Conformational Epitope-Specific Broadly ... - Journal of Virology

9 downloads 76 Views 4MB Size Report
Jan 13, 2016 - pcDNA5-FRT BG505 Furin A (10) was kindly provided by Prof John ...... DeCamp, P. B. Gilbert, B. Wood, D. C. Montefiori, J. M. Binley, G. M. ...
JVI Accepted Manuscript Posted Online 13 January 2016 J. Virol. doi:10.1128/JVI.03090-15 Copyright © 2016, American Society for Microbiology. All Rights Reserved.

1

Conformational Epitope-Specific Broadly Neutralizing Plasma Antibodies Obtained from

2

an HIV-1 Clade C Infected Elite Neutralizer Mediate Autologous Virus Escape through

3

Mutations in V1 Loop

4 5

Shilpa Patil11¶, , Rajesh Kumar1¶, , Suprit Deshpande1¶, Sweety Samal1, Tripti Shrivastava1,

6

Saikat Boliar1, Manish Bansal1, Nakul Kumar Chaudhary1, Aylur K. Srikrishnan2, Kailapuri G.

7

Murugavel2, Suniti Solomon2, Melissa Simek3, Wayne C. Koff 3, Rajat Goyal3, Bimal K.

8

Chakrabarti1, 3, Jayanta Bhattacharya1, 3, #

9 10

1. HIV Vaccine Translational Research Laboratory, Translational Health Science and

11

Technology Institute, Faridabad, Haryana- 121001, India

12

2. Y.R. Gaitonde Research and Care Center, Chennai, India

13

3. International AIDS Vaccine Initiative, New York, USA

14 15

Running title: HIV-1 clade C plasma confer cross clade neutralization

16 17

Key words: HIV-1, neutralizing antibody, envelope, plasma, clade C, protocol G, V1V2 loop

18

Abstract: 247 words

19

¶ Equal contribution

20

# Corresponding author

21

Tel: +91-01242867705

22

E-mail: [email protected] / [email protected]

23

1

24

Abstract

25

Broadly neutralizing antibodies isolated from infected patients who are elite neutralizers

26

have identified targets on HIV-1 envelope (Env) glycoprotein that are vulnerable to antibody

27

neutralization; however, it is not known whether infection established by majority of the

28

circulating clade C strains in Indian patients elicit neutralizing antibody responses against any of

29

the known targets. In the present study, we examined the specificity of a broad and potent cross

30

neutralizing plasma obtained from an Indian elite neutralizer infected with HIV-1 clade C. This

31

plasma neutralized 53/57 (93%) HIV pseudoviruses prepared with Env from distinct HIV clades

32

of different geographical origin. Mapping studies using gp120 core protein, single residue

33

knockout mutants and chimeric viruses revealed that G37080 BCN plasma lacks specificities to

34

the CD4 binding site, gp41 membrane proximal external region, N160, N332 glycans as well as

35

R166 and K169 in V1-V3 region and are known predominant targets for BCN antibodies.

36

Depletion of G37080 plasma with soluble trimeric BG505-SOSIP.664 Env (but neither with

37

monomeric gp120 nor with clade C MPER peptides), resulted in significant reduction of virus

38

neutralization, suggesting that G37080 BCN antibodies mainly target epitopes on cleaved

39

trimeric Env. Further examination of autologous circulating Envs revealed association of

40

mutation of residues in V1 loop that contributed in neutralization resistance. In summary, we

41

report identification of plasma antibodies from a clade C infected elite neutralizer that mediates

42

neutralization breadth via epitopes on trimeric gp120 not yet reported and confer autologous

43

neutralization escape via mutation of residues in V1 loop.

44

2

45

Importance

46 47

A preventive vaccine to protect against HIV-1 is urgently needed. HIV-1 envelope

48

glycoproteins are targets of neutralizing antibodies and represent a key component for

49

immunogen design. Mapping of epitopes on viral envelopes vulnerable for immune evasion will

50

aid in defining targets of vaccine immunogens. We identified novel conformational epitopes on

51

viral envelope targeted by broadly cross neutralizing antibodies elicited in natural infection in an

52

elite neutralizer infected with HIV-1 clade C. Our data extend our knowledge on neutralizing

53

epitopes associated with virus escape and would potentially contribute in immunogen design and

54

antibody based prophylactic therapy.

55

3

56 57

Introduction

58 59

Broadly neutralizing antibodies (BNAbs) target trimeric envelope glycoprotein (Env) spikes

60

of the Human Immunodeficiency Virus Type 1 (HIV-1). Characterization of the BNAbs has provided

61

key clues towards design and development of both prophylactic and therapeutic vaccines (7, 17, 30,

62

34, 35, 37). A small proportion of individuals chronically infected with HIV-1 develop BNAbs (5,

63

12, 21, 38, 52, 62, 66, 69) and isolation of several broad and potent neutralizing monoclonal

64

antibodies (bNAb) from such individuals with distinct molecular specificities to viral envelope (Env)

65

protein are reported (18, 31, 32, 64, 74, 76, 77, 79, 81). The cross neutralizing serum antibodies

66

obtained from such individuals (also referred to as ‘elite neutralizers’) with considerable breadth

67

target epitopes on structurally conserved regions of Env such as CD4 binding site (CD4bs) (11, 33,

68

65, 79), V1V2 including glycan moieties (39, 50, 74, 76), gp120-gp41 interface (3, 64) and the

69

membrane proximal external regions (MPER) (31, 45, 46, 83). Several studies have indicated that the

70

variable regions within the HIV-1 gp120 contain epitopes targeted by the autologous as well as

71

BNAbs (8, 14, 16, 28, 41, 59, 60, 73). Recently the V1V2 region has been linked to development

72

of broadly cross neutralizing (BCN) antibodies (16, 78) and the residues between 160 and 172

73

(notably R166S/K or K169A) in V1V2 have been demonstrated to be associated with virus

74

escape to autologous antibody response (16). Recent studies have further indicated that BCNAb

75

development in vivo is associated with antibody affinity maturation and co-evolution of virus

76

resulting in a considerable degree of somatic hypermutations (1, 13, 15, 16, 20, 29, 40, 63, 65, 75, 76,

77

80-82). Such information is crucial for design and development of suitable Env based immunogen

78

capable of eliciting broad and potent cross neutralizing antibodies through vaccination.

79 4

80

While a number of studies on the molecular specificities of broadly neutralizing antibodies

81

obtained from African clade C infected individuals have been reported (2, 21-23, 25, 26, 41-44, 47,

82

51, 57, 58), knowledge on immune evasion in Indian clade C infected elite neutralizers is very

83

limited (53).

84

In the present study, we examined plasma samples obtained from two hundred asymptomatic

85

and anti-retroviral therapy (ART) naïve Indian HIV-infected donors and identified plasma with cross

86

neutralizing antibodies. The molecular specificities of plasma antibodies obtained from an HIV-1

87

clade C infected elite neutralizer was characterized in detail that displayed exceptional neutralization

88

breadth across clades of different geographical origins. Interestingly, we found that neutralization

89

breadth was associated with presence of unique epitopes on the trimeric gp120.

90 91

5

92 93

Materials and Methods

94 95

Ethics statement.

96

The blood samples were collected under The IAVI Protocol G study from slow progressing anti-

97

retroviral therapy (ART) naïve HIV-1 positive donors from Nellore District of the state of

98

Andhra Pradesh, Southern India by trained clinicians at the YRG Care hospital following

99

approval and clearance from the Institutional Review Board (IRB) and the Ethics Committee.

100

The serum and plasma samples collected were shipped to the HIV Vaccine Translational

101

Research Laboratory, Translational Health Science and Technology Institute for further

102

assessment and research on the neutralizing antibody response.

103 104

Plasmids, viruses, antibodies, proteins and cells.

105

Plasmids encoding HIV-1 envelopes representing distinct clades are shown in Table 1.

106

Monoclonal antibodies used in the study and TZM-bl cells were procured from the NIH AIDS

107

Research Reagents Reference program and from the IAVI Neutralizing Antibody Consortium

108

(NAC). 293T cells were purchased from the American Type Culture Collection (ATCC).

109

Plasmid DNA encoding BG505-SOSIP.664-D7324, its purified cleaved trimeric protein (55) and

110

pcDNA5-FRT BG505 Furin A (10) was kindly provided by Prof John Moore, Weill Cornell

111

Medical College, New York. Purified gp120 TripleMut core protein (19) was obtained from Prof

112

Richard Wyatt, The Scripps Research Institute through the IAVI Neutralizing Antibody

113

Consortium (NAC). HIV-2 7312A and its chimeric constructs were provided by Prof Lynn

114

Morris, NICD, Johannesburg.

115 6

116

Purification of monomeric and trimeric Env proteins.

117

Codon optimized gp120 plasmid encoding clade C 4-2.J41 (4, 54) gp120 was cloned in pcDNA

118

3.1/V5-His-TOPO vector and transfected in 293T cells using polyethyleneimine (PEI).

119

Supernatants containing soluble gp120 were filtered through 0.45µm filter and subsequently

120

purified using Ni-NTA agarose matrix (Qiagen Inc.) by elution with phosphate buffered saline

121

(PBS) containing 300mM imidazole (pH 8.0). The purified monomeric gp120 protein was

122

extensively dialyzed with PBS (pH 7.4), concentrated using Amicon® Ultracentrifugal filers

123

(Millipore Inc.) with 30KDa cut off and stored in -80°C until further use.

124

The trimeric BG505-SOSIP.664 protein was purified using 293F cells essentially as

125

described by Sanders et al (61). Briefly, the 293F cells were transfected with plasmid DNA

126

encoding both BG505-SOSIP.664 gp140 envelope and furin (10). Supernatant containing soluble

127

BG505-SOSIP.664 gp140 was harvested 72 to 96 hours post transfection, filtered and passed

128

through a lectin agarose column obtained from Galanthus nivalis (Sigma Inc.). The

129

nonspecifically bound proteins were then washed in PBS (pH 7.4) supplemented with 0.5 M

130

NaCl. The bound proteins were then eluted using 0.5 M methyl alpha-D-manno-pyranoside,

131

extensively dialyzed with 1X PBS and concentrated. BG505-SOSIP.664 was further purified by

132

Sephadex G-200 size exclusion chromatography (AKTA, GE). Trimeric protein fractions were

133

collected, pooled, quality assessed by running in blue native polyacrylamide gel electrophoresis

134

(BN-PAGE) and favorably assessed for their ability to bind to only neutralizing and not to non-

135

neutralizing and MPER directed monoclonal antibodies as described elsewhere (55) by ELISA.

136 137

Depletion of plasma antibodies by monomeric gp120 and trimeric gp140 Env proteins.

138

Purified soluble monomeric 4-2.J41 gp120 and trimeric BG505 SOSIP.664 proteins in addition

139

to the MPER peptide (C1C; encoding clade C sequence) (71) were used for depletion of plasma 7

140

antibodies, where purified proteins were covalently coupled to the MyOne Tosylactivated

141

Dynabeads (Life Technologies Inc.) according to the manufacturer’s protocol. Briefly, 30 mg of

142

beads were used to couple with 1 mg of both monomeric and trimeric Env proteins in coupling

143

buffer (0.1M NaBO4, 1M (NH4)2SO4; pH 9.4) overnight at 37ºC for 16-24 hrs. Proteins bound to

144

magnetic beads were separated from unbound using a DynaMag™ 15 magnet (Life

145

Technologies, Inc.). Beads bound to Env proteins were next incubated with blocking buffer [PBS

146

(pH 7.4), 0.1% bovine serum albumin (BSA; Sigma) and 0.05% Tween 20] at 37ºC to block the

147

unbound sites. The antigenic integrity of both 4-2.J41 monomeric gp120 and BG505-SOSIP.664

148

bound to the beads were assessed for their ability to bind VRC01 and 4E10 MAbs (for

149

monomeric gp120) and PGT121, F105 and 4E10 MAbs (for BG505-SOSIP.664) by flow

150

cytometry (FACS Canto, Becton and Dickinson, Inc.).

151

For depletion studies, G37080 plasma was diluted to 1:50 in DMEM containing 10%

152

Fetal Bovine Sera (FBS) and 500 µl of diluted plasma was incubated with 20µl of beads at room

153

temperature for 45 minutes. Unbound plasma antibodies were separated from ones those are

154

bound to protein coated beads using a DynaMag™ 15 magnet as described above. This step was

155

repeated 4-5 times for depletion of plasma antibodies by monomeric gp120 and 10-12 times in

156

case of BG505-SOSIP.664 coated beads respectively. As a negative control, G37080 plasma

157

antibodies were depleted with uncoated beads in parallel. In addition to ELISA, percent

158

depletion of G37080 plasma antibodies was assessed by examining the sequential decrease in

159

binding of protein coated beads with depleted plasma antibodies by FACS. PGT121 MAb was

160

taken as a positive control for checking depletion by BG505-SOSIP.664 trimeric Env.

161 162

8

163

gp120 and gp140 ELISA

164

For gp120 ELISA, high binding polystyrene microtiter plate (Nunc, Inc.) was coated with 100μl

165

of monomeric 4-2.J41 gp120 (1μg/ml) in binding buffer comprising 0.1 M NaHCO3 (pH 8.6)

166

and incubated overnight at 4ºC. gp120 bound plate was washed once with 1X PBS (pH 7.4) and

167

blocked with 5% non-fat milk for 90 min at 37ºC. The plate was then washed three times with

168

1X PBS, followed by addition of 100μl of MAbs as well as the depleted and undepleted plasma

169

antibodies at different dilutions and incubated for 1hr at room temperature. The wells of the

170

ELISA plate were washed four times with PBS containing 0.1% Tween 20 (PBST) followed by

171

addition of 100µl of 1:3000 diluted HRP-conjugated anti-human IgG (Jackson Immunoresearch,

172

Inc.) and further incubated for 45 min at room temperature. Unbound conjugates were removed

173

by washing with PBST and color developed by addition of 100μl of 3, 3’, 5, 5’-

174

tetramethylbenzidine (TMB) (Life Technologies, Inc.) substrate was added. Absorbance was

175

measured at 450 nm in a spectrophotometer.

176

Binding of antibodies to BG505-SOSIP.664-D7324 trimeric protein was assessed

177

essentially as described by Sanders et al (61) in a sandwich ELISA. Briefly, high binding

178

microtiter plate (Nunc, Inc.) was first coated with D7324 antibody at 10μg/ml (Aalto Bio

179

reagents, Dublin, Ireland) followed by blocking extra unbound sites with 5% non-fat milk for 90

180

min at 37ºC. 100 µl of BG505.664-D7324 trimeric protein (300ng/ml) was then added and

181

incubated for 45 mins at room temperature. The extent of binding of G37080 plasma antibodies

182

compared to known neutralizing monoclonal antibodies were assessed by addition of primary

183

and HRP-conjugated secondary anti-human antibody as described above.

184 185

9

186

Neutralization assay.

187

Neutralization assays were carried out using TZM-bl cells as described before (54). Briefly, Env-

188

pseudotyped viruses were incubated with varying dilutions of depleted plasma antibodies and

189

incubated for an hour at 37°C CO2 incubator under humidified condition and subsequently 1 X

190

104 TZM-bl cells were added into the mixture in presence of 25 μg/ml DEAE-dextran (Sigma,

191

Inc.). The plates were further incubated for 48 hours and the degree of virus neutralization was

192

assessed by measuring relative luminescence units (RLU) in a Luminometer (Victor X2,

193

PerkinElmer Inc.).

194 195

Amplification, cloning and mutagenesis of autologous HIV-1 envs.

196

Autologous complete env genes were obtained from G37080 plasma as described previously

197

with slight modification (54). Briefly, viral RNA were extracted using High Pure viral RNA kit

198

(Roche Inc.) following manufacturer’s protocol and cDNA prepared by RT-PCR using

199

Superscript-III first strand synthesis kit (Invitrogen Inc.). rev-gp160 env genes were amplified

200

using a Phusion hi fidelity DNA polymerase (New England Biolabs Inc.). The gp160 amplicons

201

were purified and ligated into pcDNA 3.1/V5-His-TOPO (Invitrogen Inc.) vector. Chimeric Envs

202

were prepared by overlapping PCR and point substitutions were made by Quikchange II kit

203

(Agilent technologies Inc.) following manufacturer’s protocol and as described previously (49).

204 205

Preparation of envelope pseudotyped viruses.

206

Pseudotyped viruses were prepared by co-transfection of envelope expressing plasmid with env-

207

deleted HIV-1 backbone plasmid (pSG3ΔEnv) into 293T cells in 6-well tissue culture plates

208

using FuGENE6 Transfection kit (Promega Inc.). Cell supernatants containing pseudotyped

209

viruses were harvested 48 hours post-transfection and then stored at -80°C until further use. The 10

210

infectivity assays were done in TZM-bl cells (1 X 105cells/ml) containing DEAE-Dextran (25

211

μg/ml) in 96-well microtiter plates and the infectivity titers were determined by measuring the

212

luciferase activity using Britelite luciferase substrate (Perkin Elmer Inc.) with a Victor X2

213

Luminometer (Perkin Elmer Inc.).

214

11

215

Results

216 217

Identification of an elite neutralizer with HIV-1 clade C infection whose plasma showed

218

exceptional neutralization breadth.

219

The present study under The IAVI Protocol G was designed (i) to screen and identify

220

plasma antibodies obtained from chronically infected Indian donors with HIV-1 clade C with

221

substantial breadth towards neutralizing cross clade HIV-1 primary variants and (ii) to elucidate

222

their molecular specificities associated with neutralization breadth. Our hypothesis was that the

223

genetic distinctness of clade C viruses of Indian and non-Indian origin, as well due to likely

224

differences in host genetics between populations with differences in their ancestral origin

225

associated with modulation of humoral immune responses, the specificities of antibodies

226

developed in vivo associated with neutralization breadth and potency would be different.

227

Through screening of two hundred plasma samples obtained from chronically-infected

228

ART naïve Indian patients against a panel of 57 pseudoviruses containing Envs of distinct clades

229

and geographical origins (Figure 1A), we identified one donor (G37080), whose plasma showed

230

exceptional neutralization breadth. Donor G37080 serum neutralized >90% of the 57 different

231

pseudoviruses tested with median ID50 value of 533.03 (Table 1, Figure 1B).

232

Follow up plasma sample from this donor (G37080) was subsequently obtained after

233

eight months to assess whether the neutralization breadth and potencies along with their

234

molecular specificities were retained and/or improved, expecting that during the course of

235

disease, breadth and potency of neutralizing antibodies broadens through somatic

236

hypermutations (70) and/or clonal selection processes. As shown in Figure 1B and Table 1,

237

follow up plasma antibodies of G37080 donor (referred to as Visit-2 samples) were found to

238

exhibit comparable neutralization breadth to that of visit 1 plasma. Overall, G37080 BCN plasma

12

239

was found to potently neutralize pseudoviruses containing Indian clade C Env with a

240

neutralization score of 2.5 (66). Furthermore, the neutralization sensitivity of Env-pseudotyped

241

viruses was found to be correlated with the serum IgG (data not shown), suggesting that the

242

broad neutralization was associated with IgG-specific response. Taken together, our data indicate

243

that a strong humoral immune response to HIV-1 was mounted in G37080 donor and was

244

maintained overtime.

245 246

Evidence that G37080 BCN plasma antibodies do not target epitopes in CD4bs, MPER and

247

known glycan and non-glycan residues in variable domains of Env

248

First, we examined whether the G37080 BCN plasma contains antibodies directed to

249

CD4bs on Env. Plasma samples obtained from both visits were pre-treated with 25µg/ml of

250

TripleMut core protein (19), which was a concentration that we found to inhibit neutralization of

251

25711-2.4 pseudovirus by VRC01 mAb by >95%. Pre-treated plasma was subsequently used to

252

neutralize pseudovirus 25711-2.4 Env, and as shown in Figure 2, no perturbation of G37080

253

neutralizing activity was observed against pseudovirus 25711-2.4. A similar observation was

254

made when these plasma antibodies were pre-treated with RSC3 core protein (79). In addition,

255

the G37080 BCN plasma antibodies were found to efficiently neutralize IgG1b12 and VRC01

256

resistant viruses (data not shown). Our data indicated that the G37080 BCN plasma antibodies do

257

not contain CD4bs directed neutralizing antibodies.

258

To elucidate whether the BCN plasma antibodies are directed to MPER in gp41, we used

259

HIV-2/HIV-1 chimeric viruses (24) that expressed minimal residues of HIV-1 MPER containing

260

epitopes required for MPER directed mAbs such as 2F5, 4E10, Z13e and 10E8. As shown in

261

Table 2, the G37080 BCN plasma from both the visits was found to show modest neutralization

262

of HIV-2 expressing HIV-1 clade C MPER (7312-C1C) with ID50 values of 306.42 and 371.02, 13

263

respectively. We also found that depletion of G37080 plasma with a clade C MPER peptide

264

(C1C) completely abolished the sensitivity of 7312A-C1C virus to G37080 plasma (Table 3).

265

Our data suggest that although the G37080 BCN plasma neutralized 7312-C1C, presence of

266

MPER directed antibodies was not associated with neutralization breadth.

267

We next investigated whether the plasma antibodies of the donor G37080 target residues

268

in variable loops, particularly in V1V2 and V3 region that have been shown in several studies as

269

epitopes targeted by BCN antibodies on HIV-1 Env. First, we tested the extent of neutralization

270

by G37080 BCN plasma antibodies of Env pseudotyped viruses lacking glycans at 160 (N160)

271

and at 332 (N332) position in the V2 region and V3 base respectively, and also R166 and K169

272

in V2 region, which are major targets of recently identified broad and potent neutralizing

273

monoclonal antibodies. In order to test this, two clade C Envs (25711-2.4 and CAP239.G3)

274

containing N160A and N332A substitutions were tested and as shown in Table 2. Our data

275

indicate that the pseudoviruses containing Env expressing N160 or N332 substitutions have

276

identical sensitivities to G37080 plasma antibodies. Similar observations were found with

277

R166A and K169A in 93IN905 Env backbone. Taken together, our observations indicate that

278

G37080 BCN plasma antibodies did not utilize these residues in V2 and V3 regions for

279

neutralization breadth; which have been identified as important epitopes recognized by broadly

280

neutralizing antibodies elicited in clade C infection described before (16, 42, 78).

281 282

Association of neutralization breadth of G37080 plasma with recognition of conformational

283

epitopes on cleaved trimeric Env but not with that in monomeric gp120 or MPER.

284

In order to examine whether broad neutralization conferred by the G37080 plasma

285

antibodies was through recognition of epitopes on monomeric gp120 or cleaved near native Env

286

trimers, we tested binding of G37080 serum IgG to monomeric 4-2.J41 gp120 and soluble gp140 14

287

(BG505-SOSIP.664) by ELISA. We found that in addition to the monomeric 4-2.J41 gp120

288

(Figure 3A), G37080 serum polyclonal IgG was found to efficiently bind to the BG505

289

SOSIP.664-D7324 soluble trimeric Env (Figure 3B), indicating that the G37080 plasma

290

primarily contains neutralizing antibodies that targets epitopes on cleaved Env trimers.

291

We next examined whether binding of the G37080 plasma antibodies to epitopes on

292

cleaved BG505-SOSIP.664 trimeric envelope was associated with neutralization breadth. For

293

this, we tested the ability of G37080 plasma antibodies depleted with both monomeric and

294

trimeric Envs as well as with MPER peptides to neutralize a set of Env-pseudotyped viruses,

295

which were found to be sensitive to this particular plasma sample. Purified 4-2.J41 monomeric

296

gp120, BG505-SOSIP.664 trimeric gp140 and C1C MPER peptide bound to the magnetic beads

297

were used to deplete G37080 plasma antibodies as described in the ‘Materials and Methods’. The

298

depleted BCN G37080 antibodies were first assessed for their binding to 4-2.J41 gp120

299

monomers, BG505-SOSIP.664-D7324 and C1C peptide in comparison to undepleted plasma

300

antibodies by ELISA. As shown in Figure 3C and 3D, G37080 plasma depleted with monomeric

301

gp120 and trimeric gp140, respectively, had significantly reduced binding activity against

302

respective soluble proteins. Similar observation was made with MPER peptide (data not shown).

303

The depleted plasma antibodies were subsequently assessed for neutralization activity using a

304

panel of twelve Env pseudotyped viruses that were susceptible to untreated G37080 plasma

305

antibodies as mentioned above. As shown in Table 3, depletion with 4-2.J41gp120 monomer and

306

C1C peptide did not show any change in neutralization breadth of G37080 plasma antibodies,

307

while depletion with BG505-SOSIP.664 showed a significant reduction in virus neutralization. A

308

similar observations were made with the BG505-SOSIP.664 depleted PGT121 and C1C peptide

309

depleted 4E10 MAbs which lost the ability to efficiently neutralize Env-pseudotyped viruses 15

310

(16055 and ZM233.6) and HIV-2/HIV-1 (7312A-C1C) chimeric virus compared to their

311

undepleted counterparts (data not shown); thus validating our data. Interestingly, C1C peptide

312

depleted G37080 plasma failed to neutralize HIV-2/HIV-1 (7312A-C1C) chimeric virus

313

indicating that the presence of residual traces of MPER directed antibodies (as shown in Table 2)

314

are not responsible for neutralization breadth. Furthermore, examination of chimeric Envs

315

prepared between the sensitive (25711-2.4) and resistant (16055-2.3 and CAP45.G3) Envs

316

indicated that the BCN G37080 plasma antibodies predominantly target epitopes in the V1V2

317

region (Table 4) in gp120. Our data clearly indicate a correlation between neutralization breadth

318

and binding of the G37080 BCN plasma antibodies to the conformational epitopes on cleaved

319

trimeric gp120, likely in the V1V2 region; however we do not rule out the possibility that this

320

BCN plasma targets other discontinuous epitopes in the gp120, but not in MPER.

321 322

Mutations in V1 region confer resistance of autologous viruses to the G37080 plasma

323

antibodies

324

In order to decipher the specificity of the G37080 plasma antibodies, we examined the

325

degree of susceptibility of pseudoviruses prepared using env genes amplified from

326

contemporaneous autologous G37080 plasma obtained at the baseline and follow ups visits. As

327

shown in Figure 4A, both the Envs obtained from visit 2 plasma (HVTR-PG80v2.eJ38 and

328

HVTR-PG80v2.eJ41) were found to be resistant to its contemporaneous plasma antibodies,

329

while Envs obtained from visit 1 plasma (HVTR-PG80v1.eJ7 and HVTR-PG80v1.eJ19) were

330

found to be modestly sensitive to visit 2 autologous G37080 plasma antibodies. To facilitate

331

mapping G37080 BCN antibody specificity, we prepared chimeric Envs between a sensitive

332

(HVTR-PG80v1.eJ7 and HVTR-PG80v1.eJ19) and the resistant (HVTR-PG80v2.eJ38)

16

333

autologous Envs by first swapping the V1V2 regions as their amino acid sequences differed

334

maximally in this region (Figure 4B). As shown in Table 4, substitution of V1V2 sequence of

335

HVTR-PG80v1.eJ7 and HVTR-PG80v1.eJ19 into HVTR-PG80v2.eJ38 conferred Env-

336

pseudotyped viruses expressing HVTR-PG80v2.eJ38 Env with enhanced sensitivity to G37080

337

visit 2 plasma antibodies by >25 and >12-folds respectively. Conversely, the neutralization

338

susceptibilities of the Env-pseudotyped viruses expressing HVTR-PG80v1.eJ7 and HVTR-

339

PG80v1.eJ19, which contained HVTR-PG80v2.eJ38 V1V2 sequence corresponding to visit 2

340

G37080 plasma, were found to be reduced by >45 and >23 folds respectively. We noted that

341

substitution of regions other than V1V2 loop in the autologous Env did not confer any change in

342

neutralization sensitivity (Table 4). To further narrow down residues in V1V2 loop, associated

343

with neutralization sensitivity and resistance of autologous Envs, chimeric Envs and point

344

mutants were prepared and tested for their degree of modulation in susceptibility to autologous

345

G37080 plasma obtained from second visit. As shown in Table 4, we found that the V1 sequence

346

but not the V2 sequence of the sensitive Envs (HVTR-PG80v1.eJ7 and HVTR-PG80v1.eJ19)

347

when transferred to the resistant HVTR-PG80v2.eJ38 Env, increased sensitivity to G37080 BCN

348

plasma antibodies by >50 and >37 folds respectively. In agreement with this result, V1 of

349

HVTR-PG80v2.eJ38 when transferred into the sensitive Envs stated above, increased

350

neutralization resistance by >27 and >28 folds respectively to the G37080 visit 2 BCN plasma

351

antibodies. We observed that removal of a glycan at the 140 position in V1 (T140D) in the

352

HVTR-PG80v1.eJ19 mediated enhanced sensitivity of this Env to G37080 plasma by 2.64 fold

353

(Table 4). Concurrent to this observation, we found that insertion of V1 region of PG80v1.eJ19

354

with T140D substitution in PG80v2.eJ38 Env exhibited enhanced susceptibility when compared

355

with that of PG80v2.eJ38 Env chimera containing PG80v1.eJ19 V1 loop as shown in Table 4.

17

356

Our data indicate that N138 glycan potentially mask the PG80v1.eJ19 Env from being efficiently

357

neutralized by the autologous plasma compared to that of its contemporaneous counterpart

358

PG80v1.eJ7 Env. Fine scanning of V1 regions of the autologous Envs further revealed that N133

359

glycan motif and P147 residues in the PG80v2.eJ38 Env played significant role in neutralization

360

resistance to G37080 BCN autologous plasma antibodies (Figure 4B). Interestingly, all the V1

361

chimeras as well as the point mutants showed comparable sensitivities to PG9 MAb as compared

362

to their wild types (Table 5), indicating that the shift in neutralization susceptibilities were not

363

due to change in Env conformation. Moreover, we noted that both the sensitive and the resistant

364

autologous Envs contain T332 in the V3 base, clearly indicating that absence of N332 was not

365

associated with resistance to autologous neutralization. Similar observations were made with

366

respect to N160, R166 and K169 amino acid residues further consolidating that the neutralization

367

conferred by G37080 BCN plasma antibodies was not associated with antibody targeting these

368

epitopes in autologous Envs and likely for all the Envs tested against G37080 plasma antibodies.

369

18

370

Discussion

371 372

Identification of the molecular specificities of antibodies elicited in natural infection and

373

that mediate neutralization breadth and potency is key in design and development of suitable Env

374

based immunogen capable of eliciting similar antibody response upon vaccination. In the present

375

study, we characterized the molecular specificity of plasma antibodies obtained from an Indian

376

elite neutralizer (G37080) infected with HIV-1 clade C that displayed exceptional cross

377

neutralization of different clades of distinct geographical origins. The G37080 plasma was found

378

to contain the most broad and potent cross neutralizing antibodies amongst the two hundred

379

plasma samples obtained from Indian patients chronically infected with HIV-1. Plasma samples

380

collected from the G37080 donor at two time points at eight months apart showed similar

381

neutralization breadth with modest increase in potency in the follow up visit, indicating

382

association of sustained maturation of antibody producing B cells in this individual.

383

Since polyclonal plasma antibodies are not suitable for epitope mapping, we examined

384

the specificity of the G37080 BCN plasma by making use of mutant viruses with specific point

385

substitution of known neutralizing epitopes with non-specific amino acids and via depletion with

386

monomeric and trimeric Envs in addition to MPER peptide. The G37080 plasma antibodies did

387

not show dependence to the N160/K169 and N332 epitopes in V2 apex and V3 base respectively

388

Our data also is consistent with the target epitopes of the G37080 BCN antibodies being distinct

389

from those which are recognized by 2G12 (72), PGT121-128 (32) and PGT130-131, 135 (75)

390

(e.g., residues at the following positions: 295, 297, 301, 332, 334, 386, 388, 392, 394, 448, 450),

391

thus BCN G37080 antibodies appear to target a new epitope. Our data highlighting N332

392

independent development of neutralizing antibodies in a clade C infected donor G37080, also

393

differs from recent findings (27, 36, 43, 67) associating N332 with development of broad and 19

394

potent neutralizing antibody especially in clade C infection noted in African donors. Moreover,

395

recent studies indicating the role of K169 as a target of BCN antibodies obtained from a clade C

396

infected South African donor (42, 78) and the observation that vaccine-induced protection in the

397

RV144 vaccine trial was associated with antibodies targeting epitopes including K169 in V2

398

apex (39, 56) prompted us to examine whether broad neutralization of the G37080 plasma

399

antibodies was also dependent on K169 epitope. In our present study, the neutralization potency

400

of G37080 not only was unaffected by N160A/K169A knockout mutations but we also observed

401

that both sensitive and resistant autologous Envs obtained from both visits contain N160 and

402

K169 in the V2 region. Hence, owing to lack of association of neutralization breadth of the

403

G37080 BCN antibodies with N160, K169 and N332 dependences, our study further highlighted

404

that there is a likelihood of differences in development pathway of elicitation of broadly

405

neutralizing antibodies in individuals infected with HIV-1 clade C particularly those with

406

ethnically distinct.

407

Wibmer et al (78) recently demonstrated association between evolution of a broadly

408

neutralizing antibody response in a clade C infected donor with shifts in antibody specificities

409

from recognition of epitopes in V2 to the CD4bs. In the present study, the G37080 neutralizing

410

plasma antibodies obtained from both visits were found not to be absorbed out by the TripleMut

411

(9, 19) as well as the RSC3 (79) core proteins, which effectively absorb antibodies directed to the

412

CD4bs. This result indicates a lack of development of CD4bs directed neutralizing antibodies

413

during the disease course in G37080 donor. Additionally, absence of MPER directed antibodies

414

in G37080 plasma were found, although a negligible antibody titer (1:300 reciprocal dilutions) to

415

the HIV2/HIV1 (C1C) chimera was observed with both visit plasma samples. However, the

416

neutralization breadth of the G37080 plasma was not found to be associated with presence of

20

417

MPER directed antibody. Nonetheless, we do not rule out the possibility that in further course of

418

infection, this donor would possibly be able to develop MPER directed antibodies.

419

Recent studies have shown that neutralizing antibodies that targets conformational

420

epitopes binds exclusively to the cleaved near native trimeric Envs (6, 18, 48, 55). In the present

421

study, we found that absorption of G37080 plasma antibodies to soluble trimeric BG505-

422

SOSIP.664 Env was associated with depletion of neutralizing activity in G37080 BCN plasma.

423

However, we do not rule out the possibility of presence of 39F, 19b and 14e like non-

424

neutralizing antibodies that were reported to bind to BG505-SOSIP.664 trimeric Env (61). Our

425

findings indicate that the G37080 BCN antibodies target conformational epitopes in gp120. Our

426

observation also highlights that native like trimeric Envs such as BG505-SOSIP.664 can be

427

utilized in selecting antigen specific memory B cells as reported earlier (68) from G37080 donor

428

towards isolation of MAb correlating with broad neutralization displayed by the plasma

429

antibodies.

430

We made use of env clones obtained from autologous G37080 plasma from both the time

431

points to narrow down the fine specificity of the G37080 BCN plasma antibodies. By examining

432

chimeric Envs and mutant viruses we identified key residues in the V1 loop associated with

433

neutralization resistance. Interestingly, the Env chimera and mutant viruses showed comparable

434

susceptibility to PG9 MAb compared to their respective wild type Envs, indicating that they did

435

not alter Env conformation. We identified a glycan at the 133 position and a proline residue at

436

the 147 position within V1 loop of the resistant Env (PG80v2.eJ38) that were found to be

437

associated with neutralization escape, which indicated that these are contact sites for the G37080

438

BCN plasma antibodies. From our study we thus conclude that changes in V1 loop sequence are

439

associated with escape of autologous viruses to the BCN G37080 plasma. Additionally,

21

440

examination of degree of susceptibilities of pseudoviruses expressing chimeric heterologous

441

Envs to the G37080 plasma revealed that the BCN plasma antibodies predominantly target

442

epitopes in V1V2 region in gp120. However, we do not rule out the possibility of contribution of

443

other discontinuous epitopes in gp120 in mediating neutralization breadth. Isolation and

444

identification of monoclonal antibodies from this elite neutralizer donor (G37080) will help

445

precisely map specific epitope associated with neutralization breadth and potency.

446

In summary, we identified an HIV-1 infected elite neutralizer, whose plasma showed

447

exceptional neutralization breadth and provided evidence that it targets novel conformational

448

epitopes on trimeric Env predominantly in the V1V2 region not reported previously. Moreover,

449

neutralization resistance of the autologous Envs to G37080 plasma is associated with

450

substitutions of novel residues within V1 loop that form the key contact points of the BCN

451

plasma antibody. Identification of novel epitopes associated with broad neutralization of HIV-1,

452

in particular the majorly circulating clade C strains will significantly contribute in the efforts

453

towards effective immunogen design.

454 455

Funding information.

456

This study was made possible by the generous funding support of the American people through

457

the United States Agency for International Development (USAID) and support from the THSTI-

458

IAVI HIV vaccine design grant through the Department of Biotechnology, Govt. of India; partly

459

by a grant from Department of Science and Technology, Govt. of India (DST/INT/SAFR/Mega-

460

P3/2011 to Jayanta Bhattacharya) and partly by the DBT National Bioscience Research Award

461

grant (BT/HRD/NBA34/01/2012-13(iv) to Jayanta Bhattacharya). The funders had no role in

462

study design, data collection and interpretation, or the decision to submit the work for

463

publication. 22

464 465

Acknowledgements.

466

We thank all the Protocol G study participants registered with YRG Care, Chennai, and all the

467

research staffs at the Protocol G clinical center at the YRG Care, Chennai and all of the IAVI

468

Protocol G team members. We thank Dr. Albert Cupo, Prof. John P. Moore and the members the

469

SOSIP trimer HIVRAD team, Weill Cornell Medical College, New York for providing us with

470

BG505.SOSIP.664 plasmid DNA and purified protein. The following reagent was obtained

471

through the NIH AIDS Reagent Program, Division of AIDS, NIAID, NIH from Drs. John C.

472

Kappes and Xiaoyun Wu: pSG3Δenv. We thank Dr. David Montefiori, Prof Lynn Morris, Dr

473

Pascal Poignard, Dr. Richard Wyatt for making available many reagents used in our study. The

474

International AIDS Vaccine Initiative has filed a patent relating to the autologous HIV-1 clade C

475

envelope clones: U.S. Provisional Application no. 62/254,971, titled “HIV-1 clade C envelope

476

glycoproteins,” with inventors J. Bhattacharya, S. Deshpande, S. Patil, R. Kumar, B.K.

477

Chakrabarti. We sincerely thank Dr. Christopher Parks, IAVI Design and Development

478

Laboratory for providing valuable inputs in preparing the manuscript and we also thank Prof. G.

479

Balakrish Nair, Prof Sudhanshu Vrati, THSTI; Dr. Shreyasi Chatterjee and all the HVTR

480

laboratory members for support. IAVI's work was made possible by generous support from many

481

donors including: the Bill & Melinda Gates Foundation; the Ministry of Foreign Affairs of

482

Denmark; Irish Aid; the Ministry of Finance of Japan; the Ministry of Foreign Affairs of the

483

Netherlands; the Norwegian Agency for Development Cooperation (NORAD); the United

484

Kingdom Department for International Development (DFID); and the United States Agency for

485

International Development (USAID). The full list of IAVI donors is available at www.iavi.org.

486

The contents are the responsibility of the International AIDS Vaccine Initiative and do not 23

487

necessarily reflect the views of USAID or the United States Government. The contents of this

488

manuscript are the responsibility of IAVI and do not necessarily reflect the views of USAID or

489

the US Government.

490 491

24

492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541

References. 1. 2.

3.

4.

5.

6. 7.

8.

9.

10.

11.

12.

13.

Alter, G., and D. H. Barouch. 2015. Natural evolution of broadly neutralizing antibodies. Cell 161:427-428. Basu, D., C. S. Kraft, M. K. Murphy, P. J. Campbell, T. Yu, P. T. Hraber, C. Irene, A. Pinter, E. Chomba, J. Mulenga, W. Kilembe, S. A. Allen, C. A. Derdeyn, and E. Hunter. 2012. HIV-1 subtype C superinfected individuals mount low autologous neutralizing antibody responses prior to intrasubtype superinfection. Retrovirology 9:76. Blattner, C., J. H. Lee, K. Sliepen, R. Derking, E. Falkowska, A. T. de la Pena, A. Cupo, J. P. Julien, M. van Gils, P. S. Lee, W. Peng, J. C. Paulson, P. Poignard, D. R. Burton, J. P. Moore, R. W. Sanders, I. A. Wilson, and A. B. Ward. 2014. Structural delineation of a quaternary, cleavage-dependent epitope at the gp41-gp120 interface on intact HIV-1 Env trimers. Immunity 40:669-680. Boliar, S., S. Das, M. Bansal, B. N. Shukla, S. Patil, T. Shrivastava, S. Samal, S. Goswami, C. R. King, J. Bhattacharya, and B. K. Chakrabarti. 2015. An Efficiently Cleaved HIV-1 Clade C Env Selectively Binds to Neutralizing Antibodies. PloS one 10:e0122443. Braibant, M., S. Brunet, D. Costagliola, C. Rouzioux, H. Agut, H. Katinger, B. Autran, and F. Barin. 2006. Antibodies to conserved epitopes of the HIV-1 envelope in sera from longterm non-progressors: prevalence and association with neutralizing activity. Aids 20:19231930. Burton, D. R., and J. R. Mascola. 2015. Antibody responses to envelope glycoproteins in HIV-1 infection. Nature immunology 16:571-576. Caskey, M., F. Klein, J. C. Lorenzi, M. S. Seaman, A. P. West, Jr., N. Buckley, G. Kremer, L. Nogueira, M. Braunschweig, J. F. Scheid, J. A. Horwitz, I. Shimeliovich, S. BenAvraham, M. Witmer-Pack, M. Platten, C. Lehmann, L. A. Burke, T. Hawthorne, R. J. Gorelick, B. D. Walker, T. Keler, R. M. Gulick, G. Fatkenheuer, S. J. Schlesinger, and M. C. Nussenzweig. 2015. Viraemia suppressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature. Chaillon, A., M. Braibant, T. Moreau, S. Thenin, A. Moreau, B. Autran, and F. Barin. 2011. The V1V2 domain and an N-linked glycosylation site in the V3 loop of the HIV-1 envelope glycoprotein modulate neutralization sensitivity to the human broadly neutralizing antibody 2G12. Journal of virology 85:3642-3648. Chakrabarti, B. K., Y. Feng, S. K. Sharma, K. McKee, G. B. Karlsson Hedestam, C. C. Labranche, D. C. Montefiori, J. R. Mascola, and R. T. Wyatt. 2013. Robust neutralizing antibodies elicited by HIV-1 JRFL envelope glycoprotein trimers in nonhuman primates. Journal of virology 87:13239-13251. Chung, N. P., K. Matthews, H. J. Kim, T. J. Ketas, M. Golabek, K. de Los Reyes, J. Korzun, A. Yasmeen, R. W. Sanders, P. J. Klasse, I. A. Wilson, A. B. Ward, A. J. Marozsan, J. P. Moore, and A. Cupo. 2014. Stable 293 T and CHO cell lines expressing cleaved, stable HIV1 envelope glycoprotein trimers for structural and vaccine studies. Retrovirology 11:33. Diskin, R., J. F. Scheid, P. M. Marcovecchio, A. P. West, Jr., F. Klein, H. Gao, P. N. Gnanapragasam, A. Abadir, M. S. Seaman, M. C. Nussenzweig, and P. J. Bjorkman. 2011. Increasing the potency and breadth of an HIV antibody by using structure-based rational design. Science 334:1289-1293. Donners, H., B. Willems, E. Beirnaert, R. Colebunders, D. Davis, and G. van der Groen. 2002. Cross-neutralizing antibodies against primary isolates in African women infected with HIV-1. Aids 16:501-503. Doores, K. J., L. Kong, S. A. Krumm, K. M. Le, D. Sok, U. Laserson, F. Garces, P. Poignard, I. A. Wilson, and D. R. Burton. 2015. Two classes of broadly neutralizing antibodies within a single lineage directed to the high-mannose patch of HIV envelope. Journal of virology 89:1105-1118. 25

542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591

14.

15. 16.

17. 18.

19.

20.

21.

22.

23.

24.

Doria-Rose, N. A., I. Georgiev, S. O'Dell, G. Y. Chuang, R. P. Staupe, J. S. McLellan, J. Gorman, M. Pancera, M. Bonsignori, B. F. Haynes, D. R. Burton, W. C. Koff, P. D. Kwong, and J. R. Mascola. 2012. A short segment of the HIV-1 gp120 V1/V2 region is a major determinant of resistance to V1/V2 neutralizing antibodies. Journal of virology 86:83198323. Doria-Rose, N. A., and M. G. Joyce. 2015. Strategies to guide the antibody affinity maturation process. Current opinion in virology 11:137-147. Doria-Rose, N. A., C. A. Schramm, J. Gorman, P. L. Moore, J. N. Bhiman, B. J. DeKosky, M. J. Ernandes, I. S. Georgiev, H. J. Kim, M. Pancera, R. P. Staupe, H. R. Altae-Tran, R. T. Bailer, E. T. Crooks, A. Cupo, A. Druz, N. J. Garrett, K. H. Hoi, R. Kong, M. K. Louder, N. S. Longo, K. McKee, M. Nonyane, S. O'Dell, R. S. Roark, R. S. Rudicell, S. D. Schmidt, D. J. Sheward, C. Soto, C. K. Wibmer, Y. Yang, Z. Zhang, N. C. S. Program, J. C. Mullikin, J. M. Binley, R. W. Sanders, I. A. Wilson, J. P. Moore, A. B. Ward, G. Georgiou, C. Williamson, S. S. Abdool Karim, L. Morris, P. D. Kwong, L. Shapiro, and J. R. Mascola. 2014. Developmental pathway for potent V1V2-directed HIV-neutralizing antibodies. Nature 509:55-62. Esparza, J. 2013. A brief history of the global effort to develop a preventive HIV vaccine. Vaccine 31:3502-3518. Falkowska, E., K. M. Le, A. Ramos, K. J. Doores, J. H. Lee, C. Blattner, A. Ramirez, R. Derking, M. J. van Gils, C. H. Liang, R. McBride, B. von Bredow, S. S. Shivatare, C. Y. Wu, P. Y. Chan-Hui, Y. Liu, T. Feizi, M. B. Zwick, W. C. Koff, M. S. Seaman, K. Swiderek, J. P. Moore, D. Evans, J. C. Paulson, C. H. Wong, A. B. Ward, I. A. Wilson, R. W. Sanders, P. Poignard, and D. R. Burton. 2014. Broadly neutralizing HIV antibodies define a glycandependent epitope on the prefusion conformation of gp41 on cleaved envelope trimers. Immunity 40:657-668. Feng, Y., K. McKee, K. Tran, S. O'Dell, S. D. Schmidt, A. Phogat, M. N. Forsell, G. B. Karlsson Hedestam, J. R. Mascola, and R. T. Wyatt. 2012. Biochemically defined HIV-1 envelope glycoprotein variant immunogens display differential binding and neutralizing specificities to the CD4-binding site. J Biol Chem 287:5673-5686. Fera, D., A. G. Schmidt, B. F. Haynes, F. Gao, H. X. Liao, T. B. Kepler, and S. C. Harrison. 2014. Affinity maturation in an HIV broadly neutralizing B-cell lineage through reorientation of variable domains. Proceedings of the National Academy of Sciences of the United States of America 111:10275-10280. Gray, E. S., M. C. Madiga, T. Hermanus, P. L. Moore, C. K. Wibmer, N. L. Tumba, L. Werner, K. Mlisana, S. Sibeko, C. Williamson, S. S. Abdool Karim, and L. Morris. 2011. The neutralization breadth of HIV-1 develops incrementally over four years and is associated with CD4+ T cell decline and high viral load during acute infection. Journal of virology 85:4828-4840. Gray, E. S., T. Meyers, G. Gray, D. C. Montefiori, and L. Morris. 2006. Insensitivity of paediatric HIV-1 subtype C viruses to broadly neutralising monoclonal antibodies raised against subtype B. PLoS Med 3:e255. Gray, E. S., M. A. Moody, C. K. Wibmer, X. Chen, D. Marshall, J. Amos, P. L. Moore, A. Foulger, J. S. Yu, B. Lambson, S. Abdool Karim, J. Whitesides, G. D. Tomaras, B. F. Haynes, L. Morris, and H. X. Liao. 2011. Isolation of a monoclonal antibody that targets the alpha-2 helix of gp120 and represents the initial autologous neutralizing-antibody response in an HIV-1 subtype C-infected individual. Journal of virology 85:7719-7729. Gray, E. S., P. L. Moore, F. Bibollet-Ruche, H. Li, J. M. Decker, T. Meyers, G. M. Shaw, and L. Morris. 2008. 4E10-resistant variants in a human immunodeficiency virus type 1 subtype C-infected individual with an anti-membrane-proximal external regionneutralizing antibody response. Journal of virology 82:2367-2375. 26

592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641

25.

26.

27.

28.

29.

30.

31.

32.

33.

34. 35.

36.

Gray, E. S., P. L. Moore, I. A. Choge, J. M. Decker, F. Bibollet-Ruche, H. Li, N. Leseka, F. Treurnicht, K. Mlisana, G. M. Shaw, S. S. Karim, C. Williamson, and L. Morris. 2007. Neutralizing antibody responses in acute human immunodeficiency virus type 1 subtype C infection. Journal of virology 81:6187-6196. Gray, E. S., N. Taylor, D. Wycuff, P. L. Moore, G. D. Tomaras, C. K. Wibmer, A. Puren, A. DeCamp, P. B. Gilbert, B. Wood, D. C. Montefiori, J. M. Binley, G. M. Shaw, B. F. Haynes, J. R. Mascola, and L. Morris. 2009. Antibody specificities associated with neutralization breadth in plasma from human immunodeficiency virus type 1 subtype C-infected blood donors. Journal of virology 83:8925-8937. Guttman, M., A. Cupo, J. P. Julien, R. W. Sanders, I. A. Wilson, J. P. Moore, and K. K. Lee. 2015. Antibody potency relates to the ability to recognize the closed, pre-fusion form of HIV Env. Nature communications 6:6144. Harrington, P. R., J. A. Nelson, K. M. Kitrinos, and R. Swanstrom. 2007. Independent evolution of human immunodeficiency virus type 1 env V1/V2 and V4/V5 hypervariable regions during chronic infection. Journal of virology 81:5413-5417. Horiya, S., J. K. Bailey, J. S. Temme, Y. V. Guillen Schlippe, and I. J. Krauss. 2014. Directed evolution of multivalent glycopeptides tightly recognized by HIV antibody 2G12. Journal of the American Chemical Society 136:5407-5415. Hraber, P., M. S. Seaman, R. T. Bailer, J. R. Mascola, D. C. Montefiori, and B. T. Korber. 2014. Prevalence of broadly neutralizing antibody responses during chronic HIV-1 infection. Aids 28:163-169. Huang, J., G. Ofek, L. Laub, M. K. Louder, N. A. Doria-Rose, N. S. Longo, H. Imamichi, R. T. Bailer, B. Chakrabarti, S. K. Sharma, S. M. Alam, T. Wang, Y. Yang, B. Zhang, S. A. Migueles, R. Wyatt, B. F. Haynes, P. D. Kwong, J. R. Mascola, and M. Connors. 2012. Broad and potent neutralization of HIV-1 by a gp41-specific human antibody. Nature 491:406-412. Julien, J. P., D. Sok, R. Khayat, J. H. Lee, K. J. Doores, L. M. Walker, A. Ramos, D. C. Diwanji, R. Pejchal, A. Cupo, U. Katpally, R. S. Depetris, R. L. Stanfield, R. McBride, A. J. Marozsan, J. C. Paulson, R. W. Sanders, J. P. Moore, D. R. Burton, P. Poignard, A. B. Ward, and I. A. Wilson. 2013. Broadly neutralizing antibody PGT121 allosterically modulates CD4 binding via recognition of the HIV-1 gp120 V3 base and multiple surrounding glycans. PLoS pathogens 9:e1003342. Klein, F., C. Gaebler, H. Mouquet, D. N. Sather, C. Lehmann, J. F. Scheid, Z. Kraft, Y. Liu, J. Pietzsch, A. Hurley, P. Poignard, T. Feizi, L. Morris, B. D. Walker, G. Fatkenheuer, M. S. Seaman, L. Stamatatos, and M. C. Nussenzweig. 2012. Broad neutralization by a combination of antibodies recognizing the CD4 binding site and a new conformational epitope on the HIV-1 envelope protein. The Journal of experimental medicine 209:14691479. Klein, F., H. Mouquet, P. Dosenovic, J. F. Scheid, L. Scharf, and M. C. Nussenzweig. 2013. Antibodies in HIV-1 vaccine development and therapy. Science 341:1199-1204. Koff, W. C., N. D. Russell, M. Walport, M. B. Feinberg, J. W. Shiver, S. A. Karim, B. D. Walker, M. G. McGlynn, C. V. Nweneka, and G. J. Nabel. 2013. Accelerating the development of a safe and effective HIV vaccine: HIV vaccine case study for the Decade of Vaccines. Vaccine 31 Suppl 2:B204-208. Kong, L., J. H. Lee, K. J. Doores, C. D. Murin, J. P. Julien, R. McBride, Y. Liu, A. Marozsan, A. Cupo, P. J. Klasse, S. Hoffenberg, M. Caulfield, C. R. King, Y. Hua, K. M. Le, R. Khayat, M. C. Deller, T. Clayton, H. Tien, T. Feizi, R. W. Sanders, J. C. Paulson, J. P. Moore, R. L. Stanfield, D. R. Burton, A. B. Ward, and I. A. Wilson. 2013. Supersite of immune vulnerability on the glycosylated face of HIV-1 envelope glycoprotein gp120. Nature structural & molecular biology 20:796-803. 27

642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690

37.

38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

Kwong, P. D., J. R. Mascola, and G. J. Nabel. 2013. Broadly neutralizing antibodies and the search for an HIV-1 vaccine: the end of the beginning. Nature reviews. Immunology 13:693701. Li, Y., K. Svehla, M. K. Louder, D. Wycuff, S. Phogat, M. Tang, S. A. Migueles, X. Wu, A. Phogat, G. M. Shaw, M. Connors, J. Hoxie, J. R. Mascola, and R. Wyatt. 2009. Analysis of neutralization specificities in polyclonal sera derived from human immunodeficiency virus type 1-infected individuals. Journal of virology 83:1045-1059. Liao, H. X., M. Bonsignori, S. M. Alam, J. S. McLellan, G. D. Tomaras, M. A. Moody, D. M. Kozink, K. K. Hwang, X. Chen, C. Y. Tsao, P. Liu, X. Lu, R. J. Parks, D. C. Montefiori, G. Ferrari, J. Pollara, M. Rao, K. K. Peachman, S. Santra, N. L. Letvin, N. Karasavvas, Z. Y. Yang, K. Dai, M. Pancera, J. Gorman, K. Wiehe, N. I. Nicely, S. Rerks-Ngarm, S. Nitayaphan, J. Kaewkungwal, P. Pitisuttithum, J. Tartaglia, F. Sinangil, J. H. Kim, N. L. Michael, T. B. Kepler, P. D. Kwong, J. R. Mascola, G. J. Nabel, A. Pinter, S. Zolla-Pazner, and B. F. Haynes. 2013. Vaccine induction of antibodies against a structurally heterogeneous site of immune pressure within HIV-1 envelope protein variable regions 1 and 2. Immunity 38:176-186. Mikell, I., and L. Stamatatos. 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. Moore, P. L., E. S. Gray, I. A. Choge, N. Ranchobe, K. Mlisana, S. S. Abdool Karim, C. Williamson, and L. Morris. 2008. The c3-v4 region is a major target of autologous neutralizing antibodies in human immunodeficiency virus type 1 subtype C infection. Journal of virology 82:1860-1869. Moore, P. L., E. S. Gray, D. Sheward, M. Madiga, N. Ranchobe, Z. Lai, W. J. Honnen, M. Nonyane, N. Tumba, T. Hermanus, S. Sibeko, K. Mlisana, S. S. Abdool Karim, C. Williamson, A. Pinter, and L. Morris. 2011. Potent and broad neutralization of HIV-1 subtype C by plasma antibodies targeting a quaternary epitope including residues in the V2 loop. Journal of virology 85:3128-3141. Moore, P. L., E. S. Gray, C. K. Wibmer, J. N. Bhiman, M. Nonyane, D. J. Sheward, T. Hermanus, S. Bajimaya, N. L. Tumba, M. R. Abrahams, B. E. Lambson, N. Ranchobe, L. Ping, N. Ngandu, Q. Abdool Karim, S. S. Abdool Karim, R. I. Swanstrom, M. S. Seaman, C. Williamson, and L. Morris. 2012. Evolution of an HIV glycan-dependent broadly neutralizing antibody epitope through immune escape. Nature medicine 18:1688-1692. Moore, P. L., N. Ranchobe, B. E. Lambson, E. S. Gray, E. Cave, M. R. Abrahams, G. Bandawe, K. Mlisana, S. S. Abdool Karim, C. Williamson, and L. Morris. 2009. Limited neutralizing antibody specificities drive neutralization escape in early HIV-1 subtype C infection. PLoS pathogens 5:e1000598. Morris, L., X. Chen, M. Alam, G. Tomaras, R. Zhang, D. J. Marshall, B. Chen, R. Parks, A. Foulger, F. Jaeger, M. Donathan, M. Bilska, E. S. Gray, S. S. Abdool Karim, T. B. Kepler, J. Whitesides, D. Montefiori, M. A. Moody, H. X. Liao, and B. F. Haynes. 2011. Isolation of a human anti-HIV gp41 membrane proximal region neutralizing antibody by antigenspecific single B cell sorting. PloS one 6:e23532. Nelson, J. D., F. M. Brunel, R. Jensen, E. T. Crooks, R. M. Cardoso, M. Wang, A. Hessell, I. A. Wilson, J. M. Binley, P. E. Dawson, D. R. Burton, and M. B. Zwick. 2007. An affinityenhanced neutralizing antibody against the membrane-proximal external region of human immunodeficiency virus type 1 gp41 recognizes an epitope between those of 2F5 and 4E10. Journal of virology 81:4033-4043. Overbaugh, J., and L. Morris. 2012. The Antibody Response against HIV-1. Cold Spring Harb Perspect Med 2:a007039.

28

691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

Pancera, M., and R. Wyatt. 2005. Selective recognition of oligomeric HIV-1 primary isolate envelope glycoproteins by potently neutralizing ligands requires efficient precursor cleavage. Virology 332:145-156. Patil, S., I. Choudhary, N. K. Chaudhary, R. Ringe, M. Bansal, B. N. Shukla, S. Boliar, B. K. Chakrabarti, and J. Bhattacharya. 2014. Determinants in V2C2 region of HIV-1 clade C primary envelopes conferred altered neutralization susceptibilities to IgG1b12 and PG9 monoclonal antibodies in a context-dependent manner. Virology 462-463:266-272. Pejchal, R., K. J. Doores, L. M. Walker, R. Khayat, P. S. Huang, S. K. Wang, R. L. Stanfield, J. P. Julien, A. Ramos, M. Crispin, R. Depetris, U. Katpally, A. Marozsan, A. Cupo, S. Maloveste, Y. Liu, R. McBride, Y. Ito, R. W. Sanders, C. Ogohara, J. C. Paulson, T. Feizi, C. N. Scanlan, C. H. Wong, J. P. Moore, W. C. Olson, A. B. Ward, P. Poignard, W. R. Schief, D. R. Burton, and I. A. Wilson. 2011. A potent and broad neutralizing antibody recognizes and penetrates the HIV glycan shield. Science 334:1097-1103. Rademeyer, C., P. L. Moore, N. Taylor, D. P. Martin, I. A. Choge, E. S. Gray, H. W. Sheppard, C. Gray, L. Morris, and C. Williamson. 2007. Genetic characteristics of HIV-1 subtype C envelopes inducing cross-neutralizing antibodies. Virology 368:172-181. Richman, D. D., T. Wrin, S. J. Little, and C. J. Petropoulos. 2003. Rapid evolution of the neutralizing antibody response to HIV type 1 infection. Proceedings of the National Academy of Sciences of the United States of America 100:4144-4149. Ringe, R., L. Das, I. Choudhary, D. Sharma, R. Paranjape, V. S. Chauhan, and J. Bhattacharya. 2012. Unique C2V3 Sequence in HIV-1 Envelope Obtained from Broadly Neutralizing Plasma of a Slow Progressing Patient Conferred Enhanced Virus Neutralization. PloS one 7:e46713. Ringe, R., M. Thakar, and J. Bhattacharya. 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. Ringe, R. P., R. W. Sanders, A. Yasmeen, H. J. Kim, J. H. Lee, A. Cupo, J. Korzun, R. Derking, T. van Montfort, J. P. Julien, I. A. Wilson, P. J. Klasse, A. B. Ward, and J. P. Moore. 2013. Cleavage strongly influences whether soluble HIV-1 envelope glycoprotein trimers adopt a native-like conformation. Proceedings of the National Academy of Sciences of the United States of America 110:18256-18261. Rolland, M., P. T. Edlefsen, B. B. Larsen, S. Tovanabutra, E. Sanders-Buell, T. Hertz, A. C. deCamp, C. Carrico, S. Menis, C. A. Magaret, H. Ahmed, M. Juraska, L. Chen, P. Konopa, S. Nariya, J. N. Stoddard, K. Wong, H. Zhao, W. Deng, B. S. Maust, M. Bose, S. Howell, A. Bates, M. Lazzaro, A. O'Sullivan, E. Lei, A. Bradfield, G. Ibitamuno, V. Assawadarachai, R. J. O'Connell, M. S. deSouza, S. Nitayaphan, S. Rerks-Ngarm, M. L. Robb, J. S. McLellan, I. Georgiev, P. D. Kwong, J. M. Carlson, N. L. Michael, W. R. Schief, P. B. Gilbert, J. I. Mullins, and J. H. Kim. 2012. Increased HIV-1 vaccine efficacy against viruses with genetic signatures in Env V2. Nature 490:417-420. Rong, R., F. Bibollet-Ruche, J. Mulenga, S. Allen, J. L. Blackwell, and C. A. Derdeyn. 2007. Role of V1V2 and other human immunodeficiency virus type 1 envelope domains in resistance to autologous neutralization during clade C infection. Journal of virology 81:1350-1359. Rong, R., B. Li, R. M. Lynch, R. E. Haaland, M. K. Murphy, J. Mulenga, S. A. Allen, A. Pinter, G. M. Shaw, E. Hunter, J. E. Robinson, S. Gnanakaran, and C. A. Derdeyn. 2009. Escape from autologous neutralizing antibodies in acute/early subtype C HIV-1 infection requires multiple pathways. PLoS pathogens 5:e1000594. Rusert, P., A. Krarup, C. Magnus, O. F. Brandenberg, J. Weber, A. K. Ehlert, R. R. Regoes, H. F. Gunthard, and A. Trkola. 2011. Interaction of the gp120 V1V2 loop with a neighboring gp120 unit shields the HIV envelope trimer against cross-neutralizing antibodies. The Journal of experimental medicine 208:1419-1433. 29

742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791

60.

61.

62.

63.

64.

65.

66.

67.

68.

69.

70.

Sagar, M., X. Wu, S. Lee, and J. Overbaugh. 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. Journal of virology 80:9586-9598. Sanders, R. W., R. Derking, A. Cupo, J. P. Julien, A. Yasmeen, N. de Val, H. J. Kim, C. Blattner, A. T. de la Pena, J. Korzun, M. Golabek, K. de Los Reyes, T. J. Ketas, M. J. van Gils, C. R. King, I. A. Wilson, A. B. Ward, P. J. Klasse, and J. P. Moore. 2013. A nextgeneration cleaved, soluble HIV-1 Env trimer, BG505 SOSIP.664 gp140, expresses multiple epitopes for broadly neutralizing but not non-neutralizing antibodies. PLoS pathogens 9:e1003618. Sather, D. N., J. Armann, L. K. Ching, A. Mavrantoni, G. Sellhorn, Z. Caldwell, X. Yu, B. Wood, S. Self, S. Kalams, and L. Stamatatos. 2009. Factors associated with the development of cross-reactive neutralizing antibodies during human immunodeficiency virus type 1 infection. Journal of virology 83:757-769. Sather, D. N., S. Carbonetti, D. C. Malherbe, F. Pissani, A. B. Stuart, A. J. Hessell, M. D. Gray, I. Mikell, S. A. Kalams, N. L. Haigwood, and L. Stamatatos. 2014. Emergence of broadly neutralizing antibodies and viral coevolution in two subjects during the early stages of infection with human immunodeficiency virus type 1. Journal of virology 88:1296812981. Scharf, L., J. F. Scheid, J. H. Lee, A. P. West, Jr., C. Chen, H. Gao, P. N. Gnanapragasam, R. Mares, M. S. Seaman, A. B. Ward, M. C. Nussenzweig, and P. J. Bjorkman. 2014. Antibody 8ANC195 reveals a site of broad vulnerability on the HIV-1 envelope spike. Cell reports 7:785-795. Scheid, J. F., H. Mouquet, B. Ueberheide, R. Diskin, F. Klein, T. Y. Oliveira, J. Pietzsch, D. Fenyo, A. Abadir, K. Velinzon, A. Hurley, S. Myung, F. Boulad, P. Poignard, D. R. Burton, F. Pereyra, D. D. Ho, B. D. Walker, M. S. Seaman, P. J. Bjorkman, B. T. Chait, and M. C. Nussenzweig. 2011. Sequence and structural convergence of broad and potent HIV antibodies that mimic CD4 binding. Science 333:1633-1637. Simek, M. D., W. Rida, F. H. Priddy, P. Pung, E. Carrow, D. S. Laufer, J. K. Lehrman, M. Boaz, T. Tarragona-Fiol, G. Miiro, J. Birungi, A. Pozniak, D. A. McPhee, O. Manigart, E. Karita, A. Inwoley, W. Jaoko, J. Dehovitz, L. G. Bekker, P. Pitisuttithum, R. Paris, L. M. Walker, P. Poignard, T. Wrin, P. E. Fast, D. R. Burton, and W. C. Koff. 2009. Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity identified by using a high-throughput neutralization assay together with an analytical selection algorithm. Journal of virology 83:7337-7348. Sok, D., K. J. Doores, B. Briney, K. M. Le, K. L. Saye-Francisco, A. Ramos, D. W. Kulp, J. P. Julien, S. Menis, L. Wickramasinghe, M. S. Seaman, W. R. Schief, I. A. Wilson, P. Poignard, and D. R. Burton. 2014. Promiscuous glycan site recognition by antibodies to the high-mannose patch of gp120 broadens neutralization of HIV. Science translational medicine 6:236ra263. Sok, D., M. J. van Gils, M. Pauthner, J. P. Julien, K. L. Saye-Francisco, J. Hsueh, B. Briney, J. H. Lee, K. M. Le, P. S. Lee, Y. Hua, M. S. Seaman, J. P. Moore, A. B. Ward, I. A. Wilson, R. W. Sanders, and D. R. Burton. 2014. Recombinant HIV envelope trimer selects for quaternary-dependent antibodies targeting the trimer apex. Proceedings of the National Academy of Sciences of the United States of America 111:17624-17629. Stamatatos, L., L. Morris, D. R. Burton, and J. R. Mascola. 2009. Neutralizing antibodies generated during natural HIV-1 infection: good news for an HIV-1 vaccine? Nature medicine 15:866-870. Teng, G., and F. N. Papavasiliou. 2007. Immunoglobulin somatic hypermutation. Annual review of genetics 41:107-120. 30

792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842

71.

72.

73.

74.

75.

76.

77.

78.

79.

80.

81.

Tomaras, G. D., J. M. Binley, E. S. Gray, E. T. Crooks, K. Osawa, P. L. Moore, N. Tumba, T. Tong, X. Shen, N. L. Yates, J. Decker, C. K. Wibmer, F. Gao, S. M. Alam, P. Easterbrook, S. Abdool Karim, G. Kamanga, J. A. Crump, M. Cohen, G. M. Shaw, J. R. Mascola, B. F. Haynes, D. C. Montefiori, and L. Morris. 2011. Polyclonal B cell responses to conserved neutralization epitopes in a subset of HIV-1-infected individuals. Journal of virology 85:11502-11519. Trkola, A., M. Purtscher, T. Muster, C. Ballaun, A. Buchacher, N. Sullivan, K. Srinivasan, J. Sodroski, J. P. Moore, and H. Katinger. 1996. Human monoclonal antibody 2G12 defines a distinctive neutralization epitope on the gp120 glycoprotein of human immunodeficiency virus type 1. Journal of virology 70:1100-1108. van Gils, M. J., E. M. Bunnik, B. D. Boeser-Nunnink, J. A. Burger, M. Terlouw-Klein, N. Verwer, and H. Schuitemaker. 2011. Longer V1V2 region with increased number of potential N-linked glycosylation sites in the HIV-1 envelope glycoprotein protects against HIV-specific neutralizing antibodies. Journal of virology 85:6986-6995. Walker, L. M., M. Huber, K. J. Doores, E. Falkowska, R. Pejchal, J. P. Julien, S. K. Wang, A. Ramos, P. Y. Chan-Hui, M. Moyle, J. L. Mitcham, P. W. Hammond, O. A. Olsen, P. Phung, S. Fling, C. H. Wong, S. Phogat, T. Wrin, M. D. Simek, P. G. Principal Investigators, W. C. Koff, I. A. Wilson, D. R. Burton, and P. Poignard. 2011. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 477:466-470. Walker, L. M., M. Huber, K. J. Doores, E. Falkowska, R. Pejchal, J. P. Julien, S. K. Wang, A. Ramos, P. Y. Chan-Hui, M. Moyle, J. L. Mitcham, P. W. Hammond, O. A. Olsen, P. Phung, S. Fling, C. H. Wong, S. Phogat, T. Wrin, M. D. Simek, G. P. I. Protocol, W. C. Koff, I. A. Wilson, D. R. Burton, and P. Poignard. 2011. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 477:466-470. Walker, L. M., S. K. Phogat, P. Y. Chan-Hui, D. Wagner, P. Phung, J. L. Goss, T. Wrin, M. D. Simek, S. Fling, J. L. Mitcham, J. K. Lehrman, F. H. Priddy, O. A. Olsen, S. M. Frey, P. W. Hammond, S. Kaminsky, T. Zamb, M. Moyle, W. C. Koff, P. Poignard, and D. R. Burton. 2009. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 326:285-289. Walker, L. M., M. D. Simek, F. Priddy, J. S. Gach, D. Wagner, M. B. Zwick, S. K. Phogat, P. Poignard, and D. R. Burton. 2010. A limited number of antibody specificities mediate broad and potent serum neutralization in selected HIV-1 infected individuals. PLoS pathogens 6:e1001028. Wibmer, C. K., J. N. Bhiman, E. S. Gray, N. Tumba, S. S. Abdool Karim, C. Williamson, L. Morris, and P. L. Moore. 2013. Viral escape from HIV-1 neutralizing antibodies drives increased plasma neutralization breadth through sequential recognition of multiple epitopes and immunotypes. PLoS pathogens 9:e1003738. Wu, X., Z. Y. Yang, Y. Li, C. M. Hogerkorp, W. R. Schief, M. S. Seaman, T. Zhou, S. D. Schmidt, L. Wu, L. Xu, N. S. Longo, K. McKee, S. O'Dell, M. K. Louder, D. L. Wycuff, Y. Feng, M. Nason, N. Doria-Rose, M. Connors, P. D. Kwong, M. Roederer, R. T. Wyatt, G. J. Nabel, and J. R. Mascola. 2010. Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science 329:856-861. Wu, X., Z. Zhang, C. A. Schramm, M. G. Joyce, Y. Do Kwon, T. Zhou, Z. Sheng, B. Zhang, S. O'Dell, K. McKee, I. S. Georgiev, G. Y. Chuang, N. S. Longo, R. M. Lynch, K. O. Saunders, C. Soto, S. Srivatsan, Y. Yang, R. T. Bailer, M. K. Louder, N. C. S. Program, J. C. Mullikin, M. Connors, P. D. Kwong, J. R. Mascola, and L. Shapiro. 2015. Maturation and Diversity of the VRC01-Antibody Lineage over 15 Years of Chronic HIV-1 Infection. Cell 161:470-485. Wu, X., T. Zhou, J. Zhu, B. Zhang, I. Georgiev, C. Wang, X. Chen, N. S. Longo, M. Louder, K. McKee, S. O'Dell, S. Perfetto, S. D. Schmidt, W. Shi, L. Wu, Y. Yang, Z. Y. Yang, Z. Yang, Z. Zhang, M. Bonsignori, J. A. Crump, S. H. Kapiga, N. E. Sam, B. F. Haynes, M. 31

843 844 845 846 847 848 849 850 851 852 853 854 855 856 857

82.

83.

Simek, D. R. Burton, W. C. Koff, N. A. Doria-Rose, M. Connors, N. C. S. Program, J. C. Mullikin, G. J. Nabel, M. Roederer, L. Shapiro, P. D. Kwong, and J. R. Mascola. 2011. Focused evolution of HIV-1 neutralizing antibodies revealed by structures and deep sequencing. Science 333:1593-1602. Zhu, J., G. Ofek, Y. Yang, B. Zhang, M. K. Louder, G. Lu, K. McKee, M. Pancera, J. Skinner, Z. Zhang, R. Parks, J. Eudailey, K. E. Lloyd, J. Blinn, S. M. Alam, B. F. Haynes, M. Simek, D. R. Burton, W. C. Koff, N. C. S. Program, J. C. Mullikin, J. R. Mascola, L. Shapiro, and P. D. Kwong. 2013. Mining the antibodyome for HIV-1-neutralizing antibodies with next-generation sequencing and phylogenetic pairing of heavy/light chains. Proceedings of the National Academy of Sciences of the United States of America 110:64706475. Zwick, M. B., A. F. Labrijn, M. Wang, C. Spenlehauer, E. O. Saphire, J. M. Binley, J. P. Moore, G. Stiegler, H. Katinger, D. R. Burton, and P. W. Parren. 2001. Broadly neutralizing antibodies targeted to the membrane-proximal external region of human immunodeficiency virus type 1 glycoprotein gp41. Journal of virology 75:10892-10905.

858 859 860

32

861

Table and Figure Legends:

862 863

Table 1. Neutralization breadth of Protocol G G37080 plasma samples collected at two different

864

points tested against 57 panel Env-pseudotyped viruses.

865 866

Table 2. Examination of known specificity of G37080 plasma antibodies obtained at both visits

867

to HIV Env.

868 869

Table 3. Degree of shift in sensitivity of the Env-pseudotyped viruses to G37080 BCN plasma

870

depleted with the soluble monomeric and trimeric Env proteins and a clade C MPER peptide

871

(C1C).

872 873

Table 4. Mapping specificities mediating neutralization resistance of the autologous and

874

heterologous Envs to G37080 BCN plasma.

875 876

Table 5. Sensitivity of wild type, chimera and point mutants of autologous Envs to PG9 MAb

877 878

Figure 1 A. Genetic divergence of amino acid sequences of 57 HIV-1 Env (gp160) used to

879

assess neutralization breadth and potency of G37080 BCN plasma. Maximum likelihood

880

bootstrapped consensus phylogenetic tree was constructed using Jones-Taylor-Thornton (JTT)

881

substitution model with 50 bootstrapped replicates in Mega 5.2 version. Bootstrapped values are

882

shown at the nodes of each branch. Hollow circles represent envelopes (16055-2.3 and

883

92TH021) resistant to neutralization by G37080 BCN plasma. B. Neutralization breadth of the

884

G37080 BCN plasma obtained at visit 1 and visit 2 were assessed against pseudotyped viruses 33

885

expressing HIV-1 Env representing different clades and origins. Neutralization titers (median

886

ID50 values) were obtained by titrating Env-pseudotyped viruses against G37080 plasma

887

samples. Values at top of each bar graph indicate number of viruses belonging to each

888

clade/origin tested.

889 890

Figure 2. Assessing dependence of G37080 BCN antibodies to CD4 binding site (CD4bs) region

891

of HIV-1 Env. G37080 BCN plasma samples and VRC01 MAb (concentrations that neutralized

892

25711-2.4 by >80%) pre-incubated with different concentrations, as indicated, with TripleMut

893

core (A) and RSC3 (B) proteins were examined for their ability to neutralize 25711-2.4 Env

894

pseudotyped virus in TZM-bl cell neutralization assay. Note that while VRC01 pre-absorbed

895

with both TripleMut and RSC3 proteins showed inhibition to neutralize 25711-2.4 in a dose-

896

dependent manner, no such effect was observed with G37080 BCN plasma indicated absence of

897

CD4bs directed neutralizing antibodies.

898 899

Figure 3. Binding of G37080 BCN plasma IgG to 4-2.J41 monomeric gp120 (A) and BG505-

900

SOSIP.664-D7324 cleaved trimeric gp140 (B) soluble proteins were assessed by ELISA. IgG

901

purified from HIV negative healthy donor and known MAbs were used as controls. Extent of

902

binding of the depleted and undepleted G37080 BCN plasma with magnetic beads coated with 4-

903

2.J41 monomeric gp120 (C) and BG505-SOSIP.664 cleaved trimeric gp140 to their respective

904

proteins by ELISA. Note that binding to trimeric protein by ELISA was assessed by using

905

BG505-SOSIP.664 tagged with D7324 epitope to maintain native conformation of trimeric Env

906

as described before (61).

907

34

908

Figure 4. A. Neutralization susceptibility of autologous Envs to contemporaneous G37080 BCN

909

plasma and its follow up sample from the same donor. Neutralization titers (median ID50) were

910

obtained by titrating pseudotyped viruses expressing autologous Envs obtained from visit 1 and

911

follow up G37080 plasma to contemporaneous plasma antibodies. Note that both the Envs

912

obtained from follow up G37080 plasma (visit 2) were found to be resistant to contemporaneous

913

autologous plasma, while Envs obtained from visit 1 G37080 plasma were found to be sensitive

914

to follow up plasma antibodies. B. Alignment of V1V2 amino acid sequences of sensitive and

915

resistant autologous Envs obtained at both visits were done by using seqpublish available at HIV

916

Los Alamos database (www.hiv.lanl.gov). Key residues that mediate autologous neutralization

917

resistance are highlighted.

918

35

Table 1. Neutralization breadth of Protocol G G37080 plasma samples collected at two different time points tested against 57 panel Env-pseudotyped viruses

G37080 Plasma

India Clade C

Africa Clade C

Clade B

Clade A

Others

Envelope MuLV HIV-2 (7312A) 16055-2.3 16936_2.21 25710-2.3 25711-2.4 00836-2.5 2-5.J3 4.J22 4-2.J41 3-5.J25 5-4.J16 5.J41 7.J16 7.J20 11-3.J3 11-5.J12 LT-1.J1 LT1.J3 LT5.J3b LT5.J7b 93IN905 Median ID50 CAP45.G3 CAP84 CAP88 CAP239.G3 Du422.1 Du151.2 DU156.12 DU172.17 ZM109F.PB4 ZM197M.PB7 IAVIC22 Median ID50 JRFL PVO.4 TRJO4551.58 AC10.0.29 QH0692.42 REJO4549.67 SC422661.8 6535.3 RHPA 4259.7 HO61.14 92BR020 JRCSF Median ID50 Q769.ENV.b9 Q461.e2 Q842.d12 Q23.17 Q259.d2.26 BG505 94UG103 Median ID50 92TH021 LT5.J12 CH038.12 CH114.8 CH120.6 CRF 02AG_235 191727_D1_12 Median ID50

Accession No. S53043.1 JX235925.1 EF117268 EF117270 EF117271 EF117272 EF117265 GU945311.1 EU908219.1 GU945316.2 GU945314.1 GU945326.1 EU908221.1 EU908222.1 EU908223.1 GU945330.1 GU945333.1 JN400529 JN400534 JN400538 JN400540 AY669742.1

Clade NA A C C C C C C C C C C C C C C C C C C C C

DQ435682.1 EF203963.1 EF203972.1 EF203983.1 DQ411854.1 DQ411851.1 DQ411852.1 DQ411853.1 AY424138.2 DQ388515.1 --

C C C C C C C C C C C

U63632.1 AY835444.1 AY835450 AY835446 AY835439 AY835449 AY835441.1 AY835438 AY835447.1 EF210730 AY669718.1 M38429.1

B B B B B B B B B B B B

AF407157.1 AF407156 AF407160.1 AF004885 AF407152 DQ208458.1 AY669705.1

A A A A A A A

AY669775.1 FJ515876 EF042692 EF117264 EF117260 EU513195 HM215267.1

A/E B/C B/C B/C B/C A/G D

Visit-1 Visit-2