ArticlePDF Available

Comparison of intradermal and intramuscular delivery of SIV Env DNA by in vivo electroporation in macaques

Taylor & Francis
Human Vaccines & Immunotherapeutics
Authors:
  • ImQuest BioSciences

Abstract and Figures

A panel of SIVmac251 transmitted Env sequences were tested for expression, function and immunogenicity in mice and macaques. The immunogenicity of a DNA vaccine cocktail expressing SIVmac239 and three transmitted SIVmac251 Env sequences was evaluated upon intradermal or intramuscular injection followed by in vivo electroporation in macaques using sequential vaccination of gp160, gp120 and gp140 expressing DNAs. Both intradermal and intramuscular vaccination regimens using the gp160 expression plasmids induced robust humoral immune responses, which further improved using the gp120 expressing DNAs. The responses showed durability of binding and neutralizing antibody titers and high avidity for > 1 y. The intradermal DNA delivery regimen induced higher cross-reactive responses able to neutralize the heterologous tier 1B-like SIVsmE660_CG7V. Analysis of cellular immune responses showed induction of Env-specific memory responses and cytotoxic granzyme B (+) T cells in both vaccine groups, although the magnitude of the responses were ~10x higher in the intramuscular/electroporation group. The cellular responses induced by both regimens were long lasting and could be detected ~1 y after the last vaccination. These data show that both DNA delivery methods are able to induce robust and durable immune responses in macaques.
Content may be subject to copyright.
This article was downloaded by: [128.199.169.55]
On: 02 September 2015, At: 01:16
Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: 5 Howick Place,
London, SW1P 1WG
Human Vaccines & Immunotherapeutics
Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/khvi20
Comparison of intradermal and intramuscular delivery
followed by in vivo electroporation of SIV Env DNA in
macaques
Viraj Kulkarnia, Margherita Rosatib, Jenifer Beara, Guy R Pilkingtona, Rashmi Jalaha, Cristina
Bergamaschia, Ashish K Singhb, Candido Aliceaa, Bhabadeb Chowdhuryb, Gen-Mu Zhangab,
Eun-Young Kimc, Steven M Wolinskyc, Wensheng Huangd, Yongjun Guand, Celia LaBranchee,
David C Montefiorie, Kate E Broderickf, Niranjan Y Sardesaif, Antonio Valentinb, Barbara K
Felbera & George N Pavlakisb
a Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research;
National Cancer Institute; Frederick, MD USA
b Human Retrovirus Section; Vaccine Branch; Center for Cancer Research; National Cancer
Institute; Frederick, MD USA
c Division of Infectious Diseases; The Feinberg School of Medicine; Northwestern University;
Chicago, IL USA
d Institute of Human Virology; Department of Microbiology and Immunology; University of
Maryland School of Medicine; Baltimore, MD USA
e Department of Surgery; Laboratory for AIDS Vaccine Research and Development; Duke
University Medical Center; Durham, NC USA
f Inovio Pharmaceuticals, Inc.; Blue Bell, PA USA
Published online: 28 Jun 2013.
To cite this article: Viraj Kulkarni, Margherita Rosati, Jenifer Bear, Guy R Pilkington, Rashmi Jalah, Cristina Bergamaschi,
Ashish K Singh, Candido Alicea, Bhabadeb Chowdhury, Gen-Mu Zhang, Eun-Young Kim, Steven M Wolinsky, Wensheng Huang,
Yongjun Guan, Celia LaBranche, David C Montefiori, Kate E Broderick, Niranjan Y Sardesai, Antonio Valentin, Barbara K Felber
& George N Pavlakis (2013) Comparison of intradermal and intramuscular delivery followed by in vivo electroporation of SIV
Env DNA in macaques, Human Vaccines & Immunotherapeutics, 9:10, 2081-2094, DOI: 10.4161/hv.25473
To link to this article: http://dx.doi.org/10.4161/hv.25473
PLEASE SCROLL DOWN FOR ARTICLE
Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in
the publications on our platform. Taylor & Francis, our agents, and our licensors make no representations or
warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Versions
of published Taylor & Francis and Routledge Open articles and Taylor & Francis and Routledge Open Select
articles posted to institutional or subject repositories or any other third-party website are without warranty
from Taylor & Francis of any kind, either expressed or implied, including, but not limited to, warranties of
merchantability, fitness for a particular purpose, or non-infringement. Any opinions and views expressed in this
article are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The
accuracy of the Content should not be relied upon and should be independently verified with primary sources
of information. Taylor & Francis shall not be liable for any losses, actions, claims, proceedings, demands,
costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in
connection with, in relation to or arising out of the use of the Content.
This article may be used for research, teaching, and private study purposes. Terms & Conditions of access and
use can be found at http://www.tandfonline.com/page/terms-and-conditions
It is essential that you check the license status of any given Open and Open Select article to confirm
conditions of access and use.
Downloaded by [128.199.169.55] at 01:16 02 September 2015
RESEARCH PAPER
www.landesbioscience.com Human Vaccines & Immunotherapeutics 2081
Human Vaccines & Immunotherapeutics 9:10, 2081–2094; October 2013; © 2013 Landes Bioscience
RESEARCH PAPER
*Correspondence to: Barbara K Felber, Email: felberb@mail.nih.gov; George N Pavlakis, Email: pavlakig@mail.nih.gov
Submitted: 05/20/2013; Accepted: 06/20/2013
http://dx.doi.org/10.4161/hv.25473
Introduction
DNA is a compelling vaccine vehicle because of its simplicity,
scalability, and lack of immunity against the vector. Different
DNA delivery methods are being tested, including intramuscular
DNA delivery by in vivo electroporation (IM/EP) (reviewed
in refs. 1–3); Dermavir,4 liposome delivery with Vaxfectin®,5
biojector or intradermal EP (ID/EP)6,7 or gene gun.8
The development of intramuscular DNA injection followed
by in vivo electroporation9-14 brought a significant change in the
efficiency of DNA delivery, especially to higher mammals like
macaques and humans. In the field of HIV/SIV vaccine, several
groups showed that IM/EP delivery led to increased immune
responses (several fold over those induced by conventional needle
and syringe).15- 18 The magnitude of the DNA induced immune
responses could be further augmented by the inclusion of IL-12
A panel of SIVmac251 transmitted Env sequences were tested for expression, function and immunogenicity in mice and
macaques. The immunogenicity of a DNA vaccine cocktail expressing SIVmac239 and three transmitted SIVmac251 Env
sequences was evaluated upon intradermal or intramuscular injection followed by in vivo electroporation in macaques
using sequential vaccination of gp160, gp120 and gp140 expressing DNAs. Both intra dermal and intramuscular vaccination
regimens using the gp160 expression plasmids induced robust humoral immune responses, which further improved
using the gp120 expressing DNAs. The responses showed durability of binding and neutralizing antibody titers and high
avidity for >1 y. The intradermal DNA delivery regimen induced higher cross-reactive responses able to neutralize the
heterologous tier 1B-like SIVsmE660_CG7V. Analysis of cellular immune responses showed induction of Env-specic
memory responses and cytotoxic granzyme B+ T cells in both vaccine groups, although the magnitude of the responses
were ~10x higher in the intramuscular/electroporation group. The cellular responses induced by both regimens were
long lasting and could be detected ~1 y after the last vaccination. These data show that both DNA delivery methods are
able to induce robust and durable immune responses in macaques.
Comparison of intradermal and intramuscular
delivery followed by in vivo electroporation
of SIV Env DNA in macaques
Viraj Kulkarni1, Margherita Rosati2, Jenifer Bear1, Guy R Pilkington1, Rashmi Jalah1, Cristina Bergamaschi1, Ashish K Singh2,
Candido Alicea1, Bhabadeb Chowdhury2, Gen-Mu Zhang1,2, Eun-Young Kim3, Steven M Wolinsky3, Wensheng Huang4,
Yongjun Guan4, Celia LaBranche5, David C Monteori5, Kate E Broderick6, Niranjan Y Sardesai6, Antonio Valentin2,
Barbara K Felber1,*, and George N Pavlakis2,*
1Human Retrovi rus Pathogenesis Sec tion; Vaccine Branch; Center fo r Cancer Research; Nationa l Cancer Institute; Freder ick, MD USA; 2Human Retrov irus Section; Vaccine
Branch; Center f or Cancer Research; Natio nal Cancer Institute; Frede rick, MD USA; 3Divisio n of Infectious Diseas es; The Feinberg Schoo l of Medicine; Northwe stern University;
Chicago, IL USA ; 4Institute of Human Virol ogy; Departmen t of Microbiology and I mmunology; Univers ity of Maryland Sc hool of Medicine; Balt imore, MD USA; 5Depart ment of
Surgery ; Laboratory for AI DS Vaccine Research and Devel opment; Duke Universi ty Medical Center; D urham, NC USA; 6Inovio Pharm aceuticals, Inc.; Blu e Bell, PA USA
Keywords: intradermal, intramuscular, in vivo electroporation, DNA vaccine, HIV, SIVsmE660, SIVmac239, SIVmac251,
transmitted Env, binding antibody, neutralizing antibody, avidity, transitional memory, effector memory
Abbreviations: EP, electroporation; ID, intradermal; IM, intramuscular; GzmB, GranzymeB; TM, transitional memory;
EM, effector memory
DNA as adjuvant in mice and macaques.18-25 Importantly, in
macaques, the combination of optimized DNA delivery by IM/
EP and the inclusion of IL-12 DNA led to robust cellular and
humoral immune responses,15, 18, 2 4 -2 8 which resulted not only in
quantitative improvement,18,24,25 but also in improved quality
of the responses, e.g., increase of antigen-specific cytotoxic T
cells and greater breadth of the neutralizing antibody (NAb)
responses.24 Similarly, ID/EP delivery of DNA vaccines yielded
improved humoral immune responses and led to protection from
challenge as judged by lethality, viremia, and morbidity in two
independent macaque models testing a multivalent small pox
vaccine and a multivalent avian influenza (A/H5N1 vaccine).29, 30
DNA as the only vaccine component had been considered
poorly immunogenic in humans, until recent results dem-
onstrated that in vivo electroporation is more efficient (more
responders, and higher, longer-lasting immunity) than DNA
Downloaded by [128.199.169.55] at 01:16 02 September 2015
2082 Human Vaccines & Immunotherapeutics Volume 9 Issue 10
35014 (M766) and mac251_9 share a characteristic V1-V2
region found in several of our infected animals. Of note, the Env
sequence 35014 was independently identified by B. Keele et al.33
(GenBank accession #JQ086004) and also named M766, and
is also referred to with this name in this report. Env mac251_15
and mac251_2 contain a different V1-V2 region. In addition, we
included the previously reported transmitted Env CR 2.RU.3R134
as a sequence with different V1V2 from a macaque infected with
a different SIVmac251 stock.
The phylogenetic tree analysis shows the relation among the
selected SIVmac251 Env sequences, which share 4.7–6.8% AA
difference with SIVmac239 (Fig. 1C). We also include two heter-
ologous Env sequences from SIVsmE660 (CG7V and CG7G)34
which differ by ~20% from the SIVmac251 sequences and are
used in neutralization assays (see Fig. 2). We generated a col-
lection of vectors producing gp160, the trimeric gp140 and the
soluble gp120 of the different Envs from expression-optimized
cDNAs, which were cloned into the mammalian expression
vector CMVkan (Table 1). The Env plasmids were tested for
expression upon transient transfection and Western immunoblot
assay (Fig. 3A). The newly identified SIVmac251 Env proteins
were expressed at levels similar to mac239 Env. Two of the Envs
(mac251_2 and mac251_9) showed poor cleavage of gp160, with
no production of gp41, although no apparent AA changes sur-
rounding the furin cleavage site (AA 525) were noted. In addition
to the gp160 Env protein, we also generated plasmids (Fig. 3B)
expressing the trimeric gp140 (AA 1 to 686) lacking the mem-
brane spanning domain (MSD) and the soluble gp120 (A A 1 to
525). Figure 3C shows that uncleaved gp140 and gp120 were
efficiently secreted and accumulated in the extracellular com-
partment. Similar data were obtained for the other Env proteins
(not shown) and the plasmids are listed in Table 1. The sequences
were further tested for function using the pseudotype infection
assay (Fig. 3D). All sequences produced functional pseudotyped
virions, except for the 2 Env with processing defects.
To test the immunogenicity of the identified SIVmac251 Env
sequences, groups of BALB/c mice were vaccinated with four
different DNAs expressing gp160 from mac239, 251_15, 251_2,
and 251_9 by the conventional needle and syringe intramuscular
injection method (Fig. 4A). All Env vectors induced robust cel-
lular immune responses with similar levels as SIVmac239 Env,
including the 2 non-functional Env (mac251_2 and mac251_9)
(Fig. 4B). Analysis of humoral immunity by ELISA showed the
induction of robust bAb titers, except for the 2 Env which showed
impaired gp160 processing (Fig. 4C). Together, these data led to
the selection of 4 newly identified Env for the immunogenicity
study in macaques (see below).
Production of SIVmac251 Env proteins from mammalian
cells. Another goal of this work has been to select representa-
tive sequences able to efficiently produce SIV Env proteins in
mammalian cells. We tested this by the generation and screen-
ing of stable overproducing HEK293 cells. Stable high producer
HEK293 cell clones using env DNAs expressing the gp120
form of SIVmac251 35014 (M766) gp120 and the gp140 form
of SIVmac251 35014_7 were generated. In addition, we also
generated cell lines producing gp140 forms of SIVmac239 and
delivery by needle/syringe also in man,31 corroborating the find-
ings of macaque studies from us and others. Another trial, using
HIV gag DNA and IM/EP DNA delivery showed that inclu-
sion of IL-12 DNA is advantageous, resulting in both increased
frequency of responders and level of Gag-specific immunity.32
Together, these studies showed that the findings obtained in the
macaque model could be well translated to humans, which is an
encouraging step in the development of a DNA-based vaccine
against AIDS.
This report is a pilot study in macaques to compare a
SIVmac251 derived DNA vaccine based on the selection of trans-
mitted Env clones with varying sequences, and delivered by two
different routes (intradermal and intramuscular) followed by
in vivo electroporation. The vaccine candidate and the delivery
routes were designed to test whether they might induce broader
immunity compared with SIVmac239. Both vaccination regi-
mens induced potent long-lasting immune responses with IM/
EP vaccination inducing higher levels of cellular responses, and
ID/EP vaccination inducing broader humoral responses.
Results
Identification and characterization of transmitted SIVmac251
Env sequences. The Env sequence from the infectious molecu-
lar clone SIVmac239 has been frequently used as antigen to test
immunogenicity and potency of SIV vaccines in macaques. We
wished to test the hypothesis that a selection of transmitted Env
clones with varying sequences might induce broader immunity
compared with SIVmac239 Env. Toward this goal, we identified
full-length Env sequences present in the plasma early during the
peak of primary viremia from macaques infected via the mucosal
route with low dose SIVmac251. To obtain a more comprehen-
sive picture of the virus variants present in the SIVmac251 stock
and the infected animals, we also interrogated by Single Genome
Amplification (SGA) and sequencing the variable regions V1&V2
region (AA 53–260 following the numbering of SIVmac239 con-
taining V1 [AA 113–168] and V2 [AA 168–211] Fig. 1A and B),
since V1 is known to encompass most of the diversity of the SIV
Env. We noted that, despite the diversity of the stock (ref. 33 and
our unpublished analysis), viruses sharing a narrow selection of
Env sequences were found to replicate in the majority of the ana-
lyzed animals. The Env sequences were closely related to those
found in the inoculum and represented a single inferred founder
virus in 7 of 9 rhesus macaques.
From the collection of sequences, we selected Env sequences
with distinct V1-V2 regions for further analysis. Comparison
of SIVmac239 to these sequences revealed numerous changes
throughout Env in addition to the highly variable V1 region
(AA 113–168) (Fig. 1A). A cartoon of SIVmac239 indicating the
location of the variable (V) and constant (C) regions is shown
in Figure 1B. In contrast to V1 and the variable V4 (AA 402–
432), comparison to mac239 Env showed that the transmitted
sequences share several changes to the same A A in V2, V3 (AA
311–344) as well as in C1-C4 and they did not show changes in
V5 (AA 459–484) and C5, whereas numerous AA changes are
found in gp41 (AA 526–879). Of these sequences, Env 35014_7,
Downloaded by [128.199.169.55] at 01:16 02 September 2015
www.landesbioscience.com Human Vaccines & Immunotherapeutics 2083
blue and, as a representative example, the production of 35014
(M766) gp120 Env overtime is shown (Fig. 5A). A strong band
corresponding to secreted gp120 could be detected in addition
to a ~60 kDa band corresponding to albumin, introduced from
the seed culture as part of the complete culture medium that
SIVsmE660_CG7V. Selected high producer cell clones were
grown in serum-free media in a Hollow Fiber bioreactor that
allows high-density growth in defined serum-free medium.
Analysis of a 6 μl aliquots from the supernatants of daily har-
vests (20 ml) was performed on gels stained with Coomassie
Figure 1. Amino acid alignment of transmitted SIVmac251 Env sequences. (A) The identied SIVmac251 sequences and the repor ted SIVmac251
sequence CR2.RU.3R1 are compared with SIVmac239. (B) Cartoon depicts the location of the signal peptide, the variable ( V) and conserved (C) regions,
and gp41 of SIVmac239. The numbering follows SIVmac239 sequence. (C) Phylogenetic tree analysis of the SIVmac239 and the dierent SIVmac251
sequences. The tree also included 2 SIVsmE660 env CG7G and CG7V that are used in the neutralization assays.
Downloaded by [128.199.169.55] at 01:16 02 September 2015
2084 Human Vaccines & Immunotherapeutics Volume 9 Issue 10
contained fetal calf serum. The culture medium was changed
daily and typically after 7–10 d, albumin was no longer detect-
able. High level of Env production persisted for > 15 weeks of
continuous cell growth. The estimated Env production is ~3
mg/20 ml/day using purified SIV gp120 Env protein as standard.
Supernatants collected after the 1st week of culturing the cells in
the Hollow Fiber bioreactor were enriched for Env and were used
for protein isolation using standard lectin column purification.
Similar stable daily production was obtained for the other Env
proteins (not shown). After purification, the proteins were ana-
lyzed on non-denaturing gels and stained with Coomassie blue,
which revealed high quality of the produced monomeric SIVmac
35014 (M766) gp120 as well as stable trimeric forms of SIVmac
35014_7 and SIVsmE660_CG7V gp140 Env (Fig. 5B).
Vaccination via intradermal/EP and intramuscular/EP
route induces robust humoral immune responses. We compared
the immunogenicity of a combination of SIV Env DNA in rhesus
macaques using intradermal (ID) and intramuscular (IM) DNA
delivery followed by in vivo electroporation. As vaccine, we fol-
lowed a sequential immunization scheme (outlined in Fig. 6A)
using a mixture of Env DNAs expressing gp160 (EP1–3), gp120
(EP4) or gp140 (EP5). Macaques received the same DNA vac-
cine (1 mg/animal) via intradermal/EP (n = 3) or intramuscular/
EP (n = 2). Similar levels (reciprocal endpoint titer log 4.5–5)
of SIVmac251 binding Ab (bAb) were found in the plasma
(Fig. 6B) using the ID/EP or the IM/EP delivery method. The
bAb showed similar longevity during the 11 weeks of follow-up
(EP3wk2 to EP4) with a decline of < 0.5 log. Vaccination with
DNAs expressing the secreted gp120 forms of Env increased the
bAb endpoint titers in both groups (EP4wk2) to levels slightly
(~0.5 log) higher than the levels obtained upon vaccination with
DNAs expressing the gp160 forms of Env. This is likely due
to the more efficient production of gp120 from these plasmids
compared with the gp160 vectors (see Fig. 3C). Subsequent vac-
cination with plasmids expressing the trimeric gp140 showed
similar bAb levels, likely due to the fact that maximal levels were
obtained using the gp120 DNAs mixture. Together, these data
show that both DNA delivery systems efficiently induced robust
systemic binding Ab levels in the vaccinated macaques.
We added 3 animals to the IM/EP group, which were vac-
cinated with a 2 mg dose using the gp160 forms only of the same
four Env DNAs administered separately (using a slightly differ-
ent schedule with vaccinations at 0, 4, 12 weeks). These animals
showed similar reciprocal endpoint bAb titers of ~log 5 (not
shown) and allowed us to include the data from this group (after
gp160 DNA vaccination) in this study. The responses from the 1
and 2 mg dose IM/EP groups trended to be higher (~1.5 log) com-
pared those we reported from macaques (n = 8) which received
0.5 mg gp160 env DNA administered via IM/EP.24 These data
indicated that higher humoral responses could be obtained using
the IM/EP delivery by increasing the env DNA dose, with 1 mg
reflecting saturating levels in the rhesus macaque.
Intradermal DNA delivery induced more robust broadly
neutralizing antibodies. We next tested the neutralization capa-
bility of the vaccine-induced Env antibodies (Fig. 2) induced
upon ID/EP (Fig. 2, left panel) and IM/EP (Fig. 2, middle
Figure 2. Induction of neutralizing antibody in macaques immunized
via the intradermal (left panel) and the intramuscular (middle and
right panels) route followed by EP. Neutralizing antibody titers were
determined against SIVmac251-TCLA measured in M7-Luc cells (A) and
against pseudotyped viruses containing the following Env: SIVmac251
35014 (M766) (B) and 35014_7 (C), the heterologous SIVsmE660 Env
CG7G (D) and CG7V (E) were measured using the TZM-bl assay. The NAb
measurements of a group of macaques which received 3 vaccinations
of the same DNAs (total 2 mg) expressing the gp160 forms of Env via the
IM/EP deliver y is included (right panel). Neutralization titers shown are
the log of the reciprocal dilution of sample that reduced the signal by
50% compared with virus in the absence of sample.
Downloaded by [128.199.169.55] at 01:16 02 September 2015
www.landesbioscience.com Human Vaccines & Immunotherapeutics 2085
The plasma samples were also tested for their ability
to neutralize the heterologous SIVsmE660 Env (Fig. 2D
and E) CG7G and CG7V which differ by ~20% from the
SIVmac251 Envs present in the vaccine mixture (see also Fig.
1C). The Ab from both the ID/EP and the IM/EP groups
were able to neutralize the tier 1A-like SIVsmE660_CG7G
(Fig. 2D) to similar extent. Interestingly, we noted a more robust
neutralization capability to the tier 1B-like SIVsmE660_CG7V
by the antibodies induced by the ID /EP protocol, especially after
vaccination with gp120 forms of Env (EP4wk2) (Fig. 2E, left
panel) with more responders and higher titers. Of note, CG7G
and CG7V Env are 99% identical and differ by 4 AA in gp120
(1AA lies in the N-terminal portion [C1], 2 AA lie in V1, 1 AA
lies in C5) and 5 A A lie in gp41. Despite this high homology
in gp120, we noted a distinct neutralization pattern by these 2
Env in the pseudotype assay. This observation is reminiscent
to the finding using the different highly homologous gp120
proteins from SIVmac251, which we described above. Thus, it is
interesting to note that the gp120 proteins differing by very few
AA (3 A A in the SIVmac251 Envs or 4 AA in the E660 Envs)
can have such a profound distinct characteristics in the NAb
assay. This finding is agreement with a previous observations
of a single AA change in V2 of HIV Env35 and of truncation of
SIV g p4136 that also altered neutralizability.
To understand whether the inclusion of several Env in the
DNA vaccine cocktail induced Ab with broader crossreactivity,
we compared the results shown in Figure 2 to our previously
reported data of macaques vaccinated with mac239 gp160
DNA.24 Using the same panel of Env, we did not find broader
crossreactivity in macaques vaccinated with a mixture of different
SIVmac251 Env vs. only SIVmac239 Env. More importantly
however, we noted a trend that the ID/EP delivery is more potent,
inducing more consistent and higher tier NAb to the difficult to
neutralize E660_CG7V. Thus, the delivery method to the skin
panel). We also added the results from the group that received
IM/EP vaccination with the 2 mg DNA dose (Fig. 2, right panel)
which shows similar data as the 1 mg group (Fig. 2, middle panel,
compare gp160 DNA vaccination only). The neutralization capa-
bility was measured against a panel of SIV Env variants including
the homologous T cell-adapted (TCLA) SIVmac251 (Fig. 2A),
two transmitted SIVmac251 Env 35014 (M766) (Fig. 2B),
35014_7 (Fig. 2C) as well as mac251_15 (not shown) and CR2.
RU.3R1 (not shown). Plasma samples collected 2 weeks after the
gp160 (EP3wk2), gp120 (EP4wk2) and gp140 (EP5wk2) DNA
vaccinations were analyzed. Overall, similar NAb titers were
detected using ID/EP (left panel) and the IM/EP vaccination
regimens (Fig. 2A–C) upon vaccination with different forms of
Env (gp160, gp120, gp140).
We noted a distinct difference in neutralization of the trans-
mitted Env 35014_7 (Fig. 2C) compared with Env_35014
(M766 ) (Fig. 2B), with the latter being more easy to neutralize
(see also Tabl e 1) as shown by the higher NAb titers measured in
both vaccine groups. We noted this difference also upon analysis
of macaques vaccinated with SIVmac239 Env only (unpublished
observation). Thus, inclusion of 35014_7 Env in the vaccine mix-
ture did not improve the ability to neutralize 35014_7 Env pseu-
dotyped virions. It is noteworthy that the mature gp120 forms
of 35014_7 and M766 differ by only 3 AA (1 AA lies in the
N-terminal portion [C1], 2 AA lie in V4, while the remaining
8 AA changes lie in gp41; Fig. 1A). Interestingly, these 3 AA
changes in gp120 generated an Env protein with profoundly dif-
ferent behavior in the TZM-bl neutralization assay. We tested
the ability of another 2 Env (mac251_15 used in all mixtures
and CR2.RU.3R1, included in gp120 and gp140 mixtures) to
be neutralized. None of the plasma samples from the vaccinated
macaques showed any neutralizing activity against these Envs,
which like SIVmac239, appear to constitute a group of Env that
are difficult to neutralize (Table 1).
Table1. SIV Env and functional characteristics
SIV Plasmid
name
GenBank
accession #
Optimized env DNA expres-
sion plasmids (plasmid code) gp120/gp41
production from
gp160 DNA
Function in
pseudotype
assay
Ability of Env to
be neutralized
(TZM-bl assay)
Evaluation of
neutralizability of
Env
(TZM-bl assay)
gp160 gp140 gp120
mac 251
mac239 M33262 99S 237S 17 3S yes yes no difficult to neutralize
35014
(M76634)
KF003433
(JQ08600434)221S 241S 246S yes yes yes tier 1A-like
35014 _ 7 KF003434 220S 234S 230S yes yes yes tier 1B-like
251_9 K F003437 219S poor gp41
cleavage no
251_15 KF003436 217S 240S 229S yes yes no difficult to neutralize
251_ 2 KF0 03435 218 S poor gp41
cleavage no
CR2.
RU.3R1 FJ57 8 0 4 434 2 31S 242S 223S yes ND no difficult to neutralize
smE660 CG7G FJ57 838 134 251S 254S yes ND yes tier 1A-like
CG7V FJ 57 8 428 34 252S 255S yes ND Yes (ID/EP group) tier 1B-like
ND, not determined.
Downloaded by [128.199.169.55] at 01:16 02 September 2015
2086 Human Vaccines & Immunotherapeutics Volume 9 Issue 10
appeared to have an important feature to
induce antibodies with better breadth.
Longevity of humoral immune
responses. To address the persistence of the
humoral immune responses, the animals
were monitored for 14–20 mo after the
last vaccination (Fig. 7). The bAb titers to
SIVmac239 showed an initial decline during
the first ~2 mo after the last vaccination,
which was followed by similar persistence
in the intradermal/EP and intramuscular/
EP groups (Fig. 7A). We also monitored
the avidity of the SIVmac239 bAb (Table
2). The avidity index of sham DNA
injected animals or pre-samples were below
detection limit (assigned to 0.1%). The
mac239 bAb showed an avidity index with
a range of 15–47% at 2 weeks after the last
vaccination, and these values did not change
significantly overtime, indicating that the
quality of the vaccine-induced antibody
responses was maintained over time. In
addition, we also found persistence of
NAbs to SIVmac_35014 (M766) (Fig. 7B)
and the heterologous SIVsmE660_CG7G
(Fig. 7C) using both of the vaccination
methods. Together, long-term follow-up
showed that both ID/EP and the IM/EP
DNA delivery methods induced Env-specific
antibodies with similar potent durability.
Cellular immune responses induced
upon intradermal and intramuscular
delivery of Env DNA plasmids. The
induction of Env-specific cellular immune
responses was monitored upon stimulation
of PBMC with SIVmac239 Env peptide
pool (15-mer overlapping by 11 AA)
and measured by intracellular cytokine
staining and flow cytometry. The two
Figure 3. Characterization of dierent SIV
mac251 Env proteins. (A) Western immunoblot
analysis of the expression of SIVmac239 Env and
the dierent SIVmac251 Envs upon transient
transfection of HEK293 cells. (B) Cartoon depict-
ing gp160, gp140 and gp120 Env. (C) Western im-
munoblot analysis the gp160, gp140 and gp120
Env of SIVmac251_35014 (M766) upon transient
transfection of HEK293 cells. Cell-associated and
extra-cellular fractions are shown. (D) Pseu-
dotyped recombinant HIV-luc virus reporter
(NL4–3.LucRE) viruses were generated using
the SIVmac239 Env and the indicated SIVmac251
Env. Infectivity was tested in modied CEM-X174
cells that contain the HIV-LTR promoter linked
to GFP stably integrated. GFP expression, which
serves as read-out for infection, was measured
by FACS analysis.
Downloaded by [128.199.169.55] at 01:16 02 September 2015
www.landesbioscience.com Human Vaccines & Immunotherapeutics 2087
IM/EP vaccinated macaques.24 Thus, the delivery route (e.g.,
ID/EP) or inclusion of IL-12 cytokine DNA contributed sig-
nificantly to the induction of humoral responses with improved
breadth.
In contrast to the humoral responses, we found a significant
difference in the quantity of the cellular immune responses, with
the ID/EP delivery inducing lower responses. Despite this, both
vaccine regimens induced Env-specific transitional and effector
memory T cells and the cells contained the cytotoxic marker
granzyme B, a desired vaccine-induced T-cell response. Thus,
the cellular responses although different in magnitude, showed
the necessary potency as T-cell vaccine. As a matter of fact, the
potency of the vaccine reponses induced upon IM/EP DNA
delivery has been tested upon subsequent challenge of macaques
with pathogenic SIV25,26,2 8,48,49 and with SHIV50 and showed sig-
nificant reduction of viremia.
Different EP delivery methods developed by Inovio have
been reported which showed improved immunity including
HIV/SIV DNAs in macaques using intradermal low-current
EP,51 or using intradermal/subcutaneous EP,52 as well as devices
that deliver DNA simultaneously to both skin and muscle of
mice and guinea pigs53 and intradermal route in mice.54 Here,
we directly compared the immune responses induced upon IM
IM/EP immunized animals showed robust responses with
a frequency of 0.1–0.6% of Env-specific IFN-γ+ T cells
(Fig. 8A). Similar responses were measured in the 3 macaques
that were vaccinated only with gp160 DNAs (Fig. 8D). The
cellular responses induced against SIV Env were mediated
typically by both CD4+ and CD8+ T cells and, importantly,
these responses were ~10-fold higher in the IM/EP vaccinated
macaques (Fig. 8A and D) compared with the ID/EP
immunized animals (Fig. 8F). Detailed analysis of the
Env-specific responses of both groups of macaques showed
induction of transitional (CD28+CD95+CCR7) and effector
(CD28-CD95+CCR7) memory T cells (Fig. 8B, E and G)
containing the cytotoxic marker granzyme B (GzmB) (Fig. 8C
and H). Importantly, the T cell responses were also long-lasting
and could be detected for >1 y after the last vaccination. Together
these data demonstrate that both vaccination protocols induced
durable cytotoxic cellular immune responses although the IM/
EP delivery route induced higher levels of Env-specific T cell
responses.
Discussion
We have a long-standing interest in optimizing DNA-based
vaccine approaches, and thus, we have taken sequential steps
to improve efficacy of this vaccine vehicle including RNA or
codon optimization of the DNA expressing the immunogen,
optimization of expression vectors, antigen design, choice of
DNA adjuvants such as IL-12 and IL-15, and DNA delivery
methods.15,20,24-28,37-46 Our recent data on the combination of SIV
DNA and inactivated SIV particles, used as protein source in
the same vaccine regimen, revealed great potency to augment
humoral responses.28 To improve Env immunogenicity, we
describe herein the testing of a panel of transmitted SIV env
DNA and establishment of HEK293 cell lines stably producing
trimeric gp140 and soluble gp120 Env proteins, which will allow
us to further expand on this study by using purified Env proteins
instead of virus particles.
In this report, we tested different DNA-only delivery meth-
ods and compared the immunogenicity of the same cocktail of
expression-optimized SIV env DNAs using 2 different routes of
vaccine delivery in macaques: intradermal/EP and intramuscu-
lar/EP. We did not find significant differences in magnitude of
the humoral responses using these two DNA delivery methods.
Importantly, both vaccine regimens induced potent long-lasting
humoral responses, detectable for > 1 y. The IM/EP DNA deliv-
ery method was previously reported to induce long-lasting sys-
temic responses to HIV/SIV immunogens in macaques.24, 26 -28, 47
To test the breadth of the responses, we used a panel of SIVmac
and SIVsm Env sequences in pseudotype neutralization assays.
The ability to neutralize the tier 1B-like SIVsmE660_CG7V
Env served as a measure of neutralization strength. Interestingly,
we found more potent NAb to the heterologous, more difficult to
neutralize SIVsm E660_CG7V in macaques vaccinated via the
ID/EP route. These data are reminiscent of our previous report
where we found the presence of NAb to E660_CG7V only upon
inclusion of IL-12 DNA as vaccine adjuvant in the SIVmac239
Figure 4. Immunogenicity of Env DNAs in mice. (A) Outline of vaccina-
tion study of BALB/c mice. Mice (n = 5/group) were vaccinated via the
IM route (needle and syringe) at 0 and 4 weeks with SIV Env DNA for-
mulated in PBS, and were sacriced 2 week s after the 2nd vaccination.
Spleen and blood were analyzed. (B) Splenocytes were stimulated with
an Env peptide pool (15-mer, 11 AA overlap) and the IFN-γ producing T
cells were measured by ELISPOT assay. (C) The Env-specic binding an-
tibodies titers were measured by ELISA using serial dilutions of pooled
plasma samples.
Downloaded by [128.199.169.55] at 01:16 02 September 2015
2088 Human Vaccines & Immunotherapeutics Volume 9 Issue 10
Materials and Methods
Identification of transmitted SIVmac251 Env
sequences. The SIVmac251 stocks were generated
by propagating the original SIVmac251 isolate
in monkey peripheral blood mononuclear cells.56
Two stocks were used to infect rhesus macaques
(stock 2000: animals 35003, 35014, 33390; and
stock 06: animals M761, M766, M752, M757,
M764, M741) using low dose inocula. Plasma
samples collected at the peak of infection were
used for RNA extraction by PureLink viral
RNA isolation kit (Invitrogen). The full-length
env (2.86 kb) or the V1-V2 region (586 bp)
of the env gene were reverse transcribed with
Thermoscript (Invitrogen) using reverse outer
primer and amplified by nested PCR using High
Fidelity Platinum Taq Polymerase (Invitrogen)
and performed single genome sequencing assay
(SGA) as described.57 Approximately 10,000
copies of genomic RNA were amplified for each
sample at SGA. Thermal cycling conditions for
full-length env genes were as follows: an initial 1
min denaturation at 94 °C, followed by 35 cycles
of 94 °C for 15 sec, 55 °C for 30 sec, and 68 °C for
3 min; and 68 °C for 20 min extension. Thermal
cycling conditions for env V1-V2 were as follows:
an initial 1 min denaturation at 94 °C, followed
by 35 cycles of 94 °C for 30 sec, 54 °C for 30
sec, and 68 °C for 80 sec; and 68 °C for 3 min
extension. The primer position (corresponding
to SIVmac251 GenBank Accession
numberM19499) and sequence for full-length
env genes were: external sense primer: SIV251_
REV_outF 5'-CACATGCTAT TGTAAAAAGT GTTGCTA-3'
(64336459) and external antisense primer: SIV251 NEF_outR,
5'-TGTAATAAAT CCCTTCCAGT CC-3' (94409461),
internal sense primer: SIV251_REVinF, 5'-ACCATTGCCA
GTTTTGTTTT CTTA-3' (64596482), and internal
antisense primer: SIV251NEF_innerR, 5'-ATACCCCTAC
CA AGTCATCA TCT-3' (93379359). The PCR products were
sequenced with six sequencing primers as follows: SIV251_6532F
5'-TCCGAAAAAG GCTAAGGCTA ATAC-3', SIV251_6928F
5' TGAGACAGAT AGATGGGGAT TGACA-3', SIV251_8079R
5'-TTTATAATCT CCCA ACTCCA ATCG-3', SIV251_7888F
5'-AGACAAATAA TCAACACTTG GCATAAA-3',
SIV251_8404F 5'-AATGCTTGGG GATGTGCGTT TAG-
3', and SIV251_9359R 5'-ATACCCCTAC CA AGTCATCA
TCT-3'. The primer position and sequence for V1-V2 env were:
external sense primer: SIV251_V1–2_outF 5'-CTGCTTATCG
CCATCTTGCT TTTA-3' (66016624) and external
antisense primer: SIV251 V1–2_outR, 5'-CGTAATTCCT
TCTATGCCGT TCC-3' (78517873), internal sense
primer: T7-SIV251V1–2 inF, 5'-TAATACG ACT C ACTATA-
GGGGA ACA AC TCAG-3' (T7 + 67336755), and internal
antisense primer: M13R-SIV251V1–2inR, 5'-CAGGAAACAG
vs. ID delivery followed by EP of the same DNA mixtures into
macaques. Our data suggest that a combination of ID and IM
delivery is most effective in eliciting high humoral and cellular
responses. Indeed, such a delivery device has been developed and
shown superior results than either of the routes53 and should be
tested in the macaque model.
Mucosal dissemination is critical for an HIV/SIV vaccine,
since this is the route of natural infection. Although it has not
been tested yet for the intradermal/EP delivery method, the use of
intramuscular/EP elicited robust, long-lasting systemic immune
responses that disseminate into mucosal sites in vaccinated
macaques.27, 2 8 It was further shown that IM/EP did not increase
the activation state of SIV-specific CD4+ T cells in the gastro-
intestinal mucosa, thus no increase in target cells for HIV
infection was induced by this delivery method.55 Overall, our
study demonstrates that both routes of immunization followed
by EP induce robust and durable immune responses in macaques.
Thus, a combination of immune responses induced via ID and
IM delivery routes may be able to further improve immunity,
and to reduce virus acquisition. Further studies will be needed to
evaluate the efficacy of these two delivery systems upon challenge
with pathogenic SIV.
Figure 5. Production and purication of SIV Env protein from stable HEK293 cells. (A)
Samples of culture media from Fibercell grown HEK293 cells expressing SIV env protein
were assessed for purity on Coomassie blue stained SDS PAGE gels (10% TGX gel, Bio-Rad).
(B) Puried SIVsmE660_CG7V gp140, SIVmac gp140 (35014_7) and gp120 (35014/M766)
proteins were assessed for purity and quality on non-denaturing gel (10% TGX gel) and
visualized by Coomassie blue staining.
Downloaded by [128.199.169.55] at 01:16 02 September 2015
www.landesbioscience.com Human Vaccines & Immunotherapeutics 2089
CTAT-GACC ACCACC T TAGA AC-3' (73497365+M13R ).
Internal sense and antisense primers were flanking with T7 and
M13R sequence at the 5'end of the primers, respectively, so that
the sequencing was performed with T7 and M13R primers.
PCR products were amplified for sequencing using Applied
Biosystem’s BigDye Terminator v1.1 Ready Reaction mix
(Applied Biosystems). Sequencing products were detected using
Applied Biosystems 3730xl DNA analyzer. Sequencing products
were detected using Applied Biosystems 3730xl DNA analyzer.
Sequence alignments and phylogenetic analysis were performed
with the CUSTALW method using the Lasergene software
package (DNASTAR).
DNA vectors and in vitro expression. Selected Env sequences
were RNA-optimized and cloned into the CMVkan vector
comprising the CMV promoter, the bovine growth hormone
polyadenyation signal and the kanamycin gene in the plasmid
backbone.37 All env sequences a re submitted to GenBank a nd listed
in Table 1. GenBank submission for 35014 and 251_2 represent
optimize d consensus sequences. A ll plasmid DNAs were produced
in E. coli DH10B (Invitrogen) grown at 32 °C and endotoxin-
free DNA was purified (Qiagen). The DNAs were resuspended
in sterile water (Hospira, Inc.). Env production was measured in
supernatant and cell-associated fractions (1/200 of fraction) from
transfected HEK293 cells using Western immunoblot (1:5000
dilution of pooled sera from SIVmac251 infected macaques,
followed by 1:10,000 dilution of HRP-labeled anti-monkey
antiserum (Fitzgerald Industries International Inc.). The bands
were visualized using the enhanced chemiluminescence (ECL)
plus western blotting detection system (GE HealthCare).
Env protein production from stable HEK293 cells. Linearized
Env expression plasmids, purified using Nucleotide Removal Kit
Figure 7. Persistence of humoral responses in macaques vaccinated
via the intradermal and the intramuscular route. The animals were
monitored for 14–20 mo after the last vaccination. Humoral immune
responses were measured in the macaques that received DNA via the
intradermal (n = 3) and the intramuscular route (n = 5, with 3 macaques
receiving 1 mg DNA and 2 macaques receiving 2 mg DNA). (A) Endpoint
binding Ab titers to SIVmac239, (B) NAb titers to SIVmac_35014 (M766)
and (C) NAb to the heterologous SIVsmE660_CG7G (tier 1A-like) are
shown as log of reciprocal dilution of plasma that produces a 50%
reduction in signal compared with wells receiving no sample.
Figure 6. Analysis of humoral immune responses upon vaccination of
macaques with env DNA. (A) Outline of vaccination study of macaques
indicating the sequential immunization with DNA mixtures expressing
a cocktail of multivalent Env gp160 (EP1-EP3), gp120 (EP4) and gp140
(EP5). (B) Endpoint SIVmac251 Env binding Ab titers are shown for the
animals vaccinated ID/EP and the IM/EP DNA delivery regimen using
the same DNA mix tures.
Table2. Avidity of SIVmac239 Env binding antibody over time
Avidity measurements after last Env DNA vaccination
(in month)
Animal 0.5 1 3 6 10 14 20
ID/EP
M694 23.3 30.0 49.6
M710 30.3 31. 8 21. 3
M084 21.1 16. 8 ND
IM/EP
M092 21.1 32 .7 60.7
M095 47. 4 31.2 62.6
M705 25.9 20.4 20.3 30.8 35.7 32.5
M781 15. 3 14 . 2 12.8 26.2 29.1 28.8
M782 37. 0 39.9 42 .2 43.7 52.3 41.1
ND, not determined.
Downloaded by [128.199.169.55] at 01:16 02 September 2015
2090 Human Vaccines & Immunotherapeutics Volume 9 Issue 10
Figure 8. For gure legend, see page 2091.
Downloaded by [128.199.169.55] at 01:16 02 September 2015
www.landesbioscience.com Human Vaccines & Immunotherapeutics 2091
shallow ID DNA delivery using a novel 4 × 4 minimally invasive
needle array.51,61 This ID device differs from other invasive EP
devices in that the electrodes are minimally invasive (make con-
tact with the skin surface but do not penetrate) and also operate
at significantly lower voltages (15–25 V). The electrode configu-
ration is designed to achieve threshold electric fields over a wider
area at lower applied voltages. Specifically, the EP applicator con-
sists of gold-plated stainless steel needle electrodes with trocar
grinds at 1.5 mm spacing in a 4 × 4 array pattern. The surface
device was built with attachment cord for linkage to the ELGEN
1000 pulse generator (Inovio Pharmaceuticals). The following
Env DNA were used: SIVmac239 and SIVmac 251_15 DNAs
as gp160, gp140, gp120; SIVmac 35014 (M766) was included in
the gp160 and gp140 mixtures; SIVmac 35014_7 was included
in gp160 and gp120 mixtures; and SIVmac CR2.RU.3R1 was
included in the gp120 and gp140 mixtures. For ID vaccination,
macaques M084, M694 and M710 received a mixture of 1 mg
DNA (0.25 mg of each plasmid) resuspended in total of 0.25 ml
of sterile water, injected as 5 × 50 μl bleps on 3 places (2 × 50 μl
closely spaced [2× ]; 1 × 50 μl injected separately) on the back of
the animal, followed by EP. For the IM vaccination (macaques
M092 and M095) 1 mg of the env DNA mixture was resuspended
in 0.6 ml water and injected at 2 sites (0.3 ml each, left and right
internal thighs) followed by EP. The IM/EP was performed
using the ELGEN Twin Injector (Inovio Pharmaceuticals) which
consists of an outer housing with an inner wagon carrying two
standard 1 ml syringes with 21 g needles, 4 mm apart.62 A gear-
ing system presses the piston of the syringes when the wagon
slides forward in order to inject DNA during the insertion. The
needles subsequently serve as electrodes. The needles penetrate
the full depth of the targeted muscle, distributing the DNA in
a columnbular fashion throughout the muscle and co-locate the
electrical field with the delivered plasmid. The device operates at
an applied voltage of 60 and pulses twice, both of 60 ms dura-
tion. The ELGEN Twin Injector is directly linked to the ELGEN
1000 pulse generator (Inovio Pharmaceuticals). Macaques
(M705, M781, M782) received 3 vaccinations with 0.5 mg
DNA each of the same 4 gp160 DNAs resuspended separately in
0.3 ml each and injected at 4 different sites (arms and thighs).
Blood samples were collected at each vaccination and at various
time points throughout the course of vaccination to measure cel-
lular and humoral immune responses.
Humoral immune responses. The end-point binding
antibody titers to SIVmac251 gp120 were measured by ELISA
(Advanced BioScience Laboratory, Inc.) and titers greater than
the mean +3SD of normal plasma at OD450 were considered
positive. Antibody titers to SIVmac239 and antibody avidity
upon treatment with 1.5 M sodium thiocyanate (NaSCN;
Sigma-Aldrich) were measured as described.63,6 4 Neutralizing
(Qiagen), were used to transfected HEK293 cells together with
pRSVhygro as selection marker. Stable high producer HEK293
cell clones were generated using Env plasmids expressing
SIVmac251_35014 (M766) gp120 (plasmid 246S), 35014_7
gp140 (plasmid 234S) and SIVsmE660_CG7V gp120 (plasmid
261S). The following clones were among the highest producers
of 35014 (M766) gp120 (clone 36), 35014_7 gp140 (clone 47),
SIVsmE660_CG7V gp120 (clone X). Env proteins were produced
from selected stable clones grown in serum-free media in a Hollow
Fiber bioreactor that allows high-density growth without any
animal components (Fibercell®) as described.58 Supernatants (6 μl
of daily harvests of 20 ml) were monitored on gels stained with
Coomassie blue. Serial dilutions of the samples were analyzed,
together with purified SIV gp120 protein (a gift from E. Chertova)
to calibrate, after staining of the gels with Emerald Green.
Virus infectivity assay. Pseudovirus preparations were gener-
ated upon cotransfection of HEK293 cells with 5 μg pNL4–3.
LucR-E- (NIH AIDS Research and Reference reagent program,
Division of AIDS, NIAID;59) and 50 ng of the different SIV Env
expressing plasmids using the calcium phosphate method. Eight
hours following transfection, the culture medium was replaced
with fresh complete DMEM containing 10% FBS. Twenty-
four hours later, the cell supernatants were collected and 1 ml of
cell-free supernatant was added to 3 to 5 million of CEMx174-
GFP, an indicator cell line that contains the HIV-LTR promoter
linked to GFP stably integrated.60 After a 2-h incubation, 1 ml of
medium was added and the cells were incubated for 48 h. GFP
expression in infected cells was monitored by flow cytometry.
DNA vaccination of mice. Female BALB/c (6 to 8 weeks
old) were obtained from Charles River Laboratories, Inc. and
were housed at the National Cancer Institute in a temperature-
controlled, light-cycled facility. The mice were immunized by
intramuscular injection (week 0 and 4) with 100 μg plasmids
expressing the indicated Env gp160 plasmids. Two weeks after
the last vaccination, spleens and plasma were collected to mea-
sure cellular and humoral immune responses as described below.
DNA vaccination of macaques. This study was performed in
accordance with the Guide for the Care and Use of Laboratory
Animals of the National Institutes of Health. Rhesus macaques
were housed and handled in accordance with the standards of the
Association for the Assessment and Accreditation of Laboratory
Animal Care International at the Advanced BioScience
Laboratories Inc., and were approved by the Institutional Animal
Care and Use Committee (OLAW assurance number A3467-01
and USDA Certificate number 51-R-0059). Macaques received
a mixture of 4 env plasmids DNA (1 mg) via intradermal route
(ID) or via intramuscular route (IM) route followed by in vivo
electroporation (Inovio Pharmaceuticals, Inc.). The ID EP was
performed using the surface EP device (SEP) which allows for
Figure 8 (See opposite page). Comparison of magnitude and longevity of cellular responses in macaques vaccinated via the intramuscular (A–C,
D–E) and the intradermal (F–H) route. Cellular immune responses were measured in PBMC upon stimulation with a SIVmac239-specic peptide pool
(15-mer overlapping by 11 AA) at 2 weeks after vaccination with gp160 (EP3wk2), gp120 (EP4wk2) and gp140 (EP5wk2) (A–C, F–H). Animals in D–E
were monitored af ter the 3rd gp160 DNA vaccinations. All animals were monitored for ~1 y and the cellular responses were measured at EP5wk57 and
EP3wk43, respectively. The responses are shown as % Env-specic IFN-γ+ T cells among CD4+ and CD8+ (A, D, F); transitional (TM; CD28+CD95+CCR7)
and eector memory (EM; CD28CD95+CCR7) (B, E, G) T cells. The frequency of the Env-specic IFN-γ+ granzyme B+ T cells is shown (C, H). The ma-
caques of panels B and E were not analyzed for this function. Asterisk indicates that the sample scored negative for the assay.
Downloaded by [128.199.169.55] at 01:16 02 September 2015
2092 Human Vaccines & Immunotherapeutics Volume 9 Issue 10
Disclosure of Potential Conflicts of Interest
GNP and BKF are inventors on US Government-owned patents
and patent applications related to DNA vaccines and gene expres-
sion optimization that have been licensed to several companies.
There are no further patents, products in development or mar-
keted products to declare. KEB and NS are employed by Inovio
Pharmaceuticals, Inc. as such receive salary, and bonuses and
stock options as compensation.
BKF, GNP, AV: designed, coordinated the study, analyzed the
data, and wrote the paper. VK, MR, JB, GRP, RJ, CB, AKS,
CA, BC, G-MZ: performed experiments and analyzed the data.
E-YK, SMW: performed Env identification study. WH, YG: per-
formed binding Ab and avidity assays. CL, DCM: performed
and analyzed NAb assays. KEB, NYS: contributed electropora-
tion delivery methods and electroporation devices to access IM
and ID tissues for immunization.
Acknowledgments
We are grateful to D Weiss, J Treece, I Kalisz, R Pal and staff
at Advanced BioScience Laboratories, Inc., Rockville, for their
expert help. We thank A von Gegerfelt, B Keele, JJS Cadwell, and
E Chertova for discussions, R Desrosiers, R Pal, and NIAID for
SIVmac251 virus and macaque plasma samples, and T Jones for
editorial a ssistance. pNL4 -3.LucR-E- wa s provided by NI H AIDS
Research and Reference reagent program, Division of AIDS,
NIAID. This work was supported by the Intramural Research
Program of the National Cancer Institute, National Institutes of
Health (NCI/NIH) and by NIH HHSN 27201100016C.
antibody titers were determined using the M7-luc assay for the
TCLA-SIVmac251/H9 and the TZM-bl assay for the transmitted
SIVmac251 35014_7 and 35014 (M766), and the heterologous
SI Vsm E 66 0 _ C G7G a nd SIVsmE 6 6 0 _ C G7 V.65
Cellular immune responses. Mouse splenocytes were incu-
bated with a pool of overlapping 15-mer peptides (1 μg/ml final
concentration) spanning the entire gp160 region of SIVmac239
(Infinity Inc. Biotech Research and Resource) and ELIspot assays
were performed as described.38 Env-specific spots were calculated
by subtracting the cut-off value and adjusted to the number of
spot-forming cells per million splenocytes.
The frequency of antigen-specific T cells from vaccinated
macaques was determined upon stimulation with the Env pep-
tide pool followed by intracellular cytokine staining and multi-
parametric flow cytometry as described.28 The antibody panel
for surface staining consisted of: CD3-APCCy7, CD4-AmCyan,
CD95-FITC (BD PharMingen), CD8-AF-405 (Invitrogen),
CD28-PerCP Cy5.5 (BioLegend), CCR7-APC (R&D Systems
Inc.) and CD45RA-AF700 (ABD Serotec). The intracellular
staining was performed using IFN-γ-PE Cy7 (BD PharMingen)
and granzyme B-PE (Invitrogen) antibodies. PBMC were cul-
tured in medium without peptide pools as negative control or
stimulated with phorbol myristate acetate (PMA) and calcium
ionophore (Sigma) as positive control. Positive samples had a fre-
quency of IFN-γ positivity in the peptide-stimulated sample that
was > 2 fold higher than the frequency obtained in unstimulated
(minus peptide) medium only control sample. At least 105 T cells
from each sample were acquired on an LSR II flow cytometer
(BD Biosciences) and the data were analyzed using FlowJo soft-
ware (Tree Star, Inc.).
References
1. Sardesai NY, Weiner DB. Electroporation delivery
of DNA vaccines: prospects for success. Curr Opin
Immunol 2011; 23:421-9; PMID:21530212; http://
dx.doi.org/10.1016/j.coi.2011.03.008
2. Hutnick NA, Myles DJ, Bian CB, Muthumani K,
Weiner DB. Selected approaches for increasing HIV
DNA vaccine immunogenicity in vivo. Curr Opin
Virol 2011; 1:233-40; PMID:22440782; http://dx.doi.
org/10.1016/j.coviro.2011.08.003
3. Bodles-Brakhop AM, Heller R, Draghia-Akli R.
Electroporation for the delivery of DNA-based vaccines
and immunotherapeutics: current clinical develop-
ments. Mol Ther 2009; 17:585-92; PMID:19223870;
http://dx.doi.org/10.1038/mt.2009.5
4. Lori F. DermaVir: a plasmid DNA-based nano-
medicine therapeutic vaccine for the treatment of
HIV/AIDS. Expert Rev Vaccines 2011; 10:1371-
84; PMID:21988301; http://dx.doi.org/10.1586/
erv.11.118
5. Sullivan SM, Doukas J, Hartikka J, Smith L, Rolland
A. Vaxfectin: a versatile adjuvant for plasmid DNA-
and protein-based vaccines. Expert Opin Drug Deliv
2010; 7:1433-46; PMID:21118032; http://dx.doi.org
/10.1517/17425247.2010.538047
6. Hallengärd D, Bråve A, Isaguliants M, Blomberg P,
Enger J, Stout R, et al. A combination of intradermal
jet-injection and electroporation overcomes in vivo
dose restriction of DNA vaccines. Genet Vaccines
Ther 2012; 10:5; PMID:22873174; http://dx.doi.
org/10.1186/1479-0556-10-5
7. Roos AK, Eriksson F, Timmons JA, Gerhardt J, Nyman
U, Gudmundsdotter L, et al. Skin electroporation:
effects on transgene expression, DNA persistence and
local tissue environment. PLoS One 2009; 4:e7226;
PMID:19789652; http://dx.doi.org/10.1371/journal.
pone.0007226
8. Fuller DH, Simpson L, Cole KS, Clements JE, Panicali
DL, Montelaro RC, et al. Gene gun-based nucleic acid
immunization alone or in combination with recom-
binant vaccinia vectors suppresses virus burden in
rhesus macaques challenged with a heterologous SIV.
Immunol Cell Biol 1997; 75:389-96; PMID:9315483;
http://dx.doi.org/10.1038/icb.1997.61
9. Aihara H, Miyazaki J. Gene transfer into muscle by
electroporation in vivo. Nat Biotechnol 1998; 16:867-
70; PMID:9743122; http://dx.doi.org/10.1038/
nbt0998-867
10. Mathiesen I. Electropermeabilization of skeletal muscle
enhances gene transfer in vivo. Gene Ther 1999; 6:508-
14; PMID:10476210; http://dx.doi.org/10.1038/
sj.gt.3300847
11. Prud’homme GJ, Glinka Y, Khan AS, Draghia-Akli
R. Electroporation-enhanced nonviral gene transfer
for the prevention or treatment of immunological,
endocrine and neoplastic diseases. Curr Gene Ther
2006; 6:243-73; PMID:16611045; http://dx.doi.
org/10.2174/156652306776359504
12. Rizzuto G, Cappelletti M, Maione D, Savino R,
Lazzaro D, Costa P, et al. Efficient and regulated
erythropoietin production by naked DNA injection
and muscle electroporation. Proc Natl Acad Sci U S
A 1999; 96:6417-22; PMID:10339602; http://dx.doi.
org/10.1073/pnas.96.11.6417
13. Widera G, Austin M, Rabussay D, Goldbeck C,
Barnett SW, Chen M, et al. Increased DNA vaccine
delivery and immunogenicity by electroporation in
vivo. J Immunol 2000; 164:4635-40; PMID:10779767
14. Otten G, Schaefer M, Doe B, Liu H, Srivastava I,
zur Megede J, et al. Enhancement of DNA vaccine
potency in rhesus macaques by electroporation. Vaccine
2004; 22:2489-93; PMID:15193413; http://dx.doi.
org/10.1016/j.vaccine.2003.11.073
15. Rosati M, Valentin A, Jalah R, Patel V, von Gegerfelt
A, Bergamaschi C, et al. Increased immune responses
in rhesus macaques by DNA vaccination combined
with electroporation. Vaccine 2008; 26:5223-9;
PMID:18468743; http://dx.doi.org/10.1016/j.vac-
cine.2008.03.090
16. Otten GR, Schaefer M, Doe B, Liu H, Megede
JZ, Donnelly J, et al. Potent immunogenicity of an
HIV-1 gag-pol fusion DNA vaccine delivered by
in vivo electroporation. Vaccine 2006; 24:4503-9;
PMID:16181711; http://dx.doi.org/10.1016/j.vac-
cine.2005.08.017
17. Luckay A, Sidhu MK, Kjeken R, Megati S, Chong SY,
Roopchand V, et al. Effect of plasmid DNA vaccine
design and in vivo electroporation on the resulting
vaccine-specific immune responses in rhesus macaques.
J Virol 2007; 81:5257-69; PMID:17329330; http://
dx.doi.org/10.1128/JVI.00055-07
18. Hirao LA, Wu L, Khan AS, Hokey DA, Yan J, Dai A,
et al. Combined effects of IL-12 and electroporation
enhances the potency of DNA vaccination in macaques.
Vaccine 2008; 26:3112-20; PMID:18430495; http://
dx.doi.org/10.1016/j.vaccine.2008.02.036
Downloaded by [128.199.169.55] at 01:16 02 September 2015
www.landesbioscience.com Human Vaccines & Immunotherapeutics 2093
44. Valentin A, Chikhlikar P, Patel V, Rosati M, Maciel
M, Chang KH, et al. Comparison of DNA vaccines
producing HIV-1 Gag and LAMP/Gag chimera in
rhesus macaques reveals antigen-specific T-cell responses
with distinct phenotypes. Vaccine 2009; 27:4840-
9; PMID:19539586; http://dx.doi.org/10.1016/j.
vaccine.2009.05.093
45. Nasioulas G, Zolotukhin AS, Tabernero C, Solomin L,
Cunningham CP, Pavlakis GN, et al. Elements distinct
from human immunodeficiency virus type 1 splice sites
are responsible for the Rev dependence of env mRNA.
J Virol 1994; 68:2986-93; PMID:8151769
46. von Gegerfelt AS, Rosati M, Alicea C, Valentin A,
Roth P, Bear J, et al. Long-lasting decrease in vire-
mia in macaques chronically infected with simian
immunodeficiency virus SIVmac251 after therapeu-
tic DNA immunization. J Virol 2007; 81:1972-9;
PMID:17135321; http://dx.doi.org/10.1128/
JVI.01990-06
47. Cristillo AD, Weiss D, Hudacik L, Restrepo S, Galmin
L, Suschak J, et al. Persistent antibody and T cell
responses induced by HIV-1 DNA vaccine delivered
by electroporation. Biochem Biophys Res Commun
2008; 366:29-35; PMID:18036339; http://dx.doi.
org/10.1016/j.bbrc.2007.11.052
48. Hirao LA, Hokey DA, Morrow MP, Jure-Kunkel MN,
Weiner DB. Immune modulation through 4-1BB
enhances SIV vaccine protection in non-human
primates against SIVmac251 challenge. PLoS One
2011; 6:e24250; PMID:21935390; http://dx.doi.
org/10.1371/journal.pone.0024250
49. Hutnick NA, Myles DJ, Hirao L, Scott VL, Ferraro B,
Khan AS, et al. An optimized SIV DNA vaccine can
serve as a boost for Ad5 and provide partial protec-
tion from a high-dose SIVmac251 challenge. Vaccine
2012; 30:3202-8; PMID:22406458; http://dx.doi.
org/10.1016/j.vaccine.2012.02.069
50. Yin J, Dai A, Lecureux J, Arango T, Kutzler MA, Yan
J, et al. High antibody and cellular responses induced
to HIV-1 clade C envelope following DNA vaccines
delivered by electroporation. Vaccine 2011; 29:6763-
70; PMID:21195801; http://dx.doi.org/10.1016/j.vac-
cine.2010.12.055
51. Hutnick NA, Myles DJ, Ferraro B, Lucke C, Lin F,
Yan J, et al. Intradermal DNA vaccination enhanced
by low-current electroporation improves antigen
expression and induces robust cellular and humoral
immune responses. Hum Gene Ther 2012; 23:943-
50; PMID:22650607; http://dx.doi.org/10.1089/
hum.2012.055
52. Hirao LA, Wu L, Khan AS, Satishchandran A, Draghia-
Akli R, Weiner DB. Intradermal/subcutaneous immu-
nization by electroporation improves plasmid vaccine
delivery and potency in pigs and rhesus macaques.
Vaccine 2008; 26:440-8; PMID:18082294; http://
dx.doi.org/10.1016/j.vaccine.2007.10.041
53. Lin F, Shen X, McCoy JR, Mendoza JM, Yan J,
Kemmerrer SV, et al. A novel prototype device for
electroporation-enhanced DNA vaccine delivery
simultaneously to both skin and muscle. Vaccine
2011; 29:6771-80; PMID:21199706; http://dx.doi.
org/10.1016/j.vaccine.2010.12.057
54. Lin F, Shen X, Kichaev G, Mendoza JM, Yang M,
Armendi P, et al. Optimization of electroporation-
enhanced intradermal delivery of DNA vaccine using
a minimally invasive surface device. Hum Gene Ther
Methods 2012; 23:157-68; PMID:22794496; http://
dx.doi.org/10.1089/hgtb.2011.209
55. Reuter MA, Yuan S, Marx PA, Kutzler MA, Weiner
DB, Betts MR. DNA-based HIV vaccines do not
induce generalized activation in mucosal tissue T
cells. Hum Vaccin Immunother 2012; 8:1648-53;
PMID:23111167; http://dx.doi.org/10.4161/
hv.22247
56. Daniel MD, Letvin NL, King NW, Kannagi M,
Sehgal PK, Hunt RD, et al. Isolation of T-cell trop-
ic HTLV-III-like retrovirus from macaques. Science
1985; 228:1201-4; PMID:3159089; http://dx.doi.
org/10.1126/science.3159089
31. Vasan S, Hurley A, Schlesinger SJ, Hannaman D,
Gardiner DF, Dugin DP, et al. In vivo electroporation
enhances the immunogenicity of an HIV-1 DNA
vaccine candidate in healthy volunteers. PLoS One
2011; 6:e19252; PMID:21603651; http://dx.doi.
org/10.1371/journal.pone.0019252
32. Kalams SA, Parker S, Jin X, Elizaga M, Metch B, Wang
M, et al.; NIAID HIV Vaccine Trials Network. Safety
and immunogenicity of an HIV-1 gag DNA vaccine
with or without IL-12 and/or IL-15 plasmid cytokine
adjuvant in healthy, HIV-1 uninfected adults. PLoS
One 2012; 7:e29231; PMID:22242162; http://dx.doi.
org/10.1371/journal.pone.0029231
33. Del Prete GQ, Scarlotta M, Newman L, Reid C,
Parodi LM, Roser JD, et al. Comparative character-
ization of transfection- and infection-derived simian
immunodeficiency virus challenge stocks for in vivo
nonhuman primate studies. J Virol 2013; 87:4584-
95; PMID:23408608; http://dx.doi.org/10.1128/
JVI.03507-12
34. Keele BF, Li H, Learn GH, Hraber P, Giorgi EE,
Grayson T, et al. Low-dose rectal inoculation of rhesus
macaques by SIVsmE660 or SIVmac251 recapitulates
human mucosal infection by HIV-1. J Exp Med
2009; 206:1117-34; PMID:19414559; http://dx.doi.
org/10.1084/jem.20082831
35. O’Rourke SM, Schweighardt B, Phung P, Fonseca
DP, Terry K, Wrin T, et al. Mutation at a single
position in the V2 domain of the HIV-1 envelope
protein confers neutralization sensitivity to a highly
neutralization-resistant virus. J Virol 2010; 84:11200-
9; PMID:20702624; http://dx.doi.org/10.1128/
JVI.00790-10
36. Yuste E, Johnson W, Pavlakis GN, Desrosiers RC.
Virion envelope content, infectivity, and neutralization
sensitivity of simian immunodeficiency virus. J Virol
2005; 79:12455-63; PMID:16160173; http://dx.doi.
org/10.1128/JVI.79.19.12455-12463.2005
37. Rosati M, von Gegerfelt A, Roth P, Alicea C, Valentin
A, Robert-Guroff M, et al. DNA vaccines expressing
different forms of simian immunodeficiency virus
antigens decrease viremia upon SIVmac251 challenge.
J Virol 2005; 79:8480-92; PMID:15956591; http://
dx.doi.org/10.1128/JVI.79.13.8480-8492.2005
38. Kulkarni V, Jalah R, Ganneru B, Bergamaschi C, Alicea
C, von Gegerfelt A, et al. Comparison of immune
responses generated by optimized DNA vaccination
against SIV antigens in mice and macaques. Vaccine
2011; 29:6742-54; PMID:21195080; http://dx.doi.
org/10.1016/j.vaccine.2010.12.056
39. Kulkarni V, Rosati M, Valentin A, Ganneru B, Singh
AK, Yan J, et al. HIV-1 p24(gag) derived conserved
element DNA vaccine increases the breadth of
immune response in mice. PLoS One 2013; 8:e60245;
PMID:23555935; http://dx.doi.org/10.1371/journal.
pone.0060245
40. Kutzler MA, Robinson TM, Chattergoon MA, Choo
DK, Choo AY, Choe PY, et al. Coimmunization with
an optimized IL-15 plasmid results in enhanced func-
tion and longevity of CD8 T cells that are partially
independent of CD4 T cell help. J Immunol 2005;
175:112-23; PMID:15972637
41. Santra S, Liao HX, Zhang R, Muldoon M, Watson
S, Fischer W, et al. Mosaic vaccines elicit CD8+ T
lymphocyte responses that confer enhanced immune
coverage of diverse HIV strains in monkeys. Nat
Med 2010; 16:324-8; PMID:20173754; http://dx.doi.
org/10.1038/nm.2108
42. Schwartz S, Campbell M, Nasioulas G, Harrison J,
Felber BK, Pavlakis GN. Mutational inactivation of
an inhibitory sequence in human immunodeficiency
virus type 1 results in Rev-independent gag expression.
J Virol 1992; 66:7176-82; PMID:1433510
43. Schwartz S, Felber BK, Pavlakis GN. Distinct
RNA sequences in the gag region of human
immunodeficiency virus type 1 decrease RNA stability
and inhibit expression in the absence of Rev protein. J
Virol 1992; 66:150-9; PMID:1727477
19. Boyer JD, Robinson TM, Kutzler MA, Parkinson
R, Calarota SA, Sidhu MK, et al. SIV DNA vaccine
co-administered with IL-12 expression plasmid
enhances CD8 SIV cellular immune responses in
cynomolgus macaques. J Med Primatol 2005; 34:262-
70; PMID:16128921; http://dx.doi.org/10.1111/
j.1600-0684.2005.00124.x
20. Schadeck EB, Sidhu M, Egan MA, Chong SY, Piacente
P, Masood A, et al. A dose sparing effect by plasmid
encoded IL-12 adjuvant on a SIVgag-plasmid DNA
vaccine in rhesus macaques. Vaccine 2006; 24:4677-
87; PMID:16288822; http://dx.doi.org/10.1016/j.vac-
cine.2005.10.035
21. Schadeck EB, Sidhu M, Egan MA, Chong SY, Piacente
P, Masood A, et al. A dose sparing effect by plasmid
encoded IL-12 adjuvant on a SIVgag-plasmid DNA
vaccine in rhesus macaques. Vaccine 2006; 24:4677-
87; PMID:16288822; http://dx.doi.org/10.1016/j.vac-
cine.2005.10.035
22. Robinson TM, Sidhu MK, Pavlakis GN, Felber BK,
Silvera P, Lewis MG, et al. Macaques co-immu-
nized with SIVgag/pol-HIVenv and IL-12 plasmid
have increased cellular responses. J Med Primatol
2007; 36:276-84; PMID:17669216; http://dx.doi.
org/10.1111/j.1600-0684.2007.00245.x
23. Halwani R, Boyer JD, Yassine-Diab B, Haddad EK,
Robinson TM, Kumar S, et al. Therapeutic vaccination
with simian immunodeficiency virus (SIV)-DNA +
IL-12 or IL-15 induces distinct CD8 memory subsets
in SIV-infected macaques. J Immunol 2008; 180:7969-
79; PMID:18523260
24. Jalah R, Patel V, Kulkarni V, Rosati M, Alicea C,
Ganneru B, et al. IL-12 DNA as molecular vac-
cine adjuvant increases the cytotoxic T cell responses
and breadth of humoral immune responses in SIV
DNA vaccinated macaques. Hum Vaccin Immunother
2012; 8:1620-9; PMID:22894956; http://dx.doi.
org/10.4161/hv.21407
25. Winstone N, Wilson AJ, Morrow G, Boggiano C,
Chiuchiolo MJ, Lopez M, et al. Enhanced con-
trol of pathogenic Simian immunodeficiency virus
SIVmac239 replication in macaques immunized with
an interleukin-12 plasmid and a DNA prime-viral
vector boost vaccine regimen. J Virol 2011; 85:9578-
87; PMID:21734035; http://dx.doi.org/10.1128/
JVI.05060-11
26. Rosati M, Bergamaschi C, Valentin A, Kulkarni V,
Jalah R, Alicea C, et al. DNA vaccination in rhe-
sus macaques induces potent immune responses and
decreases acute and chronic viremia after SIVmac251
challenge. Proc Natl Acad Sci U S A 2009; 106:15831-
6; PMID:19717425; http://dx.doi.org/10.1073/
pnas.0902628106
27. Patel V, Valentin A, Kulkarni V, Rosati M, Bergamaschi
C, Jalah R, et al. Long-lasting humoral and cel-
lular immune responses and mucosal dissemination
after intramuscular DNA immunization. Vaccine
2010; 28:4827-36; PMID:20451642; http://dx.doi.
org/10.1016/j.vaccine.2010.04.064
28. Patel V, Jalah R, Kulkarni V, Valentin A, Rosati M,
Alicea C, et al. DNA and virus particle vaccination
protects against acquisition and confers control of vire-
mia upon heterologous simian immunodeficiency virus
challenge. Proc Natl Acad Sci U S A 2013; 110:2975-
80; PMID:23359688; http://dx.doi.org/10.1073/
pnas.1215393110
29. Hirao LA, Draghia-Akli R, Prigge JT, Yang M,
Satishchandran A, Wu L, et al. Multivalent smallpox
DNA vaccine delivered by intradermal electropora-
tion drives protective immunity in nonhuman pri-
mates against lethal monkeypox challenge. J Infect Dis
2011; 203:95-102; PMID:21148501; http://dx.doi.
org/10.1093/infdis/jiq017
30. Laddy DJ, Yan J, Khan AS, Andersen H, Cohn
A, Greenhouse J, et al. Electroporation of synthetic
DNA antigens offers protection in nonhuman primates
challenged with highly pathogenic avian influenza
virus. J Virol 2009; 83:4624-30; PMID:19211745;
http://dx.doi.org/10.1128/JVI.02335-08
Downloaded by [128.199.169.55] at 01:16 02 September 2015
2094 Human Vaccines & Immunotherapeutics Volume 9 Issue 10
63. Guan Y, Sajadi MM, Kamin-Lewis R, Fouts TR,
Dimitrov A, Zhang Z, et al. Discordant memory B cell
and circulating anti-Env antibody responses in HIV-1
infection. Proc Natl Acad Sci U S A 2009; 106:3952-
7; PMID:19225108; http://dx.doi.org/10.1073/
pnas.0813392106
64. Moore JP, Wallace LA, Follett EA, McKeating JA. An
enzyme-linked immunosorbent assay for antibodies to
the envelope glycoproteins of divergent strains of HIV-
1. AIDS 1989; 3:155-63; PMID:2540772; http://
dx.doi.org/10.1097/00002030-198903000-00006
65. Montefiori DC. Evaluating neutralizing antibodies
against HIV, SIV, and SHIV in luciferase reporter gene
assays. Curr Protoc Immunol 2005; Chapter 12:Unit
12 1.
60. von Gegerfelt AS, Liska V, Li PL, McClure HM,
Horie K, Nappi F, et al. Rev-independent simian
immunodeficiency virus strains are nonpathogenic
in neonatal macaques. J Virol 2002; 76:96-104;
PMID:11739675; http://dx.doi.org/10.1128/
JVI.76.1.96-104.2002
61. Broderick KE, Shen X, Soderholm J, Lin F, McCoy J,
Khan AS, et al. Prototype development and preclinical
immunogenicity analysis of a novel minimally inva-
sive electroporation device. Gene Ther 2011; 18:258-
65; PMID:20962869; http://dx.doi.org/10.1038/
gt.2010.137
62. Tjelle TE, Salte R, Mathiesen I, Kjeken R. A novel
electroporation device for gene delivery in large
animals and humans. Vaccine 2006; 24:4667-70;
PMID:16162367; http://dx.doi.org/10.1016/j.
vaccine.2005.08.068
57. Palmer S, Kearney M, Maldarelli F, Halvas EK, Bixby
CJ, Bazmi H, et al. Multiple, linked human immuno-
deficiency virus type 1 drug resistance mutations in
treatment-experienced patients are missed by standard
genotype analysis. J Clin Microbiol 2005; 43:406-
13; PMID:15635002; http://dx.doi.org/10.1128/
JCM.43.1.406-413.2005
58. Chertova E, Bergamaschi C, Chertov O, Sowder R,
Bear J, Roser JD, et al. Characterization and Favorable
in Vivo Properties of Heterodimeric Soluble IL-15·IL-
15Rα Cytokine Compared to IL-15 Monomer. J
Biol Chem 2013; 288:18093-103; PMID:23649624;
http://dx.doi.org/10.1074/jbc.M113.461756
59. Connor RI, Chen BK, Choe S, Landau NR. Vpr
is required for efficient replication of human
immunodeficiency virus type-1 in mononuclear
phagocytes. Virology 1995; 206:935-44;
PMID:7531918; http://dx.doi.org/10.1006/
viro.1995.1016
Downloaded by [128.199.169.55] at 01:16 02 September 2015
... Thirdly, experiments and pulsing protocols are designed and executed according to the equipment and expertise available to the research group 29 . The CELLECTRA™ 3P Electroporation device by Inovio Pharmaceuticals, a handheld array of electrodes with 16 gold plated electrodes (4 × 4 configuration), has been used to deliver EP pulses to the surfaces of rat 30 or macaque skin 25,31 . Other groups have used the Pulse Agile EO equipment and software (CytoPulse Sciences) to deliver their EP pulses through a 7 × 2 array of stainless steel acupuncture needles 29 and the commercially available DermaVax DNA vaccine delivery system (Cellectis, Paris, France, formerly CytoPulse Sciences) to vaccinate mice using a 8 × 2 32 or 6 × 2 array 33 of electrodes. ...
... This dose-sparing effect has been reported in nonhuman primate studies for Dengue virus (serotype1-4) 49 , a murine model of HIV-1 (type I envelope) 50 , and others 51,52 . The prolonged immune responses occur in resident memory cells that continue to present the antibody of interest in sufficient quantities to prevent illness against lethal challenges up to 20 months after immunization 31 . While most naturally incurring infections will not be lethal doses of virus, for people with compromised or suppressed immune systems, even a nonlethal challenge could have severe repercussions such as liver, kidney, or lung damage, as observed with the recent pandemic of SARS-CoV-2 virus (COVID-19). ...
Article
With the resurgence of illnesses, such as measles, it is of utmost importance that a high percentage of the local and global population be vaccinated. Diseases, such as measles, HepB, Hib, and others, are suppressed when 93%–95% of a population has been vaccinated against them, generating herd immunity. Lack of vaccination can lead to lifelong consequences. Barriers to vaccination include costs, lack of healthcare worker knowledge, reliance on cold chain storage, age, and personal beliefs. Vaccinations are usually administered as subcutaneous or intramuscular injections, though some oral vaccines exist. The use of intradermal or intramuscular electroporation (ID-EP and IM-EP) as an adjuvant has demonstrated benefits over traditional vaccination methods, while also having some issues, which require careful consideration. Issues for consideration include a lack of standardized equipment and pulsing protocols while benefits include dose sparing, long-term immunogenicity, and higher titer levels after a single dose compared to the gold standard. ID-EP has been shown to have better transfection rates over IM-EP, while IM-EP has demonstrated faster seroconversion rates (SRs) over IM injection alone. Improvements such as standardizing equipment with larger injection volumes, multiple EP site locations and/or disposable, predosed, lyophilized vaccine cartridges could contribute to more rapid deployment of vaccines without relying on cold chain storage and trained healthcare workers, at a reduced cost while still providing adequate levels of immunity against disease.
... However, studies have suggested that i.m. DNA vaccination, with or without EP, is more efficient at inducing cellular immune responses in both mice and macaques 23,24 . In this study we compare the use of Xcl1 and Ccl3-fusion DNA vaccines for the induction of immune responses against influenza A hemagglutinin (HA). ...
... Indeed, most studies do not includes electroporation of the injection sites 23,44,45 , or only includes electroporation for one of the delivery methods 46 . Nevertheless, delivery of DNA by i.m. injection has been reported to be a more efficient method for inducing Th1 associated immune responses such as IgG2a antibodies 23,44 , in addition to inducing stronger cytotoxic T cell responses 24,44 . In contrast, delivery of DNA i.d. has been reported to induce a more Th2/IgG1 polarized response 23,44 , and more protective antibody responses 46 . ...
Article
Full-text available
Fusing antigens to chemokines to target antigen presenting cells (APC) is a promising method for enhancing immunogenicity of DNA vaccines. However, it is unclear how different chemokines compare in terms of immune potentiating effects. Here we compare Ccl3- and Xcl1-fusion vaccines containing hemagglutinin (HA) from influenza A delivered by intramuscular (i.m.) or intradermal (i.d.) DNA vaccination. Xcl1 fusion vaccines target cDC1s, and enhance proliferation of CD4+ and CD8+ T cells in vitro. In contrast, Ccl3 target both cDC1 and cDC2, but only enhance CD4+ T cell proliferation in combination with cDC2. When Ccl3- or Xcl1-HA fusion vaccines were administered by i.m. DNA immunization, both vaccines induced Th1-polarized immune responses with antibodies of the IgG2a/IgG2b subclass and IFNγ-secreting T cells. After i.d. DNA vaccination, however, only Xcl1-HA maintained a Th1 polarized response and induced even higher numbers of IFNγ-secreting T cells. Consequently, Xcl1-HA induced superior protection against influenza infection compared to Ccl3-HA after i.d. immunization. Interestingly, i.m. immunization with Ccl3-HA induced the strongest overall in vivo cytotoxicity, despite not inducing OT-I proliferation in vitro. In summary, our results highlight important differences between Ccl3- and Xcl1- targeted DNA vaccines suggesting that chemokine fusion vaccines can be tailor-made for different diseases.
... Among the nucleic acid-based vaccines, the DNA platform elicits long-lasting adaptive responses with both CD4 + and cytotoxic CD8 + T cell responses in macaques and humans (30,(45)(46)(47)(48)(49)(50)(51)(52). mRNA vaccines are very efficient in inducing humoral immunity and mainly CD4 + T helper responses against several antigens (32,35,36,(53)(54)(55)(56). ...
Article
Full-text available
Immunogenicity of HIV-1 mRNA vaccine regimens was analyzed in a non-human primate animal model. Rhesus macaques immunized with mRNA in lipid nanoparticle (mRNA/LNP) formulation expressing HIV-1 Gag and Gag conserved regions (CE) as immunogens developed robust, durable antibody responses but low adaptive T-cell responses. Augmentation of the dose resulted in modest increases in vaccine-induced cellular immunity, with no difference in humoral responses. The gag mRNA/lipid nanoparticle (LNP) vaccine provided suboptimal priming of T cell responses for a heterologous DNA booster vaccination regimen. In contrast, a single immunization with gag mRNA/LNP efficiently boosted both humoral and cellular responses in macaques previously primed by a gag DNA-based vaccine. These anamnestic cellular responses were mediated by activated CD8⁺ T cells with a phenotype of differentiated T-bet⁺ cytotoxic memory T lymphocytes. The heterologous prime/boost regimens combining DNA and mRNA/LNP vaccine modalities maximized vaccine-induced cellular and humoral immune responses. Analysis of cytokine responses revealed a transient systemic signature characterized by the release of type I interferon, IL-15 and IFN-related chemokines. The pro-inflammatory status induced by the mRNA/LNP vaccine was also characterized by IL-23 and IL-6, concomitant with the release of IL-17 family of cytokines. Overall, the strong boost of cellular and humoral immunity induced by the mRNA/LNP vaccine suggests that it could be useful as a prophylactic vaccine in heterologous prime/boost modality and in immune therapeutic interventions against HIV infection or other chronic human diseases.
... 31,32 Besides being employed for enhancing transcutaneous gene delivery, electroporation is also helpful for promoting intracellular DNA vaccine delivery and is used locally after intramuscular injection or intradermal injection. 33 To decrease the discomfort and skin damage, a non-invasive device such as a multi-electrode array has been designed to minimize the applied voltage by maintaining a short electrode distance. 34 Moreover, the depth of penetration of an electric field produced by this electrode device is much easier to control. ...
Article
Therapeutic vaccination is a promising strategy in immunotherapy of cancers. It eradicates cancer cells by evoking and strengthening patients’ own immune system. Because of the easy access and sophisticated immune networks, the skin becomes an ideal target organ for vaccination. Genetic vaccines have been widely investigated, which have advantages of delivery of multiple antigens and less cost for production over protein/peptide vaccines. This review summarizes the advances in transcutaneous delivery of DNA/mRNA for cancer therapeutic vaccination, and gives a brief description of the immunological milieu of the skin and the importance of DC‐targeting in vaccine delivery, as well as the technologies that aim at facilitating antigen delivery and modulating antigen‐presenting cells to improve cellular responses. The applications of genetic vaccines encoding tumor antigens delivered through the skin route both in preclinical and clinical trials are outlined.
... Electroporation (EP) has been used both in pre-clinical and clinical studies to augment the plasmid DNA immunogenicity [19,20]. EP increases the transfection efficiency into antigen- presenting cells (APCs) by creating transient pores in the cell membranes increasing the uptake of DNA. ...
Article
Full-text available
Background We evaluated the safety and immunogenicity of (i) an intradermal HIV-DNA regimen given with/without intradermal electroporation (EP) as prime and (ii) the impact of boosting with modified vaccinia virus Ankara (HIV-MVA) administered with or without subtype C CN54rgp140 envelope protein adjuvanted with Glucopyranosyl Lipid A (GLA-AF) in volunteers from Tanzania and Mozambique.Methods Healthy HIV-uninfected adults (N = 191) were randomized twice; first to one of three HIV-DNA intradermal priming regimens by needle-free ZetaJet device at weeks 0, 4 and 12 (Group I: 2x0.1mL [3mg/mL], Group II: 2x0.1mL [3mg/mL] plus EP, Group III: 1x0.1mL [6mg/mL] plus EP). Second the same volunteers received 108 pfu HIV-MVA twice, alone or combined with CN54rgp140/GLA-AF, intramuscularly by syringe, 16 weeks apart. Additionally, 20 volunteers received saline placebo.ResultsVaccinations and electroporation did not raise safety concerns. After the last vaccination, the overall IFN-γ ELISpot response rate to either Gag or Env was 97%. Intradermal electroporation significantly increased ELISpot response rates to HIV-DNA-specific Gag (66% group I vs. 86% group II, p = 0.026), but not to the HIV-MVA vaccine-specific Gag or Env peptide pools nor the magnitude of responses. Co-administration of rgp140/GLA-AF with HIV-MVA did not impact the frequency of binding antibody responses against subtype B gp160, C gp140 or E gp120 antigens (95%, 99%, 79%, respectively), but significantly enhanced the magnitude against subtype B gp160 (2700 versus 300, p
... EP . 24 The predictive nature of these studies for human immunogenicity is unclear. ...
Article
Full-text available
Intradermal (i.d.) and intramuscular (i.m.) injections when administered with or without electroporation (EP) have the potential to tailor the immune response to DNA vaccination. This Phase I randomized controlled clinical trial in human immunodeficiency virus type 1-negative volunteers investigated whether the site and mode of DNA vaccination influences the quality of induced cellular and humoral immune responses following the DNA priming phase and subsequent protein boost with recombinant clade C CN54 gp140. A strategy of concurrent i.d. and i.m. DNA immunizations administered with or without EP was adopted. Subtle differences were observed in the shaping of vaccine-induced virus-specific CD4+ and CD8+ T cell-mediated immune responses between groups receiving: i.d.EP + i.m., i.d. + i.m.EP, and i.d.EP + i.m.EP regimens. The DNA priming phase induced 100% seroconversion in all of the groups. A single, non-adjuvanted protein boost induced a rapid and profound increase in binding antibodies in all groups, with a trend for higher responses in i.d.EP + i.m.EP. The magnitude of antigen-specific binding immunoglobulin G correlated with neutralization of closely matched clade C 93MW965 virus and Fc-dimer receptor binding (FcγRIIa and FcγRIIIa). These results offer new perspectives on the use of combined skin and muscle DNA immunization in priming humoral and cellular responses to recombinant protein.
... EP . 24 The predictive nature of these studies for human immunogenicity is unclear. ...
Article
Full-text available
Intradermal (ID) and intramuscular (IM) injections when administered with or without electroporation (EP), have the potential to tailor the immune response to DNA vaccination. In this Phase I randomized controlled clinical trial in HIV-1 negative volunteers we investigate whether the site and mode of DNA vaccination influences the quality of induced cellular and humoral immune responses following the DNA priming phase and subsequent protein boost with recombinant clade C CN54 gp140. We adopted a strategy of concurrent ID and IM DNA immunizations administered with or without EP. We observed subtle differences in shaping of vaccine induced virus-specific CD4+ and CD8+ T cell-mediated immune responses between groups receiving: IDEP+IM, ID+IMEP and IDEP+IMEP regimens. The DNA priming phase induced 100% seroconversion in all of the groups. A single, non-adjuvanted protein boost induced a rapid and profound increase in binding antibodies in all groups, with a trend for higher responses in the IDEP+IMEP group. The magnitude of antigen specific binding IgG correlated with neutralization of closely matched clade C 93MW965 virus and Fc-dimer receptor binding (FcgRIIa and FcgRIIIa). These results offer new perspectives on the use of combined skin and muscle DNA immunization in priming humoral and cellular responses to recombinant protein.
Article
Human cytomegalovirus (HCMV) infection, the most common cause of congenital disease globally, affecting an estimated 1 million newborns annually, can result in lifelong sequelae in infants, such as sensorineural hearing loss and brain damage. HCMV infection also leads to a significant disease burden in immunocompromised individuals. Hence, an effective HCMV vaccine is urgently needed to prevent infection and HCMV-associated diseases. Unfortunately, despite more than five decades of vaccine development, no successful HCMV vaccine is available. This review summarizes what we have learned from acquired natural immunity, including innate and adaptive immunity; the successes and failures of HCMV vaccine human clinical trials; the progress in related animal models; and the analysis of protective immune responses during natural infection and vaccination settings. Finally, we propose novel vaccine strategies that will harness the knowledge of protective immunity and employ new technology and vaccine concepts to inform next-generation HCMV vaccine development. Expected final online publication date for the Annual Review of Virology, Volume 9 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Article
Full-text available
Transdermal delivery can be accomplished through various mechanisms including formulation optimization, epidermal stratum corneum barrier disruption, or directly by removing the stratum corneum layer. Microneedling, electroporation, a combination of both and also the intradermal injection known as mesotherapy have proved efficacy in epidermal-barrier disruption. Here we analyzed the effects of these methods of epidermal-barrier disruption in the structure of the skin and the absorption of four compounds with different characteristics and properties (ketoprofen, biotin, caffein, and procaine). Swine skin (Pietrain x Durox) was used as a human analogue, both having similar structure and pharmacological release. They were biopsied at different intervals, up to 2 weeks after application. High-pressure liquid chromatography and brightfield microscopy were performed, conducting a biometric analysis and measuring histological structure and vascular status. The performed experiments led to different results in the function of the studied molecules: ketoprofen and biotin had the best concentrations with intradermal injections, while delivery methods for obtaining procaine and caffein maximum concentrations changed on the basis of the lapsed time. The studied techniques did not produce significant histological alterations after their application, except for an observed increase in Langerhans cells and melanocytes after applying electroporation, and an epidermal thinning after using microneedles, with variable results regarding dermal thickness. Although all the studied barrier disruptors can accomplish transdermal delivery, the best disruptor is dependent on the particular molecule.
Article
Full-text available
Among the nonviral techniques for gene transfer in vivo, the direct injection of plasmid DNA into muscle is simple, inexpensive, and safe. Applications of this method have been limited by the relatively low expression levels of the transferred gene. We investigated the applicability of in vivo electroporation for gene transfer into muscle, using plasmid DNA expressing interleukin-5 (IL-5) as the vector. The tibialis anterior muscles of mice were injected with the plasmid DNA, and then a pair of electrode needles were inserted into the DNA injection site to deliver electric pulses. Five days later, the serum IL-5 levels were assayed. Mice that did not receive electroporation had serum levels of 0.2 ng/ml. Electroporation enhanced the levels to over 20 ng/ml. Histochemical analysis of muscles injected with a lacZ expression plasmid showed that in vivo electroporation increased both the number of muscle fibers taking up plasmid DNA and the copy number of plasmids introduced into the cells. These results demonstrate that gene transfer into muscle by electroporation in vivo is more efficient than simple intramuscular DNA injection.
Article
Full-text available
Interleukin-15 (IL-15), a 114-amino acid cytokine related to IL-2, regulates immune homeostasis and the fate of many lymphocyte subsets. We reported that, in the blood of mice and humans, IL-15 is present as a heterodimer associated with soluble IL-15 receptor α (sIL-15Rα). Here, we show efficient production of this noncovalently linked but stable heterodimer in clonal human HEK293 cells and release of the processed IL-15·sIL-15Rα heterodimer in the medium. Purification of the IL-15 and sIL-15Rα polypeptides allowed identification of the proteolytic cleavage site of IL-15Rα and characterization of multiple glycosylation sites. Administration of the IL-15·sIL-15Rα heterodimer reconstituted from purified subunits resulted in sustained plasma IL-15 levels and in robust expansion of NK and T cells in mice, demonstrating pharmacokinetics and in vivo bioactivity superior to single chain IL-15. These identified properties of heterodimeric IL-15 provide a strong rationale for the evaluation of this molecule for clinical applications. Background IL-15 acts in vivo in association with IL-15Rα, as a heterodimeric cytokine. Results Authentically processed and glycosylated IL-15·sIL-15Rα heterodimer was purified from human cells and characterized by sequencing and functional studies. Conclusion IL-15 heterodimer shows favorable in vivo properties compared with monomeric IL-15. Significance Characterization and availability of the IL-15 heterodimer is important for functional studies and clinical applications.
Article
Full-text available
Viral diversity is considered a major impediment to the development of an effective HIV-1 vaccine. Despite this diversity, certain protein segments are nearly invariant across the known HIV-1 Group M sequences. We developed immunogens based on the highly conserved elements from the p24(gag) region according to two principles: the immunogen must (i) include strictly conserved elements of the virus that cannot mutate readily, and (ii) exclude both HIV regions capable of mutating without limiting virus viability, and also immunodominant epitopes located in variable regions. We engineered two HIV-1 p24(gag) DNA immunogens that express 7 highly Conserved Elements (CE) of 12-24 amino acids in length and differ by only 1 amino acid in each CE ('toggle site'), together covering >99% of the HIV-1 Group M sequences. Altering intracellular trafficking of the immunogens changed protein localization, stability, and also the nature of elicited immune responses. Immunization of C57BL/6 mice with p55(gag) DNA induced poor, CD4(+) mediated cellular responses, to only 2 of the 7 CE; in contrast, vaccination with p24CE DNA induced cross-clade reactive, robust T cell responses to 4 of the 7 CE. The responses were multifunctional and composed of both CD4(+) and CD8(+) T cells with mature cytotoxic phenotype. These findings provide a method to increase immune response to universally conserved Gag epitopes, using the p24CE immunogen. p24CE DNA vaccination induced humoral immune responses similar in magnitude to those induced by p55(gag), which recognize the virus encoded p24(gag) protein. The inclusion of DNA immunogens composed of conserved elements is a promising vaccine strategy to induce broader immunity by CD4(+) and CD8(+) T cells to additional regions of Gag compared to vaccination with p55(gag) DNA, achieving maximal cross-clade reactive cellular and humoral responses.
Article
Full-text available
SIV stocks for in vivo non-human primate models of AIDS are typically generated by transfection of 293T cells with molecularly-cloned viral genomes or by expansion in productively infected T cells. Although stocks are titered for infectivity in vitro prior to in vivo inoculation, virus production methods may differentially affect stock features that are not routinely analyzed but may impact in vivo infectivity, mucosal transmissibility, and early infection events. We performed a detailed analysis of nine SIV stocks, comprising five infection-derived SIVmac251 viral swarm stocks and paired infection- and transfection/293T-derived stocks of both SIVmac239 and SIVmac766. Representative stocks were evaluated for: 1) virus content, 2) infectious titer, 3) sequence diversity/polymorphism frequency by single genome amplification and 454 pyrosequencing, 4) virion-associated Env content, and 5) cytokine/chemokine content by 36-plex Luminex analysis. Regardless of production method, all stocks had comparable particle-to-infectivity ratios, with the transfection/293T stocks possessing the highest overall virus content and infectivity titers despite containing markedly lower levels of virion-associated Env than infection-derived viruses. Transfection/293T stocks also contained fewer and lower levels of cytokines/chemokines than infection-derived stocks, which had elevated levels of multiple analytes, with substantial variability among stocks. Sequencing of the infection-derived SIVmac251 stocks revealed variable levels of viral diversity between stocks, with evidence of stock-specific selection and expansion of unique viral lineages. These analyses suggest that there may be underappreciated features of SIV in vivo challenge stocks with the potential to impact early infection events, which may merit consideration when selecting virus stocks for in vivo studies.
Article
Full-text available
We show that an electric treatment in the form of high-frequency, low-voltage electric pulses can increase more than 100-fold the production and secretion of a recombinant protein from mouse skeletal muscle. Therapeutical erythopoietin (EPO) levels were achieved in mice with a single injection of as little as 1 μg of plasmid DNA, and the increase in hematocrit after EPO production was stable and long-lasting. Pharmacological regulation through a tetracycline-inducible promoter allowed regulation of serum EPO and hematocrit levels. Tissue damage after stimulation was transient. The method described thus provides a potentially safe and low-cost treatment for serum protein deficiencies.
Article
Full-text available
We have previously shown that macaques vaccinated with DNA vectors expressing SIVmac239 antigens developed potent immune responses able to reduce viremia upon high-dose SIVmac251 challenge. To further improve vaccine-induced immunity and protection, we combined the SIVmac239 DNA vaccine with protein immunization using inactivated SIVmac239 viral particles as protein source. Twenty-six weeks after the last vaccination, the animals were challenged intrarectally at weekly intervals with a titrated dose of the heterologous SIVsmE660. Two of DNA-protein coimmunized macaques did not become infected after 14 challenges, but all controls were infected by 11 challenges. Vaccinated macaques showed modest protection from SIVsmE660 acquisition compared with naïve controls (P = 0.050; stratified for TRIM5α genotype). Vaccinees had significantly lower peak (1.6 log, P = 0.0048) and chronic phase viremia (P = 0.044), with 73% of the vaccinees suppressing viral replication to levels below assay detection during the 40-wk follow-up. Vaccine-induced immune responses associated significantly with virus control: binding antibody titers and the presence of rectal IgG to SIVsmE660 Env correlated with delayed SIVsmE660 acquisition; SIV-specific cytotoxic T cells, prechallenge CD4(+) effector memory, and postchallenge CD8(+) transitional memory cells correlated with control of viremia. Thus, SIVmac239 DNA and protein-based vaccine protocols were able to achieve high, persistent, broad, and effective cellular and humoral immune responses able to delay heterologous SIVsmE660 infection and to provide long-term control of viremia. These studies support a role of DNA and protein-based vaccines for development of an efficacious HIV/AIDS vaccine.
Article
We recently developed a novel strategy to identify transmitted HIV-1 genomes in acutely infected humans using single-genome amplification and a model of random virus evolution. Here, we used this approach to determine the molecular features of simian immunodeficiency virus (SIV) transmission in 18 experimentally infected Indian rhesus macaques. Animals were inoculated intrarectally (i.r.) or intravenously (i.v.) with stocks of SIVmac251 or SIVsmE660 that exhibited sequence diversity typical of early-chronic HIV-1 infection. 987 full-length SIV env sequences (median of 48 per animal) were determined from plasma virion RNA 1--5 wk after infection. i.r. inoculation was followed by productive infection by one or a few viruses (median 1; range 1--5) that diversified randomly with near starlike phylogeny and a Poisson distribution of mutations. Consensus viral sequences from ramp-up and peak viremia were identical to viruses found in the inocula or differed from them by only one or a few nucleotides, providing direct evidence that early plasma viral sequences coalesce to transmitted/founder viruses. i.v. infection was >2,000-fold more efficient than i.r. infection, and viruses transmitted by either route represented the full genetic spectra of the inocula. These findings identify key similarities in mucosal transmission and early diversification between SIV and HIV-1, and thus validate the SIV-macaque mucosal infection model for HIV-1 vaccine and microbicide research.
Article
We have developed an enzyme-linked immunosorbent assay (ELISA) specific for antibodies to the envelope glycoproteins gp120 and gp160 of HIV-1 An antibody to a conserved epitope on gp120 is adsorbed to a solid phase and used to capture gp120 and/or gp160 from solution. This may be purified recombinant protein or in simple, non-denaturing detergent extracts of different strains of HIV-1. Human serum antibodies bound to the captured antigen are subsequently detected with an anti-human antibody coniugated to alkaline phosphatase, and the AMPAK ELISA amplification system (Novo BioLabs, Cambridge, UK). With this procedure, antibodies can be detected that recognize gp120 from a wide range of divergent HIV-1 strains. The ELISA is sufficiently sensitive to detect env antibodies in sera from HIV-positive individuals at dilutions of 1.3000 00. No repeatable false-positives were detected in a screen of 250 normal serum samples. Env antibodies were detected in all 37 strongly HIV-positive sera tested, and in four sera that were borderline or weakly positive in commercial ELISA. However, 55 sera positive in commercial ELISA but unconfirmable by Western blot ('ambiguously' positive) did not contain detectable env antibodies.
Article
HIV preferentially infects activated T cells, and activated mucosal CD4⁺ T cells are the primary sites of viral replication. One potential explanation for increased HIV acquisition rates in the STEP study is that vaccination with adenoviral (Ad) vectors increased CD4⁺ T cell activation levels at the site of infection, a concept that others and we continue to explore.¹,² Whether vaccination with HIV vaccine platforms increases the activation state of CD4⁺ T cells within peripheral tissues, such as the gastro-intestinal (GI) mucosa, is exceptionally important to determine as a vaccine safety measure, given the susceptibility of activated CD4⁺ T cells to HIV infection. In this study we examined whether vaccination with DNA plasmids and chemokine adjuvants alter the activation state of T cells within the GI mucosa, inguinal LN, and peripheral blood. T cell activation state was measured by expression of CD25, CD69, and HLA-DR over the course of the prime/boost study. DNA plasmid vaccination did not increase expression of any of these markers in the 3 tissues studied. Addition of the gut-homing chemokine TECK during DNA plasmid vaccination did not alter activation levels of CD4⁺ T cells at any of these sites. These findings indicate that DNA vaccines do not elicit generalized mucosal T cell activation. Thus, DNA platforms may be especially suitable for HIV vaccine development, where bystander activation could promote increased HIV transmission.
Article
Lin and colleagues assess the electric field strengths required to achieve efficient and balanced DNA vaccine delivery using a surface intradermal electroporation (SEP) device. They show that delivery of DNA vaccines encoding influenza virus H5 hemagglutinin (H5HA) and nucleoprotein (NP) of influenza H1N1 in mice at applied voltages of 10 to 100 V elicits robust and sustained antibody responses. Low voltage electroporation (less than 20 V), by contrast, elicits higher and more sustained cellular immune response.