+ All Categories
Home > Documents > A chimeric measles virus with a lentiviral envelope ...in Fig. 1A) was deleted from the vector...

A chimeric measles virus with a lentiviral envelope ...in Fig. 1A) was deleted from the vector...

Date post: 22-Sep-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
9
A chimeric measles virus with a lentiviral envelope replicates exclusively in CD4+/CCR5+ cells Thomas Mourez a, b, c , Mariana Mesel-Lemoine a , Chantal Combredet a , Valérie Najburg a , Nadège Cayet d , Frédéric Tangy a, a Unité de Génomique Virale et Vaccination, CNRS URA 3015, Institut Pasteur, F-75015 Paris, France b APHP, GH Saint-Louis-Lariboisière, Laboratoire de Bactériologie-Virologie, F-75010 Paris, France c Université Paris 7 Denis Diderot, F-75010 Paris, France d Institut Pasteur, Plateforme de Microscopie Ultrastructurale, F-75015 Paris, France abstract article info Article history: Received 21 May 2011 Returned to author for revision 10 June 2011 Accepted 12 August 2011 Available online 3 September 2011 Keywords: Measles Vaccine HIV SIV Chimera We generated a replicating chimeric measles virus in which the hemagglutinin and fusion surface glycoproteins were replaced with the gp160 envelope glycoprotein of simian immunodeciency virus (SIVmac239). Based on a previously cloned live-attenuated Schwarz vaccine strain of measles virus (MV), this chimera was rescued at high titers using reverse genetics in CD4+ target cells. Cytopathic effect consisted in the presence of large cell aggregates evolving to form syncytia, as observed during SIV infection. The morphology of the chimeric virus was identical to that of the parent MV particles. The presence of SIV gp160 as the only envelope protein on chimeric particles surface altered the cell tropism of the new virus from CD46+ to CD4+ cells. Used as an HIV candidate vaccine, this MV/SIVenv chimeric virus would mimic transient HIV-like infection, beneting both from HIV-like tropism and the capacity of MV to replicate in dendritic cells, macrophages and lymphocytes. © 2011 Elsevier Inc. All rights reserved. Introduction Almost thirty years after human immunodeciency virus (HIV) was identied as the causative agent of AIDS, a vaccine capable of preventing the infection remains elusive. In 2003, the rst phase III efcacy trial of an HIV candidate vaccine demonstrated that a monomeric recombinant gp120 (AIDSVax) was unable to elicit detectable neutralizing antibodies or to reduce viral load (Flynn et al., 2005; Pitisuttithum et al., 2006). In 2008, the STEP phase IIb trial of an Ad5-based vaccine challenged the rationale underlying T-cell- based vaccine strategies, as specic T cell responses induced by the vaccine also failed to prevent HIV infection or to reduce viral load (Buchbinder et al., 2008; McElrath et al., 2008). Recently, the RV144 phase III trial conducted in Thailand showed that a prime-boost strategy combining 6 doses of live-recombinant ALVAC-HIV with 2 doses of gp120 B/E envelope in at-risk healthy volunteers reduced the incidence of infection by 31% (Rerks-Ngarm et al., 2009). This very encouraging result demonstrated that both T cells and antibodies are required to prevent HIV infection, and that a prime-boost strategy is necessary to induce both arms of the immune system. However, the complex dosing schedule and weak protection provided by this strategy makes it impractical. An ideal vaccine would induce effective and long-lasting responses after one or two administrations and be amenable to low-cost mass production. To address these challenges, we have previously developed a vector derived from the pediatric live-attenuated Schwarz vaccine strain of measles virus (MV) (Combredet et al., 2003). Different HIV and SIV proteins were stably expressed from MV vectors and strong and durable specic humoral and cellular immune responses were induced in mice (Guerbois et al., 2009; Liniger et al., 2009; Lorin et al., 2004, 2005a; Wang et al., 2001). MV vaccine is a live-attenuated negative-stranded RNA virus proven to be one of the safest and most effective human vaccine products. Mass-produced in many countries and distributed at low cost through the Extended Program on Immunization, the MV vaccine induces life-long immu- nity after one or two injections. We subsequently initiated clinical development of an HIV candidate vaccine based on a recombinant MV encoding an HIV Gag-Pol-Nef antigen. This rst-generation MVHIV vector was designed to induce cellular immunity in adults with preexisting immunity to measles, to evaluate for the rst time a replicating vector as an HIV vaccine, and to prepare the GMP and regulatory logistics for such a vaccine. A phase I clinical trial of this candidate vaccine was recently launched. Measles vaccine provides very effective protection from an infection acquired through the respiratory mucosa. The effectors are both long-lived B cells that produce neutralizing antibodies to the viral glycoproteins, and long-lived CD4 and CD8 T cells. These effectors are rapidly mobilized in the respiratory mucosa on natural Virology 419 (2011) 117125 Corresponding author. Fax: + 33 140613167. E-mail address: [email protected] (F. Tangy). 0042-6822/$ see front matter © 2011 Elsevier Inc. All rights reserved. doi:10.1016/j.virol.2011.08.007 Contents lists available at SciVerse ScienceDirect Virology journal homepage: www.elsevier.com/locate/yviro
Transcript
Page 1: A chimeric measles virus with a lentiviral envelope ...in Fig. 1A) was deleted from the vector plasmid pTM-MV-Schw and replaced by a synthetic chimeric MV–SIV sequence (Fig. 1B).

Virology 419 (2011) 117–125

Contents lists available at SciVerse ScienceDirect

Virology

j ourna l homepage: www.e lsev ie r.com/ locate /yv i ro

A chimeric measles virus with a lentiviral envelope replicates exclusivelyin CD4+/CCR5+ cells

Thomas Mourez a,b,c, Mariana Mesel-Lemoine a, Chantal Combredet a, Valérie Najburg a,Nadège Cayet d, Frédéric Tangy a,⁎a Unité de Génomique Virale et Vaccination, CNRS URA 3015, Institut Pasteur, F-75015 Paris, Franceb APHP, GH Saint-Louis-Lariboisière, Laboratoire de Bactériologie-Virologie, F-75010 Paris, Francec Université Paris 7 Denis Diderot, F-75010 Paris, Franced Institut Pasteur, Plateforme de Microscopie Ultrastructurale, F-75015 Paris, France

⁎ Corresponding author. Fax: +33 140613167.E-mail address: [email protected] (F. Tangy).

0042-6822/$ – see front matter © 2011 Elsevier Inc. Aldoi:10.1016/j.virol.2011.08.007

a b s t r a c t

a r t i c l e i n f o

Article history:Received 21 May 2011Returned to author for revision 10 June 2011Accepted 12 August 2011Available online 3 September 2011

Keywords:MeaslesVaccineHIVSIVChimera

We generated a replicating chimeric measles virus in which the hemagglutinin and fusion surfaceglycoproteins were replaced with the gp160 envelope glycoprotein of simian immunodeficiency virus(SIVmac239). Based on a previously cloned live-attenuated Schwarz vaccine strain of measles virus (MV), thischimera was rescued at high titers using reverse genetics in CD4+ target cells. Cytopathic effect consistedin the presence of large cell aggregates evolving to form syncytia, as observed during SIV infection. Themorphology of the chimeric virus was identical to that of the parent MV particles. The presence of SIV gp160as the only envelope protein on chimeric particles surface altered the cell tropism of the new virus fromCD46+ to CD4+ cells. Used as an HIV candidate vaccine, this MV/SIVenv chimeric virus would mimictransient HIV-like infection, benefiting both from HIV-like tropism and the capacity of MV to replicate indendritic cells, macrophages and lymphocytes.

l rights reserved.

© 2011 Elsevier Inc. All rights reserved.

Introduction

Almost thirty years after human immunodeficiency virus (HIV)was identified as the causative agent of AIDS, a vaccine capable ofpreventing the infection remains elusive. In 2003, the first phase IIIefficacy trial of an HIV candidate vaccine demonstrated that amonomeric recombinant gp120 (AIDSVax) was unable to elicitdetectable neutralizing antibodies or to reduce viral load (Flynn etal., 2005; Pitisuttithum et al., 2006). In 2008, the STEP phase IIb trial ofan Ad5-based vaccine challenged the rationale underlying T-cell-based vaccine strategies, as specific T cell responses induced by thevaccine also failed to prevent HIV infection or to reduce viral load(Buchbinder et al., 2008; McElrath et al., 2008). Recently, the RV144phase III trial conducted in Thailand showed that a prime-booststrategy combining 6 doses of live-recombinant ALVAC-HIV with 2doses of gp120 B/E envelope in at-risk healthy volunteers reduced theincidence of infection by 31% (Rerks-Ngarm et al., 2009). This veryencouraging result demonstrated that both T cells and antibodies arerequired to prevent HIV infection, and that a prime-boost strategy isnecessary to induce both arms of the immune system. However, thecomplex dosing schedule and weak protection provided by thisstrategy makes it impractical.

An ideal vaccine would induce effective and long-lasting responsesafter one or two administrations and be amenable to low-cost massproduction. To address these challenges, we have previouslydeveloped a vector derived from the pediatric live-attenuatedSchwarz vaccine strain of measles virus (MV) (Combredet et al.,2003). Different HIV and SIV proteins were stably expressed from MVvectors and strong and durable specific humoral and cellular immuneresponses were induced in mice (Guerbois et al., 2009; Liniger et al.,2009; Lorin et al., 2004, 2005a; Wang et al., 2001). MV vaccine is alive-attenuated negative-stranded RNA virus proven to be one of thesafest and most effective human vaccine products. Mass-produced inmany countries and distributed at low cost through the ExtendedProgram on Immunization, the MV vaccine induces life-long immu-nity after one or two injections. We subsequently initiated clinicaldevelopment of an HIV candidate vaccine based on a recombinant MVencoding an HIV Gag-Pol-Nef antigen. This first-generation MV–HIVvector was designed to induce cellular immunity in adults withpreexisting immunity to measles, to evaluate for the first time areplicating vector as an HIV vaccine, and to prepare the GMP andregulatory logistics for such a vaccine. A phase I clinical trial of thiscandidate vaccine was recently launched.

Measles vaccine provides very effective protection from aninfection acquired through the respiratory mucosa. The effectors areboth long-lived B cells that produce neutralizing antibodies to theviral glycoproteins, and long-lived CD4 and CD8 T cells. Theseeffectors are rapidly mobilized in the respiratory mucosa on natural

Page 2: A chimeric measles virus with a lentiviral envelope ...in Fig. 1A) was deleted from the vector plasmid pTM-MV-Schw and replaced by a synthetic chimeric MV–SIV sequence (Fig. 1B).

118 T. Mourez et al. / Virology 419 (2011) 117–125

exposure to the virus. Recent progress achieved on the natural courseof MV infection in the macaque model has shown that the first cellsinfected are mucosal dendritic cells (de Swart, 2008; Lemon et al.,2011). Mucosal tissues are also the main portals of entry for HIV-1.After transmission, HIV-1 crosses the mucosal barrier in a few hoursand disseminates locally to draining lymph nodes after a few days(Pope and Haase, 2003). Systemic dissemination then occurs, with ahigh rate of virus replication in blood cells during the followingmonth. Thus, to induce sterilizing immunity to HIV-1, a vaccine mustinduce both neutralizing antibodies and cytolytic T lymphocytes(CTLs) at mucosal surfaces (Letvin, 2006). These effectors must eitherbe present at the time of natural exposure or be raised within a fewhours or days.

Here, to exploit the ability of the live-attenuated MV to inducepotent mucosal immunity to its surface glycoproteins, we developed anew approach based on an MV–HIV chimera rather than arecombinant MV vector. The chimeric virus was obtained by replacingthe MV fusion (F) and hemagglutinin (H) glycoprotein genes by theHIV gp160 coding sequence expressed as an MV transcript in arecombinant MV vector. The resulting virus expresses native HIVgp160 spikes on its surface, instead of MV H/F glycoproteins. It shouldtherefore have the twin advantages of HIV tropism andMV replicationin dendritic cells, macrophages and lymphocytes, thus mimickingtransient HIV infection. It should also exhibit the genetic stability andsafety features of MV vaccine strains that include no recombinationwith other segmented negative-stranded RNA viruses, a cytoplasmicreplicative cycle, no host genome integration, no persistence, and noshedding. Moreover, this chimera should be cleared from the host bythe immune reaction it induces.

With the aim of evaluating its vaccine efficacy in the macaquemodel, we first generated aMV–SIV chimera based on SIVmac239, oneof the most widely used model for AIDS in macaques. We constructedthe chimera by deleting the fusion (F) and hemagglutinin (H)glycoprotein genes from MV Schwarz vaccine strain antigenomiccDNA and replacing themby the SIVMac239 gp160 sequence as anMVtranscript. The chimeric virus was rescued with a reverse geneticssystem and produced at high titers in CEMx174 cells. SIV gp160 wasefficiently expressed on the surface of infected cells and on viral

Fig. 1. Construction of MV-SIVenv-RFP chimeric virus. Schematic representation and genomi(C) The fusion (F) and hemagglutinin (H) glycoproteins of the MVSchw vaccine strain wereIntracytoplasmic tail of the fusion protein (FIC) was conserved and placed in fusion with the g

particles. The new virus exhibited typical CD4 tropism and inducedsyncytia formation by CD4 T cells.

Results

Generation of chimeric MV expressing SIVMac239 gp160 instead of theMV H/F envelope glycoproteins

To improve the capacity of our previous pTM-MVSchw vector toelicit broad-spectrum neutralizing antibodies to HIV by expressingnative trimeric gp160 on the viral surface, we designed a new MVchimeric vector in which the MV fusion (F) and hemagglutinin (H)envelope glycoproteins genes were deleted and replaced by the gp160sequence expressed from an MV transcript. As proof-of-concept, andwith the aim of evaluating vaccine efficacy in the macaque model, wegenerated an MV–SIV chimeric virus based on SIVmac239 gp160(Fig. 1). To promote efficient expression of SIV gp160 glycoprotein atthe surface of chimeric viral particles, we replaced the intracytoplas-mic domain of gp160 with that of the MV fusion protein, thuspermitting cell membrane anchoring through interactions with theMV matrix protein, as previously demonstrated (Foley et al., 2002;Spielhofer et al., 1998). To visualize the chimeric virus in infected cells,we inserted the RFP Cherry protein sequence as an additionaltranscription unit, upstream of the MV-M matrix protein. Thisadditional transcription unit may be used in future to insert othergenes in place of RFP.

To construct the chimeric cDNA, the genomic region encompassingtheMV fusion (F) and hemagglutinin (H) genes (NarI to SpeI fragmentin Fig. 1A) was deleted from the vector plasmid pTM-MV-Schw andreplaced by a synthetic chimeric MV–SIV sequence (Fig. 1B). Thischimeric sequence (Fig. 1C) was designed to preserve the completepromoter and intergenic sequence of the MV-F gene upstream of theSIV gp160 initiation codon, such that the gp160 gene is expressed asan MV transcript. The whole gp120 and the gp41 ectodomain with itstransmembrane sequence deleted of the intracytoplasmic domainwere fused to the MV-F intracytoplasmic sequence, followed by theMV H/L intergenic region. This synthetic sequence was thenintroduced into the pTM-MVSchw-RFP-STREPtag plasmid, and the

c organization of MV (A) and chimeric MV-SIVenv (B) (N, P, M, F, H, L are MV proteins).replaced by the extracellular and transmembrane (TM) part of the gp160 of SIVmac239.p41 in order to anchor the glycoprotein with the Mmatrix protein of the measles virus.

Page 3: A chimeric measles virus with a lentiviral envelope ...in Fig. 1A) was deleted from the vector plasmid pTM-MV-Schw and replaced by a synthetic chimeric MV–SIV sequence (Fig. 1B).

Fig. 2. Rescue and culture of MV/SIVenv-RFP chimeric virus. (A) To rescue the chimericvirus, the plasmid pTM-MVSchw-Chim-SIVenv (Cherry) was transfected in HEK-293Thelper cells that were subsequently co-cultured with MT4 or CEMx174 cells. After 3 to5 days of co-culture, cytopathic effect occurred showing syncytia bubbling from cellaggregates and expressing the cherry protein. (B) CEMx174 cells were infected by cell-free MV/SIVenv chimeric virus at MOI=0.1. Typical cell aggregates and bubblingsyncytia were observed at 3 days post-infection.

119T. Mourez et al. / Virology 419 (2011) 117–125

resulting vector, designated pTM-MVSchw-Chim-SIVenv (Cherry),was fully sequenced and shown to respect the ‘rule of 6’ of the MVgenome (Calain and Roux, 1993).

MV rescue from cDNA is usually achieved by coculturingtransfected helper cells with MV target cells that express at leastone of the MV receptors (SLAM or CD46) (Vero, MRC5, CEF, B95a)(Dorig et al., 1993; Naniche et al., 1993; Tatsuo et al., 2000). Giventhe expected tropism change of the MV/SIV chimeric virus, werescued it from the pTM-MVSchw-Chim-SIVenv (Cherry) plasmid byusing the standard protocol, except that helper cells were coculturedwith SIV CD4+ target cells (MT4, CEMx174). Rescue of the chimericvirus was only successful when this modified protocol was used.Monitoring was facilitated by the presence of fluorescent cells after3 to 5 days of co-culture (Fig. 2A). A cytopathic effect (CPE) onCEMx174 cells appeared soon after fluorescence was first detected.The new chimeric virus harvested from infected cells was designat-ed MV/SIVenv. Other cells, such as MT4 and HeLa P4C5 cells, werealso successfully used to rescue the MV/SIVenv chimeric virus, witha comparable time course.

Once rescued, the chimeric virus can be amplified for weeks onCEMx174 or MT4 cells, simply by adding fresh uninfected cells tothe culture and replacing the medium, as for lentivirus amplifica-tion. Using the same procedure as for measles virus purification,cell-free virus was collected by freezing and thawing infected cellswith medium then clarification. New infection of other cells wascarried out with clarified cell-free virus. T cells infected by the MV/SIVenv chimeric virus exhibited characteristic features of lentiviralinfection with large aggregates of infected cells evolving to formsyncytia (Fig. 2B). Although the MVSchw strain is able to grow atlow titers on CD4 T cells through the use of the SLAM receptor(Dhiman et al., 2004; Yanagi et al., 2006), the chimeric MV/SIVenvvirus produced 3 log higher titers than control MVSchw onCEMx174 cells (Fig. 3A). When comparing the rate of replicationof SIV to the MV/SIVenv chimeric virus by FACS analysis (Fig. 3B),we observed that the chimeric virus replicated faster with themaximum virus yield achieved at 2–3 days post-infection, while asexpected, SIVmac251 replicated more slowly and required 8–10 days to achieve 100% of infection. Replication kinetic analysis

showed that the MV/SIVenv chimeric virus achieved titers of up to106 TCID50/ml within 48–60 h (Fig. 3C), at a similar rate than MVitself on Vero cells (Fig. 3D). Beyond 60 h the virus titer fell andmost of the cells die after few days. If additional fresh cells areadded, the infection can last several weeks. To determine the bestcell line for virus stock production and titration, we compared theviral titers obtained with CEMx174 and MT4 cells. CEMx174 cellsreproducibly produced 1 log higher virus titers than MT4 cells.CEMx174 cells are B-T hybrid lymphoblastic cells (Salter et al.,1985), a characteristic that might promote MV/SIVenv chimericvirus replication. The virus might also benefit from both the T cellentry receptor used by SIVenv glycoproteins and from the efficientreplication of MV in B lymphocytes (Kobune et al., 1990).

The MV/SIVenv chimeric virus expresses SIV gp160

To assess the expression of SIV gp160 glycoprotein by thechimeric virus, CEMx174 cells were infected with MV/SIVenv andlysed after 48 h. Cell extracts were separated by SDS-PAGEelectrophoresis and analyzed by Western blotting (Fig. 4). Lysatesof uninfected CEMx174 cells, Vero cells infected by the parentalMVSchw virus expressing the Cherry protein, and CEMx174 cellsinfected by SIV Mac239 were used as controls. Both the parentalMVSchw and chimeric MV/SIVenv viruses expressed the MVnucleoprotein N in infected cells. The SIV gp160 and its maturegp120 form were detected with the expected molecular size incells infected with MV/SIVenv chimeric virus, demonstrating thatthe MV-F promoter controls efficiently the expression of SIVglycoproteins. MV/SIVenv chimeric virus expressed a lower levelof SIV glycoproteins than SIVmac251. However, this level iscompatible with the efficient budding and spreading capacity ofthe chimeric virus, as shown in Figs. 2B and 3C.

Expression of the SIV gp160 and MV-N proteins was alsodemonstrated by immunofluorescence analysis with specific mono-clonal antibodies (Fig. 5). Both MV-N and SIV gp160 were expressedin infected cells, simultaneously with RFP cherry protein. SIV gp160was expressed on the surface of infected cells, as demonstrated bypositive surface staining of non-permeabilized cells. Large syncytia,similar to those observed with HeLa cells infected by standardMVSchw, were particularly prominent with HeLA P4C5 cells thatexpress the CD4, CXCR4 and CCR5 receptors on their surface. Thisdemonstrates that SIV gp160 expression by the chimeric virus inadherent fibroblastic-like cells expressing SIV receptors mediatesMV-like fusion.

The morphology of MV/SIVenv chimeric particles is similar to that of MVparticles

To determine whether MV-F and H glycoprotein replacement bySIV gp160 alters the morphological structure of chimeric particlescompared to parental MV, we examined infected cells by transmissionelectron microscopy. Hela P4C5 cells were infected with MV-Schw orMV/SIVenv, then fixed and stained with uranyl acetate and Reynoldslead citrate 22 h later, before electron microscopy (Fig. 6). In bothcases we observed numerous viral particles either budding or trappedin the extracytoplasmic space between infected cells, with a structureand size (150–350 nm) typical of measles virus. Moreover, a fringe ofshort spikes was observed on the surface of both MV and chimericparticles (Figs. 6C and D), corresponding to the MV and SIVglycoproteins, respectively. These observations showed that MV Fand H replacement by SIV gp160 did not affect the morphology or sizeof the resulting viral particles.

Immunoelectron microscopy was used to demonstrate thepresence of SIV gp160 anchored on the surface of the virusmembrane.Infected cells were immunostained with a specific anti-SIV gp160/120antibody before electron microscopy (Fig. 6E). The chimeric viral

Page 4: A chimeric measles virus with a lentiviral envelope ...in Fig. 1A) was deleted from the vector plasmid pTM-MV-Schw and replaced by a synthetic chimeric MV–SIV sequence (Fig. 1B).

Fig. 3. Replication kinetics of MV/SIVenv chimeric virus in CEMX174 T-cells. (A) CEMX174 cells were infected at MOI=0.1 by MVSchw or MV/SIVenv chimeric virus both of themexpressing the cherry protein. The cells were harvested 48 h post-infection and cell-associated virus titers were determined using the TCID50 method. All growth curves wereperformed in triplicate. (B) Propagation rate of MV/SIVenv chimeric virus compared to SIV. One million cells were infected in 6-well plates at a MOI=0.1 with MV/SIVenv chimericvirus (open diamonds) or SIVmac251 (black squares). The percentage of infected cells was determined at each time point by flow cytometry using either the Cherry proteinfluorescence or an intracellular SIVgag staining. (C) Growth kinetics of MV/SIVenv chimeric virus in CEMX174 T-cells. 400,000 cells were infected in a 12-well plate at a MOI=0.1with MV/SIVenv chimeric virus. Cells were collected at 16 h, 24 h, 38 h, 47 h, 71 h, and 96 h post-infection. TCID50 was determined in CEMX174 cells by the Spearman and Kärbermethod. (D) Characteristic growth kinetics of MVSchw virus in Vero cells.

120 T. Mourez et al. / Virology 419 (2011) 117–125

particles were surrounded by 10-nm gold particles, demonstratingthe anchoring of SIV gp160 to their surface. Gold particles were alsoobserved on the surface of budding viruses. In contrast, no goldparticles were observed on the surface of MVSchw particles in controlcells. We have previously observed that HIV gp160 expressed fromrecombinant MV, as an additional gene, is not incorporated on MVparticles (Guerbois et al., 2009). The present observation validates ourchoice to anchor SIVgp160 with the intracytoplasmic tail of MV-Fprotein, as supported by previous studies (Foley et al., 2002; Rouxel etal., 2009; Spielhofer et al., 1998).

Fig. 4. Expression of MV-N and SIV gp160 in MV/SIVenv infected cells. Expression ofMV-Nucleoprotein (A) and SIV gp160 (B) was detected by Western blotting using celllysates from CEMx174 cells infected by MV-SIVenv chimeric virus at MOI=0.1 andharvested 48 h post-infection. Cell lysates from Vero cells infected by MVSchw andfrom CEMx174 cells infected by SIVMac239 were used as positive controls. Both gp160and gp120 are detected indicating a correct cleavage of the gp160.

The MV–SIVenv chimeric virus acquired HIV/SIV tropism

The aim of constructing a chimeric replicating MV expressingnative SIV gp160 spikes on its surface was to obtain a virus targetingCD4+/CCR5+ cells, in the same way as HIV/SIV. Used as an HIVcandidate vaccine, this virus would exhibit both HIV tropism andreplicate in dendritic cells, macrophages and lymphocytes, thusmimicking transient HIV-like infection. To analyze the effect of theexchange of surface glycoproteins on viral tropism, we cultured theMV/SIVenv virus in different cell lines supporting the replication ofHIV/SIV or MV. Attenuated MV vaccine strains are known to replicateinefficiently in CD4 T cells, which are the main targets of HIV and SIV.We thus compared the capacity of the MV/SIVenv chimeric virus toinfect CD4+ cells, and fibroblastic cell lines that do not express CD4receptors. Cells were infected at an MOI of 0.1, and infection wasdetected by the presence of CPE and red fluorescence. The MV/SIVenvchimeric virus replicated in all the CD4+/CCR5+ cells tested, namelyCEMx174, MT4, HeLa P4C5, primary CD4+ human lymphocytes, andprimary macaque splenocytes (Table 1). Jurkat cells, which expressonly CXCR4 co-receptor, were poorly infected, confirming the tropismof SIVmac239 for CCR5. In contrast, CD4-negative cell lines such asVero, MRC5, HeLa and U937 were unable to support MV/SIVenvinfection. To confirm that the chimeric virus has lost its original CD46tropism, we infected HeLa and Vero cells, which are typical CD46+target cells for MV vaccine strains, with the parental MVSchw(Cherry) andMV/SIVenv (Cherry). We observed typical large syncytiawith red fluorescence in HeLa and Vero cells infected by MVSchw,

Page 5: A chimeric measles virus with a lentiviral envelope ...in Fig. 1A) was deleted from the vector plasmid pTM-MV-Schw and replaced by a synthetic chimeric MV–SIV sequence (Fig. 1B).

Fig. 5. Expression of SIV gp160 glycoprotein on the surface andMV-Nucleoprotein in the cytoplasm of cells infected with MV/SIVenv chimeric virus. Hela P4C5 cells were infected byMV/SIVenv virus at MOI=0.1 then fixed 24 h post-infection with PFA 4%. (A) Cell nuclei are shown in Dapi coloration. (B) The expression of MV nucleoprotein N was detected byimmunofluorescence using a FITC-labeled mouse anti-MV-Nmonoclonal antibody; the expression of SIV gp160 was detected using a mouse anti-SIV gp41 monoclonal antibody anda FITC anti-mouse IgG as secondary antibody. (C) The expression of the cherry protein is in red. (D) Merge.

121T. Mourez et al. / Virology 419 (2011) 117–125

while no signs of infection were detected in the same cells infectedwith MV/SIVenv chimeric virus (Fig. 7). In contrast, HeLa P4C5 cellswere infected with the chimera (Fig. 7). Overall, these resultsdemonstrate that this chimeric MV has acquired the capacity to infectCD4+/CCR5+ cells, as SIV/HIV, and has lost his original tropism fornon-CD4+ fibroblastic cells.

Discussion

We describe in this report the design, production and character-ization of a replicating MV/SIV chimeric virus constructed byreplacing the MV H and F surface glycoproteins with the SIV gp160envelope glycoprotein. This chimeric virus was generated from apreviously cloned live-attenuated Schwarz vaccine strain of measles

Fig. 6. Electron microscopy observation of MV/SIVenv chimeric virus. HeLa P4C5 cells were infor 22 h. Cells were fixed and analyzed by transmission electron microscopy at 80 kV. Shown×20000) and isolated particles (C, D magnification, ×80000). (E) Electron microscopy obsegp160 immunostaining with 10-nanometer colloidal gold particles was performed, showin

virus (Combredet et al., 2003). The SIV gp160 sequence was cloneddownstream of the MV-F gene promoter and was followed by the MVH/L intergenic region, such that it is expressed as an MV transcript.The transmembrane domain of SIV gp41 was preserved in order topromote trimeric gp160 assembly, and the gp41 intracytoplasmicdomain was replaced by the MV-F intracytoplasmic sequence in orderto favor anchoring of the chimeric gp160 on MV particles. Thepresence of an additional transcription unit within the chimeric virusgenome allows the expression of additional genes. We readily rescuedthe chimeric virus by coculturing transfected helper cells with CD4+target cells. In infected CEMx174 cells, a cytopathic effect is reflectedby the presence of large cell aggregates evolving to form syncytia, asobserved during SIV infection. High viral titers were obtained after 2–3 days of culture. The chimeric virus replicated at the same rate than

fected at anMOI of 0.1 with MV-SIVenv chimera (A, C, E) or MV-Schw (B, D) as a controlare typical viral particles observed trapped between infected cells (A, B magnification

rvation of MV-SIVenv chimeric virus after immunogold labeling of SIV gp160. Anti-SIVg the presence of gold particles on the surface of viral particles. Bars indicate the scale.

Page 6: A chimeric measles virus with a lentiviral envelope ...in Fig. 1A) was deleted from the vector plasmid pTM-MV-Schw and replaced by a synthetic chimeric MV–SIV sequence (Fig. 1B).

Table 1Tropism of MV/SIVenv chimeric virus.

Cells Organism CD4 Infectivitya

CEMx174 Homo sapiens + +++HeLa P4C5 Homo sapiens + +++MT4 Homo sapiens + ++Jurkat Homo sapiens + +CD4+ Lymphocytes Homo sapiens + +++Splenocytes Maccaca cynomolgus + +HeLa Homo sapiens − −MRC5 Homo sapiens − −Vero Cercopithecus aethiops (AGM) − −U937 Homo sapiens − −a Symbols: (−) no infection; (+) b10% infection; (++) 25–75% infection; (+++)

N75% infection.

122 T. Mourez et al. / Virology 419 (2011) 117–125

MV, which is much faster than SIV. This capacity might be anadvantage for its immunogenic properties in vivo. The morphology ofthe chimeric virus was identical to that of the parent MV particles,indicating that MV particle morphology is not shaped by envelopeproteins but rather by the matrix M protein (Spielhofer et al., 1998).We detected SIV gp160 expression by the chimeric virus in cell lysatesand on the surface of infected cells. The presence of gp160 anchoredon the surface of viral particles was observed by immunoelectronmicroscopy. As expected from previous studies (Foley et al., 2002;

Fig. 7. MV/SIVenv chimeric virus has lost CD46 tropism and acquired CD4/CCR5tropism. HeLa and Vero cells, which are CD46+ target cells of MV vaccine strains, areinfected with standard MV-cherry, but not with MV/SIVenv-cherry (A, B, C, D, E and F).In contrast, HeLa P4C5, which are knock-in HeLa cells expressing CD4/CCR5 receptors,are infectedbyMV/SIVenv-cherry, showing a cythopatic effect and redfluorescence (G,H).

Rouxel et al., 2009; Spielhofer et al., 1998), successful incorporation ofa foreign envelope protein on the surface of measles virus, as on rabiesvirus, depends strictly on the presence of the original intracytoplasmicdomain interacting efficiently with the matrix M protein. Whenexpressed with its own gp41 intracytoplasmic domain, HIV-1 gp160was not incorporated on MV particles (Guerbois et al., 2009). Specificsequences of HIV/SIV Env cytoplasmic tail have been shown to beresponsible for slowing and reducing its cell and viral surfaceexpression (Abrahamyan et al., 2005; Bultmann et al., 2001).Replacing the cytoplasmic tail by that of MV-F protein likely abolishedthis property. Therefore, Env incorporation into virions might beenhanced. However, enumerating the number of Env trimers on thesurface of virions is difficult and requires the use of complextechniques such as cryo-electron microscopy and specific antibodiesof very high affinity that are poorly available for SIV. The antibodiesthat we used in this work did not allow enumerating accurately thenumber of Env trimers on the surface of chimeric virus. However,whatever the number of biologically active Env trimers on its surface,the infectivity of the chimeric virus was improved as compared to SIVbecause its replicative rate was accelerated as shown in Fig. 3C.

The presence of SIV gp160 as the only envelope protein on thesurface of MV/SIVenv chimeric particles altered the cell tropism of thenew virus from CD46+ epithelial cells to CD4+/CCR5+ cells (bothcell lines and primary human and macaque cells). In contrast, CD4-negative cell lines such as Vero, MRC5 and U937, currently used toreplicate MV, did not support MV/SIVenv infection, confirming thatthe chimeric virus had lost its original CD46 tropism. MV/SIVenvchimeric particles incorporate gp160 on their surface, and theirbudding progeny exhibit efficient fusion on CD4+/CCR5+ cells,indirectly demonstrating the functional trimeric configuration ofgp160 that is required to elicit neutralizing antibodies (Mascola andMontefiori, 2010). This efficient fusion capacity of chimeric gp160 isdue to the capacity of the SIV gp41 transmembrane domain and theMV-F intracytoplasmic domain to assemble the envelope protein intrimeric form.

Used as an HIV candidate vaccine, the MV/SIVenv chimeric viruswould mimic transient HIV-like infection, benefiting both from HIV-like tropism and the capacity of MV to replicate in dendritic cells,macrophages and lymphocytes. This might be expected to confer alevel of protection similar to that provided by immunizationwith live-attenuated SIV, an approach that cannot be transposed to humans.HIV-1 mainly enters the body through mucosal sites during sexualintercourse. Thus, the presence of neutralizing antibodies and efficientcell-mediated immunity at these sites is crucial for preventing viralentry and early steps of dissemination (Montefiori and Mascola,2009). Indeed, so-called elite HIV-1 controllers, who have persistentlylow viral load in the absence of therapy, have significantly strongerand more complex mucosal immune responses than antiretroviral-treated patients (Ferre et al., 2009). It was recently reported thatmucosal antibodies induced by efficient immunization fully protectprimates from vaginal challenge (Bomsel et al., 2011).

In the Call for replicating vector prime-protein boost strategies in HIVvaccine design (Malkevitch and Robert-Guroff, 2004), it was elegantlydemonstrated that replicating vectors expressing an optimallydesigned envelope have numerous advantages for the induction ofprotective mucosal immunity. The capacity of live-attenuated MVvaccines to induce strong and durable protective immunity mediatedby CD4/CD8 T cells and persistent neutralizing antibodies to the viralenvelope is probably related to a very specific gene signature, asdemonstrated for the yellow fever vaccine YF-17D (Pulendran, 2009;Querec et al., 2009). Because such signatures result from the stressinduced by the replicating vaccine virus, our MV/SIVenv chimeramight benefit, as a candidate vaccine, from the capacity of MV toinduce very potent mucosal protection mediated by neutralizingantibodies to surface glycoproteins. Administered to subjects withpre-existing immunity to MV, this chimeric virus would escape MV-

Page 7: A chimeric measles virus with a lentiviral envelope ...in Fig. 1A) was deleted from the vector plasmid pTM-MV-Schw and replaced by a synthetic chimeric MV–SIV sequence (Fig. 1B).

123T. Mourez et al. / Virology 419 (2011) 117–125

neutralizing antibodies. It could also be used as a booster followingadministration of a recombinant MV. Live, non-persistent chimericviruses based on the safest human live attenuated vaccine have notpreviously been evaluated as HIV candidate vaccines, although similarchimeric viruses are already developed for immunization againstdengue and other arboviruses. Based on intertypic envelope exchangewithin the yellow-fever 17D vaccine backbone, this Chimerivax®technology shows promising clinical results (Guirakhoo et al., 2004;Guy et al., 2010).

There are currently no suitable mousemodels in which to evaluatethe immunogenicity and preventive efficacy of the MV/SIVenvcandidate vaccine. Genetically modified CD46-IFNAR mice thatexpress CD46 (the human receptor for MV vaccine strains) and lackthe INF-α/β receptor are susceptible to MV infection (Mrkic et al.,1998, 1999) and have been used as a model to evaluate theimmunogenicity of MV recombinants (Combredet et al., 2003;Desprès et al., 2005; Guerbois et al., 2009; Lorin et al., 2004, 2005b;Singh et al., 1999). However, these mice could not sustain thereplication of ourMV/SIVenv chimeric virus, which has lost its originaltropism.Mice expressing SLAM, the human receptor for wild-typeMVstrains (Sellin et al., 2006; Welstead et al., 2005), are also unsuitable.We will therefore evaluate the protective capacity of MV/SIVenv inthe macaque model. With its Cherry protein marker, this virus willenable the cell types supporting gp160-dependent infection to beidentified in vivo after intravenous or mucosal administration. Thehumoral and cell-mediated immunity elicited will be evaluated, andthe capacity of different vaccination schedules to protect fromautologous SIVMAC251 challenge will be assessed.

Materials and methods

Cells and tropism assay

Stable HEK-293-T7-NP human embryonic kidney cells generatedby transduction with recombinant lentiviral vectors and cloning(Tangy et al., 2006) were used for viral rescue. They were maintainedin Dulbecco's minimal essential medium (DMEM) supplemented with10% fetal calf serum. CEMx174 hybrid lymphoblastic cells weremaintained in RPMI medium supplemented with 10% fetal calf serum(Salter et al., 1985). P4C5 cells (HeLa, CD4+, CXCR4+, CCR5+,HIVLTR–LacZ+) (Marechal et al., 1998) were cultured in DMEM 10%FCS supplemented with 0.5 mg/ml G418 (Geniticin; Gibco). Fortropism assays, primary human CD4+ T lymphocytes, primaryMaccaca cynomolgus splenocytes, and the MT4, Jurkat, and U937human cell lines were cultured in RPMI supplemented with 10% fetalcalf serum.MRC5 and Vero cells weremaintained in DMEM-Glutamax(Gibco-BRL) supplemented with 10% fetal calf serum.

Plasmid construction

The chimeric plasmid was constructed from the pTM-MVSchw-eGFP plasmid (Fig. 1A), which contains an infectious MV cDNAcorresponding to the antigenome of the Schwarz MV vaccinestrain and expresses the eGFP protein in an additional transcrip-tion unit (Combredet et al., 2003). The eGFP sequence wasreplaced by the sequence coding for RFP-STREPtag protein (Cherryred fluorescent protein) between the BsiW1 and BssHII sites. Thistag protein was chosen because it is smaller and more sensitivethan eGFP. To replace the MV envelope glycoprotein genes bythose of SIV, the fragment between the two SpeI sites flanking theF and H genes in pTM-MVSchw-eGFP was removed and subclonedinto a pBS vector, designated pBS-Schw-Spe5803. A chimeric cDNAwas then synthesized (Genecust), containing successively, fromNarI to SpeI: (i) the MV M/F intergenic region, (ii) the SIVmac239gp160 sequence, in which the intracytoplasmic domain wasreplaced by that of MV-F, and (iii) the MV H/L intergenic region

(Fig. 1C). The SIVgp160 sequence was optimized to Macacca codonusage and modified to remove a SpeI restriction site and to avoidthe following cis-active elements: internal TATA-boxes, chi-sites,ribosomal entry sites, AT- or GC-rich sequences, ARE, INS and CRSsequence elements, repeated sequences, RNA secondary struc-tures, cryptic splice donor and acceptor sites, and branch points.This optimization also avoided MV editing-like sequences. Thewhole construct respected the ‘rule of 6’ of the MV genome (Calainand Roux, 1993). A BamHI site was added to the 3′ end of thesynthetic sequence to enable introduction of the cassette into thepBS-Schw-Spe5803 plasmid. To replace the MV H and F sequencesby the modified SIVenv gene, the chimeric synthetic sequence wasrecombined into the pBS-Schw-Spe5803 plasmid after NarI/BamHIdigestion. This plasmid was designated pBS-Schw-chim/siv/env.The new modified sequence was then introduced into the pTM-MVSchw-RFP-STREPtag vector after SpeI digestion. The resultingvector was designated pTM-MVSchw-Chim-SIVenv (Cherry).

Rescue of the MV/SIV chimera

The chimeric MV/SIV virus was rescued from the pTM-MVSchw-Chim-SIVenv (Cherry) plasmid as previously described (Combredet etal., 2003), by using a helper-cell-based rescue system (Radecke et al.,1995) (Parks et al., 1999), modified to recover the virus in CD4 cells.Briefly, HEK-293-T7-MV human embryonic kidney helper cells(Guerbois et al., 2009; Tangy et al., 2006) were transfected with thepTM-MVSchw-Chim-SIVenv (Cherry) plasmid and then co-culturedwith CEMx174 cells for 3 days. Positive rescue was shown by acytopathic effect (CPE) and red fluorescence generated by the Cherryprotein. CPE and red fluorescence generally appeared after 3–5 days ofco-culture. To amplify the virus, fresh CEMx174 cells were addedtwice a week and culture was continued until almost all the cells wereinfected. To harvest the virus, cells were centrifuged and freeze-thawed. After clarification, cell lysates were titrated and used as aninoculum for subsequent virus production.

Virus titration and growth kinetics

Titers of the chimeric virus were determined by using anendpoint dilution assay on CEMx174 cells. Briefly, CEMx174 cellswere infected for 2 h under low-speed centrifugation (1000 rpm)in sterile Eppendorff tubes with serial 1:10 dilutions of virussuspension in RPMI supplemented with 10% FCS. Infected cellswere seeded in 96-well plates (25000 cells in 100 μL of RPMI/well)and incubated for 24 h at 37 °C. After 24 h, 100 μL of RPMI-10% FCSwas added to each well and the cells were incubated for 4 days.The cells were observed daily for CPE and red fluorescence. Fifty-percent tissue culture infective doses (TCID50) were calculatedwith the Spearman and Kärber method. To determine thereplicative capacity of the chimeric virus, CEMx174 cells wereinfected with viral suspension for 2 h at a multiplicity of infection(MOI) of 0.1, under low-speed centrifugation (1000 rpm). InfectedCEMx174 cells were seeded in 12-well plates at 400000 cells/well.At various times post-infection, cells were harvested and centri-fuged at 1000 rpm. After a round of freeze thawing and removal ofcell debris by low-speed centrifugation, virus titers were deter-mined by limiting dilution assay on CEMx174 cells as describedabove. To compare the growth capacity of SIV to the chimericvirus, six-well plates were seeded with 106 CEMx174 cells andinfected at a MOI 0.1 by either MV/SIVenv virus or SIVmac251. Atdifferent time points, the percentage of infected cells wasdetermined by FACS analysis. Cells were washed twice with PBSand 2% FCS, and then fixed in PBS containing 4% paraformaldehyde(v/v). Cells infected by the MV/SIVenv chimeric virus weredetected with the Cherry protein fluorescence. Cells infectedwith SIV were fixed then permeabilized with PermWash buffer

Page 8: A chimeric measles virus with a lentiviral envelope ...in Fig. 1A) was deleted from the vector plasmid pTM-MV-Schw and replaced by a synthetic chimeric MV–SIV sequence (Fig. 1B).

124 T. Mourez et al. / Virology 419 (2011) 117–125

(BD Bioscience), incubated with an anti SIVgag KK64 mouseantibody (obtained through the NIBSC Centralized Facility for AIDSReagents) at 4 °C for 20 min, then with an Alexa Fluor® 488 goatanti-mouse IgG (H+L) as secondary antibody. After washing inPBS flow cytometry analysis was performed using a FACScalibur(Becton Dickinson).

Immunofluorescence detection

To detect the expression of MV-N and SIV-env proteins, immuno-fluorescence staining was performed on infected HeLa P4C5 cells fixedwith 4% PFA solution (v/v). Surface SIV-env immunostaining wasperformed after incubating the cells for 1 h in blocking solution (PBS 2%goat serum), with a 1/300-diluted mouse anti-SIVMac251gp160/gp120monoclonal antibody (KK17; obtained through the AIDS Research andReference Reagent Program, Division of AIDS, NIAID, NIH, fromDrKarenKent and Ms Caroline Powell; courtesy of the NIBSC Centralized Facilityfor AIDS Reagents). Intracytoplasmic immunostaining was performedafter incubating the cells for 1 h in blocking/permeabilizing solution(PBS, 2% goat serum, 0.1% saponin), with a mouse anti-MV-N mAb(MAB8906F; Chemicon International). In both cases the secondaryantibodywas a FITC-conjugated sheepanti-mouse IgG antibody (74641,BioRad).After2washing steps inPBS, cell nucleiwere stainedwithDAPI.

Western blotting

CEMx174 cells were infectedwith theMV-SIVenv chimeric virus at anMOIof0.1. Infectedcellswere collected48 h laterand lysedwithcompleteEDTA-free protease inhibitor (Roche) in 120 mM NaCl solution. Proteinlysates were separated (50 ng per well) by SDS-PAGE electrophoresis on4–12% NUPAGE® Bis-Tris gels with MOPS running buffer (Invitrogen)before transfer to cellulose membranes with the XCell SureLock™ system(Invitrogen). To detect the SIVgp160 andMV-N proteins, themembraneswereblottedwith amouse anti-SIVgp160/gp120V2monoclonal antibody(KK13; obtained from the Programme EVA Centre for AIDS Reagents,NIBSC, UK, supported by the EC FP6/7 Europrise Network of Excellence,AVIP and NGIN consortia and the Bill and Melinda Gates GHRC-CAVDProject and donated by Dr Karen Kent, NIBSC Centralized Facility for AIDSReagents), or a mouse anti-MV N mAb (MAB8906F; Chemicon Interna-tional). A sheep anti-mouse IgG-horseradish peroxidase (HRP) conjugate(NA931V; GE Healthcare) was used as secondary antibody. Peroxidaseactivity was visualized with an enhanced chemiluminescence detectionkit (Pierce).

Electron microscopy

HeLa P4C5 cells were seeded on glass coverslips and infected at anMOI of 0.1 with the MV–SIVenv chimera. The cells were fixed 22 hlater with 2% PFA in PBS for 5 min and with 4% PFA for 2 h at roomtemperature. Non-specific sites were saturated for 10 min with 1%bovine serum albumin, and the cells were probed for 40 min with themouse anti-SIVMac251gp160/gp120 monoclonal antibody KK17(NIH-AIDS Research and Reference Reagent Program) in PBS–1%BSA. After washing, the cells were incubated for 20 min with 10-nmcolloidal gold particles coated with protein A (University MedicalCenter, Utrecht). The cells were then fixed in 2.5% glutaraldehyde in0.1 M sodium cacodylate buffer overnight at 4 °C, postfixed with 1%osmic acid, dehydrated in ethanol, and embedded in Epon. Ultrathinsections (70–80 nm) were cut with a diamond knife, stained withuranyl acetate and Reynolds lead citrate, and observed at 80 kVaccelerating voltage in a JEOL JEM 1010 electronmicroscope equippedwith an Eloïse Mega View III camera. Non-infected cells and HeLaP4C5 cells infected with MV-Schw were used as controls.

Acknowledgments

We thank Denise Guetard (Institut Pasteur) for providing us withSIV and CEMx174 cells and for her very helpful advice. TM wassupported by APHP and by Paris VII Denis Diderot University. Thiswork was supported by Institut Pasteur and EC funds (Europrise, LSHPCT-2006-037611).

References

Abrahamyan, L.G., Mkrtchyan, S.R., Binley, J., Lu, M., Melikyan, G.B., Cohen, F.S., 2005.The cytoplasmic tail slows the folding of human immunodeficiency virus type 1 Envfrom a late prebundle configuration into the six-helix bundle. J. Virol. 79 (1),106–115.

Bomsel, M., Tudor, D., Drillet, A.S., Alfsen, A., Ganor, Y., Roger, M.G., Mouz, N., Amacker, M.,Chalifour, A., Diomede, L., Devillier, G., Cong, Z., Wei, Q., Gao, H., Qin, C., Yang, G.B.,Zurbriggen, R., Lopalco, L., Fleury, S., 2011. Immunization with HIV-1 gp41 subunitvirosomes inducesmucosal antibodiesprotecting nonhumanprimates against vaginalSHIV challenges. Immunity 34 (2), 269–280.

Buchbinder, S.P., Mehrotra, D.V., Duerr, A., Fitzgerald, D.W., Mogg, R., Li, D., Gilbert, P.B.,Lama, J.R., Marmor, M., Del Rio, C., McElrath, M.J., Casimiro, D.R., Gottesdiener, K.M.,Chodakewitz, J.A., Corey, L., Robertson, M.N., 2008. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised,placebo-controlled, test-of-concept trial. Lancet 372 (9653), 1881–1893.

Bultmann, A., Muranyi, W., Seed, B., Haas, J., 2001. Identification of two sequences in thecytoplasmic tail of the human immunodeficiency virus type 1 envelopeglycoprotein that inhibit cell surface expression. J. Virol. 75 (11), 5263–5276.

Calain, P., Roux, L., 1993. The rule of six, a basic feature for efficient replication of Sendaivirus defective interfering RNA. J. Virol. 67 (8), 4822–4830.

Combredet, C., Labrousse-Najburg, V., Mollet, L., Lorin, C., Delebecque, F., Hurtrel, B.,McClure, H., Feinberg, M., Brahic, M., Tangy, F., 2003. A molecularly cloned Schwarzstrain of measles virus vaccine induces strong immune responses in macaques andtransgenic mice. J. Virol. 77 (21), 11546–11554.

de Swart, R.L., 2008. The pathogenesis of measles revisited. Pediatr. Infect. Dis. J. 27 (10Suppl.), S84–S88.

Desprès, P., Combredet, C., Frenkiel, M., Lorin, C., Brahic, M., Tangy, F., 2005. Livemeasles vaccine expressing the secreted form of the West Nile virus envelopeglycoprotein protects against West Nile virus encephalitis. J. Infect. Dis. 191 (2),207–214.

Dhiman, N., Jacobson, R.M., Poland, G.A., 2004. Measles virus receptors: SLAM andCD46. Rev. Med. Virol. 14 (4), 217–229.

Dorig, R.E., Marcil, A., Chopra, A., Richardson, C.D., 1993. The human CD46 molecule is areceptor for measles virus (Edmonston strain). Cell 75 (2), 295–305.

Ferre, A.L., Hunt, P.W., Critchfield, J.W., Young, D.H., Morris, M.M., Garcia, J.C., Pollard, R.B.,Yee Jr., H.F., Martin, J.N., Deeks, S.G., Shacklett, B.L., 2009. Mucosal immune responsesto HIV-1 in elite controllers: a potential correlate of immune control. Blood 113 (17),3978–3989.

Flynn, N.M., Forthal, D.N., Harro, C.D., Judson, F.N., Mayer, K.H., Para, M.F., 2005. Placebo-controlled phase 3 trial of a recombinant glycoprotein 120 vaccine to prevent HIV-1infection. J. Infect. Dis. 191 (5), 654–665.

Foley, H.D., Otero, M., Orenstein, J.M., Pomerantz, R.J., Schnell, M.J., 2002. Rhabdovirus-based vectors with human immunodeficiency virus type 1 (HIV-1) envelopesdisplay HIV-1-like tropism and target human dendritic cells. J. Virol. 76 (1), 19–31.

Guerbois, M., Moris, A., Combredet, C., Najburg, V., Ruffie, C., Fevrier, M., Cayet, N.,Brandler, S., Schwartz, O., Tangy, F., 2009. Live attenuated measles vaccineexpressing HIV-1 Gag virus like particles covered with gp160DeltaV1V2 is stronglyimmunogenic. Virology 388 (1), 191–203.

Guirakhoo, F., Pugachev, K., Zhang, Z., Myers, G., Levenbook, I., Draper, K., Lang, J., Ocran,S., Mitchell, F., Parsons, M., Brown, N., Brandler, S., Fournier, C., Barrere, B., Rizvi, F.,Travassos, A., Nichols, R., Trent, D., Monath, T., 2004. Safety and efficacy of chimericyellow Fever-dengue virus tetravalent vaccine formulations in nonhumanprimates. J. Virol. 78 (9), 4761–4775.

Guy, B., Guirakhoo, F., Barban, V., Higgs, S., Monath, T.P., Lang, J., 2010. Preclinical andclinical development of YFV 17D-based chimeric vaccines against dengue, WestNile and Japanese encephalitis viruses. Vaccine 28 (3), 632–649.

Kobune, F., Sakata, H., Sugiura, A., 1990. Marmoset lymphoblastoid cells as a sensitivehost for isolation of measles virus. J. Virol. 64 (2), 700–705.

Lemon, K., de Vries, R.D., Mesman, A.W., McQuaid, S., van Amerongen, G., Yuksel, S.,Ludlow, M., Rennick, L.J., Kuiken, T., Rima, B.K., Geijtenbeek, T.B., Osterhaus, A.D.,Duprex, W.P., de Swart, R.L., 2011. Early target cells of measles virus after aerosolinfection of non-human primates. PLoS Pathog. 7 (1), e1001263.

Letvin, N.L., 2006. Progress and obstacles in the development of an AIDS vaccine. Nat.Rev. Immunol. 6 (12), 930–939.

Liniger, M., Zuniga, A., Morin, T.N., Combardiere, B., Marty, R., Wiegand, M., Ilter, O.,Knuchel, M., Naim, H.Y., 2009. Recombinant measles viruses expressing single ormultiple antigens of human immunodeficiency virus (HIV-1) induce cellular andhumoral immune responses. Vaccine 27 (25–26), 3299–3305.

Lorin, C., Mollet, L., Delebecque, F., Combredet, C., Charneau, P., Hurtrel, B., Brahic, M.,Tangy, F., 2004. A single injection of recombinant measles vaccines expressing HIV-1 Clade B envelope glycoproteins induces neutralizing antibodies and cellularimmune responses to HIV. J. Virol. 78 (1), 146–157.

Lorin, C., Delebecque, F., Labrousse, V., Da Silva, L., Lemonnier, F., Brahic, M., Tangy, F.,2005a. A recombinant live attenuated measles vaccine vector primes effective HLA-

Page 9: A chimeric measles virus with a lentiviral envelope ...in Fig. 1A) was deleted from the vector plasmid pTM-MV-Schw and replaced by a synthetic chimeric MV–SIV sequence (Fig. 1B).

125T. Mourez et al. / Virology 419 (2011) 117–125

A0201-restricted cytotoxic T lymphocytes and broadly neutralizing antibodiesagainst HIV 1 conserved epitopes. Vaccine 23 (36), 4463–4472.

Lorin, C., Saidi, H., Belaid, A., Zairi, A., Baleux, F., Hocini, H., Belec, L., Hani, K., Tangy, F.,2005b. The antimicrobial peptide dermaseptin S4 inhibits HIV-1 infectivity in vitro.Virology 334 (2), 264–275.

Malkevitch, N.V., Robert-Guroff, M., 2004. A call for replicating vector prime-protein booststrategies in HIV vaccine design. Expert Rev. Vaccines 3 (4 Suppl.), S105–S117.

Marechal, V., Clavel, F., Heard, J.M., Schwartz, O., 1998. Cytosolic Gag p24 as an index ofproductive entry of human immunodeficiency virus type 1. J. Virol. 72 (3),2208–2212.

Mascola, J.R., Montefiori, D.C., 2010. The role of antibodies in HIV vaccines. Annu. Rev.Immunol. 28, 413–444.

McElrath, M.J., De Rosa, S.C., Moodie, Z., Dubey, S., Kierstead, L., Janes, H., Defawe, O.D.,Carter, D.K., Hural, J., Akondy, R., Buchbinder, S.P., Robertson,M.N.,Mehrotra, D.V., Self,S.G., Corey, L., Shiver, J.W., Casimiro, D.R., 2008. HIV-1 vaccine-induced immunity inthe test-of-concept step study: a case-cohort analysis. Lancet 372 (9653), 1894–1905.

Montefiori, D.C., Mascola, J.R., 2009. Neutralizing antibodies against HIV-1: can we elicitthem with vaccines and how much do we need? Curr. Opin. HIV AIDS 4 (5),347–351.

Mrkic, B., Pavlovic, J., Rulicke, T., Volpe, P., Buchholz, C.J., Hourcade, D., Atkinson, J.P.,Aguzzi, A., Cattaneo, R., 1998. Measles virus spread and pathogenesis in geneticallymodified mice. J. Virol. 72 (9), 7420–7427.

Mrkic, B., Odermatt, B., Klein, M., Billeter, M., Pavlovic, J., Cattaneo, R., 1999. Lymphaticdissemination and comparative pathology of recombinant measles viruses ingenetically modified mice. J. Virol. 74 (3), 1364–1372.

Naniche, D., Varior-Krishnan, G., Cervoni, F., Wild, T.F., Rossi, B., Rabourdin-Combe, C.,Gerlier, D., 1993. Human membrane cofactor protein (CD46) acts as a cellularreceptor for measles virus. J. Virol. 67 (10), 6025–6032.

Parks, C.L., Lerch, R.A., Walpita, P., Sidhu, M.S., Udem, S.A., 1999. Enhancedmeasles viruscDNA rescue and gene expression after heat shock. J. Virol. 73 (5), 3560–3566.

Pitisuttithum, P., Gilbert, P., Gurwith, M., Heyward, W., Martin, M., van Griensven, F., Hu, D.,Tappero, J.W., Choopanya, K., 2006. Randomized, double-blind, placebo-controlledefficacy trial of a bivalent recombinant glycoprotein 120 HIV-1 vaccine among injectiondrug users in Bangkok, Thailand. J. Infect. Dis. 194 (12), 1661–1671.

Pope, M., Haase, A.T., 2003. Transmission, acute HIV-1 infection and the quest forstrategies to prevent infection. Nat. Med. 9 (7), 847–852.

Pulendran, B., 2009. Learning immunology from the yellow fever vaccine: innateimmunity to systems vaccinology. Nat. Rev. Immunol. 9 (10), 741–747.

Querec, T.D., Akondy, R.S., Lee, E.K., Cao, W., Nakaya, H.I., Teuwen, D., Pirani, A., Gernert, K.,Deng, J., Marzolf, B., Kennedy, K., Wu, H., Bennouna, S., Oluoch, H., Miller, J., Vencio, R.Z.,

Mulligan, M., Aderem, A., Ahmed, R., Pulendran, B., 2009. Systems biology approachpredicts immunogenicity of the yellow fever vaccine in humans. Nat. Immunol. 10 (1),116–125.

Radecke, F., Spielhofer, P., Schneider, H., Kaelin, K., Huber, M., Dotsch, C., Christiansen,G., Billeter, M.A., 1995. Rescue ofmeasles viruses from cloned DNA. EMBO J. 14 (23),5773–5784.

Rerks-Ngarm, S., Pitisuttithum, P., Nitayaphan, S., Kaewkungwal, J., Chiu, J., Paris, R., Premsri,N.,Namwat, C., de Souza,M., Adams, E., Benenson,M.,Gurunathan, S., Tartaglia, J.,McNeil,J.G., Francis, D.P., Stablein, D., Birx, D.L., Chunsuttiwat, S., Khamboonruang, C.,Thongcharoen, P., Robb, M.L., Michael, N.L., Kunasol, P., Kim, J.H., 2009. Vaccinationwith ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N. Engl. J. Med. 361(23), 2209–2220.

Rouxel, R.N., Svitek, N., von Messling, V., 2009. A chimeric measles virus with caninedistemper envelope protects ferrets from lethal distemper challenge. Vaccine 27(36), 4961–4966.

Salter, R.D., Howell, D.N., Cresswell, P., 1985. Genes regulating HLA class I antigenexpression in T-B lymphoblast hybrids. Immunogenetics 21 (3), 235–246.

Sellin, C.I., Davoust, N., Guillaume, V., Baas, D., Belin, M.F., Buckland, R., Wild, T.F.,Horvat, B., 2006. High pathogenicity of wild-type measles virus infection in CD150(SLAM) transgenic mice. J. Virol. 80 (13), 6420–6429.

Singh, M., Cattaneo, R., Billeter, M.A., 1999. A recombinant measles virus expressinghepatitis B virus surface antigen induces humoral immune responses in geneticallymodified mice. J. Virol. 73 (6), 4823–4828.

Spielhofer, P., Bachi, T., Fehr, T., Christiansen, G., Cattaneo, R., Kaelin, K., Billeter, M.,Naim, H., 1998. Chimeric measles viruses with a foreign envelope. J. Virol. 72,2150–2159.

Tangy, F., Charneau, P., Jacob, Y., 2006. Cells and methodology to generate non-segmented negative-strand RNA viruses. European Patent Application filed onAugust 25, 2006 N° 06291358.7.

Tatsuo, H., Ono, N., Tanaka, K., Yanagi, Y., 2000. SLAM (CDw150) is a cellular receptor formeasles virus. Nature 406 (6798), 893–897.

Wang, Z., Hangartner, T., Cornu, L., Martin, A., Zuniga, M., Billeter, M., Naim, H., 2001.Recombinant measles viruses expressing heterologous antigens of mumps andsimian immunodeficiency viruses. Vaccine 19, 2329–2336.

Welstead, G.G., Iorio, C., Draker, R., Bayani, J., Squire, J., Vongpunsawad, S., Cattaneo,R., Richardson, C.D., 2005. Measles virus replication in lymphatic cells andorgans of CD150 (SLAM) transgenic mice. Proc. Natl. Acad. Sci. U.S.A. 102 (45),16415–16420.

Yanagi, Y., Takeda, M., Ohno, S., 2006. Measles virus: cellular receptors, tropism andpathogenesis. J. Gen. Virol. 87 (Pt 10), 2767–2779.


Recommended