8
Vaccine 33 (2015) 7254–7261 Contents lists available at ScienceDirect Vaccine j o ur na l ho me page: www.elsevier.com/locate/vaccine RT-qPCR-based microneutralization assay for human cytomegalovirus using fibroblasts and epithelial cells Xiao Wang, Keith Peden, Haruhiko Murata Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA a r t i c l e i n f o Article history: Received 12 February 2015 Received in revised form 28 August 2015 Accepted 28 October 2015 Available online 6 November 2015 Keywords: Reverse transcription quantitative PCR Virus-neutralization assay Cell lysate Human cytomegalovirus Epithelial cell Fibroblast a b s t r a c t Human cytomegalovirus (HCMV) is a leading cause of congenital infection that can result in serious dis- abilities in affected children. To facilitate HCMV vaccine development, a microscale neutralization assay based on reverse transcription quantitative PCR (RT-qPCR) was developed to quantify HCMV-neutralizing antibodies. Our approach relies on the generation of crude lysates from virus-infected cells that are amenable to direct analysis by RT-qPCR, thereby circumventing rate-limiting procedures associated with sample RNA extraction and purification. By serial passaging of the laboratory HCMV strain AD169 in epithelial cells (ARPE-19), a revertant virus with restored epithelial cell tropism, designated AD169 wt131 , was obtained. AD169 and AD169 wt131 were evaluated in both epithelial cells (ARPE-19) and fibroblasts (MRC-5) by one-step RT-qPCR targeting the immediate-early gene IE1 transcript of HCMV. Expression kinetics indicated that RT-qPCR assessment could be conducted as early as 6 h post-infection. Human serum samples (n = 30) from healthy donors were tested for HCMV-specific IgG using a commercially available ELISA and for HCMV-neutralizing activity using our RT-qPCR-based neutralization assay. In agreement with the ELISA results, higher neutralizing activity was observed in the HCMV IgG seroposi- tive group when compared with the HCMV IgG seronegative group. In addition, HCMV IgG seropositive human sera exhibited higher neutralizing titers using epithelial cells compared with using fibroblasts (geometric mean titers of 344 and 8 in ARPE-19 cells and MRC-5 cells, respectively). Our assay was robust to variation in input virus dose. In addition, a simple lysis buffer containing a non-ionic detergent was successfully demonstrated to be a less costly alternative to commercial reagents for cell-lysate prepara- tion. Thus, our rapid HCMV neutralization assay may be a straightforward and flexible high-throughput tool for measuring antibody responses induced by vaccination and natural infection. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http:// creativecommons.org/licenses/by-nc-nd/4.0/). 1. Introduction Human cytomegalovirus (HCMV) is a leading cause of congen- ital infection that can result in permanent disabilities [1]. In the United States, 30,000 to 40,000 infants are born with congenital HCMV infection annually (0.6% of births), of whom more than 7000 are symptomatic (a number exceeding other congenital dis- orders for which screening is available, such as spina bifida and Down syndrome) [2–4]. An effective vaccine for prophylaxis is not yet available. A cost/benefit analysis by the Institute of Medicine Corresponding author. Food and Drug Administration, 10903 New Hamp- shire Avenue, Building 52/72, Room 1312, Silver Spring, MD 20993, USA. Tel.: +1 240 402 9767. E-mail address: [email protected] (H. Murata). has ranked a vaccine for HCMV to be a high priority for future development [5]. Neutralizing antibodies may be important for conferring protec- tion against HCMV. Prevention of congenital HCMV infection was observed in a non-randomized study in which HCMV hyperimmune globulin was administered to pregnant women with primary HCMV infection; the transmission rate was 16% in treated women com- pared with 40% in untreated women [6]. However, a subsequent randomized, placebo-controlled Phase 2 study of hyperimmune globulin treatment in pregnant women reported a smaller effect on transmission rate (30% with treatment vs. 44% with placebo), which did not meet the threshold for statistical significance [7]. Confir- mation of efficacy awaits the results of other ongoing randomized Phase 3 studies [7]. The relevant HCMV targets for neutralizing antibodies may dif- fer according to the cell type used in the neutralization assay. Entry of HCMV into epithelial and endothelial cells is mediated by a viral http://dx.doi.org/10.1016/j.vaccine.2015.10.110 0264-410X/Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

RT-qPCR-based microneutralization assay for human ... · RT-qPCR-based microneutralization assay for human cytomegalovirus using fibroblasts and epithelial cells Xiao ... 2.3. RT-qPCR-based

Embed Size (px)

Citation preview

Page 1: RT-qPCR-based microneutralization assay for human ... · RT-qPCR-based microneutralization assay for human cytomegalovirus using fibroblasts and epithelial cells Xiao ... 2.3. RT-qPCR-based

Ru

XLF

a

ARRAA

KRVCHEF

1

iUH7oDy

sT

h0

Vaccine 33 (2015) 7254–7261

Contents lists available at ScienceDirect

Vaccine

j o ur na l ho me page: www.elsev ier .com/ locate /vacc ine

T-qPCR-based microneutralization assay for human cytomegalovirussing fibroblasts and epithelial cells

iao Wang, Keith Peden, Haruhiko Murata ∗

aboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research,ood and Drug Administration, Silver Spring, MD 20993, USA

r t i c l e i n f o

rticle history:eceived 12 February 2015eceived in revised form 28 August 2015ccepted 28 October 2015vailable online 6 November 2015

eywords:everse transcription quantitative PCRirus-neutralization assayell lysateuman cytomegaloviruspithelial cellibroblast

a b s t r a c t

Human cytomegalovirus (HCMV) is a leading cause of congenital infection that can result in serious dis-abilities in affected children. To facilitate HCMV vaccine development, a microscale neutralization assaybased on reverse transcription quantitative PCR (RT-qPCR) was developed to quantify HCMV-neutralizingantibodies. Our approach relies on the generation of crude lysates from virus-infected cells that areamenable to direct analysis by RT-qPCR, thereby circumventing rate-limiting procedures associated withsample RNA extraction and purification. By serial passaging of the laboratory HCMV strain AD169 inepithelial cells (ARPE-19), a revertant virus with restored epithelial cell tropism, designated AD169wt131,was obtained. AD169 and AD169wt131 were evaluated in both epithelial cells (ARPE-19) and fibroblasts(MRC-5) by one-step RT-qPCR targeting the immediate-early gene IE1 transcript of HCMV. Expressionkinetics indicated that RT-qPCR assessment could be conducted as early as 6 h post-infection. Humanserum samples (n = 30) from healthy donors were tested for HCMV-specific IgG using a commerciallyavailable ELISA and for HCMV-neutralizing activity using our RT-qPCR-based neutralization assay. Inagreement with the ELISA results, higher neutralizing activity was observed in the HCMV IgG seroposi-tive group when compared with the HCMV IgG seronegative group. In addition, HCMV IgG seropositivehuman sera exhibited higher neutralizing titers using epithelial cells compared with using fibroblasts

(geometric mean titers of 344 and 8 in ARPE-19 cells and MRC-5 cells, respectively). Our assay was robustto variation in input virus dose. In addition, a simple lysis buffer containing a non-ionic detergent wassuccessfully demonstrated to be a less costly alternative to commercial reagents for cell-lysate prepara-tion. Thus, our rapid HCMV neutralization assay may be a straightforward and flexible high-throughputtool for measuring antibody responses induced by vaccination and natural infection.

Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://

. Introduction

Human cytomegalovirus (HCMV) is a leading cause of congen-tal infection that can result in permanent disabilities [1]. In thenited States, 30,000 to 40,000 infants are born with congenitalCMV infection annually (∼0.6% of births), of whom more than000 are symptomatic (a number exceeding other congenital dis-rders for which screening is available, such as spina bifida and

own syndrome) [2–4]. An effective vaccine for prophylaxis is notet available. A cost/benefit analysis by the Institute of Medicine

∗ Corresponding author. Food and Drug Administration, 10903 New Hamp-hire Avenue, Building 52/72, Room 1312, Silver Spring, MD 20993, USA.el.: +1 240 402 9767.

E-mail address: [email protected] (H. Murata).

ttp://dx.doi.org/10.1016/j.vaccine.2015.10.110264-410X/Published by Elsevier Ltd. This is an open access article under the CC BY-NC-N

creativecommons.org/licenses/by-nc-nd/4.0/).

has ranked a vaccine for HCMV to be a high priority for futuredevelopment [5].

Neutralizing antibodies may be important for conferring protec-tion against HCMV. Prevention of congenital HCMV infection wasobserved in a non-randomized study in which HCMV hyperimmuneglobulin was administered to pregnant women with primary HCMVinfection; the transmission rate was 16% in treated women com-pared with 40% in untreated women [6]. However, a subsequentrandomized, placebo-controlled Phase 2 study of hyperimmuneglobulin treatment in pregnant women reported a smaller effect ontransmission rate (30% with treatment vs. 44% with placebo), whichdid not meet the threshold for statistical significance [7]. Confir-mation of efficacy awaits the results of other ongoing randomized

Phase 3 studies [7].

The relevant HCMV targets for neutralizing antibodies may dif-fer according to the cell type used in the neutralization assay. Entryof HCMV into epithelial and endothelial cells is mediated by a viral

D license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Page 2: RT-qPCR-based microneutralization assay for human ... · RT-qPCR-based microneutralization assay for human cytomegalovirus using fibroblasts and epithelial cells Xiao ... 2.3. RT-qPCR-based

ine 33

pLITticfibd

rigaptcttoat

2

2

o5(AEF

Aeapw1c1dp(bGfcw

2

wDdBamR3c

X. Wang et al. / Vacc

entameric complex, which contains envelope glycoproteins H and (gH and gL) along with pUL128, pUL130, and pUL131 [8–10].n contrast, laboratory-adapted HCMV strains such as AD169 orowne are propagated efficiently only in fibroblasts; this restrictedropism is caused by mutations in the UL131-UL128 locus thatmpair proper expression of the gH/gL/pUL128/pUL130/pUL131omplex [8,10]. Induction of neutralizing antibodies with speci-city towards the gH/gL/pUL128/pUL130/pUL131 complex haseen recently considered to be an important goal in HCMV vaccineevelopment [11].

High-throughput microneutralization assays for HCMV in cur-ent use include those based on enumerating infected cells throughmmunostaining for HCMV antigens [12,13] or measuring reporter-ene activity expressed from an engineered virus [14,15]. No singlessay platform has emerged as the favored consensus. In theresent study, we have developed an alternative rapid microneu-ralization assay for HCMV using both fibroblasts and epithelialells. Our approach involves measuring the extent of virus infec-ion by subjecting crude cell lysates prepared from infected cellso direct analysis by one-step RT-qPCR. The straightforward naturef the manipulation steps in our assay may contribute to desir-ble features such as robustness, adaptability to automation, andransferability across laboratories.

. Materials and methods

.1. Cell cultures and viruses

MRC-5 fibroblasts and ARPE-19 retinal epithelial cells werebtained from American Type Culture Collection (ATCC). MRC-

cells were maintained in Eagle’s Minimum Essential MediumATCC) supplemented with 10% fetal bovine serum (FBS; Hyclone).RPE-19 cells were maintained in Advanced Dulbecco’s Modifiedagle’s Medium/F12 (Life Technologies) supplemented with 10%BS and 1X GlutaMAX (Life Technologies).

Laboratory-adapted HCMV strain AD169 was obtained fromTCC and propagated in MRC-5 cells. To obtain a virus with restoredpithelial cell tropism, ARPE-19 cells were infected with AD169 at

multiplicity of infection (MOI) of 1.25. Infected cells were seriallyassaged as described [16]. Briefly, ARPE-19 cells were passagedeekly by mixing infected cells with uninfected cells at a ratio of

:2. Virus was collected when the cytopathic effect (CPE) of theells reached 100%. The revertant virus was then passaged in ARPE-9 cells one additional time. Both AD169 and the revertant virus,esignated as AD169wt131, were titered in MRC-5 cells through end-oint dilution by calculating the 50% tissue culture infective doseTCID50). The UL131 gene was PCR-amplified and sequenced foroth AD169 and AD169wt131. AD169 matched the sequence data inenBank (X17403 and FJ527563) and contained a single-nucleotide

rame-shifting insertion in UL131 (A at position 84 after the startodon); AD169wt131 lacked this single-nucleotide insertion, butas elsewhere identical to sequence data for this strain in GenBank.

.2. Antibodies for neutralization assays

A neutralizing mouse monoclonal anti-gH antibody (clone 51C1)as obtained from a commercial source (Acris Antibodies; Saniego, CA, USA). Thirty individual serum samples from healthy maleonors ranging in age from 20 to 63 were obtained from Valleyiomedical (Winchester, VA, USA). The presence of IgG antibodiesgainst HCMV in the serum samples was evaluated using a com-

ercial ELISA kit (Zeus Scientific; ELISA Cytomegalovirus IgG Test;

aritan, NJ, USA). Serum samples were heat-inactivated at 56 ◦C for0 min prior to assessing HCMV-neutralizing activity to inactivateomplement.

(2015) 7254–7261 7255

2.3. RT-qPCR-based microneutralization assay

Cell monolayers (MRC-5 or ARPE-19) were prepared by seed-ing 96-well culture plates (15,000 cells per well). On the followingday, 2-fold serial dilutions of antibody or serum were made in theappropriate base culture medium supplemented with 1% FBS andthen mixed with equal volume of virus (AD169 or AD169wt131).Unless otherwise indicated, the final input of virus in each well(pre-neutralization) was 1000 TCID50. After incubation at 37 ◦C for1 h, the mixtures were transferred to cells in the 96-well plates.Cells were incubated for an additional 6 h and then washed twicewith phosphate-buffered saline (100 �L per well). Cell lysates weregenerated using iScript Sample Preparation Reagent (Bio-Rad) orCell-Lysis (CL) Buffer containing 10 mM Tris-HCl, pH 7.4, 0.25%Igepal CA-630, and 150 mM NaCl [17]. After exposure of cells to lysisreagent (100 �L per well) at room temperature (22 ◦C) for 2 min,the resulting cell lysates were carefully collected and stored frozenuntil analysis by RT-qPCR.

SYBR Green-based RT-qPCR analysis was performed usingthe iScript One-Step RT-PCR kit (Bio-Rad). UL123 and UL89were chosen as gene targets for RT-qPCR detection with oneof the primers from each pair spanning an exon–exon junc-tion. Both primer pairs were designed using QuantPrime [18].Primers for UL123 were AGATGTCCTGGCAGAACTCGTC (for-ward) and TTCTATGCCGCACCATGTCCAC (reverse). Primers forUL89 were CCAGTTGCTACAACACCAACAGC (forward) and AGGC-CTCTTTCTTGATGAAGTGC (reverse). The amplicon size was 62 bpfor UL123 and 87 bp for UL89. The final RT-qPCR mixture contained1X iScript One-Step SYBR Green RT-PCR mix, 300 nM each of for-ward and reverse primer, 0.2 �L of iScript reverse transcriptase,1 �L of sample cell lysate, and water to 10 �L. RT-qPCR cycling pro-tocol using the CFX-96 instrument (Bio-Rad) was as follows: 50 ◦Cfor 10 min (1X); 95 ◦C for 5 min (1X); and 95 ◦C for 10 s, 65.5 ◦C(UL123) or 61.5 ◦C (UL89) for 30 s (40X). An RNA standard wasobtained by purifying total RNA from AD169-infected MRC-5 cellsusing the RNeasy kit (Qiagen). For each run, a 10-fold dilution seriesof RNA standard (using cell lysate from uninfected cells as the dilu-ent) was included to aid in quantification; negative controls (lysatesfrom uninfected cells) and no-reverse transcription controls werealso included. Amplification efficiency usually ranged between 90%and 105%. The neutralization endpoint was defined as the concen-tration (IC90) or fold-dilution (NT90) of antibody/serum necessaryfor 90% inhibition of RT-qPCR signal in comparison with virus con-trol samples (infected cells in the absence of neutralizing activity).IC90 or NT90 values were estimated by linear interpolation derivedfrom the two experimental dilutions immediately above and imme-diately below the threshold for neutralization.

2.4. Immunostaining-based microneutralization assay

ARPE-19 cell monolayers, serum dilutions, and cell infections(using AD169wt131) were prepared as described in the RT-qPCR-based microneutralization assay. Infected cells were incubatedat 37 ◦C for 18 h. Immunostaining was performed as describedby Abai et al. [12] with minor modifications. Briefly, cells werewashed once with PBS and fixed with absolute ethanol for 20 min.Blocking was performed with 2% horse serum for 1 h, followedby treatment with 0.3% hydrogen peroxide in PBS for 30 minto quench endogenous peroxidase activity. After washing withPBS three times, cells were incubated with the primary anti-IE1monoclonal antibody (MAB810; Millipore) at a concentration of0.1 �g/mL for 1 h. The secondary biotinylated anti-mouse IgG

antibody and VECTASTAIN ABC reagent were prepared as recom-mended by the supplier (Vector Laboratories; Burlingame, CA,USA). 3,3′,5,5′-tetramethylbenzidine (TMB; Vector Laboratories)was used as the peroxidase substrate. After allowing the stain
Page 3: RT-qPCR-based microneutralization assay for human ... · RT-qPCR-based microneutralization assay for human cytomegalovirus using fibroblasts and epithelial cells Xiao ... 2.3. RT-qPCR-based

7256 X. Wang et al. / Vaccine 33 (2015) 7254–7261

F ial cel wt131

t 0. Cel1 alysisn sents

isOtdii

3

3c

rpppdtr

3

Htc

Fpac

ig. 1. IE1 transcription of HCMV in (A) MRC-5 fibroblasts and (B) ARPE-19 epithelo inoculate seeded cells in 96-well plates (15,000 cells/well) using a TCID50 of 100, 3, 6, 12, or 24 h post-infection and subjected to SYBR Green one-step RT-qPCR anumbers were normalized to the mean value at 1 h post-infection. Each point repre

ntensity to develop for 5 min, the plates were rinsed, dried, andcanned using the ImmunoSpot instrument (CTL; Shaker Heights,H, USA). The IE1-positive cells were analyzed and counted using

he BioSpot 5.0 software (CTL). The neutralization endpoint wasefined as the fold-dilution (NT90) of serum resulting in 90%

nhibition of infected foci in comparison with virus control wellsn the absence of neutralizing activity.

. Results

.1. Adaptation of a laboratory HCMV strain to restore epithelialell tropism

The tropism of laboratory-adapted HCMV strain AD169 isestricted to fibroblasts; this property results from a single base-air insertion in the gene UL131 [8,10]. AD169 was seriallyassaged in ARPE-19 cells to rescue its epithelial cell tropism. Atassage 6, extensive CPE was observed. Cell-free revertant virus,esignated as AD169wt131, was collected and sequencing confirmedhat the complete open reading frame of UL131 was restored byemoval of the single base-pair insertion found in the parental virus.

.2. Optimization of RT-qPCR for HCMV detection in cell culture

To develop an RT-qPCR-based microneutralization assay forCMV, we adapted an approach that we had successfully applied

o study RNA viruses [19,20]. Our assay relies on generating crudeell lysates that can be directly used for RT-qPCR. Cell-lysate

ig. 2. Linear range of HCMV IE1 RT-qPCR in (A) MRC-5 fibroblasts and (B) ARPE-19 epitlates (15,000 cells/well). Input virus TCID50 ranged from 125 to 8000 (two-fold series).

t 6 h post-infection followed by RT-qPCR analysis. Copy numbers were normalized agaiorresponding range (n = 3).

ls. Laboratory-adapted strain AD169 and its revertant virus AD169 were usedl lysates were prepared using iScript Sample Preparation Reagent and collected at

with primers targeting the HCMV immediate-early gene IE1 transcript. RNA copy the mean with corresponding range (n = 3).

preparation involves the following straightforward steps: (1) cellwashing, (2) cell exposure to lysis reagent, and (3) collection oflysates for immediate analysis or frozen storage. The transcript ofthe immediate-early gene IE1 (UL123) was chosen as the targetfor RT-qPCR. One of the primers was designed to span the splicejunction between exons 3 and 4 to detect target viral RNA specif-ically. No amplification from viral genomic DNA was observedas demonstrated by control reactions in the absence of reversetranscriptase (data not shown).

IE1 transcription of AD169 and AD169wt131 was evaluated by RT-qPCR using MRC-5 fibroblasts and ARPE-19 epithelial cells grownin 96-well plates (15,000 cells per well; 1000 TCID50 of virus perwell). Cell lysates were prepared at indicated times after infectionusing a commercial cell lysis reagent (Bio-Rad) and directly sub-jected to one-step SYBR Green RT-qPCR. AD169wt131 was capableof infecting both MRC-5 and ARPE-19 cells with the RT-qPCR signalnearing a plateau as early as 6 h post-infection (Fig. 1). In contrast,the parental AD169 was only able to infect efficiently MRC-5 cells(Fig. 1A); signal accumulation was modest in AD169-infected ARPE-19 cells, reflecting its deficiency in epithelial cell tropism (Fig. 1B).The observed kinetics supported using the 6-h time point for end-point assessment in our neutralization assay.

We further evaluated the relationship between RT-qPCR signaland input virus quantity. MRC-5 and ARPE-19 cells were infected

with either AD169 or AD169wt131 (2-fold increments ranging from125 to 8000 TCID50 per well), and the IE1 transcript was quan-tified at 6 h post-infection. In ARPE-19 cells, the RT-qPCR signalincreased according to input AD169wt131 in an approximately linear

helial cells. AD169 and AD169wt131 were used to inoculate seeded cells in 96-wellCell lysates were prepared using iScript Sample Preparation Reagent and collectednst the mean value of AD169 at 125 TCID50. Each point represents the mean with

Page 4: RT-qPCR-based microneutralization assay for human ... · RT-qPCR-based microneutralization assay for human cytomegalovirus using fibroblasts and epithelial cells Xiao ... 2.3. RT-qPCR-based

ine 33

miptwp

3a

ccAbwlnia(d2mfbtap

3

tMsc(I

FvvNi

X. Wang et al. / Vacc

anner (Fig. 2B). However, in MRC-5 cells, linearity between virusnput and RT-qPCR signal was observed at or below 1000 TCID50er well (Fig. 2A). Thus, the standard input virus dose for our neu-ralization assay was chosen to be 1000 TCID50 per well to remainithin the linear range while maximizing signal detectable at 6 host-infection.

.3. HCMV-neutralizing activity of a monoclonal anti-gHntibody

The neutralizing activity of a commercially available mono-lonal anti-gH antibody (clone 51C1) was determined in MRC-5ells using both AD169 and AD169wt131, and in ARPE-19 cells usingD169wt131. Two-fold serial dilutions of the antibody were incu-ated with equal volume of virus. The virus-antibody mixturesere then transferred to previously seeded cell-culture plates. Cell

ysates were collected after 6 additional hours of incubation andeutralization was assessed by RT-qPCR. A comparison of neutraliz-

ng activity in MRC-5 fibroblasts (Fig. 3A) revealed that the anti-gHntibody exhibited a higher potency (lower IC90) against AD1691.5 �g/mL) than against AD169wt131 (9.7 �g/mL). In contrast, theifference in neutralizing activity against AD169wt131 was within-fold across cell types (Fig. 3A and B). Data generated using aonoclonal antibody with known specificity to HCMV support the

easibility of measuring virus neutralization using our RT-qPCR-ased assay. Furthermore, the gH associated with AD169 appearso be more efficiently targeted for neutralization by the anti-gHntibody 51C1 compared with the gH associated with AD169wt131,ossibly due to occlusion of the gH epitope in AD169wt131.

.4. HCMV-neutralizing activity of human serum samples

Thirty human serum samples from healthy male donors wereested for HCMV-neutralizing activity using AD169wt131 in both

RC-5 and ARPE-19 cells. We first investigated whether these

erum samples contained HCMV-specific IgG antibodies using aommercial ELISA. The 30 samples were organized into two groupsSupplemental Table 1): HCMV IgG seropositive (n = 22) and HCMVgG seronegative (n = 8).

ig. 3. HCMV-neutralizing activity of an anti-gH monoclonal antibody (51C1) assessed byolume of virus for 1 h. The virus–antibody mixtures were then transferred to 96-well cultuirus quantity was 1000 TCID50 per well. Cell lysates were prepared using iScript Sample Preutralization endpoint (IC90) was defined as the antibody concentration necessary for

n the absence of neutralizing activity). Each point represents the mean with correspondi

(2015) 7254–7261 7257

Concordance was observed between neutralizing activity of serameasured in ARPE-19 cells and HCMV IgG serostatus determined byELISA (Fig. 4A). HCMV IgG positive sera were associated with sub-stantial neutralizing activity against AD169wt131 in ARPE-19 cells(NT90 ranging from 21 to 1880). In contrast, HCMV IgG negativesera were associated with substantially lower neutralizing activ-ity (NT90 ranging from <10 to 47). Geometric mean titers (GMTs)in ARPE-19 cells were 344 and 7 for positive and negative sera,respectively (Fig. 4A; NT90 titers <10 were assigned a value of 5).A marked difference in neutralizing activity was observed in MRC-5 cells compared with ARPE-19 cells. GMTs in MRC-5 cells were8 and 5 for positive and negative sera, respectively (Fig. 4A; NT90titers <10 were assigned a value of 5). Neutralization data in bothARPE-19 and MRC-5 cells for a representative sample (#11) areshown in Fig. 4B; titers for this sample were 1880 and 28 in ARPE-19cells and MRC-5 cells, respectively. For samples with determinabletiters using both cell types (n = 7; Supplemental Table 1), the meanfold-change in titer (ARPE-19/MRC-5) was 47 (ranging from 12 to67). The results suggest that sera obtained following natural infec-tion exhibit higher HCMV-neutralizing activity in epithelial cellscompared with fibroblasts.

Neutralization results obtained using our RT-qPCR-basedapproach were compared with those obtained using an approachbased on immunostaining cells expressing IE1 [12]. The data areshown in Fig. 4C. Using serum samples with measurable neutral-ization titers in both assays in ARPE-19 cells (n = 24; six sampleshad titers of <10 in both assays), a high degree of correlation wasobserved (r = 0.99).

To confirm our neutralization results, we applied our RT-qPCR-based approach using UL89 as an alternative target. UL89 isreported to be expressed with early-late kinetics [21] and encodesa subunit of terminase [22]. Again, one of the primers for UL89spanned an exon–exon junction, thereby allowing viral UL89 RNAto be specifically detected. Neutralization of AD169wt131 with UL89RNA as the detection target was assessed at 72 h post-infection

in ARPE-19 cells using a subset of our serum samples (#11, #15,#21, and #26); neutralization results were comparable with thoseobtained using IE1 RNA as the detection target (Supplemental Fig.1 and Supplemental Table 1).

RT-qPCR. Two-fold serial dilutions of anti-gH antibody were incubated with equalre plates of (A) MRC-5 cells or (B) ARPE-19 cells such that the final pre-neutralizationeparation Reagent and collected at 6 h post-infection followed by RT-qPCR analysis.90% inhibition of RT-qPCR signal compared with virus control wells (infected cellsng range (n = 2).

Page 5: RT-qPCR-based microneutralization assay for human ... · RT-qPCR-based microneutralization assay for human cytomegalovirus using fibroblasts and epithelial cells Xiao ... 2.3. RT-qPCR-based

7258 X. Wang et al. / Vaccine 33 (2015) 7254–7261

Fig. 4. HCMV-neutralizing activity of human serum samples assessed by RT-qPCR. Two-fold serial dilutions of human sera were incubated with equal volume of virus(AD169wt131) for 1 h. The virus-serum mixtures were then transferred to 96-well culture plates of MRC-5 or ARPE-19 such that the final pre-neutralization virus quantity was1000 TCID50 per well. Cell lysates were prepared using iScript Sample Preparation Reagent and collected at 6 h post-infection followed by RT-qPCR analysis. Neutralizationtiter (NT90) was defined as the dilution of serum necessary for 90% inhibition of RT-qPCR signal compared with virus control wells (infected cells in the absence of neutralizingactivity). (A) Neutralization results from the entire collection of samples (n = 30; 22 seropositives and 8 seronegatives) are summarized. Individual titers as well as geometricmean titers (GMTs) for each group are shown according to cell type used in the assay (MRC-5 or ARPE-19). For this analysis, samples with titers below the lowest dilution(<1:10) were assigned a value of 5. (B) Neutralization results from one representative serum sample (#11) are shown. Each point represents the mean with corresponding range( resultn efficie

3

iltTcT(rti

3r

ce[

n = 2). (C) RT-qPCR-based neutralization results are compared with neutralizationeutralization titers in both assays in ARPE-19 cells; n = 24; Pearson’s correlation co

.5. Assay compliance with the percentage law

We investigated whether the performance of our microneutral-zation assay conforms to predictions according to the percentageaw [23] (i.e., regardless of virus input, the proportion of virus neu-ralized by a given concentration of antibody remains constant).wo human serum samples, #11 and #21, were tested in ARPE-19ells using a range of input virus doses (1000, 2000, 4000, or 8000CID50 of AD169wt131). Titers of both sera were minimally affectedwithin 2-fold) by changes in input virus dose. The NT90 valuesanged from 1188 to 1834 for sample #11 (Fig. 5A) and from 517o 936 for sample #21 (Fig. 5B). The results suggest that our assays relatively robust to perturbations in input virus dose.

.6. A simple cell-lysis buffer as an alternative to commercialeagents

We previously reported that a simple cell-lysis buffer (CL Buffer)ontaining a non-ionic detergent (0.25% Igepal CA-630) workedffectively for preparation of cell lysates that are RT-qPCR-ready17]. Samples for our assay were generated using CL Buffer, and

s based on immunostaining cells expressing IE1 (serum samples with measurablent r = 0.99).

neutralization results were compared with those obtained usingthe commercial cell-lysis reagent (Bio-Rad). The monoclonal anti-gH antibody 51C1 was tested in MRC-5 cells using AD169 and inARPE-19 cells using AD169wt131. A subset of human serum sam-ples (#11, #15, #21, and #26) were tested in ARPE-19 cells usingAD169wt131. For the monoclonal anti-gH antibody 51C1, the IC90sobtained using CL Buffer were 1.6 �g/mL for AD169 and 9.4 �g/mLfor AD169wt131 (Supplemental Fig. 2A), consistent with valuesobtained using the commercial reagent (Fig. 3). In addition, all foursera tested using CL Buffer were associated with titers comparablewith those obtained using the commercial reagent (SupplementalFig. 2B and Supplemental Table 1). We conclude that CL Buffer isa suitable and less costly alternative for generating samples to beused in our neutralization assay.

4. Discussion

We have recently developed neutralization assays for sev-eral viruses with endpoint assessment determined by qPCR.We have emphasized an approach that can be used in

Page 6: RT-qPCR-based microneutralization assay for human ... · RT-qPCR-based microneutralization assay for human cytomegalovirus using fibroblasts and epithelial cells Xiao ... 2.3. RT-qPCR-based

X. Wang et al. / Vaccine 33 (2015) 7254–7261 7259

Fig. 5. Neutralization of HCMV is consistent with the percentage law. Two-fold serial dilutions of individual human serum samples were incubated with varying quantitiesof virus (AD169wt131) for one hour. The virus–serum mixtures were then transferred to 96-well culture plates of ARPE-19 cells; the final pre-neutralization quantities perw pt Sama . Each

htausfs(ccvRw

fotcRRdtriunttm

Hrltdt

ell were 1000, 2000, 4000, or 8000 TCID50. Cell lysates were prepared using iScrinalysis. Results of two representative human sera, (A) #11 and (B) #21, are shown

igh-throughput applications. The rate-limiting steps for conven-ional PCR workflows are usually associated with sample nucleiccid extraction/purification. In our initial proof-of-principle studysing SV40, a non-enveloped DNA virus, we successfully avoidedample DNA purification by subjecting diluted culture supernatantrom virus-infected cells to SYBR Green qPCR [24]. In subsequenttudies, we extended our approach to encompass RNA virusesinfluenza virus and respiratory syncytial virus) [19,20]. Again, weircumvented the need for sample RNA purification by using crudeell lysates as input for one-step SYBR Green RT-qPCR to quantifyiral RNA. Recent studies suggest that crude cell lysates as input forT-qPCR offer similar accuracy and superior sensitivity comparedith purified RNA [25,26].

In the present study, we have developed a neutralization assayor HCMV, an enveloped DNA virus. We did not consider samplingf culture supernatant for cell-released virus (as was done for SV40)o be practical for HCMV because its infectivity typically remainsell-associated. A decision was made to target cell-associated viralNA rather than viral DNA because (1) choosing a highly expressedNA would constitute an amplification in itself and thus facilitateetection/quantification, and (2) viral DNA replication is restrictedo the cell nucleus and thus efficient release of viral DNA mayequire harsher cell-lysis conditions. An RNA target for RT-qPCRn the case of a DNA virus such as HCMV entails additional meas-res to minimize signal contribution from viral DNA; this issue isot a concern for RNA viruses that have no DNA intermediates inheir life cycle. One of our primers spans an exon–exon splice junc-ion, which allows for specific quantification of HCMV RNA with

inimal bias due to viral DNA.Others have reported high-throughput cell-based assays for

CMV with a qPCR readout in the context of screening for antivi-al compounds [27,28]. These assays target viral DNA in diluted cell

ysates rather than viral RNA, likely a deliberate choice by the inves-igators as many of the antiviral compounds used in their studiesirectly inhibit viral DNA synthesis. Targeting viral DNA appearso be associated with lengthening of the assay (assessment at 3–4

ple Preparation Reagent and collected at 6 h post-infection followed by RT-qPCR point represents the mean with corresponding range (n = 2).

days post-infection) [27,28] compared with our approach of tar-geting the IE1 RNA transcript. Furthermore, extraction of viral DNAinvolves exposure of infected cells to a lysis buffer containing non-ionic detergents and proteinase K for 1 h at 56 ◦C; subsequently(following dilution), inactivation of proteinase K necessitates sam-ple heating for 5 min at 95 ◦C [27,28]. These steps can be a hindranceto throughput. In contrast, preparation of cell lysates using ourapproach involves cell exposure to lysis buffer for 2 min at roomtemperature; the resulting cell lysates can be directly used in RT-qPCR without dilution or other processing steps.

Other existing high-throughput cell-based HCMV assays in com-mon use rely on endpoint assessment based on immunostaining forcells expressing viral antigens [12,13,29,30] or measuring the activ-ity of a reporter gene product (fluorescent protein or luciferase)driven by a recombinant virus [14,15,31,32]. Each has advantagesas well as drawbacks. Immunostaining involves many steps, whichcan complicate adaptation to full automation. Moreover, optimiza-tion of cell fixation/permeabilization may not be trivial [30], andstaining characteristics can be dependent on cell type [12]. Gener-ation of recombinant reporter viruses can be cumbersome due tothe large size of the HCMV genome and thus may not be conduciveto studies requiring the use of numerous HCMV strains.

Our RT-qPCR-based neutralization assay for HCMV has severaladvantages. The generation of lysate samples can be achieved instraightforward steps, and the assembly of downstream reactionscan be easily automated. The dynamic range of RT-qPCR contributesto assay robustness in terms of variation in input virus dose (Fig. 5).The inherent sensitivity of PCR coupled with choosing an abundantimmediate-early gene transcript as the target allows considerableshortening of assay turn-around time with endpoint assessmentpossible as early as 6 h post-infection. The HCMV IE1 gene may besufficiently conserved such that our primers may be applicable to

many HCMV strains [33]. In any case, designing suitable primersde novo, if the need arises (for other strains or other viral genetranscript targets), is usually straightforward. In addition to a muta-tion in UL131, AD169 carries other genomic alterations secondary
Page 7: RT-qPCR-based microneutralization assay for human ... · RT-qPCR-based microneutralization assay for human cytomegalovirus using fibroblasts and epithelial cells Xiao ... 2.3. RT-qPCR-based

7 ine 33

tbrfmf

Hitnte

IFlwliimhlHgga

iesnpsm

5

aieHmttbaar

C

A

(oOgt

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

260 X. Wang et al. / Vacc

o adaptation in fibroblasts that may influence its neutralizationehavior (for example, AD169 contains a large deletion in the ULb’egion [34,35]); we intend to expand the scope of our analysis inuture studies to include clinical isolates. Finally, assay establish-

ent in new laboratories would be facilitated by distribution ofrozen RNA reagents that can serve as calibration standards.

Cost may be a constraint for throughput using our approach.owever, we have actively pursued measures to mitigate lim-

tations arising from cost. For example, we have demonstratedhe feasibility of using a simple in-house lysis buffer as an alter-ative to commercial reagents (Supplemental Fig. 2) [17]. Thus,he per-sample cost of cell-lysate preparation can be essentiallyliminated.

We observed that human sera positive for HCMV-specificgG have substantially higher neutralizing activity (∼47-fold;ig. 4 and Supplemental Table 1) when measured using epithe-ial cells compared with using fibroblasts. This is in agreement

ith prior studies [29,30,36,37]. The likely mechanism under-ying this phenomenon is the cell-type dependent differencen HCMV entry pathway. Recent data suggest that antibod-es directed against the gH/gL/pUL128/pUL130/pUL131 complex

ay comprise the majority of neutralizing activity in HCMVyperimmune globulin preparations when assayed using epithe-

ial/endothelial cells [38]. It is notable that most investigationalCMV vaccines evaluated in humans thus far have not included theH/gL/pUL128/pUL130/pUL131 complex in their repertoire of anti-ens [11]. Targeting the gH/gL/pUL128/pUL130/pUL131 complex is

recent and promising vaccination strategy [39–43].High-throughput assays to measure HCMV-neutralizing activ-

ty in both epithelial/endothelial cells and fibroblasts would bessential in support of vaccine development. Our assay may betraightforward enough such that transfer across laboratories mayot be overly burdensome. Assay transferability would be desirablearticularly in early phases of development in which generation oferoepidemiology data and evaluation of vaccine immunogenicityay be conducted by multiple independent investigators.

. Conclusion

We have demonstrated the feasibility of using an endpointssessment based on RT-qPCR performed on crude lysates fromnfected cells to measure neutralizing activity against HCMV in bothpithelial cells and fibroblasts. Neutralization titers associated withCMV IgG positive human sera were substantially higher wheneasured using epithelial cells compared with using fibroblasts,

hus highlighting the need for an assay platform flexible enougho accommodate multiple cell types. Vaccine development maye facilitated by high-throughput assays, such as our own, thatre capable of measuring cell-type dependent HCMV-neutralizingctivity while maintaining desirable features in terms of simplicity,apidity, robustness, flexibility, and transferability.

onflict of interest

The authors report no conflict of interest.

cknowledgments

This work was supported by FDA intramural research fundsCBER Targeted Funding for Modernizing Science and FDA Officef Minority Health Challenge Grant). X.W. was supported by anak Ridge Institute for Science and Education Fellowship. We arerateful to Evi Struble and Andrew M. Lewis Jr. for comments onhe manuscript.

[

[

[

(2015) 7254–7261

Appendix A. Supplementary data

Supplementary data associated with this article can be found, inthe online version, at http://dx.doi.org/10.1016/j.vaccine.2015.10.110.

References

[1] Boppana SB, Ross SA, Fowler KB. Congenital cytomegalovirus infection: clinicaloutcome. Clin Infect Dis 2013;57(Suppl 4):S178–81.

[2] Griffiths PD. Burden of disease associated with human cytomegalovirusand prospects for elimination by universal immunisation. Lancet Infect Dis2012;12:790–8.

[3] Manicklal S, Emery VC, Lazzarotto T, Boppana SB, Gupta RK. The silent globalburden of congenital cytomegalovirus. Clin Microbiol Rev 2013;26:86–102.

[4] Bate SL, Dollard SC, Cannon MJ. Cytomegalovirus seroprevalence in the UnitedStates: the National Health and Nutrition Examination Surveys, 1988–2004.Clin Infect Dis 2010;50:1439–47.

[5] Stratton K, Durch J, Lawrence R. Vaccines for the 21st century: a tool for decisionmaking. Washington, D.C.: National Academy Press; 2001.

[6] Nigro G, Adler SP, La Torre R, Best AM, Congenital Cytomegalovirus Col-laborating Group. Passive immunization during pregnancy for congenitalcytomegalovirus infection. N Engl J Med 2005;353:1350–62.

[7] Revello MG, Lazzarotto T, Guerra B, Spinillo A, Ferrazzi E, Kustermann A,et al. A randomized trial of hyperimmune globulin to prevent congenitalcytomegalovirus. N Engl J Med 2014;370:1316–26.

[8] Hahn G, Revello MG, Patrone M, Percivalle E, Campanini G, Sarasini A,et al. Human cytomegalovirus UL131-128 genes are indispensable for virusgrowth in endothelial cells and virus transfer to leukocytes. J Virol 2004;78:10023–33.

[9] Wang D, Shenk T. Human cytomegalovirus virion protein complex requiredfor epithelial and endothelial cell tropism. Proc Natl Acad Sci U S A2005;102:18153–8.

10] Wang D, Shenk T. Human cytomegalovirus UL131 open reading frame isrequired for epithelial cell tropism. J Virol 2005;79:10330–8.

11] Fu TM, An Z, Wang D. Progress on pursuit of human cytomegalovirus vac-cines for prevention of congenital infection and disease. Vaccine 2014;32:2525–33.

12] Abai AM, Smith LR, Wloch MK. Novel microneutralization assay for HCMV usingautomated data collection and analysis. J Immunol Methods 2007;322:82–93.

13] Andreoni M, Faircloth M, Vugler L, Britt WJ. A rapid microneutralizationassay for the measurement of neutralizing antibody reactive with humancytomegalovirus. J Virol Methods 1989;23:157–67.

14] Gardner TJ, Bolovan-Fritts C, Teng MW, Redmann V, Kraus TA, Sperling R,et al. Development of a high-throughput assay to measure the neutraliza-tion capability of anti-cytomegalovirus antibodies. Clin Vaccine Immunol2013;20:540–50.

15] Wang Z, Mo C, Kemble G, Duke G. Development of an efficient fluorescence-based microneutralization assay using recombinant human cytomegalovirusstrains expressing green fluorescent protein. J Virol Methods 2004;120:207–15.

16] Gerna G, Percivalle E, Sarasini A, Baldanti F, Campanini G, Revello MG. Rescue ofhuman cytomegalovirus strain AD169 tropism for both leukocytes and humanendothelial cells. J Gen Virol 2003;84:1431–6.

17] Shatzkes K, Teferedegne B, Murata H. A simple, inexpensive method for prepar-ing cell lysates suitable for downstream reverse transcription quantitative PCR.Sci Rep 2014;4:4659.

18] Arvidsson S, Kwasniewski M, Riano-Pachon DM, Mueller-Roeber B.QuantPrime—a flexible tool for reliable high-throughput primer designfor quantitative PCR. BMC Bioinformatics 2008;9:465.

19] Varada JC, Teferedegne B, Crim RL, Mdluli T, Audet S, Peden K, et al. A neu-tralization assay for respiratory syncytial virus using a quantitative PCR-basedendpoint assessment. Virol J 2013;10:195.

20] Teferedegne B, Lewis Jr AM, Peden K, Murata H. Development of a neutralizationassay for influenza virus using an endpoint assessment based on quantitativereverse-transcription PCR. PLoS One 2013;8:e56023.

21] Chambers J, Angulo A, Amaratunga D, Guo H, Jiang Y, Wan JS, et al. DNA microar-rays of the complex human cytomegalovirus genome: profiling kinetic classwith drug sensitivity of viral gene expression. J Virol 1999;73:5757–66.

22] Bogner E. Human cytomegalovirus terminase as a target for antiviralchemotherapy. Rev Med Virol 2002;12:115–27.

23] Klasse PJ, Sattentau QJ. Occupancy and mechanism in antibody-mediated neu-tralization of animal viruses. J Gen Virol 2002;83:2091–108.

24] Murata H, Teferedegne B, Lewis Jr AM, Peden K. A quantitative PCR assay forSV40 neutralization adaptable for high-throughput applications. J Virol Meth-ods 2009;162:236–44.

25] Ho YK, Xu WT, Too HP. Direct quantification of mRNA and miRNA from celllysates using reverse transcription real time PCR: a multidimensional analysisof the performance of reagents and workflows. PLoS One 2013;8:e72463.

26] Van Peer G, Mestdagh P, Vandesompele J. Accurate RT-qPCR gene expressionanalysis on cell culture lysates. Sci Rep 2012;2:222.

27] Tremblay S, Dansereau N, Balsitis S, Franti M, Lamorte L. Development of ahigh-throughput human cytomegalovirus quantitative PCR cell-based assay. JVirol Methods 2014;195:67–71.

Page 8: RT-qPCR-based microneutralization assay for human ... · RT-qPCR-based microneutralization assay for human cytomegalovirus using fibroblasts and epithelial cells Xiao ... 2.3. RT-qPCR-based

ine 33

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

X. Wang et al. / Vacc

28] Schnepf N, Boiteau N, Petit F, Alain S, Sanson-Le Pors MJ, Mazeron MC. Rapiddetermination of antiviral drug susceptibility of human cytomegalovirus byreal-time PCR. Antiviral Res 2009;81:64–7.

29] Gerna G, Sarasini A, Patrone M, Percivalle E, Fiorina L, Campanini G, et al.Human cytomegalovirus serum neutralizing antibodies block virus infection ofendothelial/epithelial cells, but not fibroblasts, early during primary infection.J Gen Virol 2008;89:853–65.

30] Tang A, Li F, Freed DC, Finnefrock AC, Casimiro DR, Wang D, et al. A novel high-throughput neutralization assay for supporting clinical evaluations of humancytomegalovirus vaccines. Vaccine 2011;29:8350–6.

31] Straschewski S, Warmer M, Frascaroli G, Hohenberg H, Mertens T, Win-kler M. Human cytomegaloviruses expressing yellow fluorescent fusionproteins—characterization and use in antiviral screening. PLoS One2010;5:e9174.

32] He R, Sandford G, Hayward GS, Burns WH, Posner GH, Forman M, et al. Recom-binant luciferase-expressing human cytomegalovirus (CMV) for evaluation ofCMV inhibitors. Virol J 2011;8:40.

33] Zweygberg Wirgart B, Brytting M, Linde A, Wahren B, Grillner L. Sequence vari-ation within three important cytomegalovirus gene regions in isolates fromfour different patient populations. J Clin Microbiol 1998;36:3662–9.

34] Murphy E, Yu D, Grimwood J, Schmutz J, Dickson M, Jarvis MA, et al. Coding

potential of laboratory and clinical strains of human cytomegalovirus. Proc NatlAcad Sci U S A 2003;100:14976–81.

35] Cha TA, Tom E, Kemble GW, Duke GM, Mocarski ES, Spaete RR. Humancytomegalovirus clinical isolates carry at least 19 genes not found in laboratorystrains. J Virol 1996;70:78–83.

[

(2015) 7254–7261 7261

36] Cui X, Meza BP, Adler SP, McVoy MA. Cytomegalovirus vaccines fail to induceepithelial entry neutralizing antibodies comparable to natural infection. Vac-cine 2008;26:5760–6.

37] Wang D, Li F, Freed DC, Finnefrock AC, Tang A, Grimes SN, et al. Quantita-tive analysis of neutralizing antibody response to human cytomegalovirus innatural infection. Vaccine 2011;29:9075–80.

38] Fouts AE, Chan P, Stephan JP, Vandlen R, Feierbach B. Antibodies againstthe gH/gL/UL128/UL130/UL131 complex comprise the majority of theanti-cytomegalovirus (anti-CMV) neutralizing antibody response in CMVhyperimmune globulin. J Virol 2012;86:7444–7.

39] Wen Y, Monroe J, Linton C, Archer J, Beard CW, Barnett SW, et al. Humancytomegalovirus gH/gL/UL128/UL130/UL131A complex elicits potently neu-tralizing antibodies in mice. Vaccine 2014;32:3796–804.

40] Kabanova A, Perez L, Lilleri D, Marcandalli J, Agatic G, Becattini S, et al. Antibody-driven design of a human cytomegalovirus gHgLpUL128L subunit vaccine thatselectively elicits potent neutralizing antibodies. Proc Natl Acad Sci U S A2014;111:17965–70.

41] Wussow F, Chiuppesi F, Martinez J, Campo J, Johnson E, Flechsig C, et al. Humancytomegalovirus vaccine based on the envelope gH/gL pentamer complex. PLoSPathog 2014;10:e1004524.

42] Fu TM, Wang D, Freed DC, Tang A, Li F, He X, et al. Restoration of viral epithelial

tropism improves immunogenicity in rabbits and rhesus macaques for a wholevirion vaccine of human cytomegalovirus. Vaccine 2012;30:7469–74.

43] Freed DC, Tang Q, Tang A, Li F, He X, Huang Z, et al. Pentameric complex ofviral glycoprotein H is the primary target for potent neutralization by a humancytomegalovirus vaccine. Proc Natl Acad Sci U S A 2013;110:E4997–5005.