ArticlePDF Available

Successful vaccination of immune suppressed recipients using Listeria vector HIV-1 vaccines in helminth infected mice

Authors:

Abstract and Figures

Vaccines for HIV, malaria and TB remain high priorities, especially for sub-Saharan populations. The question is: will vaccines currently in development for these diseases function in populations that have a high prevalence of helminth infection? Infection with helminth parasites causes immune suppression and a CD4+ Th2 skewing of the immune system, thereby impairing Th1-type vaccine efficacy. In this study, we conduct HIV vaccine trials in mice with and without chronic helminth infection to mimic the human vaccine recipient populations in Sub-Saharan Africa and other helminth parasite endemic regions of the world, as there is large overlap in global prevalence for HIV and helminth infection. Here, we demonstrate that Listeria monocytogenes functions as a vaccine vector to drive robust and functional HIV-specific cellular immune responses, irrespective of chronic helminth infection. This observation represents a significant advance in the field of vaccine research and underscores the concept that vaccines in the developmental pipeline should be effective in the target populations.
Vaccination strategy in a murine model of helminth infection. (A) To verify Th2 biasing of helminth infected mice, splenocytes from uninfected mice (open bars) or schistosome infected mice at 7 wpi (gray bars) or 18 wpi (solid bars) were stimulated for with SEA or conA ex vivo and supernatants assayed for IFN and IL-4 expression by ELISA. Results from two independent experiments are pooled and plotted as the mean + SEM of the IFN:IL-4 ratio. (B) In order to assess vaccine-specific responses in helminth infected mice, 6-8-week old female BALB/c mice were infected with Schistosoma mansoni or left naïve and helminth infection was verified by ELISA at 10 weeks post infection (wpi). Twelve wpi, age-matched mice were primed with Listeria vector HIV-1 vaccine (Lm-gag), control Listeria vector HPV vaccine (Lm-E7) or left unvaccinated. Mice were boosted 2 weeks after the prime in an identical manner. Vaccine responses were evaluated two or more weeks post last vaccination (wplv). (C-F) Splenocytes were harvested at 16 weeks post Schistosome infection from Listeria gag vaccinated, age matched mice that were uninfected (open bars) or schistosome infected (solid bars). Splenocytes were stimulated with SEA or conA ex vivo and supernatants assayed for IFN, IL-4 and IL-10 expression by ELISA. Pooled results from 3 independent experiments are plotted as the mean + SEM of the IFN:IL-4 ratio (C) and absolute IFN (D), IL-4 (E) and IL-10 values (F) (n = 16-19). Responding groups were analyzed by ANOVA with Bonferroni post test. *p < 0.05, ***p < 0.001, ****p < 0.0001.
… 
Content may be subject to copyright.
Vaccine
31 (2013) 2050–
2056
Contents
lists
available
at
SciVerse
ScienceDirect
Vaccine
j
ourna
l
ho
me
pag
e:
www.elsevier.com/locate/vaccine
Successful
vaccination
of
immune
suppressed
recipients
using
Listeria
vector
HIV-1
vaccines
in
helminth
infected
mice
Lisa
M.
Shollenbergera,
Cac
Buia,
Yvonne
Patersonb,
Kelsey
Allena,
Donald
Harna,
aDepartment
of
Infectious
Diseases
and
Center
for
Tropical
and
Emerging
Global
Diseases,
University
of
Georgia,
501
DW
Brooks
Drive,
Athens,
GA
30602-7387,
USA
bDepartment
of
Microbiology,
University
of
Pennsylvania,
3610
Hamilton
Walk,
Philadelphia,
PA
19104-6076,
USA
a
r
t
i
c
l
e
i
n
f
o
Article
history:
Received
5
September
2012
Received
in
revised
form
22
January
2013
Accepted
19
February
2013
Available online 5 March 2013
Keywords:
Chronic
helminth
infection
Endemic
schistosomiasis
Vaccine
efficacy
HIV
vaccine
Listeria
monocytogenes
a
b
s
t
r
a
c
t
Vaccines
for
HIV,
malaria
and
TB
remain
high
priorities,
especially
for
sub-Saharan
populations.
The
question
is:
will
vaccines
currently
in
development
for
these
diseases
function
in
populations
that
have
a
high
prevalence
of
helminth
infection?
Infection
with
helminth
parasites
causes
immune
suppression
and
a
CD4+
Th2
skewing
of
the
immune
system,
thereby
impairing
Th1-type
vaccine
efficacy.
In
this
study,
we
conduct
HIV
vaccine
trials
in
mice
with
and
without
chronic
helminth
infection
to
mimic
the
human
vaccine
recipient
populations
in
Sub-Saharan
Africa
and
other
helminth
parasite
endemic
regions
of
the
world,
as
there
is
large
overlap
in
global
prevalence
for
HIV
and
helminth
infection.
Here,
we
demonstrate
that
Listeria
monocytogenes
functions
as
a
vaccine
vector
to
drive
robust
and
functional
HIV-specific
cellular
immune
responses,
irrespective
of
chronic
helminth
infection.
This
observation
represents
a
significant
advance
in
the
field
of
vaccine
research
and
underscores
the
concept
that
vaccines
in
the
developmental
pipeline
should
be
effective
in
the
target
populations.
© 2013 Elsevier Ltd. All rights reserved.
1.
Introduction
Malaria,
tuberculosis
and
HIV-1
remain
tremendous
disease
burdens
for
much
of
the
world’s
population
and
vaccines
for
these
diseases
are
desperately
needed.
Recently
there
has
been
modest
success
in
HIV-1
vaccine
clinical
trials,
and
as
the
greatest
burden
of
HIV
is
found
in
Sub-Saharan
Africa,
it
is
therefore
likely
that
future
HIV-1
vaccine
trials
will
be
performed
in
those
countries
[1,2].
Globally,
in
2010,
34
million
people
were
living
with
HIV-1/AIDS,
with
68%
of
these
infected
individuals
residing
in
Sub-Saharan
Africa.
Prevalence
of
HIV-1
exceeds
20%
in
some
southern
African
countries
[3].
In
addition,
with
pronounced
geographic
coincidence,
a
large
percentage
of
these
populations
are
infected
with
one
or
more
parasitic
worms,
with
prevalence
rates
surpassing
50%
in
some
countries
[4].
Chronic
helminthiasis
causes
systemic
Th2
bias-
ing
and
IL-10
mediated
immune
suppression
[5–8].
Therefore
it
is
likely
that
helminth
infected
vaccine
recipients
will
have
compro-
mised
vaccine
specific,
cell
mediated
immune
responses
[4,6–11].
The
current
trend
of
HIV
vaccine
design
places
more
emphasis
on
eliciting
cytotoxic
T
lymphocytes
(CTL),
rather
than
humoral
Abbreviation:
SEA,
soluble
egg
antigens
from
Schistosoma
mansoni.
Corresponding
author.
Tel.:
+1
706
542
4569;
fax:
+1
706
542
5771.
E-mail
addresses:
lisaluvsciens@gmail.com
(L.M.
Shollenberger),
ctbui88@uga.edu
(C.
Bui),
yvonne@mail.med.upenn.edu
(Y.
Paterson),
kwallen10@gmail.com (K.
Allen),
dharn@uga.edu
(D.
Harn).
responses
even
though
both
arms
of
the
immune
response
are
likely
needed
for
an
effective
vaccination
strategy
[12].
In
this
regard,
sev-
eral
studies
have
suggested
that
the
effectiveness
of
HIV-1
vaccines
designed
to
induce
cell
mediated
immunity
will
be
based
on
both
the
quantity
and
quality
of
the
virus-specific
CD4+
and
CD8+
T
cell
responses
[1,13,14].
Therefore,
tremendous
effort
has
been
devoted
to
studies
on
the
design
of
prime-boost
vaccines
that
induce
strong
and
durable
HIV-1
specific
CD4+
and
CD8+
T
cell
responses
in
naïve
recipients
[12,13].
Because
of
the
negative
impact
of
helminth
infection
on
vaccine
efficacy,
we
believe
it
is
critical
that
can-
didate
vaccines
be
evaluated
in
helminth
infected
recipients
to
insure
they
are
capable
of
driving
the
desired
vaccine
specific
responses.
For
example,
efficacy
of
vaccines
for
tetanus
and
tuberculosis
were
shown
to
be
compromised
in
helminth
infected
recipients
liv-
ing
in
helminth
endemic
sites
[15–17].
Similarly,
the
only
study
to
examine
HIV-specific
CTL
responses
in
the
context
of
systemic
Th2
biasing
due
to
chronic
helminth
infection,
demonstrated
that
mice
chronically
infected
with
the
helminth
parasite
Schistosoma
man-
soni
were
not
able
to
mount
significant
HIV-1
vaccine-specific
CTL
responses
post-vaccination,
even
when
the
vaccine
was
enhanced
[18].
This
observation,
taken
together
with
other
studies
exam-
ining
virus-specific
immune
responses
in
helminth
infected
mice
suggests
that
helminth
infection
will
pose
a
significant
problem
for
the
development
of
HIV-1
vaccines
designed
to
induce
viral-
specific
Th1
type
CD4+
and
cytotoxic
CD8+
T
cell
responses.
A
goal
of
our
research
is
to
find
vaccine
vectors
able
to
drive
significant
0264-410X/$
see
front
matter ©
2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.vaccine.2013.02.037
L.M.
Shollenberger
et
al.
/
Vaccine
31 (2013) 2050–
2056 2051
HIV-specific
immune
responses
in
helminth
infected
recipients,
despite
the
immune
status
of
the
individual.
Having
effective
vac-
cine
vectors
will
facilitate
development
of
an
HIV-1
vaccine
that
functions
in
a
real
world,
helminth
endemic
setting.
To
address
the
deleterious
effect
of
immune
system
biasing
and
suppression
on
vaccine
efficacy,
de-worming
strategies
have
been
employed
to
increase
vaccine
efficacy,
but
treatment
must
be
repeated,
often
yearly,
as
re-infection
is
both
common
and
frequent
[19].
The
approach
presented
here
is
to
utilize
a
live
vaccine
vec-
tor,
Listeria
monocytogenes,
to
induce
rigorous
T
cell-based
HIV-1
vaccine-specific
immune
responses
in
helminth
infected
recipients
as
proof
of
concept,
as
viral
and
bacterial
vectors
are
known
to
induce
substantial
CTL
responses
[20].
Further,
Listeria
has
been
shown
to
overcome
pre-existing
Th2
biasing
and
reverse
vaccine
specific
immune
responses
to
third
party
antigens
from
Th2
to
Th1
type
[21].
Highlighting
both
the
promising
results
achievable
using
Listeria
as
well
as
the
continuing
acceptance
of
this
pathogen
as
a
vaccine
vector,
the
US
Department
of
Defense
has
recently
solicited
research
proposals
using
L.
monocytogenes
as
a
vaccine
vector.
The
ability
of
Listeria
to
strongly
augment
CD4+
and
CD8+
anti-viral
T
cell
responses
makes
it
an
ideal
candidate
as
a
vaccine
vector
for
HIV;
Listeria
produces
strong
mucosal
and
systemic
innate
and
adaptive
immune
responses
[22–24]
and
can
be
delivered
orally
to
target
the
mucosa,
a
major
immune
compartment
of
the
host
where
HIV
infection
is
initially
established
[22,24–26].
Listeria
lives
within
phagosomal
and
intracytoplasmic
com-
partments,
facilitating
delivery
of
HIV
antigens
to
exogenous
and
endogenous
antigen
processing
and
presentation
pathways
as
well
as
to
the
MHCII
pathway
via
cross
presentation
[27–29].
Unlike
ade-
novirus
or
vaccinia
vectors,
where
pre-existing
immunity
hampers
priming
of
immune
responses
and
repeated
delivery
of
exogenous
antigens
[30–33],
pre-existing
immunity
to
Listeria
does
not
neg-
atively
impact
the
ability
of
Listeria
to
be
administered
repeatedly
with
no
diminishment
in
the
response
to
HIV
antigens
[34–37].
Listeria
vaccines
induce
robust
immune
responses
in
macaques
and
partially
protect
against
challenge
[24,38].
Listeria
has
also
been
used
successfully
as
a
vector
to
induce
protective
immu-
nity
to
other
pathogens
including
influenza,
Francisella
tularensis,
herpes
simplex
virus
type
1,
Leishmania
major
and
human
papil-
lomavirus
[39–43],
resulting
in
induction
of
systemic
immune
responses.
In
the
event
immune
compromised
patients
were
to
exhibit
overgrowth
of
attenuated
Listeria
vectors,
these
bacteria
remain
antibiotic
susceptible.
The
overgrowth
scenario
has
not
been
observed
in
ongoing
therapeutic
cancer
vaccine
clinical
tri-
als.
Therefore,
we
believe
Listeria
is
an
ideal
vaccine
vector,
able
to
repeatedly
boost
the
CD4+
and
CD8+
T
cell
responses
to
HIV
antigens.
2.
Materials
and
methods
2.1.
Biological
reagents
The
HIV
vaccine,
an
attenuated
strain
of
L.
monocytogenes
expressing
the
HIV-1
IIIB
gag
protein
(Lm-gag)
[44],
and
the
control
strain,
which
expresses
the
E7
oncoprotein
of
the
human
papillo-
mavirus
16
(Lm-E7)
[45],
were
grown
in
BHI
supplemented
with
streptomycin.
S.
mansoni
(PR
strain)
infected
Biomphalaria
glabrata
snails
were
provided
by
the
NIAID
Schistosome
Reagent
Program.
Infectious
cercariae
were
obtained
by
exposing
infected
snails
to
direct
light.
Female
BALB/c
mice
at
5–7
weeks
old
were
purchased
from
Harlan
Laboratories,
housed
in
specific
pathogen-free
condi-
tions
and
allowed
to
acclimate
for
1
week
prior
to
manipulation.
All
animal
work
was
performed
in
accordance
with
institutional
policy
and
approved
by
the
institutional
animal
care
and
use
committee.
2.2.
Helminth
infection
of
mice
Six
to
eight-week
old
female
BALB/c
mice
were
infected
by
intraperitoneal
injection
of
35–50
infectious
cercariae
of
S.
mansoni.
Helminth
infection
was
verified
by
indirect
ELISA
for
the
presence
of
anti-egg
and/or
anti-worm
antibodies
in
sera
collected
10
weeks
post
infection
(wpi).
2.3.
Vaccination
of
mice
Age-matched
female
BALB/c
mice
were
primed
i.p.
with
0.2
LD50
(or
0.1
LD50,
as
indicated)
Lm-gag
vaccine
(106CFU),
control
Lm-
E7
vaccine
(matched
CFU
dose)
or
left
unvaccinated.
Mice
were
boosted
2
weeks
after
the
prime
in
an
identical
manner.
2.4.
ELISA
Two
weeks
post
last
vaccination
(wplv),
splenocytes
were
har-
vested
and
plated
at
1.5
million
cells
per
well
in
48-well
plates
in
complete
media
(RPMI-1640)
supplemented
with
FBS,
penicillin,
streptomycin,
amphotericin
B
and
-mercaptoethanol.
Cells
were
stimulated
with
25
g/ml
Schistosome
soluble
egg
antigen
(SEA)
or
1
g/ml
concanavalin
A
(conA)
for
72
h,
supernatants
harvested
then
analyzed
for
levels
of
IFN,
IL-4
and
IL-10
by
ELISA
(Becton
Dickinson).
2.5.
ELISpot
Two
wplv,
splenocytes
were
harvested
and
plated
at
300
K
and
150
K
cells
per
well
in
IFN
ELISpot
plates
(BD).
The
spleno-
cytes
were
re-stimulated
in
the
presence
of
media,
20
M
specific
CTL
peptide
(H2-Kd-restricted,
AMQMLKETI
from
HIV-1
IIIB
gag
protein),
20
M
irrelevant
peptide
(H2-Kd-restricted,
TYQRTRALV
from
influenza
A/PR/8/34
nucleoprotein),
20
M
specific
helper
peptide
(Class
II-restricted,
NPPIPVGEIYKRWIILGLNK
from
HIV-1
IIIB
gag
protein)
or
1
g/ml
conA
(as
a
positive
control).
Pep-
tides
were
synthesized
by
Biosynthesis,
Inc.
at
greater
than
95%
purity
and
reconstituted
in
DMSO
prior
to
dilution
in
media.
After
incubation
for
20
h,
ELISpots
were
performed
according
to
man-
ufacturer’s
instructions
and
enumerated
using
an
Immunospot
analyzer
(C.T.L.).
2.6.
Flow
cytometry
Splenocytes
were
incubated
in
the
presence
of
PMA,
Iono-
mycin
and
GolgiStop
(monensin)
for
6
h.
Surface
markers
were
stained
with
-CD8,
-CD4,
-CD25
antibodies
(BD)
and/or
HIV
gag
tetramer
(H2-Kd+AMQMLKETI,
Beckman
Coulter)
then
cells
were
fixed.
After
membrane
permeabilization,
intracellular
mark-
ers
were
stained
using
-IFN,
-IL-4
(BD)
or
-FoxP3
antibodies
(eBiosciences)
and
analyzed
by
flow
cytometry.
To
detect
HIV-
specific
CTLs
at
14
wplv,
splenocytes
were
stained
with
an
HIV
gag
tetramer
and
an
-CD8
antibody
(BD).
To
evaluate
CD8+
central
and
effector
memory
compartments
at
14
wplv,
splenocytes
were
stained
with
-CD8,
-CCR7
and
-CD62L
antibodies
(BD).
Live
cells
(as
indicated
by
using
a
LIVE/DEAD
fixable
dye,
Invitrogen)
were
acquired
and
analyzed
using
an
LSRII
flow
cytometer
running
FACS
Diva
(BD).
2.7.
In
vivo
cytotoxic
T
lymphocyte
(CTL)
assay
To
prepare
target
cells,
splenocytes
from
naïve,
syngeneic
mice
were
fluorescently
labeled
with
10
M
green
(Vybrant
CFDA
SE
Cell
Tracer
Kit)
or
20
M
violet
(CellTrace
Violet
Cell
Proliferation
Kit)
fluorescent
dye,
according
to
manufacturer’s
instructions
(Invitro-
gen).
Cells
were
then
pulsed
for
2
h
with
20
M
irrelevant
or
specific
2052 L.M.
Shollenberger
et
al.
/
Vaccine
31 (2013) 2050–
2056
CTL
peptide,
respectively.
Targets
were
mixed
and
twelve
million
cells
were
injected
intravenously
per
vaccinated
animal.
After
20
h,
splenocytes
were
collected
and
analyzed
by
flow
cytometry
for
target
recovery.
3.
Results
and
discussion
3.1.
Vaccination
strategy
in
a
murine
model
of
helminth
infection
To
address
the
impact
of
Th2
biasing
on
vaccine
efficacy,
we
test
vaccines
in
animals
chronically
infected
with
helminth
par-
asites.
Cytokine
biasing
caused
by
chronic
schistosomiasis
was
assessed
in
mice
in
the
absence
of
vaccination
with
Listeria
vectors
(Fig.
1A).
For
unvaccinated
mice,
no
cytokines
were
detected
after
media
stimulation
or
upon
SEA
stimulation,
as
expected,
since
these
mice
had
no
prior
exposure
to
schistosomal
antigens.
A
decrease
in
the
IFN:IL-4
ratio
correlates
with
an
increase
in
Th2
bias-
ing,
which
is
shown
upon
conA
stimulation
in
a
time-dependent
manner.
Th2
biasing
increases
with
duration
of
infection,
shown
by
a
97.2%
decrease
in
the
IFN:IL-4
ratio
of
conA
stimulated
cells
(between
uninfected
to
18
wpi).
At
18
wpi,
the
cytokine
ratio
is
significantly
different
from
both
uninfected
mice
and
mice
at
7
wpi,
showing
mice
become
increasingly
Th2
biased
with
time.
In
mice
and
humans,
cytokine
switching
from
Th1
to
Th2
begins
approximately
6–7
wpi,
which
is
coincident
with
egg
deposition
by
adult
worms.
Previous
studies
have
used
animals
at
8–10
wpi
as
a
model
of
helminth
infection.
However,
in
our
studies,
vacci-
nation
is
postponed
until
12
weeks
after
infection
with
S.
mansoni
to
ensure
the
mice
have
fully
developed
Th2
biasing
(Fig.
1B)
and
to
eliminate
ambiguity
regarding
its
status
as
a
chronic
infection.
The
vaccine
is
then
administered
intraperitoneally
in
a
prime-boost
strategy
with
2
weeks
between
vaccinations
and
2
weeks
between
the
last
vaccination
and
the
analysis
of
vaccine-specific
immune
responses.
To
examine
whether
Listeria
vaccinated
mice
with
chronic
helminth
infection
would
also
exhibit
Th2
biasing,
splenocytes
were
harvested
from
age-matched,
Listeria
vaccinated
mice
with
or
without
schistosome
infection
at
16
weeks,
then
stimulated
ex
vivo
for
72
h
with
SEA
(antigen-specific
stimulation)
or
conA
(non-specific
stimulation)
and
cytokine
levels
measured
by
ELISA
(Fig.
1C–F).
At
16
wpi,
Th2
biasing
of
Schistosome
infected
mice
increases
relative
to
uninfected
when
both
groups
have
been
vac-
cinated
with
Listeria
vectors,
shown
by
a
78%
decrease
in
the
IFN:IL-4
ratio
upon
conA
stimulation
(Fig.
1C).
As
shown
in
Fig.
1D,
IFN
is
produced
by
cells
from
helminth
infected
mice
in
response
to
both
SEA
and
conA,
however
is
reduced
compared
to
cells
from
uninfected
mice
stimulated
with
conA.
IL-4
and
IL-10
levels
are
markedly
increased
in
splenocytes
from
Schistosome
infected
mice
relative
to
uninfected
mice
(Fig.
1E–F).
These
results
indicate
the
helminth
infected
mice
are
immune
suppressed
in
addition
to
Th2
biased.
Taken
together,
these
results
indicate
helminth
infected,
Listeria
HIV-1
vaccinated
mice
are
Th2
biased
and
immune
sup-
pressed,
as
indicated
by
a
reduction
in
the
IFN:IL-4
ratio
and
increases
in
IL-10
levels.
Th2
biasing
is
coincident
with
schistosome
infection,
as
shown
by
a
reduction
in
IFN:IL-4
ratio
upon
conA
stimulation,
whether
the
mice
are
naïve
(Fig.
1A)
or
Lm-gag
vaccinated
(Fig.
1C–E).
Fur-
ther,
helminth
infected,
Listeria
vaccinated
mice
are
also
immune
suppressed,
shown
by
relative
increases
in
IL-10
levels
com-
pared
to
uninfected
mice
(Fig.
1F).
Therefore,
the
schistosome
infection
employed
here
is
a
good
model
for
testing
vaccine
effi-
cacy
in
the
context
of
chronic
helminth
infection,
as
it
exhibits
characteristics
similar
to
real
world
chronic
helminth
infec-
tion.
3.2.
Listeria
vector
vaccines
induce
functional
HIV-specific
T
cell
responses
despite
chronic
helminth
infection
Two
wplv,
gag-specific
responses
were
assessed
by
IFN
ELISpot
assay
by
re-stimulation
of
splenocytes
in
the
presence
of
specific
CTL
peptide
or
specific
helper
peptide.
As
shown
in
Fig.
2,
Lm-gag
induces
antigen-specific
vaccine
responses
toward
immunodom-
inant
CTL
(Fig.
2A)
and
helper
(Fig.
2B)
epitopes
during
chronic
helminth
infection.
Further,
varying
the
vaccine
dose
and
regimen
does
not
alter
the
vaccine
response
to
the
immunodominant
CTL
(Fig.
2C)
or
CD4+
helper
(Fig.
2D)
epitopes.
For
vaccination
of
ani-
mals
with
chronic
Schistosomiasis,
the
vaccine
dose
was
lowered
to
0.1
LD50 or
the
schedule
was
altered
to
eliminate
the
boost,
result-
ing
in
a
prime
only
vaccine
strategy.
No
significant
differences
were
observed
when
comparing
the
responsive
groups.
These
results
suggest
the
Listeria
vector
is
a
potent
inducer
of
the
immune
sys-
tem
for
generation
of
vaccine-specific
responses
in
the
context
of
pre-existing
chronic
helminth
infection
without
the
need
for
drug
intervention
(anthelminthics)
prior
to
vaccination
and
there
exists
ample
room
for
modification
of
vaccine
dose
and
regimen.
To
validate
that
the
responses
measured
by
ELISpot
assays
result
from
functional
effector
cells,
an
in
vivo
CTL
assay
was
performed
(Fig.
2E).
Shown
by
a
lack
of
specific
killing
in
the
Lm-E7
vacci-
nated
(control)
mice,
non-specific
effects
of
the
Listeria
vector
itself
are
not
a
cause
for
the
observed
killing.
No
significant
difference
was
observed
between
Lm-gag
vaccinated
groups
with
and
with-
out
chronic
schistosomiasis,
indicating
the
vaccine
response
is
as
effective
in
helminth
infected
mice
as
in
uninfected
mice.
Levels
of
killing
are
modest,
perhaps
owing
to
the
multitude
of
proteins
expressed
by
Listeria,
resulting
in
a
small
percentage
of
the
host’s
immune
responses
specific
to
the
HIV
IIIB
gag
protein.
However,
clear
gag-specific
killing
is
demonstrated
in
groups
vaccinated
with
Lm-gag,
indicating
the
IFN+
CD8+
T
cells
are,
indeed,
functional
CTLs
generated
by
Listeria
vectored
vaccines
in
a
Th2
environment.
3.3.
T
cell
compartment
composition
is
independent
of
chronic
helminth
infection
To
further
characterize
the
composition
of
T
cell
compartments
in
vaccinated
mice
with
and
without
chronic
helminth
infection,
splenocytes
were
harvested
2
wplv
and
analyzed
by
flow
cytome-
try.
Upon
polyclonal
stimulation
with
PMA
and
Ionomycin,
CD4+
helper
T
cells
secreted
IFN
or
doubly
secreted
IFN
and
IL-4,
but
very
few
cells
produced
IL-4
without
IFN
(Fig.
3A).
The
differ-
ences
between
helminth
infected
and
uninfected
mice
that
were
vaccinated
with
Listeria
vectors
were
not
statistically
significant
(p
>
0.05),
consistent
with
the
antigen-specific
data
shown
in
Fig.
2.
Additionally,
as
shown
in
Fig.
3B,
the
regulatory
T
cell
compartment
(CD4+CD25±FoxP3±)
was
also
unaltered
in
these
animals.
When
evaluating
the
CD8+
compartment
for
molecular
specificity
and
IFN
production,
there
were
no
significant
differences
between
the
groups.
However,
there
was
an
observed
(not
significant)
increase
in
IFN
producing
non-gag-specific
(tetramer
negative)
CD8+
T
cells
in
the
vaccinated,
helminth
infected
group.
Likely,
this
resulted
from
either
increased
antigen
exposure
in
helminth
infection
or
an
increase
in
the
variability
of
the
response.
To
reconcile
these
results
with
the
overall
increase
in
IL-4
and
IL-10
levels
after
ex
vivo
stimulation
seen
in
Fig.
1,
several
pos-
sibilities
exist,
such
as
T
cell
plasticity,
increased
expression
of
cytokines
by
the
same
number
of
producers,
IL-4
can
originate
from
non-T
cells
during
helminth
infection
[46]
and
IL-10
can
come
from
T
cell
compartments
not
investigated
in
this
paper
(CD4+CD25FoxP3IL10+)
[47].
Nevertheless,
the
composition
of
the
T
cell
compartments
in
Listeria
vaccinated
mice
is
independent
of
pre-existing
chronic
helminth
infection.
L.M.
Shollenberger
et
al.
/
Vaccine
31 (2013) 2050–
2056 2053
Fig.
1.
Vaccination
strategy
in
a
murine
model
of
helminth
infection.
(A)
To
verify
Th2
biasing
of
helminth
infected
mice,
splenocytes
from
uninfected
mice
(open
bars)
or
schistosome
infected
mice
at
7
wpi
(gray
bars)
or
18
wpi
(solid
bars)
were
stimulated
for
with
SEA
or
conA
ex
vivo
and
supernatants
assayed
for
IFNand
IL-4
expression
by
ELISA.
Results
from
two
independent
experiments
are
pooled
and
plotted
as
the
mean
+
SEM
of
the
IFN:IL-4
ratio.
(B)
In
order
to
assess
vaccine-specific
responses
in
helminth
infected
mice,
6–8-week
old
female
BALB/c
mice
were
infected
with
Schistosoma
mansoni
or
left
naïve
and
helminth
infection
was
verified
by
ELISA
at
10
weeks
post
infection
(wpi).
Twelve
wpi,
age-matched
mice
were
primed
with
Listeria
vector
HIV-1
vaccine
(Lm-gag),
control
Listeria
vector
HPV
vaccine
(Lm-E7)
or
left
unvaccinated.
Mice
were
boosted
2
weeks
after
the
prime
in
an
identical
manner.
Vaccine
responses
were
evaluated
two
or
more
weeks
post
last
vaccination
(wplv).
(C–F)
Splenocytes
were
harvested
at
16
weeks
post
Schistosome
infection
from
Listeria
gag
vaccinated,
age
matched
mice
that
were
uninfected
(open
bars)
or
schistosome
infected
(solid
bars).
Splenocytes
were
stimulated
with
SEA
or
conA
ex
vivo
and
supernatants
assayed
for
IFN,
IL-4
and
IL-10
expression
by
ELISA.
Pooled
results
from
3
independent
experiments
are
plotted
as
the
mean
+
SEM
of
the
IFN:IL-4
ratio
(C)
and
absolute
IFN
(D),
IL-4
(E)
and
IL-10
values
(F)
(n
=
16–19).
Responding
groups
were
analyzed
by
ANOVA
with
Bonferroni
post
test.
*p
<
0.05,
***p
<
0.001,
****p
<
0.0001.
Fig.
2.
Listeria
vector
vaccines
induce
functional
HIV-specific
T
cell
responses
in
helminth
(schistosome)
infected
mice.
Naïve
or
chronic
schistosome
infected
mice
were
vaccinated
with
HIV
gag
(Lm-gag),
a
control
vector
(Lm-E7)
or
left
unvaccinated.
Splenocytes
were
collected
2
wplv
and
immune
responses
to
the
immunodominant
CTL
(A)
and
CD4+
helper
(B)
epitopes
were
assayed
by
IFN
ELISpot.
Vaccine
dose
(0.1
LD50)
and
regimen
(P
=
prime
only,
P
+
B
=
prime
and
boost)
were
altered
in
schistosome
infected
mice
and
vaccine-specific
responses
against
the
CTL
(C)
and
CD4+
helper
(D)
epitopes
were
evaluated.
(A–D)
Experiments
were
performed
in
triplicate,
pooled
and
shown
as
mean
+
SEM.
Total
numbers
of
mice
per
group
are
shown
and
statistical
analyses
of
responding
groups
were
performed
using
two-way
ANOVA
with
a
Bonferroni
post-test.
(E)
To
confirm
functionality
of
the
CTL
responses
shown
in
panel
A,
an
in
vivo
CTL
assay
was
performed.
Results
from
individual
mice
are
plotted
and
the
groups
that
showed
specific
killing
were
compared
by
unpaired,
two-tailed
t-test
analysis.
2054 L.M.
Shollenberger
et
al.
/
Vaccine
31 (2013) 2050–
2056
Fig.
3.
Comparable
T
cell
compartments
exist
2
weeks
post
last
Listeria
vector
HIV
gag
vaccination
independent
of
chronic
helminth
infection.
Naïve
(open
bars)
or
chronic
schistosome
infected
(solid
bars)
mice
were
vaccinated
with
Lm-gag
using
the
standard
prime-boost
method.
Splenocytes
were
collected
2
wplv
and
assayed
by
flow
cytometry
for
Th1
and
Th2
cells
(A),
T
regulatory
cells
(B)
or
specific
CTLs
by
tetramer
staining
(C).
Percentages
(upper
panel)
and
total
numbers
of
the
cells
in
the
spleen
(lower
panel)
are
shown.
Results
(n
=
6)
are
shown
as
mean
+
SEM.
Statistical
analysis
was
performed
using
two-way
ANOVA
with
a
Bonferroni
post-test.
Fig.
4.
Listeria
vectors
drive
HIV-specific
cell-mediated
vaccine
responses
that
are
durable
in
mice
chronically
infected
with
helminth
parasites.
(A
and
B)
Splenocytes
from
uninfected
(open
bars)
and
schistosome-infected
(hatched
bars)
mice
were
collected
at
various
times
post
last
vaccination
and
immune
responses
to
the
immunodominant
CTL
(A)
and
CD4+
helper
(B)
epitopes
were
assayed
by
IFN
ELISpot,
shown
as
mean
+
SEM
and
analyzed
by
unpaired,
two-tailed
t-test.
n
=
5–6
per
group.
(C
and
D)
Splenocytes
harvested
from
uninfected
and
schistosome-infected
mice
14
wplv
were
analyzed
by
flow
cytometry
to
confirm
molecular
specificity
to
the
HIV
gag
immunodominant
epitope
(C)
and
evaluate
CD8+
memory
compartments
(D).
Total
splenocytes
were
stained
with
-CD8
antibody
and
gag-tetramer
or
memory
markers
and
live
cells
were
acquired
and
analyzed
within
the
CD8+
population.
Groups
are
compared
using
unpaired,
two-tailed
t-test
analysis.
n
=
5–6
per
group.
*p
<
0.05,
**p
<
0.01,
****p
<
0.0001.
L.M.
Shollenberger
et
al.
/
Vaccine
31 (2013) 2050–
2056 2055
3.4.
Listeria
vectors
generate
HIV-specific
cell
mediated
vaccine
responses
that
are
durable
in
pre-existing
chronic
helminth
infection
To
evaluate
the
duration
of
the
vaccine-specific
cell-mediated
immune
responses,
uninfected
or
Schistosome-infected
mice
were
analyzed
several
months
after
the
last
vaccination
(Fig.
4A
and
B).
Although
differences
were
seen
for
helper
responses
at
early
time
points
of
this
experiment
(not
normal),
no
significant
differences
were
found
for
CTL
responses,
indicating
the
effector
(and/or
effec-
tor
memory)
cell
response
to
the
vaccine
is
unchanged
over
time
between
the
groups
and
the
helper
response
equalizes
with
time.
These
results
suggest
the
vaccine-specific
cell
mediated
immune
responses
generated
by
the
Listeria
vaccine
vectors
are
durable
and
independent
of
pre-existing
chronic
helminth
infection.
Therefore,
we
hypothesize
only
the
kinetics
of
memory
generation
is
affected
by
chronic
helminth
infection
and
not
the
magnitude
of
the
resul-
tant
memory
pool.
Tetramer
staining
at
14
wplv
was
used
to
verify
the
IFN
responses
measured
by
ELISpot
arise
from
antigen-specific
CD8+
T
cells
(Fig.
4C).
Consistent
with
the
ELISpot
data,
no
significant
dif-
ference
was
found
when
comparing
tetramer-positive
populations
between
vaccinated
groups
with
and
without
chronic
helminth
infection.
Further,
CD8+
memory
populations
were
evaluated
using
flow
cytometry.
As
shown
in
Fig.
4D,
there
was
a
higher
percentage
of
CD8+
central
memory
(CCR7+CD62L+)
populations
at
14
wplv
in
uninfected
mice
than
helminth
infected
animals,
coincident
with
a
lower
CD8+
effector
memory
and
effector
(CCR7CD62L)
com-
partment.
These
results
suggest
the
similar
values
seen
at
14
wplv
from
the
IFN
ELISpot
assays
were
a
combined
result
from
CTLs
and
the
CD8+
TCM and
TEM memory
pools.
4.
Conclusions
Vaccine
development
should
take
into
consideration
underly-
ing
diseases
present
in
recipient
populations
that
may
suppress
vaccine-specific
immune
responses.
Therefore,
it
is
critical
to
test
candidate
vaccines
in
animal
models
that
represent
as
closely
as
possible,
characteristics
of
the
intended
target
human
population.
Specifically,
vaccines
for
HIV,
TB
and
malaria
need
to
be
effective
in
helminth
infected
and
uninfected
individuals.
The
results
pre-
sented
here
demonstrate
the
existence
of
vaccine
vectors
capable
of
overcoming
helminth
infection
without
the
need
to
administer
anthelminthics
prior
to
vaccination.
Remarkably,
Listeria
vectored
vaccines
are
able
to
induce
robust
and
durable
CTL
and
Th1
vaccine
responses
in
helminth
infected,
immune
suppressed
populations.
This
represents
an
important
step
toward
our
goal
of
induc-
ing
vaccine-specific
immunity
in
helminth
infected,
Th2
biased
and
immune
suppressed
populations.
Further,
our
results,
taken
together
with
other
studies,
suggest
that
attenuated
Listeria
vector
vaccines
might
be
effective
on
a
global
scale.
We
hope
these
stud-
ies
encourage
others
who
are
developing
global
health
vaccines
to
validate
vaccine
efficacy
in
helminth
infected
recipients.
Acknowledgements
The
authors
would
like
to
thank
the
NIAID
Schistosomiasis
Resource
Center
for
biological
reagents
as
well
as
the
University
of
Georgia
College
of
Veterinary
Medicine
Cytometry
Core
Facility.
The
authors
would
also
like
to
thank
Lindsay
Nyhoff
for
techni-
cal
assistance
and
Drs.
Balazs
Rada
and
Wendy
Watford
for
critical
review
of
this
manuscript.
Conflicts
of
interest:
A
patent
has
been
filed
on
behalf
of
the
authors
LS,
YP
and
DH
for
use
of
Listeria
vectored
vaccines
in
helminth
endemic
areas.
CB
and
KA
have
no
competing
financial
interests
to
declare.
YP
discloses
that
she
has
a
financial
inter-
est
in
Advaxis,
Inc.,
a
vaccine
and
therapeutic
company
that
has
licensed
or
has
an
option
to
license
all
patents
from
the
University
of
Pennsylvania
that
concern
the
use
of
Listeria
or
listerial
products
as
vaccines.
Funding
sources:
Supported
by
grants
NIH-AI-071883
and
NIH-
AI-078787
awarded
to
DAH.
References
[1]
Montefiori
DC,
Karnasuta
C,
Huang
Y,
Ahmed
H,
Gilbert
P,
de
Souza
MS,
et
al.
Magnitude
and
breadth
of
the
neutralizing
antibody
response
in
the
RV144
and
Vax003
HIV-1
vaccine
efficacy
trials.
J
Infect
Dis
2012;206:431–41.
[2]
Haynes
BF,
Gilbert
PB,
McElrath
MJ,
Zolla-Pazner
S,
Tomaras
GD,
Alam
SM,
et
al.
Immune-correlates
analysis
of
an
HIV-1
vaccine
efficacy
trial.
N
Engl
J
Med
2012;366:1275–86.
[3] UNAIDS.
Report
on
the
global
AIDS
epidemic;
2010.
[4]
Fincham
JE,
Markus
MB,
Adams
VJ.
Could
control
of
soil-transmitted
helminthic
infection
influence
the
HIV/AIDS
pandemic.
Acta
Trop
2003;86:315–33.
[5]
de
Silva
NR,
Brooker
S,
Hotez
PJ,
Montresor
A,
Engels
D,
Savioli
L.
Soil-
transmitted
helminth
infections:
updating
the
global
picture.
Trends
Parasitol
2003;19:547–51.
[6] Finkelman
FD,
Urban
Jr
JF.
The
other
side
of
the
coin:
the
protective
role
of
the
TH2
cytokines.
J
Allergy
Clin
Immunol
2001;107:772–80.
[7]
Urban
Jr
JF,
Fayer
R,
Sullivan
C,
Goldhill
J,
Shea-Donohue
T,
Madden
K,
et
al.
Local
TH1
and
TH2
responses
to
parasitic
infection
in
the
intestine:
regulation
by
IFN-gamma
and
IL-4.
Vet
Immunol
Immunopathol
1996;54:337–44.
[8]
Thomas
PG,
Harn
Jr
DA.
Immune
biasing
by
helminth
glycans.
Cell
Microbiol
2004;6:13–22.
[9]
Urban
Jr
JF,
Schopf
L,
Morris
SC,
Orekhova
T,
Madden
KB,
Betts
CJ,
et
al.
Stat6
signaling
promotes
protective
immunity
against
Trichinella
spiralis
through
a
mast
cell-
and
T
cell-dependent
mechanism.
J
Immunol
2000;164:2046–52.
[10]
Remou
F,
Rogerie
F,
Gallissot
MC,
Guyatt
HL,
Neyrinck
JL,
Diakkhate
MM,
et
al.
Sex-dependent
neutralizing
humoral
response
to
Schistosoma
mansoni
28GST
antigen
in
infected
human
populations.
J
Infect
Dis
2000;181:1855–9.
[11]
Fallon
PG,
Fookes
RE,
Wharton
GA.
Temporal
differences
in
praziquantel-
and
oxamniquine-induced
tegumental
damage
to
adult
Schistosoma
mansoni:
implications
for
drug-antibody
synergy.
Parasitology
1996;112(Pt
1):47–58.
[12] Network
Hvt
Current
Vaccine
Trials.
Available
from:
www.hvtn.org/science/trials.html [cited
08.04.12].
[13]
Hanke
T.
Vehicles
for
genetic
vaccines
against
human
immunodeficiency
virus:
induction
of
T
cell-mediated
immune
responses.
Curr
Mol
Med
2001;1:123–35.
[14] Masopust
D.
Developing
an
HIV
cytotoxic
T-lymphocyte
vaccine:
issues
of
CD8
T-cell
quantity,
quality
and
location.
J
Intern
Med
2009;265:125–37.
[15]
Sabin
EA,
Araujo
MI,
Carvalho
EM,
Pearce
EJ.
Impairment
of
tetanus
toxoid-
specific
Th1-like
immune
responses
in
humans
infected
with
Schistosoma
mansoni.
J
Infect
Dis
1996;173:269–72.
[16] Elias
D,
Akuffo
H,
Pawlowski
A,
Haile
M,
Schon
T,
Britton
S.
Schistosoma
mansoni
infection
reduces
the
protective
efficacy
of
BCG
vaccination
against
virulent
Mycobacterium
tuberculosis.
Vaccine
2005;23:1326–34.
[17] Malhotra
I,
Mungai
P,
Wamachi
A,
Kioko
J,
Ouma
JH,
Kazura
JW,
et
al.
Helminth-
and
Bacillus
Calmette–Guerin-induced
immunity
in
children
sensitized
in
utero
to
filariasis
and
schistosomiasis.
J
Immunol
1999;162:6843–8.
[18]
Da’Dara
AA,
Lautsch
N,
Dudek
T,
Novitsky
V,
Lee
TH,
Essex
M,
et
al.
Helminth
infection
suppresses
T-cell
immune
response
to
HIV-DNA-based
vaccine
in
mice.
Vaccine
2006;24:5211–9.
[19]
N‘Goran
EK,
Utzinger
J,
N‘Guessan
AN,
Müller
I,
Zamblé
K,
Lohourignon
KL,
et
al.
Reinfection
with
Schistosoma
haematobium
following
school-based
chemother-
apy
with
praziquantel
in
four
highly
endemic
villages
in
Côte
d’Ivoire.
Trop
Med
Int
Health
2001;6:817–25.
[20]
Letvin
NL.
Progress
in
the
development
of
an
HIV-1
vaccine.
Science
1998;280:1875–80.
[21]
Yeung
VP,
Gieni
RS,
Umetsu
DT,
DeKruyff
RH.
Heat-killed
Listeria
monocytogenes
as
an
adjuvant
converts
established
murine
Th2-dominated
immune
responses
into
Th1-dominated
responses.
J
Immunol
1998;161:4146–52.
[22]
Li
Z,
Zhang
M,
Zhou
C,
Zhao
X,
Iijima
N,
Frankel
FR.
Novel
vaccination
protocol
with
two
live
mucosal
vectors
elicits
strong
cell-mediated
immu-
nity
in
the
vagina
and
protects
against
vaginal
virus
challenge.
J
Immunol
2008;180:2504–13.
[23]
Bruhn
KW,
Craft
N,
Miller
JF.
Listeria
as
a
vaccine
vector.
Microbes
Infect
2007;9:1226–35.
[24] Jiang
S,
Rasmussen
RA,
Nolan
KM,
Frankel
FR,
Lieberman
J,
McClure
HM,
et
al.
Live
attenuated
Listeria
monocytogenes
expressing
HIV
Gag:
immunogenicity
in
rhesus
monkeys.
Vaccine
2007;25:7470–9.
[25]
Mehandru
S,
Dandekar
S.
Role
of
the
gastrointestinal
tract
in
establishing
infec-
tion
in
primates
and
humans.
Curr
Opin
HIV
AIDS
2008;3:22–7.
[26]
Angelakopoulos
H,
Loock
K,
Sisul
DM,
Jensen
ER,
Miller
JF,
Hohmann
EL.
Safety
and
shedding
of
an
attenuated
strain
of
Listeria
monocytogenes
with
a
deletion
of
actA/plcB
in
adult
volunteers:
a
dose
escalation
study
of
oral
inoculation.
Infect
Immun
2002;70:3592–601.
[27]
Flutter
B,
Gao
B.
MHC
class
I
antigen
presentation—recently
trimmed
and
well
presented.
Cell
Mol
Immunol
2004;1:22–30.
2056 L.M.
Shollenberger
et
al.
/
Vaccine
31 (2013) 2050–
2056
[28]
Watts
C.
The
exogenous
pathway
for
antigen
presentation
on
major
histocom-
patibility
complex
class
II
and
CD1
molecules.
Nat
Immunol
2004;5:685–92.
[29]
Shen
L,
Rock
KL.
Priming
of
T
cells
by
exogenous
antigen
cross-presented
on
MHC
class
I
molecules.
Curr
Opin
Immunol
2006;18:85–91.
[30]
Murata
K,
Garcia-Sastre
A,
Tsuji
M,
Rodrigues
M,
Rodriguez
D,
Rodriguez
JR,
et
al.
Characterization
of
in
vivo
primary
and
secondary
CD8+
T
cell
responses
induced
by
recombinant
influenza
and
vaccinia
viruses.
Cell
Immunol
1996;173:96–107.
[31]
Kundig
TM,
Kalberer
CP,
Hengartner
H,
Zinkernagel
RM.
Vaccination
with
two
different
vaccinia
recombinant
viruses:
long-term
inhibition
of
secondary
vac-
cination.
Vaccine
1993;11:1154–8.
[32] Scaria
A,
St
George
JA,
Gregory
RJ,
Noelle
RJ,
Wadsworth
SC,
Smith
AE,
et
al.
Antibody
to
CD40
ligand
inhibits
both
humoral
and
cellular
immune
responses
to
adenoviral
vectors
and
facilitates
repeated
administration
to
mouse
airway.
Gene
Ther
1997;4:611–7.
[33] Kay
MA,
Meuse
L,
Gown
AM,
Linsley
P,
Hollenbaugh
D,
Aruffo
A,
et
al.
Transient
immunomodulation
with
anti-CD40
ligand
antibody
and
CTLA4Ig
enhances
persistence
and
secondary
adenovirus-mediated
gene
transfer
into
mouse
liver.
Proc
Natl
Acad
Sci
USA
1997;94:4686–91.
[34] Leong
ML,
Hampl
J,
Liu
W,
Mathur
S,
Bahjat
KS,
Luckett
W,
et
al.
Impact
of
preexisting
vector-specific
immunity
on
vaccine
potency:
characterization
of
Listeria
monocytogenes-specific
humoral
and
cellular
immunity
in
humans
and
modeling
studies
using
recombinant
vaccines
in
mice.
Infect
Immun
2009;77:3958–68.
[35]
Starks
H,
Bruhn
KW,
Shen
H,
Barry
RA,
Dubensky
TW,
Brockstedt
D,
et
al.
Listeria
monocytogenes
as
a
vaccine
vector:
virulence
attenuation
or
existing
antivec-
tor
immunity
does
not
diminish
therapeutic
efficacy.
J
Immunol
2004;173:
420–7.
[36]
Sciaranghella
G,
Lakhashe
SK,
Ayash-Rashkovsky
M,
Mirshahidi
S,
Siddappa
NB,
Novembre
FJ,
et
al.
A
live
attenuated
Listeria
monocytogenes
vaccine
vector
expressing
SIV
Gag
is
safe
and
immunogenic
in
macaques
and
can
be
adminis-
tered
repeatedly.
Vaccine
2011;29:476–86.
[37]
Whitney
JB,
Mirshahidi
S,
Lim
SY,
Goins
L,
Ibegbu
CC,
Anderson
DC,
et
al.
Prior
exposure
to
an
attenuated
Listeria
vaccine
does
not
reduce
immunogenicity:
pre-clinical
assessment
of
the
efficacy
of
a
Listeria
vaccine
in
the
induction
of
immune
responses
against
HIV.
J
Immune
Based
Ther
Vaccines
2011;9:2.
[38]
Boyer
JD,
Robinson
TM,
Maciag
PC,
Peng
X,
Johnson
RS,
Pavlakis
G,
et
al.
DNA
prime
Listeria
boost
induces
a
cellular
immune
response
to
SIV
antigens
in
the
rhesus
macaque
model
that
is
capable
of
limited
suppression
of
SIV239
viral
replication.
Virology
2005;333:88–101.
[39]
Ikonomidis
G,
Portnoy
DA,
Gerhard
W,
Paterson
Y.
Influenza-specific
immu-
nity
induced
by
recombinant
Listeria
monocytogenes
vaccines.
Vaccine
1997;15:433–40.
[40] Jia
Q,
Lee
BY,
Clemens
DL,
Bowen
RA,
Horwitz
MA.
Recombinant
atten-
uated
Listeria
monocytogenes
vaccine
expressing
Francisella
tularensis
IglC
induces
protection
in
mice
against
aerosolized
Type
A
F.
tularensis.
Vaccine
2009;27:1216–29.
[41] Orr
MT,
Orgun
NN,
Wilson
CB,
Way
SS.
Cutting
edge:
recombinant
Listeria
mono-
cytogenes
expressing
a
single
immune-dominant
peptide
confers
protective
immunity
to
herpes
simplex
virus-1
infection.
J
Immunol
2007;178:4731–5.
[42]
Soussi
N,
Saklani-Jusforgues
H,
Colle
JH,
Milon
G,
Glaichenhaus
N,
Goossens
PL.
Effect
of
intragastric
and
intraperitoneal
immunisation
with
attenuated
and
wild-type
LACK-expressing
Listeria
monocytogenes
on
control
of
murine
Leishmania
major
infection.
Vaccine
2002;20:2702–12.
[43]
Mustafa
W,
Maciag
PC,
Pan
ZK,
Weaver
JR,
Xiao
Y,
Isaacs
SN,
et
al.
Listeria
mono-
cytogenes
delivery
of
HPV-16
major
capsid
protein
L1
induces
systemic
and
mucosal
cell-mediated
CD4+
and
CD8+
T-cell
responses
after
oral
immuniza-
tion.
Viral
Immunol
2009;22:195–204.
[44]
Mata
M,
Yao
Z-J,
Zubair
A,
Syres
K,
Paterson
Y.
Evaluation
of
a
recombinant
Listeria
monocytogenes
expressing
an
HIV
protein
that
protects
mice
against
viral
challenge.
Vaccine
2001;19:1435–45.
[45]
Gunn
GR,
Zubair
A,
Peters
C,
Pan
ZK,
Wu
TC,
Paterson
Y.
Two
Listeria
mono-
cytogenes
vaccine
vectors
that
express
different
molecular
forms
of
human
papilloma
virus-16
(HPV-16)
E7
induce
qualitatively
different
T
cell
immunity
that
correlates
with
their
ability
to
induce
regression
of
established
tumors
immortalized
by
HPV-16.
J
Immunol
2001;167:6471–9.
[46]
Metwali
A,
Andres
BD,
Blum
A,
Elliott
D,
Li
J,
Qadir
K,
et
al.
Th2-type
granuloma
development
in
acute
murine
schistosomiasis
is
only
partly
dependent
on
CD4+
T
cells
as
the
source
of
IL-4.
Eur
J
Immunol
2002;32:1242–52.
[47]
Dewals
B,
Hoving
JC,
Horsnell
WGC,
Brombacher
F.
Control
of
Schistosoma
man-
soni
egg-induced
inflammation
by
IL-4-responsive
CD4+CD25CD103+Foxp3
cells
is
IL-10-dependent.
Eur
J
Immunol
2010;40:2837–47.
... The greatest incidence of each of these diseases is in sub-Saharan Africa, where helminth infection is endemic (1,2). Helminth parasites bias immune responses to the CD4 ϩ Th2 type and can be immune suppressive (3)(4)(5)(6)(7)(8)(9)(10). To evaluate the influence of helminth infection on vaccines, we can evaluate the ability to induce vaccine-specific immune responses in helminth-infected recipients or in recipients who have been treated with immune-biasing helminth antigens (11)(12)(13). ...
... One method to overcome helminth-induced vaccine suppression is to identify vaccine vectors that are capable of producing vaccine-specific responses irrespective of helminth infection. In this regard, a Listeria vector HIV vaccine was recently shown to induce potent CD8 ϩ and Th1-type vaccine responses in schistosome-infected mice (12,13). ...
... Together, the results presented here and in our recent study (24) show that SEA functioned counterintuitively when administered with the Lm-Gag vaccine, stimulating CTL responses rather than suppressing them. Although it is well known that Listeria is a potent inducer of CD8 ϩ immunity, the specific mechanism(s) by which Listeria vector vaccines can both overcome the immunosuppressive environment of chronic helminth infection (12,13) as well as be adjuvanted by SEA is currently being investigated. ...
Article
Full-text available
Schistosome infection induces significant T helper type-2 (Th2) and anti-inflammatory immune responses and has been shown to negatively impact vaccine efficacy. Our goal was to determine if administration of schistosome soluble egg antigens (SEA) would negatively influence the induction of CTL and Th1-type T cell responses to an HIV candidate vaccine in the Th1-biased C57BL/6 mouse strain. Initial experiments failed, as we were unable to detect any response to the defined Class I epitope for HIV-1 IIIB Gag. Therefore, we initiated an epitope mapping study to identify C57BL/6 (H-2(b)) T cell epitopes in HIV-1 IIIB Gag in order to perform the experiments. The analysis defined two previously unreported minimal Class I H-2(b) and Class II I-A(b) epitopes for HIV-1 IIIB Gag. The newly defined HIV-1 IIIB Gag epitopes were used to evaluate the influence of SEA on generation of CTL and Th1-type HIV-1 IIIB Gag responses. Surprisingly, in contrast to our hypothesis, we observed that co-administration of SEA with the Listeria vector expressing HIV-1 IIIB Gag (Lm-Gag), led to significantly increased frequency of IFNγ-producing CD8+ and CD4+ T cells in C57BL/6 mice compared to mice immunized with Lm-Gag only. These observations suggest that SEA contains, in addition to Th2-type and immune suppressive molecules, substances that can act with the Lm-Gag vaccine to increase CTL and Th1-type vaccine-specific immune responses. Copyright © 2014, American Society for Microbiology. All Rights Reserved.
... However, as reinfection is common and frequent in endemic populations [16], this observation highlighted the need to discover vaccine vectors/delivery systems capable of driving potent, vaccine-specific responses, even in helminth infected recipients. To that end, we investigated whether a Listeria vector HIV-1 vaccine would drive Gag-specific CD4+ and CD8+ T cell responses in schistosome infected mice and determined that the Listeria vector HIV-1 vaccine induced robust, cell-mediated Gagspecific responses whether or not mice were infected with a model helminth parasite, Schistosoma mansoni [17]. ...
... The study by Shollenberger et al. [17] demonstrated that attenuated Listeria is an ideal vector for vaccine antigens as it generates robust cell-mediated responses that do not suffer from pre-existing vector immunity, in contrast to what is seen with adenovirus or vaccinia virus vectors [18][19][20][21][22][23][24][25]. Bacterial and viral vectors induce durable CD4+ and CD8+ antiviral T cell responses, components of an effective vaccination strategy for HIV [1,[26][27][28][29]. ...
... For these studies, we used the identical Listeria vector HIV-1 Gag vaccine (Lm-Gag), previously shown to function in mice chronically infected with S. mansoni [17]. In contrast to the initial study by Shollenberger et al. [17], here we asked if helminth infection post vaccination would impact maintenance of vaccine-specific responses, and if subsequent infection with S. mansoni would impact the ability of a second boost to drive vaccine-specific responses. ...
... Infection with the liver fluke Schistosoma mansoni reduced the cellular response to vaccination with a multi epitope T cell DNA vaccine against HIV-1 C [44]. By contrast, vaccinating with a HIV-1 gag protein expressing Lysteria monocytogenes as living carrier induced a gag-specific T cell response in S. mansoni-infected and non-infected mice to the same extent [45]. ...
Article
Full-text available
The world health organization estimates that more than a quarter of the human population is infected with parasitic worms that are called helminths. Many helminths suppress the immune system of their hosts to prolong their survival. This helminth-induced immunosuppression “spills over” to unrelated antigens and can suppress the immune response to vaccination against other pathogens. Indeed, several human studies have reported a negative correlation between helminth infections and responses to vaccinations. Using mice that are infected with the parasitic nematode Litomosoides sigmodontis as a model for chronic human filarial infections, we reported previously that concurrent helminth infection impaired the vaccination-induced protection against the human pathogenic 2009 pandemic H1N1 influenza A virus (2009 pH1N1). Vaccinated, helminth-infected mice produced less neutralizing, influenza-specific antibodies than vaccinated naïve control mice. Consequently helminth-infected and vaccinated mice were not protected against a challenge infection with influenza virus but displayed high virus burden in the lung and a transient weight loss. In the current study we tried to improve the vaccination efficacy using vaccines that are licensed for humans. We either introduced a prime-boost vaccination regimen using the non-adjuvanted anti-influenza vaccine Begripal or employed the adjuvanted influenza vaccine Fluad. Although both strategies elevated the production of influenza-specific antibodies and protected mice from the transient weight loss that is caused by an influenza challenge infection, sterile immunity was not achieved. Helminth-infected vaccinated mice still had high virus burden in the lung while non-helminth-infected vaccinated mice rapidly cleared the virus. In summary we demonstrate that basic improvements of influenza vaccination regimen are not sufficient to confer sterile immunity on the background of helminth-induced immunosuppression, despite amelioration of pathology i.e. weight loss. Our findings highlight the risk of failed vaccinations in helminth-endemic areas, especially in light of the ongoing vaccination campaign to control the COVID-19 pandemic.
... In this context, life carrier vaccines such as an antigenexpressing live Salmonella strains or irradiated sporozoites conferred protection against a P. berghei or P. chabaudi sporozoite challenge infection in helminth-infected mice in settings where DNA-or protein-based vaccines failed (15,52). Likewise, the HIV-specific cellular response to an attenuated HIV-gag expressing Listeria monocytogenes vaccine vector was not compromised in helminth-infected mice although protection could not be tested in this study (53). ...
Article
Full-text available
Helminths still infect a quarter of the human population. They manage to establish chronic infections by downmodulating the immune system of their hosts. Consequently, the immune response of helminth-infected individuals to vaccinations may be impaired as well. Here we study the impact of helminth-induced immunomodulation on vaccination efficacy in the mouse system. We have previously shown that an underlying Litomosoides sigmodontis infection reduced the antibody (Ab) response to anti-influenza vaccination in the context of a systemic expansion of type 1 regulatory T cells (Tr1). Most important, vaccine-induced protection from a challenge infection with the 2009 pandemic H1N1 influenza A virus (2009 pH1N1) was impaired in vaccinated, L. sigmodontis-infected mice. Here, we aim at the restoration of vaccination efficacy by drug-induced deworming. Treatment of mice with Flubendazole (FBZ) resulted in elimination of viable L. sigmodontis parasites in the thoracic cavity after two weeks. Simultaneous FBZ-treatment and vaccination did not restore Ab responses or protection in L. sigmodontis-infected mice. Likewise, FBZ-treatment two weeks prior to vaccination did not significantly elevate the influenza-specific Ig response and did not protect mice from a challenge infection with 2009 pH1N1. Analysis of the regulatory T cell compartment revealed that L. sigmodontis-infected and FBZ-treated mice still displayed expanded Tr1 cell populations that may contribute to the sustained suppression of vaccination responses in successfully dewormed mice. To outcompete this sustained immunomodulation in formerly helminth-infected mice, we finally combined the drug-induced deworming with an improved vaccination regimen. Two injections with the non-adjuvanted anti-influenza vaccine Begripal conferred 60% protection while MF59-adjuvanted Fluad conferred 100% protection from a 2009 pH1N1 infection in FBZ-treated, formerly L. sigmodontis-infected mice. Of note, applying this improved prime-boost regimen did not restore protection in untreated L. sigmodontis-infected mice. In summary our findings highlight the risk of failed vaccinations due to helminth infection.
... Our choice for each study was guided by the authors of those studies, and by searching for information on the immune response required to combat each of the vaccine targets. For example, vaccine-specific cytophilic antibodies or % vaccine-specific T cells were often considered the best measures of vaccine-induced immunity [17,18]. For studies that measured the same variable at multiple time-points [19,20], the effect size was calculated for each time-point and these were then averaged to provide a single effect size for that variable/study. ...
Article
Full-text available
Immune responses to vaccination are heterogeneous between individuals; the same vaccine that provides protection in one circumstance may be ineffective in another. One factor that could influence the response to vaccination is concurrent or prior infection with unrelated parasites. Here, we review both the experimental and epidemiological literature on parasite-vaccine interactions, and present a meta-analysis of the published data. In total, our review returned 101 relevant articles, 50 of which met criteria for meta-analysis. Parasite factors potentially affecting vaccination include the type of parasite involved, the stage of infection, and the timing of infection relative to vaccination. Vaccine factors affecting likelihood of interference by parasites include vaccine formulation, route of administration, and the type of immune response required to provide protection against the target antigen. Our meta-analysis of these data show three key things: (1) parasite infections at the time of vaccination result in worse immunisation outcomes, (2) chronic helminth infections are more likely to negatively impact immunisation than acute helminth infections, and (3) thymus-dependent vaccines are more susceptible to parasite interference than thymus-independent vaccines. Our findings highlight the importance of considering and mitigating parasite infections: by taking parasites into account, it should be possible to more effectively immunise individuals and populations.
... In light of these findings, this study suggests that, whilst elimination of worms can offer an affordable and a simple means of antihelminthic treatment, only partially restoration of immune responsiveness to T cell-based vaccines for HIV-1 and other infectious diseases in helminth endemic settings may be achieved. Thus, it would be important to evaluate vaccine delivery systems that can potentially overcome the negative impact of concurrent helminthiasis as previously suggested [93]. An alternative avenue would be the discovery of antihelminthic drugs which are effective in elimination of SmE from the host's tissues in addition to the elimination of the parasitic worms. ...
Article
Full-text available
Future HIV vaccines are expected to induce effective Th1 cell-mediated and Env-specific antibody responses that are necessary to offer protective immunity to HIV infection. However, HIV infections are highly prevalent in helminth endemic areas. Helminth infections induce polarised Th2 responses that may impair HIV vaccine-generated Th1 responses. In this study, we tested if Schistosoma mansoni (Sm) infection altered immune responses to SAAVI candidate HIV vaccines (DNA and MVA) and an HIV-1 gp140 Env protein vaccine (gp140) and whether parasite elimination by chemotherapy or the presence of Sm eggs (SmE) in the absence of active infection influenced the immunogenicity of these vaccines. In addition, we evaluated helminth-associated pathology in DNA and MVA vaccination groups. Mice were chronically infected with Sm and vaccinated with DNA+MVA in a prime+boost combination or MVA+gp140 in concurrent combination regimens. Some Sm-infected mice were treated with praziquantel (PZQ) prior to vaccinations. Other mice were inoculated with SmE before receiving vaccinations. Unvaccinated mice without Sm infection or SmE inoculation served as controls. HIV responses were evaluated in the blood and spleen while Sm-associated pathology was evaluated in the livers. Sm-infected mice had significantly lower magnitudes of HIV-specific cellular responses after vaccination with DNA+MVA or MVA+gp140 compared to uninfected control mice. Similarly, gp140 Env-specific antibody responses were significantly lower in vaccinated Sm-infected mice compared to controls. Treatment with PZQ partially restored cellular but not humoral immune responses in vaccinated Sm-infected mice. Gp140 Env-specific antibody responses were attenuated in mice that were inoculated with SmE compared to controls. Lastly, Sm-infected mice that were vaccinated with DNA+MVA displayed exacerbated liver pathology as indicated by larger granulomas and increased hepatosplenomegaly when compared with unvaccinated Sm-infected mice. This study shows that chronic schistosomiasis attenuates both HIV-specific T-cell and antibody responses and parasite elimination by chemotherapy may partially restore cellular but not antibody immunity, with additional data suggesting that the presence of SmE retained in the tissues after antihelminthic therapy contributes to lack of full immune restoration. Our data further suggest that helminthiasis may compromise HIV vaccine safety. Overall, these findings suggested a potential negative impact on future HIV vaccinations by helminthiasis in endemic areas.
... Despite the limits of murine models to reflect every aspect of the human situation, combined evidence gained in different mouse models for helminth infection can be informative. Since first murine studies demonstrated successful vaccination despite concurrent nematode infection by improved vaccination strategies [21,30,59,60], we suggest that development of vaccination regimes that are functional despite pre-existing nematode infection would be more promising and should be considered in addition to deworming programs before vaccination. ...
Article
Full-text available
One-third of the human population is infected with parasitic worms. To avoid being eliminated, these parasites actively dampen the immune response of their hosts. This immune modulation also suppresses immune responses to third-party antigens such as vaccines. Here, we used Litomosoides sigmodontis-infected BALB/c mice to analyse nematode-induced interference with vaccination. Chronic nematode infection led to complete suppression of the humoral response to thymus-dependent vaccination. Thereby the numbers of antigen-specific B cells as well as the serum immunoglobulin (Ig) G titres were reduced. TH2-associated IgG1 and TH1-associated IgG2 responses were both suppressed. Thus, nematode infection did not bias responses towards a TH2 response, but interfered with Ig responses in general. We provide evidence that this suppression indirectly targeted B cells via accessory T cells as number and frequency of vaccine-induced follicular B helper T cells were reduced. Moreover, vaccination using model antigens that stimulate Ig response independently of T helper cells was functional in nematode-infected mice. Using depletion experiments, we show that CD4+Foxp3+ regulatory T cells did not mediate the suppression of Ig response during chronic nematode infection. Suppression was induced by fourth stage larvae, immature adults and mature adults, and increased with the duration of the infection. By contrast, isolated microfilariae increased IgG2a responses to vaccination. This pro-inflammatory effect of microfilariae was overruled by the simultaneous presence of adults. Strikingly, a reduced humoral response was still observed if vaccination was performed more than 16 weeks after termination of L. sigmodontis infection. In summary, our results suggest that vaccination may not only fail in helminth-infected individuals, but also in individuals with a history of previous helminth infections.
... 25,26 Lm-based vaccination has also succeeded in inducing robust and functional HIV-specific cellular immune responses in mice regardless of underlying chronic infection status (e.g., helminth infections). 27 Against the parasite Leishmania major, immunization of mice with recombinant Lm delivering an L. major antigen has been shown to generate in vivo CD4 + T cells with a Th1 phenotype that exerted protective antiparasitic function. 28 Lastly, a recombinant Lm vaccine stably expressing the IglC protein of Francisella tularensis was found to induce protective immunity against lethal F. tularensis challenge in mice; this success was shown to be due to the powerful cellular CD4 + and CD8 + T cell immune responses against IglC. ...
Article
Vaccination as a medical intervention has proven capable of greatly reducing the suffering from childhood infectious disease. However, newborns and infants in particular are age groups for whom adequate vaccine-mediated protection is still largely lacking. With the challenges that the neonatal immune system faces and the required highest level of stringency for safety, designing vaccines for early life in general and the newborn in particular poses great difficulty. Nevertheless, recent advances in our understanding of neonatal immunity and its responses to vaccines and adjuvants suggest that neonatal vaccination is a task fully within reach. Among the most promising developments in neonatal vaccination is the use of Listeria monocytogenes (Lm) as a delivery platform. In this review, we will outline key properties of Lm that make it such an ideal neonatal and early life vaccine vehicle, and also discuss potential constraints of Lm as a vaccine delivery platform.
Chapter
We have coevolved with helminths in order to tolerate these parasites and minimize their virulence. However, the heterogeneity in responses to helminths leads to severe morbidity in a proportion of infected individuals. This heterogeneity is poorly understood and could be a complex relationship between host genotype, parasite genotype, and the microbiota of infected individuals. The type 2 immune response is important in limiting parasite numbers and reducing tissue damage caused by the parasites, whereas the immune regulatory network induced by helminths benefits the parasite as well as the host by limiting the effectiveness of resistance mechanisms while also reducing immune pathology. The elimination of helminths in the developed world may partly influence the rise of autoimmune disorders over the last few decades, as the immune system is no longer being tuned by the presence of helminths. There are currently efforts to determine if the reintroduction of helminths, or their biological products can be used therapeutically for the treatment of autoimmune diseases. A variety of mechanisms have been shown to play a role in suppression of inflammatory diseases by helminths and different helminths likely act through different mechanisms to suppress different inflammatory diseases. Some of the protective effects of helminth infection may be a result of indirect effects downstream of microbiota alterations. Through the combination of mechanistic clinical trials, genomic and biochemical characterization of helminths, and animal models with inflammatory diseases, pathways that mediate the coexistence of helminths and humans may be targeted as new strategies for controlling inflammatory diseases.
Article
Full-text available
Infants and children are routinely vaccinated with bacillus Calmette-Guérin (BCG) in areas of the world where worm infections are common. Because maternal helminth infection during pregnancy can sensitize the developing fetus, we studied whether this prenatal immunity persists in childhood and modifies the immune response to BCG. Children and newborns living in rural Kenya, where BCG is administered at birth and filariasis and schistosomiasis are endemic, were examined. T cells from 2- to 10-year-old children of mothers without filariasis or schistosomiasis produced 10-fold more IFN-γ in response to mycobacterial purified protein derivative than children of helminth-infected mothers (p < 0.01). This relationship was restricted to purified protein derivative because maternal infection status did not correlate with filarial Ag-driven IL-2, IFN-γ, IL-4, or IL-5 responses by children. Prospective studies initiated at birth showed that helminth-specific T cell immunity acquired in utero is maintained until at least 10–14 mo of age in the absence of infection with either Wuchereria bancrofti or Schistosoma haematobium. Purified protein derivative-driven T cell IFN-γ production evaluated 10–14 mo after BCG vaccination was 26-fold higher for infants who were not sensitized to filariae or schistosomes in utero relative to subjects who experienced prenatal sensitization (p < 0.01). These data indicate that helminth-specific immune responses acquired during gestation persist into childhood and that this prenatal sensitization biases T cell immunity induced by BCG vaccination away from type 1 IFN-γ responses associated with protection against mycobacterial infection.
Article
Full-text available
A recombinant canarypox vector expressing human immunodeficiency virus type 1 (HIV-1) Gag, Pro, and membrane-linked gp120 (vCP1521), combined with a bivalent gp120 protein boost (AIDSVAX B/E), provided modest protection against HIV-1 infection in a community-based population in Thailand (RV144 trial). No protection was observed in Thai injection drug users who received AIDSVAX B/E alone (Vax003 trial). We compared the neutralizing antibody response in these 2 trials. Neutralization was assessed with tier 1 and tier 2 strains of virus in TZM-bl and A3R5 cells. Neutralization of several tier 1 viruses was detected in both RV144 and Vax003. Peak titers were higher in Vax003 and waned rapidly in both trials. The response in RV144 was targeted in part to V3 of gp120.vCP1521 priming plus 2 boosts with gp120 protein was superior to 2 gp120 protein inoculations alone, confirming a priming effect for vCP1521. Sporadic weak neutralization of tier 2 viruses was detected only in Vax003 and A3R5 cells. The results suggest either that weak neutralizing antibody responses can be partially protective against HIV-1 in low-risk heterosexual populations or that the modest efficacy seen in RV144 was mediated by other immune responses, either alone or in combination with neutralizing antibodies.
Article
Full-text available
In the RV144 trial, the estimated efficacy of a vaccine regimen against human immunodeficiency virus type 1 (HIV-1) was 31.2%. We performed a case-control analysis to identify antibody and cellular immune correlates of infection risk. In pilot studies conducted with RV144 blood samples, 17 antibody or cellular assays met prespecified criteria, of which 6 were chosen for primary analysis to determine the roles of T-cell, IgG antibody, and IgA antibody responses in the modulation of infection risk. Assays were performed on samples from 41 vaccinees who became infected and 205 uninfected vaccinees, obtained 2 weeks after final immunization, to evaluate whether immune-response variables predicted HIV-1 infection through 42 months of follow-up. Of six primary variables, two correlated significantly with infection risk: the binding of IgG antibodies to variable regions 1 and 2 (V1V2) of HIV-1 envelope proteins (Env) correlated inversely with the rate of HIV-1 infection (estimated odds ratio, 0.57 per 1-SD increase; P=0.02; q=0.08), and the binding of plasma IgA antibodies to Env correlated directly with the rate of infection (estimated odds ratio, 1.54 per 1-SD increase; P=0.03; q=0.08). Neither low levels of V1V2 antibodies nor high levels of Env-specific IgA antibodies were associated with higher rates of infection than were found in the placebo group. Secondary analyses suggested that Env-specific IgA antibodies may mitigate the effects of potentially protective antibodies. This immune-correlates study generated the hypotheses that V1V2 antibodies may have contributed to protection against HIV-1 infection, whereas high levels of Env-specific IgA antibodies may have mitigated the effects of protective antibodies. Vaccines that are designed to induce higher levels of V1V2 antibodies and lower levels of Env-specific IgA antibodies than are induced by the RV144 vaccine may have improved efficacy against HIV-1 infection.
Article
Full-text available
The vast majority of the world's population is infected with HSV. Although antiviral therapy can reduce the incidence of reactivation and asymptomatic viral shedding, and limit morbidity and mortality from active disease, it cannot cure infection. Therefore, the development of an effective vaccine is an important global health priority. In this study, we demonstrate that recombinant Listeria monocytogenes (Lm) expressing the H-2K(b) glycoprotein B (gB)(498-505) peptide from HSV-1 triggers a robust CD8 T cell response to this Ag resulting in protective immunity to HSV infection. Following challenge with HSV-1, immune-competent mice primed with recombinant Lm-expressing gB(498-505) Ag were protected from HSV-induced paralysis. Protection was associated with dramatic reductions in recoverable virus, and early expansion of HSV-1-specific CD8 T cells in the regional lymph nodes. Thus, recombinant Lm-expressing Ag from HSV represents a promising new class of vaccines against HSV infection.
Article
Full-text available
We have evaluated an attenuated Listeria monocytogenes (Lm) candidate vaccine vector in nonhuman primates using a delivery regimen relying solely on oral vaccination. We sought to determine the impact of prior Lm vector exposure on the development of new immune responses against HIV antigens. Two groups of rhesus macaques one Lm naive, the other having documented prior Lm vector exposures, were evaluated in response to oral inoculations of the same vector expressing recombinant HIV-1 Gag protein. The efficacy of the Lm vector was determined by ELISA to assess the generation of anti-Listerial antibodies; cellular responses were measured by HIV-Gag specific ELISpot assay. Our results show that prior Lm exposures did not diminish the generation of de novo cellular responses against HIV, as compared to Listeria-naïve monkeys. Moreover, empty vector exposures did not elicit potent antibody responses, consistent with the intracellular nature of Lm. The present study demonstrates in a pre-clinical vaccine model, that prior oral immunization with an empty Lm vector does not diminish immunogenicity to Lm-expressed HIV genes. This work underscores the need for the continued development of attenuated Lm as an orally deliverable vaccine.
Article
Full-text available
Recombinant live-attenuated Listeria monocytogenes is currently being developed as a vaccine platform for treatment or prevention of malignant and infectious diseases. The effectiveness of complex biologic vaccines, such as recombinant viral and bacterial vectors, can be limited by either preexisting or vaccine-induced vector-specific immunity. We characterized the level of L. monocytogenes-specific cellular and humoral immunity present in more than 70 healthy adult subjects as a first step to understanding its possible impact on the efficacy of L. monocytogenes-based vaccines being evaluated in early-phase clinical trials. Significant L. monocytogenes-specific humoral immunity was not measured in humans, consistent with a lack of antibodies in mice immunized with wild-type L. monocytogenes. Cellular immune responses specific for listeriolysin O, a secreted bacterial protein required for potency of L. monocytogenes-derived vaccines, were detected in approximately 60% of human donors tested. In mice, while wild-type L. monocytogenes did not induce significant humoral immunity, attenuated L. monocytogenes vaccine strains induced high-titer L. monocytogenes-specific antibodies when given at high doses used for immunization. Passive transfer of L. monocytogenes-specific antiserum to naïve mice had no impact on priming antigen-specific immunity in mice immunized with a recombinant L. monocytogenes vaccine. In mice with preexisting L. monocytogenes-specific immunity, priming of naïve T cells was not prevented, and antigen-specific responses could be boosted by additional vaccinations. For the first time, our findings establish the level of L. monocytogenes-specific cellular immunity in healthy adults, and, together with modeling studies performed with mice, they support the scientific rationale for repeated L. monocytogenes vaccine immunization regimens to elicit a desired therapeutic effect.
Article
Full-text available
Neutralizing antibodies are thought to be required at mucosal surfaces to prevent human papillomavirus (HPV) transmission. However, the potential for cell-mediated immunity in mediating protection against HPV infection has not been well explored. We generated recombinant Listeria monocytogenes (Lm) constructs that secrete listeriolysin O (LLO) fused with overlapping N-terminal (LLO-L1(1-258)) or C-terminal (LLO-L1(238-474)) fragments of HPV type 16 major capsid protein L1 (HPV-16-L1). Oral immunization of mice with either construct induced IFN-gamma-producing CD8+ and CD4+ T cells in the spleen and in the Peyer's patches with the C-terminal construct. Oral immunization with both constructs resulted in diminished viral titers in the cervix and uterus of mice after intravaginal challenge with vaccinia virus expressing HPV-16-L1.
Article
Containment of the acquired immunodeficiency syndrome (AIDS) epidemic will require an effective human immunodeficiency virus type 1 (HIV-1) vaccine. Accumulating evidence suggests that such a vaccine must efficiently elicit an HIV-1–specific cytotoxic T lymphocyte (CTL) response. Nonhuman primate models will continue to provide an important tool for assessing the extent of protective immunity induced by various immunization strategies. Although replication-competent AIDS viruses attenuated for pathogenicity by selective gene deletions have provided protective immunity in nonhuman primate models, the long-term safety of such vaccines in human populations is suspect. Inactivated virus and subunit vaccines have elicited neither CTLs nor antibodies capable of neutralizing a wide array of patient HIV-1 isolates. Considerable effort is now being focused on evaluating live vector-based vaccine and plasmid DNA vaccine approaches for preventing HIV-1 infection both in animal model and human studies. Our growing understanding of the biology of HIV-1 and immune responses to this virus will continue to suggest improved vaccination approaches for exploration.
Article
Listeria monocytogenes (Lm) is known to induce strong cellular immune responses. We constructed a live-attenuated Lm vector, Lmdd-BdopSIVgag, which encodes SIVmac239 gag. Intragastric (i.g.) administration of 3 × 10(12) bacteria to rhesus macaques was safe and induced anti-Gag cellular but no humoral immune responses. Boosting of Gag-specific cellular responses was observed after i.g. administration of Lmdd-BdopSIVgag to previously vaccinated RM despite preexisting anti-Lm immunity shown by lymphoproliferative responses. Surprisingly, anti-Lm cellular responses were also detected in non-vaccinated controls, which may reflect the fact that Lm is a ubiquitous bacterium. The novel, live-attenuated Lmdd-BdopSIVgag may be an attractive platform for oral vaccine delivery.
Article
Host protection to helminth infection requires IL-4 receptor α chain (IL-4Rα) signalling and the establishment of finely regulated Th2 responses. In the current study, the role of IL-4Rα-responsive T cells in Schistosoma mansoni egg-induced inflammation was investigated. Egg-induced inflammation in IL-4Rα-responsive BALB/c mice was accompanied with Th2-biased responses, whereas T-cell-specific IL-4Rα-deficient BALB/c mice (iLck(cre)Il4ra(-) (/lox)) developed Th1-biased responses with heightened inflammation. The proportion of Foxp3(+) Treg in the draining LN of control mice did not correlate with the control of inflammation and was reduced in comparison to T-cell-specific IL-4Rα-deficient mice. This was due to IL-4-mediated inhibition of CD4(+)Foxp3(+) Treg conversion, demonstrated in adoptively transferred Rag2(-) (/) (-) mice. Interestingly, reduced footpad swelling in Il4ra(-) (/lox) mice was associated with the induction of IL-4 and IL-10-secreting CD4(+)CD25(-)CD103(+)Foxp3(-) cells, confirmed in S. mansoni infection studies. Transfer of IL-4Rα-responsive CD4(+)CD25(-)CD103(+) cells, but not CD4(+)CD25(high) or CD4(+)CD25(-)CD103(-) cells, controlled inflammation in iLck(cre)Il4ra(-) (/lox) mice. The control of inflammation depended on IL-10, as transferred CD4(+)CD25(-)CD103(+) cells from IL-10-deficient mice were not able to effectively downregulate inflammation. Together, these results demonstrate that IL-4 signalling in T cells inhibits Foxp3(+) Treg in vivo and promotes CD4(+)CD25(-)CD103(+)Foxp3(-) cells that control S. mansoni egg-induced inflammation via IL-10.