Recent progress in HIV vaccines inducing mucosal immune responses.

18
Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. EDITORIAL REVIEW Recent progress in HIV vaccines inducing mucosal immune responses Vincent Pavot a,b , Nicolas Rochereau b , Philip Lawrence c , Marc P. Girard d , Christian Genin b , Bernard Verrier a and Ste ´phane Paul b In spite of several attempts over many years at developing a HIV vaccine based on classical strategies, none has convincingly succeeded to date. As HIV is transmitted primarily by the mucosal route, particularly through sexual intercourse, understanding antiviral immunity at mucosal sites is of major importance. An ideal vaccine should elicit HIV-specific antibodies and mucosal CD8 þ cytotoxic T-lymphocyte (CTL) as a first line of defense at a very early stage of HIV infection, before the virus can disseminate into the secondary lymphoid organs in mucosal and systemic tissues. A primary focus of HIV preventive vaccine research is therefore the induction of protective immune responses in these crucial early stages of HIV infection. Numerous approaches are being studied in the field, including building upon the recent RV144 clinical trial. In this article, we will review current strategies and briefly discuss the use of adjuvants in designing HIV vaccines that induce mucosal immune responses. ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins AIDS 2014, 28:1701–1718 Keywords: adjuvants, administration routes, HIV, mucosa, vaccine Introduction Despite the extensive efforts that have been made over almost 30 years, major challenges still exist concerning HIV vaccine design. Most HIV infections by far occur through sexual contact [1]. Women are particularly vulnerable during heterosexual transmission through exposure to contaminated seminal fluids, and indeed, heterosexual women account for more than half of all individuals living with this virus [2]. Mucosal tissues involved in the sexual transmission of HIV include the cervicovaginal and rectal mucosa as well as the foreskin and oral epithelia [3]. Therefore, eliciting a strong preexisting anti-HIV immune response in mucosa- associated lymphoid tissues (MALTs) is probably of vital importance in preventing HIV infection [4]. The development of an effective vaccine is a considerable challenge, especially given the formidable propensity to immune evasion that is intrinsic to HIV. The HIV-1 envelope glycoprotein (Env) that is the target of known HIV-1-directed neutralizing antibodies (NAbs) [5–7] is protected by an evolving shield of glycans, variable immunodominant loops and conformational masking of key viral epitopes [6,8–10]. Although immunization with recombinant Env proteins or vectors encoding Env can a Institut de Biologie et Chimie des Prote ´ ines – LBTI, UMR 5305 – CNRS/University of Lyon 1, Lyon, France, b Groupe Immunite ´ des Muqueuses et Agents Pathoge `nes – INSERM CIE3 Vaccinologie, Faculte ´ de Me ´decine, Saint-Etienne, c International Centre for Research in Infectiology (CIRI), INSERM U1111 – CNRS UMR5308, University of Lyon 1, Lyon, and d French National Academy of Medicine, Paris, France. Correspondence to Dr Ste ´phane Paul, GIMAP - Faculte ´ de Me ´decine Jacques Lisfranc - 15 rue Ambroise Pare ´ - 42023 Saint- Etienne, France. Tel: +334 77 42 14 67; fax: +334 77 42 14 86; e-mail: [email protected] Received: 13 February 2014; revised: 12 April 2014; accepted: 14 April 2014. DOI:10.1097/QAD.0000000000000308 ISSN 0269-9370 Q 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins 1701

Transcript of Recent progress in HIV vaccines inducing mucosal immune responses.

EDITORI

AL REVIEW

Recent progress in HIV vaccines inducing mucosalimmune responses

Vincent Pavota,b, Nicolas Rochereaub, Philip Lawrencec,

Marc P. Girardd, Christian Geninb, Bernard Verriera

and Stephane Paulb

Copyright © L

aInstitut de BiologiMuqueuses et AgeResearch in InfectiMedicine, Paris, F

Correspondence tEtienne, France.

Tel: +334 77 42 1Received: 13 Febr

DOI:10.1097/QAD

ISSN

In spite of several attempts over many years at developing a HIV vaccine based onclassical strategies, none has convincingly succeeded to date. As HIV is transmittedprimarily by the mucosal route, particularly through sexual intercourse, understandingantiviral immunity at mucosal sites is of major importance. An ideal vaccine shouldelicit HIV-specific antibodies and mucosal CD8þ cytotoxic T-lymphocyte (CTL) as a firstline of defense at a very early stage of HIV infection, before the virus can disseminateinto the secondary lymphoid organs in mucosal and systemic tissues. A primary focus ofHIV preventive vaccine research is therefore the induction of protective immuneresponses in these crucial early stages of HIV infection. Numerous approaches arebeing studied in the field, including building upon the recent RV144 clinical trial. In thisarticle, we will review current strategies and briefly discuss the use of adjuvants indesigning HIV vaccines that induce mucosal immune responses.

� 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

AIDS 2014, 28:1701–1718

Keywords: adjuvants, administration routes, HIV, mucosa, vaccine

Introduction

Despite the extensive efforts that have been made overalmost 30 years, major challenges still exist concerningHIV vaccine design. Most HIV infections by far occurthrough sexual contact [1]. Women are particularlyvulnerable during heterosexual transmission throughexposure to contaminated seminal fluids, and indeed,heterosexual women account for more than half of allindividuals living with this virus [2]. Mucosal tissuesinvolved in the sexual transmission of HIV include thecervicovaginal and rectal mucosa as well as the foreskinand oral epithelia [3]. Therefore, eliciting a strong

ippincott Williams & Wilkins. Unaut

e et Chimie des Proteines – LBTI, UMR 5305 – CNnts Pathogenes – INSERM CIE3 Vaccinologie, Fology (CIRI), INSERM U1111 – CNRS UMR5308,rance.

o Dr Stephane Paul, GIMAP - Faculte de Medec

4 67; fax: +334 77 42 14 86; e-mail: stephane.uary 2014; revised: 12 April 2014; accepted: 1

.0000000000000308

0269-9370 Q 2014 Wolters Kluwer Hea

preexisting anti-HIV immune response in mucosa-associated lymphoid tissues (MALTs) is probably of vitalimportance in preventing HIV infection [4].

The development of an effective vaccine is a considerablechallenge, especially given the formidable propensity toimmune evasion that is intrinsic to HIV. The HIV-1envelope glycoprotein (Env) that is the target of knownHIV-1-directed neutralizing antibodies (NAbs) [5–7] isprotected by an evolving shield of glycans, variableimmunodominant loops and conformational masking ofkey viral epitopes [6,8–10]. Although immunization withrecombinant Env proteins or vectors encoding Env can

horized reproduction of this article is prohibited.

RS/University of Lyon 1, Lyon, France, bGroupe Immunite desaculte de Medecine, Saint-Etienne, cInternational Centre forUniversity of Lyon 1, Lyon, and dFrench National Academy of

ine Jacques Lisfranc - 15 rue Ambroise Pare - 42023 Saint-

[email protected] April 2014.

lth | Lippincott Williams & Wilkins 1701

Co

1702 AIDS 2014, Vol 28 No 12

induce high levels of HIV-1 specific Abs, vaccine-inducedAbs have been unable to neutralize most circulatingprimary HIV-1 isolates [6,7,11,12]. Indeed, naturalinfection predominantly induces nonneutralizing orstrain-specific Abs during the first months of infection[9,13–15]. However, NAbs are the best correlate ofprotection for many viral vaccines [16,17]. It was foundthat approximately 10–20% of HIV-1-infected individualshappen to develop broadly NAbs (bNAbs) after a few years[18–20], which is the type of humoral immune responseone would like a vaccine to elicit. These bNAbs are able toneutralize the vast majority of virus strains in a cross-clademanner and have been shown to provide robust protectionagainst mucosal challenges in the macaque model [21–23].

Our understanding of what constitutes a bNAb againstHIV has been revolutionized by the isolation of extremelybroad and potent neutralizing mAb from a number ofHIV-infected individuals [24–27]. These mAbs wereidentified by dissecting the broad neutralization activityseen in specific patient serum samples and by character-izing mAbs from B-cells [24,28,29].

Some bNAbs, when acting at the earliest steps of viralinfection, are able to prevent virus entry into host cells byblocking multiple steps of viral transmission by targetingeither the CD4þ-binding site or the glycan/V3 loop onHIV-1 gp120 [30]. The incapacity to induce bNAbs toHIV has thus been a major hurdle to HIV vaccineresearch since the beginning of the epidemic. Substantialobstacles remain in inducing bNAbs by immunizationand particularly at the mucosal level. Some studies haveshown evidence of the presence of NAbs in mucosal fluidsusing appropriate immunization vectors and deliveryregimens [31–34], but previous attempts to inducebNAbs by vaccination were fairly unsuccessful. Currently,based upon our existing understanding of the mucosalimmune system, we would expect that the quality ofhumoral and cellular immune responses at a given effectorsite would depend upon the route of vaccination [35]. It isthus essential to choose the suitable immunization routefor the desired mucosal site.

Despite the remarkable progress made in understandingthe epitopes that Abs recognize on the Env spikes of thevirion, one of the major goals of HIV vaccine research atthis time is the discovery of immunogens and immuniz-ation strategies that can elicit bNAbs. This challenge ismade even greater by the fact that it is more difficult toinduce high concentrations of NAbs at the mucosal levelthan at a systemic level [36].

It would certainly be beneficial for an HIV-1 vaccine toalso elicit immune responses capable of controlling viralreplication [37]. A wealth of data has shown that cellularimmune responses can mediate the control of viremia inHIV-1-infected humans and simian immunodeficiencyvirus (SIV)-infected rhesus macaques, including CD8þ T

pyright © Lippincott Williams & Wilkins. Unautho

lymphocytes [38–40], natural killer (NK) cells [41] andCD4þ T lymphocytes [42,43]. Moreover, vaccine trials innonhuman primates (NHPs) have shown that sustainedviremic control is achievable after heterologous SIVchallenges. For example, immunizations with an Adeno-virus serotype 26 prime and Modified Vaccinia Ankara(MVA) boost expressing SIV antigens led to a 2.32 logreduction in mean set point viral load following stringentSIVmac251 challenge, which was related to the magnitudeand breadth of the Gag-specific cellular immune responsesmeasured immediately prior to challenge [31].

Even more remarkable was the report that 50% of rhesusmacaques vaccinated with a SIV protein expressingrhesus cytomegalovirus (RhCMV/SIV) vector manifesteddurable, aviraemic control of infection with the highlypathogenic strain SIVmac239 [38]. The RhCMV/SIVvector elicited immune responses that control SIVmac239infection (regardless of the route of challenge) after viraldissemination. Over time, protected rhesus macaques lostsigns of SIV infection, showing a consistent lack ofmeasurable plasma or tissue-associated viral RNA or DNAusing ultrasensitive assays, and a loss of T-cell reactivity toSIV determinants not in the vaccine [44]. Similarly, it wasshown that protection against wild-type SIVmac239challenge by live attenuated SIV vaccines strongly corre-lated with the magnitude and function of SIV-specific,effector-differentiated T cells in the lymph nodes of theanimals. It follows from these observations that SIV-specific T cells can suppress wild-type SIVamplification atan early stage and, if present persistently in sufficient fre-quencies, can completely control and even clear infection[45].

Current assessments aim to evaluate a broad range ofmucosal immune responses and answer key questionssuch as can vaccines delivered parenterally elicit detectablemucosal responses? Whereas systemic immunizationinduces mostly immune responses in peripheral andsystemic sites, mucosal delivery of immunogens isthought to trigger primarily mucosal immune responses[46]. The second question is which mucosal immuneresponses may be associated with protection from HIVinfection? And the third, which mucosal specimens andassays are most relevant for the detection of theseresponses? Answers to these questions will be vital inclarifying which mucosal immune responses are capableof blocking HIV infection, and for developing vaccinesthat can elicit these types of responses.

Mucosal transmission of HIV-1: a rationalefor the role of HIV-specific mucosalimmune responses

Natural transmission of HIV-1 occurs through vaginalmucosa, the male genital tract, that is penile mucosa

rized reproduction of this article is prohibited.

Progress in HIV vaccines inducing mucosal responses Pavot et al. 1703

(inner foreskin, penile urethra), gastrointestinal mucosaand via breastmilk (vertical transmission). Although thereare challenges in quantifying risk by sex act, all studiesconsistently report that anal intercourse is a higher risk actthan vaginal intercourse and the probability of infectionby the vaginal route has been estimated to be one in 200or less [3]. Considering this route, HIV-1 can infect thevaginal, ectocervical and endocervical mucosa, but therelative contribution of each site to the establishment ofthe initial infection is unknown.

Both free and cell-associated HIV and SIV virions canestablish mucosal infection [1,47]. This has been showndirectly in vivo in female macaques [48–50], or ex vivousing human cervical explants [49,51], and indirectly inhumans through genetic sequence comparisons of viralisolates from acutely infected women with those fromseminal leukocytes (cell-associated virions) and plasmafrom their infected source partners [52–54]. Ex-vivostudies using human cervical explants and reconstructedvaginal mucosa have confirmed transmission of cell-freeand cell-associated HIV-1 [55–58]. Cervical mucus cantrap infected seminal cells or free virions [59,60].Conceivably, this could facilitate viral transmission byprolonging the time of contact of the virions with themucosa. However, although immobilized, the virionsmay also become more susceptible to innate antiviralsubstances or to Abs.

Several reports have shown that HIV virions bind to andenter epithelial cells in the female genital tract [61–63].Virions that are initially free, or those that are releasedfrom infected donor seminal T cells, interact withepithelial cells and traverse the epithelium by severalpathways, including transcytosis, endocytosis and sub-sequent exocytosis, by causing productive infection, ormerely by penetrating through the gaps betweenepithelial cells in the vaginal multilayered epithelium[64].

Transcytosis, which occurs across single layeredepithelia, has been shown to occur in cell lines andalso in primary cells, but has not been definitivelydemonstrated in intact mucosal tissues. Interestingly,cell-associated virions secreted from infected seminalleukocytes appear markedly more efficient at transcy-tosis than cell-free virions [65,66]. It is actually likelythat HIV is not transmitted as a naked particle, butrather as an immune complex with Env-binding IgGsthat are abundant in the semen of HIV-positive men.The low pH of the vagina and urethra also plays animportant role in transcytosis, as the neonatal FcRnreceptor that is expressed on epithelial cells of thepenile urethra and endocervix binds the IgGs at low pHand releases them at neutral pH, thus favouring thecapture of the immune virus complexes on the acidicapical (vaginal) side of the epithelium and their releasein the neutral basolateral environment [67].

Copyright © Lippincott Williams & Wilkins. Unaut

Upon release from epithelial cells, the virions can readilyinfect susceptible leukocytes [63]. It has been reportedthat virions can also productively infect the cervicalepithelial cells themselves [63,68], although this pointremains contested [69,70]. Conceivably, HIV-1 can alsobe transported through the cervicovaginal epithelium tothe draining lymph nodes by infected lymphocytes,macrophages, monocytes and dendritic cells, as has beensuggested in both in-vitro systems and mouse studies[68,71–75]. The rationale for the role of specific mucosalimmune responses in protection from HIV-1 transmissionhas been highlighted by numerous studies in human andNHPs (Table 1) [76–89].

Anatomic sites of HIV-1 persistence

Another rationale for eliciting HIV-specific mucosalimmune responses is linked to their potential to preventthe establishment of viral reservoirs within a newlyinfected host. A viral reservoir can be defined as cell typesor anatomical sites in which replication-competent formsof the virus can and do persist throughout infection and inthe presence of otherwise efficient antiretroviral therapy(ART) [90]. Gastrointestinal and vaginal mucosal tissuesare major reservoirs for initial HIV replication andamplification, and the sites of rapid CD4þ T-celldepletion [91]. Such viral reservoirs are currently thoughtto be a key factor explaining our difficulty in successfullyeradicating HIV-1 from an infected host via the currentlyavailable treatments regimes. A successful vaccine wouldneed to prevent the establishment of these reservoirs at avery early step of viral infection. This is especiallyimportant considering the fact that aggressive ART invery early acute infection can substantially decrease thesize of the viral reservoir in terms of integrated DNA andRNA concentrations [92]. Although most HIV pro-viralDNA is found in CD4þ T lymphocytes in lymphoidtissue, blood viral reservoirs may also be maintained incentral and transitional memory T cells that persistthrough mechanisms of homeostatic proliferation andrenewal. Other potential sources may include monocytesand macrophages, astrocytes and microglial cells [92].

A related aspect is also the notion of viral compartmentswithin an infected individual. A viral compartment maybe defined as a cell or tissue replication site wherein apopulation of viral variants is at least partially restricted inits ability to enter, leave and replicate and therefore displaya limited exchange of viral genetic information withother sites [90]. It is unclear as to whether all sites of viralcompartmentalization represent viral reservoirs in thestrictest sense. This anatomical compartmentalization ofHIV-1 variants has been well described for the centralnervous system (CNS), the gut-associated lymphoidtissue (GALT) [93,94] and the genital tract, althoughthere are also data for viral compartmentalization within

horized reproduction of this article is prohibited.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

1704 AIDS 2014, Vol 28 No 12

Tab

le1.

Exam

ple

sof

studie

sdem

onst

rati

ng

corr

elat

esof

muco

sal

imm

une

pro

tect

ion

inH

IV-1

infe

ctio

n.

Model

Study

des

ign

Outc

om

eR

efer

ence

s

Hum

anH

IV-1

spec

ific

muco

sal

IgA

ina

cohort

of

HIV

-1re

sist

ant

Ken

yan

fem

ale

sex

work

ers.

HIV

-1-s

pec

ific

IgA

ispre

senti

nth

ege

nit

altr

acto

fmost

HIV

-1re

sist

antK

enya

nse

xw

ork

ers.

[76,7

7]

Hum

anA

bil

ity

aten

rolm

ent

of

genit

alIg

Ato

neu

tral

ize

pri

mar

yH

IVis

ola

tes

asw

ell

assy

stem

icH

IV-s

pec

ific

cell

ula

rre

sponse

s.G

enit

alH

IV-n

eutr

aliz

ing

IgA

and

syst

emic

HIV

-spec

ific

pro

life

rati

vere

sponse

sw

ere

pro

spec

tive

lyas

soci

ated

wit

hH

IVnonac

quis

itio

n.

[78]

Hum

anH

IV-1

sero

dis

cord

antc

ouple

sin

Nai

robiw

ere

foll

ow

edquar

terl

yup

to2

year

s.C

ervi

cova

ginal

IgA

was

asse

ssed

forH

IV-1

neu

tral

izin

gac

tivi

tyby

aper

ipher

alblo

od

mononucl

earce

llbas

edas

say

usi

ng

anH

IV-1

clad

eA

pri

mar

yis

ola

te.

HES

Nw

om

enw

ere

five

tim

esm

ore

like

lyto

hav

eneu

tral

izin

gIg

Ain

cerv

icova

ginal

secr

etio

ns

than

low

-ris

kco

ntr

ol

wom

en.

Res

ponse

sw

ere

inve

rsel

yas

soci

ated

wit

hpar

tner

vira

llo

ad.

Thes

eobse

rvat

ions

support

the

exis

tence

ofan

tivi

ralac

tivi

tyin

the

muco

sal

IgA

frac

tion

foll

ow

ing

sexu

alH

IV-1

exposu

re.

[79]

Hum

anSu

b-p

reputial

swab

san

dfo

resk

inti

ssue

from

HES

Nan

dunex

pose

dco

ntr

ol

men

wer

eco

llec

ted.

Adec

reas

ein

the

rela

tive

abundan

ceof

susc

epti

ble

CD

Tce

lls,

inco

mbin

atio

nw

ith

HIV

-neu

tral

izin

gIg

Aan

da

-def

ensi

ns

may

repre

sent

apro

tect

ive

imm

une

mil

ieu

ata

site

of

HIV

exposu

re.

[80]

Inve

stig

ators

assa

yed

swab

sfo

rH

IV-n

eutr

aliz

ing

IgA

.H

um

anG

ener

atio

nof

aC

D4þ

-induce

dhum

anm

Ab,

F425A

1g8

.The

IgA

1va

rian

tdis

pla

yed

sign

ifica

nt

indep

enden

tneu

tral

izat

ion

acti

vity

agai

nst

ara

nge

of

HIV

clad

eB

isola

tes.

[81]

Char

acte

riza

tion

of

the

impac

tof

its

isoty

pe

vari

ants

on

HIV

neu

tral

izin

gac

tivi

ty.

Hum

anA

bil

ity

of

IgA

isola

ted

from

HEP

Sin

div

idual

sto

inhib

ittr

ansc

ytosi

sac

ross

ati

ght

epit

hel

ial

cell

laye

rin

vitr

o.

Inhib

itio

nw

asse

enin

thre

eof

six

cerv

icova

ginal

fluid

sam

ple

s,5/1

0sa

liva

sam

ple

san

dth

ree

of

six

pla

sma

sam

ple

sag

ainst

pri

mar

yH

IV-1

isola

tes

test

ed.

[82]

Hum

anM

uco

sal

Fab

IgA

libra

ryfr

om

HEP

San

din

-vit

roch

arac

teri

zati

on

of

seri

esof

HIV

-1Ig

As

spec

ific

for

gp41.

IgA

are

tran

scyt

osi

s-blo

ckin

gan

din

fect

ion-n

eutr

aliz

ing.

[83]

Hum

anN

inet

een

indiv

idual

sat

risk

for

HIV

infe

ctio

nw

ere

scre

ened

for

viru

s-sp

ecifi

cm

emory

CTL.

HIV

-spec

ific

CTL

wer

edet

ecte

din

seve

nof1

7ex

pose

dunin

fect

edin

div

idual

s.[8

4]

Rec

ogn

itio

nof

viru

sby

CTL

inex

pose

dunin

fect

edin

div

idual

sw

asM

HC

-Ire

stri

cted

.H

um

an37

HIV

-1-u

nin

fect

edper

sons

wit

hre

pea

ted

hig

h-r

isk

sexu

alac

tivi

tyw

ith

anH

IV-1

-infe

cted

par

tner

wer

epro

spec

tive

lyst

udie

dto

det

erm

ine

pro

tect

ing

fact

ors

agai

nst

HIV

-1tr

ansm

issi

on.

Thir

teen

of

36

dem

onst

rate

dH

IV-1

spec

ific

cyto

toxi

city

.[8

5]

Hum

anA

sses

smen

tof

HIV

-spec

ific

epit

ope

reco

gnitio

n,

pla

sma

vira

llo

adan

dex

pre

ssio

nofH

LAcl

ass

Iall

eles

ina

cohort

ofH

IV-s

eroposi

tive

bar

work

ers.

CD

8T-c

ell

reco

gnit

ion

of

mult

iple

epit

opes

wit

hin

spec

ific

Gag

regi

ons

isas

soci

ated

wit

hm

ainte

nan

ceof

alo

wst

eady-

stat

evi

rem

iain

HIV

-1se

roposi

tive

pat

ients

.

[86]

Hum

anR

ecen

tw

ork

has

focu

sed

grea

ter

atte

nti

on

on

qual

itat

ive

feat

ure

sof

the

HIV

-spec

ific

CD

T-c

ell

resp

onse

.In

vest

igat

ion

from

mult

iple

groups

has

now

focu

sed

upon

HIV

-spec

ific

CD

T-c

ell

gran

ule

-exo

cyto

sis

med

iate

dcy

toto

xici

tyas

aco

rrel

ate

of

imm

unolo

gic

contr

ol

of

HIV

.

[39]

Hum

anD

etai

led

study

of

the

inte

rpla

ybet

wee

nT-c

ell

funct

ional

attr

ibute

susi

ng

aban

kof

HIV

-spec

ific

CD

T-c

ell

clones

.H

ighly

sensi

tive

CD

Tce

lls

dis

pla

ypoly

funct

ional

pro

file

san

dpote

nt

HIV

-suppre

ssiv

eac

tivi

ty.

[87]

NH

PdIg

A1,dIg

A2

and

IgG

1ve

rsio

ns

ofnm

Ab

HG

N194

wer

eap

plied

intr

arec

tall

yin

thre

erh

esus

mac

aque

groups

bef

ore

intr

arec

tal

SHIV

chal

lenge

.dIg

A1-m

edia

ted

captu

ring

of

viri

ons

inm

uco

sal

secr

etio

ns

and

inhib

itio

noftr

ansc

ytosi

sca

npro

vide

sign

ifica

ntpre

venti

on

ofle

nti

vira

lac

quis

itio

n.

[88]

Inhib

itio

nof

tran

scyt

osi

sof

cell

-fre

evi

rus

acro

ssan

epit

hel

ial

cell

laye

rin

vitr

o.

NH

PThey

asse

ssed

whet

her

hum

anM

Absb12

and

b6

agai

nst

the

CD

-bin

din

gsi

teon

HIV

-1gp

120

and

F240

agai

nst

anim

mundom

inan

tep

itope

on

gp41

could

pre

vent

vagi

nal

tran

smis

sion

of

SHIV

.

Appli

edva

ginal

ly,

the

stro

ngl

yneu

tral

izin

gM

Ab

b12

pro

vided

ster

iliz

ing

imm

unit

yin

seve

nof

seve

nan

imal

s.[8

9]

Progress in HIV vaccines inducing mucosal responses Pavot et al. 1705

the lung, liver, kidney and breast milk (for a recent reviewsee [95]).

Env-specific antibodies to protect againstHIV-1 acquisition at mucosal surfaces

The design of immunogens able to elicit NAb remains amajor goal of HIV-1 vaccine development [96]. Manystudies in NHPs have shown that passive infusion of HIVNAbs, especially bNAbs, can prevent rectal or vaginalinfection by a chimeric simian-HIV (SHIV) containingthe env gene of HIV-1. This was initially shown using asingle oral or vaginal inoculation sufficient to infect 100%of control animals [97–100]. In this setting, protectionagainst SHIV infection was most directly associated withthe neutralization potency of the infused Abs [101,102].However, recent passive transfer studies have employedlow-dose multiple mucosal challenges to infect all controlanimals [103,104]. This model may be more physiologi-cally relevant to the relatively low probability of sexualinfection seen with HIV-1 in humans. In the low-doseNHP model, approximately 10-fold fewer Abs wererequired to mediate protection against infection thanprior studies with high-dose virus challenge: serum Abtitres sufficient to mediate 90% virus neutralization at 1 : 5serum dilution were associated with protection.

It has been suggested that Fc-mediated Ab effectorfunctions might also play an important role in conferringprotection. Indeed, although direct antibody-mediatedneutralization is highly effective against cell-free virus,increasing evidence suggests an important role for IgGFcg receptor (FcgR)-mediated inhibition of HIVreplication. Thus, bNAb IgG1 b12 showed a diminishedprotective potency after its Fc region was altered to knockout complement binding and antibody-dependent cell-mediated cytotoxicity (ADCC) activity without decreas-ing its in-vitro neutralizing activity [105]. A recent studyscreened a panel of bNAbs and nonneutralizing Abs(NoNAbs) for their ability to block HIV acquisitionand replication in vitro in either an independent orFcgR-dependent manner. In the NHP model, vaginalapplication of a gel containing the selected bNAbs2G12, 2F5 and 4E10 prevented SHIV transmission in 10out of 15 macaques after vaginal challenge, whereas theNoNAbs 246-D and 4B3 had no impact on SHIVacquisition but reduced plasma viral load [22]. Theseresults highlight that distinct neutralization and inhibitoryactivity of anti-HIV Abs affect in-vivo HIV acquisitionand replication in different ways and demonstrate thepotential interest of NAbs for microbicide and vaccinedevelopment. It follows that vaccines may not need toachieve extraordinarily high levels of HIV-1 NAbs toelicit protection at mucosal surfaces, but the Ab responsewill likely need to be durable, and NAbs will have tocross-react with a genetically diverse spectrum of HIV-1strains.

Copyright © Lippincott Williams & Wilkins. Unaut

We also do not know which type of immunoglobulins arethe best at blocking the virus at mucosal surfaces. IgG1Abs certainly can play a role, as passive infusion of suchanti-HIV bNAbs into the blood can protect animals frommucosal challenge [106,107]. However, it is known thatIgA is the predominant Ab in the majority of mucosalsecretions [108]. Mucosal IgA Ab is generated primarilyin the mucosal epithelial compartment and transportedacross the epithelial cell boundary into external secretionsby interacting with the polymeric immune globulinreceptor (pIgR) [109]. It was recently reported thatrectally applied dimeric IgA Abs derived from bNAbHGN194 could not only protect NHPs from rectalchallenge with SHIV-1157ipEL-p, but also that they didit more effectively than corresponding IgGs [88].Comparison of the IgG1 version of bNAb HGN194with its dimeric IgA versions, dIgA1 and dIgA2, showedthat the dIgA1 version protected the animals better thanthe dIgA2 and IgG1. Thus, five out of the six animalstreated with the dIgA1 version remained uninfected,whereas only one of the six dIgA2-treated animals andtwo of the six IgG1-treated animals remained virus free.All 11 untreated animals got infected.

The increased protection observed in the dIgA1-treatedanimals was initially puzzling, as all three HGN194versions neutralized the challenge virus equally well. Theexplanation could be that the dIgA1 version can bindtwice as many virus particles as the dIgA2 version. As aresult, a dIgA1 molecule can accommodate four virusparticles between its antigen-binding sites, while dIgA2can only accommodate two. This also might explain whyonly dIgA1 and not dIgA2 nor IgG1 were able to preventmost HIV particles from crossing a cultured epithelial celllayer in an in-vitro transcytosis assay [88].

Altogether, these results suggest that one should try todevelop vaccines that can elicit dIgA1s at mucosalsurfaces. These Abs do not necessarily have to beneutralizing, because it is the ability of dIgA1s to be ableto bind more HIV particles, and not necessarily bettervirus neutralization, which seems to be responsible fortheir higher level of protective efficacy.

Studies in humans have also revealed a correlation betweena high level of secretory IgA (SIgA) and protection in high-risk individuals who remain seronegative (highly exposedpersistently seronegative persons) [76,78,110,111]. Thesestudies concluded that protection could be mediated by theinteraction between these SIgA and HIV-1 on mucosalsurfaces or within epithelial cells capable of internalizingIgA-boundHIV-1. This conclusion is howeverdisputed, asthere is no evidence for IgA-mediated intraepithelial HIV-1 neutralization [112–114].

However, in addition to its ability to neutralize virus, it isthought that IgA may contribute to the elimination ofvirus in the form of exocrine immune complexes via the

horized reproduction of this article is prohibited.

Co

1706 AIDS 2014, Vol 28 No 12

lamina propria. In this manner, the mechanism of IgAprotection may be wider than that provided by IgG-mediated neutralization. It also follows that assays basedon neutralizing IgG Abs may not be suitable for assessingthe activity of mucosal IgA [115].

Vaccination strategies that elicit mucosalneutralizing antibodies

In clinical trials that show the efficacy of a vaccine, theidentification of immune responses that are predictive oftrial outcomes generates hypotheses about which of thoseresponses are responsible for protection [116,117]. TheRV144 phase 3 trial in Thailand was an opportunity toperform such a hypothesis-generating analysis for anHIV-1 vaccine. This trial of the canarypox vector vaccine(ALVAC-HIV [vCP1521]) as well as the gp120 AIDS-VAX B/E vaccine showed an estimated vaccine efficacyof 31.2% for the prevention of HIV-1 infection over aperiod of 42 months after the first of four plannedvaccinations [118]. This result enabled a systematic searchby Haynes et al. [119] who performed a case–controlanalysis to identify Ab and cellular immune correlates ofinfection risk. This immune-correlates study generatedthe hypotheses that levels of V1V2 Abs correlatedinversely with the risk of infection, whereas high levels ofEnv-specific IgA may have mitigated the effects ofprotective Abs. However, any protective role of mucosalAbs in the context of HIV-1 vaccination could not beevaluated in the RV144 trial, because mucosal sampleswere not collected.

Several studies have shown that immunization by the nasalroute (i.n.) can be most effective at eliciting Abs andcellular immunity in the female genital tract [35]. Thus,the use of live replicating recombinant Ad5hr-vectoredvaccine, in the rhesus macaques model, administered firstby i.n. and oral routes then intratracheally followed byEnv protein boosts resulted in systemic and mucosal Abresponses, including NAb, ADCC and transcytosisinhibition, together with potent cell immune responses[120]. Mucosal IgA immunity correlated with delayedacquisition following a repeated low-dose rectal SIV(mac251) challenge. The replicating Ad5 vector wasshown to disseminate across multiple mucosal sitesirrespective of delivery route [121]. These results suggestthat initial mucosal vaccination with a replicating vectorinducing NAbs in combination with a potent proteinboost may significantly reinforce protective immunityagainst SIV mucosal transmission.

As another example, four of five rhesus macaquesvaccinated first by intramuscular route (i.m.) and theni.n. with gp41-subunit antigens presented on virosomeswere protected against 13 consecutive vaginal challengeswith SHIV-SF162P3, and the fifth specimen showed only

pyright © Lippincott Williams & Wilkins. Unautho

transient infection. All of the animals displayed gp41-specific vaginal IgAs with HIV-1 transcytosis-blockingproperties and vaginal IgGs with neutralizing and/orADCC activities [32].

The immunogenicity of virosomes spiked with a gp41MPER peptide (P1) was tested in a phase I, double-blind,randomized, placebo-controlled trial in 24 healthy HIV-uninfected young women [122]. Antigen-specific serumIgGs and IgAs were elicited in all high-dose recipientsafter the first i.m. injection, but vaginal and rectal gp41-specific IgGs could be detected only after boosting via thei.n. route.

Although these data speak highly in favour of the nasalroute of immunization to elicit mucosal anti-HIV Abs inthe female genital tract, numerous studies have also shownthat parenteral immunization is able to induce protectivemucosal immune responses, notably with viral vectors(Table 2) [123–129]. Indeed, studies have demonstratedthe capacity of adenovirus/poxvirus and adenovirus/adenovirus vector based vaccines expressing HIV-1mosaic Env, Gag and Pol administered by i.m. route toprotect rhesus macaques against acquisition of infectionfollowing repetitive intrarectal inoculations of thedifficult-to-neutralize SHIV-SF162P3 or SIVmac251[31,123].

Cellular immune responses mediatecontrol of viremia

Whereas Env-specific Abs appear necessary to blockHIV-1 acquisition, Gag-specific cellular immuneresponses appear important for the control of virusreplication and viral load after infection. Gag-specificCD8þ T cells, but not Env- nor Pol-specific CD8þ Tcells, correlate with in-vivo viral load control followingSIV challenge in vaccinated monkeys [130]. This result isconsistent with studies demonstrating the association ofGag-specific cellular immune responses with viremiacontrol in HIV-1 infected individuals [131–133] andSIV-infected rhesus macaques [31,134–136]. Vif and Nefmay also contribute to viral load control in monkeys[137].

As conservation of polyfunctional HIV-specific CD8þ

T-cells appears to correlate with the control of viremia ininfected people [138], the polyfunctionality of the T-cellresponse is perceived as one of the best correlates of T-cellimmunity [87]. Thus, Ferre et al. [139] showed thatmucosal CD8þ cytotoxic T-lymphocyte (CTL) responsesin controllers are more complex and significantly strongerthan in antiretroviral-suppressed persons: HIV-controllersshow long-lasting, high avidity, polyfunctional Gag-specific CD8þ T-cell responses in mucosal compartmentsas compared with noncontrollers.

rized reproduction of this article is prohibited.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Progress in HIV vaccines inducing mucosal responses Pavot et al. 1707

Tab

le2.

Exam

ple

sof

vacc

ines

elic

itin

gpro

tect

ive

resp

onse

sag

ainst

muco

sal

chal

lenge

sin

the

NH

Pm

odel

.

Del

iver

yst

rate

gies

Route

of

imm

uniz

atio

nM

uco

sal

resp

onse

Ref

eren

ces

Ad26/A

d35/M

VA

vect

or-

bas

edva

ccin

esex

pre

ssin

gH

IV-1

mosa

icEn

v,G

ag,

and

Pol

Intr

amusc

ula

rSu

bst

anti

alpar

tial

pro

tect

ion

(�45%

)ag

ainst

repet

itiv

e,in

trar

ecta

l,het

erolo

gous

SHIV

-SF1

62P3

chal

lenge

s.[1

23]

Env-

spec

ific

bin

din

gA

bs

corr

elat

edm

ost

stro

ngl

yw

ith

pro

tect

ion.

Ad26/M

VA

or

Ad35/A

d26

vect

or-

bas

edva

ccin

esex

pre

ssin

gSI

Vsm

E543

Gag

,Pol

and

Env

anti

gens

Intr

amusc

ula

rPro

tect

ion

agai

nst

acquis

itio

nof

SIV

(mac

251)

isco

rrel

ated

wit

hEn

v-sp

ecifi

cA

bre

sponse

s.[3

1]

Vir

olo

gic

contr

ol

may

be

corr

elat

edw

ith

both

Tly

mphocy

tean

dA

bre

sponse

s(r

ecta

lse

cret

ions

and

colo

rect

albio

psi

es).

rAd26

enco

din

gSI

Vm

ac239

Gag

Intr

amusc

ula

rD

ura

ble

Gag

-spec

ific

cell

ula

rim

mune

resp

onse

sin

mult

iple

muco

sali

mm

une

com

par

tmen

ts,

incl

udin

gin

the

ora

lan

dga

stro

inte

stin

alm

uco

sa.

[128]

Pla

smid

DN

A(p

rim

e)rA

d5

(boost

)In

tram

usc

ula

rLo

wle

vels

ofN

Abs

and

anEn

v-sp

ecifi

cC

D4þ

T-c

ellr

esponse

wer

eas

soci

ated

with

vacc

ine

pro

tect

ion

agai

nst

muco

sali

nfe

ctio

nby

SIV

smE6

60

(�50%

).[1

25]

Nonre

pli

cati

ng

rAd

vect

ors

from

vari

ous

sero

types

expre

ssin

gSI

Vm

ac239

Gag

Intr

amusc

ula

rSI

V-s

pec

ific

Tly

mphocy

tere

sponse

sth

atper

sist

edfo

rove

r2

year

sin

both

per

ipher

alblo

od

and

multip

lem

uco

sal

tiss

ues

(colo

rect

al,

duoden

alan

dva

ginal

bio

psy

spec

imen

s)an

dbro

nch

oal

veola

rla

vage

fluid

.

[126]

GM

-CSF

DN

Aw

ith

DN

Apri

me

for

aSH

IV-8

9.6

vacc

ine

Intr

amusc

ula

ror

intr

ader

mal

Avi

dity

mat

ura

tion

of

anti

-Env

IgG

inblo

od.

[34]

Pre

sence

of

long-

last

ing

anti

vira

lIg

Ain

rect

alse

cret

ions.

DN

Aw

ith

afu

llSI

Vm

ac239

genom

e/rM

VA

expre

ssin

gSI

VG

ag-P

ol

and

Env

pro

tein

sIn

tram

usc

ula

ror

intr

anas

alSI

V-s

pec

ific

rect

alIg

Are

sponse

sw

ere

more

sign

ifica

ntly

per

sist

ent

ini.n.

vacc

inat

edth

anin

i.m

.va

ccin

ated

rhes

us

mac

aques

.[3

3]

I.n.

imm

uniz

atio

nin

duce

dsi

gnifi

cant

anti

-SIV

T-c

ell

resp

onse

sin

the

colo

rect

alm

uco

sa.

gp41-s

ubunit

anti

gens

graf

ted

on

viro

som

esIn

tram

usc

ula

ran

din

tran

asal

gp41-s

pec

ific

vagi

nal

IgA

s.[3

2]

HIV

-1tr

ansc

ytosi

s-blo

ckin

gpro

per

ties

.V

agin

alIg

Gs

with

neu

tral

izin

gan

d/o

rA

DC

Cac

tivi

ties

.R

ecom

bin

ant

repli

cati

ng

Vac

cinia

viru

sTia

nta

n/S

IVgp

e(p

rim

e)–

Ad5SI

Vgp

e(b

oost

)In

trao

ral

and

intr

anas

al(p

rim

e)an

din

tram

usc

ula

r(b

oost

)SI

V-s

pec

ific

CD

T-c

ellm

edia

ted

imm

unit

yag

ainst

Gag

and

Poli

sas

soci

ated

with

pro

tect

ion

agai

nst

hig

h-d

ose

intr

arec

talin

ocu

lati

on

ofSI

V(m

ac239).

[129]

RhC

MV

/SIV

Subcu

taneo

us

Induct

ion

of

SIV

-spec

ific

TEM

resp

onse

sat

pote

nti

alsi

tes

of

SIV

replica

tion.

[38]

RhC

MV

vect

ors

expre

ssin

gSI

VG

ag,R

ev/N

ef/T

at/E

nv

Subcu

taneo

us

Mai

nta

indif

fere

nti

ated

effe

ctor

mem

ory

T-c

ell

resp

onse

s.[1

24]

Mai

nta

inro

bust

SIV

-spec

ific

CD

and

CD

effe

ctor

mem

ory

T-c

ell

resp

onse

s.

Incr

ease

dre

sist

ance

toac

quis

itio

nof

SIV

mac

239

infe

ctio

n(i

ntr

arec

tal

chal

lenge

).Pri

me:

repli

cation-c

om

pet

ent

Ad5hr-

SIV

smH

4en

v/re

vm

uta

nt

and

Ad5hr-

SIV

239ga

gB

oost

:SI

Vm

ac251

gp120

pro

tein

ina

MPLA

-sta

ble

emuls

ion

Sublingu

alIn

tran

asal

/intr

atra

chea

lIn

trav

agin

alIn

trar

ecta

l

All

imm

uniz

atio

nro

ute

sel

icit

edsI

gAre

sponse

sat

mult

iple

muco

sal

site

s.Si

gnifi

cant

corr

elat

ion

of

vacc

ine-

induce

dsI

gAti

ters

inre

ctal

secr

etio

ns

with

del

ayed

acquis

itio

n.

[120]

Lact

ob

acill

us

pla

nta

rum

and

inac

tiva

ted

SIV

mac

239

Ora

lThe

tole

roge

nic

vacc

ine

induce

dM

HC

-Ib/E

-res

tric

ted

CD

Tre

gsth

atsu

ppre

ssed

SIV

-har

bori

ng

CD

T-c

ell

acti

vati

on.

[127]

Ex-v

ivo

SIV

repli

cati

on

in15/1

6an

imal

sw

ithout

induci

ng

SIV

-spec

ific

Abs

or

cyto

toxi

cT

lym

phocy

tes.

15/1

6an

imal

sw

ere

ster

ilel

ypro

tect

edaf

ter

intr

arec

tally

chal

lenge

with

SIV

mac

239

or

het

erolo

gous

stra

inSI

VB

670.

Co

1708 AIDS 2014, Vol 28 No 12

Another critical aspect of HIV cellular immune responsesis the location of the HIV-specific immune cells elicitedby immunization. Thus, the degree of protectionmediated by a live attenuated SIV vaccine stronglycorrelates with the location of SIV-specific effectorCD8þ T cells, in lymph nodes [45]. The maintenance ofthis protective T-cell response seems to be associated withpersistent replication of the live attenuated virus vaccinein follicular helper T (Tfh) cells.

Surprisingly, none of the candidate HIV vaccines tested sofar in human volunteers has been able to elicit viral loadcontrol. Neither the Step trial, based on the use of arecombinant Ad5 vector, nor the HVTN 505 trial, whichused a DNA prime followed by a recombinant Ad5 boost,nor the RV144 trial, using a recombinant Canarypoxprime followed by gp120 boosts, showed any significantimpact on viral load in vaccine recipients who becameinfected with HIV-1 [118,140,141]. There actually wassome evidence for immune selection pressure onbreakthrough HIV-1 sequences in the Step study,suggesting that, although too weak to be efficient,vaccine-elicited cellular immune responses did exertimmunologically relevant biological effects in humans[142]. The disappointing results of the Step and HVTN505 vaccine trials highlight the likely importance ofinducing mucosal immune responses that could signifi-cantly decrease virus replication in the mucosa andsubsequent viral dissemination to peripheral lymphoidtissues and blood. Another, but different example is theSIV protein encoding RhCMV, which is able to maintaindifferentiated effector memory T-cell responses at viralentry sites that show high efficacy at impairing SIVreplication at its earliest stage. This strategy can maintainrobust SIV-specific CD4þ and CD8þ effector memoryT-cell (TEM) responses that provide protection againstrepeated limiting-dose intrarectal challenge with SIV-mac239 [124].

Studies of the early kinetics of T-cell responses inpreviously vaccinated, acutely SIV-infected NHPs willallow the determination of whether an initial influx ofvirus-specific CD4þ T cells precedes robust CTLresponses and correlates with early containment [143].Alternatively, CD4þ CTL may directly contribute tocontainment of HIV infection [43].

Vaccination strategies that elicit cellularmucosal immune responses

Hansen et al. [38] reported that RhCMV/SIV vectorsused by subcutaneous route (s.c.) in the rhesus macaquesmodel are able to induce immune-mediated control ofhighly pathogenic SIVmac239 after repeated intrarectalchallenges and prior to irreversible establishment ofinfection. An early complete control of SIV was observed

pyright © Lippincott Williams & Wilkins. Unautho

in 13 of 24 rhesus macaques receiving either RhCMValone or RhCMV (s.c. prime)/Ad5 (i.m. boost) vectors,and a long-term protection (�1 year) was observed in 12of these 13 animals [124]. The immunologic assaysperformed in mononuclear cell preparations from bloodand tissues suggest that this control is related to the highfrequency of SIV-specific T-cell responses (CD8þ, andpossibly CD4þ). These responses are located both inmucosal portals of entry and at potential sites of distantviral spread and are indefinitely maintained by thepersistent RhCMV vectors, and can protect withoutanamnestic expansion.

The finding that RhCMV/SIV vector-protected rhesusmacaques are able to control haematogenous SIVdissemination after both intrarectal and intravaginalchallenge suggested that the immune responses elicitedby these vectors might provide protection even whenmucosal surfaces are bypassed [44]. Thus, persistentvectors such as CMV and their associated TEM responsesmight significantly contribute to an efficacious HIV/AIDS vaccine.

The NHP model was also used to test the vaccinationapproach using a plasmid DNA prime/rAd5 boostvaccine developed to induce both CD8þ T lymphocyteresponses and Env-specific Ab responses [125]. Afterrepeated intrarectal challenges, the vaccine failed toprotect against SIVmac251, but 50% of vaccinatedmonkeys were protected from infection withSIVsmE660. Although the exploration of immunecorrelates suggests that a NAb may be responsible forthe conferred protection against mucosal acquisition ofSIVsmE660, the reduction in peak plasma virus RNAimplicates CTL in the control of SIV replication onceinfection is established.

Intramuscular vaccination of rhesus macaques withAd/MVA or Ad/Ad vector expressing SIV Gag, Poland Env antigens, were also investigated for their capacityto induce CD8þT lymphocytes and to test whether theseresponses predict virologic control following SIV mucosalchallenge [130].

They observed that CD8þ cell mediated SIV inhibitionwas significantly associated with Gag-specific cellularimmunity but not Pol or Env-specific cellular immunityand that CD8þ lymphocytes from 23 vaccinated rhesusmacaques inhibited replication of the virus in vitro.Moreover, the level of inhibition prior to challenge wasinversely correlated with set point SIV plasma viral loadafter intrarectal challenge. These findings demonstratethat in-vitro viral inhibition following vaccinationlargely reflects Gag-specific cellular immune responsesand correlates with in-vivo control of viremia followinginfection. These data suggest the importance ofincluding Gag in an HIV-1 vaccine in which controlis desired.

rized reproduction of this article is prohibited.

Progress in HIV vaccines inducing mucosal responses Pavot et al. 1709

Rhesus macaques immunized by the i.n. route with a SIVDNA/MVA prime-boost regimen also demonstratedsignificant anti-SIV CTL responses in the colorectalmucosa and a better control of rectal SIVmac251infection when compared with macaques given the samevaccine by the i.m. route [33]. However, it was reportedthat an i.m. injection of nonreplicating recombinantAdenovirus vectors into rhesus macaques is able tosignificantly induce SIV-specific CTL responses thatpersist for over 2 years in multiple mucosal tissues, such ascolorectal, duodenal and vaginal biopsy specimens [126].

Despite the fact that mucosal vaccination often elicitslower magnitude HIV-specific T-cell responses whencompared with systemic vaccination, mucosal immuniz-ation can elicit better protection against HIV challengemediated by higher avidity CTLs in macaques, as assessedin systemic fluids [144]. Interestingly, interleukin (IL)-13seems to be detrimental to the efficient avidity of theseT-cells in the mouse model [145]. Recombinant HIV-1vaccines that coexpress the soluble or membrane-boundforms of the IL-13 receptor a2 (IL-13Ra2), and whichcan block IL-13 activity at the immunization site, wereused to make wild-type mice comparable to IL-13knock-out animals [146]. After an i.n./i.m. prime-boostvaccination, these vaccines adjuvanted with IL-13Ra2were shown to induce multifunctional mucosal CD8þ

T-cell responses in the lung, genito-rectal nodes andPeyer’s patches with greatly enhanced functional avidityand broader cytokine/chemokine profiles that providedgreater protection against a surrogate mucosal HIV-1challenge [146].

As mucosal prime-boost immunizations elicit significantnumbers of high avidity effector memory CD8þ CTL inmucosal and systemic compartments, they appear to be anessential component of any immunization approach thataims at establishing protective frontline defenses againstHIV-1 infection.

Exploring mucosal routes of immunization

Systemically delivered viral vectors can induce mucosalimmune responses against HIV-1 or SIV, most notably inthe gut, rectal and genital mucosa [147,148]. However,the strength of these responses is generally poor. Forexample, Ad-vectored vaccines have been shown toinduce low levels of mucosal immune responses aftersystemic inoculation, which are approximately 10 timeslower than the immune responses induced at systemicsites. Low-level mucosal immune responses have alsobeen seen in individuals inoculated i.m. with arecombinant pox virus vector [149]. However, at thistime, there are very few validated mucosal vaccinesagainst any infectious disease [35] and the mucosalvaccines already available provide protection only via

Copyright © Lippincott Williams & Wilkins. Unaut

induction of Ab responses. There are no current mucosalvaccines that are known to induce strong protectivecellular immune responses at the systemic or mucosallevel. Therefore, understanding the biology of themucosal immune system in order to develop bettermucosal vaccines that can induce both humoral andcellular immunity is needed.

Mucosal vaccines using oral or nasal routes have the greatadvantages of being painless, easy to administer on a largescale and easier to store and to deliver than currentsystemic vaccines [35]. Vaccination at a mucosal sitestimulates local immunity as well as immunity in othermucosal sites and usually also induces systemic immuneresponses detectable in the blood, spleen and peripherallymph nodes. This is in contrast to systemically deliveredvaccines, which are usually limited in their ability tostimulate an immune response in mucosal tissues[150,151].

Vaccines that target the nasal, oral, rectal or urogenitalmucosa have been under investigation for some time,using attenuated virus, inactivated virus, recombinantvirus, DNA, dendritic cells or peptides [152–155]. Oralimmunization strategies have been shown to induceHIV/SIV-specific immune responses in the gastrointes-tinal tract [156–158], whereas nasal immunizationstrategies have been reported to induce robust immuneresponses in the colorectal mucosa and genitourinarytracts in the NHP model [32,33,120]. Therefore, amucosal immunization strategy using both the oral andnasal routes should be able to induce potent immuneresponses at the mucosal surfaces potentially involved inHIV entry.

A promising approach to mucosal vaccination has beenthe use of virus-like particle (VLP) vaccines. VLPs aregenomeless viral particles (pseudovirions), obtained byspontaneous assembly of viral capsid proteins. They aresimilar in size and conformation to intact virions but arenonreplicating and nonpathogenic. These immunogenscan be administered as purified particles or as DNAplasmids expressing the viral proteins necessary to formVLPs in vivo [159,160]. Several successful VLP vaccineshave been developed against the sexually transmittedHPVs and tested in human trials (influenza) attesting tothe potential efficacy of VLPs as HIV-1 vaccine candidates[161,162]. VLPs can be used as potent mucosal HIV-1vaccine candidates (HIV-VLPs). Their administration byi.n. (prime) and i.m. (boost) has been shown to elicitvaginal and systemic humoral immune responses in therhesus macaques model [163]. Despite the fact that i.n.vaccines delivered into the nostrils are an attractive modeof immunization, one should be cautious of the risks ofpassage into the brain through olfactory nerves that couldbe the source of important adverse effects. As an example,the i.n. vaccine NasalFlu (Berna Biotech, Switzerland),containing an enzymatically active Escherichia coli labile

horized reproduction of this article is prohibited.

Co

1710 AIDS 2014, Vol 28 No 12

toxin adjuvant, was recalled after the establishment of anassociation with facial nerve paralysis (Bell’s palsy) [164].

Another concept that has been recently assayed in therhesus macaques model demonstrated that induction ofimmunological tolerance with a tolerogenic vaccine bymucosal route can prevent SIV infection [127]. The oraladministration of iSIVmac239 and Lactobacillus plantarum,a commensal bacterium of the digestive tract that isknown to induce immunologic tolerance, stimulatedmacaques to develop a thus far unrecognized type ofSIV-specific tolerance. This tolerance was characterizedby the suppression of SIV-specific Ab and CTL responses,and activation of a subset of CD8þ T cells that areSIV-specific, noncytolytic and MHC-Ib/E restricted.These cells apparently have the ability to suppress CD4þ

T cells activated by SIV and thereby prevent theestablishment of productive SIV infection both in vivoand in vitro.

Adjuvants as tools to orientate mucosalimmune responses

Adjuvants can be defined as substances that enhance theimmune response to the antigen(s) with which they arecoadministered. Despite their potentially critical role inthe efficacy of vaccines, relatively few adjuvants arecurrently used in commercial vaccines. Both the choiceof the adjuvant and the route of administration can greatlyaffect the type and potency of the immune responseelicited. To date, a number of approaches have beendeveloped in an effort to increase the immunogenicity ofHIV vaccines, including the use of molecular adjuvantsand cytokine adjuvants for protein antigens (Table 3)[165–177].

The addition of toxins or nontoxic derivatives of choleratoxin or mutant E. coli labile toxin to mucosalimmunization regimens has been shown to enhancesystemic immune responses [178]. The adjuvant activityof cholera toxin or labile toxin (and derivatives) can beexplained by their ability to affect several steps involved inthe induction of the immune response such as anincreased permeability of intestinal epithelium resultingin increased antigen uptake, enhancing antigen presen-tation, the promotion of IgA formation via B-cell isotypedifferentiation as well as effects on T-cell proliferation andcytokine production [179]. However, these adjuvants arenot devoid of a possible risk of severe adverse effects, asseen with the NasalFlu labile toxin adjuvanted vaccine[164].

Regarding the potential role of cytokines as adjuvants inmucosal HIV vaccine development, early clinical studiesusing protein antigens have shown that using pro-inflammatory cytokine adjuvants such as IL-1 by the i.n.

pyright © Lippincott Williams & Wilkins. Unautho

route effectively induced not only serum and vaginal IgGsbut also vaginal IgAs [180]. Many of the cytokineapproaches that have been tested in HIV vaccinedevelopment have been covered in a recent review[177] and will not be addressed here in further detail.Various combinations of IL-12, IL-15 and/or granulo-cyte-macrophage colony-stimulating factor (GM-CSF)have yielded mixed results, although, in combinationwith a DNA/MVA prime boost regimen, GM-CSF wasshown to effectively induce protective mucosal IgG andIgA production [34]. Of note, promising results havebeen observed in rhesus macaques using an IL-2adjuvanted DNA vaccine, which allowed control ofviremia and prevention of AIDS in an NHP model [181].

Over the past 10 years, there have been considerableadvances in both our understanding of the signallingpathways and receptors involved in recognition ofpathogens by the innate immune system and in theimportance of this system in then influencing an adaptiveimmune response. Detection of microbes by the innateimmune system is largely driven by pattern recognitionreceptors, including the toll-like receptors (TLRs) thatrecognize common molecular structures found on thosemicrobial agents that represent a potential danger for thedefending host organism. For HIV, it has been shown thatpolymorphisms in TLR4, 7, 8 and 9 can play a role inboth disease progression and viral load. This improvedunderstanding is now leading to the development ofnovel HIV vaccine adjuvants. TLR3 shows promisingresults when used with vaccine Ags and selective DEC-205/CD205 Ab delivery to dendritic cells. Similarly,TLR7/8 and TLR9 vaccine conjugates have been shownto enhance immune responses. Used together, IL-15 andagonists for TLR2/6, 3 and 9 synergistically upregulatedvaccine responses to recombinant MVA virus expressingviral proteins from SIVmac239 [182]. The activation ofTLR9 via unmethylated CpG motifs or related syntheticoligodeoxynucleotides (CpG ODN) mimicking bacterialDNA and thus acting as a danger signal of bacterialinvasion has also been shown to rapidly activate a varietyof innate immune cells through the Toll/IL-1 pathway toproduce Th1 cytokines and activation of APCs andB-cells. Vaginal administration of CpG ODN can inducethe rapid production of Th1 cytokines such as interferon-gamma (IFN-g), IL-12 or IL-18 in the female genitaltract [183]. These studies highlight the potential of theTLRs as agonists for HIV vaccines. Similarly, severalvaccine approaches using the TLR agonist Poly I:C andderivatives have revealed their capacity to stimulate HIVspecific immune responses in specific cell types and at sitesof mucosal exposure to pathogens [184–187]. Likewise,monophosphoryl lipid A (MPLA) from lipopolysacchar-ide (LPS) of Salmonella minnesota used as an adjuvant inparenterally administered vaccines has been shown toinduce antigen-specific mucosal and systemic cellularimmunity and Ab responses following oral or i.n. delivery,probably through activation of TLR2 and 4 [188,189].

rized reproduction of this article is prohibited.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibi

Progress in HIV vaccines inducing mucosal responses Pavot et al. 1711

Tab

le3.

Exam

ple

sof

vacc

inat

ion

stra

tegi

esto

induce

spec

ific

muco

sal

imm

une

resp

onse

sag

ainst

HIV

anti

gens.

Adju

vant

Del

iver

yst

rate

gyR

oute

of

imm

uniz

atio

nM

odel

Muco

sal

imm

une

resp

onse

Ref

eren

ces

GM

-CSF

DN

AD

NA

/SH

IV.8

9.6

.VLP

and

MV

A/S

HIV

-89.6

Intr

ader

mal

or

intr

amusc

ula

rR

hes

us

mac

aque

GM

-CSF

contr

ibute

sto

pro

tect

ion

by

enhan

cing

the

avid

ity

mat

ura

tion

of

anti

-Env

IgG

inblo

od

and

the

pre

sence

of

long-

last

ing

antivi

ral

IgA

inre

ctal

secr

etio

ns.

[34]

Pla

smid

CC

L19

or

CC

L28

HIV

-1gp

140

Intr

anas

alor

intr

amusc

ula

rM

ouse

pC

CL1

9en

han

ced

vagi

nal

-spec

ific

IgG

resp

onse

sw

hen

adm

inis

tere

di.

m.

or

i.n.

[165]

pC

CL2

8en

han

ced

vagi

nal

IgG

resp

onse

sfo

llow

ing

i.n.

but

not

i.m

.pC

CL1

9an

dpC

CL2

8en

han

ced

vagi

nal

IgA

by

i.m

.pC

CL1

9an

dpC

CL2

8en

han

ced

IgA

leve

lsw

ith

abal

ance

dIg

G:IgA

resp

onse

by

i.n.

CC

L19

and

CC

L28

also

incr

ease

dIg

cell

sin

colo

rect

alti

ssue.

CC

L19

and

CC

L28

pro

mote

den

han

ced

neu

tral

izin

gre

sponse

sin

sera

and

vagi

nal

secr

etio

ns.

Chole

rato

xin

(CT)

HIV

-1pep

tide

Intr

arec

tal,

intr

anas

ally

or

intr

agas

tric

ally

Mouse

Intr

arec

talim

muniz

atio

nw

ith

CT

induce

dlo

ng-

last

ing,

antige

n-s

pec

ific

CTL

mem

ory

inPey

er’s

pat

ches

,la

min

ap

rop

ria

and

sple

en.

[166]

Syst

emic

imm

uniz

atio

nin

duce

dsp

ecifi

cC

TL

only

inth

esp

leen

.C

TA

1-D

DM

onom

eric

or

trim

eric

HIV

-1En

vIn

tran

asal

or

intr

aper

itonea

lM

ouse

CTA

1-D

Dst

imula

tes

HIV

-1an

ti-E

nv

seru

mIg

Gan

dm

uco

sal

IgA

foll

ow

ing

i.n.

adm

inis

trat

ion.

[167]

Muta

nt

E.co

lila

bile

toxi

nLT

(R192G

)H

IV-1

pep

tide

Intr

arec

tal

Mouse

LT(R

192G

)w

asas

effe

ctiv

eas

or

more

effe

ctiv

eth

anC

Tat

induci

ng

am

uco

sal

CTL

resp

onse

.

[168]

GM

-CSF

syner

gize

dw

ith

LT(R

192G

)Poly

I:C

(TLR

3)

HIV

pep

tides

and

anti

bodie

sto

DEC

-205/C

D205

Subcu

taneo

us

or

intr

aper

itonea

lM

ouse

DEC

-tar

gete

d,

HIV

Gag

p24

along

wit

hpoly

I:C

induce

pro

tect

ive

CD

Tce

llre

sponse

sat

airw

aym

uco

sal

surf

aces

.

[169]

MPLA

(TLR

4)

HIV

-1gp

140

Subli

ngu

al,

intr

anas

al,

intr

avag

inal

or

subcu

taneo

us

Mouse

MPLA

enhan

ced

resp

onse

saf

ter

i.n.

or

s.c.

[170]

Vag

inal

spec

ific

IgA

:N

onad

juva

nte

dgp

140:

s.l.>

i.n.¼

s.c.

Adju

vante

dgp

140:

i.n.>

s.l.>

s.c.

I.V

agfa

iled

toin

duce

any

det

ecta

ble

muco

sal

resp

onse

sev

enin

the

pre

sence

of

MPLA

.

ted.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

1712 AIDS 2014, Vol 28 No 12

Tab

le3

(continued

)

Adju

vant

Del

iver

yst

rate

gyR

oute

of

imm

uniz

atio

nM

odel

Muco

sal

imm

une

resp

onse

Ref

eren

ces

Glu

copyr

anosy

lLi

pid

Adju

vant

(GLA

,TLR

4)

DN

A-M

VA

-Pro

tein

vacc

ine

Intr

amusc

ula

rM

ouse

and

rabbit

GLA

impro

ved

spec

ific

IgG

vagi

nal

resp

onse

s.[1

71]

GLA

(TLR

4)

HIV

-1gp

140

Intr

anas

alM

ouse

GLA

induce

dhig

hIg

Gan

dIg

Atitr

esin

nas

al,

vagi

nal

lava

ges

and

faec

es.

[172]

Hig

hnum

ber

sof

gp140-s

pec

ific

Ab

secr

etin

gce

lls

inth

ege

nit

altr

act.

Bac

teri

alflag

elli

n(F

liC

)or

trunca

ted

FliC

(tFl

iC)

(TLR

5)

HIV

VLP

sIn

tram

usc

ula

ror

intr

anas

alG

uin

eapig

Flag

elli

n-c

onta

inin

gV

LPs

elic

ithig

hle

vels

of

syst

emic

Ab

resp

onse

sby

eith

eri.

m.

or

i.n.,

asw

ell

asboth

syst

emic

and

muco

sal

imm

unit

yby

i.n.

[173]

VLP

s-Fl

iCw

ere

more

effe

ctiv

ein

induci

ng

syst

emic

resp

onse

s,w

hil

eV

LPs-

tFli

Cw

ere

more

effe

ctiv

ein

induci

ng

vagi

nal

IgA

resp

onse

sby

i.n.

CpG

OD

N(T

LR9)

Inac

tiva

ted

gp120-d

eple

ted

HIV

-1vi

rions

Intr

anas

alM

ouse

Lym

phocy

tes

isola

ted

from

genit

altr

acts

of

mic

eim

muniz

edw

ith

Ag

and

CpG

show

edsi

gnifi

cant

spec

ific

pro

life

rati

on

and

pro

duce

dsi

gnifi

cantl

yhig

her

leve

lsof

IFN

-g.

[174]

CD

lym

phocy

tes

wer

ein

crea

sed

inth

ege

nital

trac

tsof

mic

eim

muniz

edw

ith

HIV

-1A

gan

dC

pG

TLR

2,

3,

9ag

onis

ts/I

L-15

Pep

tide

(pri

me)

and

MV

A(b

oost

)In

trar

ecta

lR

hes

us

mac

aque

Com

bin

atio

nof

IL-1

5an

dTLR

agonis

tsm

edia

ted

par

tial

pro

tect

ion

agai

nst

SIV

rect

alch

alle

nge

.

[175]

Pre

serv

atio

nof

CD

Tce

llnum

ber

sin

the

colo

nm

uco

sa.

Hig

her

leve

lof

SIV

-spec

ific

tetr

amerþ

CD

resp

onse

sin

the

colo

ns

of

adju

vante

dan

imal

s.

Progress in HIV vaccines inducing mucosal responses Pavot et al. 1713

Other studies have shown that liposomes containing lipidA and HIV-1 proteins or peptide antigens could induceneutralizing ‘multispecific’ Abs in which the antigen-binding site of the Ab simultaneously binds both to theimmunizing lipid and protein epitope [190,191].

Conclusion

The challenges involved in the development of a HIVprophylactic vaccine are unprecedented in the history ofvaccinology. Three decades after the discovery of thevirus, the quest for a vaccine is still actively ongoing. Themajor obstacles met in the development of an efficaciousvaccine are the mutational variability and global diversityof the virus, which allow its easy escape from both thecellular and humoral responses of the host. Moreover,HIV-1 mainly infects the organism through the mucosalsites of the genital and intestinal tracts and rapidlyintegrates into memory T cells that become latent viralreservoirs. An efficient vaccine will therefore need to notonly induce potent and functional virus-specific Abs ableto block virus entry at the site of initial infection but alsoCD8þ T cells for virological control in lymphoid tissuesand lymph nodes.

As illustrated by the example of the human papillomavirus(HPV) vaccine, the parenteral route of immunizationcould be an efficient way to elicit protective mucosalimmunity, and indeed, numerous studies on HIV/SIVvaccines have shown that it is possible to induceprotection against rectal or vaginal challenges in NHPmodels by systemic active or passive immunization.However, systemic immunization usually generates onlylow humoral responses at mucosal sites that stem from thetransudation of IgGs from the blood into the genitour-inary tract, and can also induce the secretion ofimmunoglobulins that act through interaction with theneonatal Fc receptor. It is nevertheless possible thatmucosal immune responses induced by parenteralimmunization will be improved in the future by thedevelopment of specific adjuvants that would amplify andfavourize such a response.

Immunization by the mucosal route preferentially inducesIgA responses at the site of antigen delivery, as well as insecretions from anatomically remote mucosal sites. Thus,an effective mucosal route of immunization able to elicitspecific IgAs and CTL immunity in the genital mucosaappears to be the nasal mucosa and the aerodigestive tract.Data collected so far show that MALT-targeted adju-vanted vaccine design could be universally applied to anyform of HIV vaccine candidate, including peptides,subunit vaccines, VLPs, DNA or live recombinantvaccines. However, initial mucosal HIV-1 immunizationof immunologically naive individuals may induce a stateof mucosal tolerance. Systemic priming followed by

Copyright © Lippincott Williams & Wilkins. Unaut

mucosal boosting is likely to prevent this undesirableoutcome. Furthermore, such a sequence of immuniz-ations should elicit humoral immune responses in boththe systemic and the mucosal compartments.

Unfortunately, at this time, mucosal immunization hasbeen tested in only a limited number of studies, mainlybecause of the relatively inefficient uptake of antigens bymucosal surfaces and the unavailability of mucosaladjuvants approved for human use. Also, whereas systemicimmunity can readily be assessed from peripheral bloodsamples, systemic responses do not necessarily reflectresponses in mucosal compartments. Thus, in their NHPvaccination study, Bomsel et al. [32] observed highprotection after intravaginal challenge that was correlatedwith HIV-1 blocking Abs developed in the mucosalcompartment, but not in serum. Thus, antiviral mucosalimmune responses may be missed in peripheral blood.Numerous studies have assayed HIV-specific mucosalresponses in preclinical and clinical research, but anumber of difficulties have slowed progress in incorpor-ating such measurements. Indeed, processing mucosalsamples is more challenging and sampling proceduresprovide lower amounts of fluid or cells than bloodsampling. Moreover, mucosal sampling is more invasivethan blood sampling and takes more time and training ofclinical personnel.

If we are to ever fully realize the potential benefits ofmucosal vaccines to control HIV/AIDS, current researchshould be extended to the development of innovativeimmunological tools such as safe adjuvants, targetingmolecules and delivery vectors.

Acknowledgements

Authors would like to thank the ANRS (AgenceNationale de la Recherche sur le SIDA) to S.P. andN.R., the European Commission FP7 ADITEC program(HEALTH-F4-2011-280873) and FP7 Cut’hivac(HEALTH- 241904) to V.P. and B.V., the ANR (grantANR PECSDELLI and Euronanomed iNanoDCs;support to V.P., S.P. and B.V.) and the grant from theFondation pour la Recherche Medicale to V.P.

Conflicts of interestThe authors declare that there are no conflicts of interest.

References

1. Tebit DM, Ndembi N, Weinberg A, Quinones-Mateu ME.Mucosal transmission of human immunodeficiency virus. CurrHIV Res 2012; 10:3–8.

2. UNAIDS. Global report: UNAIDS report on the global AIDSepidemic. Geneva, Switzerland: UNAIDS; 2012 (http://www.unaids.org/en/media/unaids/contentassets/documents/epidemiology/2012/gr2012/20121120_unaids_global_report_2012_with_annexes_en.pdf).

horized reproduction of this article is prohibited.

Co

1714 AIDS 2014, Vol 28 No 12

3. Hladik F, McElrath MJ. Setting the stage: host invasion by HIV.Nat Rev Immunol 2008; 8:447–457.

4. Poudrier J, Thibodeau V, Roger M. Natural immunity to HIV: adelicate balance between strength and control. Clin DevImmunol 2012; 2012:875821.

5. Wyatt R, Sodroski J. The HIV-1 envelope glycoproteins: fuso-gens, antigens, and immunogens. Science 1998; 280:1884–1888.

6. Pantophlet R, Burton DR. GP120: target for neutralizing HIV-1antibodies. Annu Rev Immunol 2006; 24:739–769.

7. Overbaugh J, Morris L. The antibody response against HIV-1.Cold Spring Harb Perspect Med 2012; 2:a007039.

8. Kwong PD, Doyle ML, Casper DJ, Cicala C, Leavitt SA, MajeedS, et al. HIV-1 evades antibody-mediated neutralizationthrough conformational masking of receptor-binding sites.Nature 2002; 420:678–682.

9. Wei X, Decker JM, Wang S, Hui H, Kappes JC, Wu X, et al.Antibody neutralization and escape by HIV-1. Nature 2003;422:307–312.

10. Burton DR, Stanfield RL, Wilson IA, Antibody VS. HIV in aclash of evolutionary titans. Proc Natl Acad Sci U S A 2005;102:14943–14948.

11. Mascola JR, Montefiori DC. The role of antibodies in HIVvaccines. Annu Rev Immunol 2010; 28:413–444.

12. McElrath MJ, Haynes BF. Induction of immunity to humanimmunodeficiency virus type-1 by vaccination. Immunity2010; 33:542–554.

13. Gray ES, Moore PL, Choge IA, Decker JM, Bibollet-Ruche F, LiH, et al. Neutralizing antibody responses in acute humanimmunodeficiency virus type 1 subtype C infection. J Virol2007; 81:6187–6196.

14. Mikell I, Sather DN, Kalams SA, Altfeld M, Alter G, StamatatosL. Characteristics of the earliest cross-neutralizing antibodyresponse to HIV-1. PLoS Pathog 2011; 7:e1001251.

15. Tomaras GD, Binley JM, Gray ES, Crooks ET, Osawa K, MoorePL, et al. Polyclonal B cell responses to conserved neutraliza-tion epitopes in a subset of HIV-1-infected individuals. J Virol2011; 85:11502–11519.

16. Plotkin SA. Correlates of protection induced by vaccination.Clin Vaccine Immunol 2010; 17:1055–1065.

17. Amanna IJ, Slifka MK. Contributions of humoral and cellularimmunity to vaccine-induced protection in humans. Virology2011; 411:206–215.

18. Stamatatos L, Morris L, Burton DR, Mascola JR. Neutralizingantibodies generated during natural HIV-1 infection: goodnews for an HIV-1 vaccine? Nat Med 2009; 15:866–870.

19. Walker LM, Simek MD, Priddy F, Gach JS, Wagner D, ZwickMB, et al. A limited number of antibody specificities mediatebroad and potent serum neutralization in selected HIV-1infected individuals. PLoS Pathog 2010; 6:e1001028.

20. Kwong PD, Mascola JR. Human antibodies that neutralizeHIV-1: identification, structures, and B cell ontogenies.Immunity 2012; 37:412–425.

21. Moldt B, Rakasz EG, Schultz N, Chan-Hui PY, Swiderek K,Weisgrau KL, et al. Highly potent HIV-specific antibodyneutralization in vitro translates into effective protectionagainst mucosal SHIV challenge in vivo. Proc Natl Acad SciU S A 2012; 109:18921–18925.

22. Moog C, Dereuddre-Bosquet N, Teillaud JL, Biedma ME, HollV, Van Ham G, et al. Protective effect of vaginal application ofneutralizing and nonneutralizing inhibitory antibodies againstvaginal SHIV challenge in macaques. Mucosal Immunol 2014;7:46–56.

23. Kwong PD, Mascola JR, Nabel GJ. Broadly neutralizing anti-bodies and the search for an HIV-1 vaccine: the end of thebeginning. Nat Rev Immunol 2013; 13:693–701.

24. Walker LM, Phogat SK, Chan-Hui PY, Wagner D, Phung P,Goss JL, et al. Broad and potent neutralizing antibodies froman African donor reveal a new HIV-1 vaccine target. Science2009; 326:285–289.

25. Wu X, Yang ZY, Li Y, Hogerkorp CM, Schief WR, Seaman MS,et al. Rational design of envelope identifies broadly neutraliz-ing human monoclonal antibodies to HIV-1. Science 2010;329:856–861.

26. Walker LM, Huber M, Doores KJ, Falkowska E, Pejchal R,Julien JP, et al. Broad neutralization coverage of HIV bymultiple highly potent antibodies. Nature 2011; 477:466–470.

pyright © Lippincott Williams & Wilkins. Unautho

27. Scheid JF, Mouquet H, Ueberheide B, Diskin R, Klein F,Oliveira TY, et al. Sequence and structural convergence ofbroad and potent HIV antibodies that mimic CD4 binding.Science 2011; 333:1633–1637.

28. Scheid JF, Mouquet H, Feldhahn N, Seaman MS, Velinzon K,Pietzsch J, et al. Broad diversity of neutralizing antibodiesisolated from memory B cells in HIV-infected individuals.Nature 2009; 458:636–640.

29. Simek MD, Rida W, Priddy FH, Pung P, Carrow E, Laufer DS,et al. Human immunodeficiency virus type 1 elite neutrali-zers: individuals with broad and potent neutralizing activityidentified by using a high-throughput neutralization assaytogether with an analytical selection algorithm. J Virol2009; 83:7337–7348.

30. Malbec M, Porrot F, Rua R, Horwitz J, Klein F, Halper-Strom-berg A, et al. Broadly neutralizing antibodies that inhibitHIV-1 cell to cell transmission. J Exp Med 2013; 210:2813–2821.

31. Barouch DH, Liu J, Li H, Maxfield LF, Abbink P, Lynch DM,et al. Vaccine protection against acquisition of neutralization-resistant SIV challenges in rhesus monkeys. Nature 2012;482:89–93.

32. Bomsel M, Tudor D, Drillet AS, Alfsen A, Ganor Y, Roger MG,et al. Immunization with HIV-1 gp41 subunit virosomes in-duces mucosal antibodies protecting nonhuman primatesagainst vaginal SHIV challenges. Immunity 2011; 34:269–280.

33. Manrique M, Kozlowski PA, Wang SW, Wilson RL, MicewiczE, Montefiori DC, et al. Nasal DNA-MVA SIV vaccinationprovides more significant protection from progression toAIDS than a similar intramuscular vaccination. Mucosal Im-munol 2009; 2:536–550.

34. Lai L, Vodros D, Kozlowski PA, Montefiori DC, Wilson RL,Akerstrom VL, et al. GM-CSF DNA: an adjuvant for higheravidity IgG, rectal IgA, and increased protection against theacute phase of a SHIV-89.6P challenge by a DNA/MVAimmunodeficiency virus vaccine. Virology 2007; 369:153–167.

35. Pavot V, Rochereau N, Genin C, Verrier B, Paul S. Newinsights in mucosal vaccine development. Vaccine 2012;30:142–154.

36. Wright PF, Mestecky J, McElrath MJ, Keefer MC, Gorse GJ,Goepfert PA, et al. Comparison of systemic and mucosaldelivery of 2 canarypox virus vaccines expressing eitherHIV-1 genes or the gene for rabies virus G protein. J InfectDis 2004; 189:1221–1231.

37. Koup RA, Douek DC. Vaccine design for CD8 T lymphocyteresponses. Cold Spring Harb Perspect Med 2011; 1:a007252.

38. Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes CM,Coyne-Johnson L, et al. Profound early control of highlypathogenic SIV by an effector memory T-cell vaccine. Nature2011; 473:523–527.

39. Hersperger AR, Migueles SA, Betts MR, Connors M. Qualita-tive features of the HIV-specific CD8R T-cell response asso-ciated with immunologic control. Curr Opin HIV AIDS 2011;6:169–173.

40. McDermott AB, Koup RA. CD8(R) T cells in preventing HIVinfection and disease. AIDS 2012; 26:1281–1292.

41. Jost S, Altfeld M. Evasion from NK cell-mediated immuneresponses by HIV-1. Microbes Infect 2012; 14:904–915.

42. Ranasinghe S, Flanders M, Cutler S, Soghoian DZ, Ghebre-michael M, Davis I, et al. HIV-specific CD4 T cell responses todifferent viral proteins have discordant associations with viralload and clinical outcome. J Virol 2012; 86:277–283.

43. Soghoian DZ, Jessen H, Flanders M, Sierra-Davidson K, CutlerS, Pertel T, et al. HIV-specific cytolytic CD4 T cell responsesduring acute HIV infection predict disease outcome. Sci TranslMed 2012; 4:123ra125.

44. Hansen SG, Piatak M Jr, Ventura AB, Hughes CM, GilbrideRM, Ford JC, et al. Immune clearance of highly pathogenic SIVinfection. Nature 2013; 502:100–104.

45. Fukazawa Y, Park H, Cameron MJ, Lefebvre F, Lum R,Coombes N, et al. Lymph node T cell responses predict theefficacy of live attenuated SIV vaccines. Nat Med 2012;18:1673–1681.

46. Holmgren J, Czerkinsky C, Eriksson K, Mharandi A. Mucosalimmunisation and adjuvants: a brief overview of recent ad-vances and challenges. Vaccine 2003; 21 (Suppl 2):S89–S95.

47. Hladik F, Hope TJ. HIV infection of the genital mucosa inwomen. Curr HIV/AIDS Rep 2009; 6:20–28.

rized reproduction of this article is prohibited.

Progress in HIV vaccines inducing mucosal responses Pavot et al. 1715

48. Kaizu M, Weiler AM, Weisgrau KL, Vielhuber KA, May G,Piaskowski SM, et al. Repeated intravaginal inoculation withcell-associated simian immunodeficiency virus results in per-sistent infection of nonhuman primates. J Infect Dis 2006;194:912–916.

49. Carias AM, McCoombe S, McRaven M, Anderson M, Gallo-way N, Vandergrift N, et al. Defining the interaction of HIV-1with the mucosal barriers of the female reproductive tract. JVirol 2013; 87:11388–11400.

50. Salle B, Brochard P, Bourry O, Mannioui A, Andrieu T, PrevotS, et al. Infection of macaques after vaginal exposure to cell-associated simian immunodeficiency virus. J Infect Dis 2010;202:337–344.

51. Kolodkin-Gal D, Hulot SL, Korioth-Schmitz B, Gombos RB,Zheng Y, Owuor J, et al. Efficiency of cell-free and cell-associated virus in mucosal transmission of human immuno-deficiency virus type 1 and simian immunodeficiency virus.J Virol 2013; 87:13589–13597.

52. Salazar-Gonzalez JF, Bailes E, Pham KT, Salazar MG, GuffeyMB, Keele BF, et al. Deciphering human immunodeficiencyvirus type 1 transmission and early envelope diversification bysingle-genome amplification and sequencing. J Virol 2008;82:3952–3970.

53. Salazar-Gonzalez JF, Salazar MG, Keele BF, Learn GH, GiorgiEE, Li H, et al. Genetic identity, biological phenotype, andevolutionary pathways of transmitted/founder viruses in acuteand early HIV-1 infection. J Exp Med 2009; 206:1273–1289.

54. Shaw GM, Hunter E. HIV transmission. Cold Spring HarbPerspect Med 2012:2.

55. Gupta P, Collins KB, Ratner D, Watkins S, Naus GJ, LandersDV, et al. Memory CD4(R) T cells are the earliest detectablehuman immunodeficiency virus type 1 (HIV-1)-infected cellsin the female genital mucosal tissue during HIV-1 transmis-sion in an organ culture system. J Virol 2002; 76:9868–9876.

56. Zussman A, Lara L, Lara HH, Bentwich Z, Borkow G. Blockingof cell-free and cell-associated HIV-1 transmission throughhuman cervix organ culture with UC781. AIDS 2003; 17:653–661.

57. Sivard P, Berlier W, Picard B, Sabido O, Genin C, Misery L.HIV-1 infection of Langerhans cells in a reconstructed vaginalmucosa. J Infect Dis 2004; 190:227–235.

58. Bouschbacher M, Bomsel M, Verronese E, Gofflo S, Ganor Y,Dezutter-Dambuyant C, et al. Early events in HIV transmissionthrough a human reconstructed vaginal mucosa. AIDS 2008;22:1257–1266.

59. Maher D, Wu X, Schacker T, Horbul J, Southern P. HIV binding,penetration, and primary infection in human cervicovaginaltissue. Proc Natl Acad Sci U S A 2005; 102:11504–11509.

60. Miller CJ, Li Q, Abel K, Kim EY, Ma ZM, Wietgrefe S, et al.Propagation and dissemination of infection after vaginaltransmission of simian immunodeficiency virus. J Virol2005; 79:9217–9227.

61. Wu Z, Chen Z, Phillips DM. Human genital epithelial cellscapture cell-free human immunodeficiency virus type 1 andtransmit the virus to CD4R cells: implications for mechanismsof sexual transmission. J Infect Dis 2003; 188:1473–1482.

62. Berlier W, Bourlet T, Lawrence P, Hamzeh H, Lambert C,Genin C, et al. Selective sequestration of X4 isolates by humangenital epithelial cells: implication for virus tropism selectionprocess during sexual transmission of HIV. J Med Virol 2005;77:465–474.

63. Micsenyi AM, Zony C, Alvarez RA, Durham ND, Chen BK,Klotman ME. Postintegration HIV-1 infection of cervicalepithelial cells mediates contact-dependent productive infec-tion of T cells. J Infect Dis 2013; 208:1756–1767.

64. Margolis L, Shattock R. Selective transmission of CCR5-utiliz-ing HIV-1: the ‘gatekeeper’ problem resolved? Nat Rev Micro-biol 2006; 4:312–317.

65. Alfsen A, Yu H, Magerus-Chatinet A, Schmitt A, Bomsel M.HIV-1-infected blood mononuclear cells form an integrin-and agrin-dependent viral synapse to induce efficient HIV-1transcytosis across epithelial cell monolayer. Mol Biol Cell2005; 16:4267–4279.

66. Van Herrewege Y, Michiels J, Waeytens A, De Boeck G,Salden E, Heyndrickx L, et al. A dual chamber model of femalecervical mucosa for the study of HIV transmission and for theevaluation of candidate HIV microbicides. Antiviral Res 2007;74:111–124.

Copyright © Lippincott Williams & Wilkins. Unaut

67. Gorlani A, Forthal DN. Antibody-dependent enhancementand the risk of HIV infection. Curr HIV Res 2013; 11:421–426.

68. Phillips DM, Tan X, Perotti ME, Zacharopoulos VR. Mechan-ism of monocyte-macrophage-mediated transmission of HIV.AIDS Res Hum Retroviruses 1998; 14 (Suppl 1):S67–S70.

69. Dezzutti CS, Guenthner PC, Cummins JE Jr, Cabrera T, Mar-shall JH, Dillberger A, et al. Cervical and prostate primaryepithelial cells are not productively infected but sequesterhuman immunodeficiency virus type 1. J Infect Dis 2001;183:1204–1213.

70. Bobardt MD, Chatterji U, Selvarajah S, Van der Schueren B,David G, Kahn B, et al. Cell-free human immunodeficiencyvirus type 1 transcytosis through primary genital epithelialcells. J Virol 2007; 81:395–405.

71. Ibata B, Parr EL, King NJ, Parr MB. Migration of foreign lympho-cytes from the mouse vagina into the cervicovaginal mucosaand to the iliac lymph nodes. Biol Reprod 1997; 56:537–543.

72. Anderson DJ, Politch JA, Nadolski AM, Blaskewicz CD, Pud-ney J, Mayer KH. Targeting Trojan horse leukocytes for HIVprevention. AIDS 2010; 24:163–187.

73. Gali Y, Arien KK, Praet M, Van den Bergh R, Temmerman M,Delezay O, et al. Development of an in vitro dual-chambermodel of the female genital tract as a screening tool forepithelial toxicity. J Virol Methods 2010; 165:186–197.

74. Lawrence P, Portran D, Terrasse R, Palle S, Olivier T, Fantini J,et al. Selective transmigration of monocyte-associated HIV-1across a human cervical monolayer and its modulation byseminal plasma. AIDS 2012; 26:785–796.

75. Piguet V, Steinman RM. The interaction of HIV with dendriticcells: outcomes and pathways. Trends Immunol 2007; 28:503–510.

76. Kaul R, Plummer F, Clerici M, Bomsel M, Lopalco L, BrolidenK. Mucosal IgA in exposed, uninfected subjects: evidence for arole in protection against HIV infection. AIDS 2001; 15:431–432.

77. Kaul R, Trabattoni D, Bwayo JJ, Arienti D, Zagliani A, MwangiFM, et al. HIV-1-specific mucosal IgA in a cohort of HIV-1-resistant Kenyan sex workers. AIDS 1999; 13:23–29.

78. Hirbod T, Kaul R, Reichard C, Kimani J, Ngugi E, Bwayo JJ,et al. HIV-neutralizing immunoglobulin A and HIV-specificproliferation are independently associated with reduced HIVacquisition in Kenyan sex workers. AIDS 2008; 22:727–735.

79. Choi RY, Levinson P, Guthrie BL, Lohman-Payne B, Bosire R, LiuAY, etal. Cervicovaginal HIV-1-neutralizing immunoglobulin Adetected among HIV-1-exposed seronegative female partnersin HIV-1-discordant couples. AIDS 2012; 26:2155–2163.

80. Prodger JL, Hirbod T, Kigozi G, Nalugoda F, Reynolds SJ,Galiwango R, et al. Immune correlates of HIV exposurewithout infection in foreskins of men from Rakai, Uganda.Mucosal Immunol 2014; 7:634–644.

81. Yu X, Duval M, Lewis C, Gawron MA, Wang R, Posner MR,et al. Impact of IgA constant domain on HIV-1 neutralizingfunction of monoclonal antibody F425A1g8. J Immunol 2013;190:205–210.

82. Devito C, Broliden K, Kaul R, Svensson L, Johansen K, Kiama P,et al. Mucosal and plasma IgA from HIV-1-exposed uninfectedindividuals inhibit HIV-1 transcytosis across human epithelialcells. J Immunol 2000; 165:5170–5176.

83. Tudor D, Derrien M, Diomede L, Drillet AS, Houimel M, MoogC, et al. HIV-1 gp41-specific monoclonal mucosal IgAs de-rived from highly exposed but IgG-seronegative individualsblock HIV-1 epithelial transcytosis and neutralize CD4(R)cell infection: an IgA gene and functional analysis. MucosalImmunol 2009; 2:412–426.

84. Bernard NF, Yannakis CM, Lee JS, Tsoukas CM. Human im-munodeficiency virus (HIV)-specific cytotoxic T lymphocyteactivity in HIV-exposed seronegative persons. J Infect Dis1999; 179:538–547.

85. Goh WC, Markee J, Akridge RE, Meldorf M, Musey L, Karch-mer T, et al. Protection against human immunodeficiencyvirus type 1 infection in persons with repeated exposure:evidence for T cell immunity in the absence of inheritedCCR5 coreceptor defects. J Infect Dis 1999; 179:548–557.

86. Geldmacher C, Currier JR, Herrmann E, Haule A, Kuta E,McCutchan F, et al. CD8 T-cell recognition of multiple epitopeswithin specific Gag regions is associated with maintenance of alow steady-state viremia in human immunodeficiency virustype 1-seropositive patients. J Virol 2007; 81:2440–2448.

horized reproduction of this article is prohibited.

Co

1716 AIDS 2014, Vol 28 No 12

87. Almeida JR, Sauce D, Price DA, Papagno L, Shin SY, Moris A,et al. Antigen sensitivity is a major determinant of CD8R T-cell polyfunctionality and HIV-suppressive activity. Blood2009; 113:6351–6360.

88. Watkins JD, Sholukh AM, Mukhtar MM, Siddappa NB, LakhasheSK, Kim M, et al. Anti-HIV IgA isotypes: differential virioncapture and inhibition of transcytosis are linked to preventionof mucosal R5 SHIV transmission. AIDS 2013; 27:F13–F20.

89. Burton DR, Hessell AJ, Keele BF, Klasse PJ, Ketas TA, Moldt B,et al. Limited or no protection by weakly or nonneutralizingantibodies against vaginal SHIV challenge of macaques com-pared with a strongly neutralizing antibody. Proc Natl AcadSci U S A 2011; 108:11181–11186.

90. Eisele E, Siliciano RF. Redefining the viral reservoirs thatprevent HIV-1 eradication. Immunity 2012; 37:377–388.

91. Mehandru S, Poles MA, Tenner-Racz K, Horowitz A, Hurley A,Hogan C, et al. Primary HIV-1 infection is associated withpreferential depletion of CD4R T lymphocytes from effectorsites in the gastrointestinal tract. J Exp Med 2004; 200:761–770.

92. Katlama C, Deeks SG, Autran B, Martinez-Picado J, vanLunzen J, Rouzioux C, et al. Barriers to a cure for HIV:new ways to target and eradicate HIV-1 reservoirs. Lancet2013; 381:2109–2117.

93. Chun TW, Nickle DC, Justement JS, Meyers JH, Roby G,Hallahan CW, et al. Persistence of HIV in gut-associatedlymphoid tissue despite long-term antiretroviral therapy. JInfect Dis 2008; 197:714–720.

94. Schnell G, Joseph S, Spudich S, Price RW, Swanstrom R. HIV-1replication in the central nervous system occurs in two dis-tinct cell types. PLoS Pathog 2011; 7:e1002286.

95. Blackard JT. HIV compartmentalization: a review on a clini-cally important phenomenon. Curr HIV Res 2012; 10:133–142.

96. Benjelloun F, Lawrence P, Verrier B, Genin C, Paul S. Role ofhuman immunodeficiency virus type 1 envelope structure inthe induction of broadly neutralizing antibodies. J Virol 2012;86:13152–13163.

97. Baba TW, Liska V, Hofmann-Lehmann R, Vlasak J, Xu W,Ayehunie S, et al. Human neutralizing monoclonal antibodiesof the IgG1 subtype protect against mucosal simian-humanimmunodeficiency virus infection. Nat Med 2000; 6:200–206.

98. Mascola JR, Stiegler G, VanCott TC, Katinger H, Carpenter CB,Hanson CE, et al. Protection of macaques against vaginaltransmission of a pathogenic HIV-1/SIV chimeric virus bypassive infusion of neutralizing antibodies. Nat Med 2000;6:207–210.

99. Hofmann-Lehmann R, Vlasak J, Rasmussen RA, Smith BA,Baba TW, Liska V, et al. Postnatal passive immunization ofneonatal macaques with a triple combination of humanmonoclonal antibodies against oral simian-human immuno-deficiency virus challenge. J Virol 2001; 75:7470–7480.

100. Parren PW, Marx PA, Hessell AJ, Luckay A, Harouse J, Cheng-Mayer C, et al. Antibody protects macaques against vaginalchallenge with a pathogenic R5 simian/human immunodefi-ciency virus at serum levels giving complete neutralization invitro. J Virol 2001; 75:8340–8347.

101. Nishimura Y, Igarashi T, Haigwood N, Sadjadpour R, PlishkaRJ, Buckler-White A, et al. Determination of a statisticallyvalid neutralization titer in plasma that confers protectionagainst simian-human immunodeficiency virus challenge fol-lowing passive transfer of high-titered neutralizing antibo-dies. J Virol 2002; 76:2123–2130.

102. Mascola JR. Defining the protective antibody response forHIV-1. Curr Mol Med 2003; 3:209–216.

103. Hessell AJ, Poignard P, Hunter M, Hangartner L, Tehrani DM,Bleeker WK, et al. Effective, low-titer antibody protectionagainst low-dose repeated mucosal SHIV challenge in maca-ques. Nat Med 2009; 15:951–954.

104. Hessell AJ, Rakasz EG, Poignard P, Hangartner L, Landucci G,Forthal DN, et al. Broadly neutralizing human anti-HIV anti-body 2G12 is effective in protection against mucosal SHIVchallenge even at low serum neutralizing titers. PLoS Pathog2009; 5:e1000433.

105. Hessell AJ, Hangartner L, Hunter M, Havenith CE, BeurskensFJ, Bakker JM, et al. Fc receptor but not complement binding isimportant in antibody protection against HIV. Nature 2007;449:101–104.

pyright © Lippincott Williams & Wilkins. Unautho

106. Armbruster C, Stiegler GM, Vcelar BA, Jager W, Koller U, JilchR, et al. Passive immunization with the anti-HIV-1 humanmonoclonal antibody (hMAb) 4E10 and the hMAb combina-tion 4E10/2F5/2G12. J Antimicrob Chemother 2004; 54:915–920.

107. Klein K, Veazey RS, Warrier R, Hraber P, Doyle-Meyers LA,Buffa V, et al. Neutralizing IgG at the portal of infectionmediates protection against vaginal simian/human immuno-deficiency virus challenge. J Virol 2013; 87:11604–11616.

108. Alexander R, Mestecky J. Neutralizing antibodies in mucosalsecretions: IgG or IgA? Curr HIV Res 2007; 5:588–593.

109. Davids BJ, Palm JE, Housley MP, Smith JR, Andersen YS,Martin MG, et al. Polymeric immunoglobulin receptor inintestinal immune defense against the lumen-dwelling proto-zoan parasite Giardia. J Immunol 2006; 177:6281–6290.

110. Iqbal SM, Kaul R. Mucosal innate immunity as a determinantof HIV susceptibility. Am J Reprod Immunol 2008; 59:44–54.

111. Benjelloun F, Dawood R, Urcuqui-Inchima S, Rochereau N,Chanut B, Verrier B, et al. Secretory IgA specific for MPER canprotect from HIV-1 infection in vitro. AIDS 2013; 27:1992–1995.

112. Belec L, Georges AJ, Steenman G, Martin PM. Antibodies tohuman immunodeficiency virus in vaginal secretions of het-erosexual women. J Infect Dis 1989; 160:385–391.

113. Dorrell L, Hessell AJ, Wang M, Whittle H, Sabally S, Rowland-Jones S, et al. Absence of specific mucosal antibody responsesin HIV-exposed uninfected sex workers from the Gambia.AIDS 2000; 14:1117–1122.

114. Fiore JR, Laddago V, Lepera A, La Grasta L, Di Stefano M,Saracino A, et al. Limited secretory-IgA response in cervi-covaginal secretions from HIV-1 infected, but not high riskseronegative women: lack of correlation to genital viralshedding. New Microbiol 2000; 23:85–92.

115. Azizi A, Ghunaim H, Diaz-Mitoma F, Mestecky J. MucosalHIV vaccines: a holy grail or a dud? Vaccine 2010; 28:4015–4026.

116. Plotkin SA. Vaccines: correlates of vaccine-induced immu-nity. Clin Infect Dis 2008; 47:401–409.

117. Plotkin SA. Complex correlates of protection after vaccina-tion. Clin Infect Dis 2013; 56:1458–1465.

118. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kaewkungwal J,Chiu J, Paris R, et al. Vaccination with ALVAC and AIDSVAXto prevent HIV-1 infection in Thailand. N Engl J Med 2009;361:2209–2220.

119. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, TomarasGD, Alam SM, et al. Immune-correlates analysis of an HIV-1vaccine efficacy trial. N Engl J Med 2012; 366:1275–1286.

120. Xiao P, Patterson LJ, Kuate S, Brocca-Cofano E, Thomas MA,Venzon D, et al. Replicating adenovirus-simian immunodefi-ciency virus (SIV) recombinant priming and envelope proteinboosting elicits localized, mucosal IgA immunity in rhesusmacaques correlated with delayed acquisition following arepeated low-dose rectal SIV(mac251) challenge. J Virol2012; 86:4644–4657.

121. Patterson LJ, Kuate S, Daltabuit-Test M, Li Q, Xiao P, McKinnonK, et al. Replicating adenovirus-simian immunodeficiency virus(SIV) vectors efficiently prime SIV-specific systemic and mu-cosal immune responses by targeting myeloid dendritic cellsand persisting in rectal macrophages, regardless of immuniza-tion route. Clin Vaccine Immunol 2012; 19:629–637.

122. Leroux-Roels G, Maes C, Clement F, van Engelenburg F, vanden Dobbelsteen M, Adler M, et al. Randomized phase I:safety, immunogenicity and mucosal antiviral activity inyoung healthy women vaccinated with HIV-1 Gp41 P1 pep-tide on virosomes. PLoS One 2013; 8:e55438.

123. Barouch DH, Stephenson KE, Borducchi EN, Smith K, StanleyK, McNally AG, et al. Protective efficacy of a global HIV-1mosaic vaccine against heterologous SHIV challenges inrhesus monkeys. Cell 2013; 155:531–539.

124. Hansen SG, Vieville C, Whizin N, Coyne-Johnson L, Siess DC,Drummond DD, et al. Effector memory T cell responses areassociated with protection of rhesus monkeys from mucosalsimian immunodeficiency virus challenge. Nat Med 2009;15:293–299.

125. Letvin NL, Rao SS, Montefiori DC, Seaman MS, Sun Y, Lim SY,et al. Immune and genetic correlates of vaccine protectionagainst mucosal infection by SIV in monkeys. Sci Transl Med2011; 3:81ra36.

rized reproduction of this article is prohibited.

Progress in HIV vaccines inducing mucosal responses Pavot et al. 1717

126. Li H, Liu J, Carville A, Mansfield KG, Lynch D, Barouch DH.Durable mucosal simian immunodeficiency virus-specificeffector memory T lymphocyte responses elicited by recom-binant adenovirus vectors in rhesus monkeys. J Virol 2011;85:11007–11015.

127. Lu W, Chen S, Lai C, Guo W, Fu L, Andrieu JM. Induction ofCD8R regulatory T cells protects macaques against SIVchallenge. Cell Rep 2012; 2:1736–1746.

128. Liu J, Li H, Iampietro MJ, Barouch DH. Accelerated hetero-logous adenovirus prime-boost SIV vaccine in neonatal rhesusmonkeys. J Virol 2012; 86:7829–7835.

129. Sun C, Chen Z, Tang X, Zhang Y, Feng L, Du Y, et al. Mucosalpriming with a replicating-vaccinia virus-based vaccineelicits protective immunity to simian immunodeficiencyvirus challenge in rhesus monkeys. J Virol 2013; 87:5669–5677.

130. Stephenson KE, Li H, Walker BD, Michael NL, Barouch DH.Gag-specific cellular immunity determines in vitro viral in-hibition and in vivo virologic control following simian im-munodeficiency virus challenges of vaccinated rhesusmonkeys. J Virol 2012; 86:9583–9589.

131. Kiepiela P, Ngumbela K, Thobakgale C, Ramduth D, Honey-borne I, Moodley E, et al. CD8R T-cell responses to differentHIV proteins have discordant associations with viral load. NatMed 2007; 13:46–53.

132. Dahirel V, Shekhar K, Pereyra F, Miura T, Artyomov M,Talsania S, et al. Coordinate linkage of HIV evolution revealsregions of immunological vulnerability. Proc Natl Acad SciU S A 2011; 108:11530–11535.

133. Yamamoto T, Johnson MJ, Price DA, Wolinsky DI, Almeida JR,Petrovas C, et al. Virus inhibition activity of effector memoryCD8(R) T cells determines simian immunodeficiency virusload in vaccinated monkeys after vaccine breakthroughinfection. J Virol 2012; 86:5877–5884.

134. Kawada M, Tsukamoto T, Yamamoto H, Iwamoto N, KuriharaK, Takeda A, et al. Gag-specific cytotoxic T-lymphocyte-basedcontrol of primary simian immunodeficiency virus replicationin a vaccine trial. J Virol 2008; 82:10199–10206.

135. Liu J, O’Brien KL, Lynch DM, Simmons NL, La Porte A,Riggs AM, et al. Immune control of an SIV challenge by aT-cell-based vaccine in rhesus monkeys. Nature 2009; 457:87–91.

136. Iwamoto N, Tsukamoto T, Kawada M, Takeda A, YamamotoH, Takeuchi H, et al. Broadening of CD8R cell responses invaccine-based simian immunodeficiency virus controllers.AIDS 2010; 24:2777–2787.

137. Mudd PA, Martins MA, Ericsen AJ, Tully DC, Power KA, BeanAT, et al. Vaccine-induced CD8R T cells control AIDS virusreplication. Nature 2012; 491:129–133.

138. Betts MR, Nason MC, West SM, De Rosa SC, Migueles SA,Abraham J, et al. HIV nonprogressors preferentially maintainhighly functional HIV-specific CD8R T cells. Blood 2006;107:4781–4789.

139. Ferre AL, Hunt PW, Critchfield JW, Young DH, Morris MM,Garcia JC, et al. Mucosal immune responses to HIV-1 in elitecontrollers: a potential correlate of immune control. Blood2009; 113:3978–3989.

140. Flynn NM, Forthal DN, Harro CD, Judson FN, Mayer KH, ParaMF, et al. Placebo-controlled phase 3 trial of a recombinantglycoprotein 120 vaccine to prevent HIV-1 infection. J InfectDis 2005; 191:654–665.

141. Pitisuttithum P, Gilbert P, Gurwith M, Heyward W, Martin M,van Griensven F, et al. Randomized, double-blind, placebo-controlled efficacy trial of a bivalent recombinant glycopro-tein 120 HIV-1 vaccine among injection drug users in Bang-kok, Thailand. J Infect Dis 2006; 194:1661–1671.

142. Rolland M, Tovanabutra S, deCamp AC, Frahm N, Gilbert PB,Sanders-Buell E, et al. Genetic impact of vaccination onbreakthrough HIV-1 sequences from the STEP trial. NatMed 2011; 17:366–371.

143. Kumamoto Y, Mattei LM, Sellers S, Payne GW, Iwasaki A.CD4R T cells support cytotoxic T lymphocyte priming bycontrolling lymph node input. Proc Natl Acad Sci U S A 2011;108:8749–8754.

144. Belyakov IM, Kuznetsov VA, Kelsall B, Klinman D, MoniuszkoM, Lemon M, et al. Impact of vaccine-induced mucosal high-avidity CD8R CTLs in delay of AIDS viral dissemination frommucosa. Blood 2006; 107:3258–3264.

Copyright © Lippincott Williams & Wilkins. Unaut

145. Ranasinghe C, Ramshaw IA. Immunisation route-dependentexpression of IL-4/IL-13 can modulate HIV-specific CD8(R)CTL avidity. Eur J Immunol 2009; 39:1819–1830.

146. Ranasinghe C, Trivedi S, Stambas J, Jackson RJ. UniqueIL-13Ralpha2-based HIV-1 vaccine strategy to enhancemucosal immunity, CD8(R) T-cell avidity and protectiveimmunity. Mucosal Immunol 2013; 6:1068–1080.

147. Lin SW, Cun AS, Harris-McCoy K, Ertl HC. Intramuscularrather than oral administration of replication-defectiveadenoviral vaccine vector induces specific CD8R T cellresponses in the gut. Vaccine 2007; 25:2187–2193.

148. Tatsis N, Lin SW, Harris-McCoy K, Garber DA, Feinberg MB,Ertl HC. Multiple immunizations with adenovirus and MVAvectors improve CD8R T cell functionality and mucosalhoming. Virology 2007; 367:156–167.

149. Gherardi MM, Esteban M. Recombinant poxviruses asmucosal vaccine vectors. J Gen Virol 2005; 86:2925–2936.

150. Mowat AM. Anatomical basis of tolerance and immunityto intestinal antigens. Nat Rev Immunol 2003; 3:331–341.

151. Bolton DL, Song K, Wilson RL, Kozlowski PA, Tomaras GD,Keele BF, et al. Comparison of systemic and mucosal vaccina-tion: impact on intravenous and rectal SIV challenge. MucosalImmunol 2012; 5:41–52.

152. McMichael A, Hanke T. The quest for an AIDS vaccine: is theCD8R T-cell approach feasible? Nat Rev Immunol 2002;2:283–291.

153. Gebril A, Alsaadi M, Acevedo R, Mullen AB, Ferro VA.Optimizing efficacy of mucosal vaccines. Expert Rev Vaccines2012; 11:1139–1155.

154. Dlugonska H, Grzybowski M. Mucosal vaccination –an oldbut still vital strategy. Ann Parasitol 2012; 58:1–8.

155. Holmgren J, Svennerholm AM. Vaccines against mucosalinfections. Curr Opin Immunol 2012; 24:343–353.

156. Jirathitikal V, Bourinbaiar AS. Effect of an oral therapeuticHIV-1 vaccine on AIDS patients with CD4 count above250 cells/mm3. Acta Virol 2004; 48:73–78.

157. Pegu P, Helmus R, Gupta P, Tarwater P, Caruso L, Shen C, et al.Induction of strong anti-HIV cellular immunity by a combina-tion of Clostridium perfringens expressing HIV gag and viruslike particles. Curr HIV Res 2011; 9:613–622.

158. Kajikawa A, Zhang L, Long J, Nordone S, Stoeker L, LaVoy A,et al. Construction and immunological evaluation of dual cellsurface display of HIV-1 gag and Salmonella enterica serovarTyphimurium FliC in Lactobacillus acidophilus for vaccinedelivery. Clin Vaccine Immunol 2012; 19:1374–1381.

159. Doan LX, Li M, Chen C, Yao Q. Virus-like particles as HIV-1vaccines. Rev Med Virol 2005; 15:75–88.

160. Young KR, McBurney SP, Karkhanis LU, Ross TM. Virus-likeparticles: designing an effective AIDS vaccine. Methods 2006;40:98–117.

161. Stanley M, Pinto LA, Trimble C. Human papillomavirus vac-cines – immune responses. Vaccine 2012; 30 (Suppl 5):F83–F87.

162. Landry N, Ward BJ, Trepanier S, Montomoli E, Dargis M,Lapini G, et al. Preclinical and clinical development ofplant-made virus-like particle vaccine against avian H5N1influenza. PLoS One 2010; 5:e15559.

163. Buonaguro L, Tagliamonte M, Visciano ML, Andersen H,Lewis M, Pal R, et al. Immunogenicity of HIV virus-likeparticles in rhesus macaques by intranasal administration.Clin Vaccine Immunol 2012; 19:970–973.

164. Lewis DJ, Huo Z, Barnett S, Kromann I, Giemza R, Galiza E,et al. Transient facial nerve paralysis (Bell’s palsy) followingintranasal delivery of a genetically detoxified mutant ofEscherichia coli heat labile toxin. PLoS One 2009; 4:e6999.

165. Hu K, Luo S, Tong L, Huang X, Jin W, Huang W, et al. CCL19and CCL28 augment mucosal and systemic immune responsesto HIV-1 gp140 by mobilizing responsive immunocytes intosecondary lymph nodes and mucosal tissue. J Immunol 2013;191:1935–1947.

166. Belyakov IM, Derby MA, Ahlers JD, Kelsall BL, Earl P, Moss B,et al. Mucosal immunization with HIV-1 peptide vaccineinduces mucosal and systemic cytotoxic T lymphocytes andprotective immunity in mice against intrarectal recombinantHIV-vaccinia challenge. Proc Natl Acad Sci U S A 1998;95:1709–1714.

horized reproduction of this article is prohibited.

Co

1718 AIDS 2014, Vol 28 No 12

167. Sundling C, Schon K, Morner A, Forsell MN, Wyatt RT,Thorstensson R, et al. CTA1-DD adjuvant promotes strongimmunity against human immunodeficiency virus type 1envelope glycoproteins following mucosal immunization.J General Virol 2008; 89 (Pt 12):2954–2964.

168. Belyakov IM, Ahlers JD, Clements JD, Strober W, Berzofsky JA.Interplay of cytokines and adjuvants in the regulation ofmucosal and systemic HIV-specific CTL. J Immunol 2000;165:6454–6462.

169. Trumpfheller C, Caskey M, Nchinda G, Longhi MP, MizeninaO, Huang Y, et al. The microbial mimic poly IC inducesdurable and protective CD4R T cell immunity together witha dendritic cell targeted vaccine. Proc Natl Acad Sci U S A2008; 105:2574–2579.

170. Buffa V, Klein K, Fischetti L, Shattock RJ. Evaluationof TLR agonists as potential mucosal adjuvants for HIVgp140 and tetanus toxoid in mice. PLoS One 2012; 7:e50529.

171. McKay PF, Cope AV, Mann JF, Joseph S, Esteban M,Tatoud R, et al. Glucopyranosyl lipid A adjuvant significantlyenhances HIV specific T and B cell responses elicited by aDNA-MVA-protein vaccine regimen. PLoS One 2014;9:e84707.

172. Arias MA, Van Roey GA, Tregoning JS, Moutaftsi M, Coler RN,Windish HP, et al. Glucopyranosyl lipid adjuvant (GLA), asynthetic TLR4 agonist, promotes potent systemic and muco-sal responses to intranasal immunization with HIVgp140.PLoS One 2012; 7:e41144.

173. Vassilieva EV, Wang BZ, Vzorov AN, Wang L, Wang YC, BozjaJ, et al. Enhanced mucosal immune responses to HIV virus-likeparticles containing a membrane-anchored adjuvant. mBio2011; 2:e00328-e00310.

174. Jiang JQ, Patrick A, Moss RB, Rosenthal KL. CD8R T-cell-mediated cross-clade protection in the genital tract followingintranasal immunization with inactivated human immunode-ficiency virus antigen plus CpG oligodeoxynucleotides. J Virol2005; 79:393–400.

175. Sui Y, Gagnon S, Dzutsev A, Zhu Q, Yu H, Hogg A, et al. TLRagonists and/or IL-15 adjuvanted mucosal SIV vaccinereduced gut CD4(R) memory T cell loss in SIVmac251-challenged rhesus macaques. Vaccine 2011; 30:59–68.

176. Hutnick NA, Myles DJ, Bian CB, Muthumani K, Weiner DB.Selected approaches for increasing HIV DNA vaccineimmunogenicity in vivo. Curr Opin Virol 2011; 1:233–240.

177. Keating SM, Jacobs ES, Norris PJ. Soluble mediators of inflam-mation in HIV and their implications for therapeutics andvaccine development. Cytokine Growth Factor Rev 2012;23:193–206.

178. Belyakov IM, Hel Z, Kelsall B, Kuznetsov VA, Ahlers JD, NacsaJ, et al. Mucosal AIDS vaccine reduces disease and viral loadin gut reservoir and blood after mucosal infection of maca-ques. Nat Med 2001; 7:1320–1326.

pyright © Lippincott Williams & Wilkins. Unautho

179. Holmgren J, Adamsson J, Anjuere F, Clemens J, Czerkinsky C,Eriksson K, et al. Mucosal adjuvants and antiinfectionand antiimmunopathology vaccines based on cholera toxin,cholera toxin B subunit and CpG DNA. Immunol Lett 2005;97:181–188.

180. Staats HF, Ennis FA Jr. IL-1 is an effective adjuvant for mucosaland systemic immune responses when coadministered withprotein immunogens. J Immunol 1999; 162:6141–6147.

181. Barouch DH, Santra S, Schmitz JE, Kuroda MJ, Fu TM, WagnerW, et al. Control of viremia and prevention of clinical AIDS inrhesus monkeys by cytokine-augmented DNA vaccination.Science 2000; 290:486–492.

182. Stevceva L. Toll-like receptor agonists as adjuvants for HIVvaccines. Curr Med Chem 2011; 18:5079–5082.

183. Harandi AM, Eriksson K, Holmgren J. A protective role oflocally administered immunostimulatory CpG oligodeoxynu-cleotide in a mouse model of genital herpes infection. J Virol2003; 77:953–962.

184. Ichinohe T, Kawaguchi A, Tamura S, Takahashi H, Sawa H,Ninomiya A, et al. Intranasal immunization with H5N1 vac-cine plus Poly I:Poly C12U, a Toll-like receptor agonist,protects mice against homologous and heterologous viruschallenge. Microbes Infect 2007; 9:1333–1340.

185. De Haes W, De Koker S, Pollard C, Atkinson D, Vlieghe E,Hoste J, et al. Polyelectrolyte capsules-containing HIV-1 p24and poly I:C modulate dendritic cells to stimulate HIV-1-specific immune responses. Mol Ther 2010; 18:1408–1416.

186. Trumpfheller C, Longhi MP, Caskey M, Idoyaga J, Bozzacco L,Keler T, et al. Dendritic cell-targeted protein vaccines: a novelapproach to induce T-cell immunity. J Intern Med 2012;271:183–192.

187. San Roman B, De Andres X, Munoz PM, Obregon P, AsensioAC, Garrido V, et al. The extradomain A of fibronectin (EDA)combined with poly(I:C) enhances the immune response toHIV-1 p24 protein and the protection against recombinantListeria monocytogenes-Gag infection in the mouse model.Vaccine 2012; 30:2564–2569.

188. Childers NK, Miller KL, Tong G, Llarena JC, Greenway T,Ulrich JT, et al. Adjuvant activity of monophosphoryl lipid Afor nasal and oral immunization with soluble or liposome-associated antigen. Infect Immun 2000; 68:5509–5516.

189. Martin M, Michalek SM, Katz J. Role of innate immune factorsin the adjuvant activity of monophosphoryl lipid A. InfectImmun 2003; 71:2498–2507.

190. Beck Z, Karasavvas N, Matyas GR, Alving CR. Membrane-specific antibodies induced by liposomes can simultaneouslybind to HIV-1 protein, peptide, and membrane lipid epitopes.J Drug Target 2008; 16:535–542.

191. Matyas GR, Wieczorek L, Beck Z, Ochsenbauer-Jambor C,Kappes JC, Michael NL, et al. Neutralizing antibodies inducedby liposomal HIV-1 glycoprotein 41 peptide simultaneouslybind to both the 2F5 or 4E10 epitope and lipid epitopes. AIDS2009; 23:2069–2077.

rized reproduction of this article is prohibited.