• No results found

University of Groningen In vitro approaches for the evaluation of human vaccines Signorazzi, Aurora

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen In vitro approaches for the evaluation of human vaccines Signorazzi, Aurora"

Copied!
29
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

In vitro approaches for the evaluation of human vaccines

Signorazzi, Aurora

DOI:

10.33612/diss.166150822

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2021

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Signorazzi, A. (2021). In vitro approaches for the evaluation of human vaccines. University of Groningen.

https://doi.org/10.33612/diss.166150822

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter 6

Summarizing discussion

(3)

162 Cha pte r 6

Background

As highlighted in Chapter 1, the development of vaccines is a complex process,

traditionally dominated by trial and error [1]. Long and costly investigations, often

including animal studies, are required. Many vaccine candidates fail to reach the final

stages of development because the results obtained in animals are not translatable

to humans [2]. In addition to their use in research towards new vaccines, in vivo

methods are also heavily employed during batch surveillance of commercially

available vaccines [3]. However, animal tests – despite being the standard evaluation

method for many vaccines – suffer from large variability in the results [4]. The

development of alternative methods is therefore necessary to obtain more consistent

results and to decrease the ethical and economical burdens of animal studies [5,6].

Different types of in vitro assays can be used for the refinement, reduction or

replacement of animal tests during vaccine development and production [7].

Analytical methods study the physico- and immunochemical properties of the

vaccine candidate and its components, such as the antigens’ characteristics and

functionality [8]. Cellular assays can instead be used to characterize the reaction of

immune cells to the formulation [9]. In this thesis, both analytical and cell-based

assays were used to evaluate different properties of two model viral vaccines for

human use, tick-borne encephalitis (TBE) vaccine and influenza vaccine.

Characterizing TBE vaccine-specific immune responses

In Chapters 2-4, we aimed to study the interaction of human immune cells with the

TBE vaccine. Additionally, we harnessed the knowledge gained in these studies to

develop a cell-based assay that could contribute to the replacement of mandatory

in vivo potency testing of TBE vaccine batches. The findings of these chapters are

summarized in Figure 1.

The knowledge on the immune responses to the TBE vaccine is currently limited to

the induction of envelope protein-directed antibodies and of TBEV-specific CD4

+

T

cells [10–12]. Innate immune responses, that provide useful information on the

mechanism of action of a vaccine and that can be easily evaluated for assessing the

quality of the formulation, have not been characterized yet. In Chapter 2, we aimed

to investigate the early responses of human immune cells to the TBE vaccine. Primary

cells such as peripheral blood mononuclear cells (PBMCs), despite presenting

donor-dependent variability [13], are a versatile platform for the characterization of vaccine-

or pathogen-associated responses [14–16].

(4)

163

Cha

pte

r 6

Figure 1. Graphical summary on the development, assessment and application of an in

vitro system for the evaluation of the TBE vaccine. ❶ Using cell-based assays, we found

that freshly isolated or cryopreserved PBMCs can respond consistently and specifically to I-TBEV, but not to the adjuvanted TBE vaccine. ❷ THP-1-derived cells, instead, do not respond specifically to either formulation. ❸ In freshly isolated PBMCs, the surface protein and the viral genome (sensed via Toll-like receptor 7, red endosomal receptor in the figure) of I-TBEV stimulate IFN-producing plasmacytoid dendritic cells. IFN production induces differentiation of plasmablasts and increase in total antibodies production. ❹ Cryopreserved PBMCs can distinguish high- from low-quality I-TBEV batches, as they induce distinct expression levels of IFN-stimulated genes (ISGs). The expression of ISGs correlates with the percentage of high-quality formulation in a mixed sample. ❺ Using the same platform, we characterized the innate responses in vitro to inactivated or live TBEV; both induce significant upregulation in the transcription of ISGs and of RIG-I-like receptor (RLR) genes, and a limited inflammatory response. In this platform, I-TBEV sensing involves RLRs (blue cytosolic receptor), while the live virus is sensed by Toll-like as well as RIG-I-like receptors. Image created with BioRender.

(5)

164

Cha

pte

r 6

In this study, freshly isolated PBMCs were stimulated with the adjuvanted TBE vaccine

and its primary component, formalin-inactivated TBEV (I-TBEV). The cellular

responses were then studied in several cell subsets.

Characterization of molecular markers and proteins expressed and produced by

PBMCs revealed that I-TBEV induced type I interferon (IFN) responses, important

mediators in immunity against live TBEV [17–19]. The I-TBEV-induced IFN-α

production was found to be critical for the differentiation of B cells to plasmablasts.

Additionally, stimulation with I-TBEV resulted in increased production of total IgG

and IgM as compared to untreated cells. The adjuvanted vaccine formulation, on the

other hand, was not able to induce upregulated expression or production of IFN and

interferon-stimulated genes, nor increased plasmablast differentiation or total

antibody production. Aluminum hydroxide, present in the adjuvanted vaccine as well

as in the excipient, had an inhibitory effect on the induction of I-TBEV-associated

responses and caused a decrease in cell viability. Indeed, Al(OH)

3

is known to

interfere with readouts in cellular assays, due to both its in vitro toxicity and

antigen-aggregating mechanism [20–23] – notwithstanding its proven safety and adjuvanting

potency in humans [24,25]. Thus, the characterization of the cell responses to the TBE

vaccine was further performed using the vaccine antigen-carrying component,

I-TBEV.

After identifying the induction of type I interferon as characteristic of the response

to I-TBEV, we aimed to pinpoint the cell population(s) involved in the signaling.

Plasmacytoid dendritic cells (pDCs) were found to be in large part responsible for the

I-TBEV-induced IFN-α production. Additionally, I-TBEV promoted the differentiation

of a subpopulation of pDCs, P1-pDCs, previously shown to be highly specialized in

interferon production following virus stimulation [26].

The immunostimulatory mechanism of I-TBEV was shown to require a functional

surface protein, needed for cell binding and entry, and an intact viral genome, sensed

upon internalization of the pathogen. In fact, antibody-mediated virus neutralization,

denaturation of the glycoprotein or inhibition of the RNA-sensing Toll-like receptor

7 (TLR7) all resulted in the abrogation of I-TBEV-induced production of IFN, cytokines

and antibodies, and in the inhibition of cell differentiation. The important role of TLR7

in flavivirus sensing was brought to light by previous in vivo studies, which found a

link between TLR7 deficiency in animal models and increased replication of Langat

virus (a naturally attenuated flavivirus in the TBEV serogroup) as well as of Japanese

encephalitis virus (a mosquito-borne neurotropic flavivirus) [27,28]. Triggering of

TLR7 in TBEV sensing has not been previously proven, but only postulated [18]; here,

we show that this receptor is involved in the response of freshly isolated PBMCs to

inactivated TBEV. Thus, the stimulating properties of the genomic RNA appear to be

retained upon virus inactivation by formaldehyde.

(6)

165

Cha

pte

r 6

These results highlight that the functional integrity of the E glycoprotein and of the

viral genome are preserved during manufacturing of the TBE vaccine and are

required for the induction of type I interferon, an important link between innate and

adaptive immunity [29,30].

Establishing an in vitro evaluation system for the TBE

vaccine

As for other vaccines, the potency of the TBE vaccine is evaluated for each newly

produced batch using a lethal challenge test on immunized mice [31]. Recently,

several in vitro assays have been developed as alternatives to animal-based tests for

the quality control of vaccines [9,15,32–36]. A strategy that could reduce the need of

in vivo tests for vaccine batch release is the consistency approach: newly produced

batches of a vaccine are compared to reference ones (of proven in vivo potency) to

verify their conformity with respect to a series of quality attributes assessed in vitro

[8]. In Chapter 3, we applied the knowledge gained in Chapter 2 to develop a

cell-based method for assessing the conformity of TBE vaccine batches with respect to

the induction of innate immune responses. Together with other in vitro assays, this

method could contribute to the reduction (or replacement) of in vivo potency tests

for lot release [3]. The cellular platform for such an assay should display distinctive

responses to high- and low-quality vaccine batches and, to be applicable in a quality

control environment, should respond in a reproducible manner.

Antigen-presenting cells (APCs), as the sentinels of the immune system and main

targets of TBEV in the first stages of infection [37], are an interesting candidate

platform for measuring quality-related biomarkers. We assessed two different

APC-like platforms: one cell line-based and one primary cell-based. Cell lines are

advantageous as a consistent source of cells not subject to donor-dependent

variation. In this study we used THP-1 cells, long used as a monocyte, macrophage

and dendritic cell model [38–40]. Primary cells, on the other hand, are more closely

representative of in vivo responses than cell lines [41], and were identified in Chapter

2 as able to display I-TBEV-induced responses. Here, we used frozen-thawed PBMCs,

which would be more convenient than freshly isolated cells in an industry setting.

Using RT-qPCR as readout, we showed that the THP-1-derived platforms were

unsuitable for our purposes, as the cells showed no vaccine-specific activation. Yet,

THP-1 cells were able to generate an antiviral response after stimulation with live

TBEV. Indeed, THP-1 cells are permissive to TBEV infection [42], but produce lower

amounts of TBEV-induced chemokines than other cell lines [43]. The responses of

THP-1 cells to I-TBEV, a less potent stimulus than the live virus, might therefore have

been too low to reach the limit of detection.

(7)

166

Cha

pte

r 6

Primary cell-based platforms, in contrast, were able to respond specifically to I-TBEV

through increased transcription of interferon-stimulated genes (ISGs), in line with the

results from our previous study. Unsurprisingly, also in this platform the presence of

alum in the adjuvanted vaccine induced cytotoxicity; thus, we focused on assessing

quality parameters in I-TBEV.

Using the inactivated virus to stimulate PBMCs, we found that, in multiple donors,

the expression of several ISGs was similarly induced by different (conforming) vaccine

batches and differentially regulated in response to high- and low-quality batches.

Additionally, we evaluated the sensitivity of the assay, and showed that it could

distinguish between I-TBEV batches with differences in the formulation quality as low

as 20%.

These results support the suitability of a cell- (and specifically PBMC-) based platform

for batch conformity testing of the non-adjuvanted TBE vaccine. Such a platform

could be used for in-process control testing of vaccine batches, as well as for

screening vaccine candidates during development [14,15,44–46].

Applying in vitro evaluation systems: analysis of TBE

vaccine-associated pathways

Applying the platform presented in Chapter 3, in Chapter 4 we aimed to further

study the functions and pathways induced by I-TBEV. Additionally, we set out to

compare differences in the response to replicating and inactivated virus, to analyze

possible effects of the inactivation process on the immunogenicity of the vaccine.

Using RNA sequencing, we assessed the innate immune responses of PBMCs to live

and inactivated TBEV by analyzing the cells’ transcriptome. The results show that

both stimuli induced a distinct interferon-dominated signature in PBMCs, confirming

our previous findings on the important role of type I IFN in TBEV-associated

responses and in line with data on common early immune signatures induced by

several vaccines [17,47].

Inflammatory responses, on the other hand, were selectively regulated. After

treatment of the PBMCs with (I-)TBEV, the JAK-STAT pathway was upregulated, while

the expression of genes involved in interleukin signaling, antigen presentation and

lymphoid–non-lymphoid cell interaction was downregulated. These results are in

accordance with our earlier findings [48], but appear characteristic of the pathogen.

Indeed, other whole inactivated vaccines (WIVs), such as those against influenza virus

and respiratory syncytial virus, induce upregulation of some of the genes we found

downregulated in (I-)TBEV-treated cells [44,49–51].

(8)

167

Cha

pte

r 6

After characterizing the downstream response of (I-)TBEV-stimulated PBMCs, we

next focused on identifying the pattern recognition receptors (PRRs) involved in the

transduction of the signal. RNA-Seq had identified, next to the characteristic IFN

signature, an upregulation of cytoplasmic RIG-I-like receptors (RLRs) in cells

stimulated with live or inactivated TBEV. Thus, we analyzed the involvement in (I-)

TBEV sensing of RLRs – as well as of TLRs, prompted by the findings in Chapter 2.

Using pathway-specific inhibitors and reporter cell lines, we demonstrated that RLRs

are involved in the initiation of the antiviral response to I-TBEV and live TBEV. Indeed,

activation of RLRs is known as a predominant mechanism in sensing flaviviruses

[52,53]. TLR pathways were implicated in the induction of the antiviral response

against live TBEV, but did not appear to be involved in the sensing of the inactivated

virus in this study. This result is in apparent contrast with the TLR7 involvement

identified in Chapter 2 in freshly isolated cells. However, the use of frozen-thawed

PBMCs in Chapter 4 might explain these differences.

Indeed, cryopreservation of PBMCs has been shown to affect the pDC population

[54], identified in Chapter 2 as the primary source of I-TBEV-induced IFN-α. Further

investigation corroborated the hypothesis of a detrimental effect of cryopreservation

on pDC functionality, as a reduction by 75% of the I-TBEV-induced IFN-α was

observed in PBMCs that have undergone the freezing and thawing process (Etna et

al., unpublished results). Our working theory is therefore that pDCs, in which the

RIG-I pathway is dispensable for an RIG-IFN response [17], would preferably sense and

respond to I-TBEV through the TLR7 pathway. Other myeloid cells, less affected by

the cryopreservation process [55], could sense I-TBEV mainly through RLRs instead.

The predominant role of RIG-I in I-TBEV sensing by myeloid cells other than pDCs

would also explain the induction of the type I interferon response and of the

JAK-STAT pathways [56] and the unsuccessful activation of the THP-1 cells in Chapter 3,

as these monocytic tumor cells show a much lower expression of this receptor

compared to primary cells [57] and other cell lines [58].

Overall, Chapter 2 and 4 highlight the importance of PRR stimulation by the viral

RNA for the induction of an immune response by the TBE vaccine. The

immuno-stimulating properties conferred by the presence of the viral genome in a vaccine are

already known; for instance, influenza vaccines containing the viral RNA are more

immunogenic than formulations lacking it [44,49,59]. In the absence of the viral

genome (as in vaccines based on virus-like particles), TLR7 or RIG-I agonists have

been proposed as adjuvants to enhance poorly immunogenic vaccine candidates

[60–64]. While such an adjuvanting strategy is attractive from an immunogenic point

of view, its implementation has been hindered by safety concerns [65]. The

preservation of the viral genome during the production of whole inactivated vaccines

is therefore paramount, as this feature provides important self-adjuvanting

properties to the formulation.

(9)

168

Cha

pte

r 6

Collectively, the data presented in Chapters 2-4 brings new insights into the innate

response elicited and the signaling pathways engaged by live TBEV and by the

inactivated virus – the primary constituent of the TBE vaccine. Additionally, the results

obtained prove the applicability of the designed PBMC-based system for a variety of

purposes, from mechanistic immunological studies (Chapter 2 and 4) to batch

release testing (Chapter 3).

Applying in vitro evaluation systems: analysis of

inactivation-associated effects

The data presented in the previous chapters underscored the importance of

maintaining the structure and functionality of the viral particles and their

components during the preparation of inactivated vaccines. The integrity of the viral

proteins and genome can be affected by the inactivation process through the

introduction of nicks or cross-links [66,67]. In Chapter 5, using immunochemical and

cell-based methods, we assessed different inactivation procedures for their effect on

the virions’ integrity and functionality – here with influenza as a model.

In the case of influenza vaccines, all formulations derived from whole viral particles

(with the exception of live-attenuated vaccines) share a virus inactivation step [68].

The concept behind the inactivation procedure is that the process must inhibit the

replication of the virus without impacting on its antigenicity (for example by altering

the vaccine antigens) [69–71]. Influenza inactivation is usually achieved by exposure

of the live virus to β-propiolactone (BPL) or formaldehyde (FA), two common

inactivating chemicals [72]. Pharmacopoeias specify the maximal concentration of

these compounds to be used during the inactivation procedure; yet, the

standardization of all other parameters is left to the manufacturers. Studies have

shown that excessive inactivation with FA or BPL may cause alterations to the

antigens that result in diminished functionality of the viral particles in the vaccine

[73–78]. The effects of FA on the physicochemical properties of proteins are known

[79], while BPL is traditionally thought to mainly affect nucleic acids [67,80]. However,

recent studies have highlighted that BPL could have an effect on proteins as well

[68,81].

In this study, we assessed the impact of the two inactivating agents on the

immunochemical properties of the virus particles in several influenza virus strains, to

provide insights for the improvement and harmonization of influenza vaccine

production. Our results show that both BPL and FA caused alterations in the

properties of the virions, as summarized in Table 1. Additionally, the magnitude of

the effects caused by the treatments was found to be strain-dependent.

(10)

169

Cha

pte

r 6

Table 1. Impact of different chemical inactivation methods on influenza virus infectivity, binding, fusion and TLR7 stimulation capacity. These properties were assessed in live and

inactivated virus preparations and compared across several influenza virus strains. The number of symbols indicates the degree of efficacy (green checks) or of the detrimental effects on the virions (red arrows) of the inactivation treatments.

Generally, BPL was more efficient in inactivating the viral particles than formaldehyde,

as assessed by measuring the residual infectivity. This result was not unexpected, as

low temperature FA-treatment was previously found to cause incomplete

inactivation [82]. Following the particles’ interaction with cells, we then assessed the

ability of the inactivated virus to bind to cell membranes (measured using the

hemagglutinating properties of influenza virus [83]), to fuse in conditions of low pH

[84] and to activate TLR7, a receptor present in the endosomal compartment,

indicating successful cell entry [85]. Both BPL and FA impacted on the binding and

fusion capacity of the viral particles, in accordance with previous studies [81,86,87].

In our head-to-head comparison, BPL caused greater loss of these properties than

FA. The stimulation of a TLR7 reporter cell line was also affected by the inactivation

treatments. In this case, the inactivation with FA impacted more on influenza-induced

TLR7 activation than the BPL treatment. While we did not investigate the direct effect

of the inactivating agents on the viral RNA, other studies showed that the FA

inactivation procedure could affect the integrity of the genome [87–89]. However,

the effects of an inactivating agent on the RNA-TLR7 interaction alone are difficult

to estimate. Stimulation of an endosomal receptor presupposes binding and

internalization of the virus, processes that can be affected by inactivation-induced

protein alterations.

In summary, this study shows that chemical inactivation impacts on various

properties of influenza virus in a strain-dependent way, and that many other factors

beyond the concentration of the inactivating agent itself – most prominently the

temperature used during the inactivation process [72,82,90,91] – have to be

considered. While subtle discrepancies in differently-inactivated viruses do not

necessarily translate to significantly distinct safety and potency of the vaccine in vivo

[92], it should be noted that a thorough investigation has not yet been presented.

For such an analysis, this study describes relatively simple but highly informative

methods that could be used in an in vitro vs in vivo evaluation to assess and

ultimately select better inactivation strategies.

(11)

170

Cha

pte

r 6

In vitro vaccine evaluation systems: challenges and

opportunities

This thesis has presented a number of in vitro vaccine evaluation systems; a relevant

question that remains to be addressed is for which applications can these methods

actually be used, and at what stages of the vaccine development process. In vitro

evaluation systems can in general be applied for different purposes:

Research on vaccines and vaccine-induced responses.

In vitro methods can be used to study antigen conformation, alterations,

vaccines’ mechanisms of action and relevant pathways. For analyses focused on

the properties of the vaccine itself – such as antigen assembly, quantification and

adsorption to the adjuvant – immunochemical methods are already extensively

used as they provide a level of detail not achievable with animal studies [93–96].

The evaluation of vaccine-induced immune responses in vitro presents a

different level of complexity, as the immune system responds to extraneous

agents through dynamic interactions between several cell types and

environments. Innate immune responses are relatively easily measurable, since

they can be generated by individual cell types – in particular APCs. Cellular assays

focused on the interaction of APCs with vaccines have identified selected

pathogen-specific vaccine-induced responses [9,44,46,97,98]. Omics approaches

have pushed this even further, allowing us to obtain a complete picture of the in

vitro innate responses to a particular vaccine (as was done in Chapter 4) and to

draw more general conclusions regarding vaccine-associated signatures [99–

103]. For what concerns the evaluation of adaptive responses (the induction of

which is the actual goal of vaccination), the use of in vitro methods can prove

more complicated. Adaptive responses necessitate cell interactions;

antigen-specific activation of B or T cell lymphocytes requires the presence of unique

cells with a particular B- or T-cell receptor molecule [104,105]. Nevertheless,

novel in vitro platforms for the evaluation of B and T cell responses are being

developed [14,106], and will eventually allow prediction of adaptive responses

upon vaccination, particularly when using systems based on several modules

[107]. Thus, basic research on vaccines and vaccine-induced immune responses

remains one of the major applications of immunochemical and cell-based in vitro

methods.

Selection and comparison of promising vaccine candidates.

Apart from their application in vaccine research, in vitro methods offer

interesting possibilities for the pre-clinical development stage. In this phase,

cellular methods are already in use – however mainly for toxicity assessments

rather than efficacy determination, for which animal studies remain the go-to

approach [108].

(12)

171

Cha

pte

r 6

The use of animal models has resulted in the identification of many vaccine

candidates. Yet, a number of them did not perform adequately in subsequent

clinical trials, calling into question the relevance of such animal models for the

prediction of responses in humans [109,110]. Most notably, research into HIV

vaccines has faced many setbacks, as more than 90 vaccine candidates that were

successful in animal studies failed in humans [111]. This result is especially

disappointing considering that for HIV vaccines the animal models often

included non-human primates, expected to generate highly translational results.

In hindsight, this turned out to be a dangerous assumption, as it led researchers

to dismiss unsupportive in vitro data [112,113]. Therefore, the use of alternative

methods for the determination of vaccine candidates’ efficacy should not be

secondary but parallel – or even prior – to in vivo studies. The establishment of

panels of assays could further improve the translational value of the in vitro

results, by offering a comprehensive overview of the vaccine candidates and of

the associated responses. The use of high-throughput techniques could further

speed up this selection process [114]. In addition to their application in

pre-clinical development, in vitro methods can support the selection of the most

promising vaccine candidates in the clinical phases through head-to-head

comparisons of several candidates.

Assessment of the quality of commercial vaccines.

As abundantly discussed, one of the most extensive uses of animals in the

vaccine development and production environment lies in lot release testing

[115]. Despite the fact that these in vivo tests have been used in quality control

for several years, intrinsic issues pertaining the reproducibility of results from

animal experiments can cause high variations [116]. In adherence to the

consistency approach, in vitro testing systems can be used to replace, reduce or

refine animal tests – thereby addressing scientific, economical and ethical

concerns. In vitro assay can evaluate the vaccine quality and batch-to-batch

consistency using different strategies. Immunochemical binding assays, allowing

antigen quantification as a measure of vaccine efficacy, are in use in parallel to

(or proposed as replacement of) in vivo assays for some human as well as

veterinary vaccines [33,117–120]. While still not commonly used for the quality

control of vaccines, several types of cell-based in vitro methods can assess the

biological activity as relevant indication of the potency in vivo. Cellular assays

developed to evaluate the quality or conformity of the formulation do so by

measuring the production of vaccine-encoded genes, the cell responses to the

vaccine or the vaccine-conferred in vitro protection [9,15,121–123]. A number of

these alternative methods are in the process of being accepted and implemented

by pharmacopeias, as the use of in vitro assays for the purpose of batch release

testing of vaccines is being promoted by the scientific, industrial and regulatory

communities alike [124].

(13)

172

Cha

pte

r 6

Keeping these purposes in mind, the in vitro methods – and especially the cell-based

assays – presented in this thesis can be re-evaluated with respect to their potential

application. A summary of the conclusions drawn is shown in Table 2.

For basic research on human vaccines, in vitro studies offer the possibility of testing

the effect of the formulations on the target species’ cells – which, as previously

discussed, can respond quite differently than what results from animal models might

suggest. As shown in Chapters 2 and 4, the cell-based methods described allow the

investigation of vaccine-associated innate responses. At the research stage of vaccine

development, in vitro systems using primary cell platforms may provide more

representative data than that obtained with cell lines. As shown in Chapter 3,

immortalized cells might present different permissiveness and responsiveness to the

pathogen in its live or inactivated form.

Since basic research is often performed across several institutions, the methods and

platforms need to be relatively universally available, simple and inexpensive. Such

was the case for the assays employed in this thesis to study cytokine production,

expression of genes and surface markers, and functionality of the virions. An example

of basic research in which the PBMC-based assay described would be valuable is the

in vitro investigation of the immunological basis of vaccine failure. Unsuccessful

immunization following vaccination occurs most frequently in the elderly [125], and

has been observed for viral vaccines against TBE, influenza, herpes zoster, hepatitis

B, mumps and measles [126–131]. Analyzing immune cells from subjects in which the

vaccine did not provide protection and comparing the induction of

vaccine-associated biomarkers between donors from different age groups may help unravel

the mechanisms of immunosenescence, and could lead to improved immunization

efficacy.

Similarly to their application in vaccine research, the cell-based methods described

can be employed in the pre-clinical and clinical phases. In parallel – or prior – to the

in vivo studies performed to evaluate potency and safety, in vitro methods such as

those presented in this thesis could systematically assess several formulations and

expedite the selection of the most promising vaccine candidates. For assays

monitoring the activation of immune cells, an initial investigation into the cellular

responses occurring in the animal models following immunization should be

performed to confirm the relevance of the potential biomarkers analyzed in vitro.

Besides supporting the pre-clinical development phase, primary cell-based methods

(such as those used in Chapters 2 and 3) can also be applied during the clinical stage:

in particular, ex vivo analyses of the responses in immune cells from the vaccinated

subjects can offer an in-depth characterization of the vaccines’ function in humans.

These screenings are indeed becoming increasingly common during early clinical

studies of new vaccines [132–135], as they provide a better understanding of the

underlying immunization mechanism.

(14)

173

Cha

pte

r 6

Table 2. Applicability of cellular platforms at the different stages of vaccine research, development and production. 3 green checks indicate that the platforms are already suitable

and applicable. 2 or 1 checks indicate platforms that are not as suitable or that require further optimization (superscripts). A red cross indicates a platform unsuitable for the scope.

The most difficult stage for the implementation of primary cell-based methods is

arguably the batch release testing for commercial vaccines. In this phase more than

in all the others, the system(s) used (be it animal models, cellular platforms or

analytical methods) must be practical - due to the high frequency of testing – and as

consistent as possible, to allow comparisons across multiple batches. So far,

cell-based assays are used only for toxicity assays, while immunochemical methods are

accepted as alternatives for potency assessments [136,137]. As the interest in

replacing (or at least reducing) animal testing is now increasing in all sectors involved

in the vaccine industry, more and more cell-based assays are being developed for

potency assessments [124]. The implementation of such methods for the evaluation

of vaccine batches is therefore a question of when, not if.

Yet, some cellular platforms present more challenges than others; primary cells are

obviously less easily retrievable than cell lines, and, as shown in Chapter 3, can

exhibit donor-specific responses – at the very least quantitatively. These hurdles can

be overcome by bulk processing of PBMCs, which could be isolated, screened and

selected all at once, thereby allowing consecutive use of consistent aliquots.

Additionally, pooling of PBMCs from multiple donors could further increase their

applicability by mitigating variations in responses [13,138]. PBMC pooling, however,

would only be applicable for assessing the innate immune responses to a vaccine

after short-term stimulation. Longer incubation might lead to allogeneic responses

in cells from mixed donors [139], which would affect the assay readouts.

(15)

174

Cha

pte

r 6

The solutions proposed can minimize the intrinsic problematics derived from using

primary cells. However, for the purpose of ensuring batch-to-batch conformity,

the consistency approach prescribes the use of in vitro assays not necessarily for

predicting the potency in vivo, but for analyzing the consistency of vaccine batches

based on quality attributes [8], which – once identified – could be assessed in the

most convenient platform. Thus, the choice of platform does not have to favor the

cells that preserve the most in vivo properties (such as PBMCs), but rather those that

present the least complexity of usage and the highest reproducibility. Consequently,

cell lines remain the most homogeneous platform for the purpose of vaccine batch

assessment, and were our initial choice for the assay described in Chapter 3. If a cell

line responsive to the vaccine is identified, the development and adoption of a cell

line-based vaccine testing system would prove more feasible than the

standardization of a primary cell platform.

For the identification of such a cell line (and of potential readouts)

,

primary cell-based

assays can nevertheless be of help: once the pathways and receptors involved in the

specific response to the vaccine are characterized (as was done in Chapters 2 and

4), the candidate platforms could be efficiently screened and selected. In the case of

the TBE vaccine, for example, a potential platform (other than the PBMC-based one)

should possess functional RIG-I-like receptors and potent interferon responses.

Some commercial cell lines with these characteristics are available and even include

reporter constructs, which would allow fast and simple readouts [140,141]. If these

commercial platforms were to present low sensitivity or non-responsiveness,

transfecting the receptor of interest to create a custom-made stable cell line (an

approach already taken for some toxicological assays [142]) could be considered.

In vitro vaccine evaluation systems: future perspectives

With the continuous advancements in bioinformatics and the steady decrease in cost

of omics approaches, it is safe to predict that multiparametric analyses will become

more and more common in the vaccinology field [143]. For basic research purposes,

a more in-depth analysis into the cell-specific responses to a vaccine could be

obtained through single-cell sequencing of in vitro vaccine-stimulated cells [144].

This method is similar to that described in Chapter 4 but offers the possibility of

zooming in on the responses of individual cells to identify cell type-specific functions

and pathways induced. Deconvolution methods, which can associate observations in

bulk transcriptomics data (from unfractionated PBMCs) to specific immune subsets

[145], can already partially address this task by inferring cell type proportions

[146,147]. However, the profiling of cellular immune responses in individual subsets

enables a highly detailed characterization of the vaccine-elicited cellular immunity,

especially if combined with proteomics or metabolomics analysis [102]. Furthermore,

single-cell sequencing could potentially suggest the most appropriate cell types for

the development of cellular platforms to be used in subsequent batch release testing.

(16)

175

Cha

pte

r 6

Besides advanced readout methods, several promising and increasingly

sophisticated cellular platforms are being developed. These cell-based models try to

replicate the complexity of the immune system, which exerts its response through an

extensive range of organs, tissues and cell types with specialized functions. Complex

systems (usually) present the disadvantage of being expensive and difficult to

standardize across different laboratories. Yet, they may also provide an opportunity

to overcome the necessarily reductionist approaches of conventional in vitro

methods to the investigation of immune responses.

The MIMIC® (modular immune in vitro construct) system represents one such an

example. This system consists of several modules, that aim to predict the migration

of APCs in peripheral tissues or the induction of naïve and recall B and T cell

responses in lymphoid tissues using isolated PBMCs and tissue constructs [148]. An

advantage of the MIMIC® system is that it offers the possibility to automate the

testing process and evaluate the responses of a large number of donors

simultaneously, features that favor its use in a pre-clinical development setting. A

comparison of the immune responses in vivo, ex vivo and in the MIMIC®-based assay

showed the suitability of the system to support vaccine development also during

clinical studies [149]. In this study, the platform was in fact able to reflect

age-associated differences in vaccination-induced immunity, showing reduced

influenza-specific B and T cell responses in elderly donors that paralleled the in vivo antibody

responses.

Adding another layer of complexity are the three-dimensional systems using

micro-organoids and organ- or tissue-on-a-chip models [150,151]. Many of these platforms

are being developed for cancer immunology research [152–154], but could very well

be applied for the investigation of vaccine-associated responses. A recent work by

Wagar et al. describes a functional organotypic system that recapitulates key

germinal center features, including the production of antigen-specific antibodies,

somatic hypermutation, plasmablast differentiation and class-switch recombination,

allowing the investigation of adaptive immune responses upon vaccination in vitro

using human cells [155].

These novel platforms and technologies have the potential of further improving our

understanding of vaccine immunology. For the purpose of lot-to-lot testing of

vaccines, however, they might prove difficult to harmonize across multiple vaccine

manufacturing sites and contract research organizations, and complex to validate for

the approval by health agencies. Simpler methods, such as those described in this

thesis – not attempting to fully mimic the in vivo immune response, but evaluating

(several) key aspects of vaccine quality – could in the near future advance the

implementation of the consistency approach, eventually leading to a vaccine

development and testing environment that is both more effective for humans and

less painful to animals.

(17)

176 Cha pte r 6

References

[1] Raeven RHM, van Riet E, Meiring HD, Metz B, Kersten GFA. Systems vaccinology and big data in the vaccine development chain. Immunology 2019;156:33–46.

https://doi.org/10.1111/imm.13012.

[2] Jameson SC, Masopust D. What Is the Predictive Value of Animal Models for Vaccine Efficacy in Humans? Reevaluating the Potential of Mouse Models for the Human Immune System. Cold Spring Harb Perspect Biol 2018;10. https://doi.org/10.1101/cshperspect.a029132.

[3] De Mattia F, Chapsal J-M, Descamps J, Halder M, Jarrett N, Kross I, et al. The consistency approach for quality control of vaccines e A strategy to improve quality control and implement 3Rs. Biologicals 2011;39:59–65. https://doi.org/10.1016/j.biologicals.2010.12.001.

[4] Verthelyi D, Casey W, Arciniega J, Isbrucker R, Schmitt M, Finn T, et al. Non-animal replacement methods for human vaccine potency testing: state of the science and future directions. Procedia Vaccinol 2011;5:16–32. https://doi.org/10.1016/J.PROVAC.2011.10.002. [5] Bottini AA, Hartung T. Food for thought... on the economics of animal testing. ALTEX

2009;26:3–16. https://doi.org/10.14573/altex.2009.1.3.

[6] Ferdowsian HR, Beck N. Ethical and scientific considerations regarding animal testing and research. PLoS One 2011;6:e24059. https://doi.org/10.1371/journal.pone.0024059.

[7] Lewis DJM, Lythgoe MP. Application of “Systems Vaccinology” to Evaluate Inflammation and Reactogenicity of Adjuvanted Preventative Vaccines. J Immunol Res 2015;2015:1–11. https://doi.org/10.1155/2015/909406.

[8] Hendriksen CFM. Replacement reduction and refinement alternatives to animal use in vaccine potency measurement. Expert Rev Vaccines 2009;8:313–22.

https://doi.org/10.1586/14760584.8.3.313.

[9] Hoonakker ME, Verhagen LM, Hendriksen CF, van Els CA, Vandebriel RJ, Sloots A, et al. In vitro innate immune cell based models to assess whole cell Bordetella pertussis vaccine quality: a proof of principle. Biologicals 2015;43:100–9. https://doi.org/10.1016/j.biologicals.2014.12.002. [10] Jarmer J, Zlatkovic J, Tsouchnikas G, Vratskikh O, Strauss J, Aberle JH, et al. Variation of the

Specificity of the Human Antibody Responses after Tick-Borne Encephalitis Virus Infection and Vaccination. J Virol 2014;88:13845–57. https://doi.org/10.1128/jvi.02086-14.

[11] Aberle JH, Schwaiger J, Aberle SW, Stiasny K, Scheinost O, Kundi M, et al. Human CD4+ T Helper Cell Responses after Tick-Borne Encephalitis Vaccination and Infection. PLoS One 2015;10:e0140545. https://doi.org/10.1371/journal.pone.0140545.

[12] Kubinski M, Beicht J, Gerlach T, Volz A, Sutter G, Rimmelzwaan GF. Tick-Borne Encephalitis Virus: A Quest for Better Vaccines against a Virus on the Rise. Vaccines 2020;8:451. https://doi.org/10.3390/vaccines8030451.

[13] Wieczorek L, Brown BK, DelSarto Macedo C, Wesberry-Schmierer M, Ngauy V, Rosa Borges A, et al. Mitigation of variation observed in a peripheral blood mononuclear cell (PBMC) based HIV-1 neutralization assay by donor cell pooling. Virology 2013;447:240–8.

(18)

177

Cha

pte

r 6

https://doi.org/10.1016/J.VIROL.2013.09.014.

[14] Tapia-Calle G, Born PA, Koutsoumpli G, Gonzalez-Rodriguez MI, Hinrichs WLJ, Huckriede ALW. A PBMC-Based System to Assess Human T Cell Responses to Influenza Vaccine Candidates In Vitro. Vaccines 2019;7. https://doi.org/10.3390/vaccines7040181.

[15] Ming M, Bernardo L, Williams K, Kolattukudy P, Kapoor N, Chan LG, et al. An in vitro functional assay to measure the biological activity of TB vaccine candidate H4-IC31. Vaccine

2019;37:2960–6. https://doi.org/10.1016/J.VACCINE.2019.04.035.

[16] Alvarez-Rueda N, Rouges C, Touahri A, Misme-Aucouturier B, Albassier M, Pape P Le. In vitro immune responses of human PBMCs against Candida albicans reveals fungal and leucocyte phenotypes associated with fungal persistence. Sci Rep 2020;10.

https://doi.org/10.1038/s41598-020-63344-6.

[17] Carletti T, Zakaria MK, Marcello A. The host cell response to tick-borne encephalitis virus. Biochem Biophys Res Commun 2017. https://doi.org/10.1016/.

[18] Robertson SJ, Mitzel DN, Taylor RT, Best SM, Bloom ME. Tick-borne flaviviruses: dissecting host immune responses and virus countermeasures. Immunol Res 2009;43:172–86.

https://doi.org/10.1007/s12026-008-8065-6.

[19] Yang Q, You J, Zhou Y, Wang Y, Pei R, Chen X, et al. Tick-borne encephalitis virus NS4A ubiquitination antagonizes type I interferon-stimulated STAT1/2 signalling pathway. Emerg Microbes Infect 2020;9:714–26. https://doi.org/10.1080/22221751.2020.1745094.

[20] Mold M, Shardlow E, Exley C. Insight into the cellular fate and toxicity of aluminium adjuvants used in clinically approved human vaccinations. Sci Rep 2016;6.

https://doi.org/10.1038/SREP31578.

[21] Heydenreich B, Bellinghausen I, Lund L, Henmar H, Lund G, Adler Würtzen P, et al. Adjuvant effects of aluminium hydroxide-adsorbed allergens and allergoids - differences in vivo and in vitro. Clin Exp Immunol 2014;176:310–9. https://doi.org/10.1111/cei.12294.

[22] Ko E-J, Lee Y-T, Kim K-H, Lee Y, Jung Y-J, Kim M-C, et al. Roles of Aluminum Hydroxide and Monophosphoryl Lipid A Adjuvants in Overcoming CD4+ T Cell Deficiency To Induce Isotype-Switched IgG Antibody Responses and Protection by T-Dependent Influenza Vaccine. J Immunol 2017;198:279–91. https://doi.org/10.4049/JIMMUNOL.1600173.

[23] Minor PD. Assaying the Potency of Influenza Vaccines. Vaccines 2015;3:90. https://doi.org/10.3390/VACCINES3010090.

[24] Petrovsky N. Comparative Safety of Vaccine Adjuvants: A Summary of Current Evidence and Future Needs. Drug Saf 2015;38:1059–74. https://doi.org/10.1007/s40264-015-0350-4. [25] HogenEsch H, O’Hagan DT, Fox CB. Optimizing the utilization of aluminum adjuvants in

vaccines: you might just get what you want. Npj Vaccines 2018;3:51. https://doi.org/10.1038/s41541-018-0089-x.

[26] Alculumbre SG, Saint-André V, Di Domizio J, Vargas P, Sirven P, Bost P, et al. Diversification of human plasmacytoid predendritic cells in response to a single stimulus article. Nat Immunol

(19)

178 Cha pte r 6 2018;19:63–75. https://doi.org/10.1038/s41590-017-0012-z.

[27] Baker DG, Woods TA, Butchi NB, Morgan TM, Taylor RT, Sunyakumthorn P, et al. Toll-like receptor 7 suppresses virus replication in neurons but does not affect viral pathogenesis in a mouse model of Langat virus infection. J Gen Virol 2013;94:336–47.

https://doi.org/10.1099/vir.0.043984-0.

[28] Nazmi A, Mukherjee S, Kundu K, Dutta K, Mahadevan A, Shankar SK, et al. TLR7 is a key regulator of innate immunity against Japanese encephalitis virus infection. Neurobiol Dis 2014;69:235–47. https://doi.org/10.1016/j.nbd.2014.05.036.

[29] Le Bon A, Tough DF. Links between innate and adaptive immunity via type I interferon. Curr Opin Immunol 2002;14:432–6. https://doi.org/10.1016/S0952-7915(02)00354-0.

[30] Kadowaki N, Antonenko S, Lau JYN, Liu YJ. Natural interferon α/β-producing cells link innate and adaptive immunity. J Exp Med 2000;192:219–25. https://doi.org/10.1084/jem.192.2.219. [31] Tick-Borne Encephalitis Vaccine (Inactivated) - Monograph. Eur. Pharmacopoeia (Ph. Eur.) 8th

Ed., 2008, p. 908–10.

[32] Hoefnagel MHN, Vermeulen JP, Scheper RJ, Vandebriel RJ. Response of MUTZ-3 dendritic cells to the different components of the Haemophilus influenzae type B conjugate vaccine: Towards an in vitro assay for vaccine immunogenicity. Vaccine 2011;29:5114–21.

https://doi.org/10.1016/j.vaccine.2011.05.050.

[33] Shank-Retzlaff M, Wang F, Morley T, Anderson C, Hamm M, Brown M, et al. Correlation Between Mouse Potency and in Vitro Relative Potency for Human Papillomavirus Type 16 Virus-Like Particles and Gardasil Vaccine Samples. Hum Vaccin 2005;1.

https://doi.org/10.4161/HV.1.5.2126.

[34] Xing D, Markey K, Das RG, Feavers I. Whole-cell pertussis vaccine potency assays: The Kendrick test and alternative assays. Expert Rev Vaccines 2014;13:1175–82.

https://doi.org/10.1586/14760584.2014.939636.

[35] Williams MS. Single-radial-immunodiffusion as an in vitro potency assay for human inactivated viral vaccines. Vet Microbiol 1993;37:253–62. https://doi.org/10.1016/0378-1135(93)90027-5. [36] Leenaars PPA., Kersten GF., de Bruijn ML., Hendriksen CF. An in vitro approach in quality control

of toxoid vaccines. Vaccine 2001;19:2729–33. https://doi.org/10.1016/S0264-410X(00)00510-7. [37] Růžek D, Avšič Županc T, Borde J, Chrdle A, Eyer L, Karganova G, et al. Tick-borne encephalitis

in Europe and Russia: Review of pathogenesis, clinical features, therapy, and vaccines. Antiviral Res 2019;164:23–51. https://doi.org/10.1016/j.antiviral.2019.01.014.

[38] Berges C, Naujokat C, Tinapp S, Wieczorek H, Höh A, Sadeghi M, et al. A cell line model for the differentiation of human dendritic cells. Biochem Biophys Res Commun 2005;333:896–907. https://doi.org/10.1016/j.bbrc.2005.05.171.

[39] Bosshart H, Heinzelmann M. THP-1 cells as a model for human monocytes. Ann Transl Med 2016;4:438. https://doi.org/10.21037/atm.2016.08.53.

(20)

179

Cha

pte

r 6

[40] Chanput W, Mes JJ, Wichers HJ. THP-1 cell line: An in vitro cell model for immune modulation approach. Int Immunopharmacol 2014;23:37–45. https://doi.org/10.1016/j.intimp.2014.08.002. [41] Ryu W-S. Diagnosis and Methods. Mol. Virol. Hum. Pathog. Viruses, Academic Press; 2017, p.

47–62. https://doi.org/10.1016/B978-0-12-800838-6.00004-7.

[42] Wei J, Kang X, Li Y, Wu X, Zhang Y, Yang Y. Pathogenicity of tick-borne encephalitis virus to monocytes. Wei Sheng Wu Xue Bao 2013;53:1221–5.

[43] Zhang X, Zheng Z, Liu X, Shu B, Mao P, Bai B, et al. Tick-borne encephalitis virus induces chemokine RANTES expression via activation of IRF-3 pathway. J Neuroinflammation 2016;13. https://doi.org/10.1186/s12974-016-0665-9.

[44] Tapia-Calle G, Stoel M, de Vries-Idema J, Huckriede A. Distinctive Responses in an In Vitro Human Dendritic Cell-Based System upon Stimulation with Different Influenza Vaccine Formulations. Vaccines 2017;5:21. https://doi.org/10.3390/vaccines5030021.

[45] Sasaki E, Momose H, Hiradate Y, Ishii KJ, Mizukami T, Hamaguchi I. In vitro marker gene expression analyses in human peripheral blood mononuclear cells: A tool to assess safety of influenza vaccines in humans. J Immunotoxicol 2018;15:53–62.

https://doi.org/10.1080/1547691X.2018.1447052.

[46] Chirkova T, Ha B, Rimawi BH, Oomens AGP, Hartert T V., Anderson LJ. In vitro model for the assessment of human immune responses to subunit RSV vaccines. PLoS One 2020;15. https://doi.org/10.1371/journal.pone.0229660.

[47] Zhang J, Shao J, Wu X, Mao Q, Wang Y, Gao F, et al. Type I interferon related genes are common genes on the early stage after vaccination by meta-analysis of microarray data. Hum Vaccin Immunother 2015;11:739. https://doi.org/10.1080/21645515.2015.1008884.

[48] Signorazzi A, Etna MP, Coccia EM, Huckriede A. In vitro assessment of tick-borne encephalitis vaccine: suitable human cell platforms and potential biomarkers. ALTEX - Altern Anim Exp 2021. https://doi.org/10.14573/altex.20100811.

[49] Stoel M, Pool J, de Vries-Idema J, Zaaraoui-Boutahar F, Bijl M, Andeweg AC, et al. Innate responses induced by whole inactivated virus or subunit influenza vaccines in cultured dendritic cells correlate with immune responses in vivo. PLoS One 2015;10:e0125228.

https://doi.org/10.1371/journal.pone.0125228.

[50] Geeraedts F, Bungener L, Pool J, ter Veer W, Wilschut J, Huckriede A. Whole inactivated virus influenza vaccine is superior to subunit vaccine in inducing immune responses and secretion of proinflammatory cytokines by DCs. Influenza Other Respi Viruses 2008;2:41–51.

https://doi.org/10.1111/j.1750-2659.2008.00038.x.

[51] Waris ME, Tsou C, Erdman DD, Zaki SR, Anderson LJ. Respiratory synctial virus infection in BALB/c mice previously immunized with formalin-inactivated virus induces enhanced pulmonary inflammatory response with a predominant Th2-like cytokine pattern. J Virol 1996;70.

[52] Chang TH, Liao CL, Lin YL. Flavivirus induces interferon-beta gene expression through a pathway involving RIG-I-dependent IRF-3 and PI3K-dependent NF-κB activation. Microbes

(21)

180 Cha pte r 6 Infect 2006;8:157–71. https://doi.org/10.1016/j.micinf.2005.06.014.

[53] Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, et al. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature 2006;441:101–5. https://doi.org/10.1038/nature04734.

[54] Chowdhury F, Johnson P, Williams AP. Enumeration and phenotypic assessment of human plasmacytoid and myeloid dendritic cells in whole blood. Cytom Part A 2010;77:328–37. https://doi.org/10.1002/cyto.a.20872.

[55] Westermann J, Körner IJ, Kopp J, Kurz S, Zenke M, Dörken B, et al. Cryopreservation of mature monocyte-derived human dendritic cells for vaccination: Influence on phenotype and functional properties. Cancer Immunol Immunother 2003;52:194–8.

https://doi.org/10.1007/s00262-002-0355-6.

[56] Liu Y, Olagnier D, Lin R. Host and viral modulation of RIG-I-mediated antiviral immunity. Front Immunol 2017;7:1. https://doi.org/10.3389/fimmu.2016.00662.

[57] Ranjbar S, Haridas V, Nambu A, Jasenosky LD, Sadhukhan S, Ebert TS, et al. Cytoplasmic RNA Sensor Pathways and Nitazoxanide Broadly Inhibit Intracellular Mycobacterium tuberculosis Growth. IScience 2019;22:299–313. https://doi.org/10.1016/j.isci.2019.11.001.

[58] Li Y, Xie J, Wu S, Xia J, Zhang P, Liu C, et al. Protein Kinase Regulated by dsRNA Downregulates the Interferon Production in Dengue Virus- and dsRNA-Stimulated Human Lung Epithelial Cells. PLoS One 2013;8:e55108. https://doi.org/10.1371/journal.pone.0055108.

[59] Talbot HK, Nian H, Zhu Y, Chen Q, Williams J V., Griffin MR. Clinical effectiveness of split-virion versus subunit trivalent influenza vaccines in older adults. Clin Infect Dis 2015;60:1170–5. https://doi.org/10.1093/cid/civ019.

[60] Akache B, Weeratna R, Deora A, Thorn J, Champion B, Merson J, et al. Anti-IgE Qb-VLP Conjugate Vaccine Self-Adjuvants through Activation of TLR7. Vaccines 2016;4:3. https://doi.org/10.3390/vaccines4010003.

[61] Beljanski V, Chiang C, Kirchenbaum GA, Olagnier D, Bloom CE, Wong T, et al. Enhanced Influenza Virus-Like Particle Vaccination with a Structurally Optimized RIG-I Agonist as Adjuvant. J Virol 2015;89:10612–24. https://doi.org/10.1128/jvi.01526-15.

[62] Yong HY, Luo D. RIG-I-like receptors as novel targets for pan-antivirals and vaccine adjuvants against emerging and re-emerging viral infections. Front Immunol 2018;9:1379.

https://doi.org/10.3389/fimmu.2018.01379.

[63] Wong PT, Goff PH, Sun RJ, Ruge MJ, Ermler ME, Sebring A, et al. Combined Intranasal Nanoemulsion and RIG-I Activating RNA Adjuvants Enhance Mucosal, Humoral, and Cellular Immunity to Influenza Virus. Mol Pharm 2020.

https://doi.org/10.1021/acs.molpharmaceut.0c00315.

[64] Ziegler A, Soldner C, Lienenklaus S, Spanier J, Trittel S, Riese P, et al. A New RNA-Based Adjuvant Enhances Virus-Specific Vaccine Responses by Locally Triggering TLR- and RLH-Dependent Effects. J Immunol 2017;198:1595–605. https://doi.org/10.4049/jimmunol.1601129.

(22)

181

Cha

pte

r 6

[65] Coffman RL, Sher A, Seder RA. Vaccine adjuvants: Putting innate immunity to work. Immunity 2010;33:492–503. https://doi.org/10.1016/j.immuni.2010.10.002.

[66] Goldstein MA, Tauraso NM. Effect of formalin, beta-propiolactone, merthiolate, and ultraviolet light upon influenza virus infectivity chicken cell agglutination, hemagglutination, and antigenicity. Appl Microbiol 1970;19:290–4.

[67] Budowsky EI, Friedman EA, Zheleznova NV, Noskov FS. Principles of selective inactivation of viral genome. VI. Inactivation of the infectivity of the influenza virus by the action of fl-propiolactone. Vaccine 1991;9:398–402.

[68] She YM, Cheng K, Farnsworth A, Li X, Cyr TD. Surface modifications of influenza proteins upon virus inactivation by β-propiolactone. Proteomics 2013;13:3537–47.

https://doi.org/10.1002/pmic.201300096.

[69] The European Agency for the Evaluation of Medical Products. Note for guidence on

harmonisation of requirements for influenza vaccines. Www.Eudra.Org/Emea.Html, 1997, p. 19. [70] World Health Organization (WHO). Annex 4 - Recommendations to assure the quality, safety

and efficacy of influenza vaccines (human, live attenuated) for intranasal administration. vol. 977. 2013.

[71] Edwards K, Lynfield R, Chair Janet Englund A, Kotloff K, Levy O, Long S, et al. Summary Minutes - 142nd Vaccines and Related Biological Products Advisory Committee Meeting, 2016, p. 4. [72] Pawar SD, Murtadak VB, Kale SD, Shinde P V., Parkhi SS. Evaluation of different inactivation

methods for high and low pathogenic avian influenza viruses in egg-fluids for antigen preparation. J Virol Methods 2015;222:28–33. https://doi.org/10.1016/j.jviromet.2015.05.004. [73] Uittenbogaard JP, Zomer B, Hoogerhout P, Metz B. Reactions of beta-propiolactone with

nucleobase analogues, nucleosides, and peptides: implications for the inactivation of viruses. J Biol Chem 2011;286:36198–214. https://doi.org/10.1074/jbc.M111.279232.

[74] Dembinski JL, Hungnes O, Hauge AG, Kristoffersen A-C, Haneberg B, Mjaaland S. Hydrogen peroxide inactivation of influenza virus preserves antigenic structure and immunogenicity. J Virol Methods 2014;207:232–7. https://doi.org/10.1016/j.jviromet.2014.07.003.

[75] Furuya Y, Regner M, Lobigs M, Koskinen A, Müllbacher A, Alsharifi M. Effect of inactivation method on the cross-protective immunity induced by whole “killed” influenza A viruses and commercial vaccine preparations. J Gen Virol 2010;91:1450–60.

https://doi.org/10.1099/vir.0.018168-0.

[76] King DJ. Evaluation of different methods of inactivation of Newcastle disease virus and avian influenza virus in egg fluids and serum. Avian Dis 1991;35:505–14.

https://doi.org/10.2307/1591214.

[77] Nicholson KG, Wood JM, Zambon M. Influenza. Lancet 2003;362:1733–45. https://doi.org/10.1016/S0140-6736(03)14854-4.

[78] Jonges M, Liu WM, van der Vries E, Jacobi R, Pronk I, Boog C, et al. Influenza virus inactivation for studies of antigenicity and phenotypic neuraminidase inhibitor resistance profiling. J Clin

(23)

182 Cha pte r 6 Microbiol 2010;48:928–40. https://doi.org/10.1128/JCM.02045-09.

[79] Metz B, Kersten GFA, Baart GJE, de Jong A, Meiring H, ten Hove J, et al. Identification of formaldehyde-induced modifications in proteins: reactions with insulin. Bioconjug Chem 2006;17:815–22. https://doi.org/10.1021/bc050340f.

[80] Perrin P, Morgeaux S. Inactivation of DNA by β-propiolactone. Biologicals 1995;23:207–11. https://doi.org/10.1006/biol.1995.0034.

[81] Bonnafous P, Nicolaï M-C, Taveau J-C, Chevalier M, Barrière F, Medina J, et al. Treatment of influenza virus with beta-propiolactone alters viral membrane fusion. Biochim Biophys Acta 2014;1838:355–63. https://doi.org/10.1016/j.bbamem.2013.09.021.

[82] Möller L, Schünadel L, Nitsche A, Schwebke I, Hanisch M, Laue M. Evaluation of virus inactivation by formaldehyde to enhance biosafety of diagnostic electron microscopy. Viruses 2015;7:666–79. https://doi.org/10.3390/v7020666.

[83] Eisfeld AJ, Neumann G, Kawaoka Y. Influenza A virus isolation, culture and identification. Nat Protoc 2014;9:2663–81. https://doi.org/10.1038/nprot.2014.180.

[84] Costello DA, Whittaker GR, Daniel S. Variations in pH Sensitivity, Acid Stability, and Fusogenicity of Three Influenza Virus H3 Subtypes. J Virol 2015;89:350–60.

https://doi.org/10.1128/JVI.01927-14.

[85] Luo M. Influenza Virus Entry. Viral Mol. Mach., Springer, Boston, MA; 2012, p. 201–21. https://doi.org/10.1007/978-1-4614-0980-9_9.

[86] Desbat B, Lancelot E, Krell T, Nicolaï MC, Vogel F, Chevalier M, et al. Effect of the β-propiolactone treatment on the adsorption and fusion of influenza A/Brisbane/59/2007 and A/New Caledonia/20/1999 virus H1N1 on a dimyristoylphosphatidylcholine/ganglioside GM3 mixed phospholipids monolayer at the air-water interface. Langmuir 2011;27:13675–83. https://doi.org/10.1021/la2027175.

[87] Wilton T, Dunn G, Eastwood D, Minor PD, Martin J. Effect of Formaldehyde Inactivation on Poliovirus. J Virol 2014;88:11955–64. https://doi.org/10.1128/jvi.01809-14.

[88] Kuykendall JR, Bogdanffy MS. Efficiency of DNA-histone crosslinking induced by saturated and unsaturated aldehydes in vitro. Mutat Res Lett 1992;283:131–6. https://doi.org/10.1016/0165-7992(92)90145-8.

[89] Ma TH, Harris MM. Review of the genotoxicity of formaldehyde. Mutat Res Genet Toxicol 1988;196:37–59. https://doi.org/10.1016/0165-1110(88)90027-9.

[90] Marshall D, Sealy R, Sangster M, Coleclough C. TH cells primed during influenza virus infection provide help for qualitatively distinct antibody responses to subsequent immunization. J Immunol 1999;163:4673–82.

[91] Eftekhari Z, Mehrbod P, Vaez J, Hosseini S, Eslami Jodairi S. Evaluation of the effects of concentration, duration and temperature of incubation with Beta-Propiolactone on inactivation of cell-derived rabies virus. Vaccine Res 2016;3:26–9.

(24)

183

Cha

pte

r 6

[92] Jagt HJM, Bekkers MLE, van Bommel SAJT, van der Marel P, Schrier CC. The influence of the inactivating agent on the antigen content of inactivated Newcastle disease vaccines assessed by the in vitro potency test. Biologicals 2010;38:128–34.

https://doi.org/10.1016/j.biologicals.2009.07.006.

[93] Yang Y, Li H, Li Z, Zhang Y, Zhang S, Chen Y, et al. Size-exclusion HPLC provides a simple, rapid, and versatile alternative method for quality control of vaccines by characterizing the assembly of antigens. Vaccine 2015;33:1143–50. https://doi.org/10.1016/j.vaccine.2015.01.031.

[94] Ugozzoli M, Laera D, Nuti S, Skibinski DAG, Bufali S, Sammicheli C, et al. Flow cytometry: An alternative method for direct quantification of antigens adsorbed to aluminum hydroxide adjuvant. Anal Biochem 2011;418:224–30. https://doi.org/10.1016/j.ab.2011.07.012.

[95] Régnier M, Metz B, Tilstra W, Hendriksen C, Jiskoot W, Norde W, et al. Structural perturbation of diphtheria toxoid upon adsorption to aluminium hydroxide adjuvant. Vaccine 2012;30:6783– 8. https://doi.org/10.1016/j.vaccine.2012.09.020.

[96] Metz B, Jiskoot W, Hennink WE, Crommelin DJA, Kersten GFA. Physicochemical and immunochemical techniques predict the quality of diphtheria toxoid vaccines. Vaccine 2003;22:156–67. https://doi.org/10.1016/j.vaccine.2003.08.003.

[97] Etna MP, Giacomini E, Severa M, Pardini M, Aguilo N, Martin C, et al. A human dendritic cell-based in Vitro model to assess Mycobacterium Tuberculosis SO2 vaccine immunogenicity. ALTEX 2014;31:397–406. https://doi.org/10.14573/altex.1311041.

[98] Vandebriel RJ, Hoefnagel MHN. Dendritic cell-based in vitro assays for vaccine immunogenicity. Hum Vaccines Immunother 2012;8:1323–5. https://doi.org/10.4161/hv.21350.

[99] Rechtien A, Richert L, Lorenzo H, Martrus G, Hejblum B, Dahlke C, et al. Systems Vaccinology Identifies an Early Innate Immune Signature as a Correlate of Antibody Responses to the Ebola Vaccine rVSV-ZEBOV. Cell Rep 2017;20:2251–61. https://doi.org/10.1016/j.celrep.2017.08.023. [100] Galassie AC, Link AJ. Proteomic contributions to our understanding of vaccine and immune

responses. Proteomics Clin Appl 2015;9:972–89. https://doi.org/10.1002/prca.201500054. [101] Raeven RHM, Brummelman J, Pennings JLA, Nijst OEM, Kuipers B, Blok LER, et al. Molecular

signatures of the evolving immune response in mice following a Bordetella pertussis infection. PLoS One 2014;9:19–22. https://doi.org/10.1371/journal.pone.0104548.

[102] Waickman AT, Victor K, Li T, Hatch K, Rutvisuttinunt W, Medin C, et al. Dissecting the

heterogeneity of DENV vaccine-elicited cellular immunity using single-cell RNA sequencing and metabolic profiling. Nat Commun 2019;10:1–16. https://doi.org/10.1038/s41467-019-11634-7. [103] Luciani F, Bull RA, Lloyd AR. Next generation deep sequencing and vaccine design: Today and

tomorrow. Trends Biotechnol 2012;30:443–52. https://doi.org/10.1016/j.tibtech.2012.05.005. [104] Fauci AS, Pratt KRK, Whalen G. Activation of Human B Lymphocytes | The Journal of

Immunology. J Immunol 1976;117:2100–4.

[105] Borrebaeck CAK, Danielsson L, Moller SA. Human monoclonal antibodies produced by primary in vitro immunization of peripheral blood lymphocytes. Proc Natl Acad Sci U S A 1988;85:3995–

(25)

184 Cha pte r 6 9. https://doi.org/10.1073/pnas.85.11.3995.

[106] Nandin IS, Fong C, Deantonio C, Torreno-Pina JA, Pecetta S, Maldonado P, et al. Novel in vitro booster vaccination to rapidly generate antigen-specific human monoclonal antibodies. J Exp Med 2017;214:2471–90. https://doi.org/10.1084/jem.20170633.

[107] Drake DR, Singh I, Nguyen MN, Kachurin A, Wittman V, Parkhill R, et al. In Vitro Biomimetic Model of the Human Immune System for Predictive Vaccine Assessments. Disruptive Sci Technol 2012;1:28–40. https://doi.org/10.1089/dst.2012.0006.

[108] World Health Organization (WHO). Annex 1 WHO guidelines on nonclinical evaluation of vaccines. 2005.

[109] MacLeod M. Learning lessons from MVA85A, a failed booster vaccine for BCG: We must review how we use animal data to underpin clinical trials in humans. BMJ 2018;360.

https://doi.org/10.1136/bmj.k66.

[110] Greek R. Animal Models and the Development of an HIV Vaccine. J AIDS Clin Res 2012;2012:8. https://doi.org/10.4172/2155-6113.S8-001.

[111] Akhtar A. The Flaws and Human Harms of Animal Experimentation. Cambridge Q Healthc Ethics 2015;24:407–19. https://doi.org/10.1017/S0963180115000079.

[112] Tonks A. HIV research: Quest for the AIDS vaccine. BMJ 2007;334:1346–8.

[113] Johnston MI, Fauci AS. An HIV Vaccine — Evolving Concepts. N Engl J Med 2007;356:2073–81. https://doi.org/10.1056/nejmra066267.

[114] Wong PT, Leroueil PR, Smith DM, Ciotti S, Bielinska AU, Janczak KW, et al. Formulation, High Throughput In Vitro Screening and In Vivo Functional Characterization of Nanoemulsion-Based Intranasal Vaccine Adjuvants. PLoS One 2015;10:e0126120.

https://doi.org/10.1371/journal.pone.0126120.

[115] Nederlandse Voedsel- en Warenautoriteit. Jaaroverzicht dierproeven en proefdieren | Inspectieresultaat. 2018.

[116] Jilka RL. The Road to Reproducibility in Animal Research. J Bone Miner Res 2016;31:1317–9. https://doi.org/10.1002/jbmr.2881.

[117] Gibert R, Alberti M, Poirier B, Jallet C, Tordo N, Morgeaux S. A relevant in vitro ELISA test in alternative to the in vivo NIH test for human rabies vaccine batch release. Vaccine 2013;31:6022–9. https://doi.org/10.1016/j.vaccine.2013.10.019.

[118] Maas PA, de Winter MPM, Venema S, Oei HL, Claassen IJTM. Antigen quantification as in vitro alternative for potency testing of inactivated viral poultry vaccines. Vet Q 2000;22:223–7. https://doi.org/10.1080/01652176.2000.9695063.

[119] Perrin P, Morgeaux S, Sureau P. In vitro rabies vaccine potency appraisal by ELISA: Advantages of the immunocapture method with a neutralizing anti-glycoprotein monoclonal antibody. Biologicals 1990;18:321–30. https://doi.org/10.1016/1045-1056(90)90037-Z.

Referenties

GERELATEERDE DOCUMENTEN

Despite the experimental limitations in studying the formulated vaccine Encepur, likely due to its aluminum hydroxide content, we obtained important information on the viral

Using RT-qPCR analysis, we showed that human peripheral blood mononuclear cells (PBMCs) stimulated with non-adjuvanted TBE vaccine (NAV) displayed changes in the expression of

Studies report that FA inactivation damaged the binding ability of the virus at conditions (incubation time, concentration and temperature) similar to those tested in our

Seguendo l’interazione delle particelle con le cellule, abbiamo quindi valutato la capacità del virus inattivato di legarsi alle membrane cellulari (sfruttando le proprietà

A primary cell-based system can successfully be included in an array of in vitro methods for release testing of non-adjuvanted TBE vaccine batches. Activation of the type

Through comparing the different cross-protective immune mechanisms induced by sequential live virus infection and immunization, we conclude that cross-reactive antibodies,

Het werk beschreven in dit proefschrift draagt bij aan een beter begrip van kruisbeschermende immuunmechanismen door te achterhalen welke rol verschillende immuunreacties hebben in

Through comparing the different cross-protective immune mechanisms induced by sequential live virus infection and immunization, we conclude that cross-reactive antibodies,