12
n engl j med 366;14 nejm.org april 5, 2012 1275 The new england journal of medicine established in 1812 april 5, 2012 vol. 366 no. 14 Immune-Correlates Analysis of an HIV-1 Vaccine Efficacy Trial Barton F. Haynes, M.D., Peter B. Gilbert, Ph.D., M. Juliana McElrath, M.D., Ph.D., Susan Zolla-Pazner, Ph.D., Georgia D. Tomaras, Ph.D., S. Munir Alam, Ph.D., David T. Evans, Ph.D., David C. Montefiori, Ph.D., Chitraporn Karnasuta, Ph.D., Ruengpueng Sutthent, M.D., Ph.D., Hua-Xin Liao, M.D., Ph.D., Anthony L. DeVico, Ph.D., George K. Lewis, Ph.D., Constance Williams, B.S., Abraham Pinter, Ph.D., Youyi Fong, Ph.D., Holly Janes, Ph.D., Allan DeCamp, M.S., Yunda Huang, Ph.D., Mangala Rao, Ph.D., Erik Billings, Ph.D., Nicos Karasavvas, Ph.D., Merlin L. Robb, M.D., Viseth Ngauy, M.D., Mark S. de Souza, Ph.D., Robert Paris, M.D., Guido Ferrari, M.D., Robert T. Bailer, Ph.D., Kelly A. Soderberg, Ph.D., Charla Andrews, Sc.M., Phillip W. Berman, Ph.D., Nicole Frahm, Ph.D., Stephen C. De Rosa, M.D., Michael D. Alpert, Ph.D., Nicole L. Yates, Ph.D., Xiaoying Shen, Ph.D., Richard A. Koup, M.D., Punnee Pitisuttithum, M.D., D.T.M.H., Jaranit Kaewkungwal, Ph.D., Sorachai Nitayaphan, M.D., Ph.D., Supachai Rerks-Ngarm, M.D., Nelson L. Michael, M.D., Ph.D., and Jerome H. Kim, M.D. Abstract The authors’ affiliations are listed in the Appendix. Address reprint requests to Dr. Haynes at Duke Human Vaccine Institute, Duke University Medical Center, 2 Genome Ct., Box 103020, Durham, NC 27710, or at [email protected]. N Engl J Med 2012;366:1275-86. Copyright © 2012 Massachusetts Medical Society. Background In the RV144 trial, the estimated efficacy of a vaccine regimen against human immu- nodeficiency virus type 1 (HIV-1) was 31.2%. We performed a case–control analysis to identify antibody and cellular immune correlates of infection risk. Methods In pilot studies conducted with RV144 blood samples, 17 antibody or cellular assays met prespecified criteria, of which 6 were chosen for primary analysis to determine the roles of T-cell, IgG antibody, and IgA antibody responses in the modulation of infection risk. Assays were performed on samples from 41 vaccinees who became infected and 205 uninfected vaccinees, obtained 2 weeks after final immunization, to evaluate whether immune-response variables predicted HIV-1 infection through 42 months of follow-up. Results Of six primary variables, two correlated significantly with infection risk: the bind- ing of IgG antibodies to variable regions 1 and 2 (V1V2) of HIV-1 envelope proteins (Env) correlated inversely with the rate of HIV-1 infection (estimated odds ratio, 0.57 per 1-SD increase; P = 0.02; q = 0.08), and the binding of plasma IgA antibodies to Env correlated directly with the rate of infection (estimated odds ratio, 1.54 per 1-SD increase; P = 0.03; q = 0.08). Neither low levels of V1V2 antibodies nor high levels of Env-specific IgA antibodies were associated with higher rates of infection than were found in the placebo group. Secondary analyses suggested that Env- specific IgA antibodies may mitigate the effects of potentially protective antibodies. Conclusions This immune-correlates study generated the hypotheses that V1V2 antibodies may have contributed to protection against HIV-1 infection, whereas high levels of Env- specific IgA antibodies may have mitigated the effects of protective antibodies. Vaccines that are designed to induce higher levels of V1V2 antibodies and lower levels of Env-specific IgA antibodies than are induced by the RV144 vaccine may have improved efficacy against HIV-1 infection. The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission. Copyright © 2012 Massachusetts Medical Society. All rights reserved.

The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

n engl j med 366;14 nejm.org april 5, 2012 1275

The new england journal of medicineestablished in 1812 april 5, 2012 vol. 366 no. 14

Immune-Correlates Analysis of an HIV-1 Vaccine Efficacy TrialBarton F. Haynes, M.D., Peter B. Gilbert, Ph.D., M. Juliana McElrath, M.D., Ph.D., Susan Zolla-Pazner, Ph.D.,

Georgia D. Tomaras, Ph.D., S. Munir Alam, Ph.D., David T. Evans, Ph.D., David C. Montefiori, Ph.D., Chitraporn Karnasuta, Ph.D., Ruengpueng Sutthent, M.D., Ph.D., Hua-Xin Liao, M.D., Ph.D., Anthony L. DeVico, Ph.D.,

George K. Lewis, Ph.D., Constance Williams, B.S., Abraham Pinter, Ph.D., Youyi Fong, Ph.D., Holly Janes, Ph.D., Allan DeCamp, M.S., Yunda Huang, Ph.D., Mangala Rao, Ph.D., Erik Billings, Ph.D., Nicos Karasavvas, Ph.D., Merlin L. Robb, M.D., Viseth Ngauy, M.D., Mark S. de Souza, Ph.D., Robert Paris, M.D., Guido Ferrari, M.D.,

Robert T. Bailer, Ph.D., Kelly A. Soderberg, Ph.D., Charla Andrews, Sc.M., Phillip W. Berman, Ph.D., Nicole Frahm, Ph.D., Stephen C. De Rosa, M.D., Michael D. Alpert, Ph.D., Nicole L. Yates, Ph.D., Xiaoying Shen, Ph.D., Richard A. Koup, M.D.,

Punnee Pitisuttithum, M.D., D.T.M.H., Jaranit Kaewkungwal, Ph.D., Sorachai Nitayaphan, M.D., Ph.D., Supachai Rerks-Ngarm, M.D., Nelson L. Michael, M.D., Ph.D., and Jerome H. Kim, M.D.

A bs tr ac t

The authors’ affiliations are listed in the Appendix. Address reprint requests to Dr. Haynes at Duke Human Vaccine Institute, Duke University Medical Center, 2 Genome Ct., Box 103020, Durham, NC 27710, or at [email protected].

N Engl J Med 2012;366:1275-86.Copyright © 2012 Massachusetts Medical Society.

Background

In the RV144 trial, the estimated efficacy of a vaccine regimen against human immu-nodeficiency virus type 1 (HIV-1) was 31.2%. We performed a case–control analysis to identify antibody and cellular immune correlates of infection risk.

Methods

In pilot studies conducted with RV144 blood samples, 17 antibody or cellular assays met prespecified criteria, of which 6 were chosen for primary analysis to determine the roles of T-cell, IgG antibody, and IgA antibody responses in the modulation of infection risk. Assays were performed on samples from 41 vaccinees who became infected and 205 uninfected vaccinees, obtained 2 weeks after final immunization, to evaluate whether immune-response variables predicted HIV-1 infection through 42 months of follow-up.

Results

Of six primary variables, two correlated significantly with infection risk: the bind-ing of IgG antibodies to variable regions 1 and 2 (V1V2) of HIV-1 envelope proteins (Env) correlated inversely with the rate of HIV-1 infection (estimated odds ratio, 0.57 per 1-SD increase; P = 0.02; q = 0.08), and the binding of plasma IgA antibodies to Env correlated directly with the rate of infection (estimated odds ratio, 1.54 per 1-SD increase; P = 0.03; q = 0.08). Neither low levels of V1V2 antibodies nor high levels of Env-specific IgA antibodies were associated with higher rates of infection than were found in the placebo group. Secondary analyses suggested that Env-specific IgA antibodies may mitigate the effects of potentially protective antibodies.

Conclusions

This immune-correlates study generated the hypotheses that V1V2 antibodies may have contributed to protection against HIV-1 infection, whereas high levels of Env-specific IgA antibodies may have mitigated the effects of protective antibodies. Vaccines that are designed to induce higher levels of V1V2 antibodies and lower levels of Env-specific IgA antibodies than are induced by the RV144 vaccine may have improved efficacy against HIV-1 infection.

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.

Page 2: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

n engl j med 366;14 nejm.org april 5, 20121276

In clinical trials that show the effi-cacy of a vaccine, the identification of immune responses that are predictive of trial outcomes

generates hypotheses about which of those re-sponses are responsible for protection.1-3 The RV144 phase 3 trial in Thailand (ClinicalTrials .gov number, NCT00223080) was an opportunity to perform such a hypothesis-generating analysis for a human immunodeficiency virus type 1 (HIV-1) vaccine.4 Studies involving patients with HIV-1 in-fection in whom the disease did not progress in the long term have shown that cellular responses control the disease,5 and passive infusion of neu-tralizing antibodies prevents infection with chi-meric simian–human immunodeficiency virus (SHIV).6,7 Antibodies as well as T-cell responses to HIV-1 have been shown to protect vaccinated nonhuman primates from infection with simian immunodeficiency virus (SIV) or SHIV.8-15 An analy sis of a phase 3 trial of a glycoprotein 120 (gp120) B/B vaccine (AIDSVAX B/B), which did not show efficacy against HIV-1, showed that vaccine-specific neutralizing antibody, antibody inhibition of CD4 molecule binding to HIV-1 envelope pro-teins (Env), and antibody-dependent, cell-mediated viral inhibition were associated with reduced in-fection rates among vaccine recipients.16,17

The RV144 trial of the canarypox vector vac-cine (ALVAC-HIV [vCP1521]) plus the gp120 AIDSVAX B/E vaccine showed an estimated vac-cine efficacy of 31.2% for the prevention of HIV-1 infection over a period of 42 months after the first of four planned vaccinations.4 This result enabled a systematic search for immune corre-lates of infection risk that may be relevant for protection. Building on prior work,18,19 our con-sortium conducted a two-stage evaluation of vac-cine-evoked antibody responses, innate immune responses, and cellular immune responses.20 First, 17 assay types were selected from 32 pilot assay types on the basis of reproducibility, ability to detect postvaccine responses, and uniqueness of responses detected, from which 6 primary assay variables were selected. Second, the selected as-says in primary analyses (6 assays) and second-ary analyses (152 assays) were performed on cryopreserved blood samples from vaccinees who became infected (case patients) and on a fre-quency-matched set of samples from uninfected vaccinees (controls) to determine the association of immune-response variables with HIV-1 infec-tion risk.

Me thods

Study ProceduresCase–Control Sampling DesignPatients enrolled in the RV144 trial were vacci-nated at weeks 0, 4, 12, and 24, and immune re-sponses at week 26 were evaluated as immune correlates of infection risk4 (Fig. 1). We assessed vaccine-induced immune responses at peak im-munogenicity (week 26 [2 weeks after the final immunization]) in vaccinees who acquired HIV-1 infection after week 26 (41 vaccinated case pa-tients) as compared with vaccinees who did not acquire infection over a follow-up period of 42 months (205 vaccinated controls). Vaccinated case patients were documented to be free of HIV-1 in-fection at week 24 and to have later received a diagnosis of infection.4 The control vaccinees were selected from a stratified random sample of vac-cine recipients who were documented to be free of HIV-1 infection at 42 months. Patients were strat-ified according to sex, the number of vaccina-tions received (of four planned), and per-protocol

Figure 1 (facing page). Sample Selection for the Case–Control Study.

Patients enrolled in the RV144 study were vaccinated at weeks 0, 4, 12, and 24, and immune responses at week 26 were evaluated as immune correlates of infection risk. The vaccinated case patients were documented as not having HIV-1 infection at week 24 and as having later re-ceived a diagnosis of infection. The vaccine recipients who served as controls were selected from a stratified random sample of vaccine recipients who were docu-mented as not having HIV-1 infection at the last study visit, at 42 months. Of the 7010 HIV-uninfected vaccinat-ed controls eligible for the case–control sample, only those for whom plasma and peripheral-blood mononu-clear cell (PBMC) specimens were available at all later time points and who were not part of previous immuno-genicity-testing cohorts (6899 patients) were included. For vaccine recipients who were included in the sample, 6 strata with 1 or more case patients are shown; the re-maining strata had 0 case patients and 111 controls. All humoral assays were performed in plasma samples from all case patients and all controls (row A). Data on intra-cellular cytokine staining of PBMCs (row B) were missing for 15% of patients (owing to assay quality-control is-sues, including an aberrant batch of samples in 24 pa-tients and high values for the assay negative control in 18). Data on multiplex bead assay (Luminex) of PBMCs (row C) were missing for 13% of patients (owing to high values for the assay negative control in 36 patients). Inj denotes injection with vaccine or placebo, and PP per-protocol cohort (i.e., patients who received all four injec-tions as previously described4).

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.

Page 3: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

Immune-Correlates Analysis of an HIV-1 Vaccine Trial

n engl j med 366;14 nejm.org april 5, 2012 1277

16,3

95 D

id n

ot h

ave

HIV

-1 in

fect

ion

16,4

02 P

atie

nts

unde

rwen

t ran

dom

izat

ion 7

Wer

e H

IV-in

fect

ed a

t ran

dom

izat

ion

and

wer

e ex

clud

ed

8197

Wer

e as

sign

ed to

rec

eive

vac

cine

51 B

ecam

e H

IV-in

fect

ed d

urin

g th

e st

udy

8146

Rem

aine

d H

IV-u

ninf

ecte

d du

ring

the

stud

y

8198

Wer

e as

sign

ed to

rec

eive

pla

cebo

74 B

ecam

e H

IV-in

fect

ed d

urin

g th

e st

udy

8124

Rem

aine

d H

IV-u

ninf

ecte

d du

ring

the

stud

y

Vac

cine

cas

e–co

ntro

l coh

ort e

xclu

sion

s10

Wer

e H

IV-in

fect

ed5

Wer

e in

fect

ed b

efor

e w

k 26

5 H

ad m

issi

ng p

lasm

a an

d PB

MC

sam

ples

at w

k 26

1136

Wer

e H

IV-u

ninf

ecte

d74

With

drew

bef

ore

wk

2623

7 H

ad m

issi

ng in

fect

ion

stat

us82

5 H

ad m

issi

ng p

lasm

a an

dPB

MC

sam

ples

at w

k 26

Plac

ebo

case

–con

trol

coh

ort e

xclu

sion

s 2

4 W

ere

HIV

-infe

cted

and

wer

e no

tin

clud

ed in

per

-pro

toco

l pop

u-la

tion

1884

Wer

e H

IV-u

ninf

ecte

d18

08 W

ere

not i

nclu

ded

in p

er-

prot

ocol

pop

ulat

ion

20 W

ithdr

ew b

efor

e w

k 26

56 H

ad m

issi

ng p

lasm

a an

dPB

MC

sam

ples

at w

k 26

7051

Wer

e el

igib

le fo

r ca

se–c

ontr

ol s

ampl

ing

(41

wer

e H

IV-in

fect

ed c

ase

patie

nts;

701

0 w

ere

HIV

-uni

nfec

ted

cont

rols

) st

ratif

ied

acco

rdin

g to

sex,

per

-pro

toco

l sta

tus,

and

no.

of v

acci

natio

ns

6290

Wer

e el

igib

le fo

r ca

se–c

ontr

ol s

ampl

ing

(50

wer

e H

IV-in

fect

ed c

ase

patie

nts;

624

0 w

ere

HIV

-uni

nfec

ted

cont

rols

) st

ratif

ied

acco

rdin

g to

sex,

per

-pro

toco

l sta

tus,

and

no.

of v

acci

natio

ns

Wom

en, 4

Inj P

P12

Cas

e pa

-tie

nts

2377

Con

trol

s

Wom

en, 4

Inj P

P1

Cas

e pa

-tie

nt22

8 C

ontr

ols

Wom

en, 3

Inj P

P1

Cas

e pa

-tie

nt62

Con

trol

s

Wom

en, 2

Inj P

P1

Cas

e pa

-tie

nt53

Con

trol

s

Men

, 4 In

j PP

24 C

ase

pa-

tient

s36

96 C

ontr

ols

Wom

en, 4

Inj P

P21

Cas

e pa

-tie

nts

2420

Con

trol

s

Men

, 4 In

j PP

29 C

ase

pa-

tient

s38

20 C

ontr

ols

Men

, 4 In

j PP

2 C

ase

pa-

tient

s48

3 C

ontr

ols

All

HIV

-infe

cted

cas

e pa

tient

s in

clud

ed in

sam

ple;

5:1

str

atifi

ed r

ando

m s

ampl

e of

con

trol

s:ca

se p

atie

nts

Stra

tifie

d ra

ndom

sam

ple

of 2

0 H

IV-

infe

cted

cas

e pa

tient

s an

d 20

con

trol

s

A B C

12 C

ase

patie

nts

60 C

ontr

ols

10 C

ase

patie

nts

55 C

ontr

ols

12 C

ase

patie

nts

50 C

ontr

ols

1 C

ase

patie

nt5

Con

trol

s

1 C

ase

patie

nt4

Con

trol

s

1 C

ase

patie

nt4

Con

trol

s

1 C

ase

patie

nt5

Con

trol

s

1 C

ase

patie

nt5

Con

trol

s

1 C

ase

patie

nt5

Con

trol

s

1 C

ase

patie

nt5

Con

trol

s

1 C

ase

patie

nt4

Con

trol

s

1 C

ase

patie

nt4

Con

trol

s

24 C

ase

patie

nts

120

Con

trol

s

23 C

ase

patie

nts

97 C

ontr

ols

23 C

ase

patie

nts

102

Con

trol

s

2 C

ase

patie

nts

10 C

ontr

ols

2 C

ase

patie

nts

7 C

ontr

ols

2 C

ase

patie

nts

10 C

ontr

ols

10 C

ase

patie

nts

10 C

ontr

ols

10 C

ase

patie

nts

10 C

ontr

ols

9 C

ase

patie

nts

6 C

ontr

ols

9 C

ase

patie

nts

10 C

ontr

ols

8 C

ase

patie

nts

9 C

ontr

ols

8 C

ase

patie

nts

10 C

ontr

ols

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.

Page 4: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

n engl j med 366;14 nejm.org april 5, 20121278

status, as previously defined.4 For each of the eight strata, the number of vaccinated case pa-tients was noted, and samples from five times as many vaccinated controls were obtained. The as-says were also performed on random samples from 20 infected placebo recipients and 20 unin-fected placebo-recipient controls (Fig. 1).

Immune-Response Variables and Tiered Structure of the Correlates AnalysisThe correlates study was preceded by pilot stud-ies from November 2009 through July 201120 (Fig. S1 in the Supplementary Appendix, avail-able with the full text of this article at NEJM.org). Pilot assays were performed on samples taken at baseline and week 26 from 50 to 100 uninfected RV144 participants (80% of whom were vaccine recipients and 20% of whom were placebo recipi-ents) and scored according to four statistical cri-teria: a low false positive rate on the basis of samples from placebo and vaccine recipients at baseline, a large dynamic range of vaccine-induced immune responses, nonredundancy of responses (low correlations), and high reproducibility.

Of the 32 types of antibody, T-cell, and innate immunity assays evaluated in pilot studies, 17 met these criteria, from which 6 primary variables were chosen for assessment as correlates of in-fection risk. The purpose was to restrict the primary analysis to a limited number of vari-ables in order to optimize the statistical power for showing a correlation of risk between vacci-nated persons who acquired versus those who did not acquire HIV-1. The primary variables included 5 Env-specific antibody responses and 1 cellular response: the binding of plasma IgA antibodies to Env, the avidity of IgG antibodies for Env, antibody-dependent cellular cytotoxicity, HIV-1 neutralizing antibodies, the binding of IgG antibodies to variable regions 1 and 2 (V1V2) of the gp120 Env, and the level of Env-specific CD4+ T cells (for details, see the Supplementary Appendix). All 17 types of immune assays and their 152 component variables were also included in the secondary correlates analyses (Tables S1 and S2 in the Supplementary Appendix).

Secondary variables were drawn from the re-maining 152 assays selected from pilot assay studies; they were evaluated to help interpret the results of the primary analysis and to generate additional hypotheses (Table S1 in the Supple-mentary Appendix). For the sensitivity analysis, immune-response variables that were closely re-

lated to the six primary variables (within the same assay type) were substituted for each of the primary variables into the multivariable model (eight variables, with three individual variables paired to the primary variable of neutralizing antibodies) (Table S2 in the Supplementary Ap-pendix). All assays were performed by personnel who were unaware of treatment assignments and case–control status.

Statistical Analysis

The statistical analysis plan was finalized before data analysis, and the primary results were con-firmed by an independent statistical group (EMMES). This statistical analysis plan prescribed the statistical methods and the definitions of the immune-response variables (for details, see the Supplementary Appendix). In the primary analy-sis, logistic-regression and Cox proportional-hazards models that accounted for the sampling design were used.21,22 The analyses controlled for sex and baseline self-reported behavioral risk factors, as defined previously.4

The six primary variables were evaluated in multivariate and univariate models. The immune-response variables were modeled quantitatively and with the use of categories based on thirds of response (low, medium, and high) in the vac-cine group. The q value is the minimal false dis-covery rate at which a statistical test result may be called significant. The q values were used for multiplicity correction, with a significance thresh-old of less than 0.20, indicating that any detected correlate can have up to a 20% chance of false positivity. This approach was designed to opti-mize the discovery of correlates at the expense of an acceptable risk of false positive results.

Because of the small number of infected vac-cinees, this study had statistical power to detect only strong correlates of infection risk, with 80% power to detect a 50% reduction in the infection rate per 1-SD increment in normally distributed immune responses. The 152 secondary variables were assessed with the same univariate regres-sion analyses as the primary variables were, to generate exploratory hypotheses for further study (Table S1 in the Supplementary Appendix).

R esult s

Primary Variables in Case–Control Analyses

Vaccine-induced immune responses were detect-ed with all primary assay variables, with suffi-

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.

Page 5: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

Immune-Correlates Analysis of an HIV-1 Vaccine Trial

n engl j med 366;14 nejm.org april 5, 2012 1279

cient dynamic ranges to support regression anal-yses (Fig. 2). Figure S2 in the Supplementary Appendix shows that the six primary variables were only weakly correlated with each other, ver-ifying that the process for selecting the primary variables yielded nonredundant primary im-mune-response variables.

First, when we analyzed the six quantitative variables together in multivariate logistic-regres-sion models, there was a trend toward the predic-tion of infection risk by the variables (P = 0.08 for all six variables together). In this model, IgG avidity, antibody-dependent cellular cytotoxicity, neutralizing antibodies, and level of Env-specific CD4+ T cells did not significantly predict the HIV-1 infection rate (q>0.20). However, IgG binding to a scaffolded V1V2 antigen was in-versely correlated with infection (estimated odds ratio, 0.57 per 1-SD increase; P = 0.02; q = 0.08), and composite IgA antibody binding to an Env panel was directly correlated with infection (es-timated odds ratio, 1.54 per 1-SD increase; P = 0.03; q = 0.08) (Table 1). The univariate analy-ses of V1V2 and IgA responses yielded odds-ratio estimates of 0.70 and 1.39, respectively, with slightly reduced significance (P = 0.06, q=0.19, and P=0.05, q= 0.19, respectively) (Table 1).

Parallel multivariate analyses with the Cox model yielded similar results, with an overall multivariate P value of 0.06 and multivariate haz-ard-ratio estimates of 0.57 for the V1V2 response (P = 0.01, q = 0.06) and 1.58 for the IgA response (P = 0.02, q = 0.06) (Table S3 in the Supplemen-tary Appendix). When the multivariate analysis was repeated with only the V1V2 and IgA im-mune-response variables, the overall P value was 0.01 for both the logistic-regression and Cox re-gression models.

The logistic-regression analyses of the six primary variables categorized into low, medium, and high levels of response yielded odds-ratio estimates that were consistent with those in the quantitative variable analysis. There was no evi-dence that IgG avidity, antibody-dependent cel-lular cytotoxicity, neutralizing antibodies, or Env-specific CD4+ T cells were associated with infection risk (q>0.20). Comparison of high and low levels showed an inverse correlation be-tween V1V2 antibody levels and the risk of infec-tion (estimated odds ratio, 0.29; P = 0.02) and a trend toward a direct correlation of Env-specific IgA antibody level with infection risk (estimated odds ratio, 1.89; P = 0.17) (Table 1). However, these

categorical-model results had reduced significance levels for testing an equal infection rate across low, medium, and high responses (V1V2 antibod-ies, q = 0.23; Env-specific IgA antibodies, q = 0.23), which may be related to the division of responses into thirds, which can reduce statistical power.

Figure 3 shows curves for the cumulative in-cidence of HIV-1 infection with each primary variable among vaccine recipients according to the level of response (low, medium, or high) and for all placebo recipients who were negative for HIV-1 infection at week 24. These curves under-score the increased rate of infection among vac-cine recipients with high levels of Env-specific IgA antibodies, as compared with other vaccine recipients, and the decreased rate of infection among vaccine recipients with high levels of V1V2 antibodies.

Risk of Infection with Vaccine, According to V1V2 or Env-Specific IgA Antibody Level, as Compared with Placebo

Env-specific IgA responses were directly associ-ated with infection risk in the vaccine group, raising the possibility that a vaccine-elicited plas-ma Env-specific IgA response increased the risk of infection in the RV144 trial. To evaluate this possibility, we used logistic and Cox regression to estimate vaccine efficacy as 1 minus the odds (hazard) ratio for infection among vaccinees with low, medium, and high Env-specific IgA re-sponses, as compared with all placebo recipients who were HIV-1-negative at week 24 (Fig. S3 in the Supplementary Appendix). We found that neither low levels of V1V2 antibodies nor high levels of Env-specific IgA antibodies in vaccinees were associated with higher rates of infection than were found among placebo recipients (Fig. S3 in the Supplementary Appendix). These data suggest that vaccine-induced IgA levels did not confer an added risk of infection, as compared with placebo, and therefore were not infection-enhancing antibodies.

Interaction analyses were performed with lo-gistic-regression and Cox regression models to test for interactions of Env-specific IgA antibod-ies and of V1V2 antibodies with the other five primary variables. The analysis showed no inter-action of any primary variables with V1V2 anti-bodies but did show significant interactions of Env-specific IgA antibodies with IgG avidity, antibody-dependent cellular cytotoxicity, neu-tralizing antibodies, and CD4+ T cells (q<0.20).

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.

Page 6: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

n engl j med 366;14 nejm.org april 5, 20121280

Optical Density at a Wavelengthof 405 nm

2.0

1.0

0.2

0.5

Not

Infe

cted

Infe

cted

Not

Infe

cted

Infe

cted

BA

IgG

Ant

ibod

ies

Bin

ding

to V

1V2

Vac

cine

, Not

Infe

cted

Vac

cine

, Inf

ecte

d32

.239

.032

.239

.035

.622

.0

Low

Med

ium

Imm

une

Res

pons

e (%

)H

igh

Plac

ebo

Vac

cine

[Response Units×(1÷DissociationRate in sec)]×10−5

50.0 5.0

20.0

100.

0

0.2

10.0 2.0

1.0

Not

Infe

cted

Infe

cted

Not

Infe

cted

Infe

cted

Avi

dity

of I

gG A

ntib

odie

s fo

r En

v

Vac

cine

, Not

Infe

cted

Vac

cine

, Inf

ecte

d32

.234

.133

.731

.733

.234

.1

Low

Med

ium

Imm

une

Res

pons

e (%

)H

igh

Plac

ebo

Vac

cine

Partial Area between theCurves (RLUs)

0.15

0.10

−0.0

5

0.05

0.00

Not

Infe

cted

Infe

cted

Not

Infe

cted

Infe

cted

Ant

ibod

y-D

epen

dent

Cel

lula

r C

ytot

oxic

ity

Vac

cine

, Not

Infe

cted

Vac

cine

, Inf

ecte

d49

.353

.724

.426

.826

.319

.5

Low

Med

ium

Imm

une

Res

pons

e (%

)H

igh

Plac

ebo

Vac

cine

Log MFI

6 4 02

AUC-MB

2.2

2.0

1.4

1.6

1.0

1.2

1.8

Not

Infe

cted

Infe

cted

Not

Infe

cted

Infe

cted

IgA

Ant

ibod

ies

Bin

ding

to E

nv

Vac

cine

, Not

Infe

cted

Vac

cine

, Inf

ecte

d33

.729

.336

.124

.430

.246

.3

Low

Med

ium

Imm

une

Res

pons

e (%

)H

igh

Plac

ebo

Vac

cine

Not

Infe

cted

Infe

cted

Not

Infe

cted

Infe

cted

Neu

tral

izin

g A

ntib

odie

s

Vac

cine

, Not

Infe

cted

Vac

cine

, Inf

ecte

d35

.124

.431

.741

.533

.234

.1

Low

Med

ium

Imm

une

Res

pons

e (%

)H

igh

Plac

ebo

Vac

cine

Net Percentage of Cytokine-Expressing CD4+ Cells

1.2

1.0

0.6

0.4

0.2

0.0

0.8

Not

Infe

cted

Infe

cted

Not

Infe

cted

Infe

cted

Env-

Spec

ific

CD

4+ T

Cel

ls

Vac

cine

, Not

Infe

cted

Vac

cine

, Inf

ecte

d31

.442

.133

.731

.634

.926

.3

Low

Med

ium

Imm

une

Res

pons

e (%

)H

igh

Plac

ebo

Vac

cine

Wom

en w

ith h

igh

imm

une

resp

onse

Wom

en w

ith m

ediu

m im

mun

ere

spon

seW

omen

with

low

imm

une

resp

onse

Men

with

low

imm

une

resp

onse

Men

with

hig

h im

mun

ere

spon

seM

en w

ith m

ediu

m im

mun

ere

spon

se

***

***

**

0.5

Figu

re 2

. Dis

trib

utio

n of

the

Six

Pri

mar

y Im

mun

e-R

espo

nse

Vari

able

s in

Inf

ecte

d an

d U

ninf

ecte

d Va

ccin

e an

d Pl

aceb

o R

ecip

ient

s in

the

Cas

e–C

ontr

ol S

tudy

.

Pane

l A in

clud

es th

e tw

o id

entif

ied

imm

une

corr

elat

es o

f ris

k. P

anel

B in

clud

es th

e re

mai

ning

four

prim

ary

imm

une-

resp

onse

var

iabl

es. B

ox p

lots

sho

w th

e 25

th p

erce

ntile

(lo

wer

edg

e of

th

e bo

x), 5

0th

perc

entil

e (h

oriz

onta

l lin

e in

the

box)

, and

75t

h pe

rcen

tile

(upp

er e

dge

of th

e bo

x) fo

r th

e si

x pr

imar

y va

riabl

es, w

ith p

atie

nts

stra

tifie

d ac

cord

ing

to H

IV-1

infe

ctio

n st

atus

and

tr

eatm

ent a

ssig

nmen

t. A

dditi

onal

cha

ract

eris

tics

of th

ese

patie

nts,

incl

udin

g se

x an

d im

mun

e-re

spon

se c

ateg

orie

s, a

re in

dica

ted

by th

e co

lor

and

shap

e of

the

poin

ts. L

ow, m

ediu

m, a

nd

high

imm

une

resp

onse

s at

wee

k 26

wer

e us

ed to

div

ide

the

vacc

ine

grou

p in

to th

irds;

med

ium

res

pons

e is

indi

cate

d by

the

gray

hor

izon

tal b

ar. O

ptic

al d

ensi

ty w

as m

easu

red

by m

eans

of

an e

nzym

e-lin

ked

imm

unos

orbe

nt a

ssay

at a

wav

elen

gth

of 4

05 n

m. L

og M

FI is

the

natu

ral l

og tr

ansf

orm

atio

n of

the

med

ian

fluor

esce

nce

inte

nsit

y (M

FI).

The

avid

ity

scor

e is

[res

pons

e un

its ×

(1 ÷

dis

soci

atio

n ra

te in

sec

onds

)] ×

10−5

. The

par

tial a

rea

betw

een

the

curv

es is

the

sum

of t

he d

iffer

ence

s ov

er th

e fir

st fo

ur d

ilutio

ns b

etw

een

the

read

outs

at w

eek

0 an

d w

eek

26,

mea

sure

d in

log 1

0-t

rans

form

ed r

elat

ive

light

uni

ts (

RLU

s). T

he a

rea

unde

r th

e m

agni

tude

–bre

adth

cur

ve (

AU

C-M

B)

is th

e av

erag

e lo

g 10-t

rans

form

ed 5

0% in

hibi

tory

con

cent

ratio

n in

re-

spon

se to

a p

anel

of s

ix p

seud

oviru

ses.

The

net

per

cent

age

of c

ytok

ine-

expr

essi

ng C

D4+

T c

ells

is th

e pe

rcen

tage

of l

ive

CD

3+ C

D4+

T c

ells

exp

ress

ing

CD

154,

inte

rleu

kin-

2, in

terf

eron

-γ, o

r tu

mor

nec

rosi

s fa

ctor

α m

inus

the

nega

tive

cont

rol v

alue

. The

I ba

rs in

dica

te th

e m

ost e

xtre

me

data

poi

nts,

whi

ch a

re n

o m

ore

than

1.5

tim

es th

e in

terq

uart

ile r

ange

from

the

box.

The

dis

-tr

ibut

ion

plot

s of

the

six

prim

ary

varia

bles

and

sen

sitiv

ity

varia

bles

are

sho

wn

in F

igur

es S

5 th

roug

h S1

1 in

the

Supp

lem

enta

ry A

ppen

dix.

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.

Page 7: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

Immune-Correlates Analysis of an HIV-1 Vaccine Trial

n engl j med 366;14 nejm.org april 5, 2012 1281

Table 1. Odds Ratios for HIV-1 Infection in Univariate and Multivariate Analyses of the Six Primary Variables.*

Variable Multivariate Logistic Regression Univariate Logistic Regression

Odds Ratio(95% CI) P Value q Value

Odds Ratio(95% CI) P Value q Value

Quantitative variables

IgA antibodies binding to Env 1.54 (1.05–2.25) 0.03 0.08 1.39 (1.00–1.93) 0.05 0.19

Avidity of IgG antibodies for Env 0.81 (0.50–1.30) 0.37 0.56 0.93 (0.65–1.34) 0.70 0.81

ADCC 0.92 (0.62–1.37) 0.68 0.68 0.96 (0.68–1.35) 0.81 0.81

Neutralizing antibodies 1.37 (0.82–2.27) 0.23 0.45 1.08 (0.76–1.54) 0.67 0.81

gp70-V1V2 binding 0.57 (0.36–0.90) 0.02 0.08 0.70 (0.49–1.02) 0.06 0.19

CD4+ T cells 1.09 (0.79–1.49) 0.61 0.68 1.08 (0.79–1.47) 0.64 0.81

Categorical variables

IgA antibodies binding to Env

Medium vs. low response 0.68 (0.25–1.83) 0.45 0.71 (0.28–1.77) 0.46

High vs. low response 1.89 (0.75–4.74) 0.17 1.55 (0.69–3.48) 0.29

Overall response† 0.07 0.23 0.18 0.54

Avidity of IgG antibodies for Env

Medium vs. low response 0.67 (0.26–1.75) 0.42 0.79 (0.34–1.84) 0.59

High vs. low response 0.83 (0.30–2.30) 0.72 0.87 (0.38–2.01) 0.75

Overall response† 0.71 0.84 0.86 0.86

ADCC

Medium vs. low response 0.93 (0.39–2.19) 0.86 0.96 (0.43–2.14) 0.92

High vs. low response 0.59 (0.22–1.59) 0.30 0.60 (0.24–1.48) 0.27

Overall response† 0.57 0.84 0.53 0.71

Neutralizing antibodies

Medium vs. low response 2.08 (0.78–5.57) 0.14 1.58 (0.66–3.76) 0.31

High vs. low response 2.43 (0.77–7.67) 0.13 1.29 (0.53–3.14) 0.57

Overall response† 0.26 0.52 0.59 0.71

gp70-V1V2 binding

Medium vs. low response 0.68 (0.28–1.62) 0.38 0.84 (0.38–1.84) 0.67

High vs. low response 0.29 (0.10–0.86) 0.02 0.43 (0.18–1.05) 0.06

Overall response† 0.08 0.23 0.16 0.54

CD4+ T cells

Medium vs. low response 0.84 (0.35–2.02) 0.69 0.62 (0.27–1.45) 0.27

High vs. low response 0.77 (0.31–1.91) 0.57 0.51 (0.21–1.23) 0.13

Overall response† 0.84 0.84 0.28 0.57

* For each variable, the odds ratio is reported per 1-SD increase and for the comparison of medium and high responses with low responses. ADCC denotes antibody-dependent cellular cytotoxicity, and gp70-V1V2 the binding level of IgG anti-bodies to HIV-1 Env V1V2.

† To test the overall response, a two-sided P value from a Wald test of the null hypothesis of an equal infection rate in the low, medium, and high response subgroups was calculated.

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.

Page 8: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

n engl j med 366;14 nejm.org april 5, 20121282

Thus, in the presence of high levels of Env-spe-cific IgA antibodies, none of these four variables correlated with the risk of infection, whereas with low levels of Env-specific IgA antibodies, all four variables had inverse correlations with the risk of infection that were of borderline sig-nificance (Table S4 in the Supplementary Ap-pendix).

Secondary and Exploratory Analyses

In the sensitivity analysis that substituted each of the secondary analysis variables with the prima-ry variable, the significance levels tended to be similar or lower, with the exceptions that neu-tralization of TH023.6, neutralization of clade AE viruses in the A3R5 assay, and magnitude of induced cytokines measured in peripheral-blood mononuclear cells had q values of less than 0.20 (although at P>0.05) (Tables S2 and S5 in the Supplementary Appendix).

Of the 152 secondary variables analyzed, only 2 had q values of less than 0.20. These 2 vari-ables were IgA antibody binding to group A consensus Env gp140 (odds ratio for positive vs. negative responses, 3.71; P = 0.001; q = 0.10) and IgA antibody binding to a gp120 Env first con-stant (C1) region peptide (MQEDVISLWDQSLKP-CVKLTPLCV) (odds ratio for positive vs. negative responses, 3.15; P = 0.003; q = 0.13) (Table S1 in the Supplementary Appendix).

Discussion

We report the results of an immune-correlates analysis of the RV144 HIV-1 vaccine efficacy trial. This correlates study was designed to be hypoth-esis-generating and sensitive for discovering strong correlates of infection risk.23 An identi-fied correlate of infection risk could be a cause of vaccine-induced protection against HIV-1 infec-tion, a surrogate for other unidentified immune responses that are actually responsible for pro-tection, or a marker of HIV-1 exposure or suscep-tibility to infection.1-3 To determine whether a correlate of infection risk is a cause of vaccine protection, it must be tested in additional clinical vaccine efficacy trials or tested in animal mod-els.1-3 Extensive pilot immunogenicity studies re-vealed 17 T-cell, antibody, and innate immunity assays that were prioritized into prespecified pri-mary and secondary analyses in order to maxi-

A

Prob

abili

ty o

f Acq

uiri

ng H

IV In

fect

ion

Prob

abili

ty o

f Acq

uiri

ng H

IV In

fect

ion

1.0

0.8

0.6

0.4

0.2

0.00 12 24 36

0.008

0.006

0.004

0.002

0.0000 12 24 36

Months since Wk 26 Visit

IgG Antibodies Binding to V1V2

No. at Risk (no. of infections)PlaceboLow responseMedium responseHigh response

6267 (0)2111 (0)2312 (0)2563 (0)

6199 (24)2105 (6)2304 (8)2560 (3)

6127 (11)2099 (6)2302 (2)2556 (4)

693 (13)210 (4)180 (6)264 (2)

1.0

0.8

0.6

0.4

0.2

0.00 12 24 36

0.008

0.006

0.004

0.002

0.0000 12 24 36

Months since Wk 26 Visit

IgA Antibodies Binding to Env

No. at Risk (no. of infections)PlaceboLow responseMedium responseHigh response

6267 (0)2276 (0)2539 (0)2172 (0)

6199 (24)2273 (3)2535 (4)2162 (10)

6127 (11)2268 (5)2533 (2)2157 (5)

693 (13)282 (4)202 (4)170 (4)

Placebo

Vaccine, low response

Vaccine, medium response

Vaccine, high response

Figure 3 (and facing page). Estimated Cumulative HIV-1 Incidence Curves for the Six Primary Immune-Response Variables.

Because the overall infection rate in the RV144 trial was low, at 0.234 cases per 100 person-years for the vaccine and placebo groups combined, expres-sion of the cumulative HIV-1 incidence curves for the six primary immune-response variables on a scale of infection probabilities from 0.0 to 1.0 does not allow for an analysis of relative cumulative incidences (indicated by the flat cumulative-incidence curves at the bottom of each graph in Panels A and B). The inset for each graph, which shows an expanded lower range of the infection probability scale, reveals patterns of cumulative risk across the participant groups. Panel A includes the two identified immune corre-lates of risk.

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.

Page 9: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

Immune-Correlates Analysis of an HIV-1 Vaccine Trial

n engl j med 366;14 nejm.org april 5, 2012 1283

mize statistical power in the primary analysis to detect correlates of infection risk.

Of the six assay variables chosen for the pri-mary analysis, two showed significant correla-

tions with infection among vaccine recipients: IgG antibody binding to scaffolded V1V2 Env correlated inversely with infection, and IgA anti-body binding to Env correlated directly with in-

Placebo

Vaccine, low response

Vaccine, medium response

Vaccine, high response

B

Prob

abili

ty o

f Acq

uiri

ng H

IV In

fect

ion 1.0

0.8

0.6

0.4

0.2

0.00 12 24 36

0.008

0.006

0.004

0.002

0.0000 12 24 36

Months since Wk 26 Visit

Avidity of IgG Antibodies for Env

No. at Risk (no. of infections)PlaceboLow responseMedium responseHigh response

6267 (0)2191 (0)2417 (0)2378 (0)

6199 (24)2187 (4)2410 (7)2372 (6)

6127 (11)2182 (5)2408 (2)2367 (5)

693 (13)179 (5) 92 (4)383 (3)

Prob

abili

ty o

f Acq

uiri

ng H

IV In

fect

ion

1.0

0.8

0.6

0.4

0.2

0.00 12 24 36

0.008

0.006

0.004

0.002

0.0000 12 24 36

Months since Wk 26 Visit

Neutralizing Antibodies

No. at Risk (no. of infections)PlaceboLow responseMedium responseHigh response

6267 (0)2299 (0)2330 (0)2358 (0)

6199 (24)2296 (3)2323 (7)2351 (7)

6127 (11)2292 (4)2320 (3)2346 (5)

693 (13)232 (3)180 (7)242 (2)

1.0

0.8

0.6

0.4

0.2

0.00 12 24 36

0.008

0.006

0.004

0.002

0.0000 12 24 36

Months since Wk 26 Visit

Antibody-Dependent Cellular Cytotoxicity

6267 (0)3362 (0)1719 (0)1906 (0)

6199 (24)3354 (8)1713 (6)1903 (3)

6127 (11)3348 (6)1711 (2)1899 (4)

693 (13)210 (8)202 (3)242 (1)

1.0

0.8

0.6

0.4

0.2

0.00 12 24 36

0.008

0.006

0.004

0.002

0.0000 12 24 36

Months since Wk 26 Visit

Env-Specific CD4+ T Cells

6267 (0)2385 (0)2167 (0)2435 (0)

6199 (24)2378 (7)2162 (5)2430 (5)

6127 (11)2372 (6)2160 (2)2426 (4)

693 (13)258 (4)132 (5)264 (3)

Figure 3. (Continued.)

Panel B includes the remaining four primary immune-response variables. For each primary variable, 41 vaccinated case patients were stratified into subgroups divided into thirds according to the immune response (low, medium, and high) at week 26 in the vaccine group in the case–control study. The estimated cumulative incidence of HIV-1 infection over time since the measurement of immune response at week 26 is shown for the three vaccine subgroups and for placebo recipients who were negative for HIV-1 infection at week 24.

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.

Page 10: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

n engl j med 366;14 nejm.org april 5, 20121284

fection. These two correlates of risk, taken to-gether, were highly correlated with the infection rate and may generate important hypotheses about immune responses required for protection from HIV-1,1-3 improve the selection of primary end points in subsequent HIV-1 vaccine trials,24 and lead to improved vaccines. If protection con-ferred by V1V2 IgG antibodies can be confirmed, then the design of vaccines to induce high lev-els of V1V2 antibodies and low levels of Env-specific IgA antibodies might augment vaccine efficacy.

Several lines of evidence suggest that vaccine-induced antibodies recognize conformational epitopes in the scaffolded V1V2 reagent, which has been shown to detect conformational V1V2 antibodies.25,26 The results of an analysis of breakthrough viruses from patients in the RV144 trial were consistent with immune pressure fo-cused on amino acid patterns in and flanking the V1V2 region of HIV-1 Env.27 This region serves critical functions, such as participating in CD4-receptor and chemokine-receptor binding, binding to α4β7 integrin,28 and serving as the binding site of neutralizing antibodies.29-32

In the Step HIV-1 vaccine trial (ClinicalTrials .gov number, NCT00095576), which was designed to induce HIV-1 T-cell responses, the hazard ra-tio for HIV-1 infection in the vaccine group as compared with the placebo group was higher in selected subgroups of vaccine recipients.33 Al-though the notion of antibody-mediated en-hancement of intrauterine infection has been raised in a clinical trial of treatment of HIV-1–infected pregnant women with infusion of im-mune globlulin,34 no vaccine-associated increase in the risk of infection was seen in the RV144 trial, and in analyses that compared infection rates in vaccine-recipient subgroups with the rate in the placebo group, no increase was seen with high levels of vaccine-induced Env-specific plasma IgA antibodies (Fig. S3 in the Supple-mentary Appendix). However, a limitation of the analyses that compared infection risk among vaccine and placebo recipients is that the com-parator groups could not be randomized, and there may have been residual confounding be-cause the analysis controlled only for sex and baseline behavioral risk factors.

The significant interactions of Env-specific IgA antibodies with other primary variables fur-ther support the importance of IgA-binding an-tibodies in predicting the risk of infection (Table

S4 in the Supplementary Appendix). In vaccinees with low levels of Env-specific IgA antibodies, four of the other five primary variables — IgG avidity, antibody-dependent cellular cytotoxicity, neutralizing antibodies, and Env-specific CD4+ T cells — were inversely correlated with infec-tion, whereas in vaccinees with high levels of Env-specific IgA antibodies, there was no corre-lation between these variables and infection (Table S4 in the Supplementary Appendix). The observed interactions generated the hypothesis that plasma IgA antibody levels interfere with protective IgG effector functions, a phenomenon that has been observed with other pathogens,35,36 in the regulation of autoantibody function,37 and in immune responses to cancer.38

We found that vaccinees with IgA antibodies to the first conserved region (C1) of gp120 had a higher risk of infection than vaccinees without these antibodies (odds ratio, 3.15; P = 0.003; q = 0.13). The gp120 C1 region contains an epi-tope that can be a target on the surface of virus-infected cells for antibodies that mediate anti-body-dependent cellular cytotoxicity.39 Another possible scenario is that high levels of Env-spe-cific IgA antibodies is a surrogate marker for HIV-1 exposure that was not fully accounted for by adjustment for baseline self-reported behav-ioral risk factors in the regression models. The primary variable of Env-specific IgA antibodies was not significantly associated with baseline behavioral risk factors (P = 0.28), nor did IgA antibodies to the individual Env proteins in-cluded in the primary IgA variable correlate with baseline behavioral risk factors (Table S6 in the Supplementary Appendix). Plasma IgA is primar-ily monomeric IgA, whereas mucosal IgA is pri-marily dimeric.40 Any protective role of mucosal dimeric IgA in the context of HIV-1 vaccination could not be evaluated in the RV144 trial, be-cause mucosal samples were not collected.

The relevance of these findings to different HIV-1 risk populations receiving ALVAC-HIV, AIDSVAX B/E, or other HIV-1 vaccine regimens cannot be inferred and must be prospectively determined. Moreover, further studies are re-quired to determine causality — whether V1V2 antibodies mediate vaccine-induced protection from infection or whether Env-specific IgA anti-bodies interfere with protection. Nonetheless, the identification of immune correlates of the risk of HIV-1 infection in the RV144 trial pro-vides plausible biologic hypotheses for the origi-

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.

Page 11: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

Immune-Correlates Analysis of an HIV-1 Vaccine Trial

n engl j med 366;14 nejm.org april 5, 2012 1285

nal clinical observation of vaccine efficacy.4 Elucidation of the potential roles of V1V2 and Env-specific IgA antibodies in the modulation of HIV-1 infection risk may accelerate the clinical development of vaccine candidates that can im-prove on the results of the RV144 clinical trial.

The views expressed in this article are those of the authors and should not be construed as official or as representing the views of the Department of Health and Human Services, the National Institute of Allergy and Infectious Diseases (NIAID), the Department of Defense, the Department of the Army, or the Department of Veterans Affairs.

Supported in part by grants from the Bill and Melinda Gates Foundation Collaboration for AIDS Vaccine Discovery (to Drs. Haynes, Koup, and Montefiori); an interagency agreement be-tween the U.S. Army Medical Research and Materiel Command and the NIAID (Y1-AI-2642-12); a cooperative agreement be-tween the Henry M. Jackson Foundation for the Advancement of Military Medicine and the Department of Defense (W81XWH- 07-2-0067); and grants from the NIAID to the Center for HIV/AIDS Vaccine Immunology (U01-AI-067854) and the HIV Vac-cine Trials Network Laboratory Program (UM1-AI-068618).

Disclosure forms provided by the authors are available with the full text of this article at NEJM.org.

We thank Judith T. Lucas, Vicki C. Ashley, R. Glenn Overman,

Claire Beard, Michele Donathan, Doris Murray, Kelly Seaton, Yong Lin, Robert Parks, Amanda Martelli, Corrine Dary, Wen-hong Feng, Xing Yuan, Kara John, Dora de Fonseca, and Robert O’Connell for project management or assay performance; Cris-tine Cooper-Trenbeath, Cheryl DeBoer, Cindy Molitor, Mark Bol-lenbeck, Linda Harris, and Craig Magaret for assay data process-ing; Daryl Morris, Alicia Sato, Ying Huang, Xuesong Yu, Raphael Gottardo, Barbara Metch, Nidhi Kochar, Hasan Ahmed, Zoe Moodie, and Maggie Wang for statistical analysis; Drienna Hol-man, Laura Saganic, Huguette Redinger, Kelli Greene, Hongmei Gao, Raul Louzao, Whitney Binz, Marcella Sarzotti-Kelsoe, and Caroline Cockrell for project management or project quality control or both; Corey Nathe, Shane Coultas, Scott Langley, and Sravani Cheeti for Atlas portal design; John Mascola for Env proteins; Dennis Burton and Ann Hessell for IgA 1b12 control antibody; Rebecca Putnam, Dave Friedrich, Don Carter, Olivier Defawe, Julie Williams, Jason Stuckey, Terry Stewart, Nathaniel Chartrand, Paul Newling, Jane Vasilyeva, Paula Walsh, Rebecca Smith, Jonathan Karademos, Carol Marty, David Chambliss, Leo French, and Bryce Manso for the management and conduct of the cellular assays; Ryan Wiley and Anne Mullin for discussions; Donald Francis, Carter Lee, Keith Higgins, and Faruk Sinangil for Env gp120s; James Tartaglia, Sanjay Gurunathan, and Sanjay Phogat for advice and discussions; the RV144 Humoral/Innate and Cellular RV144 Working Groups for helpful discussions and pilot analyses; and the RV144 Steering Committee for discus-sions and oversight (see the Supplementary Appendix for full lists of the Working Groups and Steering Committee).

appendixThe authors’ affiliations are as follows: the Duke University Human Vaccine Institute and the Center for HIV/AIDS Vaccine Immunology, Duke University School of Medicine, Durham, NC (B.F.H., G.D.T., S.M.A., D.C.M., H.-X.L., G.F., K.A.S., N.L.Y., X.S.); the Statistical Center for HIV/AIDS Research and Prevention (P.B.G., Y.F., H.J., A.D., Y.H.) and the Clinical Research Division, Vaccine and Infectious Disease Division (M.J.M., N.F., S.C.D.R.), Fred Hutchinson Cancer Research Center, Seattle; the Veterans Affairs New York Harbor Health-care System and the Department of Pathology, New York University School of Medicine — both in New York (S.Z.-P., C.W.); the Depart-ment of Microbiology and Immunobiology, Harvard Medical School, New England Regional Primate Center, Southborough, MA (D.T.E., M.D.A.); the U.S. Military Research Program, Walter Reed Army Institute of Research, Silver Spring (M.R., E.B., M.L.R., R.P., C.A., N.L.M., J.H.K.), and the Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda (R.T.B., R.A.K.) — both in Maryland; the U.S. Army Component (C.K., N.K., V.N., M.S.S.) and the Royal Thai Army (S.N.), Armed Forces Research Institute of Medical Sciences, Bangkok, the Department of Microbiology, National HIV Repository and Bioinfor-matic Center, Siriraj Hospital, Bangkok (R.S.), and the Faculty of Tropical Medicine, Mahidol University, Bangkok (P.P., J.K.), and the De-partment of Disease Control, Ministry of Public Health, Nonthaburi (S.R.-N.) — all in Thailand; the Department of Biomolecular Engi-neering, University of California, Santa Cruz, Santa Cruz (P.W.B.); the Institute of Human Virology, University of Maryland School of Medicine, Baltimore (A.L.D., G.K.L.); and the University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark (A.P.).

References

1. Plotkin SA. Vaccines: correlates of vaccine-induced immunity. Clin Infect Dis 2008;47:401-9.2. Idem. Correlates of protection induced by vaccination. Clin Vaccine Immunol 2010;17:1055-65.3. Qin L, Gilbert PB, Corey L, McElrath MJ, Self SG. A framework for assessing immunological correlates of protection in vaccine trials. J Infect Dis 2007;196: 1304-12.4. Rerks-Ngarm S, Pitisuttithum P, Nitaya-phan S, et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med 2009;361:2209-20.5. Walker BD. Elite control of HIV Infec-tion: implications for vaccines and treat-ment. Top HIV Med 2007;15:134-6.6. Mascola JR. Defining the protective antibody response for HIV-1. Curr Mol Med 2003;3:209-16.7. Hessell AJ, Rakasz EG, Tehrani DM, et al. Broadly neutralizing monoclonal

antibodies 2F5 and 4E10 directed against the human immunodeficiency virus type 1 gp41 membrane-proximal external region protect against mucosal challenge by simian-human immunodeficiency virus SHIVBa-L. J Virol 2010;84:1302-13.8. Barnett SW, Burke B, Sun Y, et al. Antibody-mediated protection against mu-cosal simian-human immunodeficiency virus challenge of macaques immunized with alphavirus replicon particles and boosted with trimeric envelope glycopro-tein in MF59 adjuvant. J Virol 2010;84: 5975-85.9. Barnett SW, Srivastava IK, Kan E, et al. Protection of macaques against vaginal SHIV challenge by systemic or mucosal and systemic vaccinations with HIV-enve-lope. AIDS 2008;22:339-48.10. Letvin NL, Mascola JR, Sun Y, et al. Preserved CD4+ central memory T cells and survival in vaccinated SIV-challenged monkeys. Science 2006;312:1530-3.

11. Letvin NL, Rao SS, Montefiori DC, et al. Immune and genetic correlates of vaccine protection against mucosal infection by SIV in monkeys. Sci Transl Med 2011; 3:81ra36.12. Hansen SG, Ford JC, Lewis MS, et al. Profound early control of highly patho-genic SIV by an effector memory T-cell vaccine. Nature 2011;473:523-7.13. Picker LJ, Hansen SG, Lifson JD. New paradigms for HIV/AIDS vaccine develop-ment. Annu Rev Med 2012;63:95-111.14. Cole KS, Rowles JL, Jagerski BA, et al. Evolution of envelope-specific antibody responses in monkeys experimentally in-fected or immunized with simian immu-nodeficiency virus and its association with the development of protective immunity. J Virol 1997;71:5069-79.15. Barouch DH, Liu J, Li H, et al. Vaccine protection against acquisition of neutral-ization-resistant SIV challenges in rhesus monkeys. Nature 2012;482:89-93.

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.

Page 12: The new england journal medicine - University of …faculty.washington.edu › peterg › papers › HaynesetalNEJM...The new engl and journal of medicine 1276 n engl j med 366;14

n engl j med 366;14 nejm.org april 5, 20121286

Immune-Correlates Analysis of an HIV-1 Vaccine Trial

16. Forthal DN, Gilbert PB, Landucci G, Phan T. Recombinant gp120 vaccine- induced antibodies inhibit clinical strains of HIV-1 in the presence of Fc receptor-bearing effector cells and correlate in-versely with HIV infection rate. J Immu-nol 2007;178:6596-603.17. Gilbert PB, Peterson ML, Follmann D, et al. Correlation between immunologic responses to a recombinant glycoprotein 120 vaccine and incidence of HIV-1 infec-tion in a phase 3 HIV-1 preventive vaccine trial. J Infect Dis 2005;191:666-77.18. Karnasuta C, Paris RM, Cox JH, et al. Antibody-dependent cell-mediated cyto-toxic responses in participants enrolled in a phase I/II ALVAC-HIV/AIDSVAX B/E prime-boost HIV-1 vaccine trial in Thai-land. Vaccine 2005;23:2522-9.19. Nitayaphan S, Pitisuttithum P, Karna-suta C, et al. Safety and immunogenicity of an HIV subtype B and E prime-boost vaccine combination in HIV-negative Thai adults. J Infect Dis 2004;190:702-6.20. Rolland M, Gilbert P. Evaluating im-mune correlates in HIV type 1 vaccine ef-ficacy trials: what RV144 may provide. AIDS Res Hum Retroviruses 2011 Sep-tember 27 (Epub ahead of print).21. Breslow NE, Holubkov R. Maximum likelihood estimation of logistic regres-sion parameters under two-phase, out-come-dependent sampling. J R Stat Soc B 1997;59:447-61.22. Borgan O, Langholz B, Samuelsen SO, Goldstein L, Pogoda J. Exposure stratified case-cohort designs. Lifetime Data Anal 2000;6:39-58.23. Rothman KJ. No adjustments are needed for multiple comparisons. Epide-miology 1990;1:43-6.24. Gilbert P, Hudgens MG, Wolfson J. Commentary on “Principal stratification — a goal or a tool?” by Judea Pearl. Int J Biostat 2011;7:36.

25. Pinter A, Honnen WJ, Kayman SC, Trochev O, Wu Z. Potent neutralization of primary HIV-1 isolates by antibodies di-rected against epitopes present in the V1/V2 domain of HIV-1 gp120. Vaccine 1998;16:1803-11.26. Zolla-Pazner S, Cardozo T, DeCamp A, et al. V2-Reactive antibodies in RV144 vaccinees’ plasma. AIDS Res Hum Retro-viruses 2011;27:A-21. abstract.27. Edlefsen P, Hertz T, Magaret C, De-Camp A, Gilbert P. Sieve analysis of RV144. Presented at AIDS Vaccine 2011, Bangkok, Thailand, September 12–15, 2011. abstract.28. Newaz F, Cicala C, Van Ryk D, et al. The genotype of early-transmitting HIV gp120s promotes α (4) β(7)-reactivity, re-vealing α (4) β(7) +/CD4+ T cells as key tar-gets in mucosal transmission. PLoS Pat-hog 2011;7(2):e1001301.29. Gorny MK, Stamatatos L, Volsky B, et al. Identification of a new quaternary neutralizing epitope on human immuno-deficiency virus type 1 virus particles. J Virol 2005;79:5232-7.30. Walker LM, Phogat SK, Chan-Hui PY, et al. Broad and potent neutralizing anti-bodies from an African donor reveal a new HIV-1 vaccine target. Science 2009; 326:285-9.31. Changela A, Wu X, Yang Y, et al. Crys-tal structure of human antibody 2909 re-veals conserved features of quaternary structure-specific antibodies that potent-ly neutralize HIV-1. J Virol 2011;85:2524-35.32. McLellan JS, Pancera M, Carrico C, et al. Structure of HIV-1 gp120 V1/V2 do-main with broadly neutralizing antibody PG9. Nature 2011;480:336-43.33. Buchbinder SP, Mehrotra DV, Duerr A, et al. Efficacy assessment of a cell-mediat-ed immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, pla-

cebo-controlled, test-of-concept trial. Lancet 2008;372:1881-93.34. Onyango-Makumbi C, Omer SB, Mu-biru M, et al. Safety and efficacy of HIV hyperimmune globulin for prevention of mother-to-child HIV transmission in HIV-1-infected pregnant women and their in-fants in Kampala, Uganda (HIVIGLOB/NVP STUDY). J Acquir Immune Defic Syndr 2011;58:399-407.35. Griffiss JM, Goroff DK. IgA blocks IgM and IgG-initiated immune lysis by separate molecular mechanisms. J Immu-nol 1983;130:2882-5.36. Jarvis GA, Griffiss JM. Human IgA1 blockade of IgG-initiated lysis of Neisse-ria meningitidis is a function of antigen-binding fragment binding to the polysac-charide capsule. J Immunol 1991;147: 1962-7.37. Quan CP, Watanabe S, Forestier F, Bouvet JP. Human amniotic IgA inhibits natural IgG autoantibodies of maternal or unrelated origin. Eur J Immunol 1998;28: 4001-9.38. Mathew GD, Qualtiere LF, Neel HB III, Pearson GR. IgA antibody, antibody-dependent cellular cytotoxicity and prog-nosis in patients with nasopharyngeal carcinoma. Int J Cancer 1981;27:175-80.39. Ferrari G, Pollara J, Kozink D, et al. An HIV-1 gp120 envelope human mono-clonal antibody that recognizes a C1 conformational epitope mediates potent antibody-dependent cellular cytotoxicity (ADCC) activity and defines a common ADCC epitope in human HIV-1 serum. J Virol 2011;85:7029-36.40. Mestecky J, Moro I, Kerr MA, Woof JM. Mucosal immunoglobulins. In: Mes-tecky J, Lamm ME, Strober W, Bienen-stock J, McGhee JR, Mayer L, eds. Mucosal immunology. 3rd ed. Burlington, MA: Elsevier Academic Press, 2005:153-82.Copyright © 2012 Massachusetts Medical Society.

nejm 200th anniversary articles

The NEJM 200th Anniversary celebration includes publication of a series of invited review and Perspective articles throughout 2012.

Each article explores a story of progress in medicine over the past 200 years. The collection of articles is available at the NEJM 200th Anniversary website,

http://NEJM200.NEJM.org.

The New England Journal of Medicine Downloaded from nejm.org at FRED HUTCHINSON CANCER RESEARCH on May 25, 2013. For personal use only. No other uses without permission.

Copyright © 2012 Massachusetts Medical Society. All rights reserved.