SlideShare a Scribd company logo
1 of 12
Download to read offline
Conformational Epitope-Specific Broadly Neutralizing Plasma
Antibodies Obtained from an HIV-1 Clade C-Infected Elite
Neutralizer Mediate Autologous Virus Escape through Mutations in
the V1 Loop
Shilpa Patil,a
Rajesh Kumar,a
Suprit Deshpande,a
Sweety Samal,a
Tripti Shrivastava,a
Saikat Boliar,a
Manish Bansal,a
Nakul Kumar Chaudhary,a
Aylur K. Srikrishnan,b
Kailapuri G. Murugavel,b
Suniti Solomon,b
Melissa Simek,c
Wayne C. Koff,c
Rajat Goyal,c
Bimal K. Chakrabarti,a,c
Jayanta Bhattacharyaa,c
HIV Vaccine Translational Research Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, Indiaa
; Y. R. Gaitonde Research and Care Center,
Chennai, Indiab
; International AIDS Vaccine Initiative, New York, New York, USAc
ABSTRACT
Broadly neutralizing antibodies isolated from infected patients who are elite neutralizers have identified targets on HIV-1 enve-
lope (Env) glycoprotein that are vulnerable to antibody neutralization; however, it is not known whether infection established by
the majority of the circulating clade C strains in Indian patients elicit neutralizing antibody responses against any of the known
targets. In the present study, we examined the specificity of a broad and potent cross-neutralizing plasma obtained from an In-
dian elite neutralizer infected with HIV-1 clade C. This plasma neutralized 53/57 (93%) HIV pseudoviruses prepared with Env
from distinct HIV clades of different geographical origins. Mapping studies using gp120 core protein, single-residue knockout
mutants, and chimeric viruses revealed that G37080 broadly cross-neutralizing (BCN) plasma lacks specificities to the CD4 bind-
ing site, gp41 membrane-proximal external region, N160 and N332 glycans, and R166 and K169 in the V1-V3 region and are
known predominant targets for BCN antibodies. Depletion of G37080 plasma with soluble trimeric BG505-SOSIP.664 Env (but
with neither monomeric gp120 nor clade C membrane-proximal external region peptides) resulted in significant reduction of
virus neutralization, suggesting that G37080 BCN antibodies mainly target epitopes on cleaved trimeric Env. Further examina-
tion of autologous circulating Envs revealed the association of mutation of residues in the V1 loop that contributed to neutral-
ization resistance. In summary, we report the identification of plasma antibodies from a clade C-infected elite neutralizer that
mediate neutralization breadth via epitopes on trimeric gp120 not yet reported and confer autologous neutralization escape via
mutation of residues in the V1 loop.
IMPORTANCE
A preventive vaccine to protect against HIV-1 is urgently needed. HIV-1 envelope glycoproteins are targets of neutralizing anti-
bodies and represent a key component for immunogen design. The mapping of epitopes on viral envelopes vulnerable to im-
mune evasion will aid in defining targets of vaccine immunogens. We identified novel conformational epitopes on the viral enve-
lope targeted by broadly cross-neutralizing antibodies elicited in natural infection in an elite neutralizer infected with HIV-1
clade C. Our data extend our knowledge on neutralizing epitopes associated with virus escape and potentially contribute to im-
munogen design and antibody-based prophylactic therapy.
Broadly neutralizing antibodies (BNAbs) target trimeric enve-
lope glycoprotein (Env) spikes of human immunodeficiency
virus type 1 (HIV-1). Characterization of the BNAbs has provided
key clues toward the design and development of both prophylactic
and therapeutic vaccines (1–6). A small proportion of individuals
chronically infected with HIV-1 develop BNAbs (7–14), and the
isolation of several broad and potent neutralizing monoclonal an-
tibodies (MAbs) from such individuals with distinct molecular
specificities to viral envelope protein has been reported (15–23).
The cross-neutralizing serum antibodies obtained from such in-
dividuals (also referred to as elite neutralizers), which have con-
siderable breadth, target epitopes on structurally conserved re-
gions of Env such as the CD4 binding site (CD4bs) (22, 24–26),
V1V2, including glycan moieties (19, 20, 27, 28), the gp120-gp41
interface (18, 29), and the membrane-proximal external regions
(MPER) (16, 30–32). Several studies have indicated that the vari-
able regions within HIV-1 gp120 contain epitopes targeted by
autologous antibodies as well as BNAbs (33–40). Recently the
V1V2 region has been linked to the development of broadly cross-
neutralizing (BCN) antibodies (35, 41), and the residues between
160 and 172 (notably R166S/K or K169A) in V1V2 have been
demonstrated to be associated with virus escape from autologous
Received 9 December 2015 Accepted 7 January 2016
Accepted manuscript posted online 13 January 2016
Citation Patil S, Kumar R, Deshpande S, Samal S, Shrivastava T, Boliar S, Bansal M,
Chaudhary NK, Srikrishnan AK, Murugavel KG, Solomon S, Simek M, Koff WC, Goyal
R, Chakrabarti BK, Bhattacharya J. 2016. Conformational epitope-specific broadly
neutralizing plasma antibodies obtained from an HIV-1 clade C-infected elite
neutralizer mediate autologous virus escape through mutations in the V1 loop.
J Virol 90:3446–3457. doi:10.1128/JVI.03090-15.
Editor: W. I. Sundquist, University of Utah
Address correspondence to Jayanta Bhattacharya, JBhattacharya@iavi.org.
S.P., R.K., and S.D. contributed equally to this work.
Copyright © 2016, American Society for Microbiology. All Rights Reserved.
crossmark
3446 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
antibody response (35). Recent studies have further indicated that
BCNAb development in vivo is associated with antibody affinity
maturation and coevolution of virus, resulting in a considerable
degree of somatic hypermutations (19, 20, 23, 26, 35, 42–50). Such
information is crucial for the design and development of suitable
Env-based immunogens capable of eliciting broad and potent
cross-neutralizing antibodies through vaccination.
While a number of studies on the molecular specificities of
broadly neutralizing antibodies obtained from African clade C-in-
fected individuals have been reported (9, 37, 51–62), knowledge
on immune evasion in Indian clade C-infected elite neutralizers is
very limited (63).
In the present study, we examined plasma samples obtained
from two hundred asymptomatic and antiretroviral therapy
(ART) naive Indian HIV-infected donors and identified plasma
with cross-neutralizing antibodies. The molecular specificities of
plasma antibodies obtained from an HIV-1 clade C-infected elite
neutralizer was characterized in detail that displayed exceptional
neutralization breadth across clades of different geographical or-
igins. Interestingly, we found that neutralization breadth was as-
sociated with the presence of unique epitopes on the trimeric
gp120.
MATERIALS AND METHODS
Ethics statement. The blood samples were collected under the IAVI Pro-
tocol G study from slow-progressing ART naive HIV-1-positive donors
from Nellore District of the state of Andhra Pradesh, southern India, by
trained clinicians at the YRG Care Hospital following approval and clear-
ance from the Institutional Review Board (IRB) and the Ethics Commit-
tee. The serum and plasma samples collected were shipped to the HIV
Vaccine Translational Research Laboratory, Translational Health Science
and Technology Institute, for further assessment and research on the neu-
tralizing antibody response.
Plasmids, viruses, antibodies, proteins, and cells. Plasmids encoding
HIV-1 envelopes representing distinct clades are shown in Table 1. Mono-
clonal antibodies used in the study and TZM-bl cells were procured from
the NIH AIDS Research and Reagents Reference program and from the
IAVI Neutralizing Antibody Consortium (NAC). 293T cells were pur-
chased from the American Type Culture Collection (ATCC). Plasmid
DNA encoding BG505-SOSIP.664-D7324, its purified cleaved trimeric
protein (64), and pcDNA5-FRT BG505 furin A (65) were kindly provided
by John Moore, Weill Cornell Medical College, New York. Purified gp120
TripleMut core protein (66) was obtained from Richard Wyatt, The
Scripps Research Institute, through the NAC. HIV-2 7312A and its chi-
meric constructs were provided by Lynn Morris, NICD, Johannesburg,
South Africa.
Purification of monomeric and trimeric Env proteins. Codon-opti-
mized gp120 plasmid encoding clade C 4-2.J41 (67, 68) gp120 was cloned
in pcDNA 3.1/V5-His-TOPO vector and transfected into 293T cells using
polyethyleneimine (PEI). Supernatants containing soluble gp120 were fil-
tered through 0.45-␮m-pore-size filters and subsequently purified using
nickel-nitrilotriacetic acid (Ni-NTA) agarose matrix (Qiagen Inc.) by elution
with phosphate-buffered saline (PBS) containing 300 mM imidazole (pH
8.0). The purified monomeric gp120 protein was extensively dialyzed with
PBS (pH 7.4), concentrated using Amicon ultracentrifugal filters (Millipore
Inc.) with a 30-kDa cutoff, and stored at Ϫ80°C until further use.
The trimeric BG505-SOSIP.664 protein was purified using 293F
cells essentially as described by Sanders et al. (69). Briefly, the 293F
cells were transfected with plasmid DNA encoding both BG505-SO-
SIP.664 gp140 envelope and furin (65). Supernatant containing solu-
ble BG505-SOSIP.664 gp140 was harvested 72 to 96 h posttransfection,
filtered, and passed through a lectin agarose column obtained from
Galanthus nivalis (Sigma Inc.). The nonspecifically bound proteins then
were washed in PBS (pH 7.4) supplemented with 0.5 M NaCl. The bound
proteins then were eluted using 0.5 M methyl ␣-D-mannopyranoside,
extensively dialyzed with 1ϫ PBS, and concentrated. BG505-SOSIP.664
was further purified by Sephadex G-200 size exclusion chromatography
(AKTA; GE). Trimeric protein fractions were collected and pooled, their
quality was assessed by running in blue native polyacrylamide gel electro-
TABLE 1 Neutralization breadth of Protocol G G37080 plasma samples
collected at two different time points tested against panel of 57 Env-
pseudotyped viruses
HIV-1 Clade C Plasma Confers Cross-Clade Neutralization
April 2016 Volume 90 Number 7 jvi.asm.org 3447Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
phoresis (BN-PAGE), and they were favorably assessed for their ability to
bind to only neutralizing and not to nonneutralizing and MPER-directed
monoclonal antibodies as described elsewhere (64) by enzyme-linked im-
munosorbent assay (ELISA).
Depletion of plasma antibodies by monomeric gp120 and trimeric
gp140 Env proteins. Purified soluble monomeric 4-2.J41 gp120 and tri-
meric BG505 SOSIP.664 proteins, in addition to the MPER peptide (C1C;
encoding clade C sequence) (70), were used for the depletion of plasma
antibodies where purified proteins were covalently coupled to tosylacti-
vated MyOne Dynabeads (Life Technologies Inc.) according to the man-
ufacturer’s protocol. Briefly, 30 mg of beads was coupled with 1 mg of
both monomeric and trimeric Env proteins in coupling buffer [0.1 M
NaBO4, 1 M (NH4)2SO4; pH 9.4] overnight at 37°C for 16 to 24 h. Proteins
bound to magnetic beads were separated from unbound proteins using a
DynaMag 15 magnet (Life Technologies, Inc.). Beads bound to Env pro-
teins next were incubated with blocking buffer (PBS [pH 7.4], 0.1% bo-
vine serum albumin [BSA; Sigma], and 0.05% Tween 20) at 37°C to block
the unbound sites. The antigenic integrity of both 4-2.J41 monomeric
gp120 and BG505-SOSIP.664 bound to the beads was assessed for their
ability to bind VRC01 and 4E10 MAbs (for monomeric gp120) and
PGT121, F105, and 4E10 MAbs (for BG505-SOSIP.664) by flow cytom-
etry (FACSCanto; Becton and Dickinson, Inc.).
For depletion studies, G37080 plasma was diluted to 1:50 in Dulbec-
co’s modified Eagle’s medium (DMEM) containing 10% fetal bovine se-
rum (FBS), and 500 ␮l of diluted plasma was incubated with 20 ␮l of beads
at room temperature for 45 min. Unbound plasma antibodies were sepa-
rated from those that were bound to protein-coated beads using a
DynaMag 15 magnet as described above. This step was repeated 4 to 5
times for the depletion of plasma antibodies by monomeric gp120 and 10
to 12 times in the case of BG505-SOSIP.664-coated beads. As a negative
control, G37080 plasma antibodies were depleted with uncoated beads in
parallel. In addition to ELISA, the percent depletion of G37080 plasma
antibodies was assessed by examining the sequential decrease in binding
of protein-coated beads with depleted plasma antibodies by fluorescence-
activated cell sorting (FACS). PGT121 MAb was taken as a positive con-
trol for checking depletion by BG505-SOSIP.664 trimeric Env.
gp120 and gp140 ELISA. For gp120 ELISA, a high-binding polysty-
rene microtiter plate (Nunc, Inc.) was coated with 100 ␮l of monomeric
4-2.J41 gp120 (1 ␮g/ml) in binding buffer containing 0.1 M NaHCO3 (pH
8.6) and incubated overnight at 4°C. The gp120-plate was washed once
with 1ϫ PBS (pH 7.4) and blocked with 5% nonfat milk for 90 min at
37°C. The plate then was washed three times with 1ϫ PBS followed by the
addition of 100 ␮l of MAbs, as well as the depleted and undepleted plasma
antibodies at different dilutions, and incubated for 1 h at room tempera-
ture. The wells of the ELISA plate were washed four times with PBS con-
taining 0.1% Tween 20 (PBST), followed by the addition of 100 ␮l of
1:3,000-diluted horseradish peroxidase (HRP)-conjugated anti-human
IgG (Jackson ImmunoResearch, Inc.), and further incubated for 45 min at
room temperature. Unbound conjugates were removed by washing with
PBST, and color was developed by the addition of 100 ␮l of 3,3=,5,5=-
tetramethylbenzidine (TMB) (Life Technologies, Inc.) substrate. Absor-
bance was measured at 450 nm in a spectrophotometer.
Binding of antibodies to BG505-SOSIP.664-D7324 trimeric protein
was assessed essentially as described by Sanders et al. (69) in a sandwich
ELISA. Briefly, a high-binding microtiter plate (Nunc, Inc.) first was
coated with D7324 antibody at 10 ␮g/ml (Aalto Bio Reagents, Dublin,
Ireland) followed by blocking extra unbound sites with 5% nonfat milk
for 90 min at 37°C. One hundred microliters of BG505.664-D7324 trim-
eric protein (300 ng/ml) then was added and incubated for 45 min at room
temperature. The extent of binding of G37080 plasma antibodies com-
pared to that of known neutralizing monoclonal antibodies was assessed
by the addition of primary and HRP-conjugated secondary anti-human
antibody as described above.
Neutralization assay. Neutralization assays were carried out using
TZM-bl cells as described before (68). Briefly, Env-pseudotyped viruses
were incubated with various dilutions of depleted plasma antibodies and
incubated for an hour at 37°C in a CO2 incubator under humidified con-
ditions, and subsequently 1 ϫ 104
TZM-bl cells were added to the mixture
in the presence of 25 ␮g/ml DEAE-dextran (Sigma, Inc.). The plates were
further incubated for 48 h, and the degree of virus neutralization was
assessed by measuring relative luminescence units (RLU) in a luminom-
eter (Victor X2; PerkinElmer Inc.).
Amplification, cloning, and mutagenesis of autologous HIV-1 env
genes. Autologous complete env genes were obtained from G37080
plasma as described previously, with slight modifications (68). Briefly,
viral RNA was extracted using a high-pure viral RNA kit (Roche Inc.) by
following manufacturer’s protocol, and cDNA was prepared by reverse
transcription-PCR (RT-PCR) using a Superscript III first-strand synthesis
kit (Invitrogen Inc.). rev-gp160 env genes were amplified using a Phusion
high-fidelity DNA polymerase (New England BioLabs Inc.). The gp160
amplicons were purified and ligated into pcDNA 3.1/V5-His-TOPO (In-
vitrogen Inc.) vector. Chimeric Env proteins were prepared by overlap-
ping PCR, and point substitutions were made with a QuikChange II kit
(Agilent Technologies Inc.) by following the manufacturer’s protocol and
as described previously (71).
Preparation of envelope-pseudotyped viruses. Pseudotyped viruses
were prepared by the cotransfection of envelope-expressing plasmid with
an env-deleted HIV-1 backbone plasmid (pSG3⌬Env) into 293T cells in
6-well tissue culture plates using a FuGENE6 transfection kit (Promega
Inc.). Cell supernatants containing pseudotyped viruses were harvested 48
h posttransfection and then stored at Ϫ80°C until further use. The infec-
tivity assays were done in TZM-bl cells (1 ϫ 105
cells/ml) containing
DEAE-dextran (25 ␮g/ml) in 96-well microtiter plates, and the infectivity
titers were determined by measuring the luciferase activity using Britelite
luciferase substrate (PerkinElmer Inc.) with a Victor X2 luminometer
(PerkinElmer Inc.).
RESULTS
Identification of an elite neutralizer with HIV-1 clade C infec-
tion whose plasma showed exceptional neutralization breadth.
The present study, under IAVI Protocol G, was designed (i) to
screen and identify plasma antibodies obtained from Indian do-
nors chronically infected with HIV-1 clade C, with substantial
breadthtowardneutralizingcross-cladeHIV-1primaryvariants,and
(ii) to elucidate the molecular specificities associated with neutraliza-
tionbreadth.Ourhypothesiswasthatgiventhegeneticdistinctnessof
clade C viruses of Indian and non-Indian origin, as well the likely
differences in host genetics between populations and their ancestral
origins associated with the modulation of humoral immune re-
sponses, the specificities of antibodies developed in vivo associated
with neutralization breadth and potency would be different.
Through screening of two hundred plasma samples obtained
from chronically infected ART naive Indian patients against a
panel of 57 pseudoviruses containing Envs of distinct clades and
geographical origins (Fig. 1A), we identified one donor (G37080)
whose plasma showed exceptional neutralization breadth. Donor
G37080 serum neutralized Ͼ90% of the 57 different pseudovi-
ruses tested, with a median 50% inhibitory dose (ID50) value of
533.03 (Table 1 and Fig. 1B).
Follow-up plasma sample from this donor (G37080) subse-
quently was obtained after 8 months to assess whether the neu-
tralization breadth and potencies, along with their molecular
specificities, were retained and/or improved, as we expected that
during the course of disease, the breadth and potency of neutral-
izing antibodies broadens through somatic hypermutations (72)
and/or clonal selection processes. As shown in Fig. 1B and Table 1,
follow-up plasma antibodies of donor G37080 (referred to as the
visit 2 samples) were found to exhibit neutralization breadth com-
Patil et al.
3448 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
parable to that of visit 1 plasma. Overall, G37080 BCN plasma was
found to potently neutralize pseudoviruses containing Indian
clade C Env with a neutralization score of 2.5 (13). Furthermore,
the neutralization sensitivity of Env-pseudotyped viruses was
found to be correlated with the serum IgG (data not shown), sug-
gesting that the broad neutralization was associated with IgG-
specific responses. Taken together, our data indicate that a strong
humoral immune response to HIV-1 was mounted in donor
G37080 and was maintained over time.
Evidence that G37080 BCN plasma antibodies do not target
epitopes in CD4bs, MPER, and known glycan and nonglycan
residues in variable domains of Env. We first examined whether
the G37080 BCN plasma contains antibodies directed to the
CD4bs on Env. Plasma samples obtained from both visits were
pretreated with 25 ␮g/ml of TripleMut core protein (66), which
was a concentration that we found to inhibit the neutralization of
25711-2.4 pseudovirus by VRC01 MAb by Ͼ95%. Pretreated
plasma subsequently was used to neutralize pseudovirus 25711-
2.4 Env, and as shown in Fig. 2, no perturbation of G37080 neu-
tralizing activity was observed against pseudovirus 25711-2.4. A
similar observation was made when these plasma antibodies were
pretreated with RSC3 core protein (22). In addition, the G37080
BCN plasma antibodies were found to efficiently neutralize
IgG1b12- and VRC01-resistant viruses (data not shown). Our
data indicated that the G37080 BCN plasma antibodies do not
contain CD4bs-directed neutralizing antibodies.
To elucidate whether the BCN plasma antibodies are directed
to MPER in gp41, we used HIV-2/HIV-1 chimeric viruses (73)
that expressed minimal residues of HIV-1 MPER containing
epitopes required for MPER-directed MAbs, such as 2F5, 4E10,
Z13e, and 10E8. As shown in Table 2, the G37080 BCN plasma
from both visits was found to show modest neutralization of
HIV-2 expressing HIV-1 clade C MPER (7312-C1C), with ID50
values of 306.42 and 371.02, respectively. We also found that the
depletion of G37080 plasma with a clade C MPER peptide (C1C)
completely abolished the sensitivity of 7312A-C1C virus to
FIG1 (A) Genetic divergence of amino acid sequences of 57 HIV-1 Env (gp160) pseudoviruses used to assess neutralization breadth and potency of G37080 BCN
plasma. The maximum likelihood bootstrapped consensus phylogenetic tree was constructed using the Jones-Taylor-Thornton (JTT) substitution model with
50 bootstrapped replicates in Mega 5.2. Bootstrapped values are shown at the nodes of each branch. Hollow circles represent envelopes (16055-2.3 and 92TH021)
resistant to neutralization by G37080 BCN plasma. (B) Neutralization breadth of the G37080 BCN plasma obtained at visit 1 and visit 2 were assessed against
pseudotyped viruses expressing HIV-1 Env representing different clades and origins. Neutralization titers (median ID50 values) were obtained by titrating
Env-pseudotyped viruses against G37080 plasma samples. Values at the top of each bar graph indicate the number of viruses belonging to each clade/origin tested.
FIG 2 Assessing dependence of G37080 BCN antibodies to CD4 binding site (CD4bs) region of HIV-1 Env. G37080 BCN plasma samples and VRC01 MAb
(concentrations that neutralized 25711-2.4 by Ͼ80%) preincubated with different concentrations, as indicated, with TripleMut core (A) and RSC3 (B) proteins
were examined for their ability to neutralize 25711-2.4 Env pseudotyped virus in a TZM-bl cell neutralization assay. Note that while VRC01 preabsorbed with
both TripleMut and RSC3 proteins showed inhibited neutralization of 25711-2.4 in a dose-dependent manner, no such effect was observed with G37080 BCN
plasma, indicating the absence of CD4bs-directed neutralizing antibodies.
HIV-1 Clade C Plasma Confers Cross-Clade Neutralization
April 2016 Volume 90 Number 7 jvi.asm.org 3449Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
G37080 plasma (Table 3). Our data suggest that although the
G37080 BCN plasma neutralized 7312-C1C, the presence of
MPER-directed antibodies was not associated with neutralization
breadth.
We next investigated whether the plasma antibodies of the do-
nor G37080 target residues in variable loops, particularly in V1V2
and V3 regions, which have been shown in several studies to be
epitopes targeted by BCN antibodies on HIV-1 Env. We first
tested the extent of neutralization by G37080 BCN plasma anti-
bodies of Env-pseudotyped viruses lacking glycans at positions
160 (N160) and 332 (N332) in the V2 region and V3 base, respec-
tively, and also R166 and K169 in the V2 region, which are major
targets of recently identified broad and potent neutralizing mono-
clonal antibodies. In order to test this, two clade C Envs (25711-
2.4 and CAP239.G3), containing N160A and N332A substitutions,
were tested (Table 2). Our data indicate that the pseudoviruses con-
taining Env expressing the N160 or N332 substitution have sensi-
tivities identical to those of G37080 plasma antibodies. Similar
observations were found with R166A and K169A in the 93IN905
Env backbone. Taken together, our observations indicate that
G37080 BCN plasma antibodies did not utilize these residues in
V2 and V3 regions for neutralization breadth; these have been
identified as important epitopes recognized by broadly neutraliz-
ing antibodies elicited in clade C infection as described before (35,
41, 56).
AssociationofneutralizationbreadthofG37080plasmawith
recognition of conformational epitopes on cleaved trimeric Env
but not with that in monomeric gp120 or MPER. In order to
examine whether broad neutralization conferred by the G37080
plasma antibodies was through the recognition of epitopes on
monomeric gp120 or cleaved near-native Env trimers, we tested
the binding of G37080 serum IgG to monomeric 4-2.J41 gp120
and soluble gp140 (BG505-SOSIP.664) by ELISA. We found that
in addition to the monomeric 4-2.J41 gp120 (Fig. 3A), G37080
serum polyclonal IgG was found to efficiently bind to the BG505
SOSIP.664-D7324 soluble trimeric Env (Fig. 3B), indicating that
the G37080 plasma primarily contains neutralizing antibodies
that target epitopes on cleaved Env trimers.
We next examined whether binding of the G37080 plasma an-
tibodies to epitopes on cleaved BG505-SOSIP.664 trimeric enve-
lope was associated with neutralization breadth. For this, we tested
the ability of G37080 plasma antibodies depleted of both mono-
meric and trimeric Envs, as well as of MPER peptides, to neutralize
a set of Env-pseudotyped viruses, which were found to be sensitive
to this particular plasma sample. Purified 4-2.J41 monomeric
gp120, BG505-SOSIP.664 trimeric gp140, and C1C MPER pep-
tide bound to the magnetic beads were used to deplete G37080
plasma antibodies as described in Materials and Methods. The
depleted BCN G37080 antibodies first were assessed for their
binding to 4-2.J41 gp120 monomers, BG505-SOSIP.664-D7324,
and C1C peptide and compared to undepleted plasma antibodies
by ELISA. As shown in Fig. 3C and D, G37080 plasma depleted
with monomeric gp120 and trimeric gp140, respectively, had sig-
nificantly reduced binding activity against the respective soluble
proteins. Similar observation was made with MPER peptide (data
not shown). The depleted plasma antibodies subsequently were
assessed for neutralization activity using a panel of 12 Env-pseu-
dotyped viruses that were susceptible to untreated G37080 plasma
antibodies as mentioned above. As shown in Table 3, depletion
with 4-2.J41gp120 monomer and C1C peptide did not show any
change in neutralization breadth of G37080 plasma antibodies,
while depletion with BG505-SOSIP.664 showed a significant re-
duction in virus neutralization. Similar observations were made
with the BG505-SOSIP.664-depleted PGT121 and C1C peptide-
depleted 4E10 MAbs, which lost the ability to efficiently neutralize
Env-pseudotyped viruses (16055 and ZM233.6) and HIV-2/
HIV-1 (7312A-C1C) chimeric virus compared to their undepleted
counterparts (data not shown), validating our data. Interestingly,
C1C peptide-depleted G37080 plasma failed to neutralize HIV-2/
TABLE 3 Degree of shift in sensitivity of Env-pseudotyped viruses to
G37080 BCN plasma depleted with monomeric and trimeric Envs and
C1C peptide
Env-pseudotyped
virus
Fold reduction in neutralization (ID50)a
gp140 trimer
(BG505-SOSIP.664)
gp120 monomer
(4-2.J41)
MPER (C1C
peptide)
25710-2.3 Ͼ10.30 1.3 0.83
25711-2.4 Ͼ8.52 1.4 1.44
3-5.J25 Ͼ7.85 0.9 0.84
4-2.J41 12.11 1.1 1.04
IAVI_C22 Ͼ15.92 1.2 1.18
92BR020 Ͼ35.08 1.1 1.34
93IN905 3.41 1.2 0.94
JRCSF Ͼ8.75 0.5 0.93
Q23.17 Ͼ23.28 1.0 0.98
Du156.12 Ͼ15.73 0.8 1.61
HVTR-PG80v1.eJ7 Ͼ10.03 0.9 1.12
HVTR-PG80v1.eJ19 Ͼ15.60 0.5 1.18
HIV-2 7312A-C1C Ͼ10
a
Fold reduction in virus neutralization was obtained by comparing the neutralization
titer (ID50 values) of panel viruses against undepleted and depleted G37080 plasma.
ID50 values are reciprocal dilutions at which the undepleted and depleted plasma
conferred 50% neutralization of the Env-pseudotyped viruses in TZM-bl cells.
TABLE 2 Examination of specificity of G37080 plasma antibodies
obtained at both visits to HIV Env
HIV type Region
Fold decrease in
ID50
a
for plasma
at visit:
1 2
HIV-1 Env mutants
HIV-1 25711-2.4 N160A V2 1.02 Ͻ1
HIV-1 25711-2.4 R166A Ͻ1 Ͻ1
HIV-1 25711-2.4 K169E Ͻ1 Ͻ1
HIV-1 93IN905 R166A Ͻ1 Ͻ1
HIV-1 93IN905 K169A Ͻ1 Ͻ1
HIV-1 25711-2.4 N332A V3 1.52 Ͻ1
HIV-1 CAP239.G3 N332A 1.35 1.32
HIV-2/HIV-1 chimera Region of HIV-1 ID50 ID50
HIV-2 7312A HIV-2 wild type Ͻ20 Ͻ20
HIV-2 7312A-C1C Clade C MPER 306.42 371.02
HIV-2 7312A-C3 2F5 epitope Ͻ20 Ͻ20
HIV-2 7312A-C4 4E10, Z13e1, and
10E8 epitopes
334.34 371.27
HIV-2 7312A-C6 4E10 minimal epitope Ͻ20 223.90
HIV-2 7312A-C7 2F5 minimal epitope Ͻ20 Ͻ20
a
ID50 values refer to the reciprocal dilution that conferred 50% neutralization of
viruses in a TZM-bl assay. Assays were done in duplicate and were repeated more
than three times.
Patil et al.
3450 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
HIV-1 (7312A-C1C) chimeric virus, indicating that the presence
of residual traces of MPER-directed antibodies (as shown in
Table 2) is not responsible for neutralization breadth. Further-
more, the examination of sensitive (25711-2.4) and resistant
(16055-2.3 and CAP45.G3) chimeric Envs indicated that the BCN
G37080 plasma antibodies predominantly target epitopes in the
V1V2 region (Table 4) in gp120. Our data clearly indicate a cor-
relation between neutralization breadth and binding of the
G37080 BCN plasma antibodies to the conformational epitopes
on cleaved trimeric gp120, likely in the V1V2 region; however, we
do not rule out the possibility that this BCN plasma targets other
discontinuous epitopes in gp120 but not in MPER.
Mutations in V1 region confer resistance to autologous vi-
ruses to the G37080 plasma antibodies. In order to decipher the
specificity of the G37080 plasma antibodies, we examined the de-
gree of susceptibility of pseudoviruses prepared using env genes
amplified from contemporaneous autologous G37080 plasma ob-
tained at the baseline and follow-up visits. As shown in Fig. 4A,
both of the Env proteins obtained from visit 2 plasma (HVTR-
PG80v2.eJ38 and HVTR-PG80v2.eJ41) were found to be resistant
to its contemporaneous plasma antibodies, while Env proteins
obtained from visit 1 plasma (HVTR-PG80v1.eJ7 and HVTR-
PG80v1.eJ19) were found to be modestly sensitive to visit 2 autol-
ogous G37080 plasma antibodies. To facilitate mapping G37080
BCN antibody specificity, we prepared chimeric Envs between
a sensitive (HVTR-PG80v1.eJ7 and HVTR-PG80v1.eJ19) and a
resistant (HVTR-PG80v2.eJ38) autologous Env by first swap-
ping the V1V2 regions, as their amino acid sequences differed
maximally in this region (Fig. 4B). As shown in Table 4, the
insertion of the V1V2 sequences of HVTR-PG80v1.eJ7 and
HVTR-PG80v1.eJ19 into HVTR-PG80v2.eJ38 conferred Env-
pseudotyped viruses expressing HVTR-PG80v2.eJ38 Env with
sensitivity to G37080 visit 2 plasma antibodies enhanced by
Ͼ25- and Ͼ12-fold, respectively. Conversely, the neutraliza-
tion susceptibilities of the Env-pseudotyped viruses expressing
HVTR-PG80v1.eJ7 and HVTR-PG80v1.eJ19, which contained
HVTR-PG80v2.eJ38 V1V2 sequence corresponding to visit 2
G37080 plasma, were found to be reduced by Ͼ45- and Ͼ23-
fold, respectively. We noted that alterations of regions other
than the V1V2 loop in the autologous Env did not confer any
change in neutralization sensitivity (Table 4). To further spec-
ify residues in the V1V2 loop associated with neutralization
sensitivity and resistance of autologous Envs, chimeric Envs
and point mutants were prepared and tested for their degree of
modulation in susceptibility to autologous G37080 plasma ob-
tained from the second visit. As shown in Table 4, we found that
the V1 sequence, but not the V2 sequence, of the sensitive Envs
(HVTR-PG80v1.eJ7 and HVTR-PG80v1.eJ19) increased sensitiv-
ity to G37080 BCN plasma antibodies by Ͼ50 and Ͼ37-fold, re-
spectively, when transferred to the resistant HVTR-PG80v2.eJ38
FIG 3 Binding of G37080 BCN plasma IgG to 4-2.J41 monomeric gp120 (A) and BG505-SOSIP.664-D7324 cleaved trimeric gp140 (B) soluble proteins was
assessed by ELISA. IgG purified from HIV-negative healthy donor and known MAbs were used as controls. The extent of binding of the depleted and undepleted
G37080 BCN plasma with magnetic beads coated with 4-2.J41 monomeric gp120 (C) and BG505-SOSIP.664 cleaved trimeric gp140 to their respective proteins
by ELISA. Note that binding to trimeric protein by ELISA was assessed by using BG505-SOSIP.664 tagged with the D7324 epitope to maintain the native
conformation of trimeric Env as described before (69).
HIV-1 Clade C Plasma Confers Cross-Clade Neutralization
April 2016 Volume 90 Number 7 jvi.asm.org 3451Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
Env. In agreement with this result, V1 of HVTR-PG80v2.eJ38,
when transferred into the sensitive Envs described above, in-
creased neutralization resistance by Ͼ27- and Ͼ28-fold, respec-
tively, to the G37080 visit 2 BCN plasma antibodies. We observed
that the removal of a glycan at the 138 position in V1 (T140D) in
HVTR-PG80v1.eJ19 mediated the enhanced sensitivity of this Env
to G37080 plasma by 2.64-fold (Table 4). Concurrent with this
observation, we found that the alteration of the V1 region of
PG80v1.eJ19 with the T140D substitution in PG80v2.eJ38 Env
exhibited enhanced susceptibility compared to that of the
PG80v2.eJ38 Env chimera containing the PG80v1.eJ19 V1 loop, as
shown in Table 4. Our data indicate that N138 glycan masks the
PG80v1.eJ19 Env from being efficiently neutralized by the autol-
ogous plasma compared to that of its contemporaneous counter-
part, PG80v1.eJ7 Env. Fine scanning of V1 regions of the autolo-
gous Envs further revealed that the N133 glycan motif and P147
residues in the PG80v2.eJ38 Env played a significant role in neu-
tralization resistance to G37080 BCN autologous plasma antibod-
ies (Fig. 4B). Interestingly, all of the V1 chimeras as well as the
point mutants showed sensitivities to PG9 MAb comparable to
those of their wild types (Table 5), indicating that the shifts in
neutralization susceptibilities were not due to changes in Env con-
formation. Moreover, we noted that both the sensitive and the
resistant autologous Envs contain T332 in the V3 base, clearly
indicating that the absence of N332 was not associated with resis-
tance to autologous neutralization. Similar observations were
made with respect to N160, R166, and K169 amino acid residues,
further consolidating that the neutralization conferred by G37080
BCN plasma antibodies was not associated with antibody target-
ing these epitopes in autologous Envs, and is likely the case for all
of the Envs tested against G37080 plasma antibodies.
DISCUSSION
The identification of the molecular specificities of antibodies elic-
ited in natural infection and that mediate neutralization breadth
and potency is key in the design and development of suitable Env-
based immunogens capable of eliciting similar antibody responses
upon vaccination. In the present study, we characterized the mo-
lecular specificity of plasma antibodies obtained from an Indian
elite neutralizer (G37080) infected with HIV-1 clade C that dis-
played exceptional cross-neutralization of different clades of distinct
geographical origins. The G37080 plasma was found to contain the
most broad and potent cross-neutralizing antibodies among the two
hundred plasma samples obtained from Indian patients chronically
infected with HIV-1. Plasma samples collected from the G37080 do-
nor at two time points at 8 months apart showed similar neutraliza-
tion breadth with modest increase in potency in the follow-up visit,
indicating an association with the sustained maturation of antibody-
producing B cells in this individual.
Since polyclonal plasma antibodies are not suitable for epitope
mapping, we examined the specificity of the G37080 BCN plasma
by making use of mutant viruses with specific point substitutions
of known neutralizing epitopes with nonspecific amino acids and
via depletion with monomeric and trimeric Envs in addition to
MPER peptide. The G37080 plasma antibodies did not show de-
pendence on the N160/K169 and N332 epitopes in the V2 apex
and V3 base, respectively. Our data also are consistent with the
target epitopes of the G37080 BCN antibodies being distinct from
those which are recognized by 2G12 (74), PGT121-128 (17), and
PGT130-131 and PGT135 (19) (e.g., residues at the following po-
TABLE 4 Dissection of specificity for autologous neutralization
resistance
Chimera and point mutant
Neutralization potency
(ID50)
Fold
changea
Effectb
PG80v1.eJ7 Env backbone
V1V2 loop
PG80v2.eJ38 (V1V2) in v1.eJ7 45.75 Decrease
PG80v2.eJ38 (V1) in v1.eJ7 27.97 Decrease
PG80v2.eJ38 (V2) in v1.eJ7 1.35 No effect
Point mutations
PG80v1.eJ7 (D133N) 3.25 Decrease
PG80v1.eJ7 (S143G) 0.87 No effect
PG80v1.eJ7 (D133N ϩ S143G) 2.88 Decrease
PG80v1.eJ7 (T147P) 8.21 Decrease
PG80v1.eJ19 Env backbone
V1V2 loop
PG80v2.eJ38(V1V2) in v1.eJ19 23.05 Decrease
PG80v2.eJ38 (V1) in v1.eJ19 28.61 Decrease
PG80v2.eJ38 (V2) in v1.eJ19 1.87 Increase
Point mutations
PG80v1.eJ19 (D133N) 2.51 Decrease
PG80v1.eJ19 (T139A) 0.99 No effect
PG80v1.eJ19 (T139A ϩ T140D) 2.64 Increase
PG80v1.eJ19 (N143G) 1.38 No effect
PG80v1.eJ19 (T139A ϩ T140D ϩ N143G) 2.16 Increase
PG80v1.eJ19 (T145P) 3.24 Decrease
PG80v2.eJ38 Env backbone
V1V2 loop
PG80v1.eJ7 (V1V2) in v2.eJ38 26.56 Increase
PG80v1.eJ7 (V1) in v2.eJ38 49.62 Increase
PG80v1.eJ7 (V2) in v2.eJ38 1.07 No effect
PG80v1.eJ19(V1V2) in v2.eJ38 12.81 Increase
PG80v1.eJ19 (V1) in v2.eJ38 37.60 Increase
PG80v1.eJ19 (V2) in v2.eJ38 0.94 No effect
Other regions in gp120
PG80v1.eJ7 (V3C3) in v2.eJ38 0.84 No effect
PG80v1.eJ7 (V3C3V4C4) in v2.eJ38 0.90 No effect
PG80v1.eJ7 (C4V5C5) in v2.eJ38 1.08 No effect
PG80v1.eJ19 (V3C3) in PG80v2.eJ38 1.09 No effect
PG80v1.eJ19 (V3C3V4C4) in PG80v2.eJ38 1.15 No effect
PG80v1.eJ19 (C4V5C5) in PG80v2.eJ38 0.96 No effect
Point mutations
PG80v1.eJ19 V1 (T139AϩT140D) in
v2.eJ38
53.91 Increase
PG80v2.eJ38 (N133D) 4.72 Increase
PG80v2.eJ38 (G143S) 0.94 No effect
PG80v2.eJ38 (N133DϩG143S) 3.63 Increase
PG80v2.eJ38 (P147T) 4.11 Increase
Heterologous Env chimera
V1V2 loop
16055-2.3 (25711-2.4 V1V2) 18.38 Increase
25711-2.4 (16055-2.3 V1V2) 2.03 Decrease
CAP45 (25711-2.4 V1V2) 16.84 Increase
25711-2.4 (CAP45-V1V2) 10.54 Decrease
a
Fold changes in reciprocal dilution of plasma mediating 50% virus neutralization
(ID50).
b
Fold increase or decrease in neutralization titer (ID50 values).
Patil et al.
3452 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
sitions: 295, 297, 301, 332, 334, 386, 388, 392, 394, 448, and 450);
thus, BCN G37080 antibodies appear to target a new epitope. Our
data highlighting the N332-independent development of neutral-
izing antibodies in clade C-infected donor G37080 also differ from
recent findings (57, 75–77) associating N332 with the develop-
ment of broad and potent neutralizing antibody, especially in
clade C infection in African donors. Moreover, recent studies in-
dicating the role of K169 as a target of BCN antibodies obtained
from a clade C-infected South African donor (41, 56) and the
observation that vaccine-induced protection in the RV144 vaccine
trial was associated with antibodies targeting epitopes, including
K169 in the V2 apex (27, 78), prompted us to examine whether
broad neutralization of the G37080 plasma antibodies also was
dependent on the K169 epitope. In the present study, the neutral-
ization potency of G37080 was unaffected by N160A/K169A
knockout mutations, and we also observed that both sensitive and
resistant autologous Envs obtained from both visits contain N160
and K169 in the V2 region. Hence, owing to the lack of association
of neutralization breadth of the G37080 BCN antibodies with
N160, K169, and N332 dependencies, our study further high-
lighted that there is a likelihood of differences in the development
pathway of elicitation of broadly neutralizing antibodies in indi-
viduals infected with HIV-1 clade C, particularly those with eth-
nically distinct variants.
Wibmer et al. (41) recently demonstrated an association be-
tween the evolution of a broadly neutralizing antibody response in
a clade C-infected donor with shifts in antibody specificities from
the recognition of epitopes in V2 to the CD4bs. In the present
study, the G37080 neutralizing plasma antibodies obtained from
both visits were found not to be absorbed by the TripleMut (66,
79) and RSC3 (22) core proteins, which effectively absorb anti-
bodies directed to the CD4bs. This result indicates a lack of devel-
opment of CD4bs-directed neutralizing antibodies during the dis-
ease course in the G37080 donor. Additionally, the absence of
MPER-directed antibodies from G37080 plasma was found, al-
though a negligible antibody titer (1:300 reciprocal dilutions) to
the HIV2/HIV1 (C1C) chimera was observed with plasma sam-
FIG 4 (A) Neutralization susceptibility of autologous Envs to contemporaneous G37080 BCN plasma and its follow-up sample from the same donor.
Neutralization titers (median ID50) were obtained by titrating pseudotyped viruses expressing autologous Envs obtained from visit 1 and follow-up G37080
plasma to contemporaneous plasma antibodies. Note that both of the Envs obtained from follow-up G37080 plasma (visit 2) were found to be resistant to
contemporaneous autologous plasma, while Envs obtained from visit 1 G37080 plasma were found to be sensitive to follow-up plasma antibodies. (B) Alignment
of V1V2 amino acid sequences of sensitive and resistant autologous Envs obtained at both visits was done by using seqpublish, available at the HIV Los Alamos
database (www.hiv.lanl.gov). Key residues that mediate autologous neutralization resistance are highlighted.
TABLE 5 Sensitivity of wild type, chimera, and point mutants of
autologous Envs to PG9 MAb
Env chimera and mutant ID50
PG80v1.eJ7 (wild type) 0.12
PG80v1.eJ19 (wild type) 0.97
PG80v1.eJ19 (T139A ϩ T140D) 0.77
PG80v2.eJ38 (wild type) 0.02
PG80v1.eJ7 (V1) in v2.eJ38 0.04
PG80v1.eJ19 (V1) in v2.eJ38 0.05
PG80v1.eJ19 (V1) (T139A ϩ T140D) in v2.eJ38 0.06
PG80v2.eJ38 (N133D) 0.01
PG80v2.eJ38 (P147T) 0.04
HIV-1 Clade C Plasma Confers Cross-Clade Neutralization
April 2016 Volume 90 Number 7 jvi.asm.org 3453Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
ples from both visits. However, the neutralization breadth of the
G37080 plasma was not found to be associated with the presence
of MPER-directed antibody. Nonetheless, we do not rule out the
possibility that in the further course of infection, this donor may
be able to develop MPER-directed antibodies.
Recentstudieshaveshownthatneutralizingantibodiesthattarget
conformational epitopes bind exclusively to the cleaved near-native
trimeric Envs (15, 64, 80, 81). In the present study, we found that the
absorption of G37080 plasma antibodies to soluble trimeric BG505-
SOSIP.664 Env was associated with the depletion of neutralizing ac-
tivity in G37080 BCN plasma. However, we do not rule out the pos-
sibility of the presence of 39F-, 19b-, and 14e-like nonneutralizing
antibodies that were reported to bind to BG505-SOSIP.664 trimeric
Env (69). Our findings indicate that the G37080 BCN antibodies tar-
get conformational epitopes in gp120. Our observation also high-
lights that native-like trimeric Envs, such as BG505-SOSIP.664, can
be utilized in selecting antigen-specific memory B cells, as reported
earlier(82),fromdonorG37080towardisolationofMAbcorrelating
with broad neutralization displayed by the plasma antibodies.
We made use of env clones obtained from autologous G37080
plasma from both time points to refine the fine specificity of the
G37080 BCN plasma antibodies. By examining chimeric Envs and
mutant viruses, we identified key residues in the V1 loop associ-
ated with neutralization resistance. Interestingly, the Env chimera
and mutant viruses showed susceptibility to PG9 MAb compara-
ble to that of their respective wild-type Envs, indicating that they
did not alter Env conformation. We identified a glycan at the 133
position and a proline residue at the 147 position within the V1
loop of the resistant Env (PG80v2.eJ38) that were found to be
associated with neutralization escape, which indicated that these
are contact sites for the G37080 BCN plasma antibodies. Thus,
from our study we conclude that changes in V1 loop sequence are
associated with the escape of autologous viruses to the BCN
G37080 plasma. Additionally, an examination of the degree of
susceptibilities of pseudoviruses expressing chimeric heterolo-
gous Envs to the G37080 plasma revealed that the BCN plasma
antibodies predominantly target epitopes in the V1V2 region in
gp120. However, we do not rule out the possibility of the contri-
bution of other discontinuous epitopes in gp120 in mediating
neutralization breadth. The isolation and identification of mono-
clonal antibodies from this elite neutralizer donor (G37080) will
help precisely map specific epitopes associated with neutralization
breadth and potency.
In summary, we identified an HIV-1-infected elite neutralizer
whose plasma showed exceptional neutralization breadth, and we
provided evidence that it targets novel conformational epitopes
on trimeric Env, predominantly in the V1V2 region, not reported
previously. Moreover, the neutralization resistance of the autolo-
gous Envs to G37080 plasma is associated with substitutions of
novel residues within the V1 loop that form the key contact points
of the BCN plasma antibody. The identification of novel epitopes
associated with broad neutralization of HIV-1, in particular the
major circulating clade C strains, will significantly contribute to
efforts toward effective immunogen design.
ACKNOWLEDGMENTS
We thank all of the Protocol G study participants registered with YRG
Care, Chennai, all of the research staff members at the Protocol G clinical
center at YRG Care, Chennai, and all of the IAVI Protocol G team mem-
bers. We sincerely thank Christopher Parks, IAVI Design and Develop-
ment Laboratory, for providing valuable input in preparing the manu-
script, and we also thank G. Balakrish Nair and Sudhanshu Vrati (THSTI),
Shreyasi Chatterjee, and all of the HVTR laboratory members for support.
We thank Albert Cupo, John P. Moore, and the members the SOSIP
trimer HIVRAD team, Weill Cornell Medical College, New York, for pro-
viding us with BG505.SOSIP.664 plasmid DNA and purified protein. We
thank David Montefiori, Lynn Morris, Pascal Poignard, and Richard Wy-
att for making available many reagents used in our study. The following
reagent was obtained through the NIH AIDS Reagent Program, Division
of AIDS, NIAID, NIH, from John C. Kappes and Xiaoyun Wu: pSG3 env.
IAVI’s work was made possible by generous support from many do-
nors, including the Bill & Melinda Gates Foundation, the Ministry of
Foreign Affairs of Denmark, Irish Aid, the Ministry of Finance of Japan,
the Ministry of Foreign Affairs of the Netherlands, the Norwegian Agency
for Development Cooperation (NORAD), the United Kingdom Depart-
ment for International Development (DFID), and the United States
Agency for International Development (USAID). The full list of IAVI
donors is available at www.iavi.org. The contents are the responsibility of
the International AIDS Vaccine Initiative and do not necessarily reflect
the views of USAID or the United States Government.
The International AIDS Vaccine Initiative has filed a patent relating to
the autologous HIV-1 clade C envelope clones (J. Bhattacharya, S. Desh-
pande, S. Patil, R. Kumar, and B. K. Chakrabarti, U.S. patent application
62/254,971).
FUNDING INFORMATION
This study was made possible by the generous support of the American
people through the United States Agency for International Development
(USAID) through the IAVI, support from a THSTI-IAVI HIV Vaccine
Design Program grant through the Department of Biotechnology, Gov-
ernment of India, in part by a grant from the Department of Science and
Technology, Government of India (DST/INT/SAFR/Mega-P3/2011 to
J.B.), and in part by a DBT National Bioscience Research Award [BT/
HRD/NBA34/01/2012-13(iv) to J.B.]. The funders had no role in study
design, data collection and interpretation, or the decision to submit the
work for publication.
REFERENCES
1. Caskey M, Klein F, Lorenzi JC, Seaman MS, West AP, Jr, Buckley N,
Kremer G, Nogueira L, Braunschweig M, Scheid JF, Horwitz JA, Shime-
liovich I, Ben-Avraham S, Witmer-Pack M, Platten M, Lehmann C,
Burke LA, Hawthorne T, Gorelick RJ, Walker BD, Keler T, Gulick RM,
Fatkenheuer G, Schlesinger SJ, Nussenzweig MC. 2015. Viraemia sup-
pressed in HIV-1-infected humans by broadly neutralizing antibody
3BNC117. Nature 522:487–491. http://dx.doi.org/10.1038/nature14411.
2. Esparza J. 2013. A brief history of the global effort to develop a preventive
HIV vaccine. Vaccine 31:3502–3518. http://dx.doi.org/10.1016/j.vaccine
.2013.05.018.
3. Hraber P, Seaman MS, Bailer RT, Mascola JR, Montefiori DC, Korber
BT. 2014. Prevalence of broadly neutralizing antibody responses during
chronic HIV-1 infection. AIDS 28:163–169. http://dx.doi.org/10.1097
/QAD.0000000000000106.
4. Klein F, Mouquet H, Dosenovic P, Scheid JF, Scharf L, Nussenzweig
MC. 2013. Antibodies in HIV-1 vaccine development and therapy. Sci-
ence 341:1199–1204. http://dx.doi.org/10.1126/science.1241144.
5. Koff WC, Russell ND, Walport M, Feinberg MB, Shiver JW, Karim SA,
Walker BD, McGlynn MG, Nweneka CV, Nabel GJ. 2013. Accelerating
the development of a safe and effective HIV vaccine: HIV vaccine case
study for the decade of vaccines. Vaccine 31(Suppl 2):B204–B208. http:
//dx.doi.org/10.1016/j.vaccine.2012.10.115.
6. Kwong PD, Mascola JR, Nabel GJ. 2013. Broadly neutralizing antibodies
and the search for an HIV-1 vaccine: the end of the beginning. Nat Rev
Immunol 13:693–701. http://dx.doi.org/10.1038/nri3516.
7. Braibant M, Brunet S, Costagliola D, Rouzioux C, Agut H, Katinger H,
Autran B, Barin F. 2006. Antibodies to conserved epitopes of the HIV-1
envelope in sera from long-term non-progressors: prevalence and associ-
ation with neutralizing activity. AIDS 20:1923–1930. http://dx.doi.org/10
.1097/01.aids.0000247113.43714.5e.
Patil et al.
3454 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
8. Donners H, Willems B, Beirnaert E, Colebunders R, Davis D, van der
Groen G. 2002. Cross-neutralizing antibodies against primary isolates in
African women infected with HIV-1. AIDS 16:501–503. http://dx.doi.org
/10.1097/00002030-200202150-00030.
9. Gray ES, Madiga MC, Hermanus T, Moore PL, Wibmer CK, Tumba
NL, Werner L, Mlisana K, Sibeko S, Williamson C, Abdool Karim SS,
Morris L. 2011. The neutralization breadth of HIV-1 develops incremen-
tally over four years and is associated with CD4ϩ T cell decline and high
viral load during acute infection. J Virol 85:4828–4840. http://dx.doi.org
/10.1128/JVI.00198-11.
10. Li Y, Svehla K, Louder MK, Wycuff D, Phogat S, Tang M, Migueles SA,
Wu X, Phogat A, Shaw GM, Connors M, Hoxie J, Mascola JR, Wyatt R.
2009. Analysis of neutralization specificities in polyclonal sera derived
from human immunodeficiency virus type 1-infected individuals. J Virol
83:1045–1059. http://dx.doi.org/10.1128/JVI.01992-08.
11. Richman DD, Wrin T, Little SJ, Petropoulos CJ. 2003. Rapid evolu-
tion of the neutralizing antibody response to HIV type 1 infection.
Proc Natl Acad Sci U S A 100:4144–4149. http://dx.doi.org/10.1073
/pnas.0630530100.
12. Sather DN, Armann J, Ching LK, Mavrantoni A, Sellhorn G, Caldwell
Z, Yu X, Wood B, Self S, Kalams S, Stamatatos L. 2009. Factors
associated with the development of cross-reactive neutralizing antibodies
during human immunodeficiency virus type 1 infection. J Virol 83:757–
769. http://dx.doi.org/10.1128/JVI.02036-08.
13. Simek MD, Rida W, Priddy FH, Pung P, Carrow E, Laufer DS, Lehrman
JK, Boaz M, Tarragona-Fiol T, Miiro G, Birungi J, Pozniak A, McPhee
DA, Manigart O, Karita E, Inwoley A, Jaoko W, Dehovitz J, Bekker LG,
Pitisuttithum P, Paris R, Walker LM, Poignard P, Wrin T, Fast PE,
Burton DR, Koff WC. 2009. Human immunodeficiency virus type 1 elite
neutralizers: individuals with broad and potent neutralizing activity iden-
tified by using a high-throughput neutralization assay together with an
analytical selection algorithm. J Virol 83:7337–7348. http://dx.doi.org/10
.1128/JVI.00110-09.
14. Stamatatos L, Morris L, Burton DR, Mascola JR. 2009. Neutralizing
antibodies generated during natural HIV-1 infection: good news for an
HIV-1 vaccine? Nat Med 15:866–870.
15. Falkowska E, Le KM, Ramos A, Doores KJ, Lee JH, Blattner C, Ramirez
A, Derking R, MJ van Gils Liang CH, McBride R, von Bredow B,
Shivatare SS, Wu CY, Chan-Hui PY, Liu Y, Feizi T, Zwick MB, Koff
WC, Seaman MS, Swiderek K, Moore JP, Evans D, Paulson JC, Wong
CH, Ward AB, Wilson IA, Sanders RW, Poignard P, Burton DR. 2014.
Broadly neutralizing HIV antibodies define a glycan-dependent epitope
on the prefusion conformation of gp41 on cleaved envelope trimers. Im-
munity 40:657–668. http://dx.doi.org/10.1016/j.immuni.2014.04.009.
16. Huang J, Ofek G, Laub L, Louder MK, Doria-Rose NA, Longo NS,
Imamichi H, Bailer RT, Chakrabarti B, Sharma SK, Alam SM, Wang T,
Yang Y, Zhang B, Migueles SA, Wyatt R, Haynes BF, Kwong PD,
Mascola JR, Connors M. 2012. Broad and potent neutralization of HIV-1
by a gp41-specific human antibody. Nature 491:406–412. http://dx.doi
.org/10.1038/nature11544.
17. Julien JP, Sok D, Khayat R, Lee JH, Doores KJ, Walker LM, Ramos A,
Diwanji DC, Pejchal R, Cupo A, Katpally U, Depetris RS, Stanfield RL,
McBride R, Marozsan AJ, Paulson JC, Sanders RW, Moore JP, Burton
DR, Poignard P, Ward AB, Wilson IA. 2013. Broadly neutralizing anti-
body PGT121 allosterically modulates CD4 binding via recognition of the
HIV-1 gp120 V3 base and multiple surrounding glycans. PLoS Pathog
9:e1003342. http://dx.doi.org/10.1371/journal.ppat.1003342.
18. Scharf L, Scheid JF, Lee JH, West AP, Jr, Chen C, Gao H, Gnanapra-
gasam PN, Mares R, Seaman MS, Ward AB, Nussenzweig MC, Bjork-
man PJ. 2014. Antibody 8ANC195 reveals a site of broad vulnerability on
the HIV-1 envelope spike. Cell Rep 7:785–795. http://dx.doi.org/10.1016
/j.celrep.2014.04.001.
19. Walker LM, Huber M, Doores KJ, Falkowska E, Pejchal R, Julien JP,
Wang SK, Ramos A, Chan-Hui PY, Moyle M, Mitcham JL, Hammond
PW, Olsen OA, Phung P, Fling S, Wong CH, Phogat S, Wrin T, Simek
MD, Protocol G Principal Investigators, Koff WC, Wilson IA, Burton
DR, Poignard P. 2011. Broad neutralization coverage of HIV by multiple
highly potent antibodies. Nature 477:466–470. http://dx.doi.org/10.1038
/nature10373.
20. Walker LM, Phogat SK, Chan-Hui PY, Wagner D, Phung P, Goss JL,
Wrin T, Simek MD, Fling S, Mitcham JL, Lehrman JK, Priddy FH,
Olsen OA, Frey SM, Hammond PW, Kaminsky S, Zamb T, Moyle M,
Koff WC, Poignard P, Burton DR. 2009. Broad and potent neutralizing
antibodies from an African donor reveal a new HIV-1 vaccine target. Sci-
ence 326:285–289. http://dx.doi.org/10.1126/science.1178746.
21. Walker LM, Simek MD, Priddy F, Gach JS, Wagner D, Zwick MB,
Phogat SK, Poignard P, Burton DR. 2010. A limited number of antibody
specificities mediate broad and potent serum neutralization in selected
HIV-1 infected individuals. PLoS Pathog 6:e1001028. http://dx.doi.org/10
.1371/journal.ppat.1001028.
22. Wu X, Yang ZY, Li Y, Hogerkorp CM, Schief WR, Seaman MS, Zhou
T, Schmidt SD, Wu L, Xu L, Longo NS, McKee K, O’Dell S, Louder MK,
Wycuff DL, Feng Y, Nason M, Doria-Rose N, Connors M, Kwong PD,
Roederer M, Wyatt RT, Nabel GJ, Mascola JR. 2010. Rational design of
envelope identifies broadly neutralizing human monoclonal antibodies to
HIV-1. Science 329:856–861. http://dx.doi.org/10.1126/science.1187659.
23. Wu X, Zhou T, Zhu J, Zhang B, Georgiev I, Wang C, Chen X, Longo
NS, Louder M, McKee K, O’Dell S, Perfetto S, Schmidt SD, Shi W, Wu
L, Yang Y, Yang ZY, Yang Z, Zhang Z, Bonsignori M, Crump JA,
Kapiga SH, Sam NE, Haynes BF, Simek M, Burton DR, Koff WC,
Doria-Rose NA, Connors M, NISC Comparative Sequencing Program,
Mullikin JC, Nabel GJ, Roederer M, Shapiro L, Kwong PD, Mascola JR.
2011. Focused evolution of HIV-1 neutralizing antibodies revealed by
structures and deep sequencing. Science 333:1593–1602. http://dx.doi.org
/10.1126/science.1207532.
24. Diskin R, Scheid JF, Marcovecchio PM, West AP, Jr, Klein F, Gao H,
Gnanapragasam PN, Abadir A, Seaman MS, Nussenzweig MC, Bjork-
man PJ. 2011. Increasing the potency and breadth of an HIV antibody by
using structure-based rational design. Science 334:1289–1293. http://dx
.doi.org/10.1126/science.1213782.
25. Klein F, Gaebler C, Mouquet H, Sather DN, Lehmann C, Scheid JF,
Kraft Z, Liu Y, Pietzsch J, Hurley A, Poignard P, Feizi T, Morris L,
Walker BD, Fatkenheuer G, Seaman MS, Stamatatos L, Nussenzweig
MC. 2012. Broad neutralization by a combination of antibodies recogniz-
ing the CD4 binding site and a new conformational epitope on the HIV-1
envelope protein. J Exp Med 209:1469–1479. http://dx.doi.org/10.1084
/jem.20120423.
26. Scheid JF, Mouquet H, Ueberheide B, Diskin R, Klein F, Oliveira
TY, Pietzsch J, Fenyo D, Abadir A, Velinzon K, Hurley A, Myung S,
Boulad F, Poignard P, Burton DR, Pereyra F, Ho DD, Walker BD,
Seaman MS, Bjorkman PJ, Chait BT, Nussenzweig MC. 2011. Se-
quence and structural convergence of broad and potent HIV antibod-
ies that mimic CD4 binding. Science 333:1633–1637. http://dx.doi.org
/10.1126/science.1207227.
27. Liao HX, Bonsignori M, Alam SM, McLellan JS, Tomaras GD, Moody
MA, Kozink DM, Hwang KK, Chen X, Tsao CY, Liu P, Lu X, Parks RJ,
Montefiori DC, Ferrari G, Pollara J, Rao M, Peachman KK, Santra S,
Letvin NL, Karasavvas N, Yang ZY, Dai K, Pancera M, Gorman J,
Wiehe K, Nicely NI, Rerks-Ngarm S, Nitayaphan S, Kaewkungwal J,
Pitisuttithum P, Tartaglia J, Sinangil F, Kim JH, Michael NL, Kepler
TB, Kwong PD, Mascola JR, Nabel GJ, Pinter A, Zolla-Pazner S, Haynes
BF. 2013. Vaccine induction of antibodies against a structurally heteroge-
neous site of immune pressure within HIV-1 envelope protein variable
regions 1 and 2. Immunity 38:176–186. http://dx.doi.org/10.1016/j
.immuni.2012.11.011.
28. Pejchal R, Doores KJ, Walker LM, Khayat R, Huang PS, Wang SK,
Stanfield RL, Julien JP, Ramos A, Crispin M, Depetris R, Katpally U,
Marozsan A, Cupo A, Maloveste S, Liu Y, McBride R, Ito Y, Sanders
RW, Ogohara C, Paulson JC, Feizi T, Scanlan CN, Wong CH, Moore
JP, Olson WC, Ward AB, Poignard P, Schief WR, Burton DR, Wilson
IA. 2011. A potent and broad neutralizing antibody recognizes and pen-
etrates the HIV glycan shield. Science 334:1097–1103. http://dx.doi.org
/10.1126/science.1213256.
29. Blattner C, Lee JH, Sliepen K, Derking R, Falkowska E, de la Pena AT,
Cupo A, Julien JP, van Gils M, Lee PS, Peng W, Paulson JC, Poignard
P, Burton DR, Moore JP, Sanders RW, Wilson IA, Ward AB. 2014.
Structural delineation of a quaternary, cleavage-dependent epitope at the
gp41-gp120 interface on intact HIV-1 Env trimers. Immunity 40:669–
680. http://dx.doi.org/10.1016/j.immuni.2014.04.008.
30. Morris L, Chen X, Alam M, Tomaras G, Zhang R, Marshall DJ, Chen
B, Parks R, Foulger A, Jaeger F, Donathan M, Bilska M, Gray ES,
Abdool Karim SS, Kepler TB, Whitesides J, Montefiori D, Moody MA,
Liao HX, Haynes BF. 2011. Isolation of a human anti-HIV gp41 mem-
brane proximal region neutralizing antibody by antigen-specific single B
cell sorting. PLoS One 6:e23532. http://dx.doi.org/10.1371/journal.pone
.0023532.
HIV-1 Clade C Plasma Confers Cross-Clade Neutralization
April 2016 Volume 90 Number 7 jvi.asm.org 3455Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
31. Nelson JD, Brunel FM, Jensen R, Crooks ET, Cardoso RM, Wang M,
Hessell A, Wilson IA, Binley JM, Dawson PE, Burton DR, Zwick MB.
2007. An affinity-enhanced neutralizing antibody against the membrane-
proximal external region of human immunodeficiency virus type 1 gp41
recognizes an epitope between those of 2F5 and 4E10. J Virol 81:4033–
4043. http://dx.doi.org/10.1128/JVI.02588-06.
32. Zwick MB, Labrijn AF, Wang M, Spenlehauer C, Saphire EO, Binley
JM, Moore JP, Stiegler G, Katinger H, Burton DR, Parren PW. 2001.
Broadly neutralizing antibodies targeted to the membrane-proximal
external region of human immunodeficiency virus type 1 glycoprotein
gp41. J Virol 75:10892–10905. http://dx.doi.org/10.1128/JVI.75.22
.10892-10905.2001.
33. Chaillon A, Braibant M, Moreau T, Thenin S, Moreau A, Autran B,
Barin F. 2011. The V1V2 domain and an N-linked glycosylation site in the
V3 loop of the HIV-1 envelope glycoprotein modulate neutralization sen-
sitivity to the human broadly neutralizing antibody 2G12. J Virol 85:3642–
3648. http://dx.doi.org/10.1128/JVI.02424-10.
34. Doria-Rose NA, Georgiev I, O’Dell S, Chuang GY, Staupe RP, McLellan JS,
Gorman J, Pancera M, Bonsignori M, Haynes BF, Burton DR, Koff WC,
Kwong PD, Mascola JR. 2012. A short segment of the HIV-1 gp120 V1/V2
region is a major determinant of resistance to V1/V2 neutralizing antibodies.
J Virol 86:8319–8323. http://dx.doi.org/10.1128/JVI.00696-12.
35. Doria-Rose NA, Schramm CA, Gorman J, Moore PL, Bhiman JN,
DeKosky BJ, Ernandes MJ, Georgiev IS, Kim HJ, Pancera M, Staupe RP,
Altae-Tran HR, Bailer RT, Crooks ET, Cupo A, Druz A, Garrett NJ, Hoi
KH, Kong R, Louder MK, Longo NS, McKee K, Nonyane M, O’Dell S,
Roark RS, Rudicell RS, Schmidt SD, Sheward DJ, Soto C, Wibmer CK,
Yang Y, Zhang Z, NISC Comparative Sequencing Program, Mullikin
JC, Binley JM, Sanders RW, Wilson IA, Moore JP, Ward AB, Georgiou
G, Williamson C, Abdool Karim SS, Morris L, Kwong PD, Shapiro L,
Mascola JR. 2014. Developmental pathway for potent V1V2-directed
HIV-neutralizing antibodies. Nature 509:55–62. http://dx.doi.org/10
.1038/nature13036.
36. Harrington PR, Nelson JA, Kitrinos KM, Swanstrom R. 2007. Indepen-
dent evolution of human immunodeficiency virus type 1 env V1/V2 and
V4/V5 hypervariable regions during chronic infection. J Virol 81:5413–
5417. http://dx.doi.org/10.1128/JVI.02554-06.
37. Moore PL, Gray ES, Choge IA, Ranchobe N, Mlisana K, Abdool Karim
SS, Williamson C, Morris L. 2008. The c3-v4 region is a major target of
autologous neutralizing antibodies in human immunodeficiency virus
type 1 subtype C infection. J Virol 82:1860–1869. http://dx.doi.org/10
.1128/JVI.02187-07.
38. Rusert P, Krarup A, Magnus C, Brandenberg OF, Weber J, Ehlert AK,
Regoes RR, Gunthard HF, Trkola A. 2011. Interaction of the gp120 V1V2
loop with a neighboring gp120 unit shields the HIV envelope trimer
against cross-neutralizing antibodies. J Exp Med 208:1419–1433. http:
//dx.doi.org/10.1084/jem.20110196.
39. Sagar M, Wu X, Lee S, Overbaugh J. 2006. Human immunodeficiency
virus type 1 V1-V2 envelope loop sequences expand and add glycosylation
sites over the course of infection, and these modifications affect antibody
neutralization sensitivity. J Virol 80:9586–9598. http://dx.doi.org/10.1128
/JVI.00141-06.
40. van Gils MJ, Bunnik EM, Boeser-Nunnink BD, Burger JA, Terlouw-
Klein M, Verwer N, Schuitemaker H. 2011. Longer V1V2 region with
increased number of potential N-linked glycosylation sites in the HIV-1
envelope glycoprotein protects against HIV-specific neutralizing antibod-
ies. J Virol 85:6986–6995. http://dx.doi.org/10.1128/JVI.00268-11.
41. Wibmer CK, Bhiman JN, Gray ES, Tumba N, Abdool Karim SS,
Williamson C, Morris L, Moore PL. 2013. Viral escape from HIV-1
neutralizing antibodies drives increased plasma neutralization breadth
through sequential recognition of multiple epitopes and immuno-
types. PLoS Pathog 9:e1003738. http://dx.doi.org/10.1371/journal
.ppat.1003738.
42. Alter G, Barouch DH. 2015. Natural evolution of broadly neutralizing
antibodies. Cell 161:427–428. http://dx.doi.org/10.1016/j.cell.2015.04
.007.
43. Doores KJ, Kong L, Krumm SA, Le KM, Sok D, Laserson U, Garces F,
Poignard P, Wilson IA, Burton DR. 2015. Two classes of broadly neu-
tralizing antibodies within a single lineage directed to the high-mannose
patch of HIV envelope. J Virol 89:1105–1118. http://dx.doi.org/10.1128
/JVI.02905-14.
44. Doria-Rose NA, Joyce MG. 2015. Strategies to guide the antibody
affinity maturation process. Curr Opin Virol 11:137–147. http://dx.doi
.org/10.1016/j.coviro.2015.04.002.
45. Fera D, Schmidt AG, Haynes BF, Gao F, Liao HX, Kepler TB, Harrison
SC. 2014. Affinity maturation in an HIV broadly neutralizing B-cell lin-
eage through reorientation of variable domains. Proc Natl Acad Sci U S A
111:10275–10280. http://dx.doi.org/10.1073/pnas.1409954111.
46. Horiya S, Bailey JK, Temme JS, Guillen Schlippe YV, Krauss IJ. 2014.
Directed evolution of multivalent glycopeptides tightly recognized by HIV
antibody 2G12. J Am Chem Soc 136:5407–5415. http://dx.doi.org/10.1021
/ja500678v.
47. Mikell I, Stamatatos L. 2012. Evolution of cross-neutralizing antibody
specificities to the CD4-BS and the carbohydrate cloak of the HIV Env in
an HIV-1-infected subject. PLoS One 7:e49610. http://dx.doi.org/10.1371
/journal.pone.0049610.
48. Sather DN, Carbonetti S, Malherbe DC, Pissani F, Stuart AB, Hessell
AJ, Gray MD, Mikell I, Kalams SA, Haigwood NL, Stamatatos L. 2014.
Emergence of broadly neutralizing antibodies and viral coevolution in two
subjects during the early stages of infection with human immunodefi-
ciency virus type 1. J Virol 88:12968–12981. http://dx.doi.org/10.1128/JVI
.01816-14.
49. Wu X, Zhang Z, Schramm CA, Joyce MG, Do Kwon Y, Zhou T, Sheng
Z, Zhang B, O’Dell S, McKee K, Georgiev IS, Chuang GY, Longo NS,
Lynch RM, Saunders KO, Soto C, Srivatsan S, Yang Y, Bailer RT,
Louder MK, NISC Comparative Sequencing Program, Mullikin JC,
Connors M, Kwong PD, Mascola JR, Shapiro L. 2015. Maturation and
diversity of the VRC01-antibody lineage over 15 years of chronic HIV-1
infection. Cell 161:470–485. http://dx.doi.org/10.1016/j.cell.2015.03.004.
50. Zhu J, Ofek G, Yang Y, Zhang B, Louder MK, Lu G, McKee K, Pancera
M, Skinner J, Zhang Z, Parks R, Eudailey J, Lloyd KE, Blinn J, Alam
SM, Haynes BF, Simek M, Burton DR, Koff WC, NISC Comparative
Sequencing Program, Mullikin JC, Mascola JR, Shapiro L, Kwong PD.
2013. Mining the antibodyome for HIV-1-neutralizing antibodies with
next-generation sequencing and phylogenetic pairing of heavy/light
chains. Proc Natl Acad Sci U S A 110:6470–6475. http://dx.doi.org/10
.1073/pnas.1219320110.
51. Basu D, Kraft CS, Murphy MK, Campbell PJ, Yu T, Hraber PT, Irene
C, Pinter A, Chomba E, Mulenga J, Kilembe W, Allen SA, Derdeyn CA,
Hunter E. 2012. HIV-1 subtype C superinfected individuals mount low
autologous neutralizing antibody responses prior to intrasubtype super-
infection. Retrovirology 9:76. http://dx.doi.org/10.1186/1742-4690-9-76.
52. Gray ES, Meyers T, Gray G, Montefiori DC, Morris L. 2006. Insensi-
tivity of paediatric HIV-1 subtype C viruses to broadly neutralising mono-
clonal antibodies raised against subtype B. PLoS Med 3:e255. http://dx.doi
.org/10.1371/journal.pmed.0030255.
53. Gray ES, Moody MA, Wibmer CK, Chen X, Marshall D, Amos J, Moore
PL, Foulger A, Yu JS, Lambson B, Abdool Karim S, Whitesides J,
Tomaras GD, Haynes BF, Morris L, Liao HX. 2011. Isolation of a
monoclonal antibody that targets the alpha-2 helix of gp120 and repre-
sents the initial autologous neutralizing-antibody response in an HIV-1
subtype C-infected individual. J Virol 85:7719–7729. http://dx.doi.org/10
.1128/JVI.00563-11.
54. Gray ES, Moore PL, Choge IA, Decker JM, Bibollet-Ruche F, Li H,
Leseka N, Treurnicht F, Mlisana K, Shaw GM, Karim SS, Williamson C,
Morris L. 2007. Neutralizing antibody responses in acute human immu-
nodeficiency virus type 1 subtype C infection. J Virol 81:6187–6196. http:
//dx.doi.org/10.1128/JVI.00239-07.
55. Gray ES, Taylor N, Wycuff D, Moore PL, Tomaras GD, Wibmer CK,
Puren A, DeCamp A, Gilbert PB, Wood B, Montefiori DC, Binley JM,
Shaw GM, Haynes BF, Mascola JR, Morris L. 2009. Antibody specific-
ities associated with neutralization breadth in plasma from human immu-
nodeficiency virus type 1 subtype C-infected blood donors. J Virol 83:
8925–8937. http://dx.doi.org/10.1128/JVI.00758-09.
56. Moore PL, Gray ES, Sheward D, Madiga M, Ranchobe N, Lai Z,
Honnen WJ, Nonyane M, Tumba N, Hermanus T, Sibeko S, Mlisana K,
Abdool Karim SS, Williamson C, Pinter A, Morris L. 2011. Potent and
broad neutralization of HIV-1 subtype C by plasma antibodies targeting a
quaternary epitope including residues in the V2 loop. J Virol 85:3128–
3141. http://dx.doi.org/10.1128/JVI.02658-10.
57. Moore PL, Gray ES, Wibmer CK, Bhiman JN, Nonyane M, Sheward DJ,
Hermanus T, Bajimaya S, Tumba NL, Abrahams MR, Lambson BE,
Ranchobe N, Ping L, Ngandu N, Abdool Karim Q, Abdool Karim SS,
Swanstrom RI, Seaman MS, Williamson C, Morris L. 2012. Evolution of
an HIV glycan-dependent broadly neutralizing antibody epitope through
Patil et al.
3456 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
immune escape. Nat Med 18:1688–1692. http://dx.doi.org/10.1038/nm
.2985.
58. Moore PL, Ranchobe N, Lambson BE, Gray ES, Cave E, Abrahams MR,
Bandawe G, Mlisana K, Abdool Karim SS, Williamson C, Morris L.
2009. Limited neutralizing antibody specificities drive neutralization es-
cape in early HIV-1 subtype C infection. PLoS Pathog 5:e1000598. http:
//dx.doi.org/10.1371/journal.ppat.1000598.
59. Overbaugh J, Morris L. 2012. The antibody response against HIV-1. Cold
Spring Harb Perspect Med 2:a007039.
60. Rademeyer C, Moore PL, Taylor N, Martin DP, Choge IA, Gray ES,
Sheppard HW, Gray C, Morris L, Williamson C. 2007. Genetic charac-
teristics of HIV-1 subtype C envelopes inducing cross-neutralizing anti-
bodies. Virology 368:172–181. http://dx.doi.org/10.1016/j.virol.2007.06
.013.
61. Rong R, Bibollet-Ruche F, Mulenga J, Allen S, Blackwell JL, Derdeyn
CA. 2007. Role of V1V2 and other human immunodeficiency virus
type 1 envelope domains in resistance to autologous neutralization
during clade C infection. J Virol 81:1350–1359. http://dx.doi.org/10
.1128/JVI.01839-06.
62. Rong R, Li B, Lynch RM, Haaland RE, Murphy MK, Mulenga J, Allen
SA, Pinter A, Shaw GM, Hunter E, Robinson JE, Gnanakaran S,
Derdeyn CA. 2009. Escape from autologous neutralizing antibodies in
acute/early subtype C HIV-1 infection requires multiple pathways. PLoS
Pathog 5:e1000594. http://dx.doi.org/10.1371/journal.ppat.1000594.
63. Ringe R, Das L, Choudhary I, Sharma D, Paranjape R, Chauhan VS,
Bhattacharya J. 2012. Unique C2V3 sequence in HIV-1 envelope ob-
tained from broadly neutralizing plasma of a slow progressing patient
conferred enhanced virus neutralization. PLoS One 7:e46713. http://dx
.doi.org/10.1371/journal.pone.0046713.
64. Ringe RP, Sanders RW, Yasmeen A, Kim HJ, Lee JH, Cupo A, Korzun
J, Derking R, van Montfort T, Julien JP, Wilson IA, Klasse PJ, Ward
AB, Moore JP. 2013. Cleavage strongly influences whether soluble HIV-1
envelope glycoprotein trimers adopt a native-like conformation. Proc
Natl Acad Sci U S A 110:18256–18261. http://dx.doi.org/10.1073/pnas
.1314351110.
65. Chung NP, Matthews K, Kim HJ, Ketas TJ, Golabek M, de Los Reyes K,
Korzun J, Yasmeen A, Sanders RW, Klasse PJ, Wilson IA, Ward AB,
Marozsan AJ, Moore JP, Cupo A. 2014. Stable 293 T and CHO cell lines
expressing cleaved, stable HIV-1 envelope glycoprotein trimers for struc-
tural and vaccine studies. Retrovirology 11:33. http://dx.doi.org/10.1186
/1742-4690-11-33.
66. Feng Y, McKee K, Tran K, O’Dell S, Schmidt SD, Phogat A, Forsell MN,
Karlsson Hedestam GB, Mascola JR, Wyatt RT. 2012. Biochemically
defined HIV-1 envelope glycoprotein variant immunogens display differ-
ential binding and neutralizing specificities to the CD4-binding site. J Biol
Chem 287:5673–5686. http://dx.doi.org/10.1074/jbc.M111.317776.
67. Boliar S, Das S, Bansal M, Shukla BN, Patil S, Shrivastava T, Samal S,
Goswami S, King CR, Bhattacharya J, Chakrabarti BK. 2015. An effi-
ciently cleaved HIV-1 clade C Env selectively binds to neutralizing anti-
bodies. PLoS One 10:e0122443. http://dx.doi.org/10.1371/journal.pone
.0122443.
68. Ringe R, Thakar M, Bhattacharya J. 2010. Variations in autologous
neutralization and CD4 dependence of b12 resistant HIV-1 clade C env
clones obtained at different time points from antiretroviral naive Indian
patients with recent infection. Retrovirology 7:76. http://dx.doi.org/10
.1186/1742-4690-7-76.
69. Sanders RW, Derking R, Cupo A, Julien JP, Yasmeen A, de Val N, Kim
HJ, Blattner C, de la Pena AT, Korzun J, Golabek M, de Los Reyes K,
Ketas TJ, van Gils MJ, King CR, Wilson IA, Ward AB, Klasse PJ, Moore
JP. 2013. A next-generation cleaved, soluble HIV-1 Env trimer, BG505
SOSIP.664 gp140, expresses multiple epitopes for broadly neutralizing but
not non-neutralizing antibodies. PLoS Pathog 9:e1003618. http://dx.doi
.org/10.1371/journal.ppat.1003618.
70. Tomaras GD, Binley JM, Gray ES, Crooks ET, Osawa K, Moore PL,
Tumba N, Tong T, Shen X, Yates NL, Decker J, Wibmer CK, Gao F,
Alam SM, Easterbrook P, Abdool Karim S, Kamanga G, Crump JA,
Cohen M, Shaw GM, Mascola JR, Haynes BF, Montefiori DC, Morris L.
2011. Polyclonal B cell responses to conserved neutralization epitopes in a
subset of HIV-1-infected individuals. J Virol 85:11502–11519. http://dx
.doi.org/10.1128/JVI.05363-11.
71. Patil S, Choudhary I, Chaudhary NK, Ringe R, Bansal M, Shukla BN,
Boliar S, Chakrabarti BK, Bhattacharya J. 2014. Determinants in V2C2
region of HIV-1 clade C primary envelopes conferred altered neutraliza-
tion susceptibilities to IgG1b12 and PG9 monoclonal antibodies in a con-
text-dependent manner. Virology 462-463:266–272.
72. Teng G, Papavasiliou FN. 2007. Immunoglobulin somatic hypermuta-
tion. Annu Rev Genet 41:107–120. http://dx.doi.org/10.1146/annurev
.genet.41.110306.130340.
73. Gray ES, Moore PL, Bibollet-Ruche F, Li H, Decker JM, Meyers T, Shaw
GM, Morris L. 2008. 4E10-resistant variants in a human immunodefi-
ciency virus type 1 subtype C-infected individual with an anti-membrane-
proximal external region-neutralizing antibody response. J Virol 82:2367–
2375. http://dx.doi.org/10.1128/JVI.02161-07.
74. Trkola A, Purtscher M, Muster T, Ballaun C, Buchacher A, Sullivan N,
Srinivasan K, Sodroski J, Moore JP, Katinger H. 1996. Human mono-
clonal antibody 2G12 defines a distinctive neutralization epitope on the
gp120 glycoprotein of human immunodeficiency virus type 1. J Virol 70:
1100–1108.
75. Guttman M, Cupo A, Julien JP, Sanders RW, Wilson IA, Moore JP, Lee
KK. 2015. Antibody potency relates to the ability to recognize the closed,
pre-fusion form of HIV Env. Nat Commun 6:6144. http://dx.doi.org/10
.1038/ncomms7144.
76. Kong L, Lee JH, Doores KJ, Murin CD, Julien JP, McBride R, Liu Y,
Marozsan A, Cupo A, Klasse PJ, Hoffenberg S, Caulfield M, King CR,
Hua Y, Le KM, Khayat R, Deller MC, Clayton T, Tien H, Feizi T,
Sanders RW, Paulson JC, Moore JP, Stanfield RL, Burton DR, Ward
AB, Wilson IA. 2013. Supersite of immune vulnerability on the glycosy-
lated face of HIV-1 envelope glycoprotein gp120. Nat Struct Mol Biol
20:796–803. http://dx.doi.org/10.1038/nsmb.2594.
77. Sok D, Doores KJ, Briney B, Le KM, Saye-Francisco KL, Ramos A, Kulp
DW, Julien JP, Menis S, Wickramasinghe L, Seaman MS, Schief WR,
Wilson IA, Poignard P, Burton DR. 2014. Promiscuous glycan site
recognition by antibodies to the high-mannose patch of gp120 broadens
neutralization of HIV. Sci Transl Med 6:236ra263.
78. Rolland M, Edlefsen PT, Larsen BB, Tovanabutra S, Sanders-Buell E,
Hertz T, deCamp AC, Carrico C, Menis S, Magaret CA, Ahmed H,
Juraska M, Chen L, Konopa P, Nariya S, Stoddard JN, Wong K, Zhao
H, Deng W, Maust BS, Bose M, Howell S, Bates A, Lazzaro M,
O’Sullivan A, Lei E, Bradfield A, Ibitamuno G, Assawadarachai V,
O’Connell RJ, de Souza MS, Nitayaphan S, Rerks-Ngarm S, Robb ML,
McLellan JS, Georgiev I, Kwong PD, Carlson JM, Michael NL, Schief
WR, Gilbert PB, Mullins JI, Kim JH. 2012. Increased HIV-1 vaccine
efficacy against viruses with genetic signatures in Env V2. Nature 490:417–
420. http://dx.doi.org/10.1038/nature11519.
79. Chakrabarti BK, Feng Y, Sharma SK, McKee K, Karlsson Hedestam GB,
Labranche CC, Montefiori DC, Mascola JR, Wyatt RT. 2013. Robust
neutralizing antibodies elicited by HIV-1 JRFL envelope glycoprotein
trimers in nonhuman primates. J Virol 87:13239–13251. http://dx.doi.org
/10.1128/JVI.01247-13.
80. Burton DR, Mascola JR. 2015. Antibody responses to envelope glycopro-
teins in HIV-1 infection. Nat Immunol 16:571–576. http://dx.doi.org/10
.1038/ni.3158.
81. Pancera M, Wyatt R. 2005. Selective recognition of oligomeric HIV-1
primary isolate envelope glycoproteins by potently neutralizing ligands
requires efficient precursor cleavage. Virology 332:145–156. http://dx.doi
.org/10.1016/j.virol.2004.10.042.
82. Sok D, van Gils MJ, Pauthner M, Julien JP, Saye-Francisco KL, Hsueh
J, Briney B, Lee JH, Le KM, Lee PS, Hua Y, Seaman MS, Moore JP,
Ward AB, Wilson IA, Sanders RW, Burton DR. 2014. Recombinant HIV
envelope trimer selects for quaternary-dependent antibodies targeting the
trimer apex. Proc Natl Acad Sci U S A 111:17624–17629. http://dx.doi.org
/10.1073/pnas.1415789111.
HIV-1 Clade C Plasma Confers Cross-Clade Neutralization
April 2016 Volume 90 Number 7 jvi.asm.org 3457Journal of Virology
onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom

More Related Content

What's hot

SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...
SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...
SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...Guy Boulianne
 
Dr Susan E Caldwell Publication Samples
Dr Susan E Caldwell Publication SamplesDr Susan E Caldwell Publication Samples
Dr Susan E Caldwell Publication SamplesSusan E Caldwell, PhD
 
Singapore grouper iridovirus induced parapoptosis-like death in host cells vi...
Singapore grouper iridovirus induced parapoptosis-like death in host cells vi...Singapore grouper iridovirus induced parapoptosis-like death in host cells vi...
Singapore grouper iridovirus induced parapoptosis-like death in host cells vi...mgray11
 
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...Dr. Md. Ehsanul Haque
 
Aeras schofield 21112013
Aeras schofield 21112013Aeras schofield 21112013
Aeras schofield 21112013AerasGlobalTB
 
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...ILRI
 
Karn lessons-hiv-latency-2014-10-06
Karn lessons-hiv-latency-2014-10-06Karn lessons-hiv-latency-2014-10-06
Karn lessons-hiv-latency-2014-10-06Kimberly Schafer
 
New developments in vaccines against African swine fever
New developments in vaccines against African swine feverNew developments in vaccines against African swine fever
New developments in vaccines against African swine feverILRI
 
Cocciodioidomycosis by Davinder Singh
Cocciodioidomycosis by Davinder SinghCocciodioidomycosis by Davinder Singh
Cocciodioidomycosis by Davinder SinghDavinder Sidhu
 
Candidemia in HIV-positive patients in Dschang District Hospital (West Region...
Candidemia in HIV-positive patients in Dschang District Hospital (West Region...Candidemia in HIV-positive patients in Dschang District Hospital (West Region...
Candidemia in HIV-positive patients in Dschang District Hospital (West Region...Claude Nangwat
 
Sigma xi presentation final1
Sigma xi presentation   final1Sigma xi presentation   final1
Sigma xi presentation final1mukta936
 
NK cells in HBV and HCV
NK cells in HBV and HCVNK cells in HBV and HCV
NK cells in HBV and HCVMario Mondelli
 
CORONAVIRUS AKA SARS PATENT
CORONAVIRUS AKA SARS PATENT CORONAVIRUS AKA SARS PATENT
CORONAVIRUS AKA SARS PATENT ICJ-ICC
 

What's hot (19)

SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...
SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...
SARS–CoV–2 Spike Impairs DNA Damage Repair and Inhibits V(D)J Recombination I...
 
Mismatch Amplification Mutation Assay (MAMA) PCR Reveals Altered El Tor Vibri...
Mismatch Amplification Mutation Assay (MAMA) PCR Reveals Altered El Tor Vibri...Mismatch Amplification Mutation Assay (MAMA) PCR Reveals Altered El Tor Vibri...
Mismatch Amplification Mutation Assay (MAMA) PCR Reveals Altered El Tor Vibri...
 
Dr Susan E Caldwell Publication Samples
Dr Susan E Caldwell Publication SamplesDr Susan E Caldwell Publication Samples
Dr Susan E Caldwell Publication Samples
 
Coronavirus disease
Coronavirus disease Coronavirus disease
Coronavirus disease
 
Singapore grouper iridovirus induced parapoptosis-like death in host cells vi...
Singapore grouper iridovirus induced parapoptosis-like death in host cells vi...Singapore grouper iridovirus induced parapoptosis-like death in host cells vi...
Singapore grouper iridovirus induced parapoptosis-like death in host cells vi...
 
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
Sensitivity and Specificity of an In-house Sandwich ELISA Kit for Newcastle D...
 
1584-09
1584-091584-09
1584-09
 
Aeras schofield 21112013
Aeras schofield 21112013Aeras schofield 21112013
Aeras schofield 21112013
 
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...
Development of diagnostic tools to reduce antimicrobial (mis)use: Novel ident...
 
Karn lessons-hiv-latency-2014-10-06
Karn lessons-hiv-latency-2014-10-06Karn lessons-hiv-latency-2014-10-06
Karn lessons-hiv-latency-2014-10-06
 
Science
ScienceScience
Science
 
Publication 3 - 3rd Author
Publication 3 - 3rd AuthorPublication 3 - 3rd Author
Publication 3 - 3rd Author
 
New developments in vaccines against African swine fever
New developments in vaccines against African swine feverNew developments in vaccines against African swine fever
New developments in vaccines against African swine fever
 
Cocciodioidomycosis by Davinder Singh
Cocciodioidomycosis by Davinder SinghCocciodioidomycosis by Davinder Singh
Cocciodioidomycosis by Davinder Singh
 
Candidemia in HIV-positive patients in Dschang District Hospital (West Region...
Candidemia in HIV-positive patients in Dschang District Hospital (West Region...Candidemia in HIV-positive patients in Dschang District Hospital (West Region...
Candidemia in HIV-positive patients in Dschang District Hospital (West Region...
 
Sigma xi presentation final1
Sigma xi presentation   final1Sigma xi presentation   final1
Sigma xi presentation final1
 
NK cells in HBV and HCV
NK cells in HBV and HCVNK cells in HBV and HCV
NK cells in HBV and HCV
 
Chronic granulomatous disease
Chronic granulomatous diseaseChronic granulomatous disease
Chronic granulomatous disease
 
CORONAVIRUS AKA SARS PATENT
CORONAVIRUS AKA SARS PATENT CORONAVIRUS AKA SARS PATENT
CORONAVIRUS AKA SARS PATENT
 

Viewers also liked

Jack Baker CV-2
Jack Baker CV-2Jack Baker CV-2
Jack Baker CV-2Jack Baker
 
CV-Terry_Potter with Admin
CV-Terry_Potter with AdminCV-Terry_Potter with Admin
CV-Terry_Potter with AdminTerry Potter
 
ThiagoDetanico Resume 13May2016
ThiagoDetanico Resume 13May2016ThiagoDetanico Resume 13May2016
ThiagoDetanico Resume 13May2016Thiago Detanico
 
Svoboda Ben - Resume 16-0411
Svoboda Ben - Resume 16-0411Svoboda Ben - Resume 16-0411
Svoboda Ben - Resume 16-0411Benjamin Svoboda
 
P_Flanagan_Resume project_managment 23JUN2016
P_Flanagan_Resume project_managment 23JUN2016P_Flanagan_Resume project_managment 23JUN2016
P_Flanagan_Resume project_managment 23JUN2016Patrick Flanagan
 

Viewers also liked (7)

Jack Baker CV-2
Jack Baker CV-2Jack Baker CV-2
Jack Baker CV-2
 
Michael Valentine CV
Michael Valentine CVMichael Valentine CV
Michael Valentine CV
 
CV-Terry_Potter with Admin
CV-Terry_Potter with AdminCV-Terry_Potter with Admin
CV-Terry_Potter with Admin
 
ThiagoDetanico Resume 13May2016
ThiagoDetanico Resume 13May2016ThiagoDetanico Resume 13May2016
ThiagoDetanico Resume 13May2016
 
Svoboda Ben - Resume 16-0411
Svoboda Ben - Resume 16-0411Svoboda Ben - Resume 16-0411
Svoboda Ben - Resume 16-0411
 
P_Flanagan_Resume project_managment 23JUN2016
P_Flanagan_Resume project_managment 23JUN2016P_Flanagan_Resume project_managment 23JUN2016
P_Flanagan_Resume project_managment 23JUN2016
 
Dr. Khaled CV
Dr. Khaled CVDr. Khaled CV
Dr. Khaled CV
 

Similar to Broadly neutralizing antibodies from an HIV-1 clade C elite neutralizer target novel trimeric Env epitopes

2008Phylogenetic Analysis and Isolation of CDV
2008Phylogenetic Analysis and Isolation of CDV2008Phylogenetic Analysis and Isolation of CDV
2008Phylogenetic Analysis and Isolation of CDVDavid Chung -Tiang Liang
 
Deshpande S et al RETROVIROLOGY_b 2016
Deshpande S et al RETROVIROLOGY_b 2016Deshpande S et al RETROVIROLOGY_b 2016
Deshpande S et al RETROVIROLOGY_b 2016SHILPA PATIL
 
Association of human leukocyte antigen class II allele and haplotypes Trans R...
Association of human leukocyte antigen class II allele and haplotypes Trans R...Association of human leukocyte antigen class II allele and haplotypes Trans R...
Association of human leukocyte antigen class II allele and haplotypes Trans R...Subrat Thanapati
 
Virology Journal Club
Virology Journal ClubVirology Journal Club
Virology Journal ClubPrenesh
 
African Swine Fever (ASF) virus genomics and diagnostics
African Swine Fever (ASF) virus genomics and diagnosticsAfrican Swine Fever (ASF) virus genomics and diagnostics
African Swine Fever (ASF) virus genomics and diagnosticsILRI
 
Deshpande et al. Retrovirology (2016)
Deshpande et al. Retrovirology (2016)Deshpande et al. Retrovirology (2016)
Deshpande et al. Retrovirology (2016)SHILPA PATIL
 
ex-lab field report.docx
ex-lab field report.docxex-lab field report.docx
ex-lab field report.docxMesele Tilahun
 
A simple and rapid dna extraction method from FINA nd qPCR
A simple and rapid dna extraction method from FINA nd qPCRA simple and rapid dna extraction method from FINA nd qPCR
A simple and rapid dna extraction method from FINA nd qPCRManish Thakur
 
Clinical Application of Stem Cells in HIV
Clinical Application of Stem Cells in HIVClinical Application of Stem Cells in HIV
Clinical Application of Stem Cells in HIVahmedicine
 
JPT_Poster_EPS_2016_final
JPT_Poster_EPS_2016_finalJPT_Poster_EPS_2016_final
JPT_Poster_EPS_2016_finalPaul Von Hoegen
 
Methodology for development of defined deletion mutant vaccine for salmonellosis
Methodology for development of defined deletion mutant vaccine for salmonellosisMethodology for development of defined deletion mutant vaccine for salmonellosis
Methodology for development of defined deletion mutant vaccine for salmonellosisBhoj Raj Singh
 
ProImmune Antigen Characterization Summit Sanja Selak
ProImmune Antigen Characterization Summit Sanja SelakProImmune Antigen Characterization Summit Sanja Selak
ProImmune Antigen Characterization Summit Sanja Selakamandacturner
 
Central Nervous System Fungal Infection
 Central Nervous System Fungal Infection Central Nervous System Fungal Infection
Central Nervous System Fungal InfectionDR.SITI HAWA HAMZAH
 

Similar to Broadly neutralizing antibodies from an HIV-1 clade C elite neutralizer target novel trimeric Env epitopes (20)

Indo-SA HIVR4P
Indo-SA HIVR4PIndo-SA HIVR4P
Indo-SA HIVR4P
 
2008Phylogenetic Analysis and Isolation of CDV
2008Phylogenetic Analysis and Isolation of CDV2008Phylogenetic Analysis and Isolation of CDV
2008Phylogenetic Analysis and Isolation of CDV
 
Deshpande S et al RETROVIROLOGY_b 2016
Deshpande S et al RETROVIROLOGY_b 2016Deshpande S et al RETROVIROLOGY_b 2016
Deshpande S et al RETROVIROLOGY_b 2016
 
Zoonotic Viral Disease.pptx
Zoonotic Viral Disease.pptxZoonotic Viral Disease.pptx
Zoonotic Viral Disease.pptx
 
Sales 2
Sales 2Sales 2
Sales 2
 
M Ezzat El Zowalaty EUSOM
M Ezzat El Zowalaty EUSOMM Ezzat El Zowalaty EUSOM
M Ezzat El Zowalaty EUSOM
 
Association of human leukocyte antigen class II allele and haplotypes Trans R...
Association of human leukocyte antigen class II allele and haplotypes Trans R...Association of human leukocyte antigen class II allele and haplotypes Trans R...
Association of human leukocyte antigen class II allele and haplotypes Trans R...
 
Virology Journal Club
Virology Journal ClubVirology Journal Club
Virology Journal Club
 
African Swine Fever (ASF) virus genomics and diagnostics
African Swine Fever (ASF) virus genomics and diagnosticsAfrican Swine Fever (ASF) virus genomics and diagnostics
African Swine Fever (ASF) virus genomics and diagnostics
 
Deshpande et al. Retrovirology (2016)
Deshpande et al. Retrovirology (2016)Deshpande et al. Retrovirology (2016)
Deshpande et al. Retrovirology (2016)
 
Deshpande et al.,Retrovirology 2016
Deshpande et al.,Retrovirology 2016Deshpande et al.,Retrovirology 2016
Deshpande et al.,Retrovirology 2016
 
ex-lab field report.docx
ex-lab field report.docxex-lab field report.docx
ex-lab field report.docx
 
A simple and rapid dna extraction method from FINA nd qPCR
A simple and rapid dna extraction method from FINA nd qPCRA simple and rapid dna extraction method from FINA nd qPCR
A simple and rapid dna extraction method from FINA nd qPCR
 
Clinical Application of Stem Cells in HIV
Clinical Application of Stem Cells in HIVClinical Application of Stem Cells in HIV
Clinical Application of Stem Cells in HIV
 
Clonal b cells in patients with hepatitis c virus–associated mixed
Clonal b cells in patients with hepatitis c virus–associated mixedClonal b cells in patients with hepatitis c virus–associated mixed
Clonal b cells in patients with hepatitis c virus–associated mixed
 
JPT_Poster_EPS_2016_final
JPT_Poster_EPS_2016_finalJPT_Poster_EPS_2016_final
JPT_Poster_EPS_2016_final
 
Methodology for development of defined deletion mutant vaccine for salmonellosis
Methodology for development of defined deletion mutant vaccine for salmonellosisMethodology for development of defined deletion mutant vaccine for salmonellosis
Methodology for development of defined deletion mutant vaccine for salmonellosis
 
ProImmune Antigen Characterization Summit Sanja Selak
ProImmune Antigen Characterization Summit Sanja SelakProImmune Antigen Characterization Summit Sanja Selak
ProImmune Antigen Characterization Summit Sanja Selak
 
Minh's Poster
Minh's PosterMinh's Poster
Minh's Poster
 
Central Nervous System Fungal Infection
 Central Nervous System Fungal Infection Central Nervous System Fungal Infection
Central Nervous System Fungal Infection
 

Broadly neutralizing antibodies from an HIV-1 clade C elite neutralizer target novel trimeric Env epitopes

  • 1. Conformational Epitope-Specific Broadly Neutralizing Plasma Antibodies Obtained from an HIV-1 Clade C-Infected Elite Neutralizer Mediate Autologous Virus Escape through Mutations in the V1 Loop Shilpa Patil,a Rajesh Kumar,a Suprit Deshpande,a Sweety Samal,a Tripti Shrivastava,a Saikat Boliar,a Manish Bansal,a Nakul Kumar Chaudhary,a Aylur K. Srikrishnan,b Kailapuri G. Murugavel,b Suniti Solomon,b Melissa Simek,c Wayne C. Koff,c Rajat Goyal,c Bimal K. Chakrabarti,a,c Jayanta Bhattacharyaa,c HIV Vaccine Translational Research Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, Indiaa ; Y. R. Gaitonde Research and Care Center, Chennai, Indiab ; International AIDS Vaccine Initiative, New York, New York, USAc ABSTRACT Broadly neutralizing antibodies isolated from infected patients who are elite neutralizers have identified targets on HIV-1 enve- lope (Env) glycoprotein that are vulnerable to antibody neutralization; however, it is not known whether infection established by the majority of the circulating clade C strains in Indian patients elicit neutralizing antibody responses against any of the known targets. In the present study, we examined the specificity of a broad and potent cross-neutralizing plasma obtained from an In- dian elite neutralizer infected with HIV-1 clade C. This plasma neutralized 53/57 (93%) HIV pseudoviruses prepared with Env from distinct HIV clades of different geographical origins. Mapping studies using gp120 core protein, single-residue knockout mutants, and chimeric viruses revealed that G37080 broadly cross-neutralizing (BCN) plasma lacks specificities to the CD4 bind- ing site, gp41 membrane-proximal external region, N160 and N332 glycans, and R166 and K169 in the V1-V3 region and are known predominant targets for BCN antibodies. Depletion of G37080 plasma with soluble trimeric BG505-SOSIP.664 Env (but with neither monomeric gp120 nor clade C membrane-proximal external region peptides) resulted in significant reduction of virus neutralization, suggesting that G37080 BCN antibodies mainly target epitopes on cleaved trimeric Env. Further examina- tion of autologous circulating Envs revealed the association of mutation of residues in the V1 loop that contributed to neutral- ization resistance. In summary, we report the identification of plasma antibodies from a clade C-infected elite neutralizer that mediate neutralization breadth via epitopes on trimeric gp120 not yet reported and confer autologous neutralization escape via mutation of residues in the V1 loop. IMPORTANCE A preventive vaccine to protect against HIV-1 is urgently needed. HIV-1 envelope glycoproteins are targets of neutralizing anti- bodies and represent a key component for immunogen design. The mapping of epitopes on viral envelopes vulnerable to im- mune evasion will aid in defining targets of vaccine immunogens. We identified novel conformational epitopes on the viral enve- lope targeted by broadly cross-neutralizing antibodies elicited in natural infection in an elite neutralizer infected with HIV-1 clade C. Our data extend our knowledge on neutralizing epitopes associated with virus escape and potentially contribute to im- munogen design and antibody-based prophylactic therapy. Broadly neutralizing antibodies (BNAbs) target trimeric enve- lope glycoprotein (Env) spikes of human immunodeficiency virus type 1 (HIV-1). Characterization of the BNAbs has provided key clues toward the design and development of both prophylactic and therapeutic vaccines (1–6). A small proportion of individuals chronically infected with HIV-1 develop BNAbs (7–14), and the isolation of several broad and potent neutralizing monoclonal an- tibodies (MAbs) from such individuals with distinct molecular specificities to viral envelope protein has been reported (15–23). The cross-neutralizing serum antibodies obtained from such in- dividuals (also referred to as elite neutralizers), which have con- siderable breadth, target epitopes on structurally conserved re- gions of Env such as the CD4 binding site (CD4bs) (22, 24–26), V1V2, including glycan moieties (19, 20, 27, 28), the gp120-gp41 interface (18, 29), and the membrane-proximal external regions (MPER) (16, 30–32). Several studies have indicated that the vari- able regions within HIV-1 gp120 contain epitopes targeted by autologous antibodies as well as BNAbs (33–40). Recently the V1V2 region has been linked to the development of broadly cross- neutralizing (BCN) antibodies (35, 41), and the residues between 160 and 172 (notably R166S/K or K169A) in V1V2 have been demonstrated to be associated with virus escape from autologous Received 9 December 2015 Accepted 7 January 2016 Accepted manuscript posted online 13 January 2016 Citation Patil S, Kumar R, Deshpande S, Samal S, Shrivastava T, Boliar S, Bansal M, Chaudhary NK, Srikrishnan AK, Murugavel KG, Solomon S, Simek M, Koff WC, Goyal R, Chakrabarti BK, Bhattacharya J. 2016. Conformational epitope-specific broadly neutralizing plasma antibodies obtained from an HIV-1 clade C-infected elite neutralizer mediate autologous virus escape through mutations in the V1 loop. J Virol 90:3446–3457. doi:10.1128/JVI.03090-15. Editor: W. I. Sundquist, University of Utah Address correspondence to Jayanta Bhattacharya, JBhattacharya@iavi.org. S.P., R.K., and S.D. contributed equally to this work. Copyright © 2016, American Society for Microbiology. All Rights Reserved. crossmark 3446 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
  • 2. antibody response (35). Recent studies have further indicated that BCNAb development in vivo is associated with antibody affinity maturation and coevolution of virus, resulting in a considerable degree of somatic hypermutations (19, 20, 23, 26, 35, 42–50). Such information is crucial for the design and development of suitable Env-based immunogens capable of eliciting broad and potent cross-neutralizing antibodies through vaccination. While a number of studies on the molecular specificities of broadly neutralizing antibodies obtained from African clade C-in- fected individuals have been reported (9, 37, 51–62), knowledge on immune evasion in Indian clade C-infected elite neutralizers is very limited (63). In the present study, we examined plasma samples obtained from two hundred asymptomatic and antiretroviral therapy (ART) naive Indian HIV-infected donors and identified plasma with cross-neutralizing antibodies. The molecular specificities of plasma antibodies obtained from an HIV-1 clade C-infected elite neutralizer was characterized in detail that displayed exceptional neutralization breadth across clades of different geographical or- igins. Interestingly, we found that neutralization breadth was as- sociated with the presence of unique epitopes on the trimeric gp120. MATERIALS AND METHODS Ethics statement. The blood samples were collected under the IAVI Pro- tocol G study from slow-progressing ART naive HIV-1-positive donors from Nellore District of the state of Andhra Pradesh, southern India, by trained clinicians at the YRG Care Hospital following approval and clear- ance from the Institutional Review Board (IRB) and the Ethics Commit- tee. The serum and plasma samples collected were shipped to the HIV Vaccine Translational Research Laboratory, Translational Health Science and Technology Institute, for further assessment and research on the neu- tralizing antibody response. Plasmids, viruses, antibodies, proteins, and cells. Plasmids encoding HIV-1 envelopes representing distinct clades are shown in Table 1. Mono- clonal antibodies used in the study and TZM-bl cells were procured from the NIH AIDS Research and Reagents Reference program and from the IAVI Neutralizing Antibody Consortium (NAC). 293T cells were pur- chased from the American Type Culture Collection (ATCC). Plasmid DNA encoding BG505-SOSIP.664-D7324, its purified cleaved trimeric protein (64), and pcDNA5-FRT BG505 furin A (65) were kindly provided by John Moore, Weill Cornell Medical College, New York. Purified gp120 TripleMut core protein (66) was obtained from Richard Wyatt, The Scripps Research Institute, through the NAC. HIV-2 7312A and its chi- meric constructs were provided by Lynn Morris, NICD, Johannesburg, South Africa. Purification of monomeric and trimeric Env proteins. Codon-opti- mized gp120 plasmid encoding clade C 4-2.J41 (67, 68) gp120 was cloned in pcDNA 3.1/V5-His-TOPO vector and transfected into 293T cells using polyethyleneimine (PEI). Supernatants containing soluble gp120 were fil- tered through 0.45-␮m-pore-size filters and subsequently purified using nickel-nitrilotriacetic acid (Ni-NTA) agarose matrix (Qiagen Inc.) by elution with phosphate-buffered saline (PBS) containing 300 mM imidazole (pH 8.0). The purified monomeric gp120 protein was extensively dialyzed with PBS (pH 7.4), concentrated using Amicon ultracentrifugal filters (Millipore Inc.) with a 30-kDa cutoff, and stored at Ϫ80°C until further use. The trimeric BG505-SOSIP.664 protein was purified using 293F cells essentially as described by Sanders et al. (69). Briefly, the 293F cells were transfected with plasmid DNA encoding both BG505-SO- SIP.664 gp140 envelope and furin (65). Supernatant containing solu- ble BG505-SOSIP.664 gp140 was harvested 72 to 96 h posttransfection, filtered, and passed through a lectin agarose column obtained from Galanthus nivalis (Sigma Inc.). The nonspecifically bound proteins then were washed in PBS (pH 7.4) supplemented with 0.5 M NaCl. The bound proteins then were eluted using 0.5 M methyl ␣-D-mannopyranoside, extensively dialyzed with 1ϫ PBS, and concentrated. BG505-SOSIP.664 was further purified by Sephadex G-200 size exclusion chromatography (AKTA; GE). Trimeric protein fractions were collected and pooled, their quality was assessed by running in blue native polyacrylamide gel electro- TABLE 1 Neutralization breadth of Protocol G G37080 plasma samples collected at two different time points tested against panel of 57 Env- pseudotyped viruses HIV-1 Clade C Plasma Confers Cross-Clade Neutralization April 2016 Volume 90 Number 7 jvi.asm.org 3447Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
  • 3. phoresis (BN-PAGE), and they were favorably assessed for their ability to bind to only neutralizing and not to nonneutralizing and MPER-directed monoclonal antibodies as described elsewhere (64) by enzyme-linked im- munosorbent assay (ELISA). Depletion of plasma antibodies by monomeric gp120 and trimeric gp140 Env proteins. Purified soluble monomeric 4-2.J41 gp120 and tri- meric BG505 SOSIP.664 proteins, in addition to the MPER peptide (C1C; encoding clade C sequence) (70), were used for the depletion of plasma antibodies where purified proteins were covalently coupled to tosylacti- vated MyOne Dynabeads (Life Technologies Inc.) according to the man- ufacturer’s protocol. Briefly, 30 mg of beads was coupled with 1 mg of both monomeric and trimeric Env proteins in coupling buffer [0.1 M NaBO4, 1 M (NH4)2SO4; pH 9.4] overnight at 37°C for 16 to 24 h. Proteins bound to magnetic beads were separated from unbound proteins using a DynaMag 15 magnet (Life Technologies, Inc.). Beads bound to Env pro- teins next were incubated with blocking buffer (PBS [pH 7.4], 0.1% bo- vine serum albumin [BSA; Sigma], and 0.05% Tween 20) at 37°C to block the unbound sites. The antigenic integrity of both 4-2.J41 monomeric gp120 and BG505-SOSIP.664 bound to the beads was assessed for their ability to bind VRC01 and 4E10 MAbs (for monomeric gp120) and PGT121, F105, and 4E10 MAbs (for BG505-SOSIP.664) by flow cytom- etry (FACSCanto; Becton and Dickinson, Inc.). For depletion studies, G37080 plasma was diluted to 1:50 in Dulbec- co’s modified Eagle’s medium (DMEM) containing 10% fetal bovine se- rum (FBS), and 500 ␮l of diluted plasma was incubated with 20 ␮l of beads at room temperature for 45 min. Unbound plasma antibodies were sepa- rated from those that were bound to protein-coated beads using a DynaMag 15 magnet as described above. This step was repeated 4 to 5 times for the depletion of plasma antibodies by monomeric gp120 and 10 to 12 times in the case of BG505-SOSIP.664-coated beads. As a negative control, G37080 plasma antibodies were depleted with uncoated beads in parallel. In addition to ELISA, the percent depletion of G37080 plasma antibodies was assessed by examining the sequential decrease in binding of protein-coated beads with depleted plasma antibodies by fluorescence- activated cell sorting (FACS). PGT121 MAb was taken as a positive con- trol for checking depletion by BG505-SOSIP.664 trimeric Env. gp120 and gp140 ELISA. For gp120 ELISA, a high-binding polysty- rene microtiter plate (Nunc, Inc.) was coated with 100 ␮l of monomeric 4-2.J41 gp120 (1 ␮g/ml) in binding buffer containing 0.1 M NaHCO3 (pH 8.6) and incubated overnight at 4°C. The gp120-plate was washed once with 1ϫ PBS (pH 7.4) and blocked with 5% nonfat milk for 90 min at 37°C. The plate then was washed three times with 1ϫ PBS followed by the addition of 100 ␮l of MAbs, as well as the depleted and undepleted plasma antibodies at different dilutions, and incubated for 1 h at room tempera- ture. The wells of the ELISA plate were washed four times with PBS con- taining 0.1% Tween 20 (PBST), followed by the addition of 100 ␮l of 1:3,000-diluted horseradish peroxidase (HRP)-conjugated anti-human IgG (Jackson ImmunoResearch, Inc.), and further incubated for 45 min at room temperature. Unbound conjugates were removed by washing with PBST, and color was developed by the addition of 100 ␮l of 3,3=,5,5=- tetramethylbenzidine (TMB) (Life Technologies, Inc.) substrate. Absor- bance was measured at 450 nm in a spectrophotometer. Binding of antibodies to BG505-SOSIP.664-D7324 trimeric protein was assessed essentially as described by Sanders et al. (69) in a sandwich ELISA. Briefly, a high-binding microtiter plate (Nunc, Inc.) first was coated with D7324 antibody at 10 ␮g/ml (Aalto Bio Reagents, Dublin, Ireland) followed by blocking extra unbound sites with 5% nonfat milk for 90 min at 37°C. One hundred microliters of BG505.664-D7324 trim- eric protein (300 ng/ml) then was added and incubated for 45 min at room temperature. The extent of binding of G37080 plasma antibodies com- pared to that of known neutralizing monoclonal antibodies was assessed by the addition of primary and HRP-conjugated secondary anti-human antibody as described above. Neutralization assay. Neutralization assays were carried out using TZM-bl cells as described before (68). Briefly, Env-pseudotyped viruses were incubated with various dilutions of depleted plasma antibodies and incubated for an hour at 37°C in a CO2 incubator under humidified con- ditions, and subsequently 1 ϫ 104 TZM-bl cells were added to the mixture in the presence of 25 ␮g/ml DEAE-dextran (Sigma, Inc.). The plates were further incubated for 48 h, and the degree of virus neutralization was assessed by measuring relative luminescence units (RLU) in a luminom- eter (Victor X2; PerkinElmer Inc.). Amplification, cloning, and mutagenesis of autologous HIV-1 env genes. Autologous complete env genes were obtained from G37080 plasma as described previously, with slight modifications (68). Briefly, viral RNA was extracted using a high-pure viral RNA kit (Roche Inc.) by following manufacturer’s protocol, and cDNA was prepared by reverse transcription-PCR (RT-PCR) using a Superscript III first-strand synthesis kit (Invitrogen Inc.). rev-gp160 env genes were amplified using a Phusion high-fidelity DNA polymerase (New England BioLabs Inc.). The gp160 amplicons were purified and ligated into pcDNA 3.1/V5-His-TOPO (In- vitrogen Inc.) vector. Chimeric Env proteins were prepared by overlap- ping PCR, and point substitutions were made with a QuikChange II kit (Agilent Technologies Inc.) by following the manufacturer’s protocol and as described previously (71). Preparation of envelope-pseudotyped viruses. Pseudotyped viruses were prepared by the cotransfection of envelope-expressing plasmid with an env-deleted HIV-1 backbone plasmid (pSG3⌬Env) into 293T cells in 6-well tissue culture plates using a FuGENE6 transfection kit (Promega Inc.). Cell supernatants containing pseudotyped viruses were harvested 48 h posttransfection and then stored at Ϫ80°C until further use. The infec- tivity assays were done in TZM-bl cells (1 ϫ 105 cells/ml) containing DEAE-dextran (25 ␮g/ml) in 96-well microtiter plates, and the infectivity titers were determined by measuring the luciferase activity using Britelite luciferase substrate (PerkinElmer Inc.) with a Victor X2 luminometer (PerkinElmer Inc.). RESULTS Identification of an elite neutralizer with HIV-1 clade C infec- tion whose plasma showed exceptional neutralization breadth. The present study, under IAVI Protocol G, was designed (i) to screen and identify plasma antibodies obtained from Indian do- nors chronically infected with HIV-1 clade C, with substantial breadthtowardneutralizingcross-cladeHIV-1primaryvariants,and (ii) to elucidate the molecular specificities associated with neutraliza- tionbreadth.Ourhypothesiswasthatgiventhegeneticdistinctnessof clade C viruses of Indian and non-Indian origin, as well the likely differences in host genetics between populations and their ancestral origins associated with the modulation of humoral immune re- sponses, the specificities of antibodies developed in vivo associated with neutralization breadth and potency would be different. Through screening of two hundred plasma samples obtained from chronically infected ART naive Indian patients against a panel of 57 pseudoviruses containing Envs of distinct clades and geographical origins (Fig. 1A), we identified one donor (G37080) whose plasma showed exceptional neutralization breadth. Donor G37080 serum neutralized Ͼ90% of the 57 different pseudovi- ruses tested, with a median 50% inhibitory dose (ID50) value of 533.03 (Table 1 and Fig. 1B). Follow-up plasma sample from this donor (G37080) subse- quently was obtained after 8 months to assess whether the neu- tralization breadth and potencies, along with their molecular specificities, were retained and/or improved, as we expected that during the course of disease, the breadth and potency of neutral- izing antibodies broadens through somatic hypermutations (72) and/or clonal selection processes. As shown in Fig. 1B and Table 1, follow-up plasma antibodies of donor G37080 (referred to as the visit 2 samples) were found to exhibit neutralization breadth com- Patil et al. 3448 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
  • 4. parable to that of visit 1 plasma. Overall, G37080 BCN plasma was found to potently neutralize pseudoviruses containing Indian clade C Env with a neutralization score of 2.5 (13). Furthermore, the neutralization sensitivity of Env-pseudotyped viruses was found to be correlated with the serum IgG (data not shown), sug- gesting that the broad neutralization was associated with IgG- specific responses. Taken together, our data indicate that a strong humoral immune response to HIV-1 was mounted in donor G37080 and was maintained over time. Evidence that G37080 BCN plasma antibodies do not target epitopes in CD4bs, MPER, and known glycan and nonglycan residues in variable domains of Env. We first examined whether the G37080 BCN plasma contains antibodies directed to the CD4bs on Env. Plasma samples obtained from both visits were pretreated with 25 ␮g/ml of TripleMut core protein (66), which was a concentration that we found to inhibit the neutralization of 25711-2.4 pseudovirus by VRC01 MAb by Ͼ95%. Pretreated plasma subsequently was used to neutralize pseudovirus 25711- 2.4 Env, and as shown in Fig. 2, no perturbation of G37080 neu- tralizing activity was observed against pseudovirus 25711-2.4. A similar observation was made when these plasma antibodies were pretreated with RSC3 core protein (22). In addition, the G37080 BCN plasma antibodies were found to efficiently neutralize IgG1b12- and VRC01-resistant viruses (data not shown). Our data indicated that the G37080 BCN plasma antibodies do not contain CD4bs-directed neutralizing antibodies. To elucidate whether the BCN plasma antibodies are directed to MPER in gp41, we used HIV-2/HIV-1 chimeric viruses (73) that expressed minimal residues of HIV-1 MPER containing epitopes required for MPER-directed MAbs, such as 2F5, 4E10, Z13e, and 10E8. As shown in Table 2, the G37080 BCN plasma from both visits was found to show modest neutralization of HIV-2 expressing HIV-1 clade C MPER (7312-C1C), with ID50 values of 306.42 and 371.02, respectively. We also found that the depletion of G37080 plasma with a clade C MPER peptide (C1C) completely abolished the sensitivity of 7312A-C1C virus to FIG1 (A) Genetic divergence of amino acid sequences of 57 HIV-1 Env (gp160) pseudoviruses used to assess neutralization breadth and potency of G37080 BCN plasma. The maximum likelihood bootstrapped consensus phylogenetic tree was constructed using the Jones-Taylor-Thornton (JTT) substitution model with 50 bootstrapped replicates in Mega 5.2. Bootstrapped values are shown at the nodes of each branch. Hollow circles represent envelopes (16055-2.3 and 92TH021) resistant to neutralization by G37080 BCN plasma. (B) Neutralization breadth of the G37080 BCN plasma obtained at visit 1 and visit 2 were assessed against pseudotyped viruses expressing HIV-1 Env representing different clades and origins. Neutralization titers (median ID50 values) were obtained by titrating Env-pseudotyped viruses against G37080 plasma samples. Values at the top of each bar graph indicate the number of viruses belonging to each clade/origin tested. FIG 2 Assessing dependence of G37080 BCN antibodies to CD4 binding site (CD4bs) region of HIV-1 Env. G37080 BCN plasma samples and VRC01 MAb (concentrations that neutralized 25711-2.4 by Ͼ80%) preincubated with different concentrations, as indicated, with TripleMut core (A) and RSC3 (B) proteins were examined for their ability to neutralize 25711-2.4 Env pseudotyped virus in a TZM-bl cell neutralization assay. Note that while VRC01 preabsorbed with both TripleMut and RSC3 proteins showed inhibited neutralization of 25711-2.4 in a dose-dependent manner, no such effect was observed with G37080 BCN plasma, indicating the absence of CD4bs-directed neutralizing antibodies. HIV-1 Clade C Plasma Confers Cross-Clade Neutralization April 2016 Volume 90 Number 7 jvi.asm.org 3449Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
  • 5. G37080 plasma (Table 3). Our data suggest that although the G37080 BCN plasma neutralized 7312-C1C, the presence of MPER-directed antibodies was not associated with neutralization breadth. We next investigated whether the plasma antibodies of the do- nor G37080 target residues in variable loops, particularly in V1V2 and V3 regions, which have been shown in several studies to be epitopes targeted by BCN antibodies on HIV-1 Env. We first tested the extent of neutralization by G37080 BCN plasma anti- bodies of Env-pseudotyped viruses lacking glycans at positions 160 (N160) and 332 (N332) in the V2 region and V3 base, respec- tively, and also R166 and K169 in the V2 region, which are major targets of recently identified broad and potent neutralizing mono- clonal antibodies. In order to test this, two clade C Envs (25711- 2.4 and CAP239.G3), containing N160A and N332A substitutions, were tested (Table 2). Our data indicate that the pseudoviruses con- taining Env expressing the N160 or N332 substitution have sensi- tivities identical to those of G37080 plasma antibodies. Similar observations were found with R166A and K169A in the 93IN905 Env backbone. Taken together, our observations indicate that G37080 BCN plasma antibodies did not utilize these residues in V2 and V3 regions for neutralization breadth; these have been identified as important epitopes recognized by broadly neutraliz- ing antibodies elicited in clade C infection as described before (35, 41, 56). AssociationofneutralizationbreadthofG37080plasmawith recognition of conformational epitopes on cleaved trimeric Env but not with that in monomeric gp120 or MPER. In order to examine whether broad neutralization conferred by the G37080 plasma antibodies was through the recognition of epitopes on monomeric gp120 or cleaved near-native Env trimers, we tested the binding of G37080 serum IgG to monomeric 4-2.J41 gp120 and soluble gp140 (BG505-SOSIP.664) by ELISA. We found that in addition to the monomeric 4-2.J41 gp120 (Fig. 3A), G37080 serum polyclonal IgG was found to efficiently bind to the BG505 SOSIP.664-D7324 soluble trimeric Env (Fig. 3B), indicating that the G37080 plasma primarily contains neutralizing antibodies that target epitopes on cleaved Env trimers. We next examined whether binding of the G37080 plasma an- tibodies to epitopes on cleaved BG505-SOSIP.664 trimeric enve- lope was associated with neutralization breadth. For this, we tested the ability of G37080 plasma antibodies depleted of both mono- meric and trimeric Envs, as well as of MPER peptides, to neutralize a set of Env-pseudotyped viruses, which were found to be sensitive to this particular plasma sample. Purified 4-2.J41 monomeric gp120, BG505-SOSIP.664 trimeric gp140, and C1C MPER pep- tide bound to the magnetic beads were used to deplete G37080 plasma antibodies as described in Materials and Methods. The depleted BCN G37080 antibodies first were assessed for their binding to 4-2.J41 gp120 monomers, BG505-SOSIP.664-D7324, and C1C peptide and compared to undepleted plasma antibodies by ELISA. As shown in Fig. 3C and D, G37080 plasma depleted with monomeric gp120 and trimeric gp140, respectively, had sig- nificantly reduced binding activity against the respective soluble proteins. Similar observation was made with MPER peptide (data not shown). The depleted plasma antibodies subsequently were assessed for neutralization activity using a panel of 12 Env-pseu- dotyped viruses that were susceptible to untreated G37080 plasma antibodies as mentioned above. As shown in Table 3, depletion with 4-2.J41gp120 monomer and C1C peptide did not show any change in neutralization breadth of G37080 plasma antibodies, while depletion with BG505-SOSIP.664 showed a significant re- duction in virus neutralization. Similar observations were made with the BG505-SOSIP.664-depleted PGT121 and C1C peptide- depleted 4E10 MAbs, which lost the ability to efficiently neutralize Env-pseudotyped viruses (16055 and ZM233.6) and HIV-2/ HIV-1 (7312A-C1C) chimeric virus compared to their undepleted counterparts (data not shown), validating our data. Interestingly, C1C peptide-depleted G37080 plasma failed to neutralize HIV-2/ TABLE 3 Degree of shift in sensitivity of Env-pseudotyped viruses to G37080 BCN plasma depleted with monomeric and trimeric Envs and C1C peptide Env-pseudotyped virus Fold reduction in neutralization (ID50)a gp140 trimer (BG505-SOSIP.664) gp120 monomer (4-2.J41) MPER (C1C peptide) 25710-2.3 Ͼ10.30 1.3 0.83 25711-2.4 Ͼ8.52 1.4 1.44 3-5.J25 Ͼ7.85 0.9 0.84 4-2.J41 12.11 1.1 1.04 IAVI_C22 Ͼ15.92 1.2 1.18 92BR020 Ͼ35.08 1.1 1.34 93IN905 3.41 1.2 0.94 JRCSF Ͼ8.75 0.5 0.93 Q23.17 Ͼ23.28 1.0 0.98 Du156.12 Ͼ15.73 0.8 1.61 HVTR-PG80v1.eJ7 Ͼ10.03 0.9 1.12 HVTR-PG80v1.eJ19 Ͼ15.60 0.5 1.18 HIV-2 7312A-C1C Ͼ10 a Fold reduction in virus neutralization was obtained by comparing the neutralization titer (ID50 values) of panel viruses against undepleted and depleted G37080 plasma. ID50 values are reciprocal dilutions at which the undepleted and depleted plasma conferred 50% neutralization of the Env-pseudotyped viruses in TZM-bl cells. TABLE 2 Examination of specificity of G37080 plasma antibodies obtained at both visits to HIV Env HIV type Region Fold decrease in ID50 a for plasma at visit: 1 2 HIV-1 Env mutants HIV-1 25711-2.4 N160A V2 1.02 Ͻ1 HIV-1 25711-2.4 R166A Ͻ1 Ͻ1 HIV-1 25711-2.4 K169E Ͻ1 Ͻ1 HIV-1 93IN905 R166A Ͻ1 Ͻ1 HIV-1 93IN905 K169A Ͻ1 Ͻ1 HIV-1 25711-2.4 N332A V3 1.52 Ͻ1 HIV-1 CAP239.G3 N332A 1.35 1.32 HIV-2/HIV-1 chimera Region of HIV-1 ID50 ID50 HIV-2 7312A HIV-2 wild type Ͻ20 Ͻ20 HIV-2 7312A-C1C Clade C MPER 306.42 371.02 HIV-2 7312A-C3 2F5 epitope Ͻ20 Ͻ20 HIV-2 7312A-C4 4E10, Z13e1, and 10E8 epitopes 334.34 371.27 HIV-2 7312A-C6 4E10 minimal epitope Ͻ20 223.90 HIV-2 7312A-C7 2F5 minimal epitope Ͻ20 Ͻ20 a ID50 values refer to the reciprocal dilution that conferred 50% neutralization of viruses in a TZM-bl assay. Assays were done in duplicate and were repeated more than three times. Patil et al. 3450 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
  • 6. HIV-1 (7312A-C1C) chimeric virus, indicating that the presence of residual traces of MPER-directed antibodies (as shown in Table 2) is not responsible for neutralization breadth. Further- more, the examination of sensitive (25711-2.4) and resistant (16055-2.3 and CAP45.G3) chimeric Envs indicated that the BCN G37080 plasma antibodies predominantly target epitopes in the V1V2 region (Table 4) in gp120. Our data clearly indicate a cor- relation between neutralization breadth and binding of the G37080 BCN plasma antibodies to the conformational epitopes on cleaved trimeric gp120, likely in the V1V2 region; however, we do not rule out the possibility that this BCN plasma targets other discontinuous epitopes in gp120 but not in MPER. Mutations in V1 region confer resistance to autologous vi- ruses to the G37080 plasma antibodies. In order to decipher the specificity of the G37080 plasma antibodies, we examined the de- gree of susceptibility of pseudoviruses prepared using env genes amplified from contemporaneous autologous G37080 plasma ob- tained at the baseline and follow-up visits. As shown in Fig. 4A, both of the Env proteins obtained from visit 2 plasma (HVTR- PG80v2.eJ38 and HVTR-PG80v2.eJ41) were found to be resistant to its contemporaneous plasma antibodies, while Env proteins obtained from visit 1 plasma (HVTR-PG80v1.eJ7 and HVTR- PG80v1.eJ19) were found to be modestly sensitive to visit 2 autol- ogous G37080 plasma antibodies. To facilitate mapping G37080 BCN antibody specificity, we prepared chimeric Envs between a sensitive (HVTR-PG80v1.eJ7 and HVTR-PG80v1.eJ19) and a resistant (HVTR-PG80v2.eJ38) autologous Env by first swap- ping the V1V2 regions, as their amino acid sequences differed maximally in this region (Fig. 4B). As shown in Table 4, the insertion of the V1V2 sequences of HVTR-PG80v1.eJ7 and HVTR-PG80v1.eJ19 into HVTR-PG80v2.eJ38 conferred Env- pseudotyped viruses expressing HVTR-PG80v2.eJ38 Env with sensitivity to G37080 visit 2 plasma antibodies enhanced by Ͼ25- and Ͼ12-fold, respectively. Conversely, the neutraliza- tion susceptibilities of the Env-pseudotyped viruses expressing HVTR-PG80v1.eJ7 and HVTR-PG80v1.eJ19, which contained HVTR-PG80v2.eJ38 V1V2 sequence corresponding to visit 2 G37080 plasma, were found to be reduced by Ͼ45- and Ͼ23- fold, respectively. We noted that alterations of regions other than the V1V2 loop in the autologous Env did not confer any change in neutralization sensitivity (Table 4). To further spec- ify residues in the V1V2 loop associated with neutralization sensitivity and resistance of autologous Envs, chimeric Envs and point mutants were prepared and tested for their degree of modulation in susceptibility to autologous G37080 plasma ob- tained from the second visit. As shown in Table 4, we found that the V1 sequence, but not the V2 sequence, of the sensitive Envs (HVTR-PG80v1.eJ7 and HVTR-PG80v1.eJ19) increased sensitiv- ity to G37080 BCN plasma antibodies by Ͼ50 and Ͼ37-fold, re- spectively, when transferred to the resistant HVTR-PG80v2.eJ38 FIG 3 Binding of G37080 BCN plasma IgG to 4-2.J41 monomeric gp120 (A) and BG505-SOSIP.664-D7324 cleaved trimeric gp140 (B) soluble proteins was assessed by ELISA. IgG purified from HIV-negative healthy donor and known MAbs were used as controls. The extent of binding of the depleted and undepleted G37080 BCN plasma with magnetic beads coated with 4-2.J41 monomeric gp120 (C) and BG505-SOSIP.664 cleaved trimeric gp140 to their respective proteins by ELISA. Note that binding to trimeric protein by ELISA was assessed by using BG505-SOSIP.664 tagged with the D7324 epitope to maintain the native conformation of trimeric Env as described before (69). HIV-1 Clade C Plasma Confers Cross-Clade Neutralization April 2016 Volume 90 Number 7 jvi.asm.org 3451Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
  • 7. Env. In agreement with this result, V1 of HVTR-PG80v2.eJ38, when transferred into the sensitive Envs described above, in- creased neutralization resistance by Ͼ27- and Ͼ28-fold, respec- tively, to the G37080 visit 2 BCN plasma antibodies. We observed that the removal of a glycan at the 138 position in V1 (T140D) in HVTR-PG80v1.eJ19 mediated the enhanced sensitivity of this Env to G37080 plasma by 2.64-fold (Table 4). Concurrent with this observation, we found that the alteration of the V1 region of PG80v1.eJ19 with the T140D substitution in PG80v2.eJ38 Env exhibited enhanced susceptibility compared to that of the PG80v2.eJ38 Env chimera containing the PG80v1.eJ19 V1 loop, as shown in Table 4. Our data indicate that N138 glycan masks the PG80v1.eJ19 Env from being efficiently neutralized by the autol- ogous plasma compared to that of its contemporaneous counter- part, PG80v1.eJ7 Env. Fine scanning of V1 regions of the autolo- gous Envs further revealed that the N133 glycan motif and P147 residues in the PG80v2.eJ38 Env played a significant role in neu- tralization resistance to G37080 BCN autologous plasma antibod- ies (Fig. 4B). Interestingly, all of the V1 chimeras as well as the point mutants showed sensitivities to PG9 MAb comparable to those of their wild types (Table 5), indicating that the shifts in neutralization susceptibilities were not due to changes in Env con- formation. Moreover, we noted that both the sensitive and the resistant autologous Envs contain T332 in the V3 base, clearly indicating that the absence of N332 was not associated with resis- tance to autologous neutralization. Similar observations were made with respect to N160, R166, and K169 amino acid residues, further consolidating that the neutralization conferred by G37080 BCN plasma antibodies was not associated with antibody target- ing these epitopes in autologous Envs, and is likely the case for all of the Envs tested against G37080 plasma antibodies. DISCUSSION The identification of the molecular specificities of antibodies elic- ited in natural infection and that mediate neutralization breadth and potency is key in the design and development of suitable Env- based immunogens capable of eliciting similar antibody responses upon vaccination. In the present study, we characterized the mo- lecular specificity of plasma antibodies obtained from an Indian elite neutralizer (G37080) infected with HIV-1 clade C that dis- played exceptional cross-neutralization of different clades of distinct geographical origins. The G37080 plasma was found to contain the most broad and potent cross-neutralizing antibodies among the two hundred plasma samples obtained from Indian patients chronically infected with HIV-1. Plasma samples collected from the G37080 do- nor at two time points at 8 months apart showed similar neutraliza- tion breadth with modest increase in potency in the follow-up visit, indicating an association with the sustained maturation of antibody- producing B cells in this individual. Since polyclonal plasma antibodies are not suitable for epitope mapping, we examined the specificity of the G37080 BCN plasma by making use of mutant viruses with specific point substitutions of known neutralizing epitopes with nonspecific amino acids and via depletion with monomeric and trimeric Envs in addition to MPER peptide. The G37080 plasma antibodies did not show de- pendence on the N160/K169 and N332 epitopes in the V2 apex and V3 base, respectively. Our data also are consistent with the target epitopes of the G37080 BCN antibodies being distinct from those which are recognized by 2G12 (74), PGT121-128 (17), and PGT130-131 and PGT135 (19) (e.g., residues at the following po- TABLE 4 Dissection of specificity for autologous neutralization resistance Chimera and point mutant Neutralization potency (ID50) Fold changea Effectb PG80v1.eJ7 Env backbone V1V2 loop PG80v2.eJ38 (V1V2) in v1.eJ7 45.75 Decrease PG80v2.eJ38 (V1) in v1.eJ7 27.97 Decrease PG80v2.eJ38 (V2) in v1.eJ7 1.35 No effect Point mutations PG80v1.eJ7 (D133N) 3.25 Decrease PG80v1.eJ7 (S143G) 0.87 No effect PG80v1.eJ7 (D133N ϩ S143G) 2.88 Decrease PG80v1.eJ7 (T147P) 8.21 Decrease PG80v1.eJ19 Env backbone V1V2 loop PG80v2.eJ38(V1V2) in v1.eJ19 23.05 Decrease PG80v2.eJ38 (V1) in v1.eJ19 28.61 Decrease PG80v2.eJ38 (V2) in v1.eJ19 1.87 Increase Point mutations PG80v1.eJ19 (D133N) 2.51 Decrease PG80v1.eJ19 (T139A) 0.99 No effect PG80v1.eJ19 (T139A ϩ T140D) 2.64 Increase PG80v1.eJ19 (N143G) 1.38 No effect PG80v1.eJ19 (T139A ϩ T140D ϩ N143G) 2.16 Increase PG80v1.eJ19 (T145P) 3.24 Decrease PG80v2.eJ38 Env backbone V1V2 loop PG80v1.eJ7 (V1V2) in v2.eJ38 26.56 Increase PG80v1.eJ7 (V1) in v2.eJ38 49.62 Increase PG80v1.eJ7 (V2) in v2.eJ38 1.07 No effect PG80v1.eJ19(V1V2) in v2.eJ38 12.81 Increase PG80v1.eJ19 (V1) in v2.eJ38 37.60 Increase PG80v1.eJ19 (V2) in v2.eJ38 0.94 No effect Other regions in gp120 PG80v1.eJ7 (V3C3) in v2.eJ38 0.84 No effect PG80v1.eJ7 (V3C3V4C4) in v2.eJ38 0.90 No effect PG80v1.eJ7 (C4V5C5) in v2.eJ38 1.08 No effect PG80v1.eJ19 (V3C3) in PG80v2.eJ38 1.09 No effect PG80v1.eJ19 (V3C3V4C4) in PG80v2.eJ38 1.15 No effect PG80v1.eJ19 (C4V5C5) in PG80v2.eJ38 0.96 No effect Point mutations PG80v1.eJ19 V1 (T139AϩT140D) in v2.eJ38 53.91 Increase PG80v2.eJ38 (N133D) 4.72 Increase PG80v2.eJ38 (G143S) 0.94 No effect PG80v2.eJ38 (N133DϩG143S) 3.63 Increase PG80v2.eJ38 (P147T) 4.11 Increase Heterologous Env chimera V1V2 loop 16055-2.3 (25711-2.4 V1V2) 18.38 Increase 25711-2.4 (16055-2.3 V1V2) 2.03 Decrease CAP45 (25711-2.4 V1V2) 16.84 Increase 25711-2.4 (CAP45-V1V2) 10.54 Decrease a Fold changes in reciprocal dilution of plasma mediating 50% virus neutralization (ID50). b Fold increase or decrease in neutralization titer (ID50 values). Patil et al. 3452 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
  • 8. sitions: 295, 297, 301, 332, 334, 386, 388, 392, 394, 448, and 450); thus, BCN G37080 antibodies appear to target a new epitope. Our data highlighting the N332-independent development of neutral- izing antibodies in clade C-infected donor G37080 also differ from recent findings (57, 75–77) associating N332 with the develop- ment of broad and potent neutralizing antibody, especially in clade C infection in African donors. Moreover, recent studies in- dicating the role of K169 as a target of BCN antibodies obtained from a clade C-infected South African donor (41, 56) and the observation that vaccine-induced protection in the RV144 vaccine trial was associated with antibodies targeting epitopes, including K169 in the V2 apex (27, 78), prompted us to examine whether broad neutralization of the G37080 plasma antibodies also was dependent on the K169 epitope. In the present study, the neutral- ization potency of G37080 was unaffected by N160A/K169A knockout mutations, and we also observed that both sensitive and resistant autologous Envs obtained from both visits contain N160 and K169 in the V2 region. Hence, owing to the lack of association of neutralization breadth of the G37080 BCN antibodies with N160, K169, and N332 dependencies, our study further high- lighted that there is a likelihood of differences in the development pathway of elicitation of broadly neutralizing antibodies in indi- viduals infected with HIV-1 clade C, particularly those with eth- nically distinct variants. Wibmer et al. (41) recently demonstrated an association be- tween the evolution of a broadly neutralizing antibody response in a clade C-infected donor with shifts in antibody specificities from the recognition of epitopes in V2 to the CD4bs. In the present study, the G37080 neutralizing plasma antibodies obtained from both visits were found not to be absorbed by the TripleMut (66, 79) and RSC3 (22) core proteins, which effectively absorb anti- bodies directed to the CD4bs. This result indicates a lack of devel- opment of CD4bs-directed neutralizing antibodies during the dis- ease course in the G37080 donor. Additionally, the absence of MPER-directed antibodies from G37080 plasma was found, al- though a negligible antibody titer (1:300 reciprocal dilutions) to the HIV2/HIV1 (C1C) chimera was observed with plasma sam- FIG 4 (A) Neutralization susceptibility of autologous Envs to contemporaneous G37080 BCN plasma and its follow-up sample from the same donor. Neutralization titers (median ID50) were obtained by titrating pseudotyped viruses expressing autologous Envs obtained from visit 1 and follow-up G37080 plasma to contemporaneous plasma antibodies. Note that both of the Envs obtained from follow-up G37080 plasma (visit 2) were found to be resistant to contemporaneous autologous plasma, while Envs obtained from visit 1 G37080 plasma were found to be sensitive to follow-up plasma antibodies. (B) Alignment of V1V2 amino acid sequences of sensitive and resistant autologous Envs obtained at both visits was done by using seqpublish, available at the HIV Los Alamos database (www.hiv.lanl.gov). Key residues that mediate autologous neutralization resistance are highlighted. TABLE 5 Sensitivity of wild type, chimera, and point mutants of autologous Envs to PG9 MAb Env chimera and mutant ID50 PG80v1.eJ7 (wild type) 0.12 PG80v1.eJ19 (wild type) 0.97 PG80v1.eJ19 (T139A ϩ T140D) 0.77 PG80v2.eJ38 (wild type) 0.02 PG80v1.eJ7 (V1) in v2.eJ38 0.04 PG80v1.eJ19 (V1) in v2.eJ38 0.05 PG80v1.eJ19 (V1) (T139A ϩ T140D) in v2.eJ38 0.06 PG80v2.eJ38 (N133D) 0.01 PG80v2.eJ38 (P147T) 0.04 HIV-1 Clade C Plasma Confers Cross-Clade Neutralization April 2016 Volume 90 Number 7 jvi.asm.org 3453Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
  • 9. ples from both visits. However, the neutralization breadth of the G37080 plasma was not found to be associated with the presence of MPER-directed antibody. Nonetheless, we do not rule out the possibility that in the further course of infection, this donor may be able to develop MPER-directed antibodies. Recentstudieshaveshownthatneutralizingantibodiesthattarget conformational epitopes bind exclusively to the cleaved near-native trimeric Envs (15, 64, 80, 81). In the present study, we found that the absorption of G37080 plasma antibodies to soluble trimeric BG505- SOSIP.664 Env was associated with the depletion of neutralizing ac- tivity in G37080 BCN plasma. However, we do not rule out the pos- sibility of the presence of 39F-, 19b-, and 14e-like nonneutralizing antibodies that were reported to bind to BG505-SOSIP.664 trimeric Env (69). Our findings indicate that the G37080 BCN antibodies tar- get conformational epitopes in gp120. Our observation also high- lights that native-like trimeric Envs, such as BG505-SOSIP.664, can be utilized in selecting antigen-specific memory B cells, as reported earlier(82),fromdonorG37080towardisolationofMAbcorrelating with broad neutralization displayed by the plasma antibodies. We made use of env clones obtained from autologous G37080 plasma from both time points to refine the fine specificity of the G37080 BCN plasma antibodies. By examining chimeric Envs and mutant viruses, we identified key residues in the V1 loop associ- ated with neutralization resistance. Interestingly, the Env chimera and mutant viruses showed susceptibility to PG9 MAb compara- ble to that of their respective wild-type Envs, indicating that they did not alter Env conformation. We identified a glycan at the 133 position and a proline residue at the 147 position within the V1 loop of the resistant Env (PG80v2.eJ38) that were found to be associated with neutralization escape, which indicated that these are contact sites for the G37080 BCN plasma antibodies. Thus, from our study we conclude that changes in V1 loop sequence are associated with the escape of autologous viruses to the BCN G37080 plasma. Additionally, an examination of the degree of susceptibilities of pseudoviruses expressing chimeric heterolo- gous Envs to the G37080 plasma revealed that the BCN plasma antibodies predominantly target epitopes in the V1V2 region in gp120. However, we do not rule out the possibility of the contri- bution of other discontinuous epitopes in gp120 in mediating neutralization breadth. The isolation and identification of mono- clonal antibodies from this elite neutralizer donor (G37080) will help precisely map specific epitopes associated with neutralization breadth and potency. In summary, we identified an HIV-1-infected elite neutralizer whose plasma showed exceptional neutralization breadth, and we provided evidence that it targets novel conformational epitopes on trimeric Env, predominantly in the V1V2 region, not reported previously. Moreover, the neutralization resistance of the autolo- gous Envs to G37080 plasma is associated with substitutions of novel residues within the V1 loop that form the key contact points of the BCN plasma antibody. The identification of novel epitopes associated with broad neutralization of HIV-1, in particular the major circulating clade C strains, will significantly contribute to efforts toward effective immunogen design. ACKNOWLEDGMENTS We thank all of the Protocol G study participants registered with YRG Care, Chennai, all of the research staff members at the Protocol G clinical center at YRG Care, Chennai, and all of the IAVI Protocol G team mem- bers. We sincerely thank Christopher Parks, IAVI Design and Develop- ment Laboratory, for providing valuable input in preparing the manu- script, and we also thank G. Balakrish Nair and Sudhanshu Vrati (THSTI), Shreyasi Chatterjee, and all of the HVTR laboratory members for support. We thank Albert Cupo, John P. Moore, and the members the SOSIP trimer HIVRAD team, Weill Cornell Medical College, New York, for pro- viding us with BG505.SOSIP.664 plasmid DNA and purified protein. We thank David Montefiori, Lynn Morris, Pascal Poignard, and Richard Wy- att for making available many reagents used in our study. The following reagent was obtained through the NIH AIDS Reagent Program, Division of AIDS, NIAID, NIH, from John C. Kappes and Xiaoyun Wu: pSG3 env. IAVI’s work was made possible by generous support from many do- nors, including the Bill & Melinda Gates Foundation, the Ministry of Foreign Affairs of Denmark, Irish Aid, the Ministry of Finance of Japan, the Ministry of Foreign Affairs of the Netherlands, the Norwegian Agency for Development Cooperation (NORAD), the United Kingdom Depart- ment for International Development (DFID), and the United States Agency for International Development (USAID). The full list of IAVI donors is available at www.iavi.org. The contents are the responsibility of the International AIDS Vaccine Initiative and do not necessarily reflect the views of USAID or the United States Government. The International AIDS Vaccine Initiative has filed a patent relating to the autologous HIV-1 clade C envelope clones (J. Bhattacharya, S. Desh- pande, S. Patil, R. Kumar, and B. K. Chakrabarti, U.S. patent application 62/254,971). FUNDING INFORMATION This study was made possible by the generous support of the American people through the United States Agency for International Development (USAID) through the IAVI, support from a THSTI-IAVI HIV Vaccine Design Program grant through the Department of Biotechnology, Gov- ernment of India, in part by a grant from the Department of Science and Technology, Government of India (DST/INT/SAFR/Mega-P3/2011 to J.B.), and in part by a DBT National Bioscience Research Award [BT/ HRD/NBA34/01/2012-13(iv) to J.B.]. The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication. REFERENCES 1. Caskey M, Klein F, Lorenzi JC, Seaman MS, West AP, Jr, Buckley N, Kremer G, Nogueira L, Braunschweig M, Scheid JF, Horwitz JA, Shime- liovich I, Ben-Avraham S, Witmer-Pack M, Platten M, Lehmann C, Burke LA, Hawthorne T, Gorelick RJ, Walker BD, Keler T, Gulick RM, Fatkenheuer G, Schlesinger SJ, Nussenzweig MC. 2015. Viraemia sup- pressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature 522:487–491. http://dx.doi.org/10.1038/nature14411. 2. Esparza J. 2013. A brief history of the global effort to develop a preventive HIV vaccine. Vaccine 31:3502–3518. http://dx.doi.org/10.1016/j.vaccine .2013.05.018. 3. Hraber P, Seaman MS, Bailer RT, Mascola JR, Montefiori DC, Korber BT. 2014. Prevalence of broadly neutralizing antibody responses during chronic HIV-1 infection. AIDS 28:163–169. http://dx.doi.org/10.1097 /QAD.0000000000000106. 4. Klein F, Mouquet H, Dosenovic P, Scheid JF, Scharf L, Nussenzweig MC. 2013. Antibodies in HIV-1 vaccine development and therapy. Sci- ence 341:1199–1204. http://dx.doi.org/10.1126/science.1241144. 5. Koff WC, Russell ND, Walport M, Feinberg MB, Shiver JW, Karim SA, Walker BD, McGlynn MG, Nweneka CV, Nabel GJ. 2013. Accelerating the development of a safe and effective HIV vaccine: HIV vaccine case study for the decade of vaccines. Vaccine 31(Suppl 2):B204–B208. http: //dx.doi.org/10.1016/j.vaccine.2012.10.115. 6. Kwong PD, Mascola JR, Nabel GJ. 2013. Broadly neutralizing antibodies and the search for an HIV-1 vaccine: the end of the beginning. Nat Rev Immunol 13:693–701. http://dx.doi.org/10.1038/nri3516. 7. Braibant M, Brunet S, Costagliola D, Rouzioux C, Agut H, Katinger H, Autran B, Barin F. 2006. Antibodies to conserved epitopes of the HIV-1 envelope in sera from long-term non-progressors: prevalence and associ- ation with neutralizing activity. AIDS 20:1923–1930. http://dx.doi.org/10 .1097/01.aids.0000247113.43714.5e. Patil et al. 3454 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
  • 10. 8. Donners H, Willems B, Beirnaert E, Colebunders R, Davis D, van der Groen G. 2002. Cross-neutralizing antibodies against primary isolates in African women infected with HIV-1. AIDS 16:501–503. http://dx.doi.org /10.1097/00002030-200202150-00030. 9. Gray ES, Madiga MC, Hermanus T, Moore PL, Wibmer CK, Tumba NL, Werner L, Mlisana K, Sibeko S, Williamson C, Abdool Karim SS, Morris L. 2011. The neutralization breadth of HIV-1 develops incremen- tally over four years and is associated with CD4ϩ T cell decline and high viral load during acute infection. J Virol 85:4828–4840. http://dx.doi.org /10.1128/JVI.00198-11. 10. Li Y, Svehla K, Louder MK, Wycuff D, Phogat S, Tang M, Migueles SA, Wu X, Phogat A, Shaw GM, Connors M, Hoxie J, Mascola JR, Wyatt R. 2009. Analysis of neutralization specificities in polyclonal sera derived from human immunodeficiency virus type 1-infected individuals. J Virol 83:1045–1059. http://dx.doi.org/10.1128/JVI.01992-08. 11. Richman DD, Wrin T, Little SJ, Petropoulos CJ. 2003. Rapid evolu- tion of the neutralizing antibody response to HIV type 1 infection. Proc Natl Acad Sci U S A 100:4144–4149. http://dx.doi.org/10.1073 /pnas.0630530100. 12. Sather DN, Armann J, Ching LK, Mavrantoni A, Sellhorn G, Caldwell Z, Yu X, Wood B, Self S, Kalams S, Stamatatos L. 2009. Factors associated with the development of cross-reactive neutralizing antibodies during human immunodeficiency virus type 1 infection. J Virol 83:757– 769. http://dx.doi.org/10.1128/JVI.02036-08. 13. Simek MD, Rida W, Priddy FH, Pung P, Carrow E, Laufer DS, Lehrman JK, Boaz M, Tarragona-Fiol T, Miiro G, Birungi J, Pozniak A, McPhee DA, Manigart O, Karita E, Inwoley A, Jaoko W, Dehovitz J, Bekker LG, Pitisuttithum P, Paris R, Walker LM, Poignard P, Wrin T, Fast PE, Burton DR, Koff WC. 2009. Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity iden- tified by using a high-throughput neutralization assay together with an analytical selection algorithm. J Virol 83:7337–7348. http://dx.doi.org/10 .1128/JVI.00110-09. 14. Stamatatos L, Morris L, Burton DR, Mascola JR. 2009. Neutralizing antibodies generated during natural HIV-1 infection: good news for an HIV-1 vaccine? Nat Med 15:866–870. 15. Falkowska E, Le KM, Ramos A, Doores KJ, Lee JH, Blattner C, Ramirez A, Derking R, MJ van Gils Liang CH, McBride R, von Bredow B, Shivatare SS, Wu CY, Chan-Hui PY, Liu Y, Feizi T, Zwick MB, Koff WC, Seaman MS, Swiderek K, Moore JP, Evans D, Paulson JC, Wong CH, Ward AB, Wilson IA, Sanders RW, Poignard P, Burton DR. 2014. Broadly neutralizing HIV antibodies define a glycan-dependent epitope on the prefusion conformation of gp41 on cleaved envelope trimers. Im- munity 40:657–668. http://dx.doi.org/10.1016/j.immuni.2014.04.009. 16. Huang J, Ofek G, Laub L, Louder MK, Doria-Rose NA, Longo NS, Imamichi H, Bailer RT, Chakrabarti B, Sharma SK, Alam SM, Wang T, Yang Y, Zhang B, Migueles SA, Wyatt R, Haynes BF, Kwong PD, Mascola JR, Connors M. 2012. Broad and potent neutralization of HIV-1 by a gp41-specific human antibody. Nature 491:406–412. http://dx.doi .org/10.1038/nature11544. 17. Julien JP, Sok D, Khayat R, Lee JH, Doores KJ, Walker LM, Ramos A, Diwanji DC, Pejchal R, Cupo A, Katpally U, Depetris RS, Stanfield RL, McBride R, Marozsan AJ, Paulson JC, Sanders RW, Moore JP, Burton DR, Poignard P, Ward AB, Wilson IA. 2013. Broadly neutralizing anti- body PGT121 allosterically modulates CD4 binding via recognition of the HIV-1 gp120 V3 base and multiple surrounding glycans. PLoS Pathog 9:e1003342. http://dx.doi.org/10.1371/journal.ppat.1003342. 18. Scharf L, Scheid JF, Lee JH, West AP, Jr, Chen C, Gao H, Gnanapra- gasam PN, Mares R, Seaman MS, Ward AB, Nussenzweig MC, Bjork- man PJ. 2014. Antibody 8ANC195 reveals a site of broad vulnerability on the HIV-1 envelope spike. Cell Rep 7:785–795. http://dx.doi.org/10.1016 /j.celrep.2014.04.001. 19. Walker LM, Huber M, Doores KJ, Falkowska E, Pejchal R, Julien JP, Wang SK, Ramos A, Chan-Hui PY, Moyle M, Mitcham JL, Hammond PW, Olsen OA, Phung P, Fling S, Wong CH, Phogat S, Wrin T, Simek MD, Protocol G Principal Investigators, Koff WC, Wilson IA, Burton DR, Poignard P. 2011. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 477:466–470. http://dx.doi.org/10.1038 /nature10373. 20. Walker LM, Phogat SK, Chan-Hui PY, Wagner D, Phung P, Goss JL, Wrin T, Simek MD, Fling S, Mitcham JL, Lehrman JK, Priddy FH, Olsen OA, Frey SM, Hammond PW, Kaminsky S, Zamb T, Moyle M, Koff WC, Poignard P, Burton DR. 2009. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Sci- ence 326:285–289. http://dx.doi.org/10.1126/science.1178746. 21. Walker LM, Simek MD, Priddy F, Gach JS, Wagner D, Zwick MB, Phogat SK, Poignard P, Burton DR. 2010. A limited number of antibody specificities mediate broad and potent serum neutralization in selected HIV-1 infected individuals. PLoS Pathog 6:e1001028. http://dx.doi.org/10 .1371/journal.ppat.1001028. 22. Wu X, Yang ZY, Li Y, Hogerkorp CM, Schief WR, Seaman MS, Zhou T, Schmidt SD, Wu L, Xu L, Longo NS, McKee K, O’Dell S, Louder MK, Wycuff DL, Feng Y, Nason M, Doria-Rose N, Connors M, Kwong PD, Roederer M, Wyatt RT, Nabel GJ, Mascola JR. 2010. Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science 329:856–861. http://dx.doi.org/10.1126/science.1187659. 23. Wu X, Zhou T, Zhu J, Zhang B, Georgiev I, Wang C, Chen X, Longo NS, Louder M, McKee K, O’Dell S, Perfetto S, Schmidt SD, Shi W, Wu L, Yang Y, Yang ZY, Yang Z, Zhang Z, Bonsignori M, Crump JA, Kapiga SH, Sam NE, Haynes BF, Simek M, Burton DR, Koff WC, Doria-Rose NA, Connors M, NISC Comparative Sequencing Program, Mullikin JC, Nabel GJ, Roederer M, Shapiro L, Kwong PD, Mascola JR. 2011. Focused evolution of HIV-1 neutralizing antibodies revealed by structures and deep sequencing. Science 333:1593–1602. http://dx.doi.org /10.1126/science.1207532. 24. Diskin R, Scheid JF, Marcovecchio PM, West AP, Jr, Klein F, Gao H, Gnanapragasam PN, Abadir A, Seaman MS, Nussenzweig MC, Bjork- man PJ. 2011. Increasing the potency and breadth of an HIV antibody by using structure-based rational design. Science 334:1289–1293. http://dx .doi.org/10.1126/science.1213782. 25. Klein F, Gaebler C, Mouquet H, Sather DN, Lehmann C, Scheid JF, Kraft Z, Liu Y, Pietzsch J, Hurley A, Poignard P, Feizi T, Morris L, Walker BD, Fatkenheuer G, Seaman MS, Stamatatos L, Nussenzweig MC. 2012. Broad neutralization by a combination of antibodies recogniz- ing the CD4 binding site and a new conformational epitope on the HIV-1 envelope protein. J Exp Med 209:1469–1479. http://dx.doi.org/10.1084 /jem.20120423. 26. Scheid JF, Mouquet H, Ueberheide B, Diskin R, Klein F, Oliveira TY, Pietzsch J, Fenyo D, Abadir A, Velinzon K, Hurley A, Myung S, Boulad F, Poignard P, Burton DR, Pereyra F, Ho DD, Walker BD, Seaman MS, Bjorkman PJ, Chait BT, Nussenzweig MC. 2011. Se- quence and structural convergence of broad and potent HIV antibod- ies that mimic CD4 binding. Science 333:1633–1637. http://dx.doi.org /10.1126/science.1207227. 27. Liao HX, Bonsignori M, Alam SM, McLellan JS, Tomaras GD, Moody MA, Kozink DM, Hwang KK, Chen X, Tsao CY, Liu P, Lu X, Parks RJ, Montefiori DC, Ferrari G, Pollara J, Rao M, Peachman KK, Santra S, Letvin NL, Karasavvas N, Yang ZY, Dai K, Pancera M, Gorman J, Wiehe K, Nicely NI, Rerks-Ngarm S, Nitayaphan S, Kaewkungwal J, Pitisuttithum P, Tartaglia J, Sinangil F, Kim JH, Michael NL, Kepler TB, Kwong PD, Mascola JR, Nabel GJ, Pinter A, Zolla-Pazner S, Haynes BF. 2013. Vaccine induction of antibodies against a structurally heteroge- neous site of immune pressure within HIV-1 envelope protein variable regions 1 and 2. Immunity 38:176–186. http://dx.doi.org/10.1016/j .immuni.2012.11.011. 28. Pejchal R, Doores KJ, Walker LM, Khayat R, Huang PS, Wang SK, Stanfield RL, Julien JP, Ramos A, Crispin M, Depetris R, Katpally U, Marozsan A, Cupo A, Maloveste S, Liu Y, McBride R, Ito Y, Sanders RW, Ogohara C, Paulson JC, Feizi T, Scanlan CN, Wong CH, Moore JP, Olson WC, Ward AB, Poignard P, Schief WR, Burton DR, Wilson IA. 2011. A potent and broad neutralizing antibody recognizes and pen- etrates the HIV glycan shield. Science 334:1097–1103. http://dx.doi.org /10.1126/science.1213256. 29. Blattner C, Lee JH, Sliepen K, Derking R, Falkowska E, de la Pena AT, Cupo A, Julien JP, van Gils M, Lee PS, Peng W, Paulson JC, Poignard P, Burton DR, Moore JP, Sanders RW, Wilson IA, Ward AB. 2014. Structural delineation of a quaternary, cleavage-dependent epitope at the gp41-gp120 interface on intact HIV-1 Env trimers. Immunity 40:669– 680. http://dx.doi.org/10.1016/j.immuni.2014.04.008. 30. Morris L, Chen X, Alam M, Tomaras G, Zhang R, Marshall DJ, Chen B, Parks R, Foulger A, Jaeger F, Donathan M, Bilska M, Gray ES, Abdool Karim SS, Kepler TB, Whitesides J, Montefiori D, Moody MA, Liao HX, Haynes BF. 2011. Isolation of a human anti-HIV gp41 mem- brane proximal region neutralizing antibody by antigen-specific single B cell sorting. PLoS One 6:e23532. http://dx.doi.org/10.1371/journal.pone .0023532. HIV-1 Clade C Plasma Confers Cross-Clade Neutralization April 2016 Volume 90 Number 7 jvi.asm.org 3455Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
  • 11. 31. Nelson JD, Brunel FM, Jensen R, Crooks ET, Cardoso RM, Wang M, Hessell A, Wilson IA, Binley JM, Dawson PE, Burton DR, Zwick MB. 2007. An affinity-enhanced neutralizing antibody against the membrane- proximal external region of human immunodeficiency virus type 1 gp41 recognizes an epitope between those of 2F5 and 4E10. J Virol 81:4033– 4043. http://dx.doi.org/10.1128/JVI.02588-06. 32. Zwick MB, Labrijn AF, Wang M, Spenlehauer C, Saphire EO, Binley JM, Moore JP, Stiegler G, Katinger H, Burton DR, Parren PW. 2001. Broadly neutralizing antibodies targeted to the membrane-proximal external region of human immunodeficiency virus type 1 glycoprotein gp41. J Virol 75:10892–10905. http://dx.doi.org/10.1128/JVI.75.22 .10892-10905.2001. 33. Chaillon A, Braibant M, Moreau T, Thenin S, Moreau A, Autran B, Barin F. 2011. The V1V2 domain and an N-linked glycosylation site in the V3 loop of the HIV-1 envelope glycoprotein modulate neutralization sen- sitivity to the human broadly neutralizing antibody 2G12. J Virol 85:3642– 3648. http://dx.doi.org/10.1128/JVI.02424-10. 34. Doria-Rose NA, Georgiev I, O’Dell S, Chuang GY, Staupe RP, McLellan JS, Gorman J, Pancera M, Bonsignori M, Haynes BF, Burton DR, Koff WC, Kwong PD, Mascola JR. 2012. A short segment of the HIV-1 gp120 V1/V2 region is a major determinant of resistance to V1/V2 neutralizing antibodies. J Virol 86:8319–8323. http://dx.doi.org/10.1128/JVI.00696-12. 35. Doria-Rose NA, Schramm CA, Gorman J, Moore PL, Bhiman JN, DeKosky BJ, Ernandes MJ, Georgiev IS, Kim HJ, Pancera M, Staupe RP, Altae-Tran HR, Bailer RT, Crooks ET, Cupo A, Druz A, Garrett NJ, Hoi KH, Kong R, Louder MK, Longo NS, McKee K, Nonyane M, O’Dell S, Roark RS, Rudicell RS, Schmidt SD, Sheward DJ, Soto C, Wibmer CK, Yang Y, Zhang Z, NISC Comparative Sequencing Program, Mullikin JC, Binley JM, Sanders RW, Wilson IA, Moore JP, Ward AB, Georgiou G, Williamson C, Abdool Karim SS, Morris L, Kwong PD, Shapiro L, Mascola JR. 2014. Developmental pathway for potent V1V2-directed HIV-neutralizing antibodies. Nature 509:55–62. http://dx.doi.org/10 .1038/nature13036. 36. Harrington PR, Nelson JA, Kitrinos KM, Swanstrom R. 2007. Indepen- dent evolution of human immunodeficiency virus type 1 env V1/V2 and V4/V5 hypervariable regions during chronic infection. J Virol 81:5413– 5417. http://dx.doi.org/10.1128/JVI.02554-06. 37. Moore PL, Gray ES, Choge IA, Ranchobe N, Mlisana K, Abdool Karim SS, Williamson C, Morris L. 2008. The c3-v4 region is a major target of autologous neutralizing antibodies in human immunodeficiency virus type 1 subtype C infection. J Virol 82:1860–1869. http://dx.doi.org/10 .1128/JVI.02187-07. 38. Rusert P, Krarup A, Magnus C, Brandenberg OF, Weber J, Ehlert AK, Regoes RR, Gunthard HF, Trkola A. 2011. Interaction of the gp120 V1V2 loop with a neighboring gp120 unit shields the HIV envelope trimer against cross-neutralizing antibodies. J Exp Med 208:1419–1433. http: //dx.doi.org/10.1084/jem.20110196. 39. Sagar M, Wu X, Lee S, Overbaugh J. 2006. Human immunodeficiency virus type 1 V1-V2 envelope loop sequences expand and add glycosylation sites over the course of infection, and these modifications affect antibody neutralization sensitivity. J Virol 80:9586–9598. http://dx.doi.org/10.1128 /JVI.00141-06. 40. van Gils MJ, Bunnik EM, Boeser-Nunnink BD, Burger JA, Terlouw- Klein M, Verwer N, Schuitemaker H. 2011. Longer V1V2 region with increased number of potential N-linked glycosylation sites in the HIV-1 envelope glycoprotein protects against HIV-specific neutralizing antibod- ies. J Virol 85:6986–6995. http://dx.doi.org/10.1128/JVI.00268-11. 41. Wibmer CK, Bhiman JN, Gray ES, Tumba N, Abdool Karim SS, Williamson C, Morris L, Moore PL. 2013. Viral escape from HIV-1 neutralizing antibodies drives increased plasma neutralization breadth through sequential recognition of multiple epitopes and immuno- types. PLoS Pathog 9:e1003738. http://dx.doi.org/10.1371/journal .ppat.1003738. 42. Alter G, Barouch DH. 2015. Natural evolution of broadly neutralizing antibodies. Cell 161:427–428. http://dx.doi.org/10.1016/j.cell.2015.04 .007. 43. Doores KJ, Kong L, Krumm SA, Le KM, Sok D, Laserson U, Garces F, Poignard P, Wilson IA, Burton DR. 2015. Two classes of broadly neu- tralizing antibodies within a single lineage directed to the high-mannose patch of HIV envelope. J Virol 89:1105–1118. http://dx.doi.org/10.1128 /JVI.02905-14. 44. Doria-Rose NA, Joyce MG. 2015. Strategies to guide the antibody affinity maturation process. Curr Opin Virol 11:137–147. http://dx.doi .org/10.1016/j.coviro.2015.04.002. 45. Fera D, Schmidt AG, Haynes BF, Gao F, Liao HX, Kepler TB, Harrison SC. 2014. Affinity maturation in an HIV broadly neutralizing B-cell lin- eage through reorientation of variable domains. Proc Natl Acad Sci U S A 111:10275–10280. http://dx.doi.org/10.1073/pnas.1409954111. 46. Horiya S, Bailey JK, Temme JS, Guillen Schlippe YV, Krauss IJ. 2014. Directed evolution of multivalent glycopeptides tightly recognized by HIV antibody 2G12. J Am Chem Soc 136:5407–5415. http://dx.doi.org/10.1021 /ja500678v. 47. Mikell I, Stamatatos L. 2012. Evolution of cross-neutralizing antibody specificities to the CD4-BS and the carbohydrate cloak of the HIV Env in an HIV-1-infected subject. PLoS One 7:e49610. http://dx.doi.org/10.1371 /journal.pone.0049610. 48. Sather DN, Carbonetti S, Malherbe DC, Pissani F, Stuart AB, Hessell AJ, Gray MD, Mikell I, Kalams SA, Haigwood NL, Stamatatos L. 2014. Emergence of broadly neutralizing antibodies and viral coevolution in two subjects during the early stages of infection with human immunodefi- ciency virus type 1. J Virol 88:12968–12981. http://dx.doi.org/10.1128/JVI .01816-14. 49. Wu X, Zhang Z, Schramm CA, Joyce MG, Do Kwon Y, Zhou T, Sheng Z, Zhang B, O’Dell S, McKee K, Georgiev IS, Chuang GY, Longo NS, Lynch RM, Saunders KO, Soto C, Srivatsan S, Yang Y, Bailer RT, Louder MK, NISC Comparative Sequencing Program, Mullikin JC, Connors M, Kwong PD, Mascola JR, Shapiro L. 2015. Maturation and diversity of the VRC01-antibody lineage over 15 years of chronic HIV-1 infection. Cell 161:470–485. http://dx.doi.org/10.1016/j.cell.2015.03.004. 50. Zhu J, Ofek G, Yang Y, Zhang B, Louder MK, Lu G, McKee K, Pancera M, Skinner J, Zhang Z, Parks R, Eudailey J, Lloyd KE, Blinn J, Alam SM, Haynes BF, Simek M, Burton DR, Koff WC, NISC Comparative Sequencing Program, Mullikin JC, Mascola JR, Shapiro L, Kwong PD. 2013. Mining the antibodyome for HIV-1-neutralizing antibodies with next-generation sequencing and phylogenetic pairing of heavy/light chains. Proc Natl Acad Sci U S A 110:6470–6475. http://dx.doi.org/10 .1073/pnas.1219320110. 51. Basu D, Kraft CS, Murphy MK, Campbell PJ, Yu T, Hraber PT, Irene C, Pinter A, Chomba E, Mulenga J, Kilembe W, Allen SA, Derdeyn CA, Hunter E. 2012. HIV-1 subtype C superinfected individuals mount low autologous neutralizing antibody responses prior to intrasubtype super- infection. Retrovirology 9:76. http://dx.doi.org/10.1186/1742-4690-9-76. 52. Gray ES, Meyers T, Gray G, Montefiori DC, Morris L. 2006. Insensi- tivity of paediatric HIV-1 subtype C viruses to broadly neutralising mono- clonal antibodies raised against subtype B. PLoS Med 3:e255. http://dx.doi .org/10.1371/journal.pmed.0030255. 53. Gray ES, Moody MA, Wibmer CK, Chen X, Marshall D, Amos J, Moore PL, Foulger A, Yu JS, Lambson B, Abdool Karim S, Whitesides J, Tomaras GD, Haynes BF, Morris L, Liao HX. 2011. Isolation of a monoclonal antibody that targets the alpha-2 helix of gp120 and repre- sents the initial autologous neutralizing-antibody response in an HIV-1 subtype C-infected individual. J Virol 85:7719–7729. http://dx.doi.org/10 .1128/JVI.00563-11. 54. Gray ES, Moore PL, Choge IA, Decker JM, Bibollet-Ruche F, Li H, Leseka N, Treurnicht F, Mlisana K, Shaw GM, Karim SS, Williamson C, Morris L. 2007. Neutralizing antibody responses in acute human immu- nodeficiency virus type 1 subtype C infection. J Virol 81:6187–6196. http: //dx.doi.org/10.1128/JVI.00239-07. 55. Gray ES, Taylor N, Wycuff D, Moore PL, Tomaras GD, Wibmer CK, Puren A, DeCamp A, Gilbert PB, Wood B, Montefiori DC, Binley JM, Shaw GM, Haynes BF, Mascola JR, Morris L. 2009. Antibody specific- ities associated with neutralization breadth in plasma from human immu- nodeficiency virus type 1 subtype C-infected blood donors. J Virol 83: 8925–8937. http://dx.doi.org/10.1128/JVI.00758-09. 56. Moore PL, Gray ES, Sheward D, Madiga M, Ranchobe N, Lai Z, Honnen WJ, Nonyane M, Tumba N, Hermanus T, Sibeko S, Mlisana K, Abdool Karim SS, Williamson C, Pinter A, Morris L. 2011. Potent and broad neutralization of HIV-1 subtype C by plasma antibodies targeting a quaternary epitope including residues in the V2 loop. J Virol 85:3128– 3141. http://dx.doi.org/10.1128/JVI.02658-10. 57. Moore PL, Gray ES, Wibmer CK, Bhiman JN, Nonyane M, Sheward DJ, Hermanus T, Bajimaya S, Tumba NL, Abrahams MR, Lambson BE, Ranchobe N, Ping L, Ngandu N, Abdool Karim Q, Abdool Karim SS, Swanstrom RI, Seaman MS, Williamson C, Morris L. 2012. Evolution of an HIV glycan-dependent broadly neutralizing antibody epitope through Patil et al. 3456 jvi.asm.org April 2016 Volume 90 Number 7Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom
  • 12. immune escape. Nat Med 18:1688–1692. http://dx.doi.org/10.1038/nm .2985. 58. Moore PL, Ranchobe N, Lambson BE, Gray ES, Cave E, Abrahams MR, Bandawe G, Mlisana K, Abdool Karim SS, Williamson C, Morris L. 2009. Limited neutralizing antibody specificities drive neutralization es- cape in early HIV-1 subtype C infection. PLoS Pathog 5:e1000598. http: //dx.doi.org/10.1371/journal.ppat.1000598. 59. Overbaugh J, Morris L. 2012. The antibody response against HIV-1. Cold Spring Harb Perspect Med 2:a007039. 60. Rademeyer C, Moore PL, Taylor N, Martin DP, Choge IA, Gray ES, Sheppard HW, Gray C, Morris L, Williamson C. 2007. Genetic charac- teristics of HIV-1 subtype C envelopes inducing cross-neutralizing anti- bodies. Virology 368:172–181. http://dx.doi.org/10.1016/j.virol.2007.06 .013. 61. Rong R, Bibollet-Ruche F, Mulenga J, Allen S, Blackwell JL, Derdeyn CA. 2007. Role of V1V2 and other human immunodeficiency virus type 1 envelope domains in resistance to autologous neutralization during clade C infection. J Virol 81:1350–1359. http://dx.doi.org/10 .1128/JVI.01839-06. 62. Rong R, Li B, Lynch RM, Haaland RE, Murphy MK, Mulenga J, Allen SA, Pinter A, Shaw GM, Hunter E, Robinson JE, Gnanakaran S, Derdeyn CA. 2009. Escape from autologous neutralizing antibodies in acute/early subtype C HIV-1 infection requires multiple pathways. PLoS Pathog 5:e1000594. http://dx.doi.org/10.1371/journal.ppat.1000594. 63. Ringe R, Das L, Choudhary I, Sharma D, Paranjape R, Chauhan VS, Bhattacharya J. 2012. Unique C2V3 sequence in HIV-1 envelope ob- tained from broadly neutralizing plasma of a slow progressing patient conferred enhanced virus neutralization. PLoS One 7:e46713. http://dx .doi.org/10.1371/journal.pone.0046713. 64. Ringe RP, Sanders RW, Yasmeen A, Kim HJ, Lee JH, Cupo A, Korzun J, Derking R, van Montfort T, Julien JP, Wilson IA, Klasse PJ, Ward AB, Moore JP. 2013. Cleavage strongly influences whether soluble HIV-1 envelope glycoprotein trimers adopt a native-like conformation. Proc Natl Acad Sci U S A 110:18256–18261. http://dx.doi.org/10.1073/pnas .1314351110. 65. Chung NP, Matthews K, Kim HJ, Ketas TJ, Golabek M, de Los Reyes K, Korzun J, Yasmeen A, Sanders RW, Klasse PJ, Wilson IA, Ward AB, Marozsan AJ, Moore JP, Cupo A. 2014. Stable 293 T and CHO cell lines expressing cleaved, stable HIV-1 envelope glycoprotein trimers for struc- tural and vaccine studies. Retrovirology 11:33. http://dx.doi.org/10.1186 /1742-4690-11-33. 66. Feng Y, McKee K, Tran K, O’Dell S, Schmidt SD, Phogat A, Forsell MN, Karlsson Hedestam GB, Mascola JR, Wyatt RT. 2012. Biochemically defined HIV-1 envelope glycoprotein variant immunogens display differ- ential binding and neutralizing specificities to the CD4-binding site. J Biol Chem 287:5673–5686. http://dx.doi.org/10.1074/jbc.M111.317776. 67. Boliar S, Das S, Bansal M, Shukla BN, Patil S, Shrivastava T, Samal S, Goswami S, King CR, Bhattacharya J, Chakrabarti BK. 2015. An effi- ciently cleaved HIV-1 clade C Env selectively binds to neutralizing anti- bodies. PLoS One 10:e0122443. http://dx.doi.org/10.1371/journal.pone .0122443. 68. Ringe R, Thakar M, Bhattacharya J. 2010. Variations in autologous neutralization and CD4 dependence of b12 resistant HIV-1 clade C env clones obtained at different time points from antiretroviral naive Indian patients with recent infection. Retrovirology 7:76. http://dx.doi.org/10 .1186/1742-4690-7-76. 69. Sanders RW, Derking R, Cupo A, Julien JP, Yasmeen A, de Val N, Kim HJ, Blattner C, de la Pena AT, Korzun J, Golabek M, de Los Reyes K, Ketas TJ, van Gils MJ, King CR, Wilson IA, Ward AB, Klasse PJ, Moore JP. 2013. A next-generation cleaved, soluble HIV-1 Env trimer, BG505 SOSIP.664 gp140, expresses multiple epitopes for broadly neutralizing but not non-neutralizing antibodies. PLoS Pathog 9:e1003618. http://dx.doi .org/10.1371/journal.ppat.1003618. 70. Tomaras GD, Binley JM, Gray ES, Crooks ET, Osawa K, Moore PL, Tumba N, Tong T, Shen X, Yates NL, Decker J, Wibmer CK, Gao F, Alam SM, Easterbrook P, Abdool Karim S, Kamanga G, Crump JA, Cohen M, Shaw GM, Mascola JR, Haynes BF, Montefiori DC, Morris L. 2011. Polyclonal B cell responses to conserved neutralization epitopes in a subset of HIV-1-infected individuals. J Virol 85:11502–11519. http://dx .doi.org/10.1128/JVI.05363-11. 71. Patil S, Choudhary I, Chaudhary NK, Ringe R, Bansal M, Shukla BN, Boliar S, Chakrabarti BK, Bhattacharya J. 2014. Determinants in V2C2 region of HIV-1 clade C primary envelopes conferred altered neutraliza- tion susceptibilities to IgG1b12 and PG9 monoclonal antibodies in a con- text-dependent manner. Virology 462-463:266–272. 72. Teng G, Papavasiliou FN. 2007. Immunoglobulin somatic hypermuta- tion. Annu Rev Genet 41:107–120. http://dx.doi.org/10.1146/annurev .genet.41.110306.130340. 73. Gray ES, Moore PL, Bibollet-Ruche F, Li H, Decker JM, Meyers T, Shaw GM, Morris L. 2008. 4E10-resistant variants in a human immunodefi- ciency virus type 1 subtype C-infected individual with an anti-membrane- proximal external region-neutralizing antibody response. J Virol 82:2367– 2375. http://dx.doi.org/10.1128/JVI.02161-07. 74. Trkola A, Purtscher M, Muster T, Ballaun C, Buchacher A, Sullivan N, Srinivasan K, Sodroski J, Moore JP, Katinger H. 1996. Human mono- clonal antibody 2G12 defines a distinctive neutralization epitope on the gp120 glycoprotein of human immunodeficiency virus type 1. J Virol 70: 1100–1108. 75. Guttman M, Cupo A, Julien JP, Sanders RW, Wilson IA, Moore JP, Lee KK. 2015. Antibody potency relates to the ability to recognize the closed, pre-fusion form of HIV Env. Nat Commun 6:6144. http://dx.doi.org/10 .1038/ncomms7144. 76. Kong L, Lee JH, Doores KJ, Murin CD, Julien JP, McBride R, Liu Y, Marozsan A, Cupo A, Klasse PJ, Hoffenberg S, Caulfield M, King CR, Hua Y, Le KM, Khayat R, Deller MC, Clayton T, Tien H, Feizi T, Sanders RW, Paulson JC, Moore JP, Stanfield RL, Burton DR, Ward AB, Wilson IA. 2013. Supersite of immune vulnerability on the glycosy- lated face of HIV-1 envelope glycoprotein gp120. Nat Struct Mol Biol 20:796–803. http://dx.doi.org/10.1038/nsmb.2594. 77. Sok D, Doores KJ, Briney B, Le KM, Saye-Francisco KL, Ramos A, Kulp DW, Julien JP, Menis S, Wickramasinghe L, Seaman MS, Schief WR, Wilson IA, Poignard P, Burton DR. 2014. Promiscuous glycan site recognition by antibodies to the high-mannose patch of gp120 broadens neutralization of HIV. Sci Transl Med 6:236ra263. 78. Rolland M, Edlefsen PT, Larsen BB, Tovanabutra S, Sanders-Buell E, Hertz T, deCamp AC, Carrico C, Menis S, Magaret CA, Ahmed H, Juraska M, Chen L, Konopa P, Nariya S, Stoddard JN, Wong K, Zhao H, Deng W, Maust BS, Bose M, Howell S, Bates A, Lazzaro M, O’Sullivan A, Lei E, Bradfield A, Ibitamuno G, Assawadarachai V, O’Connell RJ, de Souza MS, Nitayaphan S, Rerks-Ngarm S, Robb ML, McLellan JS, Georgiev I, Kwong PD, Carlson JM, Michael NL, Schief WR, Gilbert PB, Mullins JI, Kim JH. 2012. Increased HIV-1 vaccine efficacy against viruses with genetic signatures in Env V2. Nature 490:417– 420. http://dx.doi.org/10.1038/nature11519. 79. Chakrabarti BK, Feng Y, Sharma SK, McKee K, Karlsson Hedestam GB, Labranche CC, Montefiori DC, Mascola JR, Wyatt RT. 2013. Robust neutralizing antibodies elicited by HIV-1 JRFL envelope glycoprotein trimers in nonhuman primates. J Virol 87:13239–13251. http://dx.doi.org /10.1128/JVI.01247-13. 80. Burton DR, Mascola JR. 2015. Antibody responses to envelope glycopro- teins in HIV-1 infection. Nat Immunol 16:571–576. http://dx.doi.org/10 .1038/ni.3158. 81. Pancera M, Wyatt R. 2005. Selective recognition of oligomeric HIV-1 primary isolate envelope glycoproteins by potently neutralizing ligands requires efficient precursor cleavage. Virology 332:145–156. http://dx.doi .org/10.1016/j.virol.2004.10.042. 82. Sok D, van Gils MJ, Pauthner M, Julien JP, Saye-Francisco KL, Hsueh J, Briney B, Lee JH, Le KM, Lee PS, Hua Y, Seaman MS, Moore JP, Ward AB, Wilson IA, Sanders RW, Burton DR. 2014. Recombinant HIV envelope trimer selects for quaternary-dependent antibodies targeting the trimer apex. Proc Natl Acad Sci U S A 111:17624–17629. http://dx.doi.org /10.1073/pnas.1415789111. HIV-1 Clade C Plasma Confers Cross-Clade Neutralization April 2016 Volume 90 Number 7 jvi.asm.org 3457Journal of Virology onMarch11,2016byguesthttp://jvi.asm.org/Downloadedfrom