery

vehicle

First approved

for use as

adjuvant in

vaccine

Emulsions Early 1900’s

[187, 188]

Intralipid –

1962 [189]

Propofol – 1989 [190] MF59 – 1996 [7]

Liposomes 1964 [191] Doxil – 1995

[190]

Doxil – 1995

[190]

Invlexal V –1997

[192]

Microparticles

(PLGA)

Early 1970’s

[134]

Decapeptyl

LP – 1986

[134]

Decapeptyl LP – 1986

[134]

N/A

Page 52: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

Figure 1

Page 53: Designing and building the next generation of improved vaccine adjuvants

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

Graphical abstract