ArticlePDF Available

Ebola Virus‐Like Particle–Based Vaccine Protects Nonhuman Primates against Lethal Ebola Virus Challenge

Authors:

Abstract

Currently, there are no licensed vaccines or therapeutics for the prevention or treatment of infection by the highly lethal filoviruses, Ebola virus (EBOV) and Marburg virus (MARV), in humans. We previously had demonstrated the protective efficacy of virus-like particle (VLP)-based vaccines against EBOV and MARV infection in rodents. To determine the efficacy of vaccination with Ebola VLPs (eVLPs) in nonhuman primates, we vaccinated cynomolgus macaques with eVLPs containing EBOV glycoprotein (GP), nucleoprotein (NP), and VP40 matrix protein and challenged the macaques with 1000 pfu of EBOV. Serum samples from the eVLP-vaccinated nonhuman primates demonstrated EBOV-specific antibody titers, as measured by enzyme-linked immunosorbent assay, complement-mediated lysis assay, and antibody-dependent cell-mediated cytotoxicity assay. CD44+ T cells from eVLP-vaccinated macaques but not from a naive macaque responded with vigorous production of tumor necrosis factor- alpha after EBOV-peptide stimulation. All 5 eVLP-vaccinated monkeys survived challenge without clinical or laboratory signs of EBOV infection, whereas the control animal died of infection. On the basis of safety and efficacy, eVLPs represent a promising filovirus vaccine for use in humans.
S430 JID 2007:196 (Suppl 2) Warfield et al.
SUPPLEMENT ARTICLE
Ebola Virus-Like Particle–Based Vaccine Protects
Nonhuman Primates against Lethal Ebola Virus
Challenge
Kelly L. Warfield,
a
Dana L. Swenson,
a
Gene G. Olinger, Warren V. Kalina, M. Javad Aman,
b
and Sina Bavari
US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
Background. Currently, there are no licensed vaccines or therapeutics for the prevention or treatment of
infection by the highly lethal filoviruses, Ebola virus (EBOV) and Marburg virus (MARV), in humans. We previously
had demonstrated the protective efficacy of virus-like particle (VLP)–based vaccines against EBOV and MARV
infection in rodents.
Methods. To determine the efficacy of vaccination with Ebola VLPs (eVLPs) in nonhuman primates, we
vaccinated cynomolgus macaques with eVLPs containing EBOV glycoprotein (GP), nucleoprotein (NP), and VP40
matrix protein and challenged the macaques with 1000 pfu of EBOV.
Results. Serum samples from the eVLP-vaccinated nonhuman primates demonstrated EBOV-specific antibody
titers, as measured by enzyme-linked immunosorbent assay, complement-mediated lysis assay, and antibody-
dependent cell-mediated cytotoxicity assay. CD44
+
T cells from eVLP-vaccinated macaques but not from a naive
macaque responded with vigorous production of tumor necrosis factor–aafter EBOV-peptide stimulation. All 5
eVLP-vaccinated monkeys survived challenge without clinical or laboratory signs of EBOV infection, whereas the
control animal died of infection.
Conclusion. On the basis of safety and efficacy, eVLPs represent a promising filovirus vaccine for use in
humans.
During the past 20 years, owing to advances in molec-
ular biology and in the understanding of basic virology,
scientists have been able to develop subunit vaccines
based on virus-like particles (VLPs). VLPs for many
viruses have been developed and are based on the
knowledge that expression of specific viral structural
proteins results in the self-assembly of particles that
morphologically resemble the authentic virus [1]. Some
Potential conflicts of interest: K.L.W., D.L.S., M.J.A., and S.B. hold patent rights
to Ebola and Marburg virus-like particle–based vaccines. G.G.O. and W.V.K.: none
reported.
Presented in part: Filoviruses: Recent Advances and Future Challenges,
International Centre for Infectious Diseases Symposium, Winnipeg, Manitoba,
Canada, 17–19 September 2006.
Financial support: Defense Threat Reduction Agency. Supplement sponsorship
is detailed in the Acknowledgments.
Opinions, interpretations, conclusions, and recommendations are those of the
authors and are not necessarily endorsed by the US Army.
a
K.L.W. and D.L.S. contributed equally to this work.
b
Present affiliation: Integrated BioTherapeutics Inc., Frederick, Maryland.
Reprints or correspondence: Dr. Sina Bavari or Dr. Kelly Warfield, US Army
Medical Research Institute of Infectious Diseases, 1425 Porter St., Fort Detrick,
MD 21702 (sina.bavari@us.army.mil or kelly.warfield@us.army.mil).
The Journal of Infectious Diseases 2007;196:S430–7
This article is in the public domain, and no copyright is claimed.
0022-1899/2007/19610S2-0043
DOI: 10.1086/520583
of the many advantages of using VLPs as vaccines include
(1) their similar morphology to the live enveloped or
nonenveloped viruses from which they are derived; (2)
a strong safety profile, since they are nonreplicating; (3)
no concerns regarding viral vector or preexisting anti-
vector immunity; (4) the fact that they can be generated
in large quantities by use of mammalian or insect cell
lines; (5) their ability to generate innate, humoral, and
cellular immunity; and (6) the fact that they have been
safely and effectively administered in humans [1–7].
We and others have demonstrated previously the
generation of enveloped Ebola VLPs (eVLPs) in mam-
malian and insect cell-expression systems [8–13]. VLPs
containing glycoprotein (GP) and VP40 derived from
Ebola virus (EBOV) have been used successfully to vac-
cinate rodents [2, 13–17]. Both BALB/c and C57BL/6
mice have been protected against a range of challenge
doses (10–1,000 pfu or 300–30,000 LD
50
) by means
of dose-dependent eVLP vaccination in the presence or
absence of adjuvant [2, 14, 15]. Addition of saponin-
derived QS-21 or RIBI adjuvant to the eVLP-vaccine
regimen allows administration of a decreased dose of
the vaccine and completely protects mice and guinea
by guest on September 20, 2014http://jid.oxfordjournals.org/Downloaded from
VLP-Based Vaccine in Nonhuman Primates JID 2007:196 (Suppl 2) S431
pigs against challenge, even after only 1 inoculation (authors’
unpublished data) [18]. eVLP vaccination completely prevents
viremia and clinical symptoms after EBOV challenge in rodents
but does not induce sterile immunity, as evidenced by an ex-
pansion of immune responses to viral proteins not present in
the vaccine, after challenge [2, 14, 15, 18]. However, the ques-
tion of whether eVLPs would be able to protect nonhuman
primates against EBOV infection has remained. Therefore, the
goal of the current study was to determine whether eVLPs
would be viable vaccine candidates for the protection of pri-
mates against lethal EBOV challenge.
MATERIALS AND METHODS
Generation and characterization of eVLPs. 293T-derived
eVLPs containing EBOV GP, nucleoprotein (NP), and VP40
were prepared essentially as described elsewhere [9, 14, 16, 17].
Total protein concentrations in the vaccine preparations were
determined in the presence of detergent, by use of a Bradford
protein assay (BioRad). Expression of GP, NP, and VP40 in
each vaccine preparation was verified by Western blot analysis
[9, 17, 18]. eVLPs were processed and imaged via electron
microscopy, as described elsewhere [9, 14, 16, 17]. Endotoxin
levels in all eVLP preparations used in this study were !0.03
endotoxin units/mg, as determined by the Limulus amebocyte
lysate test (Invitrogen).
Animals. In testing before the start of this study, the cy-
nomolgus macaques used were found to be antibody negative
for filovirus, simian T cell leukemia virus–1, simian immu-
nodeficiency virus, and herpes B virus. The eVLP-vaccinated
monkeys received 3 intramuscular injections, at 42-day inter-
vals, of 1.0 mL of sterile saline containing 250 mg of eVLPs
and 0.5 mL of RIBI adjuvant (Corixa). Blood samples were
obtained from the femoral vein of monkeys under anesthesia.
Female cynomolgus macaques of 3–4 kg in weight were chal-
lenged by intramuscular injection of 1000 pfu of Zaire EBOV
(ZEBOV; 1995 outbreak strain) [19]. Viremia was determined
by means of a traditional plaque assay [20]. Hematology and
kidney- and liver-associated enzymes were assessed as described
elsewhere [21]. For ethical reasons, the use of relevant historical
control animals was required by the Laboratory Animal Care
and Use Committee of the US Army Medical Research Institute
of Infectious Diseases (Fort Detrick, MD), to reduce the num-
ber of nonhuman primates needed in these studies. For this
reason, the control monkey in the current study was not treated,
so that, for the data analysis, results for this monkey could be
combined with those for 23 historical control animals chal-
lenged with the same seed stock of ZEBOV at the same dose
and via the same route.
All EBOV-infected animals and their samples were handled
under maximum containment in a biosafety level 4 laboratory
at the US Army Medical Research Institute of Infectious Dis-
eases (Fort Detrick, MD). Research was conducted in compli-
ance with the Animal Welfare Act and other federal statutes
and regulations relating to animals and experiments involving
animals and adhered to principles stated in the Guide for the
Care and Use of Laboratory Animals [22]. The facility where
this research was conducted is fully accredited by the Associ-
ation for Assessment and Accreditation of Laboratory Animal
Care International (Rockville, MD).
Total antibody responses against EBOV. Levels of EBOV-
specific antibodies were determined from serum or plasma sam-
ples by use of ELISA, as described elsewhere [14]. Antibody
titers were defined as the reciprocal of the highest dilution
giving a net optical density value 0.2.
Determination of complement-mediated lysis by EBOV GP–
specific antibody. Antibodies recognizing viral antigens on
infected cells can bind complement via their Fc region and can
initiate activation of the complement cascade, resulting in kill-
ing of the virus-infected cell. To determine whether eVLP vac-
cination generated antibodies capable of inducingcomplement-
mediated lysis, Vero cells were infected at an MOI of 10 with
EBOV GP–expressing Venezuelan equine encephalitis virus
(VEE) replicons, produced as described elsewhere [23], and
were incubated in a humidified, 5% CO
2
incubator at 37Cfor
16 h. After incubation, Vero cells were removed from the plate
by means of trypsinization and were labeled with 100 mCi of
51
Cr for 1 h. Cells were washed 3 times in RPMI 1640 without
fetal bovine serum (FBS) and were resuspended to 100,000cells/
mL in RPMI 1640 containing 10% FBS. Next, 100 mLofVero
cells were plated in each well of a 96-well plate containing
various dilutions of plasma samples from eVLP-vaccinated and
control monkeys. Then, low-endotoxin guinea pig complement
was added, at a final dilution of 1: 20, and samples were in-
cubated for 3 h. The amount of radioactivity released into the
supernatants was determined with a g-radiation counter. Spon-
taneous lysis was measured in VEE replicon–expressing
EBOV GP–infected cells [23] with complement added but no
antibodies present, and total lysis was measured in Vero cells
incubated with 1% Triton X-100. Percentage of specific
lysis was calculated as [(experimental releasespontaneous
.
release)/(maximum release spontaneous release)]10 0
Antibody-dependent cell-mediated cytotoxicity (ADCC)
assay. ADCC occurs when virus-specific antibodies coat tar-
get infected cells and make them vulnerable to killing during
coculture with other cytolytic cells. To determine whether an-
tibodies stimulated by eVLP vaccination are capable ofinducing
ADCC, Vero cells were infected with VEE replicon–expressing
ZEBOV GP for 16 h and were labeled with 100 mCi of
51
Cr, as
described above. Labeled Vero cells were plated in a 96-well
plate (10,000 cells/well). Primate plasma from previously vac-
cinated or naive (negative control) animals was added at various
dilutions to labeled, GP-expressing Vero cells. Purified effector
by guest on September 20, 2014http://jid.oxfordjournals.org/Downloaded from
S432 JID 2007:196 (Suppl 2) Warfield et al.
Figure 1. Antibody responses in nonhuman primates vaccinated with
Ebola virus-like particles (eVLPs) containing glycoprotein (GP), nucleopro-
tein, and VP40. Cynomolgus macaques were vaccinated 3 times, at 6-
week intervals, with 250 mg of eVLPs in RIBI adjuvant. A, Total anti–
Ebola virus (EBOV) antibody titers for individual animals (VLP1–5), as
determined by ELISA. B, Induction of complement-mediated lysis indicated
in plasma samples from eVLP-vaccinated macaques. EBOV GP–expressing
Vero cells were incubated with guinea pig complement and the indicated
dilutions of plasma. The percentage lysis was determined as compared
with that in untreated cells. C, Results of lysis of EBOV GP–expressing
target cells by an antibody-dependent cell-mediated cytotoxicity (ADCC)
assay. Target cells incubated with plasma from eVLP-vaccinated macaques
or an unvaccinated animal (naive) and human effector cells (effector-to-
target cell ratio, 40:1) showed various levels of ADCC antibody titers.
Error bars indicate SDs.
cells (peripheral blood mononuclear cells [PBMCs]), resus-
pended in RPMI 1640 with 10% FBS, were added to antibody-
coated target cells at the following effector-to-target cell (ET)
ratios: 1: 10, 1: 20, 1:40, and 1:80. Optimization of the assay
was determined by choosing the ET ratio that produced the
least background in wells with no antibody or with plasma
from unvaccinated animals. Each plate was incubated for 4 h
at 37C in the presence of 5% CO
2
. After 4 h, centrifugation
of each plate was done at 250 gto pellet the cells, 50 mLof
supernatant was removed, and
51
Cr released into supernatant
was quantified by use of a g-radiation counter. Spontaneous
lysis was measured in VEE replicon–expressing EBOV GP–in-
fected cells [23] with effector cells added but no antibodies
present, and total lysis was measured in Vero cells incubated
with 1% Triton X-100. Percentage of specific lysis was calculated
as [(experimental release spontaneous release)/(maximum
.release spontaneous release)]10 0
T cell responses to EBOV peptides in eVLP-vaccinated non-
human primates. Epitopes recognized by circulating CD4
+
and CD8
+
T cells were determined as described elsewhere [15,
24], with several minor modifications. In brief, EBOV-specific
responses were analyzed by culturing PBMCs with 1–5 mgof
overlapping 15-residue peptides representing EBOV GP, NP, or
VP40 (Mimotopes) or 1 mg/mL staphylococcal enterotoxin B
(SEB) in complete RPMI 1640 containing 10 mg/mL brefeldin
A. After 18 h of culture, the cells were stained with anti-CD44,
-CD8, or -CD4 (Pharmingen) in brefeldin A. After cell-surface
staining, cells were fixed in 1% formaldehyde, permeabilized
with saponin, and stained with anti–tumor necrosis factor
(TNF)–aphycoerythrin (Pharmingen). Samples with an in-
crease in the frequency of TNF-a–positive cells of 12-fold above
background, as assessed by no peptide stimulation or by re-
sponse to an irrelevant peptide (Lassa virus N: RPLSA-
GVYMGNLSSQ), were considered to be positive.
RESULTS
Humoral responses to EBOV infection, after eVLP vaccination.
To determine whether eVLP vaccination elicits humoral re-
sponses in nonhuman primates, total antibody responses in
blood from the eVLP-vaccinated macaques were determined,
by ELISA using irradiated whole EBOV virions, immediately
before each vaccination and before challenge (figure 1A). Total
EBOV-specific antibodies in the eVLP-vaccinated macaques
rose 3–10-fold after the first vaccination and plateaued after 2
vaccinations (figure 1A).
Neutralizing titers were observed in the eVLP-vaccinated ma-
caques, consistent with our results in previous studies of rodents
[14, 16], with 80% plaque reduction/neutralization titers rang-
ing from 1:20 to 1:160 (data not shown). Additional studies
of antibody function revealed that eVLP vaccination of non-
human primates elicited both complement-mediated lysis and
ADCC (figure 1Band 1C). Although the interaction between
antibodies and antigen provides the specificity of the response,
the complement system is likely to provide the actual protective
response by destroying antigen-coated cells. Antibodies from
the eVLP-vaccinated animals were able to induce complement-
mediated lysis of cells expressing EBOV GP, in a dilution-de-
pendent manner (figure 1B). ADCC caused by EBOV GP–
specific antibodies coating target infected cells, making them
vulnerable to killing during coculture with other cytolytic cells,
by guest on September 20, 2014http://jid.oxfordjournals.org/Downloaded from
VLP-Based Vaccine in Nonhuman Primates JID 2007:196 (Suppl 2) S433
Figure 2. Vigorous CD44
+
T cell response to Ebola virus (EBOV) glycoprotein (GP) and nucleoprotein (NP) peptides, in nonhuman primates vaccinated
with Ebola virus-like particles (eVLPs). Peripheral blood leukocytes from eVLP-vaccinated cynomolgus macaques were collected 10 days after vaccination
and were used ex vivo for identification of peptides that induced intracellular tumor necrosis factor (TNF)–ain CD4
+
/CD44
+
or CD8
+
/CD44
+
T cells.
Percentages of TNF-a–producing cells that were 12-fold higher than the background percentage (no peptide or irrelevant peptide) were considered
to be positive. Representative responses to EBOV GP and NP peptides in an eVLP-vaccinated animal (VLP4) and a naive control animal are shown.
SEB, staphylococcal enterotoxin B.
also was observed when serum samples from the eVLP-vac-
cinated monkeys were used (figure 1C).
Cellular immune responses of eVLP-vaccinated nonhuman
primates. Representative flow-cytometry data are shown for
the naive control animal and for monkey VLP4, at 10 days
after their third vaccination (figure 2). Although this particular
eVLP-vaccinated monkey appeared to have the lowest total
antibody titers, as assessed by ELISA (figure 1A), it demon-
strated strong T cell responses to EBOV-peptide stimulation
(figures 2 and 3). In figure 2, we plotted responses from both
the naive and the eVLP-vaccinated monkeys and also show
TNF-aproduction in T cells stimulated overnight with SEB,
irrelevant peptide, or EBOV-specific peptides. The SEB-stim-
ulated cells were used as a positive control for T cell activation
and cytokine secretion, while several wells with no peptide or
an irrelevant peptide (such as Lassa virus N [15, 24]) were
included as negative controls, to determine the assay back-
ground (figure 2). Interestingly, specific pools from the majority
of the eVLP-vaccinated macaques were recognized, such as GP
pools 9 and 10 and NP pools 3, 4, and 11 (figure 3Aand 3B).
However, epitopes within the VP40 matrix protein were not
strongly recognized in samples from the eVLP-vaccinated an-
imals (figure 3C).
Protection of nonhuman primates against EBOV infection,
by eVLP vaccination. All 5 eVLP-vaccinated monkeys and
the single naive control monkey were challenged with 1000
pfu of ZEBOV at 4 weeks after the last vaccination (figure 4A).
As typically observed in cynomolgus macaques, the control
monkey developed clinical and laboratory signs of EBOV in-
fection beginning 4–5 days after challenge and died from the
disease 6 days after challenge (figures 4 and 5). Results for a
cohort of 23 naive control monkeys infected with the same
challenge virus via the same means are shown in figure 4A, to
indicate the normal course of disease and the mean time to
death after EBOV challenge. The eVLP-vaccinated monkeys
were completely protected against disease after lethal EBOV
challenge. We observed no signs of clinical disease, such as rash,
anorexia, or weight loss, in any of the eVLP-vaccinated mon-
keys. In addition, viremia was not detected by standard plaque
assay at any of the time points when measurements were taken
by guest on September 20, 2014http://jid.oxfordjournals.org/Downloaded from
S434 JID 2007:196 (Suppl 2) Warfield et al.
Figure 3. Cellular immune responses of nonhuman primates vaccinated
with Ebola virus-like particles (eVLPs) to Ebola virus (EBOV)–based pep-
tides. Peripheral blood mononuclear cells from eVLP-vaccinated animals
(VLP1–5) were collected 10 days after vaccination and were used ex vivo
for identification of peptides from EBOV glycoprotein (GP; A), nucleoprotein
(NP; B), and VP40 (C) that induced CD4
+
/CD44
+
or CD8
+
/CD44
+
T cells
expressing tumor necrosis factor (TNF)–a. Percentages of TNF-a–pro-
ducing cells that were 12-fold higher than the background percentage
(dashed line) were considered to be positive. All cells were tested against
duplicate pools of peptides, and responses to 1 set are shown here.
Figure 4. Protection of nonhuman primates vaccinated with Ebola vi-
rus-like particles (eVLPs) containing glycoprotein, nucleoprotein,andVP40.
Cynomolgus macaques ( ) were vaccinated 3 times with eVLPs innp5
RIBI adjuvant. A, Survival after challenge with 1000 pfu of Zaire Ebola
virus (ZEBOV), assessed 4 weeks after the last vaccination. For the pur-
poses of representation on the Kaplan-Meier plot, results for the control
monkey in the current study were combined with those for 23 historical
control animals challenged with the same seed stock of ZEBOV, at the
same dose and via the same route. B, Virus titers in the plasma of ZEBOV-
challenged eVLP-vaccinated monkeys were determined by use of a stan-
dard plaque assay. The laboratory values for the naive control monkey
are shown from an unscheduled blood sample at 6 days after challenge,
obtained immediately before euthanasia. C, Rectal temperature after chal-
lenge. Data are presented as the mean value for the eVLP-vaccinated
animals and the single control animal. Error bars indicate SDs.
(figure 4B). We did observe a mild elevation (2F–3F) in core
body temperature in 2 eVLP-vaccinated animals on day 8 after
challenge, although this increase was transient and not typical
of the high fever usually observed in filovirus-infected monkeys
(figure 4C).
We observed only minor hematologic and liver-enzyme
changes in the eVLP-vaccinated monkeys after EBOV challenge
(figure 5). Liver enzymes, such as aspartate transaminase, al-
kaline phosphatase, and alanine transaminase, were not altered
after EBOV infection of the eVLP-vaccinated monkeys (figure
5A–5C). The numbers of circulating platelets did not decrease
in the eVLP-vaccinated monkeys after EBOV challenge (figure
5D). Not surprisingly, total white blood cell (WBC) count and
percentage of lymphocytes in the blood were slightly more
variable in all the animals after challenge (figure 5Eand 5F).
As expected, the naive control animal in this experiment ex-
hibited an increase in total WBC count, with a concomitant
decrease in the percentage of circulating lymphocytes (figure
5Eand 5F).
DISCUSSION
Numerous vaccine approaches have been used against lethal
filoviral infections, including classic vaccine preparations of in-
by guest on September 20, 2014http://jid.oxfordjournals.org/Downloaded from
VLP-Based Vaccine in Nonhuman Primates JID 2007:196 (Suppl 2) S435
Figure 5. Clinical laboratory results for nonhuman primates vaccinated with Ebola virus-like particles (eVLPs) containing glycoprotein, nucleoprotein,
and VP40. Cynomolgus macaques ( ) were vaccinated 3 times with eVLPs in RIBI adjuvant. Liver function was assessed by analysis of serumnp5
samples from eVLP-vaccinated monkeys after challenge. Levels of aspartate transaminase (AST; A), alkaline phosphatase (ALP; B), and alanine
transaminase (ALT; C) were measured at the indicated time points. Platelet count (D), total white blood cell (WBC) count (E), and percentage of
lymphocytes (F) in blood also were assessed after challenge and are presented as the mean (SD) of the eVLP-vaccinated animals and the single
control animal. The laboratory values for the naive control monkey are shown from an unscheduled blood sample at 6 days after challenge, obtained
immediately before euthanasia.
activated or avirulent virus [25, 26]; viral vectors such as ad-
enovirus, VEE, paramyxovirus, vaccinia, and vesicular stoma-
titis virus that encode protective filoviral proteins [24, 26–29],
DNA [27, 30–32]; and other subunit vaccines [2, 26, 33]. In
this study, we have shown that VLPs are a promising vaccine
candidate for protection of nonhuman primates against lethal
EBOV infection. Some of the advantages of VLPs as filovirus
vaccines include presentation of antigen in its native form, an
excellent safety profile, no interference by a vector backbone,
lack of problems related to vector immunity, and induction of
strong antibody and T cell responses.
In our studies, we have found high levels of antibodies in
the serum of eVLP-vaccinated nonhuman primates, using 4
different assays. Not only did the vaccinated animals develop
high levels of total EBOV-specific antibodies, as determined by
ELISA, but they also developed antibodies that were highly
active in vitro. These antibodies induced virus-neutralizing
(data not shown), ADCC, and complement-mediated lysis ac-
tivity (figure 1Band 1C). Data suggest that our VLP vaccines
induced EBOV-specific antibodies that are multifunctional. On
the basis of our studies of nonhuman primates thus far (figure
1 and authors’ unpublished data), whether total levels of an-
tibodies correlate with protection against filovirus challenge is
not clear. We had shown previously that, although B cells are
by guest on September 20, 2014http://jid.oxfordjournals.org/Downloaded from
S436 JID 2007:196 (Suppl 2) Warfield et al.
absolutely required for eVLP-mediated protection in mice,
transfer of serum from eVLP-vaccinated mice to naive recip-
ients did not confer protection against EBOV infection, but
this result may have been dependent on the amount of antibody
transferred [15]. However, the role of B cells and antibodies in
protection against filoviral infection needs further investigation,
and their necessity for the protection of nonhuman primates
against EBOV infection is uncertain [34–38].
The eVLP-vaccinated monkeys developed strong T cell re-
sponses to EBOV epitopes, as assessed by intracellular cytokine
(TNF-a) staining of peptide-stimulated PBMCs. More re-
sponses to GP and NP were observed than to the VP40 matrix
protein (figure 3). Although responses to the peptides varied
among the monkeys, certain epitopes were recognized by more
than 1 monkey. Unfortunately, at this time, the reagents that
are available to describe the T cell responses of cynomolgus
macaques are limited, and further characterization of these re-
sponses may be required [39]. The importance of T cell re-
sponses in VLP-mediated protection against EBOV infection
has been demonstrated by use of knockout mice: CD4
+
knock-
out mice were protected only partially against EBOV infection,
and CD8
+
T cells were absolutely required [15]. On the basis
of our studies of mice, induction of both EBOV-specific an-
tibodies, to impede early viral infection and replication, and
cytotoxic T cells, to destroy virus-infected cells, was found to
be necessary for immunity and protection against EBOV in-
fection. Future studies of nonhuman primates and clinicaltrials
with humans will be required, to identify correlates of immunity
for VLPs and filoviral infection.
Proper glycosylation and presentation of viral proteins, as
well as vaccine dose, are critical factors for successful filovirus
vaccines [14, 27, 30, 33, 40]. We previously have shown that
eVLP-mediated protection against EBOV challenge is dose de-
pendent but that the addition of adjuvant can help reduce the
VLP dose and the number of injections required to mitigate
protection [14–16, 18, 41]. Kinetic studies of antibody titers
and T cell responses in eVLP-vaccinated nonhuman primates
indicated that 3 doses of VLPs did not appear to boost the
gross immune responses, compared with the responses ob-
served after 2 vaccines (figure 1 and data not shown). None-
theless, future studies are required in order to refine the VLP
vaccination schedule, dose, and requirement for adjuvant in
nonhuman primates. Studies of mice and guinea pigs have
indicated that protection after only 1 VLP vaccination is an
achievable goal [2, 18]. Currently, we also are examining the
role of a variety of adjuvants in enhancing VLP responses.
The eVLPs provided robust protection in thevaccinated non-
human primates. Significant clinical or laboratory signs of
EBOV infection, including detectable viremia, anorexia, or con-
siderably elevated levels of liver enzymes, were not found in
the eVLP-vaccinated monkeys. However, eVLP vaccination did
not induce sterile immunity against this lethal EBOV challenge.
A half-log increase in total anti-EBOV antibody titers was ob-
served in the eVLP-vaccinated monkeys at 28 days after chal-
lenge (figure 1A). Furthermore, we observed a broadening of
the T cell repertoire in the eVLP-vaccinated monkeys after
EBOV challenge. This was demonstrated by the recognition of
T cell epitopes in proteins not included in the vaccine, such as
VP24 and VP35, in lymphocytes after challenge but not before
challenge (data not shown). This observation was not surpris-
ing, since we previously had noted an increase in EBOV-specific
antibodies and the development of cytotoxic T lymphocytes
recognizing VP24 and VP35 in eVLP-vaccinated rodents after
challenge [14, 15, 18]. Since the vaccinated nonhuman primates
were completely protected against clinical signs and symptoms
of EBOV infection, the induction of sterile immunity by an
EBOV vaccine candidate does not seem to be critical.
Although VLPs produced in mammalian cells are highly im-
munogenic, we currently are exploring alternative strategies for
vaccine production, to accelerate the development process to-
ward the use of eVLPs and Marburg VLPs (mVLPs) in humans.
We and others have successfully generated eVLPs and mVLPs
in a baculovirus expression system [13, 41]. Both insect cell–
derived eVLPs and mVLPs were able to mature and activate
human myeloid dendritic cells (data not shown) [41]. Fur-
thermore, mice vaccinated with insect cell–derived eVLPs sur-
vived lethal challenge with mouse-adapted EBOV, suggesting
that the baculovirus-derived eVLPs are as effective as those
produced in mammalian cells [41].
Since our current data indicate that eVLPs are highly im-
munogenic in monkeys and stimulate virus-specific humoral
and cellular responses, our next major goal is to demonstrate
the efficacy of mVLPs against Marburg virus (MARV) infection
[16] and also of a mixture of eVLPs and mVLPs as a pan-
filovirus vaccine against both EBOV and MARV infections [18].
In addition, we currently are planning preclinical experiments
in preparation for future clinical trials with humans. Besides
use as a vaccine, VLPs also are being used to dissect innate
immune responses to filoviruses, with the goal of developing
immunotherapeutics for the treatment of EBOV and MARV
infections [42], as well as a safe surrogate model for the ex-
amination of filoviral replication, entry, and assembly [2].
Acknowledgments
We especially would like to thank M. A. Fernandez, as well as D. K.
Reed, S. Van Tongeren, K. Kuehl, A. Pace, N. Posten, J. Smith, C. Rice,
and J. Stockman, for excellent technical assistance.
Supplement sponsorship. This article was published as part of a sup-
plement entitled “Filoviruses: Recent Advances and Future Challenges,”
sponsored by the Public Health Agency of Canada, the National Institutes
of Health, the Canadian Institutes of Health Research, Cangene, CUH2A,
Smith Carter, Hemisphere Engineering, Crucell, and the International Cen-
tre for Infectious Diseases.
by guest on September 20, 2014http://jid.oxfordjournals.org/Downloaded from
VLP-Based Vaccine in Nonhuman Primates JID 2007:196 (Suppl 2) S437
References
1. Noad R, Roy P. Virus-like particles as immunogens. Trends Microbiol
2003; 11:438–44.
2. Warfield KL, Swenson DL, Demmin G, Bavari S. Filovirus-like particles
as vaccines and discovery tools. Expert Rev Vaccines 2005;4:429–40.
3. Stanley MA. Human papillomavirus vaccines. Curr Opin Mol Ther
2002; 4:15–22.
4. Furumoto H, Irahara M. Human papillomavirus (HPV) and cervical
cancer. J Med Invest 2002; 49:124–33.
5. Lo-Man R, Rueda P, Sedlik C, Deriaud E, Casal I, Leclerc C. A recom-
binant virus-like particle system derived from parvovirus as an efficient
antigen carrier to elicit a polarized Th1 immune response without ad-
juvant. Eur J Immunol 1998; 28:1401–7.
6. Estes MK, Ball JM, Crawford SE, et al. Virus-like particle vaccines for
mucosal immunization. Adv Exp Med Biol 1997; 412:387–95.
7. Sapp M, Volpers C, Streeck RE. Synthesis, properties and applicationsof
papillomavirus-like particles. Intervirology 1996; 39:49–53.
8. Noda T, Sagara H, Suzuki E, Takada A, Kida H, Kawaoka Y. Ebola virus
VP40 drives the formation of virus-like filamentous particles along with
GP. J Virol 2002; 76:4855–65.
9. Bavari S, Bosio CM, Wiegand E, et al. Lipid raft microdomains: a gateway
for compartmentalized trafficking of Ebola and Marburg viruses. J Exp
Med 2002; 195:593–602.
10. Yasuda J, Nakao M, Kawaoka Y, Shida H. Nedd4 regulates egress of Ebola
virus-like particles from host cells. J Virol 2003; 77:9987–92.
11. Licata JM, Johnson RF, Han Z, Harty RN. Contribution of Ebola virus
glycoprotein, nucleoprotein, and VP24 to budding of VP40 virus-like
particles. J Virol 2004; 78:7344–51.
12. Han Z, Boshra H, Sunyer JO, Zwiers SH, Paragas J, Harty RN. Bio-
chemical and functional characterization of the Ebola virus VP24 protein:
implications for a role in virus assembly and budding. J Virol 2003;77:
1793–800.
13. Ye L, Lin J, Sun Y, et al. Ebola virus-like particles produced in insect cells
exhibit dendritic cell stimulating activity and induce neutralizing anti-
bodies. Virology 2006; 351:260–70.
14. Warfield KL, Bosio CM, Welcher BC, et al. Ebola virus-like particles
protect from lethal Ebola virus infection. Proc Natl Acad Sci USA
2003; 100:15889–94.
15. Warfield KL, Olinger G, Deal EM, et al. Induction of humoral and CD8
+
T cell responses are required for protection against lethal Ebola virus
infection. J Immunol 2005; 175:1184–91.
16. Warfield KL, Swenson DL, Negley DL, Schmaljohn AL, Aman MJ, Bavari
S. Marburg virus-like particles protect guinea pigs from lethal Marburg
virus infection. Vaccine 2004;22:3495–502.
17. Swenson DL, Warfield KL, Kuehl K, et al. Generation of Marburg virus-
like particles by co-expression of glycoprotein and matrix protein. FEMS
Immunol Med Microbiol 2004; 40:27–31.
18. Swenson DL, Warfield KL, Negley DL, Schmaljohn A, Aman MJ, Bavari
S. Virus-like particles exhibit potential as a pan-filovirus vaccine for both
Ebola and Marburg viral infections. Vaccine 2005;23:3033–42.
19. Jahrling PB, Geisbert TW, Geisbert JB, et al. Evaluation of immune
globulin and recombinant interferon-2b for treatment of experimental
Ebola virus infections. J Infect Dis 1999; 179(Suppl 1):S224–34.
20. Moe JB, Lambert RD, Lupton HW. Plaque assay for Ebola virus. J Clin
Microbiol 1981; 13:791–3.
21. Geisbert TW, Hensley LE, Jahrling PB, et al. Treatment of Ebola virus
infection with a recombinant inhibitor of factor VIIa/tissue factor: a study
in rhesus monkeys. Lancet 2003; 362:1953–8.
22. Committee on the Care and Use of Laboratory Animals of the Institute
of Laboratory Animals Resources Commission of Life Sciences, National
Research Council. Guide for the care and use of laboratory animals.
Washington, DC: National Academy Press, 1996.
23. Pushko P, Bray M, Ludwig GV, et al. Recombinant RNA replicons derived
from attenuated Venezuelan equine encephalitis virus protect guinea pigs
and mice from Ebola hemorrhagic fever virus. Vaccine 2000;19:142–53.
24. Olinger GG, Bailey MA, Dye JM, et al. Protective cytotoxic T-cell re-
sponses induced by Venezuelan equine encephalitis virus replicons ex-
pressing Ebola virus proteins. J Virol 2005;79:14189–96.
25. Geisbert TW, Pushko P, Anderson K, Smith J, Davis KJ, Jahrling PB.
Evaluation in nonhuman primates of vaccines against Ebola virus.Emerg
Infect Dis 2002; 8:503–7.
26. Hevey M, Negley D, Vanderzanden L, et al. Marburg virus vaccines:
comparing classical and new approaches. Vaccine 2001; 20:586–93.
27. Sullivan NJ, Geisbert TW, Geisbert JB, et al. Immune protection of non-
human primates against Ebola virus with single low-dose adenovirus
vectors encoding modified GPs. PLoS Med 2006; 3:865–77.
28. Bukreyev A, Yang L, Zaki SR, et al. A single intranasal inoculation with
a paramyxovirus-vectored vaccine protects guinea pigs against a lethal-
dose Ebola virus challenge. J Virol 2006;80:2267–79.
29. Jones SM, Feldmann H, Stroher U, et al. Live attenuated recombinant
vaccine protects nonhuman primates against Ebola and Marburg viruses.
Nat Med 2005; 11:786–90.
30. Dowling W, Thompson E, Badger C, et al. Influences of glycosylation
on antigenicity, immunogenicity, and protective efficacy of Ebola virus
GP DNA vaccines. J Virol 2007; 81:1821–37.
31. Riemenschneider J, Garrison A, Geisber t J, et al. Comparisonof individual
and combination DNA vaccines for B. anthracis, Ebola virus, Marburg
virus and Venezuelan equine encephalitis virus. Vaccine2003; 21:4071–80.
32. Vanderzanden L, Bray M, Fuller D, et al. DNA vaccines expressing either
the GP or NP genes of Ebola virus protect mice from lethal challenge.
Virology 1998; 246:134–44.
33. Hevey M, Negley D, Geisbert J, Jahrling P, Schmaljohn A. Antigenicity
and vaccine potential of Marburg virus glycoprotein expressed by bac-
ulovirus recombinants. Virology 1997; 239:206–16.
34. Wilson JA, Bray M, Bakken R, Hart MK. Vaccine potential of Ebola virus
VP24, VP30, VP35, and VP40 proteins. Virology 2001;286:384–90.
35. Wilson JA, Hevey M, Bakken R, et al. Epitopes involved in antibody-
mediated protection from Ebola virus. Science 2000; 287:1664–6.
36. Hevey M, Negley D, Schmaljohn A. Characterization of monoclonal
antibodies to Marburg virus (strain Musoke) glycoprotein and identifi-
cation of two protective epitopes. Virology 2003; 314:350–7.
37. Takada A, Ebihara H, Jones S, Feldmann H, Kawaoka Y. Protectiveefficacy
of neutralizing antibodies against Ebola virus infection. Vaccine 2007; 25:
993–9.
38. Oswald WB, Geisbert TW, Davis KJ, et al. Neutralizing antibody fails to
impact the course of Ebola virus infection in monkeys. PLoS Pathog
2007; 3:62–6.
39. Watkins DI. The evolution of major histocompatibility class I genes in
primates. Crit Rev Immunol 1995; 15:1–29.
40. Hevey M, Negley D, Pushko P, Smith J, Schmaljohn A. Marburg virus
vaccines based upon alphavirus replicons protect guinea pigs and non-
human primates. Virology 1998; 251:28–37.
41. Warfield KL, Posten NA, Swenson DL, et al. Filovirus-like particles pro-
duced in insect cells: immunogenicity and protection in rodents. J Infect
Dis 2007; 196(Suppl 2):S421–9 (in this supplement).
42. Warfield KL, Perkins JG, Swenson DL, et al. Role of natural killer cells
in innate protection against lethal Ebola virus infection. J Exp Med
2004; 200:169–79.
by guest on September 20, 2014http://jid.oxfordjournals.org/Downloaded from
... In mouse and guinea pig efficacy tests, VLPs made of VP40 and GP provided 100 percent protection from deadly ZEBOV infection [6]. Three vaccinations of non-human primates (NHPs) with VLPs containing glycoprotein, nucleoprotein, VP40, and an adjuvant (RIBI) resulted in immunological responses in the animals which were protective against deadly ZEBOV [7][8][9]. ...
... ZEBOV and Marburg virus (MARV) were tested in guinea pig animal models utilizing chimeric VLPs and vice versa for the cross-protection as well as a combination of ZEBOV-and MARV-like particles on this platform. The findings confirmed the necessity of GP for protection and the efficacy of blended VLPs over chimeric VLPs [7][8][9]. ...
Article
Full-text available
The global outgoing outbreaks of Ebola virus disease (EVD) in different regions of Sudan, Yuganda and Western Africa has focused the inadequacies and restrictions of pre-designed vac-cines to battle against the EVD, which has affirmed the emergency for the development of a systematic protocol to produce Ebola vaccines prior to the outbreak. There are several vaccines available being developed by preclinical trials and human based clinical trials. The group of vac-cines includes Viral particles similar vaccines, DNA based vaccines, whole virus recombinant vaccines, incompetent replication originated vaccines, and competent replication vaccines. The limitations and challenges faced in development of Ebola vaccines are selection of Immunogen, rapid-responsiveness, cross-Protective immunity-based vaccination with assurance of prolonged protection. Another issue of manufacturing and distribution of vaccines involves post authoriza-tion, licensing, and surveillance to ensure vaccine’s efficacy towards combating the Ebola out-break. The current review focusses on the development process, the current perspective of the development of Ebola vaccine and futuristic challenges to combat the future emerging Ebola in-fectious disease.
... Accumulating evidence suggests that key global pathogens, such as HIV, for which vaccine development has been challenging, may require vaccine strategies that can induce both neutralizing and non-neutralizing antibody functions (1)(2)(3)(4)(5)(6). Since the discovery of antibodies with extensive neutralization breadth [known as broadly neutralizing antibodies (bnAbs)] against HIV strains elicited by natural infection, the induction of this type of antibodies has been the ultimate goal of HIV vaccinologists (7). ...
... This observation suggests that the moderate protection achieved by the RV144 vaccine was mediated by nonneutralizing antibody responses (8,9). A number of studies have shown that non-neutralizing antibody functions such as antibodydependent cellular phagocytosis (ADCP) and antibody-dependent cellular cytotoxicity (ADCC) are crucial for controlling HIV (1), influenza (2), herpes simplex virus (3), Ebola virus (4), and cytomegalovirus (CMV) (5,6). A follow-up study to the RV144 trial (HVTN 702) using a similar vaccine strategy in Africa was recently discontinued because of lack of efficacy (10), and although more investigation is needed to understand the differences between HVTN 702 and RV144, the discrepancy is suggestive that an optimal HIV vaccine may require elicitation of both neutralizing and non-neutralizing antibody responses. ...
Article
Full-text available
To develop vaccines for certain key global pathogens such as HIV, it is crucial to elicit both neutralizing and non-neutralizing Fc-mediated effector antibody functions. Clinical evidence indicates that non-neutralizing antibody functions including antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) contribute to protection against several pathogens. In this study, we demonstrated that conjugation of HIV Envelope (Env) antigen gp120 to a self-assembling nanofiber material named Q11 induced antibodies with higher breadth and functionality when compared to soluble gp120. Immunization with Q11-conjugated gp120 vaccine (gp120-Q11) demonstrated higher tier 1 neutralization, ADCP, and ADCC as compared to soluble gp120. Moreover, Q11 conjugation altered the Fc N-glycosylation profile of antigen-specific antibodies, leading to a phenotype associated with increased ADCC in animals immunized with gp120-Q11. Thus, this nanomaterial vaccine strategy can enhance non-neutralizing antibody functions possibly through modulation of immunoglobulin G Fc N-glycosylation.
... 99,100 In mice and guinea pigs, vaccination with eVLPs or hybrid (e/m) VLPs could elicit rapid and broad EBOV-specific humoral and cellular responses, and could achieve complete protection. [100][101][102][103] Furthermore, the three-component eVLPs combining GP, VP40, and NP complemented by adjuvants provided 100% protection in NHPs with no signs of illness, [104][105][106] highlighting their superior performance as vaccine candidates. Meanwhile, baculovirus expression systems were adopted to increase transfection efficiency and yield. ...
Article
Full-text available
Ebola virus (EBOV) is classified as a category A pathogen as it causes viral hemorrhagic fever, one of the most-deadly virus-related diseases. Since its discovery in 1976, EBOV has caused a number of global public health incidents, which have posed a serious threat to both humans and non-human primates. Thus, numerous preventive vaccine studies are underway, including research on inactivated vaccines, DNA vaccines, subunit vaccines, virus-like particles, Venezuelan equine encephalitis virus replicon particles, and several viral vector vaccines. The vesicular stomatitis virus-based vaccine Ervebo was recently approved by the Food and Drug Administration and the European Union, and several other vaccines have also been proven to confer potent protection in non-human primates against EBOV lethal challenge. This review provides a brief background of EBOV, with a focus on the epidemiology, available animal models, and advances in preventive approaches for EBOV infection.
... In the last two decades, multiple studies have been conducted to develop an ebolavirus vaccine and therapeutics [147][148][149][150][151][152][153][154][155][156][157][158][159][160][161][162][163][164]. In 2014, a randomized, double-blind, placebo-controlled, phase I clinical trial demonstrated the safety and immunogenicity of an ebolavirus vaccine [165]. ...
Article
Full-text available
Since the inception of the ebolavirus in 1976, 32 outbreaks have resulted in nearly 15,350 deaths in more than ten countries of the African continent. In the last decade, the largest (2013–2016) and second largest (2018–2020) ebolavirus outbreaks have occurred in West Africa (mainly Guinea, Liberia, and Sierra Leone) and the Democratic Republic of the Congo, respectively. The 2013–2016 outbreak indicated an alarming geographical spread of the virus and was the first to qualify as an epidemic. Hence, it is imperative to halt ebolavirus progression and develop effective countermeasures. Despite several research efforts, ebolaviruses’ natural hosts and secondary reservoirs still elude the scientific world. The primary source responsible for infecting the index case is also unknown for most outbreaks. In this review, we summarize the history of ebolavirus outbreaks with a focus on etiology, natural hosts, zoonotic reservoirs, and transmission mechanisms. We also discuss the reasons why the African continent is the most affected region and identify steps to contain this virus.
... ENGERIX-B is very efficacious, highly immunogenic, and safe (Elhanan et al. 2018). VLP-based vaccines have also been developed and tested to prevent other viruses such as EBOV (Swenson et al. 2005), Influenza (Pushko et al. 2007(Pushko et al. , 2010 (Mahmood et al. 2008), MARV (Swenson et al. 2005;Warfield et al. 2007), Dengue virus (Murawski et al. 2010), West Nile virus (Spohn et al. 2010), Hepatitis C (Mihailova et al. 2006), HIV (Kang et al. 1999), and Chikungunya (Akahata et al. 2010). ...
Chapter
Full-text available
Lassa Fever (LF) is a viral hemorrhagic fever endemic in West Africa. LF begins with flu-like symptoms that are difficult to distinguish from other common endemic diseases such as malaria, dengue, and yellow fever making it hard to diagnose clinically. Availability of a rapid diagnostic test and other serological and molecular assays facilitates accurate diagnosis of LF. Lassa virus therapeutics are currently in different stages of preclinical development. Arevirumab, a cocktail of monoclonal antibodies, demonstrates a great safety and efficacy profile in non-human primates. Major efforts have been made in the development of a Lassa virus vaccine. Two vaccine candidates, MeV-NP and pLASV-GPC are undergoing evaluation in phase I clinical trials.
... Moreover, it was demonstrated that VLPs activated cellular immunity and that CD8+ T-cells are required for protection [617]. The protective efficacy of VLPs seen in rodent models has also been observed in non-human primates by Warfield and colleagues [618]. These studies showed encouraging results versus both Ebola and Marburg viruses [619,620]. ...
Article
Full-text available
The environmental conditions generated by war and characterized by poverty, undernutrition, stress, difficult access to safe water and food as well as lack of environmental and personal hygiene favor the spread of many infectious diseases. Epidemic typhus, plague, malaria, cholera, typhoid fever, hepatitis, tetanus, and smallpox have nearly constantly accompanied wars, frequently deeply conditioning the outcome of battles/wars more than weapons and military strategy. At the end of the nineteenth century, with the birth of bacteriology, military medical researchers in Germany, the United Kingdom, and France were active in discovering the etiological agents of some diseases and in developing preventive vaccines. Emil von Behring, Ronald Ross and Charles Laveran, who were or served as military physicians, won the first, the second, and the seventh Nobel Prize for Physiology or Medicine for discovering passive anti-diphtheria/tetanus immunotherapy and for identifying mosquito Anopheline as a malaria vector and plasmodium as its etiological agent, respectively. Meanwhile, Major Walter Reed in the United States of America discovered the mosquito vector of yellow fever, thus paving the way for its prevention by vector control. In this work, the military relevance of some vaccine-preventable and non-vaccine-preventable infectious diseases, as well as of biological weapons, and the military contributions to their control will be described. Currently, the civil–military medical collaboration is getting closer and becoming interdependent, from research and development for the prevention of infectious diseases to disasters and emergencies management, as recently demonstrated in Ebola and Zika outbreaks and the COVID-19 pandemic, even with the high biocontainment aeromedical evacuation, in a sort of global health diplomacy.
Article
Full-text available
Viruses have threatened human lives for decades, causing both chronic and acute infections accompanied by mild to severe symptoms. During the long journey of confrontation, humans have developed intricate immune systems to combat viral infections. In parallel, vaccines are invented and administrated to induce strong protective immunity while generating few adverse effects. With advancements in biochemistry and biophysics, different kinds of vaccines in versatile forms have been utilized to prevent virus infections, although the safety and effectiveness of these vaccines are diverse from each other. In this review, we first listed and described major pathogenic viruses and their pandemics that emerged in the past two centuries. Furthermore, we summarized the distinctive characteristics of different antiviral vaccines and adjuvants. Subsequently, in the main body, we reviewed recent advances of nanoparticles in the development of next-generation vaccines against influenza viruses, coronaviruses, HIV, hepatitis viruses, and many others. Specifically, we described applications of self-assembling protein polymers, virus-like particles, nano-carriers, and nano-adjuvants in antiviral vaccines. We also discussed the therapeutic potential of nanoparticles in developing safe and effective mucosal vaccines. Nanoparticle techniques could be promising platforms for developing broad-spectrum, preventive, or therapeutic antiviral vaccines.
Article
Full-text available
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Article
Full-text available
Ebola virus is the primary contributor to the global threat of filovirus severe hemorrhagic fever, and Ebola virus disease has a case fatality rate of 50–90%. An inactivated, bivalent filovirus/rabies virus vaccine, FILORAB1, consists of recombinant rabies virus virions expressing the Ebola virus glycoprotein. FILORAB1 is immunogenic and protective from Ebola virus challenge in mice and non-human primates, and protection is enhanced when formulated with toll-like receptor 4 agonist Glucopyranosyl lipid adjuvant (GLA) in a squalene oil-in-water emulsion (SE). Through an adjuvant comparison in mice, we demonstrate that GLA-SE improves FILORAB1 efficacy by activating the innate immune system and shaping a Th1-biased adaptive immune response. GLA-SE adjuvanted mice and those adjuvanted with the SE component are better protected from surrogate challenge, while Th2 alum adjuvanted mice are not. Additionally, the immune response to FILORAB1 is long-lasting, as exhibited by highly-maintained serum antibody titers and long-lived cells in the spleen and bone marrow.
Article
Full-text available
Spatiotemporal aspects of filovirus entry and release are poorly understood. Lipid rafts act as functional platforms for multiple cellular signaling and trafficking processes. Here, we report the compartmentalization of Ebola and Marburg viral proteins within lipid rafts during viral assembly and budding. Filoviruses released from infected cells incorporated raft-associated molecules, suggesting that viral exit occurs at the rafts. Ectopic expression of Ebola matrix protein and glycoprotein supported raft-dependent release of filamentous, virus-like particles (VLPs), strikingly similar to live virus as revealed by electron microscopy. Our findings also revealed that the entry of filoviruses requires functional rafts, identifying rafts as the site of virus attack. The identification of rafts as the gateway for the entry and exit of filoviruses and raft-dependent generation of VLPs have important implications for development of therapeutics and vaccination strategies against infections with Ebola and Marburg viruses.
Article
Subunit vaccines based on recombinant proteins can suffer from poor immunogenicity owing to incorrect folding of the target protein or poor presentation to the immune system. Virus-like particles (VLPs) represent a specific class of subunit vaccine that mimic the structure of authentic virus particles. They are recognized readily by the immune system and present viral antigens in a more authentic conformation than other subunit vaccines. VLPs have therefore shown dramatic effectiveness as candidate vaccines. Here, we review the current status of VLPs as vaccines, and discuss the characteristics and problems associated with producing VLPs for different viruses.
Article
A plaque assay for Ebola virus is reported. The procedure has real potential for future research, although it is less sensitive than indirect fluorescent-antibody and mouse inoculation tests.
Article
MHC class I genes evolve by recombination, largely within loci, and selection (presumably pathogen-driven) maintains these new alleles in the population. Although chimpanzees and humans share an A locus allelic lineage, the B locus molecules of the chimpanzee were less similar to human B locus molecules. The A and B locus molecules in rhesus and cynomolgus monkeys were even less similar to their human counterparts, with little conservation of allelic lineages between macaques and humans. In contrast to the instability of MHC class I alleles and allelic lineages, the MHC class I loci themselves are well conserved during the evolution of Great Ape and Old World primates. Homologues of HLA-A, -B, -E, and -F have been found in macaques. The C locus, however, has only been found in gorillas and chimpanzees, whereas in orangutans and rhesus monkeys it is possible that the A and B loci have been duplicated. Classical New World monkey MHC class I genes are all more similar to the nonclassical HLA-G gene and a nonclassical F-like gene is present in the cotton-top tamarin. Duplication and either subsequent deletion or expansion of MHC class I loci, therefore, appear to be the modus operandi of the evolution of these genes in primates.
Article
Papillomavirus-like particles can be obtained by expression of the major capsid protein L1 alone or by coexpression with the minor capsid protein L2 in various systems. Virus-like particles (VPLs) and virions have very similar capsid structures. Immunization with VLPs yields antibodies neutralizing virions in vitro. Vaccination of animals with VLPs has been shown to protect against viral challenge. VLPs of human papillomavirus (HPV) are therefore the most promising vaccine candidate to prevent infections with HPVs associated with cervical cancer, the most frequent carcinoma in women worldwide.
Article
Viruses which infect the gastrointestinal tract are well suited for examining the immune response(s) to oral delivery of antigen and exploring the advantages and pitfalls of oral vaccines. We have used recombinant DNA techniques to produce nonreplicating self-assembled virus-like particles (VLPs) from two gastrointestinal viruses, rotavirus and Norwalk virus. Both of these viruses normally cause acute gastroenteritis in man or animals. The VLPs are morphologically and antigenically similar to the native virus and quite stable, features which are advantageous for their use as subunit vaccines. In addition, these VLPs could be useful as carriers of foreign epitopes from heterologous pathogens or of drugs which need to be delivered to the gastrointestinal track. This paper briefly reviews the properties of these VLPs made in insect cells and data showing their potential as subunit vaccines for parenteral or oral delivery.
Article
There is no effective vaccine for Marburg virus (MBGV) or any other filovirus, nor enough pertinent information to expedite rational vaccine development. To ascertain some of the minimal requirements for a MBGV vaccine, we determined whether whole inactivated MBGV, or a baculovirus-expressed virion subunit, could be used to immunize guinea pigs against a lethal infection. Baculovirus recombinants were made to express the MBGV glycoprotein (GP) either as a full-length, cell-associated molecule or a slightly truncated (5.4%) product secreted into medium; the latter, for its far greater ease in manipulation, was tested for its vaccine potential. Like MBGV GP, both the full-length and truncated GP expressed by baculovirus recombinants were abundantly glycosylated with both N- and O-linked glycans; differences in glycosylation were detectable, but these could not be shown to affect antigenicity with respect to available antibodies. The recombinant truncated glycoprotein elicited protection against lethal challenge with the MBGV isolate from which it was constructed and less effectively against an antigenically disparate MBGV isolate. Killed (irradiated) MBGV antigen was protective, in a reciprocal fashion, against both MBGV types. In a preliminary assessment of possible protective mechanisms, serum antibodies from immune animals were shown to be sufficient for protecting naive guinea pigs from lethal MBGV infections
Article
Hybrid virus-like particles (VLP) were prepared by self-assembly of the modified porcine parvovirus (PPV) VP2 capsid protein carrying a CD8+ or CD4+ T cell epitope. Immunization of mice with a single dose of these hybrid pseudo-particles, without adjuvant, induced strong cytotoxic T lymphocyte and T helper (Th) responses against the reporter epitope. The Th response was characterized by a Th1 phenotype. We also analyzed in vitro the uptake mechanism of these parvovirus-like particles and the processing requirements associated with presentation by MHC molecules. Although previously shown to be presented by MHC class I molecules, these particles also enter very efficiently the MHC class II endocytic pathway, and behave as conventional exogenous antigens. Indeed, the processing of chimeric PPV:VLP was performed in endosomal/lysosomal acidic vesicles and the presentation of the foreign epitope carried by these particles was sensitive to brefeldin A and cycloheximide, showing that the foreign peptide was loaded on nascent MHC class II molecules. These results give some indication of how PPV:VLP can be presented by MHC class I and class II molecules, and underscore the wide potency of such VLP system to deliver foreign antigens for vaccine design.
Article
DNA vaccines expressing the envelope glycoprotein (GP) or nucleocapsid protein (NP) genes of Ebola virus were evaluated in adult, immunocompetent mice. The vaccines were delivered into the skin by particle bombardment of DNA-coated gold beads with the Powderject-XR gene gun. Both vaccines elicited antibody responses as measured by ELISA and elicited cytotoxic T cell responses as measured by chromium release assays. From one to four vaccinations with 0.5 microgram of the GP DNA vaccine resulted in a dose-dependent protection from Ebola virus challenge. Maximal protection (78% survival) was achieved after four vaccinations. Mice were completely protected with a priming dose of 0.5 microgram of GP DNA followed by three or four subsequent vaccinations with 1.5 micrograms of DNA. Partial protection could be observed for at least 9 months after three immunizations with 0.5 microgram of the GP DNA vaccine. Comparing the GP and NP vaccines indicated that approximately the same level of protection could be achieved with either vaccine.