34
1 1 2 3 4 5 6 Th1 immune response to Plasmodium falciparum circumsporozoite protein is 7 boosted by adenovirus vectors 35 and 26 with homologous insert 8 9 Katarina Radošević 1* , Ariane Rodriguez 1^ , Angelique A.C. Lemckert 1^† , Marjolein van 10 der Meer 1 , Gert Gillissen 1 , Carolien Warnar 1 , Rie von Eyben 1 , Maria Grazia Pau 1 and 11 Jaap Goudsmit 1,2 12 13 1 Crucell Holland B. V., Leiden, The Netherlands and 2 Center of Poverty-related 14 Communicable Diseases, Academic Medical Center, Amsterdam, The Netherlands 15 16 Running Title: A 3-component heterologous prime-boost vaccination for malaria 17 18 ^ Both authors contributed equally 19 † Present address: Batavia Bioservices B.V., Leiden, The Netherlands 20 * Corresponding Author. Mailing address: Crucell Holland BV, Archimedesweg 4-6, 21 2333 CN Leiden, The Netherlands. Phone: 31 (0)71 5199286. Fax: 31 (0)71 5199800. 22 E-mail: [email protected]. 23 Copyright © 2010, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved. Clin. Vaccine Immunol. doi:10.1128/CVI.00311-10 CVI Accepts, published online ahead of print on 8 September 2010 on February 21, 2021 by guest http://cvi.asm.org/ Downloaded from on February 21, 2021 by guest http://cvi.asm.org/ Downloaded from on February 21, 2021 by guest http://cvi.asm.org/ Downloaded from

Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

1

1

2

3

4

5

6

Th1 immune response to Plasmodium falciparum circumsporozoite protein is 7

boosted by adenovirus vectors 35 and 26 with homologous insert 8

9

Katarina Radošević1*

, Ariane Rodriguez1^

, Angelique A.C. Lemckert

1^†, Marjolein van 10

der Meer1, Gert Gillissen

1, Carolien Warnar

1, Rie von Eyben

1, Maria Grazia Pau

1 and 11

Jaap Goudsmit1,2

12

13

1Crucell Holland B. V., Leiden, The Netherlands and

2Center of Poverty-related 14

Communicable Diseases, Academic Medical Center, Amsterdam, The Netherlands

15

16

Running Title: A 3-component heterologous prime-boost vaccination for malaria 17

18

^ Both authors contributed equally 19

† Present address: Batavia Bioservices B.V., Leiden, The Netherlands 20

* Corresponding Author. Mailing address: Crucell Holland BV, Archimedesweg 4-6, 21

2333 CN Leiden, The Netherlands. Phone: 31 (0)71 5199286. Fax: 31 (0)71 5199800. 22

E-mail: [email protected]. 23

Copyright © 2010, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved.Clin. Vaccine Immunol. doi:10.1128/CVI.00311-10 CVI Accepts, published online ahead of print on 8 September 2010

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 2: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

2

ABSTRACT 1

The most advanced malaria vaccine, RTS,S, is comprised of an adjuvanted portion of the 2

Plasmodium falciparum circumsporozoite (CS) protein, fused to and admixed with the 3

hepatitis B surface antigen. This vaccine confers short term protection against malaria 4

infection with an efficacy of about 50% and induces particularly B-cell and CD4+ T-cell 5

responses. In the present study the hypothesis is tested that Th1 immune response to CS 6

protein, in particular the CD8+ T-cell response, which is needed for strong and lasting 7

malaria immunity, is boosted to sustainable levels using the Ad35.CS/Ad26.CS 8

combination. In this study we evaluated immune responses induced with vaccination 9

regimens based on an adjuvanted yeast-produced complete CS protein followed by two 10

recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 11

Ad35.CS (subgroup B) and Ad26.CS (subgroup D). Our results show that (i) the yeast-12

produced adjuvanted full-length CS protein is highly potent in inducing high CS-specific 13

humoral responses in mice, but poor T-cell response (ii) the Ad35.CS vector boosts the 14

IFNγ+ CD8+ T-cell response induced by the CS protein immunization and shifts the 15

immune response towards the Th1 type and (iii) a three-component heterologous 16

vaccination comprised of a CS protein prime, followed by boosts with Ad35.CS and 17

Ad26.CS, elicits an even more robust and sustainable IFNγ+ CD8+ T-cell response as 18

compared to one or two component regimens. The Ad35.CS/Ad26.CS combination 19

boosted particularly the IFNγ+ and TNFα+ T cells, confirming the shift of the immune 20

response from the Th2 to Th1 type. 21

These results support the notion of first immunizations of infants with an adjuvanted CS 22

protein vaccine, followed by a booster Ad35.CS/Ad26.CS vaccine at later age, to induce 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 3: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

3

lasting protection against malaria for which the Th1 response and immune memory is 1

required. 2

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 4: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

4

INTRODUCTION 1

Almost forty years after the feasibility of vaccination against malaria was first 2

demonstrated by means of irradiated sporozoites (9), a vaccine modality that efficiently 3

induces long-lived protective immunity remains elusive. The most advanced CS-based 4

malaria vaccine candidate to date is RTS,S, a vaccine based on a fragment of 5

Plasmodium falciparum circumsporozoite (CS) protein, fused to and admixed with 6

hepatitis B surface protein. In adults, RTS,S with adjuvant AS02 has consistently 7

conferred 40% protection against malaria infection upon sporozoite challenge (53). Even 8

though RTS,S/AS02 induces high level CS-specific antibody responses, the induced T-9

cell responses are weak (21). As Th1 response, and in particular IFNγ and CD8+ T cells, 10

are associated with protection, novel adjuvant systems were developed with the aim to 11

improve the induced T-cell response while maintaining potent levels of CS-specific 12

antibody responses. One of these novel adjuvant systems, AS01, demonstrated its 13

suitability in mice as it improved CS-specific CD4+ T-cell responses and led to induction 14

of CD8+ T cells (31). Non-human primate studies also demonstrated that RTS,S with 15

AS01 adjuvant induces strong CS-specific antibody responses as well as higher mean 16

frequencies of IFNγ- and TNFα-producing CD4+ T cells compared to the RTS,S with 17

AS02 adjuvant. However, the induction of CD8+ T cells was not confirmed in this non-18

human primate study (31). In humans, RTS,S/AS01 has shown to induce high titers of 19

CS-specific antibodies, higher numbers of Th1 CD4+ T cells compared to RTS,S/AS02, 20

but no CS-specific CD8+ T cells (22). Though, RTS,S/AS01 was able to afford 50% 21

protection against malaria infection in adults upon sporozoite challenge (22) and 53% 22

efficacy against disease in children between the ages of 5 to 17 months (5). These results, 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 5: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

5

albeit far of being optimal, supported the progress of RTS,S/AS01 to phase III clinical 1

trial testing in early 2009, enrolling children at the age of 6 weeks to 17 months at 2

multiple sites in Sub-Saharan Africa. It is anticipated that RTS,S/AS01 will be the first 3

licensed malaria vaccine, provided its efficacy is confirmed in the phase III trial. 4

Albeit our understanding about the correlate(s) of protection for malaria is limited, there 5

is ample evidence that circumsporozoite (CS) protein-specific antibodies, CD8+ T cells 6

and Th1 cytokines, and in particular IFNγ, play a central role in controlling the pre-7

erythrocytic and early liver stages of malaria (19, 20, 34, 46, 56). Adenoviral vectors are 8

particularly suited for induction of IFNγ-producing CD8+ T cells required to combat 9

malaria infection (32, 42), due to intracellular expression of a transgene inserted in the 10

vector genome and efficient routing of expressed protein towards the class I presentation 11

pathway. Recently, we have demonstrated the advantage of utilizing two recombinant 12

adenoviral vectors derived from distinct serotypes, Ad35.CS and Ad5.CS, in a 13

heterologous prime-boost regimen in mice and non-human primates (45). This 14

heterologous prime-boost regimen elicited high CS-specific IFNγ+ T-cell response as 15

well CS-specific Th1 type antibodies able to bind malaria parasites. Though the Ad5-16

based vectors are very potent vaccines, the high prevalence of pre-existing immunity 17

towards Ad5 in the human population hampers their immunogenicity and clinical utility 18

(8, 37). Low seroprevalence of Ad5 neutralizing antibodies in infants of 6 months to 1.5 19

years of age offers an opportunity to administer Ad5-based vaccines to this population 20

without antibodies interfering and neutralizing the vaccine efficacy (41), however, 21

acceptance of this approach by regulatory agencies remains elusive. Novel vaccine 22

vectors based on rare low seroprevalent Ad serotypes have an advantage of not being 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 6: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

6

hampered by anti-Ad5 immunity while inducing a strong immune response (1, 4, 28, 32, 1

40). 2

Within the current study we evaluate whether vaccination with Ad35.CS and Ad26.CS 3

can enhance the CS-specific immune response induced by a yeast-produced full-length 4

CS protein vaccine, and in particular, whether the combined vaccination sustainably 5

potentiate the Th1 responses necessary for protection against malaria. The Ad35.CS 6

vaccine candidate is currently being evaluated in a Phase 1 clinical study, in partnership 7

with the National Institute of Allergy and Infectious Diseases, and so far has shown safe. 8

Ad35-based vaccine candidates against other infectious diseases, i.e. tuberculosis and 9

HIV, have also been clinically evaluated, and demonstrated safe and immunogenic. 10

Recently, an Ad26-vectored vaccine against HIV was also been clinically assessed in a 11

Phase I study, which showed that a 3-dose regimen of this HIV candidate vaccine is safe 12

and immunogenic. Based upon encouraging results, the clinical testing of the 13

combination of Ad35- and Ad26-based vaccines against malaria and HIV is in 14

preparation. 15

16

17

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 7: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

7

MATERIALS AND METHODS 1

Vector and protein construction, production and purification. 2

E1/E3-deleted, replication-incompetent Ad26 and Ad35 vectors expressing the same P. 3

falciparum CS gene were generated in E1-complementing PER.C6® cells and purified 4

using CsCl gradients as previously described (1, 16). Viral particles (vp) were quantified 5

by high-performance liquid chromatography (HPLC). The P. falciparum CS gene is a 6

synthetic, mammalian-codon optimized insert encoding a CS protein based on the EMBL 7

protein sequence CAH04007, and truncated for the last 14 amino acids at the C-terminus. 8

The N-terminus of this CS protein is a consensus assembled by alignment of various 9

sequences present in the GenBank, while the repeat region and the C-terminus are based 10

on the sequence of the 3D7 P. falciparum clone. The CS repeat region consisted of 27 11

NANP repeats, a cluster of 3 NVDP and one separate NVDP. CS protein of identical 12

sequence as in the adenovectors has been produced in Hansenula polymorpha RB11 13

clone by ARTES Biotechnology GmbH (Germany). A C-terminal His-tag sequence was 14

introduced into the construct to facilitate Ni-column purification of the CS protein from 15

the culture supernatant. 16

17

Characterization of the yeast-produced CS protein 18

The yeast-produced CS protein was analyzed by CS-specific Western blot and 19

InstantBlue staining, which demonstrated the identity and purity of the CS protein (more 20

than 80% pure) (Figure 1a). For the Western blot, rabbit polyclonal antibody against P. 21

falciparum CS (MRA-24, MR4/ATCC) was used in combination with goat-anti-rabbit 22

IgG conjugated to horseradish peroxide (HRP, Biorad) and enhanced chemiluminescence 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 8: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

8

(ECL+, GE healthcare) to detect CS expression. The InstantBlue staining was performed 1

according to protocol provided by the manufacturer (Expedeon). 2

A dose of the yeast-produced CS protein for prime-boost immunogenicity studies was 3

selected using immunization of BALB/c mice (n = 5 per group) with increasing dosages 4

of CS protein (5 µg, 10 µg or 25 µg), formulated with the Montanide ISA 720 (Seppic, 5

France) at a 30:70 volume-based ratio, at 3-weeks intervals. The CS-specific humoral 6

response were assessed using ELISA, which demonstrated that the yeast-produced CS 7

protein induces maximal CS-specific antibody responses already at the lowest tested dose 8

(5 µg) and after two immunizations (Figure 1b). The induced IgG response consisted 9

predominantly of IgG1 antibodies, indicating the Th2 type response (Figure 1C). 10

Analysis of the CS-specific cellular immunity using ELISPOT revealed poor induction of 11

IFNγ+ T-cells for all doses (data not shown). 12

13

Animals and vaccinations regimens. 14

Our study seek to evaluate whether vaccination with Ad35.CS and Ad26.CS can enhance 15

the CS-specific immune response induced by a protein-based vaccine (eventually RTS,S), 16

as potential vaccination strategy for malaria. Since the RTS,S vaccine was not available, 17

for these studies we have used a yeast-produced full-length CS protein vaccine. 18

All animal experiments were approved in advance by an independent Ethical Review 19

Board. Six to eight week-old female BALB/c mice were purchased from Harlan (Zeist, 20

The Netherlands) and kept at the institutional animal facility under specific-pathogen-free 21

conditions during the experiment. 22

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 9: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

9

To evaluate the immunogenicity of the heterologous CS protein/Ad prime-boost 1

regimens, BALB/c mice (n = 8 per group) were primed at week 0 with 5 µg adjuvanted 2

CS protein and boosted at week 4 with 109 vp Ad35.CS. The optimal immunization doses 3

of Ad.CS for immunization were selected from earlier dose response experiments (data 4

not shown). Another group of mice (n = 8) received a homologous prime-boost regimen 5

of 5 µg adjuvanted CS protein. As negative control group, BALB/c mice (n = 6) were 6

injected at week 0 with adjuvant montanide ISA720 and at week 4 with 109 vp 7

Ad35.Empty (adenovector without insert; indicated as sham immunization group). 8

To evaluate the 3-component heterologous prime-boost, BALB/C mice (n = 8) were 9

immunized at week 0 with 5 µg adjuvanted CS protein, boosted at week 4 with 109 vp 10

Ad35.CS and at week 8 with 1010

vp Ad26.CS. Comparator groups of BALB/C mice (n = 11

8 per group) started immunization at week 4 with 5 µg adjuvanted CS protein and were 12

boosted after 4 weeks (at week 8) with either 109 vp Ad35.CS or 5 µg adjuvanted CS 13

protein. As a negative control group, mice (n = 3) received the adjuvant montanide 14

ISA720 at week 0, 109 vp rAd35.Empty at week 4 and 10

10 vp rAd26.Empty at week 8. 15

16

CS-specific T-cell assays. 17

CS-specific cellular immune responses in vaccinated mice were assessed using 18

interferon-γ (IFNγ) ELISPOT assay, intracellular cytokine staining in combination with 19

surface staining of CD4 and CD8 markers (ICS), as described previously elsewhere (4, 20

44), and cytometric bead array (CBA) assay. 21

For the stimulation of splenocytes in the ELISPOT and ICS, a peptide pool consisting of 22

11-amino acids overlapping 15-mer peptides spanning the whole sequence of the P. 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 10: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

10

falciparum CS protein was used. The pool contained a highly immunodominant CD8+ T-1

cell epitope (NYDNAGTNL; H-2Kd), which is responsible for the main part of measured 2

responses in the ELISPOT and the CD8+ responses in the ICS. This was confirmed with 3

an experiment wherein the splenocytes were stimulated with the 9-mer peptides, which 4

generated virtually identical responses as the peptide pool (data not shown). For the 5

ELISPOT, ninety-six-well multiscreen plates (Millipore, Bedford, MA) were coated 6

overnight with 100 µl/well of 10 µg/ml anti-mouse IFN-γ (BD Pharmingen, San Diego, 7

CA) in endotoxin-free Dulbecco’s PBS (D-PBS). The plates were then washed three 8

times with D-PBS containing 0.05% Tween-20 (D-PBS/Tween), blocked for 2 h with D-9

PBS containing 5% FBS at 37oC, and rinsed with RPMI 1640 containing 10% FBS. 10

Splenocytes from individual mice were stimulated with the CS peptide pool for 18 h at 11

37oC. Following incubation the plates were washed six times with D-PBS/Tween and 12

once with distilled water. The plates were then incubated with 2 µg/ml biotinylated anti-13

mouse IFN-γ (BD Pharmingen, San Diego, CA) for 2 h at room temperature, washed six 14

times with D-PBS/Tween, and incubated for 2 h with a 1:500 dilution of streptavidin-15

alkaline phosphatase (Southern Biotechnology Associates, Birmingham, AL). Following 16

six washes with D-PBS/Tween and one with PBS, the plates were developed with nitro 17

blue tetrazolium/5-bromo-4-chloro-3-indolyl-phosphate chromogen (Pierce, Rockford, 18

IL), reaction was stopped with tap water, air dried, and read using an ELISPOT reader 19

(Aelvis GmbH). Spot-forming units (SFU) per 106 cells were calculated. In the case of 20

the ICS, splenocytes from individual animals were stimulated with the CS peptide pool or 21

cultured with medium alone. All cultures

contained monensin (GolgiStop; BD 22

Biosciences) as well as 1 µg/ml anti-CD49d (BD Biosciences). The cultured cells

were 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 11: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

11

stained with monoclonal antibodies specific for cell surface molecules (CD4 and CD8). 1

After fixing with Cytofix/Cytoperm solution (BD Biosciences), cells were permeabilized 2

and stained

with antibodies specific for mouse IFNγ. Approximately 200,000 to 3

1,000,000 events were collected per sample. The background level of cytokine staining

4

was typically lower than 0.01% for CD4+ T cells and lower than 0.05% for CD8+ T cells. 5

The T-helper response induced by the different vaccination regimens was evaluated using 6

Cytometric bead array (CBA) assay. Splenocytes from individual mice were stimulated 7

with 5 µg/ml yeast-produced CS protein. After 48 h of incubation at 37°C, supernatants 8

were harvested and analyzed for the presence of the Th1 (IFNγ, TNFα, IL-2), Th2 (IL-4, 9

IL-6, IL-10) and Th17 (IL-17) cytokines using the Mouse Th1/Th2/Th17 Cytokine Kit 10

according to protocol provided by the manufacturer (BD Biosciences). 11

12

CS-specific antibody assays. 13

CS-specific antibody responses were assessed by enzyme-linked immunosorbent assay 14

(ELISA) as previously described (33). Ninety-six-well microtiter plates (Maxisorp; 15

Nunc) were coated overnight at 4°C with 2 µg/ml of CS-specific (NANP)6C peptide in 16

0.05 M Carbonate buffer (pH 9.6). Plates were washed three times and blocked with PBS 17

containing 1% BSA and 0.05% Tween-20 for 1 h at 37°C. After the plates were washed 18

three times, 1:100-diluted individual serum samples were added to the wells and serially 19

twofold diluted in PBS containing 0.2% BSA and 0.05% Tween-20. Plates were 20

incubated for 2 h at 37°C. Plates were washed three times and incubated with biotin-21

labeled anti-mouse or anti-rabbit immunoglobulin G (IgG) (Dako, Denmark), followed 22

by horseradish peroxidase-conjugated streptavidin (Pharmingen San Diego, CA) for 30 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 12: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

12

min each at 37°C. For detection of the IgG subclasses, samples were incubated with 1

horseradish peroxidase-labeled anti-mouse IgG1 or IgG2a antibodies (Southern Biotech, 2

Birmingham, AL). Finally, the plates were washed and 100 µl of o-phenylenediamine 3

dihydrochloride (OPD) substrate (Pierce, Rockford, IL) was added to each well. After 10 4

min, the reaction was stopped by adding 100 µl/well of 1 M H2SO4. The optical density 5

was measured at 492 nm using a Bio-Tek reader (Bio-Tek Instruments, Winooski, VT). 6

The ELISA units were calculated relative to the OD curve of the serially diluted standard 7

serum, with one ELISA unit corresponding to the serum dilution at 50% of the maximum 8

of the standard curve. The IgG2a/IgG1 ratio was determined using titer values of IgG1 9

and IgG2a antibodies, which are expressed as a reverse of serum dilution. 10

11

Statistical analyses. 12

Comparisons of geometric mean immune responses were performed by student t-test after 13

logarithmic transformation to account for two test groups. Comparisons of geometric 14

mean immune responses were performed by analyses of variance (ANOVA) with Tukey 15

adjustments after logarithmic transformation to account for multiple comparisons. In all 16

cases, p-values lower than 0.05 were considered significant. 17

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 13: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

13

RESULTS 1

2

Immunogenicity of CS protein prime followed by Ad35.CS boost. 3

CS-specific humoral response induced in BALB/c mice with an CS protein prime and 4

Ad35.CS boost at two weeks post immunization was assessed by ELISA assay (Figure 5

2A and B) while the cellular immune responses were measured using IFNγ ELISPOT 6

(Figure 2C) and ICS (Figure 2D). The homologous prime-boost regimen with the CS 7

protein elicited a very potent CS-specific IgG response. The levels of the antibody 8

response elicited by the heterologous CS protein/Ad35.CS regimen were comparable to 9

that seen for the homologous CS protein prime-boost regimen (P > 0.05 comparing CS-10

specific IgG levels with ANOVA). Beside the total CS-specific IgG levels, we 11

determined the IgG2a/IgG1 ratio to obtain indications of the type of T-helper responses 12

induced by the different prime-boost regimens (Figure 2B). The homologous CS protein 13

prime-boost regimen elicited primarily IgG1 antibody responses indicating a more Th2 14

type immune response while replacing the protein boost with Ad35.CS boost resulted in a 15

more pronounced induction of IgG2a antibodies indicating shift towards a Th1 type 16

response (P < 0.05 comparing IgG2a/IgG1 ratios with ANOVA). 17

Evaluation of the CS-specific T-cell responses using ELISPOT (Figure 2C) and ICS 18

(Figure 2D) assays showed that the homologous CS protein regimen evoked a poor but 19

measurable CS-specific T-cell response. The inclusion of Ad35.CS as a boost to the CS 20

protein prime resulted in significantly increased levels of CS-specific IFNγ-producing 21

CD8+ T-cells (P < 0.05 comparing CS-specific CD8+ T-cell levels with ANOVA). This 22

correlated to the more Th1 type response for the CS protein/Ad35.CS regimens as 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 14: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

14

determined by CS-specific IgG2a/IgG1 ratio. It should be noted that IFNγ+ CD4+ 1

response might have been underestimated using the stimulation with the 15-mer peptides, 2

as we observed in another study (39). Stimulation of splenocytes with the CS-protein in 3

the current study did show higher CD4+ responses, however, the background in the assay 4

was unacceptably high (data not shown). 5

6

Immunogenicity of a three-component heterologous prime-boost regimen. 7

The combination of the yeast-produced CS protein with the Ad35.CS in a heterologous 8

prime-boost regimen results in the induction of high level of IFNγ+ CD8+ T cells, 9

maintained high level of CS-specific IgG response and the antibody response was shifted 10

towards the Th1 type. We next investigated whether a prime-boost regimen comprised of 11

the three components, CS protein, Ad35.CS and Ad26.CS, might result in an even more 12

robust and sustained Th1 immune response. Our earlier experiments demonstrated that 13

the Ad35.CS/Ad26.CS combination induces significantly higher immune responses than 14

the Ad35.CS/Ad35.CS combination (data not shown) and, therefore, the homologous 15

adenovector combination Ad35.CS/Ad35.CS was not included as a booster vaccine in the 16

current study. A group of mice received a prime with adjuvanted CS protein and a boost 17

with Ad35.CS followed by a second boost with Ad26.CS (three-component heterologous 18

prime-boost). A comparator group of mice received a prime with adjuvanted CS protein 19

followed by an Ad35.CS boost. At two weeks post the final boost immunization, mice 20

receiving the three-component heterologous prime-boost regimen showed a significantly 21

higher levels of CS-specific IFNγ-producing CD8+ T-cells compared to the mice 22

receiving the CS protein prime and Ad35.CS boost regimen (Figure 3A; P > 0.05 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 15: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

15

comparing CS-specific IFNγ-producing CD8+ T-cell levels with ANOVA). At eight 1

weeks post the final boost immunization, the IFNγ+ CD8+ T-cell response induced by the 2

three-component prime-boost regimen was still significantly higher compared to the CS 3

protein/Ad35.CS regimen (Figure 3B; P > 0.05 comparing CS-specific IFNγ-producing 4

CD8+ T-cell levels with ANOVA). Importantly, at both time points the levels of CS-5

specific IgG responses induced by the three-component prime-boost regimen were 6

comparable to that seen for the CS protein/Ad35.CS regimen (Figure 3B and D; P > 0.05 7

comparing CS-specific IgG levels with ANOVA). The IgG2a/IgG1 ratio of CS-specific 8

antibodies induced with the CS protein/Ad35.CS/Ad26.CS vaccine regimen was 9

comparable to the ratio induced with the CS protein/Ad35.CS immunization (data not 10

shown). 11

12

Cytokine profile induced by the different vaccination regimens 13

The total number of CS-specific CD4+ T cells expressing two or more immune markers, 14

being Th1 cytokines IFNγ, TNFα, IL2, and activation marker CD40L, induced upon 15

immunization with RTS,S, has been associated with protection to malaria infection in the 16

human challenge model (22). We investigated cytokine profile breadth induced in CS-17

specific T cells with three component malaria vaccine, CS protein/Ad35.CS/Ad26.CS, 18

and compared it to the cytokine profiles induced with CS-protein/CS protein or CS 19

protein/Ad35.CS regimen. Two weeks after the final boost immunization, expression 20

levels of the Th1 (IFNγ, TNFα, IL-2), Th2 (IL-4, IL-6, IL-10) and Th17 (IL-17) 21

cytokines were determined using the cytometric bead array (CBA) assay upon 48 hrs in 22

vitro stimulation of splenocytes with the CS protein. The CBA assay with protein 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 16: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

16

stimulation provides a blueprint of the type of T helper cells that have been induced with 1

the vaccination regiment. All vaccination regimens, except for the sham, induced the 2

tested cytokines, with an exception of IL-4 which was not detected (data not shown) 3

(Figure 4). The CS protein/Ad35.CS/Ad26.CS regimen induced significantly higher 4

levels of IFNγ and TNFα compared to either the CS protein or the CS protein/Ad35.CS 5

regimen (Figure 4; P < 0.05 comparing cytokine levels with ANOVA). The levels of 6

other cytokines (IL-2, IL-6, IL-10 and IL17) were comparable for all immunization 7

regimens (Figure 4; P > 0.05 comparing cytokine levels with ANOVA). 8

Summarizing, these data confirm that a prime-boost regimen comprising of the three 9

components, CS protein, Ad35.CS and Ad26.CS, results in a robust and broad Th1 type 10

immune response. 11

12

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 17: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

17

DISCUSSION 1

Immunizations with a CS protein vaccine elicit potent antibody responses, but poor 2

cellular responses. In this study we demonstrated that vaccination with the CS protein 3

followed by Ad35.CS vector in a heterologous prime-boost regimen results in 4

enhancement of IFNγ+ CD8+ T-cell responses. The boosting with Ad35.CS did not 5

hamper the level of CS-specific humoral response induced with the protein vaccination, 6

but shifted the Ig isotypes towards a Th1 type of response. In addition, we established 7

that a heterologous prime-boost regimen comprising a CS protein prime followed by 8

boosts with Ad35.CS and Ad26.CS elicits strong CS-specific Th1 type responses, with a 9

durable enhancement of the IFNγ+ CD8+ T-cells and potent antibody responses. 10

The ongoing Phase III trial of RTS,S, a CS-protein based vaccine adjuvanted with a 11

AS01 adjuvnat, represents a breakthrough for malaria vaccine development. The vaccine 12

induces primarily antibody responses and has been shown to partially protect young 13

children and infants in malaria-endemic areas, reducing the risk of clinical episodes of 14

malaria by 53 percent over an eight-month follow-up period (5). Whilst RTS,S gives a 15

better chance of surviving to the most vulnerable part of the population, it is clear that 16

more must be done in order to develop vaccines that will provide greater and more 17

sustainable levels of protection to fully eradicate malaria. 18

IFN-γ+ CD8+ T cells have been associated with protection against liver stage malaria 19

parasites as they inactivate and eliminate intracellular parasites through IFN-γ induced 20

production of nitric oxide and through cell-mediated cytotoxicity (6, 12, 43, 47). 21

Therefore, it is widely accepted that persistent protective immunity against malaria likely 22

requires both high levels of Th1 type immune responses targeting the pre-erythrocytic 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 18: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

18

stage of the malaria parasites. Such a complex immunity is not easily achieved by single 1

vaccine modalities as demonstrated by the low number of malaria vaccine regimens in 2

advanced clinical trials (38). 3

Adenoviral vectors are known to induce high levels of antigen-specific IFN-γ+ CD8+ T 4

cells (52). The combination of adenovectors with other vaccine types has proven highly 5

efficient in eliciting strong and sustainable T-cell immunity as well as humoral responses 6

(7, 14, 26, 48-50). Indeed within the current study we show that the priming with an 7

adjuvanted yeast-produced CS protein followed by the Ad35.CS boost results in the 8

induction of higher CS-specific IFN-γ+ CD8+ T-cell responses compared to exclusively 9

protein-based vaccine regimen. Importantly, while the overall CS-specific IgG levels 10

were not affected compared to the responses induced with an entirely CS protein 11

vaccination regimen, the CS protein/Ad35.CS regimen elicited a more Th1 type response. 12

These results corroborated our earlier findings in which prime-boost regimens comprised 13

of Ad35 vaccine vectors expressing CS or LSA-1, and RTS,S or a LSA-1 protein vaccine 14

resulted in potent Th1 type T-cell responses and high level humoral responses (44, 51). 15

Previously we reported on the heterologous prime-boost regimen utilizing the Ad35.CS 16

and Ad5.CS vaccine vectors that elicited high levels of CS-specific IFN-γ producing T 17

cells in both mice and non-human primates (45). These results demonstrated the potential 18

of adenovector-based heterologous prime-boost regimens to induce the type of immunity 19

required to combat malaria. Because of the high Ad5 seroprevalence in the human 20

population, considerable effort has been directed towards the development of novel low 21

seroprevalent adenoviral vaccine vectors that are able to circumvent anti-Ad5 immunity 22

and are highly immunogenic (1, 4, 13, 17, 25, 35, 55). The Ad26-based vaccine vector 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 19: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

19

has been shown to be a particular interesting vector considering the ability to induce 1

immune responses in mice (1), non-human primates (27, 28) and humans (3), and 2

particularly suited as a boost to other adenovector-based vaccines that utilize different 3

adenovirus serotypes. Given the wide diversity of adenoviruses in nature many different 4

serotypes are potentially available. In our study, the inclusion of the Ad26.CS boost to the 5

CS protein/Ad35.CS prime-boost regimen elicited an overall higher and more sustainable 6

CS-specific IFNγ+ CD8+ immune response as compared to the homologous or the 2-7

component heterologous prime-boost regimens. 8

The recent association of Th1 cytokine expressing CD4+ T cells, induced with RTS,S 9

vaccine, with protection against malaria infection in the human challenge model has 10

reinforced the view that induction of a broad immune response of Th1 type is required for 11

development of efficient malaria vaccines (22). Induction of balanced pro-inflammatory 12

and regulatory immune responses is also a key factor determining the outcome of malaria 13

infection. Failure to develop an effective pro-inflammatory response might result in 14

unrestricted parasite replication, whereas failure to control this response can lead to the 15

development of severe immunopathology (10). Boosting of the CS protein vaccine with 16

the Ad35.CS and, in particular, with the Ad35.CS/Ad26.CS combination strongly 17

enhanced the levels of Th1 cytokines IFNγ and TNFα, while the levels of Th1 cytokine 18

IL-2, Th2 cytokines IL-6 and IL-10 and Th17 cytokine IL-17 were comparable to the 19

levels induced with the CS protein vaccine alone. This result indicated the capacity of the 20

three-component regimen to stimulate an overall balanced cytokine response, with a 21

strong shift towards the Th1 responses as compared to the homologous CS protein 22

regimen inducing primarily a Th2 biased response. While the role for Th1 type response 23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 20: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

20

in protection against malaria has been well documented (6, 12, 19, 20, 43, 47), to our 1

knowledge there are no reports concerning the role of Th17 cells in malaria infection. 2

However, there is mounting evidence that IL-17 might be relevant for protection against 3

parasitic infections, as it has been indicated by the induction of this cytokine in response 4

to infection with a number of parasites such as Eimeria maxima (18), Nippostrongglyus 5

brasiliensis (59) and Leishmania donovani (36). For instance, production of IL-17 and 6

IL-22 in humans was shown to have a strong and independent association with protection 7

the Kala Azar disease, caused by L. donovani (36). Other studies have also indicated a 8

role for Th17 cells in protection against other pathogens such as, Mycobacterium 9

tuberculosis (2, 23, 24), Streptococcus pneumoniae (29, 30, 58), Helicobactor pylori (11, 10

54), and Influenza virus (15, 57). In the current study, albeit no significant difference was 11

observed in the mean level of the IL-17 cytokines between different groups, the 12

adenovector containing regimens induced more uniform IL-17 responses as compared to 13

the protein immunization. 14

The limited and short lived protection induced with the CS protein vaccine points in two 15

directions of improvement using Ad.CS-based vaccines. One by administering Ad35.CS 16

vaccine as a priming vaccine to the CS protein vaccine, to strongly increase Th1 cellular 17

responses, as described in Stewart et al (51). Second, as demonstrated in the current 18

study, by administering the Ad35.CS/Ad26.CS combination as a booster vaccine (in 19

second year of life or even at school age) following an early-in-life protein CS vaccine, to 20

induce long lasting protection for which the Th1 type response and immune memory is 21

required. 22

23

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 21: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

21

ACKNOWLEDGMENTS 1

We acknowledge financial support from the European Commission (FP6 grant for the 2

project PRIBOMAL). 3

4

5

6

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 22: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

22

REFERENCES 1

2

1. Abbink, P., A. A. Lemckert, B. A. Ewald, D. M. Lynch, M. Denholtz, S. 3

Smits, L. Holterman, I. Damen, R. Vogels, A. R. Thorner, K. L. O'Brien, A. 4

Carville, K. G. Mansfield, J. Goudsmit, M. J. Havenga, and D. H. Barouch. 5 2007. Comparative seroprevalence and immunogenicity of six rare serotype 6

recombinant adenovirus vaccine vectors from subgroups B and D. J Virol 7

81:4654-63. 8

2. Aujla, S. J., Y. R. Chan, M. Zheng, M. Fei, D. J. Askew, D. A. Pociask, T. A. 9

Reinhart, F. McAllister, J. Edeal, K. Gaus, S. Husain, J. L. Kreindler, P. J. 10

Dubin, J. M. Pilewski, M. M. Myerburg, C. A. Mason, Y. Iwakura, and J. K. 11 Kolls. 2008. IL-22 mediates mucosal host defense against Gram-negative 12

bacterial pneumonia. Nat Med 14:275-81. 13

3. Barouch, D. 2009. First-in-human phase 1 safety and immunogenicity of an 14

Adenovirus Serotype 26 HIV-1 vaccine vector, La Conférence AIDS Vaccine 15

Paris, France. 16

4. Barouch, D. H., M. G. Pau, J. H. Custers, W. Koudstaal, S. Kostense, M. J. 17

Havenga, D. M. Truitt, S. M. Sumida, M. G. Kishko, J. C. Arthur, B. 18

Korioth-Schmitz, M. H. Newberg, D. A. Gorgone, M. A. Lifton, D. L. 19 Panicali, G. J. Nabel, N. L. Letvin, and J. Goudsmit. 2004. Immunogenicity of 20

recombinant adenovirus serotype 35 vaccine in the presence of pre-existing anti-21

Ad5 immunity. J Immunol 172:6290-7. 22

5. Bejon, P., J. Lusingu, A. Olotu, A. Leach, M. Lievens, J. Vekemans, S. 23

Mshamu, T. Lang, J. Gould, M. C. Dubois, M. A. Demoitie, J. F. Stallaert, P. 24

Vansadia, T. Carter, P. Njuguna, K. O. Awuondo, A. Malabeja, O. Abdul, S. 25

Gesase, N. Mturi, C. J. Drakeley, B. Savarese, T. Villafana, W. R. Ballou, J. 26 Cohen, E. M. Riley, M. M. Lemnge, K. Marsh, and L. von Seidlein. 2008. 27

Efficacy of RTS,S/AS01E vaccine against malaria in children 5 to 17 months of 28

age. N Engl J Med 359:2521-32. 29

6. Carvalho, L. J., C. T. Daniel-Ribeiro, and H. Goto. 2002. Malaria vaccine: 30

candidate antigens, mechanisms, constraints and prospects. Scand J Immunol 31

56:327-43. 32

7. Casimiro, D. R., A. J. Bett, T. M. Fu, M. E. Davies, A. Tang, K. A. Wilson, M. 33

Chen, R. Long, T. McKelvey, M. Chastain, S. Gurunathan, J. Tartaglia, E. 34 A. Emini, and J. Shiver. 2004. Heterologous human immunodeficiency virus 35

type 1 priming-boosting immunization strategies involving replication-defective 36

adenovirus and poxvirus vaccine vectors. J Virol 78:11434-8. 37

8. Catanzaro, A. T., R. A. Koup, M. Roederer, R. T. Bailer, M. E. Enama, Z. 38

Moodie, L. Gu, J. E. Martin, L. Novik, B. K. Chakrabarti, B. T. Butman, J. 39

G. Gall, C. R. King, C. A. Andrews, R. Sheets, P. L. Gomez, J. R. Mascola, G. 40 J. Nabel, and B. S. Graham. 2006. Phase 1 safety and immunogenicity 41

evaluation of a multiclade HIV-1 candidate vaccine delivered by a replication-42

defective recombinant adenovirus vector. J Infect Dis 194:1638-49. 43

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 23: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

23

9. Clyde, D. F., H. Most, V. C. McCarthy, and J. P. Vanderberg. 1973. 1

Immunization of man against sporozite-induced falciparum malaria. Am J Med 2

Sci 266:169-77. 3

10. Couper, K. N., D. G. Blount, M. S. Wilson, J. C. Hafalla, Y. Belkaid, M. 4

Kamanaka, R. A. Flavell, J. B. de Souza, and E. M. Riley. 2008. IL-10 from 5

CD4CD25Foxp3CD127 adaptive regulatory T cells modulates parasite clearance 6

and pathology during malaria infection. PLoS Pathog 4:e1000004. 7

11. DeLyria, E. S., R. W. Redline, and T. G. Blanchard. 2009. Vaccination of mice 8

against H pylori induces a strong Th-17 response and immunity that is neutrophil 9

dependent. Gastroenterology 136:247-56. 10

12. Doolan, D. L., and N. Martinez-Alier. 2006. Immune response to pre-11

erythrocytic stages of malaria parasites. Curr Mol Med 6:169-85. 12

13. Fitzgerald, J. C., G. P. Gao, A. Reyes-Sandoval, G. N. Pavlakis, Z. Q. Xiang, 13

A. P. Wlazlo, W. Giles-Davis, J. M. Wilson, and H. C. Ertl. 2003. A simian 14

replication-defective adenoviral recombinant vaccine to HIV-1 gag. J Immunol 15

170:1416-22. 16

14. Gilbert, S. C., J. Schneider, C. M. Hannan, J. T. Hu, M. Plebanski, R. 17

Sinden, and A. V. Hill. 2002. Enhanced CD8 T cell immunogenicity and 18

protective efficacy in a mouse malaria model using a recombinant adenoviral 19

vaccine in heterologous prime-boost immunisation regimes. Vaccine 20:1039-45. 20

15. Hamada, H., L. Garcia-Hernandez Mde, J. B. Reome, S. K. Misra, T. M. 21

Strutt, K. K. McKinstry, A. M. Cooper, S. L. Swain, and R. W. Dutton. 2009. 22

Tc17, a unique subset of CD8 T cells that can protect against lethal influenza 23

challenge. J Immunol 182:3469-81. 24

16. Havenga, M., R. Vogels, D. Zuijdgeest, K. Radosevic, S. Mueller, M. 25

Sieuwerts, F. Weichold, I. Damen, J. Kaspers, A. Lemckert, M. van 26

Meerendonk, R. van der Vlugt, L. Holterman, D. Hone, Y. Skeiky, R. 27 Mintardjo, G. Gillissen, D. Barouch, J. Sadoff, and J. Goudsmit. 2006. Novel 28

replication-incompetent adenoviral B-group vectors: high vector stability and 29

yield in PER.C6 cells. J Gen Virol 87:2135-43. 30

17. Holterman, L., R. Vogels, R. van der Vlugt, M. Sieuwerts, J. Grimbergen, J. 31

Kaspers, E. Geelen, E. van der Helm, A. Lemckert, G. Gillissen, S. Verhaagh, 32

J. Custers, D. Zuijdgeest, B. Berkhout, M. Bakker, P. Quax, J. Goudsmit, 33 and M. Havenga. 2004. Novel replication-incompetent vector derived from 34

adenovirus type 11 (Ad11) for vaccination and gene therapy: low seroprevalence 35

and non-cross-reactivity with Ad5. J Virol 78:13207-15. 36

18. Hong, Y. H., H. S. Lillehoj, E. P. Lillehoj, and S. H. Lee. 2006. Changes in 37

immune-related gene expression and intestinal lymphocyte subpopulations 38

following Eimeria maxima infection of chickens. Vet Immunol Immunopathol 39

114:259-72. 40

19. John, C. C., A. M. Moormann, D. C. Pregibon, P. O. Sumba, M. M. McHugh, 41

D. L. Narum, D. E. Lanar, M. D. Schluchter, and J. W. Kazura. 2005. 42

Correlation of high levels of antibodies to multiple pre-erythrocytic Plasmodium 43

falciparum antigens and protection from infection. Am J Trop Med Hyg 73:222-8. 44

20. John, C. C., A. J. Tande, A. M. Moormann, P. O. Sumba, D. E. Lanar, X. M. 45

Min, and J. W. Kazura. 2008. Antibodies to pre-erythrocytic Plasmodium 46

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 24: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

24

falciparum antigens and risk of clinical malaria in Kenyan children. J Infect Dis 1

197:519-26. 2

21. Kester, K. E., J. F. Cummings, C. F. Ockenhouse, R. Nielsen, B. T. Hall, D. 3

M. Gordon, R. J. Schwenk, U. Krzych, C. A. Holland, G. Richmond, M. G. 4

Dowler, J. Williams, R. A. Wirtz, N. Tornieporth, L. Vigneron, M. 5

Delchambre, M. A. Demoitie, W. R. Ballou, J. Cohen, and D. G. Heppner, Jr. 6 2008. Phase 2a trial of 0, 1, and 3 month and 0, 7, and 28 day immunization 7

schedules of malaria vaccine RTS,S/AS02 in malaria-naive adults at the Walter 8

Reed Army Institute of Research. Vaccine 26:2191-202. 9

22. Kester, K. E., J. F. Cummings, O. Ofori-Anyinam, C. F. Ockenhouse, U. 10

Krzych, P. Moris, R. Schwenk, R. A. Nielsen, Z. Debebe, E. Pinelis, L. 11

Juompan, J. Williams, M. Dowler, V. A. Stewart, R. A. Wirtz, M. C. Dubois, 12 M. Lievens, J. Cohen, W. R. Ballou, and D. G. Heppner, Jr. 2009. 13

Randomized, double-blind, phase 2a trial of falciparum malaria vaccines 14

RTS,S/AS01B and RTS,S/AS02A in malaria-naive adults: safety, efficacy, and 15

immunologic associates of protection. J Infect Dis 200:337-46. 16

23. Khader, S. A., G. K. Bell, J. E. Pearl, J. J. Fountain, J. Rangel-Moreno, G. E. 17

Cilley, F. Shen, S. M. Eaton, S. L. Gaffen, S. L. Swain, R. M. Locksley, L. 18 Haynes, T. D. Randall, and A. M. Cooper. 2007. IL-23 and IL-17 in the 19

establishment of protective pulmonary CD4+ T cell responses after vaccination 20

and during Mycobacterium tuberculosis challenge. Nat Immunol 8:369-77. 21

24. Khader, S. A., J. E. Pearl, K. Sakamoto, L. Gilmartin, G. K. Bell, D. M. 22

Jelley-Gibbs, N. Ghilardi, F. deSauvage, and A. M. Cooper. 2005. IL-23 23

compensates for the absence of IL-12p70 and is essential for the IL-17 response 24

during tuberculosis but is dispensable for protection and antigen-specific IFN-25

gamma responses if IL-12p70 is available. J Immunol 175:788-95. 26

25. Lemckert, A. A., J. Grimbergen, S. Smits, E. Hartkoorn, L. Holterman, B. 27

Berkhout, D. H. Barouch, R. Vogels, P. Quax, J. Goudsmit, and M. J. 28 Havenga. 2006. Generation of a novel replication-incompetent adenoviral vector 29

derived from human adenovirus type 49: manufacture on PER.C6 cells, tropism 30

and immunogenicity. J Gen Virol 87:2891-9. 31

26. Letvin, N. L., Y. Huang, B. K. Chakrabarti, L. Xu, M. S. Seaman, K. 32

Beaudry, B. Korioth-Schmitz, F. Yu, D. Rohne, K. L. Martin, A. Miura, W. 33

P. Kong, Z. Y. Yang, R. S. Gelman, O. G. Golubeva, D. C. Montefiori, J. R. 34 Mascola, and G. J. Nabel. 2004. Heterologous envelope immunogens contribute 35

to AIDS vaccine protection in rhesus monkeys. J Virol 78:7490-7. 36

27. Liu, J., B. A. Ewald, D. M. Lynch, M. Denholtz, P. Abbink, A. A. Lemckert, 37

A. Carville, K. G. Mansfield, M. J. Havenga, J. Goudsmit, and D. H. 38 Barouch. 2008. Magnitude and phenotype of cellular immune responses elicited 39

by recombinant adenovirus vectors and heterologous prime-boost regimens in 40

rhesus monkeys. J Virol 82:4844-52. 41

28. Liu, J., K. L. O'Brien, D. M. Lynch, N. L. Simmons, A. La Porte, A. M. 42

Riggs, P. Abbink, R. T. Coffey, L. E. Grandpre, M. S. Seaman, G. Landucci, 43

D. N. Forthal, D. C. Montefiori, A. Carville, K. G. Mansfield, M. J. Havenga, 44 M. G. Pau, J. Goudsmit, and D. H. Barouch. 2009. Immune control of an SIV 45

challenge by a T-cell-based vaccine in rhesus monkeys. Nature 457:87-91. 46

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 25: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

25

29. Lu, Y. J., J. Gross, D. Bogaert, A. Finn, L. Bagrade, Q. Zhang, J. K. Kolls, A. 1

Srivastava, A. Lundgren, S. Forte, C. M. Thompson, K. F. Harney, P. W. 2 Anderson, M. Lipsitch, and R. Malley. 2008. Interleukin-17A mediates 3

acquired immunity to pneumococcal colonization. PLoS Pathog 4:e1000159. 4

30. Malley, R., A. Srivastava, M. Lipsitch, C. M. Thompson, C. Watkins, A. 5

Tzianabos, and P. W. Anderson. 2006. Antibody-independent, interleukin-17A-6

mediated, cross-serotype immunity to pneumococci in mice immunized 7

intranasally with the cell wall polysaccharide. Infect Immun 74:2187-95. 8

31. Mettens, P., P. M. Dubois, M. A. Demoitie, B. Bayat, M. N. Donner, P. 9

Bourguignon, V. A. Stewart, D. G. Heppner, Jr., N. Garcon, and J. Cohen. 10 2008. Improved T cell responses to Plasmodium falciparum circumsporozoite 11

protein in mice and monkeys induced by a novel formulation of RTS,S vaccine 12

antigen. Vaccine 26:1072-82. 13

32. Ophorst, O. J., K. Radosevic, M. J. Havenga, M. G. Pau, L. Holterman, B. 14

Berkhout, J. Goudsmit, and M. Tsuji. 2006. Immunogenicity and protection of 15

a recombinant human adenovirus serotype 35-based malaria vaccine against 16

Plasmodium yoelii in mice. Infect Immun 74:313-20. 17

33. Ophorst, O. J., K. Radosevic, K. Ouwehand, W. van Beem, R. Mintardjo, J. 18

Sijtsma, J. Kaspers, A. Companjen, L. Holterman, J. Goudsmit, and M. J. 19 Havenga. 2007. Expression and immunogenicity of the Plasmodium falciparum 20

circumsporozoite protein: the role of GPI signal sequence. Vaccine 25:1426-36. 21

34. Overstreet, M. G., I. A. Cockburn, Y. C. Chen, and F. Zavala. 2008. 22

Protective CD8 T cells against Plasmodium liver stages: immunobiology of an 23

'unnatural' immune response. Immunol Rev 225:272-83. 24

35. Pinto, A. R., J. C. Fitzgerald, W. Giles-Davis, G. P. Gao, J. M. Wilson, and H. 25

C. Ertl. 2003. Induction of CD8+ T cells to an HIV-1 antigen through a prime 26

boost regimen with heterologous E1-deleted adenoviral vaccine carriers. J 27

Immunol 171:6774-9. 28

36. Pitta, M. G., A. Romano, S. Cabantous, S. Henri, A. Hammad, B. Kouriba, L. 29

Argiro, M. el Kheir, B. Bucheton, C. Mary, S. H. El-Safi, and A. Dessein. 30 2009. IL-17 and IL-22 are associated with protection against human kala azar 31

caused by Leishmania donovani. J Clin Invest 119:2379-87. 32

37. Priddy, F. H., D. Brown, J. Kublin, K. Monahan, D. P. Wright, J. Lalezari, S. 33

Santiago, M. Marmor, M. Lally, R. M. Novak, S. J. Brown, P. Kulkarni, S. A. 34

Dubey, L. S. Kierstead, D. R. Casimiro, R. Mogg, M. J. DiNubile, J. W. 35 Shiver, R. Y. Leavitt, M. N. Robertson, D. V. Mehrotra, and E. Quirk. 2008. 36

Safety and immunogenicity of a replication-incompetent adenovirus type 5 HIV-1 37

clade B gag/pol/nef vaccine in healthy adults. Clin Infect Dis 46:1769-81. 38

38. Radosevic, K., A. Rodriguez, A. Lemckert, and J. Goudsmit. 2009. 39

Heterologous prime-boost vaccinations for poverty-related diseases: advantages 40

and future prospects. Expert Rev Vaccines 8:577-92. 41

39. Radosevic, K., A. Rodriguez, R. Mintardjo, D. Tax, K. L. Bengtsson, C. 42

Thompson, M. Zambon, G. J. Weverling, F. Uytdehaag, and J. Goudsmit. 43 2008. Antibody and T-cell responses to a virosomal adjuvanted H9N2 avian 44

influenza vaccine: impact of distinct additional adjuvants. Vaccine 26:3640-6. 45

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 26: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

26

40. Radosevic, K., C. W. Wieland, A. Rodriguez, G. J. Weverling, R. Mintardjo, 1

G. Gillissen, R. Vogels, Y. A. Skeiky, D. M. Hone, J. Sadoff, T. van der Poll, 2 M. Havenga, and J. Goudsmit. 2007. Protective immune responses to an rAd35 3

TB vaccine in two mouse strains (H-2d vs. H-2b): CD4 and CD8 T-cell epitope 4

mapping and role of IFN{gamma}. Infect Immun 75:4105-15. 5

41. Roberts, D. M., A. Nanda, M. J. Havenga, P. Abbink, D. M. Lynch, B. A. 6

Ewald, J. Liu, A. R. Thorner, P. E. Swanson, D. A. Gorgone, M. A. Lifton, A. 7

A. Lemckert, L. Holterman, B. Chen, A. Dilraj, A. Carville, K. G. Mansfield, 8 J. Goudsmit, and D. H. Barouch. 2006. Hexon-chimaeric adenovirus serotype 5 9

vectors circumvent pre-existing anti-vector immunity. Nature 441:239-43. 10

42. Rodrigues, E. G., F. Zavala, D. Eichinger, J. M. Wilson, and M. Tsuji. 1997. 11

Single immunizing dose of recombinant adenovirus efficiently induces CD8+ T 12

cell-mediated protective immunity against malaria. J Immunol 158:1268-74. 13

43. Rodrigues, M. M., A. S. Cordey, G. Arreaza, G. Corradin, P. Romero, J. L. 14

Maryanski, R. S. Nussenzweig, and F. Zavala. 1991. CD8+ cytolytic T cell 15

clones derived against the Plasmodium yoelii circumsporozoite protein protect 16

against malaria. Int Immunol 3:579-85. 17

44. Rodriguez, A., J. Goudsmit, A. Companjen, R. Mintardjo, G. Gillissen, D. 18

Tax, J. Sijtsma, G. J. Weverling, L. Holterman, D. E. Lanar, M. J. Havenga, 19 and K. Radosevic. 2008. Impact of recombinant adenovirus serotype 35 priming 20

versus boosting of a Plasmodium falciparum protein: characterization of T- and 21

B-cell responses to liver-stage antigen 1. Infect Immun 76:1709-18. 22

45. Rodriguez, A., R. Mintardjo, D. Tax, G. Gillissen, J. Custers, M. G. Pau, J. 23

Klap, S. Santra, H. Balachandran, N. L. Letvin, J. Goudsmit, and K. 24 Radosevic. 2009. Evaluation of a prime-boost vaccine schedule with distinct 25

adenovirus vectors against malaria in rhesus monkeys. Vaccine 27:6226-33. 26

46. Schofield, L., J. Villaquiran, A. Ferreira, H. Schellekens, R. Nussenzweig, 27

and V. Nussenzweig. 1987. Gamma interferon, CD8+ T cells and antibodies 28

required for immunity to malaria sporozoites. Nature 330:664-6. 29

47. Seguin, M. C., F. W. Klotz, I. Schneider, J. P. Weir, M. Goodbary, M. 30

Slayter, J. J. Raney, J. U. Aniagolu, and S. J. Green. 1994. Induction of nitric 31

oxide synthase protects against malaria in mice exposed to irradiated Plasmodium 32

berghei infected mosquitoes: involvement of interferon gamma and CD8+ T cells. 33

J Exp Med 180:353-8. 34

48. Shiver, J. W., and E. A. Emini. 2004. Recent advances in the development of 35

HIV-1 vaccines using replication-incompetent adenovirus vectors. Annu Rev Med 36

55:355-72. 37

49. Shiver, J. W., T. M. Fu, L. Chen, D. R. Casimiro, M. E. Davies, R. K. Evans, 38

Z. Q. Zhang, A. J. Simon, W. L. Trigona, S. A. Dubey, L. Huang, V. A. 39

Harris, R. S. Long, X. Liang, L. Handt, W. A. Schleif, L. Zhu, D. C. Freed, N. 40

V. Persaud, L. Guan, K. S. Punt, A. Tang, M. Chen, K. A. Wilson, K. B. 41

Collins, G. J. Heidecker, V. R. Fernandez, H. C. Perry, J. G. Joyce, K. M. 42

Grimm, J. C. Cook, P. M. Keller, D. S. Kresock, H. Mach, R. D. Troutman, 43

L. A. Isopi, D. M. Williams, Z. Xu, K. E. Bohannon, D. B. Volkin, D. C. 44

Montefiori, A. Miura, G. R. Krivulka, M. A. Lifton, M. J. Kuroda, J. E. 45

Schmitz, N. L. Letvin, M. J. Caulfield, A. J. Bett, R. Youil, D. C. Kaslow, and 46

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 27: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

27

E. A. Emini. 2002. Replication-incompetent adenoviral vaccine vector elicits 1

effective anti-immunodeficiency-virus immunity. Nature 415:331-5. 2

50. Skeiky, Y. A., and J. C. Sadoff. 2006. Advances in tuberculosis vaccine 3

strategies. Nat Rev Microbiol 4:469-76. 4

51. Stewart, V. A., S. M. McGrath, P. M. Dubois, M. G. Pau, P. Mettens, J. 5

Shott, M. Cobb, J. R. Burge, D. Larson, L. A. Ware, M. A. Demoitie, G. J. 6

Weverling, B. Bayat, J. H. Custers, M. C. Dubois, J. Cohen, J. Goudsmit, and 7 D. G. Heppner, Jr. 2007. Priming with an adenovirus 35-circumsporozoite 8

protein (CS) vaccine followed by RTS,S/AS01B boosting significantly improves 9

immunogenicity to Plasmodium falciparum CS compared to that with either 10

malaria vaccine alone. Infect Immun 75:2283-90. 11

52. Tatsis, N., and H. C. Ertl. 2004. Adenoviruses as vaccine vectors. Mol Ther 12

10:616-29. 13

53. Vekemans, J., A. Leach, and J. Cohen. 2009. Development of the RTS,S/AS 14

malaria candidate vaccine. Vaccine 27 Suppl 6:G67-71. 15

54. Velin, D., L. Favre, E. Bernasconi, D. Bachmann, C. Pythoud, E. Saiji, H. 16

Bouzourene, and P. Michetti. 2009. Interleukin-17 is a critical mediator of 17

vaccine-induced reduction of Helicobacter infection in the mouse model. 18

Gastroenterology 136:2237-2246 e1. 19

55. Vogels, R., D. Zuijdgeest, R. van Rijnsoever, E. Hartkoorn, I. Damen, M. P. 20

de Bethune, S. Kostense, G. Penders, N. Helmus, W. Koudstaal, M. Cecchini, 21

A. Wetterwald, M. Sprangers, A. Lemckert, O. Ophorst, B. Koel, M. van 22

Meerendonk, P. Quax, L. Panitti, J. Grimbergen, A. Bout, J. Goudsmit, and 23 M. Havenga. 2003. Replication-deficient human adenovirus type 35 vectors for 24

gene transfer and vaccination: efficient human cell infection and bypass of 25

preexisting adenovirus immunity. J Virol 77:8263-71. 26

56. Weiss, W. R., M. Sedegah, R. L. Beaudoin, L. H. Miller, and M. F. Good. 27

1988. CD8+ T cells (cytotoxic/suppressors) are required for protection in mice 28

immunized with malaria sporozoites. Proc Natl Acad Sci U S A 85:573-6. 29

57. Williman, J., E. Lockhart, L. Slobbe, G. Buchan, and M. Baird. 2006. The use 30

of Th1 cytokines, IL-12 and IL-23, to modulate the immune response raised to a 31

DNA vaccine delivered by gene gun. Vaccine 24:4471-4. 32

58. Zhang, Z., T. B. Clarke, and J. N. Weiser. 2009. Cellular effectors mediating 33

Th17-dependent clearance of pneumococcal colonization in mice. J Clin Invest 34

119:1899-909. 35

59. Zhugong Liu, H. L., Qian Liu, Gity Mousavi and William C. Gause 2008. The 36

parasite Nippostrongylus brasiliensis induces multiple regulatory pathways that 37

control increases of IL-17 expression and associated pathology in the lung. 38

FASEB Journal 22. 39

40

41

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 28: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

28

FIGURE LEGENDS 1

2

Figure 1. Characterization of the yeast-produced CS protein. (A) The yeast-produced 3

CS protein was analyzed by CS-specific Western blot and InstantBlue staining. (B) 4

BALB/c mice (n = 5 per group) were immunized s.c. three times with 5, 10 or 25 µg 5

montanide ISA720 adjuvanted CS protein at 3-weeks interval. CS-specific humoral 6

responses were assessed every 1.5 weeks up to 8 weeks after the initial immunization by 7

ELISA. Mean titers with 95% confidence interval are depicted. EU; ELISA units. (C) 8

IgG2a/IgG1 ratios upon measurement of CS-specific IgG2a and IgG1 responses 8 weeks 9

after the initial immunization. Bars represent geometric means IgG2a/IgG1 ratios 10

11

Figure 2. Immunogenicity of heterologous prime-boost regimen comprised of the 12

yeast-produced CS protein and Ad35.CS. BALB/c mice(n = 8 per group) were 13

immunized as indicated in the graphs. A negative control group received the adjuvant and 14

Ad35.Empty vector (sham). Two weeks after the boost immunization CS-specific 15

humoral immune responses were assessed by (A) CS-specific IgG responses using 16

ELISA and (B) IgG2a/IgG1 ratios upon measurement of CS-specific IgG2a and IgG1 17

responses. CS-specific CD8+ T-cell immune responses were assessed by (C) IFNγ 18

ELISPOT and (D) IFNγ ICS. Bars represent geometric means of (A) ELISA units (EU), 19

(B) IgG2a/IgG1 ratios, (C) spot forming units (SFU), or (D) percentage of IFNγ+ CD4+ 20

or IFNγ+ CD8+ positive cells. The background level of cytokine staining was typically 21

lower than 0.01% for the CD4+ T cells and lower than 0.05% for the CD8+ T cells. 22

23

24

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 29: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

29

Figure 3. Immunogenicity of a three-component heterologous prime-boost regimen. 1

BALB/c mice(n = 8 per group) were immunized as indicated in the graphs. A negative 2

control group received the adjuvant and Ad.Empty vectors (sham). Two-weeks (A) and 3

eight-weeks (C) after the final boost immunization CS-specific IFNγ+ CD8+ T-cell 4

responses were assessed using ELISPOT. Two-weeks (B) and eight-weeks (D) after the 5

final boost immunization CS-specific humoral immune responses were assessed by IgG 6

ELISA. Bars represent geometric means of (A, C) spot forming units (SFU) or (B, D) 7

ELISA units (EU). 8

9

Figure 4. Cytokine profile induced by the different vaccination regimens. 10

BALB/c mice (n = 8 per group) were immunized as indicated in the graphs. A negative 11

control group received the adjuvant and Ad.Empty vectors (sham). Two-weeks after the 12

final boost immunization cytokine expression was assessed by CBA assay upon 48 hrs in 13

vitro stimulation of splenocytes with the CS protein. Bars represent geometric means of 14

pg/ml IFNγ, TNFα, IL-2, IL-6, IL-10 or IL-17 cytokine levels. In none of the immunized 15

mice measurable levels of IL-4 were detected. 16

17

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 30: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

Figure 1

0 1 2 3 4 5 6 7 81

10

100

1000

10000

100000

10000005 ug

10 ug

25 ug

Prime Boost Boost

Weeks

EU

A B

20

30

40

50

60

80

100

C

0.0001

0.001

0.01

0.1

1

10

100

- + - + - + 5 ug 10 ug 25 ug

AdjuvantProtein

rati

o I

gG

2a/I

gG

1 Th1

Th2

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 31: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

A

C

B

Figure 2

0

100

200

300

400

P < 0.05

IFN

- γγ γγ S

FU

/10

6 s

ple

no

cyte

s

0.0001

0.001

0.01

0.1

1

10

100 P < 0.05

rati

o IgG

2a/IgG

1

1

10

100

1000

10000

100000

N.S.

EU

sham

sham

CS Prot

CS Prot

CS Prot

Ad35.CS

sham

sham

CS Prot

CS Prot

CS Prot

Ad35.CS

CS Prot

CS Prot

CS Prot

Ad35.CS

Th1

Th2

D

CS ProtCS Prot

CS ProtAd35.CS

0.00

0.25

0.50

0.75

1.00P < 0.05

CD4

CD8

% IFNγγ γγ+ C

D+ c

ells

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 32: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

A

C

Figure 3

B

D

0

500

1000

1500

2000

2500 P < 0.05

IFN

- γγ γγ S

FU

/ 1

06 s

ple

no

cy

tes

1

10

100

1000

10000

100000N.S.

EU

0

500

1000

1500

2000

2500P < 0.05

IFN

- γγ γγ S

FU

/ 10

6 s

ple

no

cyte

s

1

10

100

1000

10000

100000N.S.

EU

shamsham

sham

CS Prot

Ad35.CS

CS ProtAd35.CS

Ad26.CS

shamsham

sham

CS Prot

Ad35.CS

CS ProtAd35.CS

Ad26.CS

sham

shamsham

CS Prot

Ad35.CS

CS Prot

Ad35.CSAd26.CS

sham

shamsham

CS Prot

Ad35.CS

CS Prot

Ad35.CSAd26.CS

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 33: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

Figure 4

0

1000

2000

3000

4000

5000P < 0.05

P < 0.05

IFNγγ γγ (

pg

/ml)

0

100

200

300

400

500

600P < 0.05

TN

Fαα αα

(p

g/m

l)

0

100

200

300N.S.

IL-2

(p

g/m

l)

0

100

200

300

400

500

600

700

800N.S.

IL-6

(p

g/m

l)

0

100

200

300N.S.

IL-1

0 (

pg

/ml)

0

100

200

300N.S.

IL-1

7 (

pg

/ml)

sham

shamsham

CS Prot

CS Prot

CS Prot

Ad35.CSAd26.CS

CS Prot

Ad35.CS

sham

shamsham

CS Prot

CS Prot

CS Prot

Ad35.CSAd26.CS

CS Prot

Ad35.CS

sham

shamsham

CS Prot

CS Prot

CS Prot

Ad35.CSAd26.CS

CS Prot

Ad35.CS

Th1

Th2 Th17

on February 21, 2021 by guest

http://cvi.asm.org/

Dow

nloaded from

Page 34: Downloaded from on April 9, 2020 by guest · 9/8/2010  · 11 recombinant low seroprevalent adenoviruses expressing P. falciparum CS antigen, 12 Ad35.CS (subgroup B) and Ad26.CS (subgroup

CLINICAL AND VACCINE IMMUNOLOGY, Feb. 2011, p. 353 Vol. 18, No. 21556-6811/11/$12.00 doi:10.1128/CVI.00554-10Copyright © 2011, American Society for Microbiology. All Rights Reserved.

ERRATUM

The Th1 Immune Response to Plasmodium falciparum Circumsporozoite ProteinIs Boosted by Adenovirus Vectors 35 and 26 with a Homologous Insert

Katarina Radosevic, Ariane Rodriguez, Angelique A. C. Lemckert, Marjolein van der Meer,Gert Gillissen, Carolien Warnar, Rie von Eyben, Maria Grazia Pau, and Jaap Goudsmit

Crucell Holland B.V., Leiden, Netherlands, and Center for Poverty-Related Communicable Diseases,Academic Medical Center, Amsterdam, Netherlands

Volume 17, no. 11, pages 1687–1694, 2010. Page 1687, Abstract, the first sentence should read: “The most advanced malariavaccine, RTS,S, is comprised of a portion of the Plasmodium falciparum circumsporozoite (CS) protein, fused to and admixed withthe hepatitis B virus surface antigen, and an adjuvant.”

The third sentence should read: “In the present study, we tested the hypothesis that the Th1 immune response to CS protein,in particular the CD8� T-cell response, which is needed for strong and lasting malaria immunity, is boosted to sustainable levelsby adenovirus vectors 35 and 26 with a homologous insert (Ad35.CS/Ad26.CS).”

353