JVI Accepted Manuscript Posted Online 18 November 2015 J. Virol. doi:10.1128/JVI.02454-15 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

1

Title: HIV-1 monoclonal antibodies suppress acute SHIV viremia and limit seeding of cell-

2

associated viral reservoirs

3 4

Byline: Diane L. Bolton,1†# Amarendra Pegu,2† Keyun Wang,2 Kathleen McGinnis,2 Martha

5

Nason,3 Kathryn Foulds,2 Valerie Letukas,2 Stephen D. Schmidt,2 Xuejun Chen,2 John Paul

6

Todd,2 Jeffrey Lifson,4 Srinivas Rao,2§ Nelson L. Michael,5 Merlin L. Robb,1 John R. Mascola,2

7

and Richard A. Koup2

8 9

1

US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring,

10

Maryland 20910. Henry M. Jackson Foundation for the Advancement of Military Medicine,

11

Bethesda, Maryland 20818.

12

2

13

Institutes of Health, Bethesda, Maryland 20892.

14

3

15

Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892.

16

4

17

Laboratory for Cancer Research, AIDS and Cancer Virus Program, P.O. Box B, Frederick,

18

Maryland 21702.

19

5

20

Maryland 20910.

Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National

Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and

AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc./Frederick National

US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring,

21 22

Running title: Acute SHIV passive immunotherapy with HIV-1 Env mAbs

23

1

24

#Address correspondence to Diane L. Bolton, [email protected]

25

†D.L.B.

26

§

and A.P. contributed equally to this work.

Present address: SanofiPasteur, 38 Sidney St #370, Cambridge, MA 02139.

2

27

Abstract: Combination antiretroviral therapy (cART) administered shortly after HIV-1

28

infection can suppress viremia and limit seeding of the viral reservoir, but lifelong treatment is

29

required in the majority of patients. Highly potent broadly neutralizing HIV-1 monoclonal

30

antibodies (mAbs) can reduce plasma viremia when administered during chronic HIV-1

31

infection, but the therapeutic potential of these antibodies during acute infection is unknown.

32

We tested the ability of HIV-1 envelope glycoprotein specific broadly neutralizing mAbs to

33

suppress acute simian-human immunodeficiency virus (SHIV) replication in rhesus macaques.

34

Four groups of macaques were infected with SHIV-SF162P3 and received i) the CD4-binding

35

site mAb VRC01 or ii) a combination of a more potent clonal relative of VRC01 (VRC07-523)

36

and a V3 glycan-dependent mAb (PGT121) or iii) daily cART, all on day 10, just prior to

37

expected peak plasma viremia, or iv) no treatment. Daily cART was initiated 11 days after mAb

38

administration and was continued for 13 weeks in all treated animals. Over a period of 11 days

39

after a single administration, mAb treatment significantly reduced peak viremia, accelerated the

40

decay slope and reduced total viral replication as compared to untreated controls. Proviral DNA

41

in lymph node CD4 T cells was also diminished after treatment with dual mAb. These data

42

demonstrate the virological effect of potent mAbs and support future clinical trials that

43

investigate HIV-1 neutralizing mAbs as adjunctive therapy with cART during acute HIV-1

44

infection.

45 46

Importance: Treatment of chronic HIV-1 infection with potent broadly neutralizing HIV-1

47

mAbs has been shown to significantly reduce plasma viremia. However, the anti-viral effect of

48

mAb treatment during acute HIV-1 infection is unknown. Here, we demonstrate that mAbs

49

targeting the HIV-1 envelope glycoprotein both suppress acute SHIV plasma viremia and limit

3

50

CD4 T cell-associated viral DNA. These findings provide support for clinical trials of mAbs as

51

adjunctive therapy with antiretroviral therapy during acute HIV-1 infection.

4

52 53

Introduction Virological events in the first weeks following HIV-1 transmission set the stage for

54

lifelong chronic infection that remains incurable with currently available combination

55

antiretroviral therapy (cART), at least in part due to early establishment of viral reservoirs,

56

including latently infected cells, that persist despite cART and can give rise to recrudescent

57

infection when treatment is interrupted. A major hurdle to eradicating infection is the early

58

establishment of persistent viral reservoirs. Clinical cohorts in whom HIV-1 infection is

59

diagnosed during this early acute phase provide a unique opportunity to modify the course of

60

disease and better understand how viral reservoirs are established. While initiation of cART

61

during Fiebig I-III limits residual cell-associated viral DNA levels (1, 2), current antiretrovirals

62

act by blocking new rounds of infection and are thus limited in their ability to affect populations

63

of already infected cells that can contribute to viral reservoirs. Moreover, recent evidence from

64

experimentally infected rhesus macaques indicates that seeding of viral reservoirs can occur

65

before the advent of detectable viremia (3). While efforts to induce virus expression from

66

latently infected cells to facilitate elimination by immune-mediated cytotoxic or viral cytopathic

67

mechanisms are being actively explored, their efficacy remains to be demonstrated (4-7).

68

Alternative treatment options are needed, with particular focus on agents capable of targeting

69

already infected cells.

70

The recent isolation from chronically infected patients of potent broadly neutralizing

71

monoclonal antibodies (mAbs) specific for the HIV-1 Envelope glycoproteins creates new

72

possibilities for therapeutic agents (8-11). In addition to direct neutralization of virus, antibodies

73

can facilitate the recognition and elimination of infected cells through Fc-mediated mechanisms

74

such as antibody-dependent cell-mediated cytotoxicity and complement-mediated lysis (12-14).

5

75

In rhesus macaque and humanized mice animal models of SHIV and HIV-1 infection

76

respectively, HIV-1 mAbs have been shown to be effective as both immunoprophylactic and

77

immunotherapeutic agents (15-20). It has also been suggested that the number of infected cells

78

is reduced by mAb therapy in chronically infected animals (15, 17). These studies have led to

79

initiation of clinical trials testing safety and efficacy of mAbs for both prevention and treatment

80

of HIV-1 in humans (21-27). A recent report on the use of 3BNC117 to suppress viremia in

81

chronically HIV-1 infected individuals validates the animal data and provides rationale for use of

82

mAb therapy against HIV-1 infection (28). However, the impact of mAbs administered during

83

early acute infection on virus replication, reservoir establishment, and adaptive immune

84

responses is not known.

85

Here, we assessed the therapeutic activity of a single mAb, or a combination of two more

86

potent mAbs, during early acute SHIV162P3 infection in rhesus macaques. The single CD4

87

binding site directed mAb VRC01 was chosen because it is currently being studied in human

88

clinical trials. The combination of VRC07-523 and PGT121 was chosen because both mAbs are

89

under clinical development. VRC07-523 is an engineered variant related to VRC01 that is 5-10

90

fold more potent (29, 30). PGT121 is a potent mAb directed to the V3-glycan supersite region of

91

Env (31). We found that acute viremia was significantly reduced by a single infusion of either

92

VRC01 or a combination of VRC07-523 and PGT121, and that the dual mAb therapy regimen

93

dramatically diminished viral replication similarly to daily three-drug cART.

6

94

Materials and Methods

95

SHIV infection and treatment

96

Indian origin rhesus macaques were randomly assigned to each treatment group and challenged

97

intravenously with 20 TCID50 of SHIV-SF162P3. cART consisted of 20 mg/kg/day tenofovir

98

(PMPA, given subcutaneously; from Gilead Sciences, CA), 30 mg/kg/day emtricitabine (FTC,

99

given subcutaneously; from Gilead) and 100 mg raltegravir twice daily (Isentress, mixed with

100

food; from Merck, NJ). Animal weights ranged from 3.35-4.49 kg. For the antibody infusions,

101

low endotoxin preparation ( 0.05). To ensure that the elevated Env-

277

specific antibody levels reflect adaptive rhesus responses and not detection of residual infused

278

human mAb, we confirmed that the anti-rhesus Ig detection reagent used in our assay does not

279

cross-react with human Ig (data not shown). Similarly, the frequency of SHIV-specific T cells was reduced several fold in treated

280 281

animals (Fig. 3C-D). Antigen-specific CD4 and CD8 T cells were measured by in vitro peptide

282

stimulation of PBMC followed by intracellular cytokine staining for IFNγ, TNFα, and IL-2.

283

CD8 T cells targeting SIV Gag and HIV-1 Env were significantly diminished eight and twelve

284

weeks post-infection, while a similar trend was observed for both CD4 and CD8 T cells

285

following mAb treatment at week three. Treatment groups did not differ from one another. This

286

suggests that mAb therapy was effective at reducing the SHIV antigen load responsible for

287

stimulating adaptive T and B cell responses and that the suppression was similar to that seen with

288

cART.

15

289

The combination of cytokines expressed by a T cell has been shown to influence cellular

290

long-term memory potential and renewal capacity (39-41). To determine the impact of early

291

therapy on T cell functionality, we compared the cytokine distribution of Gag-specific CD4 T

292

cells. Acute infection response profiles were significantly different under cART and VRC07-

293

523+PGT121 compared to untreated infection (Fig. 3E, week 3). Analysis of the individual

294

categories revealed a shift toward IL-2-expressing cells by these regimens, while VRC01 therapy

295

and no treatment resulted in responses more likely to express only IFNγ or both IFNγ and TNFα

296

(Fig. 3F). These differences are consistent with less antigen-driven differentiation of CD4 T

297

cells under cART or VRC07-523+PGT121, the most potent suppressors of viremia. Following

298

viremia suppression by cART in the VRC01/cART group, CD4 T cell functionality evolved to

299

be more similar to the other treated groups (Fig. 3E-F, weeks 8-12), while untreated infection

300

maintained a high proportion of differentiated IFNγ+TNFα+ cells. Similarly, fewer terminally

301

differentiated single IFNγ+ SHIV-specific CD8 T cells were observed following VRC07-

302

523+PGT121 therapy (Fig. 3E-F, week 3). Thus acute immunotherapy with either VRC07-523

303

and PGT121 or daily ART facilitated the maintenance of CD4 and CD8 T cell memory potential.

304 305 306

Cell-associated viral load A major objective of therapy initiation during acute HIV-1 infection is to reduce the

307

establishment of a latently infected pool of cells, or reservoir. To determine the impact of mAb

308

immunotherapy on the number of infected cells, we measured CD4 T cell associated viral DNA

309

in lymph nodes and peripheral blood. CD4 T cells were FACS sorted into total, naïve, central

310

memory, and T follicular helper (Tfh; lymph node only) populations (Fig. 4A) and viral gag

311

(unspliced) DNA was measured by qPCR. The average proviral burden per cell was calculated

16

312

by dividing the gag DNA copy number within each population by the number of cells analyzed,

313

as determined by alb DNA copies. One week after treatment initiation (day 18), the normalized

314

level of SIV gag DNA in lymph node and PBMC CD4 T cells was reduced by all interventions

315

compared to untreated controls, to a statistically significant level for most comparisons (Fig. 4B-

316

C, p0.05). Of note, VRC07-523+PGT121 treatment achieved the

321

greatest day 18 reduction in lymph node naïve CD4 T cell viral load, which was less than the

322

assay’s detection threshold (5E-5 copies/cell) in four of the six animals. After cART initiation in

323

the mAb-treated animals, the decreased cell-associated viral DNA relative to the control group

324

was maintained and remained comparable to the group that received cART only in both LN and

325

PBMC (Fig. 4D). Pre-treatment (day 8) PBMC-associated viral DNA did not differ between any

326

of the study groups, indicating comparable baseline proviral DNA.

327

To determine whether the limited viral reservoir would impair viral resurgence upon

328

cART interruption, cART was discontinued 16 weeks after infection for all treatment groups.

329

Viremia rebound was observed for all animals ~12 days after cART cessation, with no difference

330

between mAb and cART groups with respect to the time to peak rebound viremia or the

331

magnitude of peak rebound viremia (Fig. 5).

332 333 334

Discussion

17

335

Therapeutic intervention during the acute stage of HIV-1 infection has the potential to

336

limit establishment of viral reservoirs and improve prospects for success in reservoir targeting

337

interventions. Our studies show robust in vivo activity of anti-HIV-1 broadly neutralizing mAbs

338

during acute SHIV-SF162P3 infection. All measured virological parameters, including peak

339

viremia, post-peak acute viremia, and cell-associated viral load, were reduced by dual VRC07-

340

523+PGT121 antibody treatment. While VRC01 monotherapy also curtailed viral replication, it

341

was less consistent than VRC07-523+PGT121, in line with the lower potency of this mAb.

342

VRC01 was used alone in this study because it is planned for use in clinical trials, both as an

343

early HIV-1 immunotherapeutic intervention as well as a prophylactic agent to prevent infection.

344

Since SHIV SF162P3 is about 10-fold less sensitive to VRC01 than most HIV-1 isolates (30,

345

38), we also used a combination of mAbs, VRC07-523 and PGT121, with more potent activity

346

against SHIV-SF163P3. These two mAbs are being developed for clinical use and represent the

347

potential efficacy against HIV-1 when two potent mAbs are used together. For therapeutic

348

purposes, it may also be important to use antibodies to more than one site to avoid the selection

349

of resistant strains in the setting of active virus replication and a diverse swarm of viral genetic

350

variants.

351

Recent studies using the new generation of potent HIV-1-specific monoclonal antibodies

352

in humanized mice, rhesus macaques, and humans demonstrated robust, albeit typically transient,

353

suppression of chronic viremia (15-17, 28). Early HIV-1 diagnosis prior to the peak of viremia

354

is now feasible in a field setting and has been achieved in clinical cohorts spanning multiple

355

countries (Robb et al., submitted). Here, we directly compared mAbs to cART, a benchmark for

356

effective therapy, during the early phase of acute viremia, just prior to peak, and observed

357

remarkably similar viremia suppression. Viremia was still declining 11 days following VRC07-

18

358

523+PGT121 infusion, suggesting that the mAb antiviral activity was sustained for over a week.

359

PGT121 alone was previously reported to completely suppress chronic SHIV viremia (15), and

360

thus the relative contribution of VRC07-523 and PGT121 to the suppression observed here will

361

be important to assess in future studies. Greater acute viremia control by the dual mAb arm

362

relative to VRC01 is consistent with more potent in vitro neutralization of the SHIV challenge

363

virus by both the VRC07-523 and PGT121 mAbs, and thus in vivo therapeutic efficacy was

364

associated with neutralization activity. The ability of mAbs to inhibit acute virus replication to

365

this extent indicates that they represent a potent therapy option during the exponential viral

366

growth phase. To our knowledge, this is the first demonstration of mAbs suppressing acute virus

367

replication. And since mAbs likely employ different antiviral mechanisms than drugs, as

368

discussed below, they have the potential to augment the impact of cART when used together

369

during acute infection.

370

While antibody-mediated selection of resistant viral sequences is an important

371

consideration for passive immunotherapy in chronic HIV-1 infection (28), we think immune

372

pressure by mAbs was unlikely to give rise to less sensitive SHIV variants in this acute infection

373

model due to the clonal nature of the challenge virus and the short 11-day mAb treatment

374

window. Sequence analysis of the challenge stock identified a single minor Env variant, A64W,

375

with no known association with resistance to the mAbs studied herein (data not shown).

376

Our rhesus macaque animal model likely does not adequately recapitulate some

377

important aspects of the use of human antibodies in HIV-1 infection. First, fairly rapid macaque

378

immune responses generated against the human antibodies preclude prolonged antibody activity

379

in vivo. Humoral responses specific for these mAbs were detected within three weeks of

380

infusion in all animals. As a result, mAb half-life is more limited in macaques compared to

19

381

humans. Future studies with simianized antibodies and variants employing mutations to increase

382

FcRn binding and persistence in vivo will help address the long-term potential of mAbs to

383

suppress viral replication (42). Second, some effector functions in the Fc portion of the human

384

mAb may not be optimal in rhesus macaques. While both IgG and cell-surface Fcγ receptors

385

show considerable sequence similarity between humans and macaques, there are species

386

differences with respect to Fcγ receptor tissue expression as well as specificity and affinity for

387

human IgG (43-45), which may have limited the spectrum of human mAb mediated antiviral

388

activities. Third, we used intravenous inoculation to establish a model with consistent and

389

relatively synchronous viremia, whereas most human infection is via a mucosal route. Peak viral

390

loads typically occur slightly earlier with intravenous challenge compared to mucosal delivery.

391

While cART is the cornerstone of HIV-1 treatment, antibodies may offer some potential

392

benefits when added to antiretroviral drugs. Currently available antiretrovirals act only to block

393

new rounds of infection, whereas mAb have the potential to inhibit new rounds of infection

394

through both neutralization and other virion clearance mechanisms, as well as through the

395

targeting of infected cells expressing viral antigens, via Fc receptor mediated effector functions

396

(e.g. antibody-dependent phagocytosis, cell-mediated cytotoxicity, and complement-dependent

397

cytotoxicity). These complementary mechanisms may enable mAbs to further impair virus

398

replication when used in concert with cART. In addition, some data suggest limited ART

399

penetration of lymph node (46), and thus antibodies may have greater access to infected cells

400

residing in secondary lymphoid tissues than drugs. We observed a lower frequency of SHIV-

401

infected naïve CD4 T cells in lymph node following VRC07-523+PGT121 compared to cART.

402

However, it is not clear why only naïve cells would be targeted and not other CD4 T cell subsets.

403

For these reasons, it will be important to measure mAb localization in tissues in future studies,

20

404

including an assessment of tissue penetration as a determinant of mAb antiviral activity. We also

405

saw evidence of enhanced humoral responses during acute infection with mAb therapy versus

406

cART (Fig. 3), with elevated Env-specific antibodies following either VRC01 or VRC07-

407

523+PGT121. We speculate that monoclonal antibody-antigen immune complexes may thus

408

also aid in elicitation of the endogenous adaptive humoral immune response. Antibodies

409

therefore warrant consideration for use in conjunction with cART to enhance the antiviral

410

activity of drugs.

411

In summary, our results show that mAb immunotherapy is effective in controlling acute

412

SHIV replication. Our acute SHIV rhesus macaque model reproduced key aspects of human

413

HIV-1 infection, including robust peak and set-point viremia and effective viral suppression by

414

cART. Moreover, our intent to model interventions in early acute infection clinical cohorts,

415

during Fiebig stages I-III, was achieved by initiating therapy during the days immediately prior

416

to peak viremia. These data provide rationale for assessing the efficacy of combined cART and

417

mAb therapy as well as evaluating efficacy following mucosal infection to further determine the

418

utility of antibody therapy. These pre-clinical studies will inform clinical trials currently being

419

planned in acutely infected individuals that will be required to establish therapeutic efficacy

420

during acute infection.

21

421

Funding information. This work was supported by a cooperative agreement (W81XWH-07-2-

422

0067) between the Henry M. Jackson Foundation for the Advancement of Military Medicine,

423

Inc., and the U.S. Department of Defense (DOD). This research was funded, in part, by the U.S.

424

National Institute of Allergy and Infectious Diseases and in part with federal funds under

425

Contract No. HHSN261200800001E from the National Cancer Institute. The funders had no

426

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

427

publication.

428 429 430

Acknowledgements. We thank Romas Geleziunas and Martha Vazquez from Gilead Sciences,

431

Inc. for generously providing antiretroviral drugs for use in in vivo experiments. We thank the

432

VRC Flow Cytometry Core’s Steve Perfetto and Richard Nguyen for rigorous maintenance of

433

flow cytometers used for measuring antigen-specific T cell responses and sorting T cell

434

populations for viral DNA quantitation. We thank Takuya Yamamoto for assistance with the

435

flow cytometric staining panel for isolating T follicular helper cells. Eli Boritz and Sam Darko

436

sequenced and analyzed diversity of the challenge virus stock. Plasma viral load measurements

437

were performed by Mike Piatak (deceased) with technical assistance from Randy Fast, Kelli

438

Oswald and Rebecca Shoemaker of the Quantitative Molecular Diagnostics Core of the AIDS

439

and Cancer Virus Program of the Frederick National Laboratory for Cancer Research. We thank

440

Brenda Hartman for assistance formatting manuscript figures. We thank Barney Graham for

441

critical reading of the manuscript and input on study design. The views expressed are those of

442

the authors and should not be construed to represent the positions of the Departments of the

443

Army, Defense, or Health and Human Services.

22

444

References

445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488

1.

2.

3.

4.

5. 6.

7.

8.

9. 10.

11.

12.

Archin NM, Vaidya NK, Kuruc JD, Liberty AL, Wiegand A, Kearney MF, Cohen MS, Coffin JM, Bosch RJ, Gay CL, Eron JJ, Margolis DM, Perelson AS. 2012. Immediate antiviral therapy appears to restrict resting CD4+ cell HIV-1 infection without accelerating the decay of latent infection. Proc Natl Acad Sci U S A 109:9523-9528. Ananworanich J, Schuetz A, Vandergeeten C, Sereti I, de Souza M, Rerknimitr R, Dewar R, Marovich M, van Griensven F, Sekaly R, Pinyakorn S, Phanuphak N, Trichavaroj R, Rutvisuttinunt W, Chomchey N, Paris R, Peel S, Valcour V, Maldarelli F, Chomont N, Michael N, Phanuphak P, Kim JH, Group RSS. 2012. Impact of multi-targeted antiretroviral treatment on gut T cell depletion and HIV reservoir seeding during acute HIV infection. PLoS One 7:e33948. Whitney JB, Hill AL, Sanisetty S, Penaloza-MacMaster P, Liu J, Shetty M, Parenteau L, Cabral C, Shields J, Blackmore S, Smith JY, Brinkman AL, Peter LE, Mathew SI, Smith KM, Borducchi EN, Rosenbloom DI, Lewis MG, Hattersley J, Li B, Hesselgesser J, Geleziunas R, Robb ML, Kim JH, Michael NL, Barouch DH. 2014. Rapid seeding of the viral reservoir prior to SIV viraemia in rhesus monkeys. Nature 512:74-77. Sung JA, Lam S, Garrido C, Archin N, Rooney CM, Bollard CM, Margolis DM. 2015. Expanded Cytotoxic T-cell Lymphocytes Target the Latent HIV Reservoir. J Infect Dis doi:10.1093/infdis/jiv022. Archin NM, Margolis DM. 2014. Emerging strategies to deplete the HIV reservoir. Curr Opin Infect Dis 27:29-35. Deng K, Pertea M, Rongvaux A, Wang L, Durand CM, Ghiaur G, Lai J, McHugh HL, Hao H, Zhang H, Margolick JB, Gurer C, Murphy AJ, Valenzuela DM, Yancopoulos GD, Deeks SG, Strowig T, Kumar P, Siliciano JD, Salzberg SL, Flavell RA, Shan L, Siliciano RF. 2015. Broad CTL response is required to clear latent HIV-1 due to dominance of escape mutations. Nature 517:381-385. Denton PW, Long JM, Wietgrefe SW, Sykes C, Spagnuolo RA, Snyder OD, Perkey K, Archin NM, Choudhary SK, Yang K, Hudgens MG, Pastan I, Haase AT, Kashuba AD, Berger EA, Margolis DM, Garcia JV. 2014. Targeted cytotoxic therapy kills persisting HIV infected cells during ART. PLoS Pathog 10:e1003872. Ananworanich J, McSteen B, Robb ML. 2015. Broadly neutralizing antibody and the HIV reservoir in acute HIV infection: a strategy toward HIV remission? Curr Opin HIV AIDS 10:198-206. Kwong PD, Mascola JR. 2012. Human antibodies that neutralize HIV-1: identification, structures, and B cell ontogenies. Immunity 37:412-425. Burton DR, Poignard P, Stanfield RL, Wilson IA. 2012. Broadly neutralizing antibodies present new prospects to counter highly antigenically diverse viruses. Science 337:183-186. West AP, Jr., Scharf L, Scheid JF, Klein F, Bjorkman PJ, Nussenzweig MC. 2014. Structural insights on the role of antibodies in HIV-1 vaccine and therapy. Cell 156:633-648. Lewis GK. 2014. Role of Fc-mediated antibody function in protective immunity against HIV-1. Immunology 142:46-57. 23

489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534

13.

14. 15.

16.

17.

18.

19.

20.

21.

22.

23.

Milligan C, Richardson BA, John-Stewart G, Nduati R, Overbaugh J. 2015. Passively Acquired Antibody-Dependent Cellular Cytotoxicity (ADCC) Activity in HIV-Infected Infants Is Associated with Reduced Mortality. Cell Host Microbe 17:500-506. Ackerman ME, Alter G. 2013. Opportunities to exploit non-neutralizing HIVspecific antibody activity. Curr HIV Res 11:365-377. Barouch DH, Whitney JB, Moldt B, Klein F, Oliveira TY, Liu J, Stephenson KE, Chang HW, Shekhar K, Gupta S, Nkolola JP, Seaman MS, Smith KM, Borducchi EN, Cabral C, Smith JY, Blackmore S, Sanisetty S, Perry JR, Beck M, Lewis MG, Rinaldi W, Chakraborty AK, Poignard P, Nussenzweig MC, Burton DR. 2013. Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys. Nature 503:224-228. Klein F, Halper-Stromberg A, Horwitz JA, Gruell H, Scheid JF, Bournazos S, Mouquet H, Spatz LA, Diskin R, Abadir A, Zang T, Dorner M, Billerbeck E, Labitt RN, Gaebler C, Marcovecchio PM, Incesu RB, Eisenreich TR, Bieniasz PD, Seaman MS, Bjorkman PJ, Ravetch JV, Ploss A, Nussenzweig MC. 2012. HIV therapy by a combination of broadly neutralizing antibodies in humanized mice. Nature 492:118-122. Shingai M, Nishimura Y, Klein F, Mouquet H, Donau OK, Plishka R, BucklerWhite A, Seaman M, Piatak M, Jr., Lifson JD, Dimitrov DS, Nussenzweig MC, Martin MA. 2013. Antibody-mediated immunotherapy of macaques chronically infected with SHIV suppresses viraemia. Nature 503:277-280. Moldt B, Rakasz EG, Schultz N, Chan-Hui PY, Swiderek K, Weisgrau KL, Piaskowski SM, Bergman Z, Watkins DI, Poignard P, Burton DR. 2012. Highly potent HIV-specific antibody neutralization in vitro translates into effective protection against mucosal SHIV challenge in vivo. Proc Natl Acad Sci U S A 109:18921-18925. Mascola JR, Stiegler G, VanCott TC, Katinger H, Carpenter CB, Hanson CE, Beary H, Hayes D, Frankel SS, Birx DL, Lewis MG. 2000. Protection of macaques against vaginal transmission of a pathogenic HIV-1/SIV chimeric virus by passive infusion of neutralizing antibodies. Nat Med 6:207-210. Mascola JR, Lewis MG, Stiegler G, Harris D, VanCott TC, Hayes D, Louder MK, Brown CR, Sapan CV, Frankel SS, Lu Y, Robb ML, Katinger H, Birx DL. 1999. Protection of Macaques against pathogenic simian/human immunodeficiency virus 89.6PD by passive transfer of neutralizing antibodies. J Virol 73:4009-4018. Cavacini LA, Samore MH, Gambertoglio J, Jackson B, Duval M, Wisnewski A, Hammer S, Koziel C, Trapnell C, Posner MR. 1998. Phase I study of a human monoclonal antibody directed against the CD4-binding site of HIV type 1 glycoprotein 120. AIDS Res Hum Retroviruses 14:545-550. Armbruster C, Stiegler GM, Vcelar BA, Jager W, Michael NL, Vetter N, Katinger HW. 2002. A phase I trial with two human monoclonal antibodies (hMAb 2F5, 2G12) against HIV-1. AIDS 16:227-233. Armbruster C, Stiegler GM, Vcelar BA, Jager W, Koller U, Jilch R, Ammann CG, Pruenster M, Stoiber H, Katinger HW. 2004. Passive immunization with the antiHIV-1 human monoclonal antibody (hMAb) 4E10 and the hMAb combination 4E10/2F5/2G12. J Antimicrob Chemother 54:915-920. 24

535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578

24.

25.

26.

27.

28.

29.

30.

31.

Trkola A, Kuster H, Rusert P, Joos B, Fischer M, Leemann C, Manrique A, Huber M, Rehr M, Oxenius A, Weber R, Stiegler G, Vcelar B, Katinger H, Aceto L, Gunthard HF. 2005. Delay of HIV-1 rebound after cessation of antiretroviral therapy through passive transfer of human neutralizing antibodies. Nat Med 11:615-622. Mehandru S, Vcelar B, Wrin T, Stiegler G, Joos B, Mohri H, Boden D, Galovich J, Tenner-Racz K, Racz P, Carrington M, Petropoulos C, Katinger H, Markowitz M. 2007. Adjunctive passive immunotherapy in human immunodeficiency virus type 1infected individuals treated with antiviral therapy during acute and early infection. J Virol 81:11016-11031. Matsushita S, Yoshimura K, Ramirez KP, Pisupati J, Murakami T, Group KDS. 2015. Passive transfer of neutralizing mAb KD-247 reduces plasma viral load in patients chronically infected with HIV-1. AIDS 29:453-462. Ledgerwood JE, Coates EE, Yamshchikov G, Saunders JG, Holman L, Enama ME, DeZure A, Lynch R, Gordon I, Plummer S, Hendel CS, Pegu A, Conan-Cibotti M, Sitar S, Bailer RT, Narpala S, McDermott A, Louder M, O'Dell S, Mohan S, Pandey JP, Schwartz RM, Hu Z, Koup RA, Capparelli E, Mascola JR, Graham BS, Team VRCS. 2015. Safety, Pharmacokinetics, and Neutralization of the Broadly Neutralizing HIV-1 Human Monoclonal Antibody VRC01 in Healthy Adults. Clin Exp Immunol doi:10.1111/cei.12692. Caskey M, Klein F, Lorenzi JC, Seaman MS, West AP, Jr., Buckley N, Kremer G, Nogueira L, Braunschweig M, Scheid JF, Horwitz JA, Shimeliovich I, BenAvraham 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 suppressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature 522:487-491. Rudicell RS, Kwon YD, Ko SY, Pegu A, Louder MK, Georgiev IS, Wu X, Zhu J, Boyington JC, Chen X, Shi W, Yang ZY, Doria-Rose NA, McKee K, O'Dell S, Schmidt SD, Chuang GY, Druz A, Soto C, Yang Y, Zhang B, Zhou T, Todd JP, Lloyd KE, Eudailey J, Roberts KE, Donald BR, Bailer RT, Ledgerwood J, Program NCS, Mullikin JC, Shapiro L, Koup RA, Graham BS, Nason MC, Connors M, Haynes BF, Rao SS, Roederer M, Kwong PD, Mascola JR, Nabel GJ. 2014. Enhanced potency of a broadly neutralizing HIV-1 antibody in vitro improves protection against lentiviral infection in vivo. J Virol 88:12669-12682. 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. 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 GPI, Koff WC, Wilson IA, Burton DR, Poignard P. 2011. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 477:466-470.

25

579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624

32.

33.

34.

35. 36.

37.

38.

39.

40. 41.

42.

43.

44.

Zhou T, Zhu J, Yang Y, Gorman J, Ofek G, Srivatsan S, Druz A, Lees CR, Lu G, Soto C, Stuckey J, Burton DR, Koff WC, Connors M, Kwong PD. 2014. Transplanting supersites of HIV-1 vulnerability. PLoS One 9:e99881. Wu X, Wang C, O'Dell S, Li Y, Keele BF, Yang Z, Imamichi H, Doria-Rose N, Hoxie JA, Connors M, Shaw GM, Wyatt RT, Mascola JR. 2012. Selection pressure on HIV1 envelope by broadly neutralizing antibodies to the conserved CD4-binding site. J Virol 86:5844-5856. Catanzaro AT, Roederer M, Koup RA, Bailer RT, Enama ME, Nason MC, Martin JE, Rucker S, Andrews CA, Gomez PL, Mascola JR, Nabel GJ, Graham BS, Team VRCS. 2007. Phase I clinical evaluation of a six-plasmid multiclade HIV-1 DNA candidate vaccine. Vaccine 25:4085-4092. Foulds KE, Donaldson M, Roederer M. 2012. OMIP-005: Quality and phenotype of antigen-responsive rhesus macaque T cells. Cytometry A 81:360-361. Nishimura Y, Sadjadpour R, Mattapallil JJ, Igarashi T, Lee W, Buckler-White A, Roederer M, Chun TW, Martin MA. 2009. High frequencies of resting CD4+ T cells containing integrated viral DNA are found in rhesus macaques during acute lentivirus infections. Proc Natl Acad Sci U S A 106:8015-8020. Cline AN, Bess JW, Piatak M, Jr., Lifson JD. 2005. Highly sensitive SIV plasma viral load assay: practical considerations, realistic performance expectations, and application to reverse engineering of vaccines for AIDS. J Med Primatol 34:303-312. Pegu A, Yang ZY, Boyington JC, Wu L, Ko SY, Schmidt SD, McKee K, Kong WP, Shi W, Chen X, Todd JP, Letvin NL, Huang J, Nason MC, Hoxie JA, Kwong PD, Connors M, Rao SS, Mascola JR, Nabel GJ. 2014. Neutralizing antibodies to HIV-1 envelope protect more effectively in vivo than those to the CD4 receptor. Sci Transl Med 6:243ra288. Darrah PA, Patel DT, De Luca PM, Lindsay RW, Davey DF, Flynn BJ, Hoff ST, Andersen P, Reed SG, Morris SL, Roederer M, Seder RA. 2007. Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major. Nat Med 13:843-850. Seder RA, Darrah PA, Roederer M. 2008. T-cell quality in memory and protection: implications for vaccine design. Nat Rev Immunol 8:247-258. Wu CY, Kirman JR, Rotte MJ, Davey DF, Perfetto SP, Rhee EG, Freidag BL, Hill BJ, Douek DC, Seder RA. 2002. Distinct lineages of T(H)1 cells have differential capacities for memory cell generation in vivo. Nat Immunol 3:852-858. Saunders KO, Pegu A, Georgiev IS, Zeng M, Joyce MG, Yang ZY, Ko SY, Chen X, Schmidt SD, Haase AT, Todd JP, Bao S, Kwong PD, Rao SS, Mascola JR, Nabel GJ. 2015. Sustained Delivery of a Broadly Neutralizing Antibody in Nonhuman Primates Confers Long-Term Protection against Simian/Human Immunodeficiency Virus Infection. J Virol 89:5895-5903. Trist HM, Tan PS, Wines BD, Ramsland PA, Orlowski E, Stubbs J, Gardiner EE, Pietersz GA, Kent SJ, Stratov I, Burton DR, Hogarth PM. 2014. Polymorphisms and interspecies differences of the activating and inhibitory FcgammaRII of Macaca nemestrina influence the binding of human IgG subclasses. J Immunol 192:792-803. Rogers KA, Scinicariello F, Attanasio R. 2006. IgG Fc receptor III homologues in nonhuman primate species: genetic characterization and ligand interactions. J Immunol 177:3848-3856. 26

625 626 627 628 629 630 631 632 633 634 635

45.

46.

Warncke M, Calzascia T, Coulot M, Balke N, Touil R, Kolbinger F, Heusser C. 2012. Different adaptations of IgG effector function in human and nonhuman primates and implications for therapeutic antibody treatment. J Immunol 188:44054411. Fletcher CV, Staskus K, Wietgrefe SW, Rothenberger M, Reilly C, Chipman JG, Beilman GJ, Khoruts A, Thorkelson A, Schmidt TE, Anderson J, Perkey K, Stevenson M, Perelson AS, Douek DC, Haase AT, Schacker TW. 2014. Persistent HIV-1 replication is associated with lower antiretroviral drug concentrations in lymphatic tissues. Proc Natl Acad Sci U S A 111:2307-2312.

27

636

Table 1. Neutralization potency of anti-HIV-1 mAbs. HIV-1 viruses (>195 strains)

SHIV SF162P3 Antibody

IC50 (mg/ml)

IC80 (mg/ml)

Median IC50 (mg/ml)

Median IC80 (mg/ml)

VRC01

4.28

8.63

0.378

1.01

VRC07-523

0.520

0.975

0.045

0.219

PGT121

0.101

0.190

0.014

0.094

637 638 639 640 641

Figures:

642

levels in plasma. (A) Twenty-four rhesus macaques were challenged intravenously with

643

SHIVSF162P3 and treated on day 10 with either daily ART (n=6), a single infusion of mAb VRC01

644

(n = 6), or a single infusion of two mAbs together (VRC07-523 and PGT121). Six control

645

animals remained untreated. For those animals given mAb treatment, daily ART was initiated 21

646

days after infection. (B) Antibody levels in plasma of rhesus macaques after a single infusion of

647

mAb VRC01, or a combination of mAbs VRC07-523 and PGT121, infused 10 days post SHIV-

648

challenge. Each line represents data for one animal and error bars represent the 95% confidence

649

intervals. The shaded areas correspond to the duration of daily ART treatment. Red dotted line

650

indicates the in vitro neutralization IC80 (µg/ml) against the SHIV-SF162P3 challenge stock.

651

The black dotted line indicates the limit of detection of plasma antibody levels. (C) Geometric

652

mean antibody levels in the plasma for the mAb treatment groups (n=6 per group). The arrow

653

indicates the time of antibody injection and the shaded area represents the period of daily ART.

654

(D) Measured plasma neutralization titers against the SHIV SF162P3 challenge stock.

655

Geometric mean plasma neutralization ID80 titers are shown for the mAb treatment groups (n=6

656

per group; solid red and blue lines) at the indicated time following infection. Predicted ID80

Fig. 1. Experimental treatment schema and VRC01, VRC07-523, and PGT121 antibody

28

657

values calculated from the plasma mAb concentration shown in (B) are overlaid as dotted lines.

658

The predicted ID80 values for the group that received VRC07-523 and PGT121 was a sum of the

659

predicted ID80 value for each mAb. (E) Anti-antibody responses after SHIV challenge. The

660

geometric mean end point titer values for plasma reactivity to VRC01, VRC07-523, and PGT121

661

in infused animals was determined using an ELISA-based approach. The error bars represent the

662

95% confidence interval; the dotted line indicates the limit of detection for the assay. Arrows

663

indicate timing of mAb infusion.

664 665

Fig. 2. SHIV plasma viral loads. (A) Plasma viral loads in rhesus macaques that were left

666

untreated (control), or treated as described above. Each animal is represented by a distinct line.

667

The black arrows indicate day of mAb administration and the shaded areas correspond to the

668

duration of daily ART treatment. (B) Geometric mean plasma viral loads for the untreated and

669

the 3 treatment groups (n=6 per group). The error bars represent the 95% confidence interval at

670

each time point. Peak viremia (C), day 10-21 viremia area under the curve (D), and the slope of

671

viremia decay from peak to day 21 (E) are shown for each animal by treatment group with lines

672

at top indicating significant differences relative to untreated control animals (* p

Human Immunodeficiency Virus Type 1 Monoclonal Antibodies Suppress Acute Simian-Human Immunodeficiency Virus Viremia and Limit Seeding of Cell-Associated Viral Reservoirs.

Combination antiretroviral therapy (cART) administered shortly after human immunodeficiency virus type 1 (HIV-1) infection can suppress viremia and li...
565B Sizes 0 Downloads 8 Views