university of groningen novel perspectives for …and h1n1 pandemic vaccines, several adjuvants,...

19
University of Groningen Novel perspectives for influenza vaccine formulation and administration Saluja, Vinay 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: 2010 Link to publication in University of Groningen/UMCG research database Citation for published version (APA): Saluja, V. (2010). Novel perspectives for influenza vaccine formulation and administration: L. lactis cell wall derived adjuvant, stable dry powder vaccines and mucosal vaccine delivery. Groningen: s.n. 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. Download date: 29-06-2020

Upload: others

Post on 20-Jun-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

University of Groningen

Novel perspectives for influenza vaccine formulation and administrationSaluja, Vinay

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite fromit. Please check the document version below.

Document VersionPublisher's PDF, also known as Version of record

Publication date:2010

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):Saluja, V. (2010). Novel perspectives for influenza vaccine formulation and administration: L. lactis cell wallderived adjuvant, stable dry powder vaccines and mucosal vaccine delivery. Groningen: s.n.

CopyrightOther 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 theauthor(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policyIf you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediatelyand investigate your claim.

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

Download date: 29-06-2020

Page 2: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

p a r t O N eGeM PArtIcLeS AS A PotentIAL neW AdJuVAnt For InFLuenzA VAccIneS

Page 3: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes
Page 4: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

InFLuenzA AntIGen-SPArInG By IMMune StIMuLAtIon WItH GrAM-PoSItIVe enHAncer MAtrIx (GeM) PArtIcLeS

Vinay Saluja1, Marinella r. Visser1, Wouter t. Veer2, Maarten L. van roosmalen3, Kees Leenhouts3, Wouter L.J. Hinrichs1, Anke Huckriede2

and Henderik W. Frijlink1

1department of Pharmaceutical technology and Biopharmacy, university of Groningen,A. deusinglaan 1, 9713 AV, Groningen, the netherlands

2department of Medical Microbiology, Molecular Virology Section, university Medicalcenter and university of Groningen, A. deusinglaan 1, 9713 AV, Groningen, the netherlands

3Mucosis BV, Meditech center, L.J. zielstraweg 1, 9713 Gx, Groningen, the netherlands

Vaccine (accepted)

c H a p t e r 2

Page 5: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

cHAPter 2

24

abstractGram-positive enhancer matrix (GEM) particles, produced from non-genetically modified Lactococcus lactis bacteria have an inherent immunostimulatory activity. It was investigated whether co-administration of GEM particles can reduce the amount of influenza subunit vaccine (HA) necessary to protect mice from viral infection. Decreasing HA amounts of 5, 1, 0.2, and 0.04 µg admixed with GEM particles were tested in intramuscular immunizations. Combinations of GEM and seasonal HA (A/Wisconsin/67/2005 [H3N2]) induced significantly higher systemic and better Th1/Th2-type balanced immune responses than HA alone. Addition of GEM to 0.04 µg HA resulted in similar HI titers as 1-5 µg non-adjuvanted HA. To test the protective efficacy of the adjuvanted combination, mice were immunized with influenza subunit vaccine A/PR/8/34 (H1N1) and then challenged with live virus (A/PR/8/34). Mice immunized with 1 µg HA + GEM showed undetectable virus titers in the lungs 5 days after challenge, whereas mice immunized with 1 µg HA alone showed detectable levels of virus in the lungs. Interestingly, mice vaccinated with the 0.04 µg HA + GEM vaccine demonstrated reduced lung virus titers and a reduction in weight that was similar as that in mice vaccinated with 1 µg non-adjuvanted HA. These results indicate that the use of GEM as immunostimulant allows for a strong reduction in the antigen dose as compared to the benchmark vaccine by using GEM particles. Thus, addition of GEM can strongly potentiate immunogenicity of influenza subunit vaccine both quantitatively and qualitatively.

Page 6: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

InFLuenzA AntIGen-SPArInG By GeM PArtIcLeS

2

25

1. iNtroductioNThe recent outbreak of influenza A (H1N1)swl-2009 virus showed that influenza viruses still pose a potential threat to the human population [1]. In addition to periodic pandemics, annual seasonal influenza infection cause high morbidity and mortality [2]. The most effective way of controlling and preventing influenza infection is immunization [2, 3]. However, every seasonal epidemic and occasional pandemic is caused by a different virus strain, because of the variation in the viral spike proteins, haemagglutinin and neuraminidase. Therefore, new vaccines based on the varying strains are required for each immunization [4]. Another problem is the limited immunogenicity of conventional inactivated influenza vaccines [5]. Moreover, in a pandemic scenario vaccines should be produced fast and in huge quantities, which is a burden on constrained global vaccine production facilities [6].

Traditionally, adjuvants are used to overcome problems like low immunogenicity of vaccines [7]. For the past decades, aluminium-based adjuvants have been mostly used to boost the poor immunogenicity of several vaccines [8, 9]. In the context of H5N1 and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes have been tested for their antigen-sparing and immune-enhancing potential [10-20]. In particular, the extent of antigen-sparing incurred by oil-in-water based adjuvants (MF-59 and AS03) was substantial [20].

Recently, a novel adjuvant has been developed that consists of peptidoglycan spheres produced from the food-grade bacterium Lactococcus lactis [21]. The bacterium is killed at low pH (pH 1) and high temperature (99 °C) which generates the so-called Gram-positive enhancer matrix (GEM) particles [22]. Due to the acid treatment at high temperature these particles lack the viable intracellular content and surface proteins [22]. The thick peptidoglycan based bacterial cell wall provides structural rigidity to the particles. GEM particles present a new adjuvant system which can strongly boost vaccine immunogenicity [23, 24]. The immunostimulatory activity of GEM particles is attributed to the activation of antigen presenting cells by toll like receptor 2 (TLR-2) [25]. Malaria parasite, Yersinia pestis and pneumococcal antigens when displayed on GEM particles induced high immune responses in mouse models [24-26]. Moreover, it was shown that GEM particles admixed with influenza subunit vaccine (HA) elicit strong systemic and local immune responses when administered via the intranasal route [27]. Therefore, GEM particles are considered to be a promising adjuvant for influenza immunizations.

In this study, we investigated the effect of GEM particles on HA vaccine efficacy when administered via the i.m. route. Furthermore, we investigated whether GEM particles allow vaccine dose reduction. For this purpose, mice were immunized with different doses of HA with and without the addition of a standard amount of GEM particles.

Page 7: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

cHAPter 2

26

2. materials aNd methods

2.1 virus and vaccinesInfluenza monovalent subunit vaccine of strain A/Wisconsin/67/2005 (H3N2) was kindly provided by Solvay Pharmaceuticals. The concentration of HA in the vaccine was determined using the single radial immunodiffusion assay [28]. Influenza virus A/PR/8/34 (HIN1) was obtained from NIBSC, UK. The virus was propagated in the allantoic fluid of embryonated chicken eggs and stored at -70 °C. PR8 subunit vaccine was produced as described earlier [29].

2.2 gem preparationL. lactis strain MG1363acmAΔ1 was cultured overnight. Subsequently, cells were washed once with sterile distilled water. The cells were resuspended in 10% trichloroacetic acid (TCA) and placed in a hot water bath of 99 °C for 30 minutes. The acid and heat treatment killed the bacteria. After acid and heat treatment, the GEM particles were pelleted, washed three times in phosphate buffered saline (PBS; Dulbecco’s Phosphate Buffered Saline without CaCl2 and MgCl2) and finally resuspended in PBS and stored at 4 °C. One unit (1 U) of GEM particles was defined as approximately 2.5 x 109 particles.

2.3 dose-response immunizations with influenza subunit vaccine a/wisconsin/67/2005 (h3N2)Animal experiments were approved by the Committee for Animal Experimentation of the University of Groningen, The Netherlands according to the guidelines provided by Dutch Animal Protection Act. Balb/c mice (6-8 weeks) purchased from Harlan, Zeist, The Netherlands were used for all immunization experiments. The mice were divided in 8 groups of 8 animals each. All 8 groups of animals were immunized with prime vaccination on day 0 and one booster vaccinations on day 21 with four different amounts of influenza subunit vaccine (5 μg, 1, 0.2, and 0.04 HA) with and without GEM particles (1 unit). HA and the GEM particles were admixed just before the immunization. All mice were injected i.m. in both their calf muscles with a total of 50 µl vaccine or vaccine-adjuvant mixture, equally divided over both injection sites. The mice were sacrificed one week after the booster vaccination, i.e., on day 28. After the animals were sacrificed the spleens were harvested and stored in supplemented Iscove’s Modified Dulbecco’s Glutamax medium (Invitrogen, The Netherlands) with 5 % Fetal Calf Serum, 1% penicillin/streptomycin and 50 µM β-mercaptoethanol at 4 °C.

Blood samples were drawn on day 21 (before booster) by orbital puncture and day 28 by heart puncture. Sera were obtained by centrifugation of blood at 1200 x g for 5 min and the samples were subsequently stored at -20 °C until further analysis.

Page 8: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

InFLuenzA AntIGen-SPArInG By GeM PArtIcLeS

2

27

2.4 haemagglutination inhibition assayHaemagglutination inhibition (HI) titers in serum were determined as described previously [27]. Briefly, 25 % kaolin suspension was added to inactivated sera (56 °C for 30 min). After centrifugation at 1200 x g, 50 µl of the supernatant was transferred in duplicate to 96 well round bottom plate (Greiner, Alphen a/d Rijn, Netherlands) and serially diluted twofold in PBS. Next, four haemagglutination units of inactivated A/Wisconsin/67/2005 (H3N2) influenza virus was added and incubated for 40 min at room temperature. Finally, 50 µl of 1 % guinea pig red blood cells were added and incubated for 2 h at room temperature. The highest dilution capable of preventing haemagglutination was scored as HI titer.

2.5 elisa assaysTotal influenza-specific IgG and IgG subtypes were determined using ELISA assays as described previously [27]. Briefly, the plates were incubated overnight with HA, blocked with 3% bovine serum albumin (Sigma-Aldrich, The Netherlands) and incubated with serial dilutions of sera for 1.5 h at 37 °C. Next, the plates were washed three times and incubated with horseradish peroxidase-conjugated goat antibodies directed against mouse IgG, IgG1 and IgG2a (Southern Biotech, Birmingham, AL, USA). Finally, the plates were developed using substrate solution (0.02 % 1, 2- phenylenediamine-dihydrochloride (Sigma) in 50 mM phosphate buffer pH 5.6, containing 0.006 % H2O2) for 30 min. The reaction was stopped by addition of 2 M H2SO4 and absorbance was read at 490 nm. Titers reported are the reciprocal of the calculated sample dilution corresponding with an A490 ≥ 0.2 after background correction.

2.6 elispot assayThe Elispot assay was performed as described earlier [27]. Briefly, plates were incubated overnight at 4 °C with anti-mouse interferon-γ (IFN-γ) and interleukin-4 (IL-4) (BD, Pharmingen, Belgium). After washing the plates were blocked for 1 h at 37 °C and spleen cells were added to the plates with or without subunit vaccine. Next, the cells were lysed after overnight incubation, and incubated with biotinylated anti-mouse IFN-γ and IL-4 antibodies (BD Pharmingen, Belgium) at a concentration of 0.125 µg. Plates were then incubated with Streptavidin alkaline phosphatase (BD Pharmingen, Belgium) for 1 h at 37 °C. Finally, the spots were developed using substrate solution consisting of 1 mg/ml 5-bromo-4-chloro-3-indolylphosphate, 0.92 % w/v 2-amino-2-methyl-1-propanol, 0.08 µl/ml TritonX-405, 1 M MgCl2 and 6 mg/ml agarose. The spots were scanned using an Elispot reader (A.EL.VIS GmbH, Hannover, Germany) and counted manually using Image J Software (National Institute of Health, USA).

2.7 immunization and viral challenge experiment with a/pr/8/34 (h1N1)For immunization and viral challenge experiment, mice were divided in five groups of 10 animals each. Four groups of animals were immunized with prime vaccination on

Page 9: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

cHAPter 2

28

day 0 and one booster vaccination on day 21. In the control group, mice were injected with PBS only. Mice were immunized i.m. with 1, 0.2 or 0.04 µg HA from A/PR/8/34 adjuvanted with GEM particles (1 U). One group of mice was immunized with 1 µg HA from A/PR/8/34 without adjuvant. This dose of the non-adjuvanted vaccine was chosen, as it was shown to be the minimum dose required for protection of mice from viral infection (M. Liu, submitted for publication). On day 28, mice were anaesthetized and challenged with 2 × 102 TCID50 A/PR8 influenza virus [29]. After challenge, the mice were monitored every 12 h for clinical signs by weighing and observing their activity and appearance. From each group five animals were sacrificed 3 days after challenge and the remaining mice were sacrificed 5 days after challenge. The lung lobes were collected and stored at 4 °C in PBS.

2.8 determination of virus titers in lungs of challenged miceThe lung virus titers were determined as described previously [29]. Briefly, lungs were homogenized and supernatants were collected by centrifugation. The supernatants were twofold diluted in 96 well plates. Fifty microliter of the diluted supernatant was added to a monolayer of MDCK cells and incubated for 1h. Then 7.5 μg TPCK trypsin (Sigma) was added per well followed by 72 h of incubation. After 3 days, the supernatants were transferred to round-bottom 96-well plate. Next, 50 μl of a 1 % guinea pig erythrocyte suspension were added and incubated for 2 h. Dilution at which hemagglutination was observed, indicated the virus titer in the lungs. The 10log virus titer was calculated per gram of lung tissue.

2.9 statistical analysisStatistical analyses were performed using Anova test and Bonferroni’s correction for multiple comparisons. The results are presented as mean ± standard error mean (S.E.M.) unless indicated otherwise.

3. results

3.1 effect of gem particles on the serum antibody responsesIn order to evaluate the antigen-sparing potential of GEM particles, mice were immunized with different doses of influenza subunit vaccine containing 5, 1, 0.2, and 0.04 µg HA with and without GEM particles. The standard assay for predicting the protective efficacy of an influenza vaccine is the determination of the HI titers. An HI titer > 40 ( i.e. 2log HI > 5.3) is considered to be protective in humans [30]. After one immunization, only the GEM containing vaccine doses of 5, 1, or 0.2 µg HA were able to induce HI titers > 2log5.3 (Fig. 1A). Addition of GEM particles to the different amounts of HA increased the titers by 2-5 folds. After the booster immunization, all vaccines reached HI titers > 2log5.3 (Fig. 1B). Interestingly, GEM containing HA doses still significantly enhanced the HI titers after the booster immunization as compared

Page 10: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

InFLuenzA AntIGen-SPArInG By GeM PArtIcLeS

2

29

to their non-adjuvanted counterparts (Fig. 1B). Mice vaccinated with 1 and 5 µg HA without GEM induced similar HI titers, suggesting a plateau in the immune response. Nevertheless, addition of GEM to these vaccines significantly enhanced the HI titers (Fig 1B). The lower HA dose vaccines (0.2 and 0.04 µg) with GEM induced similar titers compared to those induced by the non-adjuvanted 5 and 1 µg HA vaccines.

5µg 1µg 0.2µg 0.04µg2

3

4

5

6

7HA HA +GEMA

* **

*

2 log

HI t

iter

5µg 1µg 0.2µg 0.04µg2

4

6

8

10

12 *HA HA +GEMB

*

* *

2 log

HI t

iter

At day 21 after the first immunization, IgG responses in mice immunized with 5 µg HA without GEM particles were similar to responses in mice immunized with 1 µg HA (Fig. 2A). However, after booster immunization, antibody responses in mice immunized with 1 µg HA were significantly higher than responses in mice immunized with 5 µg HA, indicating that 1 µg HA is the optimal dose for immunization without adjuvant. GEM adjuvanted HA vaccines (5, 1, and 0.2 µg HA + GEM) induced significantly higher IgG titers than 1 µg non-adjuvanted HA after the first immunization (Fig. 2A). However, after the booster immunization no significant differences were observed between adjuvanted and non-adjuvanted HA immunizations at the corresponding HA dose, except for the 5 µg dose (Fig. 2B).

3.2 effect of gem particles on the type of immune responseIn order to determine the type and the quality of the immune response, the IgG subtypes and the numbers of IFN-γ and IL-4 producing T cells in the spleen were determined. As expected, i.m. immunization with non-adjuvanted HA induced higher IgG1 than IgG2a titers (Fig. 3A and 3B), indicating a Th2-type dominated immune response [27, 31]. Addition of GEM to HA vaccines (5, 1, and 0.04 µg) induced similar IgG1 titers compared to non-adjuvanted vaccines. Only the 0.2 µg HA + GEM vaccine induced

Figure 1. Geometric mean HI titers 3 weeks after prime and 1 week after booster immunization. Influenza antigen (A/Wisconsin/H3N2) specific HI titers on day 21 (A) after primary immunization and day 28 (B) after prime and boost immunization with decreasing HA doses (5, 1, 0.2, and 0.04 μg) without (white bars) and with (black bars) GEM particles. The dashed line represents an HI titer of 2log (40) = 5.3. Titers are given as mean per experimental group +/- S.E.M. * p < 0.05.

Page 11: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

cHAPter 2

30

significantly higher IgG1 responses than 0.2 µg non-adjuvanted vaccine. However, at an antigen dose of 5 and 0.2 µg, GEM adjuvanted vaccines induced significantly higher IgG2a responses than non-adjuvanted vaccines.

The type of immune response was further evaluated by determining the antigen-specific IFN-γ and IL-4 responses in 4 mice from each group. As shown in Fig. 4A, there was a moderate stimulatory effect on the number of INF-γ producing cells in the groups receiving HA vaccine (1, 0.2, and 0.04 µg) with GEM as compared to the non-adjuvanted groups. In addition, all the mice immunized with HA + GEM vaccines showed significantly less IL-4 producing cells than mice immunized with vaccine only.

Figure 2. Geometric mean IgG titers 3 weeks after prime and 1 week after booster immunization. Influenza antigen (A/Wisconsin/H3N2) specific IgG titers on day 21 (A) after primary immunization and day 28 (B) after prime and boost immunization with decreasing HA doses (5, 1, 0.2, and 0.04 μg) without (white bars) and with (black bars) GEM particles. Titers are given as mean per experimental group +/- S.E.M. * p < 0.05.

5µg 1µg 0.2µg 0.04µg2.0

2.5

3.0

3.0

4.0

4.5

5µg 1µg 0.2µg 0.04µg2

3

4

5

A B

***

**

10lo

g Ig

G ti

ter

10lo

g Ig

G ti

ter

HA HA +GEM HA HA +GEM

5µg 1µg 0.2µg 0.04µg2

3

4

5

6

5µg 1µg 0.2µg 0.04µg2.0

2.5

3.0

3.5

4.0

4.5A B

* *

*

10lo

g Ig

G1

titer

HA HA +GEM HA HA +GEM

10lo

g Ig

G2a

tite

r

Figure 3. Geometric mean IgG1 and IgG2a titers 1 week after booster immunization. Influenza antigen (A/Wisconsin/H3N2) specific IgG1 (A) and IgG2a (B) titers four weeks after prime and boost vaccination in sera of mice immunized with decreasing HA doses (5, 1, 0.2, and 0.04 μg) without (white bars) and with (black bars) GEM particles. Titers are given as mean per experimental group +/- S.E.M. * p < 0.05.

Page 12: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

InFLuenzA AntIGen-SPArInG By GeM PArtIcLeS

2

31

As an indication of the Th1/Th2 balance the ratio of IFN-γ/ IL-4 was determined (Fig. 4B). When admixed with lower antigen doses (1, 0.2, and 0.04 µg) GEM skewed the response towards a Th1 type. This improvement in the Th1/Th2 balance was primarily due to the reduction of IL-4 producing T-cells.

Cyt

okin

e pr

oduc

ing

cells

/106 c

ells

0

100

200

300

400

5µg 1µg 0.2µg 0.04µg5µg 1µg 0.2µg 0.04µg+GEM +GEM+GEM +GEM

IL-4 IFN-γ

A

B

5µg 1µg 0.2µg 0.04µg0

0.5

1.0

1.5

2.0

2.5

IFN

-γ /

IL-4

ratio

HA HA +GEM

Figure 4. T-helper cell responses. (A) IL-4 (white bars), IFN-γ (black bars) in mice (n=4 per group) immunized with decreasing HA doses (5, 1, 0.2 and 0.04 μg) without and with GEM particles. (B) The IFN-γ/IL-4 ratio of the effector cell population from mice immunized with influenza subunit vaccine without and with GEM particles. The ratio presented was calculated as the ratio of mean values of IFN-γ producing splenocytes and the mean of the IL-4 producing splenocytes. Numbers are given as mean per experimental group +/- S.E.M.

In conclusion, addition of GEM particles to subunit influenza vaccine offers the possibility of antigen dose sparing and/or improvement of the quality of the immune response. Accordingly, the three lower doses (1, 0.2, and 0.04 µg) of HA + GEM were chosen for investigating the protective efficacy in a mice challenge study.

3.3 immunization and live virus a/pr/8/34 (h1N1) challenge We next investigated if GEM particles allow reduction of the antigen dose without compromising the protective potential of the vaccination. Mice were immunized with 1, 0.2 or 0.04 µg A/PR/8/34 influenza subunit vaccine with GEM or with 1 µg non-adjuvanted vaccine. Phosphate buffer was administered to the control group. Mice were challenged i.n. with live virus on day 28 after immunization and then observed for clinical symptoms for 5 days. Five mice per group were sacrificed on day 3 after challenge and the remaining five mice on day 5 after challenge for the determination of lung virus titers.

Mice injected with PBS showed significant weight loss (Fig. 5A) after 3 days (9%) and 5 days (15%). In addition, after 3 days and onwards clinical signs of illness like ruffled fur and reduced activity were observed in the control (PBS) group. In contrast, no clinical signs of illness were observed in the immunized mice 5 days after challenge.

Page 13: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

cHAPter 2

32

The mice immunized with 1 µg HA + GEM showed some gain in weight, while the mice immunized with 1 µg HA showed a minor reduction in weight.

All the vaccinated mice showed significant reduction in lung virus titers (Fig. 5B) compared to the control (PBS) group. After 3 days, viral replication was detected in all mice receiving non-adjuvanted vaccine. In contrast, two out of five mice immunized with 1 µg HA + GEM had no detectable virus indicating that they were fully protected from viral replication in the lungs. Interestingly, 5 days after challenge all the mice vaccinated with 1 µg HA + GEM had undetectable lung virus titers, whereas mice immunized with 1 µg non-adjuvanted HA still showed lung virus titers in four out of five mice. In the mice vaccinated with 0.2 and 0.04 µg HA + GEM no significant differences in lung virus titers were observed compared to mice vaccinated with 1 µg HA after 3 days. On day 5, only two out of five mice showed detectable lung virus titers in the 0.2 µg HA + GEM group. After 5 days, lung virus titers in the 0.04 μg HA + GEM group were not significantly different (p = 0.81) to those in the non-adjuvanted 1 μg HA group.

Aver

age

rel.

body

wt.

(%)

Days0 1 2 3 4 5

80

85

90

95

100

105

110

PBS 1µg 1µg+GEM 0.2µg+GEM 0.04µg+GEM

A

10lo

g vi

rus

titer

s in

lung

s (p

er g

)

PBS 1µg 1µg+GEM

0.2µg+GEM

0.04µg+GEM

0

2

4

6

85/55/5

5/5

3/5

5/5

5/5

4/5

0/5

2/5

4/5

BDay 3 after challengeDay 5 after challenge

Figure 5. Immunization and challenge studies. A. Relative change in average body weight after challenge with live virus (A/PR/8/34). Mice were immunized twice i.m. with 1 µg A/PR/8/34 subunit vaccine (open circle), 1 µg subunit vaccine with GEM particles (black triangle), 0.2 µg subunit vaccine with GEM particles (open triangle) and 0.04 µg subunit vaccine with GEM particles (black square). Control mice were injected with PBS (black circle). 4 weeks after the first immunization mice were challenged by i.n. inoculation of live A/PR/8/34 virus. Weight was measured twice daily and is given as mean per experimental group +/- S.E.M. B. Lung virus titers were determined in mice 3 days (white bars) and 5 days (black bars) after challenge. Average lung virus titers +/- S.E.M. are given. The numbers above the bars indicate the number of mice per group with detectable titers.

Page 14: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

InFLuenzA AntIGen-SPArInG By GeM PArtIcLeS

2

33

4. discussioNDespite recent advances in vaccine production, still a huge gap has to be overcome to fulfill the global vaccine need in case of a pandemic [6, 32]. One of the important strategies for increased immunogenicity and antigen-sparing is to use adjuvants [33]. In this study, we showed that GEM particles strongly reduced the amount of antigen required for protection of mice from a viral infection. The systemic immune responses were significantly increased by addition of GEM to the influenza subunit vaccine. GEM particles also improved the quality of the immune response by shifting the response from a Th2-type dominated to a balanced Th1/Th2-type immune response. The addition of GEM particles also enabled to achieve protective HI titers in naïve mice after a single immunization for an HA dose as low as 0.2 μg, whereas 5 and 1 μg of HA without GEM did not reach protective HI titers at that stage.

Earlier studies have demonstrated the immunostimulatory properties of the GEM particles [23, 25-27]. Yet, this is the first study that demonstrates the antigen-sparing potential of the GEM particles. The addition of GEM particles resulted in: (i) higher HI titers, (ii) a more balanced Th1/Th2-type immune response, (iii) a reduction in viral titers in the lungs or even complete viral clearance after challenge, and (iv) protection at lower HA dose levels. The increased viral clearance in mice immunized with adjuvanted vaccine (1 and 0.2 µg HA) might be due to the induction of higher IgG2a titers and a comparatively high INF-γ/IL-4 ratio. Moreover, the high INF-γ/IL-4 ratio indicates that GEM particles skew the response towards Th1-type, which is considered beneficial [29, 34-36]. Furthermore, the highest HA dose (1 µg) without GEM and the five-fold lower HA dose (0.2 µg) with GEM induced a similar level of protection. Thus, addition of GEM particles to HA subunit vaccine allows for at least a five-fold reduction of the HA dose with full retention of the protective potential. The results indicated that even a 25-fold reduction could be possible, however, this warrants further investigations.

Several bacterial cell-wall components are known to activate the innate immune system by activation of TLRs [37]. Recently, it was shown that GEM particles bind specifically to TLR-2 and stimulate both human and mouse dendritic cells. Moreover, it was shown that activation of TLR-2 receptors in human peripheral blood mononuclear cells can shift the immune response towards the Th1-type [38]. In our study, we showed that co-administration of subunit influenza vaccine and GEM particles significantly potentiated and modulated the immune responses. Thus, it can be speculated that the activation of the TLR-2 receptors might have led to a significant decrease in IL-4 responses and a moderate increase in INF-γ response in this study. Major obstacles for adjuvant development are safety and cost issues [39]. GEM particles have several advantages over other adjuvant systems [12, 40-42]: (i) they are not viable and produced from a generally recognized as safe (GRAS) probiotic organism (L. lactis) that demonstrated it’s safety in several pre-clinical toxicity studies [23-27], (ii) the production technology for GEM particles is relatively simple and a GMP process that

Page 15: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

cHAPter 2

34

can be adapted for inexpensive large scale production is available [21], and (iii) GEM particles are very stable and can be stored for a long period even at room temperature [21]. These properties make GEM particles an attractive adjuvant for seasonal as well as pandemic influenza vaccines.

5. coNclusioNsGEM particles are a promising adjuvant for influenza subunit vaccines. The addition of GEM particles to HA subunit vaccine resulted in significantly enhanced immune responses. Additionally, the incorporation of the GEM particles in the vaccines also resulted in Th1 skewing of the immune response. Furthermore, lower doses of influenza vaccine were able to protect mice from virus growth in the lungs when the vaccines were supplemented with GEM particles. These results show the potential of GEM as adjuvant for influenza vaccines and warrant further development.

bibliography1. CDC estimates of 2009 H1N1 influenza cases,

hospitalizations and deaths in the United States, April 2009-February 13, 2010. Atlanta: Centers for Disease Control and Prevention,2010.(http://flu.gov/individualfamily/about/h1n1/estimates_2009_h1n1.html.)

2. R.B. Couch, J.A. Kasel, W.P. Glezen, T.R. Cate, H.R. Six, L.H. Taber, A.L. Frank, S.B. Greenberg, J.M. Zahradnik, W.A. Keitel, Influenza: its control in persons and populations. J Infect Dis 153(3) (1986) 431-440.

3. R.J. Cox, K.A. Brokstad, P. Ogra, Influenza virus: immunity and vaccination strategies. Comparison of the immune response to inactivated and live, attenuated influenza vaccines. Scand J Immunol 59(1) (2004) 1-15.

4. P.D. Minor, Vaccines against seasonal and pandemic influenza and the implications of changes in substrates for virus production. Clin Infect Dis 50(4) 560-565.

5. A.H. Ellebedy, R.J. Webby, Influenza vaccines. Vaccine 27 Suppl 4 (2009) D65-68.

6. M. Hoelscher, S. Gangappa, W. Zhong, L. Jayashankar, S. Sambhara, Vaccines against epidemic and pandemic influenza. Expert Opin Drug Deliv 5(10) (2008) 1139-1157.

7. S.G. Reed, S. Bertholet, R.N. Coler, M. Friede, New horizons in adjuvants for vaccine development. Trends Immunol 30(1) (2009) 23-32.

8. E.B. Lindblad, Aluminium compounds for use in vaccines. Immunol Cell Biol 82(5) (2004) 497-505.

9. R.K. Gupta, Aluminum compounds as vaccine adjuvants. Adv Drug Deliv Rev 32(3) (1998) 155-172.

10. D. Middleton, S. Rockman, M. Pearse, I. Barr, S. Lowther, J. Klippel, D. Ryan, L. Brown, Evaluation of vaccines for H5N1 influenza virus in ferrets reveals the potential for protective single-shot immunization. J Virol 83(15) (2009) 7770-7778.

11. T. Nolan, P.C. Richmond, N.T. Formica, K. Hoschler, M.V. Skeljo, T. Stoney, J. McVernon, G. Hartel, D.C. Sawlwin, J. Bennet, D. Ryan, R.L. Basser, M.C. Zambon, Safety and immunogenicity of a prototype adjuvanted inactivated split-virus influenza A (H5N1) vaccine in infants and children. Vaccine 26(50) (2008) 6383-6391.

12. Y.F. Lau, L.H. Tang, A.W. McCall, E.E. Ooi, K. Subbarao, An adjuvant for the induction of potent, protective humoral responses to an H5N1 influenza vaccine with antigen-sparing effect in mice. J Virol (2010).

13. B. Baras, K.J. Stittelaar, J.H. Simon, R.J. Thoolen, S.P. Mossman, F.H. Pistoor, G. van Amerongen, M.A. Wettendorff, E. Hanon, A.D. Osterhaus, Cross-protection against lethal H5N1 challenge in ferrets with an adjuvanted pandemic influenza vaccine. PLoS One 3(1) (2008) e1401.

14. C. Caillet, F. Piras, M.C. Bernard, A. de Montfort, F. Boudet, F.R. Vogel, A. Hoffenbach, C. Moste, I. Kusters, AF03-adjuvanted and non-adjuvanted pandemic influenza A (H1N1) 2009 vaccines induce strong antibody responses

Page 16: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

InFLuenzA AntIGen-SPArInG By GeM PArtIcLeS

2

35

in seasonal influenza vaccine-primed and unprimed mice. Vaccine 28(18) (2010) 3076-3079.

15. C.S. Waddington, W.T. Walker, C. Oeser, A. Reiner, T. John, S. Wilkins, M. Casey, P.E. Eccleston, R.J. Allen, I. Okike, S. Ladhani, E. Sheasby, K. Hoschler, N. Andrews, P. Waight, A.C. Collinson, P.T. Heath, A. Finn, S.N. Faust, M.D. Snape, E. Miller, A.J. Pollard, Safety and immunogenicity of AS03B adjuvanted split virion versus non-adjuvanted whole virion H1N1 influenza vaccine in UK children aged 6 months-12 years: open label, randomised, parallel group, multicentre study. BMJ 340 (2010) c2649.

16. W.A. Keitel, J.D. Campbell, J.J. Treanor, E.B. Walter, S.M. Patel, F. He, D.L. Noah, H. Hill, Safety and immunogenicity of an inactivated influenza A/H5N1 vaccine given with or without aluminum hydroxide to healthy adults: results of a phase I-II randomized clinical trial. J Infect Dis 198(9) (2008) 1309-1316.

17. K.G. Nicholson, A.E. Colegate, A. Podda, I. Stephenson, J. Wood, E. Ypma, M.C. Zambon, Safety and antigenicity of non-adjuvanted and MF59-adjuvanted influenza A/Duck/Singapore/97 (H5N3) vaccine: a randomised trial of two potential vaccines against H5N1 influenza. Lancet 357(9272) (2001) 1937-1943.

18. R. Bodewes, J.H. Kreijtz, G. van Amerongen, M.M. Geelhoed-Mieras, R.J. Verburgh, J.G. Heldens, J. Bedwell, J.M. van den Brand, T. Kuiken, C.A. van Baalen, R.A. Fouchier, A.D. Osterhaus, G.F. Rimmelzwaan, A single immunization with CoVaccine HTTM-adjuvanted H5N1 influenza vaccine induces protective cellular and humoral immune responses in ferrets. J Virol (2010).

19. R.C. Brady, J.J. Treanor, R.L. Atmar, W.A. Keitel, R. Edelman, W.H. Chen, P. Winokur, R. Belshe, I.L. Graham, D.L. Noah, K. Guo, H. Hill, Safety and immunogenicity of a subvirion inactivated influenza A/H5N1 vaccine with or without aluminum hydroxide among healthy elderly adults. Vaccine 27(37) (2009) 5091-5095.

20. W.A. Keitel, R.L. Atmar, Vaccines for pandemic influenza: summary of recent clinical trials. Curr Top Microbiol Immunol 333 (2009) 431-451.

21. M.L. van Roosmalen, R. Kanninga, M. El Khattabi, J. Neef, S. Audouy, T. Bosma, A. Kuipers, E. Post, A. Steen, J. Kok, G. Buist, O.P. Kuipers, G. Robillard, K. Leenhouts, Mucosal vaccine delivery of antigens tightly bound to an adjuvant particle made from food-grade bacteria. Methods 38(2) (2006) 144-149.

22. T. Bosma, R. Kanninga, J. Neef, S.A. Audouy, M.L. van Roosmalen, A. Steen, G. Buist, J. Kok, O.P. Kuipers, G. Robillard, K. Leenhouts, Novel surface display system for proteins on non-genetically modified gram-positive bacteria. Appl Environ Microbiol 72(1) (2006) 880-889.

23. S.A. Audouy, M.L. van Roosmalen, J. Neef, R. Kanninga, E. Post, M. van Deemter, H. Metselaar, S. van Selm, G.T. Robillard, K.J. Leenhouts, P.W. Hermans, Lactococcus lactis GEM particles displaying pneumococcal antigens induce local and systemic immune responses following intranasal immunization. Vaccine 24(26) (2006) 5434-5441.

24. S.A. Audouy, S. van Selm, M.L. van Roosmalen, E. Post, R. Kanninga, J. Neef, S. Estevao, E.E. Nieuwenhuis, P.V. Adrian, K. Leenhouts, P.W. Hermans, Development of lactococcal GEM-based pneumococcal vaccines. Vaccine 25(13) (2007) 2497-2506.

25. K. Ramirez, Y. Ditamo, L. Rodriguez, W.L. Picking, M.L. van Roosmalen, K. Leenhouts, M.F. Pasetti, Neonatal mucosal immunization with a non-living, non-genetically modified Lactococcus lactis vaccine carrier induces systemic and local Th1-type immunity and protects against lethal bacterial infection. Mucosal Immunol 3(2) (2010) 159-171.

26. R. Ramasamy, S. Yasawardena, A. Zomer, G. Venema, J. Kok, K. Leenhouts, Immunogenicity of a malaria parasite antigen displayed by Lactococcus lactis in oral immunisations. Vaccine 24(18) (2006) 3900-3908.

27. V. Saluja, J.P. Amorij, M.L. van Roosmalen, K. Leenhouts, A. Huckriede, W.L. Hinrichs, H.W. Frijlink, Intranasal delivery of influenza subunit vaccine formulated with GEM particles as an adjuvant. AAPS J 12(2) (2010) 109-116.

28. J.P. Amorij, J. Meulenaar, W.L. Hinrichs, T. Stegmann, A. Huckriede, F. Coenen, H.W. Frijlink, Rational design of an influenza subunit vaccine powder with sugar glass technology: preventing conformational changes of haemagglutinin during freezing and freeze-drying. Vaccine 25(35) (2007) 6447-6457.

29. L. Bungener, F. Geeraedts, W. Ter Veer, J. Medema, J. Wilschut, A. Huckriede, Alum boosts TH2-type antibody responses to whole-inactivated virus influenza vaccine in mice but does not confer superior protection. Vaccine 26(19) (2008) 2350-2359.

30. The European Agency for the Evaluation of Medicinal Products (EMEA) Committee for Proprietary Medicinal Products (CPMP). Note for guidance on harmonization of requirements

Page 17: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes

cHAPter 2

36

for influenza vaccines. CPMP/BWP/214/96, London, March 12, 1997.

31. J.P. Amorij, V. Saluja, A.H. Petersen, W.L. Hinrichs, A. Huckriede, H.W. Frijlink, Pulmonary delivery of an inulin-stabilized influenza subunit vaccine prepared by spray-freeze drying induces systemic, mucosal humoral as well as cell-mediated immune responses in BALB/c mice. Vaccine 25(52) (2007) 8707-8717.

32. N. Hehme, T. Colegate, B. Palache, L. Hessel, Influenza vaccine supply: building long-term sustainability. Vaccine 26 Suppl 4 (2008) D23-26.

33. R.L. Atmar, W.A. Keitel, Adjuvants for pandemic influenza vaccines. Curr Top Microbiol Immunol 333 (2009) 323-344.

34. A.O. Hovden, R.J. Cox, A. Madhun, L.R. Haaheim, Two doses of parenterally administered split influenza virus vaccine elicited high serum IgG concentrations which effectively limited viral shedding upon challenge in mice. Scand J Immunol 62(4) (2005) 342-352.

35. V.C. Huber, R.M. McKeon, M.N. Brackin, L.A. Miller, R. Keating, S.A. Brown, N. Makarova, D.R. Perez, G.H. Macdonald, J.A. McCullers, Distinct contributions of vaccine-induced immunoglobulin G1 (IgG1) and IgG2a antibodies to protective immunity against influenza. Clin Vaccine Immunol 13(9) (2006) 981-990.

36. S. Tamura, T. Kurata, Defense mechanisms against influenza virus infection in the

respiratory tract mucosa. Jpn J Infect Dis 57(6) (2004) 236-247.

37. A. Yoshimura, E. Lien, R.R. Ingalls, E. Tuomanen, R. Dziarski, D. Golenbock, Cutting edge: recognition of Gram-positive bacterial cell wall components by the innate immune system occurs via Toll-like receptor 2. J Immunol 163(1) (1999) 1-5.

38. F. Brull, R.P. Mensink, K. van den Hurk, A. Duijvestijn, J. Plat, TLR2 activation is essential to induce a Th1 shift in human peripheral blood mononuclear cells by plant stanols and plant sterols. J Biol Chem 285(5) 2951-2958.

39. B. Guy, The perfect mix: recent progress in adjuvant research. Nat Rev Microbiol 5(7) (2007) 505-517.

40. I. Babai, Y. Barenholz, Z. Zakay-Rones, E. Greenbaum, S. Samira, I. Hayon, M. Rochman, E. Kedar, A novel liposomal influenza vaccine (INFLUSOME-VAC) containing hemagglutinin-neuraminidase and IL-2 or GM-CSF induces protective anti-neuraminidase antibodies cross-reacting with a wide spectrum of influenza A viral strains. Vaccine 20(3-4) (2001) 505-515.

41. S. Chang, J. Warner, L. Liang, J. Fairman, A novel vaccine adjuvant for recombinant flu antigens. Biologicals 37(3) (2009) 141-147.

42. J. Hartikka, V. Bozoukova, C.K. Yang, M. Ye, D. Rusalov, M. Shlapobersky, A. Vilalta, Q. Wei, A. Rolland, L.R. Smith, Vaxfectin, a cationic lipid-based adjuvant for protein-based influenza vaccines. Vaccine 27(46) (2009) 6399-6403.

Page 18: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes
Page 19: University of Groningen Novel perspectives for …and H1N1 pandemic vaccines, several adjuvants, like oil-in-water based adjuvants, aluminium salts (alum), and immunostimulating complexes