You are on page 1of 12

ORIGINAL ARTICLE

A Multi-Subunit Chlamydial Vaccine Induces Antibody and


Cell-Mediated Immunity in Immunized Koalas (Phascolarctos
cinereus): Comparison of Three Different Adjuvants
Alison J. Carey1, Peter Timms1, Galit Rawlinson2, Jacqui Brumm2, Karen Nilsson2, Jonathon M. Harris1,
Kenneth W. Beagley1
1
Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Qld, Australia;
2
Lone Pine Koala Sanctuary, Fig Tree Pocket, Brisbane, Qld, Australia

Keywords Problem
Adjuvant, antibody, Chlamydia, koala Chlamydial infections represent a major threat to the survival of the
koala. Infections caused by Chlamydia pecorum cause blindness, infertility,
Correspondence
Kenneth W. Beagley, Institute of Health and
pneumonia and urinary tract infections and represent a threat to the
Biomedical Innovation, Queensland University survival of the species. Little is known about the immune response in
of Technology, 60 Musk Ave, Kelvin Grove, koalas, or the safety of commonly used adjuvants for induction of pro-
Qld 4059, Australia. tective systemic and mucosal immunity.
E-mail: k2.beagley@qut.edu.au
Method of study
Submitted September 1, 2009; In the present study, we immunized 18 healthy female koalas subcuta-
accepted October 9, 2009. neously with a combination of three chlamydial antigens [major outer
membrane protein (MOMP), NrdB and TC0512 (Omp85)] mixed with
Citation
Carey AJ, Timms P, Rawlinson G, Brumm J,
one of three different adjuvants [Alhydrogel, Immunostimulating Com-
Nilsson K, Harris JM, Beagley KW. A multi- plex (ISC) and TiterMax Gold].
subunit chlamydial vaccine induces antibody
and cell-mediated immunity in immunized Results
koalas (Phascolarctos cinereus): comparison of All adjuvants induced strong neutralizing IgG responses in plasma
three different adjuvants. Am J Reprod against the three antigens with prolonged responses lasting more than
Immunol 2010; 63: 161–172 270 days seen in Alhydrogel and ISC immunized animals. Cloacal IgG
responses lasting >270 days were also induced in ISC-immunized ani-
doi:10.1111/j.1600-0897.2009.00776.x
mals. Chlamydia-specific peripheral blood mononuclear cell proliferative
responses were elicited by both Alhydrogel and ISC, and these lasted
>270 days in the ISC group.

Conclusion
The data show that a multi-subunit chlamydial vaccine, given subcuta-
neously, can elicit Chlamydia-specific cell-mediated and antibody
responses in the koala demonstrating that the development of a protec-
tive vaccine is feasible.

urban development in particular, these combined


Introduction
threats mean that many koala populations in Austra-
Chlamydia continues to be the most important infec- lia are under significant threat of collapse. Koalas are
tious disease affecting the koala (Phascolarctos cinere- infected with two species of Chlamydia, C. pecorum
us).1–4 Together with habitat destruction, because of and C. pneumoniae.5 Chlamydia pecorum is always the
American Journal of Reproductive Immunology 63 (2010) 161–172
ª 2010 John Wiley & Sons A/S 161
CAREY ET AL.

most prevalent and is present at higher levels in Because MOMP is a major target of the immune
infected animals.3 Both species can be detected at response, immunization with MOMP will only elicit
both ocular and genital sites; however, it is generally protection against homologous serovars, but not
accepted that C. pecorum causes the most serious dis- against serovars that express a different MOMP.
ease. Koalas suffer a similar spectrum of chlamydial Sequencing of the MOMP-encoding ompA gene of
diseases as seen in humans. Ocular infections cause C. pecorum from animals, including koalas,18 has
keratoconjunctivitis leading to blindness.6,7 Rhinitis demonstrated a similar level of diversity to that seen
and pneumonia are associated with respiratory infec- among human C. trachomatis serovars, suggesting
tions and urinary tract infections cause cystitis and that a MOMP-based vaccine may also not be effec-
continual urine soiling or ‘wet bottom’.2 Infections tive in koalas. Recent studies by our group have
of the genital tract can cause severe inflammation identified a number of chlamydial antigens that elicit
and fibrosis resulting in cystitis and infertility.1 Many protective immunity against genital infections in
koalas carry Chlamydia without displaying symptoms, mouse models and are highly conserved across
but increasing numbers of animals will develop overt known chlamydial species. Ribonucleotide reductase
disease when stressed by environmental pressures (NrdB)19 is an enzyme that catalyses the synthesis of
such as habitat loss. Treatment options for chlamyd- the four deoxynucleoside triphosphates (dNTPs)
ial infections are presently limited to antibiotics; from host cell-derived ribonucleoside triphosphates,
however, antibiotics can adversely affect the intesti- with its activity being the rate limiting step for chla-
nal microflora and health of the animals.8 Anti- mydial DNA synthesis. Comparison of NrdB
biotics can also cause persistent infections that may sequences across the published chlamydial genomes
evade immune responses but revert to an acute demonstrates a high degree (>90%) of sequence
infectious state when treatment ceases.9 Persistent conservation. NrdB also contains predicted human
infections are also believed to enhance the inflam- and mouse helper T-cell epitopes, with the region of
matory damage that is the hallmark of chlamydial NrdB containing these epitopes being 100% con-
disease. A successful vaccine to prevent chlamydial served across the published chlamydial genomes
infection would not only overcome the limitations of including C. pecorum. TC0512 is a putative outer
current antibiotic treatments but would also allow membrane protein (omp85) from C. muridarum that
for protection of animals undergoing relocation. An was identified using expression library immunization
effective vaccine requires both an antigen that elicits as an antigen that elicits partial protection against
protective immunity combined with an adjuvant genital C. muridarum infection in mice.20 Comparison
that is safe and that elicits the appropriate immune of omp85 gene sequences across chlamydial species
response. To date, the only adjuvants that have been showed >70% sequence similarity across the entire
used in koalas are Freund’s adjuvants.10–12 These molecule. As with NrdB, there are regions of TC0512
can cause severe adverse local reactions in many that contain sequences that are more highly con-
species13 and are generally considered to be inappro- served and may contain T-cell epitopes. As vaccines
priate for use outside the research laboratory. There based on whole-killed Chlamydia have the potential
are currently no data on the safety of other adju- to exacerbate inflammatory disease15 and because
vants in the koala. this study did not involve a challenge infection, we
Early trials of human vaccines for trachoma, using combined three recombinant chlamydial antigens,
whole-killed Chlamydia, resulted in exacerbated dis- MOMP and TC0512 isolated from C. muridarum and
ease in many recipients following a subsequent nat- NrdB isolated from C. trachomatis, in our experimen-
ural infection.14,15 Since then, chlamydial vaccine tal vaccine. This allowed us to determine if animals
research has focused on defining the antigens that responded to each of the individual vaccine compo-
elicit protective immunity without exacerbating nents and also if the use of a combination of moder-
inflammatory disease. The major outer membrane ate to highly conserved antigens would elicit
protein (MOMP) of Chlamydia is the best-studied cross-protective immunity against various chlamydial
antigen to date and in small animal models, it is able species.
to elicit significant protection against infection.16,17 The immune system of the koala has been
The major problem with MOMP-based vaccines is described as ‘lazy’,11 with humoral responses taking
the sequence diversity in the surface-exposed vari- significantly longer to develop than in eutherians.
able domains of different C. trachomatis serovars. This is not supported by other studies, however, and
American Journal of Reproductive Immunology 63 (2010) 161–172
162 ª 2010 John Wiley & Sons A/S
DEVELOPING A CHLAMYDIA VACCINE FOR THE KOALA

the development of cell-mediated immunity may be


Antigens
similar to that in non-metatherian mammals.21 Koa-
las have been shown to mount an antibody response Three recombinant chlamydial proteins were used.
following immunization with the antigen bovine A recombinant maltose binding protein-major outer
serum albumin (BSA)10 and koala peripheral blood membrane protein (MBP-MOMP) from C. murida-
leukocytes (PBL) do proliferate when cultured with rum, produced in Escherichia coli, was generously pro-
mitogens such as concanavalin A (ConA) and phyto- vided by Dr. Harlan Caldwell (Rocky Mountain
haemagglutinin (PHA),22 although lymphocyte pro- Laboratories)23 and was purified as described.16
liferative responses were not detected following Ribonucleotide reductase small chain protein (NrdB)
skin-painting with the contact-sensitizing agent 2-4- from C. trachomatis serovar D was cloned in pRSET-A
dinitrofluorobenzene.11 It must be said, however, (InVitrogen, Sydney, Australia), expressed in E. coli
that knowledge of the koala immune response is and purified as described.19 TC0512 (Omp85), a
scarce and this is mirrored by the lack of reagents putative outer membrane protein from C. muridrum,
available to measure immunity in the koala. As a was produced as a recombinant His-tagged protein
first step toward developing a safe and effective chla- in E. coli using the gene fragment identified by
mydial vaccine for the koala, we immunized groups McNeilly et al.20 The three antigens were chosen
of healthy female koalas with three different adju- because each was able to elicit partial protection
vants [Immuno stimulating complex (ISC), Alhydro- against genital challenge in the mouse model with
gel and TiterMax Gold] currently used in veterinary either C. muridarum16,20 or C. trachomatis.19
and animal vaccines, admixed with a combination of
three chlamydial antigens (MOMP, NrdB, TC0512).
Adjuvants
Signs of adverse reactions at the injection sites were
monitored after each immunization. Antigen-specific Three adjuvants were evaluated in this study. Immu-
lymphocyte proliferation was measured in peripheral nostimulating complex (ISC, lot #08141a6) was a
blood leukocytes over a 10-month period, together generous gift from Dr. John Walker, Pfizer Animal
with plasma and reproductive tract antibody levels. Health, VMRD, Parkville, Australia. Alhydrogel (Alu-
Plasma antibody was also assayed for neutralizing minum hydroxide Gel, Batch #3957; Accurate
activity against three different species of Chlamydia. Chemical and Scientific Corporation, Westbury, NY,
Two of the three adjuvants were well-tolerated by USA) was obtained from Auspep Pty Ltd, Parkville
koalas. High levels of antigen-specific peripheral Australia. TiterMax Gold adjuvant was obtained
blood mononuclear cell (PBMC) proliferation were from TiterMax USA, Norcross, GA, USA.
observed in animals immunized with ISC and plasma
antibody against all three antigens and were present
Immunizations
in all animals in the ISC and Alhydrogel groups
throughout the study. Plasma antibody was able to All animals were immunized subcutaneously on days
inhibit in vitro chlamydial infections of two species of 0, 30 and 60 with 50 lg each of MBP-MOMP, TC0512
Chlamydia, to varying degrees. Our data show that a and NrdB combined with 50 lg of ISC in 1 mL of PBS
multi-subunit vaccine approach may be able to pro- (group 1), Alhydrogel (final concentration 20%) in
vide protection against chlamydial infection in the 1 mL PBS (group 2) or suspended in PBS and emulsi-
koala. fied in an equal volume of TiterMax Gold (group 3).
Blood sample was collected into EDTA vacutainer
tubes from all animals at 4-week intervals, beginning
Materials and methods
2 weeks after the second immunization, up to
270 days. Cloacal swabs were collected at monthly
Animals
intervals following the third immunization.
Eighteen female koalas aged 2–5 years were assigned
to one of three groups, each of six animals. Animals
Lymphocyte Stimulation Assay
were housed at the Lone Pine Koala Sanctuary
(Brisbane, Australia). All procedures were approved Blood samples were centrifuged at 41 · g (Beckman
by the Queensland University Animal Ethics Com- Coulter GS-6R, centrifuge with a GH3.8 rotor) for
mittee. 10 min. Plasma was collected and stored frozen at
American Journal of Reproductive Immunology 63 (2010) 161–172
ª 2010 John Wiley & Sons A/S 163
CAREY ET AL.

)20C until further use. An equal volume of Hanks plasma was initially diluted 1:500. For detection of
Balanced Salt Solution (HBSS) was added to the pel- antibody in reproductive tract secretions, cloacal
leted blood and mixed by inverting the tube several swabs were collected in 1 mL PBS-T and frozen at
times. The diluted blood was layered over Ficoll- )20C. Prior to assay, swabs were thawed and
paque premium (GE Healthcare, Rydalmere, Austra- vortexed. Swab eluate was serially diluted twofold
lia) and the tubes were centrifuged at 650 · g for starting with undiluted sample. Plates were then
25 min with the brake off. PBMC at the interface washed 4· in PBS-T and sheep anti-koala IgG
were removed and transferred to a new tube, (1:8000 in PBS-T) added and plates incubated for a
washed twice with HBSS and counted. Cells were further hour at 37C. After further three washes
then suspended at 2 · 106 cells ⁄ mL in PBS supple- (PBS-T), HRP-conjugated rabbit anti-sheep IgG
mented with 1% fetal calf serum. Carboxyfluoresce- (1:1000, Southern Biotech ⁄ Millipore, North Ryde,
in succinimidyl ester (CFSE; Sigma-Aldrich, Castle Australia) was added to wells and plates incubated
Hill, Australia) was added to give a final concentra- for a further hour. After five washes with PBS,
tion of 5 lm and cells were incubated in the dark at 50 lL ⁄ well of TMB substrate (Sigma-Aldrich) in cit-
room temperature for 5 min. Cells were then rate buffer was added and after 10 min, the reactions
washed 3· in HBSS and suspended at 2 · 106 were stopped by adding 50 lL ⁄ well 1 m H2SO4. Opti-
cells ⁄ mL in culture medium (DMEM containing 5% cal density was then read at 450 nm (Bio-Rad, North
foetal calf serum, 2 mm l-glutamine, 100 lg ⁄ mL Ryde, NSW, Australia). Endpoint titers (EPT) were
streptomycin sulfate, 2 lg ⁄ mL gentamycin and calculated as the inverse of the dilution, where
50 lm 2 b-mercaptoethanol). An aliquot of non- absorbance values were equivalent to that of the
CFSE treated cells was retained as control. A total of mean of the no sample (PBS-T) control wells + 2
100 lL ⁄ well (2 · 105 cells) was aliquoted into wells standard deviations.
of 96-well tissue culture plates (In Vitro Technolo-
gies, Noble Park North, Australia). Antigens (MBP-
Preparation of Sheep Anti-Koala IgG
MOMP, NrdB and TC0512) were added to give a
final concentration of each antigen of 50 lg ⁄ mL. Serum from three healthy koalas was pooled and
PHA (10 lg ⁄ mL) was added as a positive control and the IgG fraction isolated using a Protein A ⁄ Protein G
culture medium as a negative control. Plates were column (Zymed Laboratories, South San Francisco,
incubated for 4 days at 37C in 5% CO2. After CA, USA) according to the manufacturer’s instruc-
4 days, cells were washed once in PBS, then resus- tions. The purified IgG fraction was sent to the Insti-
pended in PBS containing 4% paraformaldehyde tute of Medical and Veterinary Science (IMVS,
and stored in the dark at 4C until analysis. Prolifer- Adelaide, Australia) where it was used to immunize
ation of PBMC was determined using a Beckman a sheep. The sheep antiserum was used at a dilution
Coulter flow cytometer (FC500, Gladesville, NSW, of 1:8000 in ELISA assays.
Australia). Proliferation was expressed as the % of
PBMC that had undergone >3 cell divisions.
In Vitro Neutralization Assay
McCoy cells were seeded in 48-well plates and
ELISAs
grown overnight until 70–80% confluent. Plasma
Wells of 96-well ELISA plates (Greiner bio-one med- samples were diluted 1:10, 1:50, 1:250 and 1: 1250.
ium binding; InterPath, West Heidelberg, Australia) Chlamydia muridarum (1000 ifu) or a koala isolate of
were coated overnight at 4C with MBP-MOMP C. pneumoniae24 was added to 0.1 mL of diluted
(0.5 lg ⁄ mL), NrdB (40 lg ⁄ mL) or TC0512 (80 lg ⁄ mL) plasma or cloacal eluate and incubated for 1 hr at
in borate-buffered saline (BBS). Wells were then 37C. Medium was removed from the McCoy cell
washed 3· with PBS containing 0.05% Tween-20 monolayers and replaced by the plasma or cloacal
(PBS-T) then blocked for 1 hr at 37C with 5% skim eluate samples and the plates incubated for 4 hr.
milk in PBS-T. After blocking, samples were serially Samples were removed and replaced with fresh med-
diluted twofold in PBS-T and added to wells, then ium containing 1 lg ⁄ mL cycloheximide. Cells were
incubated for 1 hr at 37C. For TC0512 antibody incubated for further 30 hr and then fixed with
detection, plasma was initially diluted 1:100. For 100% methanol. Inclusions were identified by stain-
detection of MOMP and NrdB-specific antibodies, ing with FITC-labeled anti-chlamydial LPS (CelLabs,
American Journal of Reproductive Immunology 63 (2010) 161–172
164 ª 2010 John Wiley & Sons A/S
DEVELOPING A CHLAMYDIA VACCINE FOR THE KOALA

Brookvale, Australia) according to the manufac- * *


30
turer’s instructions. Iscomatirx

Cells proliferating (%)


Alhydrogel
20 Titre Max Gold
Statistics
All statistical analyses were performed using Graph-
Pad Prism version 5 (GraphPad Software, LaJolla, 10
CA, USA). All data presented are the mean of six
koalas ± SE of the mean. Two-way anova with Bon-
ferroni’s post test with the P value set at <0.05 was 0

0
74
used to analyze the PBMC proliferation and the

10

15

27
ay

ay

ay

ay
D
plasma and cloacal swab antibody samples. Neutral-

D
ization studies were analyzed using unpaired t-test
Fig. 1 Koala peripheral blood mononuclear cells were isolated by
with the P value set at 0.05. centrifugation on Ficoll-paque gradients and then labeled with CFSE as
described in methods. Cells were cultured in vitro for 4 days with
50 lg ⁄ mL of MOMP, CT0512 and NrdB and proliferation determined
Results
by analysis of CFSE fluorescence using a Beckman Coulter FC500. The
Female koalas were immunized with a combination Y-axis shows the percent of PBMC that has undergone ‡3 cell divi-
sions. n = 6 animals per group. Two-way ANOVA was performed with
of three chlamydial antigens combined with one of
Bonferroni post tests. *P < 0.05.
three different adjuvants. No adverse effects were
noted in animals immunized with either the ISC or
Alhydrogel containing vaccine formulations and all background was seen in PBMC from animals immu-
six animals in each group completed the three nized with TiterMax Gold at any time-point. The
immunization schedule. In the group receiving the data show that the strongest cell-mediated response
TiterMax Gold-formulated vaccine, however, three is induced by immunization with ISC and that this is
of the six animals developed abscesses at the vacci- sustained for >200 days after the third immuniza-
nation site following the second immunization. tion. Stimulation with an irrelevant antigen (BSA)
These were obviously painful and impaired mobility resulted in <1% of PBMC proliferating and stimula-
for several days. These animals were treated with tion of PBMC from non-immunized koalas with the
non-steroidal anti-inflammatories for 2–3 days after chlamydial antigen combination did not exceed pro-
the second immunization. For reasons of the adverse liferation levels seen in medium controls (data not
reactions, animals in this group only received two shown).
immunizations on days 0 and 30 and no samples
were collected beyond day 158 of the trial.
Antibody Responses in Plasma
IgG antibodies against the individual antigens,
PBMC Proliferation
TC0512, MOMP and NrdB were measured by ELISA.
The PBMC were stimulated in vitro with the antigen
combination and the proliferation measured by
TC0512 Antibody Response
CFSE dye-dilution assay (Fig. 1). At days 74 and
102, equivalent levels of proliferation were observed At day 74, animals in all three groups had equiva-
in the animals immunized with the ISC or Alhydro- lent levels of TC0512-specific IgG with EPT of 7000–
gel-formulated vaccines. At day 270, only PBMC 10,000 (Fig. 2a). At day 102 plasma from animals
from animals that had received the ISC-formulated immunized with Alhydrogel and TiterMax Gold for-
vaccine showed in vitro proliferation with approxi- mulated vaccines had significantly higher (P < 0.05,
mately 15% of cells proliferating. Proliferation in P < 0.01 respectively) plasma levels of IgG than the
this group was significantly higher (P < 0.05) than animals immunized with ISC. At day 158, there was
in either of the other groups. Low-levels of prolifera- no significant difference in TC0512 IgG levels among
tion in both the Alhydrogel and ISC groups were the three groups, while at day 270, antibody titres
observed at day 158. The reason for this remains >5000 were still present in animals immunized with
unknown. No significant proliferation above media the ISC or Alhydrogel formulations.
American Journal of Reproductive Immunology 63 (2010) 161–172
ª 2010 John Wiley & Sons A/S 165
CAREY ET AL.

(a) † TiterMax group (EPT 80,000) than in the other


15 000 Iscomatirx groups. At day 158, antibody levels in the TiterMax
Alhydrogel group had dropped to levels equivalent to those of
*
Titre Max Gold
the ISC and Alhydrogel groups and at day 270,
10 000
EPT value

animals in the ISC and Alhydrogel groups still had


titres of 20,000. Similar to the MOMP-specific
5000 response, the highest NrdB titres were seen in ani-
mals immunized with TiterMax but longer-lasting
responses were observed in the ISC and Alhydrogel
0 groups (Fig. 2b).
74

0
10

15

27
ay

ay

ay

ay
D

# MOMP Antibody Response


(b)
100 000 # At day 74, animals immunized with TiterMax-
formulated antigens had significantly higher MOMP-
80 000
specific IgG titres (EPT of 80,000) than animals
EPT value

60 000 immunized with either the Alhydrogel or ISC formu-


lations (EPT 40,000–55,000, Fig. 2c; P < 0.001). At
40 000
day 102, MOMP IgG titres were still significantly
20 000 higher in the TiterMax animals, although EPT had
dropped to approximately 60,000, while antibody
0
titres in the Alhydrogel and ISC groups remained at
74

0
10

15

27

an EPT of approximately 40,000 (P < 0.05 and


ay

ay

ay

ay
D

P < 0.001 respectively). A similar pattern was seen


(c) # # # at day 158 while at day 270 MOMP-specific IgG was
* † still detected in plasma of animals immunized with
100 000
ISC or Alhydrogel with titres in the ISC group
80 000 slightly higher, although this was not significant
(Fig. 2c). Overall, the highest MOMP-specific IgG
EPT value

60 000
titres were observed in the animals immunized with
40 000 TiterMax; however, antibody responses were
sustained for longer periods in the ISC and Alhydro-
20 000
gel groups, possibly because these received three
0 immunizations.
74

0
10

15

27
ay

ay

ay

ay
D

Genital Tract Antibody Responses


Fig. 2 Plasma was collected on the indicated days from animals
immunized with MOMP ⁄ CT0512 ⁄ NrdB, combined with ISC, Alhydrogel
Cloacal swabs were eluted in 1 mL of PBS-T and
or TiterMax Gold. Plasma IgG specific for CT0512 (a), NrdB (b) and assayed by ELISA for IgG antibodies against TC0512,
MOMP (c) were determined by ELISA as described in methods. n = 6 MOMP and NrdB.
animals per group. Two-way ANOVA was performed with Bonferroni
post tests. *P < 0.05, !P < 0.01, #P < 0.001.
TC0512 Antibody Response
The highest TC0512-specific IgG responses in swab
eluate at all time-points were observed in the
NrdB Antibody Response
ISC-immunized animals (Fig. 3a). These levels were
At day 74, titres of NrdB-specific IgG of 50,000 significantly higher than in the Alhydrogel and Titer-
were seen in the TiterMax group and this was sig- Max-immunized groups at day 74 (P < 0.05), and
nificantly higher (P < 0.001) than IgG levels in the Alhydrogel group at day 158 (P < 0.05) and
either the ISC or Alhydrogel groups (Fig. 2b). At were continuing to increase at day 270 (Fig. 3a),
day 102, titres were still significantly higher in the reaching EPT of 300–400.
American Journal of Reproductive Immunology 63 (2010) 161–172
166 ª 2010 John Wiley & Sons A/S
DEVELOPING A CHLAMYDIA VACCINE FOR THE KOALA

(a) ISC
MOMP Antibody Response
400 * * #
Alhydrogel
At days 74, 102 and 158, there was no difference in
Titre Max Gold
300 MOMP-specific IgG antibodies in swab eluates
EPT value

among the three experimental groups (Fig. 3c). At


200 day 270, antibody levels in samples from ISC-immu-
nized animals were significantly greater than those
100 in the Alhydrogel-immunized group (P < 0.01;
Fig. 3c). Antibody levels in all groups peaked at day
0 102, but in the ISC-immunized animals, the anti-
2

0
74

body levels were maintained, while in the other two


10

15

27
y

y
Da

Da

Da

Da

groups, levels declined beyond 6 months.


(b)
250

200
In Vitro Neutralization
Two of the three chlamydial antigens in this study
EPT value

150
(MOMP and TC0512) were isolated from the mouse
100 strain C. muridarum while the third antigen (NrdB)
50
was isolated from the human strain C. trachomatis
serovar D. We therefore tested the ability of plasma
0 from immunized animals to neutralize C. muridarum
in vitro and also to neutralize in vitro infectivity of
2

0
74

27
10

15
y
Da

the unrelated C. pneumoniae species LPCoLN isolated


Da

Da

Da

(c) from koalas.24 Plasma from Alhydrogel- and ISC-


400 * * #
immunized animals, at a 1 ⁄ 10 dilution, neutralized
300
in vitro infection of McCoy cells by 80–90% at day
74 and this was sustained up to day 270 (Fig. 4).
EPT value

200 Neutralization by plasma from Alhydrogel-immu-


nized animals was slightly higher at days 74, 102
100 and 158, although this difference was not significant.
Plasma from TiterMax-immunized animals inhibited
0 in vitro C. muridarum infection by >95% at day 74;
2

0
74

10

15

27
y

y
Da

Da

Da

Da

100
Iscomatirx
Fig. 3 Cloacal swabs were collected on the indicated days from
Neutralisation (%)

90 Alhydrogel
animals immunized with MOMP ⁄ CT0512 ⁄ NrdB, combined with either Titre Max Gold
ISC, Alhydrogel or TiterMax Gold. Swabs were eluted in 1 mL of 80
PBS-T and IgG specific for CT0512 (a), NrdB (b) and MOMP (c) were
determined by ELISA as described in methods. n = 6 animals per 70
group. Two-way ANOVA was performed with Bonferroni post tests.
60
*P < 0.05, #P < 0.01.
50
74

0
10

15

27
ay

ay

ay

ay
D

NrdB Antibody Response


Fig. 4 Plasma from animals immunized with MOMP ⁄ CT0512 ⁄ NrdB,
At days 74, 102 and 158, there was no difference in combined with either ISC, Alhydrogel or TiterMax Gold was diluted
1:10 and 0.1 mL incubated with 1000 ifu of Chlamydia muridarum for
NrdB-specific IgG in swab eluates among the three
1 hr as described in methods. Samples were then plated on McCoy
experimental groups (Fig. 3b). On day 270, antibody
cell monolayers and incubated for a further 30 hr. Cells containing
was still evident in animals immunized with ISC and inclusions were identified by staining with FITC-labeled anti-chlamydial
Alhydrogel, but was undetectable in animals immu- LPS. Unpaired t-tests were performed to determine the significance of
nized with TiterMax. differences between groups at the indicated time-points.

American Journal of Reproductive Immunology 63 (2010) 161–172


ª 2010 John Wiley & Sons A/S 167
CAREY ET AL.

however, the percent neutralization steadily declined 80


Iscomatirx
and was approximately 70% at day 158 (Fig. 4).
Thus, at early stages, plasma from TiterMax-immu- Alhydrogel

Neutralisation (%)
60
nized animals showed the greatest neutralizing activ- Titre Max Gold
ity, but this was not sustained throughout the
10 months of the trial compared with that seen in 40
the other two groups. At a 1 ⁄ 250 dilution, plasma
from the Alhydrogel-immunized animals showed the
20
greatest levels of in vitro neutralization (data not
shown) at day 74. The in vitro neutralization level
dropped to around 50% at day 102 and was main- 0

74
tained at 50–60% throughout the remainder of the

0
10

15

27
ay

ay

ay

ay
trial. At this dilution, plasma from ISC-immunized

D
animals neutralized infectivity by approximately
Fig. 5 Plasma from animals immunized with MOMP ⁄ CT0512 ⁄ NrdB,
65% at day 74 and neutralization activity was sus-
combined with either ISC, Alhydrogel or TiterMax Gold was diluted
tained at 35–55% for the rest of the study. Plasma 1:10 and 0.1 mL incubated with 1000 ifu of a koala isolate of Chla-
from TiterMax-immunized animals at a dilution of mydia pneumoniae for 1 hr as described in methods. Samples were
1 ⁄ 250 caused a 40–50% reduction in infectivity at then plated on McCoy cell monolayers and incubated for a further
days 74, 102 and 158. 30 hr. Cells containing inclusions were identified by staining with FITC-
Plasma was also tested for neutralization of C. labeled anti-chlamydial LPS. Unpaired t-tests were performed to deter-
pneumoniae infection, using the LPCoLN strain, mine the significance of differences between groups at the indicated
time-points.
which was isolated from koalas. Plasma (1 ⁄ 10 dilu-
tion) from Alhydrogel-immunized koalas neutral-
ized C. pneumoniae infection by approximately 30% 50% of koalas immunized with TiterMax developed
across the entire time course of the study (Fig. 5). abscesses at the injection site after the second vacci-
Similar levels of C. pneumoniae neutralization were nation. Animals in this group therefore only
seen using plasma from ISC-immunized animals at received two of the three planned immunizations
days 74, 102 and 158, but had dropped to 10–12% and samples were not collected from these animals
neutralization at day 270 (Fig. 5). Plasma from beyond day 158. Strong PBL proliferation was still
TiterMax-immunized animals also neutralized C. present at 270 days in the ISC-immunized koalas
pneumoniae infection by 25–30% at days 74 and with almost 20% of PBL proliferating (>3 cell divi-
102. At a 1 ⁄ 50 dilution, only plasma from the sions) as determined by CFSE dye-dilution assay. At
Alhydrogel-immunized animals contained C. pneu- the early time-points (days 74 and 102), plasma
moniae-neutralizing activity throughout the entire antibody specific for all three antigens was the high-
study. At early time-points, this plasma neutralized est in the animals immunized with TiterMax; how-
30–40% of C. pneumoniae infection and this had ever, no samples were collected beyond day 158
dropped to approximately 15% by day 270 (data from this group because of the adverse reactions to
not shown). Plasma from ISC-immunized animals immunization. Antibody against all 3 antigens was
neutralized infectivity by 15% at day 74, but this sustained throughout the study in animals immu-
had dropped to baseline at day 102 while plasma nized with ISC or Alhydrogel. Overall, NrdB and
from TiterMax-immunized animals did not signifi- MOMP antibody titres in plasma were 4–8 fold
cantly inhibit in vitro infectivity throughout the higher than the TC0512 antibody titres. Plasma-neu-
entire study period. Plasma from immunized tralizing activity for C. muridarum was the highest in
animals, of any group, was not able to neutralize plasma from animals immunized with Alhydrogel
in vitro infectivity of a koala isolate of C. pecorum and ISC with slightly higher neutralizing activity
(data not shown). seen in the Alhydrogel group. Neutralizing activity
for C. pneumoniae was the greatest in plasma from
animals immunized with Alhydrogel. Significantly
Discussion
higher levels of inhibition of C. muridarum infection,
Immunization of koalas with ISC and Alhydrogel compared with C. pneumoniae infection were
containing vaccines were well tolerated, whereas observed, probably because both MOMP and TC0512
American Journal of Reproductive Immunology 63 (2010) 161–172
168 ª 2010 John Wiley & Sons A/S
DEVELOPING A CHLAMYDIA VACCINE FOR THE KOALA

were derived from C. muridarum. It is tempting to Aluminum-based adjuvants have been used in
speculate that MOMP-specific antibody and, to a les- both human and veterinary vaccines for more than
ser extent, TC512-specific antibody were responsible half a century. Veterinary vaccines containing alumi-
for neutralizing C. muridarum infection, while anti- num adjuvants have been developed to target viral,
body against the highly conserved antigen NrdB bacterial and parasite infections (reviewed in26). Alu-
neutralized C. pneumoniae infection. This is perhaps minum adjuvants induce primarily Th2 immunity,
not surprising and confirms the immunodominance driving strong antibody responses, but usually do
of MOMP as the major antigen recognized during not elicit strong cell-mediated immunity. Mecha-
the humoral response to chlamydial infection in nisms of action include formation of an antigen
most species.25 No neutralization of in vitro infection depot, production of particulate antigen to facilitate
of a koala isolate of C. pecorum was observed with targeting to APCs, activation of complement and
any of the plasma samples (data not shown). This stimulation of monocyte ⁄ macrophages.26,27 Recently,
lack of neutralization of C. pecorum does, however, it has been shown that Alum, following intraperito-
suggest that the koala response to NrdB, even neal injection, induces the differentiation of mono-
though this antigen is highly conserved, was not cytes into CD11c+ inflammatory dendritic cells that
neutralizing. The three antigens we chose to evalu- migrate to draining lymph nodes. Stimulation of
ate in the trial had different levels of amino acid these dendritic cells involves activation of the NALP3
conservation across the chlamydial species. NrdB inflammasome, the production of IL-1b and induc-
had the highest level of conservation, estimated tion of the endogenous danger signal uric acid28,29
between 90 and 92% across the seven chlamydial all of which are required for adjuvant action. Plasma
species for which full genome sequences are avail- IgG levels in koalas immunized with Alhydrogel
able. We have previously predicted both human and were less than those in the TiterMax-immunized
mouse T-cell epitope regions in this protein and group at the early time-points and equivalent to
these were 100% conserved across the chlamydial those in the ISC-immunized animals at the later
species. TC0512 (omp85) had the next highest level time-points. Chlamydia-neutralizing activity in
of amino acid conservation, 70–75%, with several plasma from Alhydrogel-immunized animals was
regions of higher level variation observed. As previ- maintained at high levels for the duration of the
ously well known, ompA (MOMP) had the lowest study. Animals immunized with Alhydrogel also
level of conservation, between 60 and 65%, again maintained a local genital IgG response up to at least
with regions of conservation interspersed with day 158, while PBMC proliferation in this group was
regions of high variability. not as great or long lasting as that seen in ISC-
Of the three adjuvants used, TiterMax Gold immunized animals, consistent with the expected
caused severe local reactions at the injection site Th2 predominance of these adjuvants.
in 50% of the animals following the second Immune stimulating complex (ISCOMATRIX) is
immunization. At the day 74 time-point, plasma composed of purified saponin (ISCOPREP) com-
IgG levels against two of the three antigens were plexed with cholesterol and phospholipid (reviewed
the highest in the animals immunized with Titer- in.30,31 ISCOMATRIX induces both strong antibody
Max Gold, whereas PBMC proliferation was the and cell-mediated responses, including cytotoxic
lowest in this group at all time-points. This is T-lymphocyte (CTL) responses30,32,33 in most species
consistent with the manufacturer’s claims that tested. ISCOMATRIX has also been shown to reduce
TiterMax adjuvants elicit antibody responses supe- the antigen dose required to elicit an antibody
rior to Freund’s complete adjuvant (CFA) in most response34 and can be delivered by the intranasal
species. TiterMax Gold is a water-in-oil emulsion route, where it is able to elicit significant mucosal
comprising the block co-polymer CRL-8300, the responses (reviewed in35). The mechanism of action
metabolizable oil squalene and a microparticle seems to depend on the induction of multiple cyto-
stabilizer. Whilst TiterMax adjuvants are claimed to kines including IL-1, IL-2, IL-4, IL-5, IL-6, IL-10,
cause less severe side effects than CFA in many IL-8, IL-12 and IFN-c, which occurs in the draining
species, the development of abscesses at the injec- lymph nodes.31 Up-regulation of CD86 and MHC
tion site that were obviously painful and impaired class II on APC may also be important for adjuvant
mobility of animals for several days make this function.36 As ISCOMATRIX is cleared rapidly from
adjuvant unacceptable for use in koalas. the injection site, formation of an antigen depot is
American Journal of Reproductive Immunology 63 (2010) 161–172
ª 2010 John Wiley & Sons A/S 169
CAREY ET AL.

probably not important for adjuvant function.30 In are currently being used in ongoing immunization
our studies, animals immunized with ISC mounted and challenge studies. However, the induction of
robust PBMC proliferative responses that were both long-lived systemic and mucosal antibody and cell-
superior to those induced by Alhydrogel and Titer- mediated immunity by immunization with a safe
Max and were maintained for the entire length of ISC-adjuvanted vaccine, that can be boosted in cap-
the study (>270 days). The longevity of this response tive animals by repeat vaccination and in wild ani-
is encouraging as healthy koalas have a life span of mals could be boosted by natural challenge, is
10–12 years in the wild. Plasma IgG levels in ISC extremely encouraging and provides evidence that
immunized animals were equivalent in magnitude protective immunity against chlamydial infection in
and duration to those in the Alhydrogel-immunized the koala can be induced by vaccination.
group, while genital IgG responses were the highest
in ISC-immunized koalas. This combination of a
Acknowledgments
robust, long-lived cell-mediated response, together
with a sustained systemic and mucosal antibody We thank Dr. John Walker VMRD, Pfizer, Australia
response, suggests that immunization with ISC for provision of the Immunostimulating Complex
should elicit protective immunity against chlamydial (ISC) adjuvant, Ms Candice Mitchell for provision of
infection.37 Studies are currently underway to the C. pneumoniae LPCoLN strain, Dr. Adam Polking-
develop assays to determine if the PBMC prolifera- horne for assistance with chlamydial gene alignment
tive response is also characterized by secretion of studies and Dr. Kelly Cunningham for review of the
IFN-c, a cytokine essential for protection against manuscript. We also gratefully acknowledge the gen-
chlamydial infection. Future studies will also investi- erous financial support provided by Lone Pine Koala
gate intranasal immunization of koalas using ISC Sanctuary.
to determine if protective immunity is elicited by
this route.
References
Despite the fact that all animals were immunized
subcutaneously, antibody against all 3 antigens was 1 McColl KA, Martin RW, Gleeson LJ, Handasyde KA,
detected in cloacal swab eluates. These antibodies Lee AK: Chlamydia infection and infertility in the
were sustained for 270 days in animals immunized female koala (Phascolarctos cinereus). Vet Rec 1984;
with Alhydrogel and ISC. Swab antibodies were of 115:655.
the IgG class, consistent with findings in humans 2 Canfield PJ, Love DN, Mearns G, Farram E:
and rodents that serum-derived IgG plays a major Chlamydial infection in a colony of captive koalas.
protective role in the female reproductive tract.38 Aust Vet J 1991; 68:167–169.
We are currently developing an IgA-specific antise- 3 Jackson M, White N, Giffard P, Timms P:
Epizootiology of Chlamydia infections in two
rum to determine if IgA is also present in the koala
free-range koala populations. Vet Microbiol 1999;
reproductive tract.
65:255–264.
In summary, our studies have shown that subcu-
4 Markey B, Wan C, Hanger J, Phillips C, Timms P: Use
taneous immunization of koalas with a multi-sub-
of quantitative real-time PCR to monitor the shedding
unit chlamydial vaccine, adjuvanted with either ISC
and treatment of chlamydiae in the koala
or Alhydrogel, is well tolerated and elicits both (Phascolarctos cinereus). Vet Microbiol 2007;
humoral and cell-mediated immune responses that 120:334–342.
last >270 days. Plasma antibodies from both groups 5 Girjes AA, Hugall AF, Timms P, Lavin MF: Two
were able to neutralize chlamydial infection in vitro. distinct forms of Chlamydia psittaci associated with
PBMC proliferative responses and genital IgG levels disease and infertility in Phascolarctos cinereus
were the highest in animals immunized with ISC (koala). Infect Immun 1988; 56:1897–1900.
suggesting that this adjuvant is best suited for induc- 6 Brown AS, Grice RG: Isolation of Chlamydia psittaci
tion of protective immunity against chlamydial infec- from koalas (Phascolarctos cinereus). Aust Vet J 1984;
tions in the koala. The studies also highlight the 61:413.
need to use antigens from Chlamydia species that 7 Brown AS, Girjes AA, Lavin MF, Timms P, Woolcock
infect koalas. We now have a collection of C. pecorum JB: Chlamydial disease in koalas. Aust Vet J 1987;
strains isolated from koalas in South-East Queens- 64:346–350.
land. Antigens have been prepared from these and
American Journal of Reproductive Immunology 63 (2010) 161–172
170 ª 2010 John Wiley & Sons A/S
DEVELOPING A CHLAMYDIA VACCINE FOR THE KOALA

8 Osawa R, Carrick FN: Use of a dietary supplement in Chlamydia muridarum genital infection. Vaccine 2007;
koalas during systemic antibiotic treatment of 25:2643–2655.
chlamydial infection. Aust Vet J 1990; 67:305–307. 21 Wilkinson R, Kotlarski I, Barton M: Further
9 Hogan RJ, Mathews SA, Mukhopadhyay S, characterisation of the immune response of the koala.
Summersgill JT, Timms P: Chlamydial persistence: Vet Immunol Immunopathol 1994; 40:325–339.
beyond the biphasic paradigm. Infect Immun 2004; 22 Wilkinson R, Kotlarski I, Barton M, Phillips P:
72:1843–1855. Isolation of koala lymphoid cells and their in vitro
10 Wilkinson R, Allanson M, Kolega V, Lawrence D, responses to mitogens. Vet Immunol Immunopathol
Neville S: Purification and initial characterisation of 1992; 31:21–33.
koala immunoglobulins. Vet Immunol Immunopathol 23 Su H, Raymond L, Rockey DD, Fischer E, Hackstadt T,
1991; 29:189–195. Caldwell HD: A recombinant Chlamydia trachomatis
11 Wilkinson R, Kotlarski I, Barton M: Koala lymphoid major outer membrane protein binds to heparan
cells: analysis of antigen-specific responses. Vet sulfate receptors on epithelial cells. Proc Natl Acad Sci
Immunol Immunopathol 1992; 33:237–247. USA 1996; 93:11143–11148.
12 Kitchener AL, Kay DJ, Walters B, Menkhorst P, 24 Mitchell CM, Mathews SA, Theodoropoulos C, Timms
McCartney CA, Buist JA, Mate KE, Rodger JC: The P: In vitro characterisation of koala Chlamydia
immune response and fertility of koalas (Phascolarctos pneumoniae: morphology, inclusion development and
cinereus) immunised with porcine zonae pellucidae doubling time. Vet Microbiol 2009; 136:91–99.
or recombinant brushtail possum ZP3 protein. 25 Caldwell HD, Schachter J: Antigenic analysis of the
J Reprod Immunol 2009; 82:40–47. major outer membrane protein of Chlamydia spp.
13 Warren HS, Vogel FR, Chedid LA: Current status of Infect Immun 1982; 35:1024–1031.
immunological adjuvants. Annu Rev Immunol 1986; 26 Lindblad EB: Aluminium compounds for use in
4:369–388. vaccines. Immunol Cell Biol 2004; 82:497–505.
14 Grayston JT: Immunisation against trachoma. Pan Am 27 Hunter RL: Overview of vaccine adjuvants: present
Health Organ Sci Publ 1965; 147:549. and future. Vaccine 2002; 3:S7–S12.
15 Grayston JT, Wang SP: The potential for vaccine 28 Kool M, Petrilli V, De Smedt T, Rolaz A, Hammad H,
against infection of the genital tract with Chlamydia van Nimwegen M, Bergen IM, Castillo R, Lambrecht
trachomatis. [Review] [19 refs]. Sex Transm Dis 1978; BN, Tschopp J: Cutting edge: alum adjuvant
5:73–77. stimulates inflammatory dendritic cells through
16 Berry LJ, Hickey DK, Skelding KA, Bao S, Rendina activation of the NALP3 inflammasome. J Immunol
AM, Hansbro PM, Gockel CM, Beagley KW: 2008; 181:3755–3759.
Transcutaneous immunization with combined cholera 29 Kool M, Soullie T, van Nimwegen M, Willart MA,
toxin and CpG adjuvant protects against Chlamydia Muskens F, Jung S, Hoogsteden HC, Hammad H,
muridarum genital tract infection. Infect Immun 2004; Lambrecht BN: Alum adjuvant boosts adaptive
72:1019–1028. immunity by inducing uric acid and activating
17 Skelding KA, Hickey DK, Horvat JC, Bao S, Roberts inflammatory dendritic cells. J Exp Med 2008;
KG, Finnie JM, Hansbro PM, Beagley KW: 205:869–882.
Comparison of intranasal and transcutaneous 30 Pearse MJ, Drane D: ISCOMATRIX adjuvant: a potent
immunization for induction of protective immunity inducer of humoral and cellular immune responses.
against Chlamydia muridarum respiratory tract Vaccine 2004; 22:2391–2395.
infection. Vaccine 2006; 24:355–366. 31 Pearse MJ, Drane D: ISCOMATRIX adjuvant for
18 Jackson M, Giffard P, Timms P: Outer membrane antigen delivery. Adv Drug Deliv Rev 2005; 57:465–474.
protein A gene sequencing demonstrates the 32 Sjolander S, Drane D, Davis R, Beezum L, Pearse M,
polyphylogenetic nature of koala Chlamydia pecorum Cox J: Intranasal immunisation with influenza-
isolates. Syst Appl Microbiol 1997; 20:187–200. ISCOM induces strong mucosal as well as systemic
19 Barker CJ, Beagley KW, Hafner LM, Timms P: In antibody and cytotoxic T-lymphocyte responses.
silico identification and in vivo analysis of a novel Vaccine 2001; 19:4072–4080.
T-cell antigen from Chlamydia, NrdB. Vaccine 2008; 33 Malliaros J, Quinn C, Arnold FH, Pearse MJ, Drane
26:1285–1296. DP, Stewart TJ, Macfarlan RI: Association of antigens
20 McNeilly CL, Beagley KW, Moore RJ, Haring V, to ISCOMATRIX adjuvant using metal chelation
Timms P, Hafner LM: Expression library leads to improved CTL responses. Vaccine 2004;
immunization confers partial protection against 22:3968–3975.

American Journal of Reproductive Immunology 63 (2010) 161–172


ª 2010 John Wiley & Sons A/S 171
CAREY ET AL.

34 Boyle J, Eastman D, Millar C, Camuglia S, Cox J, of iscom-borne antigens and presentation under MHC
Pearse M, Good J, Drane D: The utility of class I or class II restriction. Cell Immunol 1998;
ISCOMATRIX adjuvant for dose reduction of antigen 185:30–38.
for vaccines requiring antibody responses. Vaccine 37 Morrison RP, Caldwell HD: Immunity to murine
2007; 25:2541–2544. chlamydial genital infection. Infect Immun 2002;
35 Hu KF, Lovgren-Bengtsson K, Morein B: 70:2741–2751.
Immunostimulating complexes (ISCOMs) for nasal 38 Mestecky J, Moldoveanu Z, Russell MW:
vaccination. Adv Drug Deliv Rev 2001; 51:149–159. Immunologic uniqueness of the genital tract:
36 Villacres MC, Behboudi S, Nikkila T, challenge for vaccine development. Am J Reprod
Lovgren-Bengtsson K, Morein B: Internalization Immunol 2005; 53:208–214.

American Journal of Reproductive Immunology 63 (2010) 161–172


172 ª 2010 John Wiley & Sons A/S

You might also like