You are on page 1of 19

HLA-A02:01−Restricted Epitopes Identified

from the Herpes Simplex Virus Tegument


Protein VP11/12 Preferentially Recall
Polyfunctional Effector Memory CD8 + T
This information is current as Cells from Seropositive Asymptomatic
of September 19, 2020.
Individuals and Protect Humanized
HLA-A*02:01 Transgenic Mice against
Ocular Herpes
Ruchi Srivastava, Arif A. Khan, Doran Spencer, Hawa

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


Vahed, Patricia P. Lopes, Nhi Thi Uyen Thai, Christine
Wang, Thanh T. Pham, Jiawei Huang, Vanessa M. Scarfone,
Anthony B. Nesburn, Steven L. Wechsler and Lbachir
BenMohamed
J Immunol 2015; 194:2232-2248; Prepublished online 23
January 2015;
doi: 10.4049/jimmunol.1402606
http://www.jimmunol.org/content/194/5/2232

Supplementary http://www.jimmunol.org/content/suppl/2015/01/22/jimmunol.140260
Material 6.DCSupplemental
References This article cites 76 articles, 37 of which you can access for free at:
http://www.jimmunol.org/content/194/5/2232.full#ref-list-1

Why The JI? Submit online.


• Rapid Reviews! 30 days* from submission to initial decision
• No Triage! Every submission reviewed by practicing scientists
• Fast Publication! 4 weeks from acceptance to publication
*average

Subscription Information about subscribing to The Journal of Immunology is online at:


http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2015 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2015 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

HLA-A02:01–Restricted Epitopes Identified from the Herpes


Simplex Virus Tegument Protein VP11/12 Preferentially
Recall Polyfunctional Effector Memory CD8+ T Cells from
Seropositive Asymptomatic Individuals and Protect
Humanized HLA-A*02:01 Transgenic Mice against
Ocular Herpes

Ruchi Srivastava,* Arif A. Khan,* Doran Spencer,* Hawa Vahed,* Patricia P. Lopes,*
Nhi Thi Uyen Thai,* Christine Wang,* Thanh T. Pham,* Jiawei Huang,*

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


Vanessa M. Scarfone,† Anthony B. Nesburn,* Steven L. Wechsler,*,‡,x,{ and
Lbachir BenMohamed*,‖,#
The HSV type 1 tegument virion phosphoprotein (VP) 11/12 (VP11/12) is a major Ag targeted by CD8+ T cells from HSV-
seropositive individuals. However, whether and which VP11/12 epitope–specific CD8+ T cells play a role in the “natural” pro-
tection seen in seropositive healthy asymptomatic (ASYMP) individuals (who have never had clinical herpes disease) remain to be
determined. In this study, we used multiple prediction computer-assisted algorithms to identify 10 potential HLA-A*02:01–
restricted CD8+ T cell epitopes from the 718-aa sequence of VP11/12. Three of 10 epitopes exhibited high-to-moderate binding
affinity to HLA-A*02:01 molecules. In 10 sequentially studied HLA-A*02:01–positive and HSV-1–seropositive ASYMP individ-
uals, the most frequent, robust, and polyfunctional effector CD8+ T cell responses, as assessed by a combination of tetramer
frequency, granzyme B, granzyme K, perforin, CD107a/b cytotoxic degranulation, IFN-g, and multiplex cytokines assays, were
predominantly directed against three epitopes: VP11/1266–74, VP11/12220–228, and VP11/12702–710. Interestingly, ASYMP individ-
uals had a significantly higher proportion of CD45RAlowCCR7lowCD44highCD62LlowCD27lowCD28lowCD8+ effector memory CD8+
T cells (TEMs) specific to the three epitopes, compared with symptomatic individuals (with a history of numerous episodes of
recurrent ocular herpetic disease). Moreover, immunization of HLA-A*02:01 transgenic mice with the three ASYMP CD8+ TEM
cell epitopes induced robust and polyfunctional epitope-specific CD8+ TEM cells that were associated with a strong protective
immunity against ocular herpes infection and disease. Our findings outline phenotypic and functional features of protective HSV-
specific CD8+ T cells that should guide the development of an effective T cell–based herpes vaccine. The Journal of Immunology,
2015, 194: 2232–2248.

H
erpes simplex virus type 1 (HSV-1) infection is wide- virus establishes latency in the neurons of sensory ganglia after
spread in human populations (1–5). A staggering 1 bil- a primary infection (7–10). The majority of HSV-seropositive
lion individuals worldwide currently carry the virus that individuals are asymptomatic (ASYMP) (7–10). They do not ex-
causes a wide range of diseases throughout their life (1–5). perience any recurrent herpetic disease (e.g., cold sore, ocular or
Complications range from mild, such as cold sores and genital genital herpes) even though the virus spontaneously reactivates
lesion, to serious, such as permanent brain damage from en- from latency and sheds multiple times each year in their body
cephalitis in adults and neonates, and blinding corneal inflam- fluids (i.e., tears, saliva, nasal and vaginal secretions) (2, 3, 11,
mation (5, 6). HSV infections are prevalent and permanent, as the 12). In contrast, a small proportion of HSV-seropositive individ-

*Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, Address correspondence and reprint requests to Dr. Lbachir BenMohamed, Labora-
University of California Irvine, School of Medicine, Irvine, CA 92697; †Stem Cell tory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, Hewitt
Research Center, University of California Irvine, Irvine, CA 92697; ‡Virology Research, Hall, Room 2032, 843 Health Sciences Road, University of California Irvine, Irvine,
Gavin Herbert Eye Institute and Department of Ophthalmology, University of California CA 92697-4390. E-mail address: Lbenmoha@uci.edu
Irvine, School of Medicine, Irvine, CA 92697; xDepartment of Microbiology and Mo-
The online version of this article contains supplemental material.
lecular Genetics, University of California Irvine, School of Medicine, Irvine, CA 92697;
{
Center for Virus Research, University of California Irvine, Irvine, CA 92697; ‖Depart- Abbreviations used in this article: ASYMP, asymptomatic; GzmB, granzyme B;
ment of Molecular Biology and Biochemistry, University of California Irvine, School of GzmK, granzyme K; HSK, herpetic stromal keratitis; HSV-1, HSV type 1; IRB,
Medicine, Irvine, CA 92697; and #Institute for Immunology, University of California Institutional Review Board; MFI, mean fluorescence intensity; PFN, perforin; RS,
Irvine, School of Medicine, Irvine, CA 92697 rabbit skin; SYMP, symptomatic; TAP, transporter associated with Ag transport; TCM,
central memory CD8+ T cell; TEM, effector memory CD8+ T cell; Tg, transgenic; VP,
Received for publication October 14, 2014. Accepted for publication December 16,
virion phosphoprotein; VP11/12, virion phosphoprotein 11 and 12.
2014.
This work was supported by the Public Health Service (Research Grants EY14900, Copyright Ó 2015 by The American Association of Immunologists, Inc. 0022-1767/15/$25.00
EY019896, EY024618, EY013191, 1R56AI098985, and 1R56AI093133), the National
Institutes of Health, the Discovery Eye Foundation, The Discovery Center for Eye
Research, and a Research to Prevent Blindness Challenge grant.

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1402606
The Journal of Immunology 2233

uals are symptomatic (SYMP) and experience endless recurrences Table I. Cohorts of HLA-A*02:01–positive, HSV-seropositive SYMP
of herpetic disease, usually multiple times a year (13, 14), often and ASYMP individuals enrolled in this study
requiring continuous antiviral therapy (i.e., acyclovir and deriva-
tives). Notably, in some HSV-1–seropositive SYMP individuals, Subject-Level Characteristics All Subjects (n = 693)

sporadic reactivation of the virus from latency and corneal rein- Sex, no. (%)
fection can cause blinding recurrent herpetic stromal keratitis Female 355 (52)
(HSK), a T cell–mediated immunopathological lesion of the cor- Male 338 (48)
Race, no. (%)
nea (4, 5, 15). Therapeutic manipulation of the immune system White 488 (71)
(immunotherapy) is an attractive strategy to impact SYMP dis- Nonwhite 205 (29)
ease, HSV-1 shedding, and ultimately, HSV-1 transmission in the Median age (range), y 32 (18–65)
community (7–10). For this to occur, one must first identify the HSV status, no. (%)
HSV-1 Ags/epitopes involved in the apparent protection seen in HSV-1+ 261 (38)
HSV-2+ 323 (46)
seropositive ASYMP individuals, who appear to immunologically HSV-1+ and HSV-2+ 23 (3)
contain infection and disease. HSV2 86 (12)
Among the 84+ HSV-1–encoded protein Ags, perhaps the least HLA, no. (%)
characterized immunologically are the tegument proteins, which HLA-A*02:01+ 330 (52)
HLA-A*02:012 257 (48)
are located between the capsid and the envelope (7–10). We re- Herpes disease status, no. (%)
cently focused on identifying protective T cell epitopes from HSV- ASYMP 575 (94)
1 and HSV-2 tegument proteins because: 1) vaccine clinical trials SYMP 32 (6)

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


that have concentrated during the last two decades on HSV en-
velope glycoproteins (mainly gB and gD) have failed to produce
protective immunity against herpes infection and disease (16, 17); were females, and 338 were males. Among these, a cohort of 261 im-
2) tegument proteins are injected into the cytoplasm of host cells munocompetent individuals, with an age range of 18–65 (median, 32) y,
who are seropositive for HSV-1 and seronegative for HSV-2, were enrolled
during viral entry, resulting in an immediate processing by APC
in this study. All patients were negative for HIV, HBV, and had no history
and T cell activation shortly postinfection (7–10); 3) the abundant of immunodeficiency. A total of 607 patients were HSV-1, HSV-2, or
virion phosphoprotein (VP) 11 and 12 (VP11/12) tegument pro- HSV-1/HSV-2 seropositive, among which 575 patients were healthy and
tein, encoded by the UL46 gene, is an excellent target for pro- ASYMP (individuals who, in the absence of therapy, have never had any re-
teolysis, an event that precedes T cell epitope presentation in current herpes disease, ocular, genital or elsewhere, based on their self-report
and physician examination; even a single episode of any herpetic disease in
association with HLA molecules (18); and 4) using a proteome- their entire life excludes the individual from this group). The remaining 32
wide approach, covering all 84+ HSV-1–encoded proteins, we and patients were defined as HSV-seropositive SYMP, having suffered frequent and
others have found HSV tegument proteins in general, and the severe recurrent genital, ocular, and/or orofacial lesions, with 2 patients having
abundant VP11/12 tegument in particular, to be major targets of had clinically well-documented repetitive HSK (1 patient with ∼20 episodes
over 20 y that necessitated several corneal transplantations).
CD8+ T cells from HSV-seropositive healthy ASYMP individuals
Signs of recurrent disease in SYMP patients were defined as herpetic lid
(who have never had clinical herpes) (7–10, 19–22). However, the lesions, herpetic conjunctivitis, dendritic or geographic keratitis, stromal
repertoire of HSV-1 VP11/12 epitopes that induce protective CD8+ keratitis, and iritis consistent with HSK, with one or more episodes per year
T cells in ASYMP individuals and the nature of VP11/12 epitope- for the past 2 y. However, at the time of blood collection, SYMP patients had
specific CD8+ T cells involved in protective immunity remain to be no recurrent disease (other than corneal scarring) and had no recurrences
during the previous 30 d. They have no ocular disease other than HSK, have
determined. This information is necessary for the successful design no history of recurrent genital herpes, and are HSV-1 seropositive and HSV-
of an effective T cell–based therapeutic vaccine strategy. 2 seronegative. Because the spectrum of recurrent ocular herpetic disease is
In this study, we identify three novel protective ASYMP epitopes wide, our emphasis was mainly on the number of recurrent episodes, not on the
from VP11/12 tegument protein that are strongly recognized by CD8+ severity of the recurrent disease. No attempt was made to assign specific T cell
epitopes to specific severity of recurrent lesions. Patients were also excluded
T cells from HSV-seropositive healthy ASYMP individuals (VP11/
if they: 1) were pregnant or breastfeeding, or 2) were on acyclovir or other
1266–74, VP11/12220–228, and VP11/12702–710). A significantly higher related antiviral drugs or any immunosuppressive drugs at the time of blood
proportion of HSV-1 VP11/12 epitope–specific effector memory sample collection. SYMP and ASYMP groups were matched for age, sex,
CD8+ T cells (TEMs) were detected in ASYMP individuals com- serological status, and race. Eighty-six healthy control individuals were se-
pared with SYMP patients. Frequent, robust, and polyfunctional ronegative for both HSV-1 and HSV-2 and had no history of ocular herpes,
genital lesions, or orofacial herpes disease. All subjects were enrolled at the
effector CD8+ T cell responses were directed predominantly against University of California, Irvine under Institutional Review Board (IRB)–
these three epitopes in ASYMP individuals. Moreover, immuniza- approved protocols (IRB 2003-3111 and IRB 2009-6963). A written informed
tion of HLA Tg mice with the three VP11/12 CD8+ T cell epitopes consent was received from all participants before inclusion in the study.
induced strong protective immunity against ocular herpes infection
and disease, associated with strong and polyfunctional VP11/12 Bioinformatics analyses
epitope-specific CD8+ TEM cell responses. Altogether, our findings HSV-1 VP11/12 open reading frames used in this study were from strain 17
identify previously unknown protective epitopes from the VP11/12 (National Center for Biotechnology Information accession no. P 10230).
tegument protein, and delineate quantitative and qualitative features The 718-aa sequence (Supplemental Fig. 1) of the VP11/12 protein was
screened for HLA-A2.1–restricted epitopes using different computational
of a protective HSV-specific CD8+ T cell response that should be algorithms as previously described: BIMAS software (Bioinformatics and
taken into consideration during herpes vaccine development. Molecular Analysis Section, National Institutes of Health, Washington,
DC; http://www-bimas.cit.nih.gov/molbio/hla_bind/) and the SYFPEITHI
algorithm (http://www.syfpeithi.de) (1, 2). Potential cleavage sites for
Materials and Methods human proteasome were also used in this screening and were identified
Human study population using NetChop 3.0 (http://www.cbs.dtu.dk/services/NetChop/) and the
MHC Pathway database (http://www.mhc-pathway.net) (1, 2).
During the last decade (i.e., January 2003 to September 2014), we have
recruited and screened 693 individuals for HSV-1 and HSV-2 seropositivity Peptide synthesis
at Gavin Herbert Eye Institute and University of California Irvine Institute
for Clinical and Translational Science (Table I). A total of 488 individuals To determine potential vaccine candidates, we synthesized 10 HLA-A*0201
were white, 205 were nonwhite (African, Asian, Hispanic, and others), 355 binding peptides of HSV-1 (strain 17) VP11/12 with high estimated half-
2234 VP11/12-SPECIFIC CD8+ T CELLS IN SYMPTOMATIC HERPES

time of dissociation (T1/2) by Magenex (San Diego, CA) on a 9050 Pep Dickinson). The acquired data were analyzed with FlowJo software (BD
Synthesizer Instrument using solid-phase peptide synthesis and standard Biosciences, San Jose, CA).
9-fluorenylmethoxy carbonyl technology (PE Applied Biosystems, Foster
City, CA). Stock solutions were made at 1 mg/ml in PBS. All peptides Flow cytometry analysis
were aliquoted and were stored at 220˚C until assayed.
PBMCs were analyzed by flow cytometry after staining with fluorochrome-
conjugated human-specific mAbs. Anti-human Abs were CD3 (clone SK7)
Cell lines PE-Cy7, CD44 (clone G44-26) A700, CD8 (clone SK1) allophycocyanin-
The T2 (174 3 CEM.T2) mutant hybrid cell line derived from the T Cy7, CCR7 (clone 150503) Alexa Fluor 700, CD28 (clone CD28.2)
lymphoblast cell line CEM was obtained from the ATCC. T2 cells lack allophycocyanin, CD45RO PE-Cy5, CD45RA FITC, CD62L allophyco-
the functional transporter associated with Ag transport (TAP) hetero- cyanin, IFN-g Alexa Fluor 647, T-bet (clone O4-46) Alexa Fluor 488 (BD
dimer and fail to express normal amounts of HLA-A*0201 on the cell Pharmingen); granzyme B (GzmB; clone GB11) A647, CD107a (clone
surface. HLA-A*0201 surface expression can be stabilized with exogenous H4A3) FITC, CD107b (clone H4B4) FITC (BioLegend); CD27 (clone
loading of peptide that are able to bind to the MHC class I molecule. The 0323) Alexa Fluor 700, granzyme K (GzmK; clone G3H69) PerCP eFlour
T2 cell line was maintained in IMDM (ATCC) supplemented with 10% 710, perforin (PFN; clone d69) FITC, Eomes (clone WD1928) eFluor
heat-inactivated FCS and 100 U penicillin/ml, 100 U streptomycin/ml 660 (eBioscience). Draining lymph nodes and splenocytes were ana-
(Sigma-Aldrich). lyzed by flow cytometry after staining with fluorochrome-conjugated
mouse-specific mAbs. Anti-mouse Abs were CD8 (clone 53-6.7) PE-
Stabilization of HLA-A*0201 on class I HLA–transfected B 3 Cy7, CD44 (clone IM7) allophycocyanin-Cy7, CD62L (MEL-14) A700,
CD107a (clone 1D4B) FITC, CD107b (clone Ha1/29) FITC, IFN-g (clone
T hybrid cell lines XMG1.2) A700 (BD Pharmingen); GzmB (clone GB11) A647 (Bio-
To determine whether synthetic peptides could stabilize HLA-A*0201 Legend).
molecule expression on the T2 cell surface, we examined peptide-inducing Surface-staining mAbs against various cell markers were added to a total

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


HLA-A*0201 upregulation on T2 cells according to a protocol described of 1 3 106 PBMCs in PBS containing 1% FBS and 0.1% sodium azide
previously (23, 24). T2 cells (3 3 105/well) were incubated with different (FACS buffer) for 45 min at 4˚C. After washing with FACS buffer, cells
concentrations of individual VP11/12 peptide (as indicated in Fig. 1) in 48- were permeabilized for 20 min on ice using the Cytofix/Cytoperm Kit (BD
well plates for 18 h at 26˚C. Cells were then incubated at 37˚C for 3 h in Biosciences) and then washed twice with Perm/Wash Buffer (BD Biosci-
the presence of 0.7 ml/ml BD GolgiStop, to block cell-surface expression ence). Intracellular cytokine staining mAbs were then added to the cells
of newly synthesized HLA-A*0201 molecules, and human b2 micro- and incubated for 45 min on ice in the dark. Cells were washed again with
globulin (1 mg/ml). The cells were washed with FACS buffer (1% BSA and Perm/Wash and FACS buffer, and fixed in PBS containing 2% parafor-
0.1% sodium azide in PBS) and stained with anti–HLA-A2.1–specific maldehyde (Sigma-Aldrich, St. Louis, MO). For each sample, 200,000
mAb (clone BB7.2; BD Pharmingen) at 4˚C for 30 min. After incubation, total events were acquired on BD LSRII Fortessa. Ab capture beads (BD
the cells were washed with FACS buffer, fixed with 1% paraformaldehyde Biosciences) were used as individual compensation tubes for each fluo-
in PBS, and analyzed by flow cytometry using an LSRII (Becton Dick- rophore in the experiment. To define positive and negative populations, we
inson). The acquired data were analyzed with FlowJo software (BD Bio- used fluorescence minus controls for each fluorophore used in this study,
sciences, San Jose, CA). Expression was measured by LSRII flow when initially developing staining protocols. In addition, we further opti-
cytometer, and mean fluorescence intensity (MFI) was recorded. Percent mized gating by examining known negative cell populations for back-
MFI increase was calculated as follows: Percent MFI increase = (MFI with ground level expression. The gating strategy was similar to that used in our
the given peptide 2 MFI without peptide)/(MFI without peptide) 3 100. previous work (2). In brief, we gated on single cells, dump2 cells, viable
Each experiment was performed three times, and means 6 SD values were cells (Aqua Blue), lymphocytes, CD3+ cells, and CD8+ cells before the
calculated. final gating on functional cells. For epitope-specific cells, VP11/12 tetra-
mer–specific CD8+ T cells were gated for different markers used in cos-
taining. Data analysis was performed using FlowJo version 9.7.5 (TreeStar,
HSV-specific serotyping through ELISA Ashland, OR). Statistical analyses were done using GraphPad Prism ver-
The sera collected from random donors were tested for anti-HSV Abs. sion 5 (La Jolla, CA).
ELISA was performed on sterile 96-well, flat-bottom microplates. The
plates were coated with the HSV-1 Ag in coating buffer overnight at 4˚C. mRNA isolation and real-time RT-PCR analysis
The next day plates were washed with PBS-1% Tween 20 (PBST) five Total RNA was isolated from fresh PBMCs using the Direct-zol RNA
times. Nonspecific binding was blocked by incubating them with a 5% MiniPrep kit (Zymo Research) and reverse transcribed with High-Capacity
solution of skimmed milk in PBS (200 ml/well) at 4˚C for 1 h at room cDNA Reverse Transcription kit (Applied Biosystems) according to the
temperature. The microplates were washed three times with PBS-Tween manufacturer’s instructions. Quantitative real-time PCR was carried out on
and incubated with various sera at 37˚C for 2 h. After five washes, bio- a Rotor-Gene 3000 (Corbett Research) using the LightCycler 480 SYBR
tinylated rabbit anti-human IgG, diluted 1:20,000 with PBST, was used as
Green 1 Master (Roche). The data were normalized to GAPDH for tran-
the secondary Ab and incubated at 37˚C for 2 h. After five washes,
scription factor analysis. Relative mRNA expression levels were calculated
streptavidin was added at a 1:5000 dilution and incubated for 30 min at using the following formula:
room temperature. After five additional washes, the color was developed
by adding 100 ml TMB substrate. The mixture was incubated for 5–15 min DCPtarget ðcontrol2sampleÞ
at room temperature in the absence of light. The reaction was terminated Etarget
ratio ¼ DCPref ðcontrol2sampleÞ
by adding 1 M H2SO4. The absorbance was measured at 450 nm. Eref

PBMC isolation Data are expressed as fold increase.

A total of 100 ml of an individual’s blood was drawn into yellow-top Tetramer/VP11/12 peptide staining
Vacutainer tubes (Becton Dickinson). The PBMCs were isolated by gra-
Fresh PBMCs were analyzed for the frequency of CD8+ T cells recognizing
dient centrifugation using leukocyte separation medium (Cellgro). The
the VP11/12 peptide–tetramer complexes, as we previously described (7–
cells were washed in PBS and resuspended in complete culture medium
10). The cells were incubated with VP11/12 peptide–tetramer complex for
consisting of RPMI 1640 medium containing 10% FBS (Bio-Products,
30–45 min at 37˚C. The cell preparations were then washed with FACS
Woodland, CA) supplemented with 13 penicillin/L-glutamine/streptomycin,
buffer and stained with FITC-conjugated anti-human CD8 mAb (BD
13 sodium pyruvate, 13 nonessential amino acids, and 50 mM 2-ME (Life
Pharmingen). The cells were washed and fixed with 1% paraformaldehyde
Technologies, Rockville, MD). Aliquots of freshly isolated PBMCs were
in PBS. The cells were then acquired on a BD LSR II, and data were
also cryopreserved in 90% FCS and 10% DMSO in liquid nitrogen for
analyzed using FlowJo version 9.5.6 (Tree Star).
future testing.
CD107 cytotoxicity assay
HLA-A2 typing
To detect VP11/12-specific cytolytic CD8+ T cells in PBMCs and cornea
The HLA-A2 status was confirmed by PBMC staining with 2 ml anti–HLA- cells, we performed intracellular CD107a/b cytotoxicity assay as described
A2 mAb, (clone BB7.2; BD Pharmingen), at 4˚C for 30 min. The cells by Betts et al. (25, 26) with a few modifications. In brief, 1 3 106 PBMCs
were washed and analyzed by flow cytometry using an LSRII (Becton from patients, in addition to spleen cells, draining lymph node cells, and
The Journal of Immunology 2235

trigeminal ganglia cells from mice infected with HSV, were transferred Results
into 96-well V-bottom FACS plates (BD) in R10 medium and stimulated
In silico prediction of potential HLA-A*02:01–restricted T cell
with 10 different VP11/12 peptides (10 mg/ml) in the presence of anti–
CD107a-FITC and anti–CD107b-FITC (BD Pharmingen) and BD Golgi- epitopes from the HSV-1 VP11/12 protein
Stop (10 mg/ml) for 5–6 h at 37˚C. ConA (10 mg/ml; Sigma) and no The amino acid sequence of HSV-1 VP11/12 tegument protein
peptide were used as positive and negative controls, respectively. At the
end of the incubation period, the cells were harvested into separate tubes (strain 17) was screened for potential HLA-A*02:01–binding
and washed once with FACS buffer and then stained with PE-conjugated regions using BIMAS, SYFPEITHI, and MAPPP predictive com-
anti-human CD8 Ab for 30 min. Cells were fixed, permeabilized, and putational algorithms (1). The HLA-A*02:01 haplotype is preva-
stained with additional Abs against IFN-g using FIX/PERM and PERM/ lent in .50% of the world’s population irrespective of sex and
Wash solution (BD).
ethnicity (29). Based on these analyses, 10 potential peptide
HLA-A*0201 transgenic mice epitopes with high predicted affinity to HLA-A*0201 molecules were
selected (Table II). All 10 VP11/12 peptide epitopes shared the HLA-
HLA-A*02:01 transgenic (Tg) mice (previously described in Refs. 27–29) A*0201–binding motifs: leucine or valine at the second position and
were kindly provided by Dr. Francois Lemonier (Pasteur Institute, Paris,
France) and were bred at University of California Irvine. These mice rep-
a leucine, valine, methionine, or alanine at the ninth position. Based
resent the F1 generation resulting from a cross between HLA-A*02:01/Kb on the earlier computational algorithms, these VP11/12 peptides bear
Tg mice (expressing a chimeric gene consisting of the 1 and 2 domains putative antigenic and immunogenic HLA-A*0201–binding epitopes,
of HLA-A*02:01 and the 3 domain of H-2Kb) created on the BALB/c and thus are more likely to be less constrained than other parts of the
genetic background. The genotype of the HLA Tg mice used in this VP11/12 molecule, resulting in increased accessibility to proteolysis,
study was confirmed as HLA-A*02:01, the most common A*02 subtype,
supporting that the immunogenic peptide epitopes reported in this study are an event that precedes T cell epitope presentation in association with

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


likely presented by the HLA-A*02:01 molecule. All animal studies were HLA molecules (28, 32–36).
conducted in facilities approved by the Association for Assessment and
Accreditation of Laboratory Animal Care and according to Institutional Three VP11/12 epitopes bind and stabilize HLA-A*02:01
Animal Care and Use Committee–approved animal protocols (202-2372).
molecules on the surface of target cells
Virus production Peptides corresponding to the 10 highest probable VP11/12 epitope
HSV-1 (strain McKrae) was grown and titrated on rabbit skin (RS) cells. peptides were synthesized and experimentally tested for binding
UV-inactivated HSV-1 was generated as we described previously (3). HSV affinity to soluble HLA-A*0201 molecules (Table II, far right
inactivation was confirmed by the inability to produce plaques when tested column). The VP11/1266–74 and VP11/12197–205 peptides showed
on RS cells. highest affinity binding to HLA-A*0201 molecules (KD affinity
values of 81 and 83 nM, respectively). Peptides VP11/12702–710
Ocular challenge of humanized HLA Tg mice with HSV-1
and VP11/12220–228 showed medium binding affinity to soluble
Two groups of age-matched female HLA-A*02:01 Tg mice were chal- HLA-A*0201 molecules (KD of 313 and 709 nM, respectively).
lenged with 2 3 105 PFU of strain McKrae as eye drops. Control mice None of the remaining six peptides binds with significant affinity
were inoculated using mock samples of virus. After ocular challenge, mice
were monitored for ocular herpes infection and disease. Some of them to HLA-A*0201 molecules.
remained ASYMP (exhibited no symptoms or disease), whereas some of Cell-surface expression of HLA-A*02:01 molecules depends
them developed symptoms and were considered SYMP. primarily on peptide transport into the ER/Golgi by the TAP. Class
I-HLA–transfected B 3 T hybrid cell lines (T2 lines) are deficient
Immunization of HLA-A*02:01 Tg mice with VP11/12 in TAP but still express low amounts of MHC class I on the
immunodominant peptide epitopes surface of the cells (1). We next performed a stabilization assay of
Three groups of age-matched female HLA-A*02:01 Tg mice (n = 10 each) HLA-A*02:01 molecules on T2 cells. The T2 cell binding assay is
were immunized s.c. with the immunodominant CD8+ T cell human based upon the ability of peptides to stabilize the MHC class I
epitopes (VP11/1266–74, VP11/12220–228, and VP11/12702–710) delivered complex on the surface of the T2 cell line. Each of the 10 VP11/12
with the CD4+ T cell VP11/12129–143 epitope emulsified in CpG1826 ad-
juvant (GROUP 1), with the immunodominant CD8+ T cell human epi- peptides was tested at a concentration of 0.12, 0.6, 3, 15, and 30
topes (VP11/1266–74, VP11/12220–228, and VP11/12702–710) delivered with mM (Fig. 1). Of 10 VP11/12 peptides, VP11/12220–228 showed the
the CD4+ T cell PADRE epitope emulsified in CpG1826 adjuvant (GROUP highest binding affinity to HLA-A*02:01 molecules. VP11/12220–228
2), or with the CpG1826 adjuvant alone (MOCK) on days 0 and 21. All significantly increased levels of HLA-A*02:01 molecules on the
immunizations were carried out with 100 mM of each peptide.
surface of the T2 cells in a dose-dependent manner (p , 0.001).
Monitoring of ocular herpes infection and disease From the remaining nine peptides, VP11/12127–135, VP11/1266–74,
and VP11/12702–710 were medium binders to HLA-A*02:01
Animals were examined for signs of ocular disease by slit lamp. Clinical molecules (Fig. 1). Despite many attempts, the remaining six
assessments were made immediately before inoculation and on days 1, 3, 5, 7,
10, 14, and 21 thereafter. The examination was performed by investigators peptides produced no significant stabilization of HLA-A*02:01
blinded to the treatment regimen of the mice and scored according to a standard molecules on the surface of T2 cells (Fig. 1). Notably, the VP11/
0–4 scale: 0, no disease; 1, 25%; 2, 50%; 3, 75%; and 4, 100% staining, as 12197–205 peptide that appeared to bind to soluble HLA-A*02:01
previously described (30). To quantify replication and clearance of HSV-1 did not significantly increase the levels of HLA-A*02:01 mole-
from the eyes, we swabbed mice daily with moist, type 1 calcium alginate
cules on the surface of the T2 cells. Inversely, the VP11/12127–135
swabs. Swabs were placed in 1.0 ml titration media (Media 199, 2%
penicillin/streptomycin, 2% newborn calf serum) and frozen at 280˚C peptide that appeared to increase the levels of HLA-A*02:01
until titrated on RS cell monolayers, as described previously (30). Mice molecules on the surface of the T2 cells did not bind to soluble
were also examined for survival in a window of 30 d after challenge, as HLA-A*02:01 molecules.
we previously described (30). Altogether, the earlier results suggest that 3 of 10 potential
VP11/12 peptides, VP11/1266–74, VP11/12220–228, and VP11/
Statistical analyses
12702–710, bind with high affinity to soluble HLA-A*02:01 mole-
Data for each assay were compared by ANOVA and Student t test using cules and significantly increase the levels of HLA-A*02:01 mol-
GraphPad Prism version 5 (La Jolla, CA). Differences between the groups
ecules on the surface of the T2 cells. These three peptides might
were identified by ANOVA and multiple comparison procedures, as we
previously described (31). Data are expressed as the mean 6 SD. Results therefore lead to efficient functional presentation to HLA-A*02:01
were considered statistically significant at p , 0.05. molecules and stimulation of CD8+ T cells.
2236 VP11/12-SPECIFIC CD8+ T CELLS IN SYMPTOMATIC HERPES

Table II. Potential HLA-A*02:01–restricted epitopes selected from HSV virion protein (VP11/12)

Peptide Sequence m.w. No. (aa) BIMASa SYFPEITHIa MAPPPa MHCPreda HLA-A*0201 (IC50 nM)

VP11/1239–47 CLLPTPEGL 942.1 9 38.730 25 ND 0.4 ND


VP11/1266–74 FLTCTDRSV 1041.1 9 63.988 22 0.9727 0.15 83
VP11/12127–135 ILTQYWKYL 1227.4 9 339.555 21 0.9576 0.49 ND
VP11/12197–205 RIQQYMFFM 1263.5 9 50.119 13 1.0000 0.27 81
VP11/12220–228 RLNELLAYV 1090.2 9 3607.314 30 0.8284 0.23 709
VP11/12230–238 VLYRWASWM 1211.4 9 148.643 16 0.7441 0.27 ND
VP11/12238–246 MLWTTDKHV 1130.3 9 981.379 20 0.7346 0.21 1293
VP11/12346–354 TLTGYGVWA 967.0 9 27.324 18 0.5100 ND ND
VP11/12622–630 RVYEEIPWM 1222.4 9 72.146 16 ND 0.37 ND
VP11/12702–710 ALSALLTKL 929.1 9 49.134 29 0.7905 0.55 313
The sequence of HSV-1 virion protein (VP11/12) was submitted to screening of potential HLA-A*02:01 epitopes using several computer algorithms. Ten peptides were
selected on the basis of HLA-A*0201 binding motif sequence from HSV-1 VP11/12.
a
The values in these columns show predicted IC50 as calculated by BIMAS (http://www-bimas.cit.nih.gov/molbio/hla_bind), SYFPEIT (http://www.syfpeithi.de), MAPPP
(http://www.mpiib-berlin.mpg.de/MAPPP), and MHCPred (http://www.mhc-pathway.net). The sequences of synthesized peptides are based on the HSV-1 strain 17.

Frequent VP11/12220–228– and VP11/12702–710–specific CD8+ on acyclovir or other antiviral or anti-inflammatory drug treat-
T cells detected in HLA-A*02:01–positive HSV-seropositive ments at the time of blood sample collections.

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


ASYMP individuals We compared the frequency of CD8+ T cells specific to each of
The characteristics of the SYMP and ASYMP study populations the 10 VP11/12 peptide epitopes, using HLA-A*02:01–specific
used in this study, with respect to age, sex, HLA-A*02:01 fre- tetramers/anti-CD8 mAbs, in the peripheral blood of 10 HLA-
quency distribution, HSV-1/HSV-2 seropositivity, and status of A*02:01–positive, HSV-1–seropositive ASYMP and 10 HLA-
ocular herpetic disease are presented in Table I and detailed in A*02:01–positive, HSV-1–seropositive SYMP individuals, as
Materials and Methods. Because HSV-1 is the main cause of described earlier (Fig. 1). All 10 peptides were tested in this study
ocular herpes, only individuals who are HSV-1 seropositive and because lack of binding may not always translate into lack of
HSV-2 seronegative were enrolled in this study. These HSV-1– frequency or function (1, 2). The low frequencies of PBMC-
seropositive individuals were segregated into two groups: 1) 10 derived, HSV-specific CD8+ T cells complicate a direct ex vivo
HLA-A*02:01–positive HSV-1–infected ASYMP individuals, detection with tetramers using a typical number of PBMCs (∼106
who have never had detectable levels of any clinical herpes dis- cells). In addition, a prior expansion of CD8+ T cells by HSV-1 or
ease; and 2) 10 HLA-A*02:01–positive HSV-1–infected SYMP peptide stimulation in an in vitro culture would hamper a reliable
individuals with a history of numerous episodes of well-documented determination of the frequency, phenotype, and function of
recurrent clinical herpes diseases, such as herpetic lid lesions, her- epitope-specific CD8+ T cells. We therefore opted to measure the
petic conjunctivitis, dendritic or geographic keratitis, stromal kera- frequencies of VP11/12 epitope-specific CD8+ T cells ex vivo
titis, and iritis consistent with recurrent HSK, with one or more using 103 the number of PBMCs (∼10 3 106) per tetramer/CD8
episodes per year for the past 2 y. None of the SYMP patients was mAbs panel. The representative dot plots shown in Fig. 2A indicate

FIGURE 1. Stabilization of HLA-A*0201 molecules by VP11/12 peptides on the surface of T2 cells. T2 cells (3 3 105) were incubated with serial
dilutions of the indicated VP11/12 peptide, as described in Materials and Methods. Cells were then stained with FITC-conjugated anti–HLA-A2
mAb (BB7.2). The graph represents the percent of MFI reflecting an increase in the expression of HLA-A2 molecules on the surface of T2 cells triggered
by various concentrations of VP11/12 peptides. The percent of MFI increase was calculated as follows: Percent of MFI increase = (MFI with the given
peptide 2 MFI without peptide)/(MFI without peptide) 3 100. Solid lines represent peptides that bind with high to moderate affinity to HLA-A02:01
molecules, as determined by stabilization of high levels of HLA-A*02:01 molecules on the surface of T2 cells when incubated with the indicated molarity
of VP11/12 peptide. Broken lines represent low-affinity peptide binders as determined by low levels of HLA-A*02:01 molecules stabilized on the surface
of T2 cells. Error bars show SD obtained from three independent experiments.
The Journal of Immunology 2237

similar frequencies of CD8+ T cells specific to subdominant VP11/ epitopes, there was a significantly higher frequency of recognition
1266–74 epitope and higher frequencies of CD8+ T cells specific to by CD8+ T cells from ASYMP individuals.
two immunodominant VP11/12220–228 and VP11/12702–710 epitopes
detected in one ASYMP individual (top row) compared with one ASYMP individuals have a higher frequency of VP11/12
SYMP individual (lower row). Fig. 2B shows median frequencies epitope–specific CD8+ TEMs, whereas SYMP individuals have
detected in 10 SYMP and 10 ASYMP individuals. The highest and a higher frequency of VP11/12 epitope–specific CD8+ central
most significant frequencies of tetramer+ CD8+ T cells that were memory CD8+ T cells
consistently detected in both SYMP and ASYMP individuals were HSV-specific memory CD8+ T cells are categorized into two major
recorded against VP11/12220–228 and VP11/12702–710 epitopes (7.1– phenotypically distinct TEM and central memory CD8+ T cell (TCM)
14.7%). Significantly higher frequencies of CD8+ T cells specific to subpopulations (1, 4, 11, 37). We next investigated whether there
both VP11/12220–228 and VP11/12702–710 epitopes were detected in is a difference in the frequencies of CD8+ TEM and TCM sub-
ASYMP versus SYMP individuals (p = 0.03 and p = 0.02, re- populations specific to HSV-1 VP11/12 epitopes in SYMP versus
spectively). Similarly, low frequencies of CD8+ T cells were de- ASYMP individuals. Because the frequency of total CD8+ T cells
tected in both SYMP and ASYMP individuals against VP11/1266–74 specific to VP11/1266–74 epitope was low, we focused on CD8+
epitope (p = 0.4). Despite repeated attempts, with and without T cells specific to the immunodominant VP11/12220–228 and VP11/
in vitro expansions, the remaining seven VP11/12 peptide/HLA- 12702–710 epitopes, using specific tetramers, as described earlier.
A*02:01 tetramers consistently detected a low and nonsignificant ASYMP individuals had significantly higher percentages of
frequency of CD8+ T cells in HLA-A*02:01–positive HSV- CD44highCC62LlowCD8+ TEMs specific to VP11/12220–228 (Fig. 3A,
seropositive SYMP and ASYMP individuals (data not shown). 3B) and VP11/12702–710 (Fig. 3C, 3D) epitopes compared with

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


Thus, although CD8+ T cells from ASYMP and SYMP both SYMP individuals (p , 0.05, ANOVA test). In contrast, signifi-
frequently recognized the VP11/12220–228 and VP11/12702–710 cantly higher percentages of VP11/12220–228 and VP11/12702–710
epitope–specific CD8+CD44highCC62Lhigh TCMs were consistently
detected in SYMP individuals compared with ASYMP individuals
(p , 0.01, ANOVA test).
Using different markers of CD8+ TEM and TCM subpopulations,
we similarly found that ASYMP individuals had significantly
higher percentages of CD45RAlowCCR7lowCD8+ TEMs specific to
VP11/12220–228 (Fig. 3E, 3F) and VP11/12702–710 (Fig. 3G, 3H)
epitopes, whereas SYMP individuals had significantly greater
percentages of CD45RAlow CCR7highCD8+ TCM s (p , 0.01,
ANOVA test).
Altogether, the phenotypic properties of HSV-1 VP11/12 epi-
tope–specific memory CD8+ T cells revealed a clear dichotomy in
HSV-specific memory CD8+ T cell subpopulations, with ASYMP
individuals featuring higher frequencies of the “experienced”
HSV-specific CD8+ TEMs. Thus, the results suggest that at a sec-
ond pathogen encounter (e.g., after HSV-1 reactivation from
latency or reinfection), ASYMP individuals, but not SYMP indi-
viduals, would better contain herpes infection and disease by
mounting faster and stronger protective HSV-specific CD8+ TEM
cell responses.
HSV-1 VP11/12 epitope–specific effector CD8+ T cells from
ASYMP individuals are multifunctional
FIGURE 2. Frequencies of CD8+ T cells specific to VP11/12220–228 and We compared the function of HSV-VP11/12 epitope–specific ef-
VP11/12702–710 epitopes detected in HLA-A*02:01–positive HSV-sero- fector CD8+ T cells from SYMP versus ASYMP individuals. We
positive ASYMP individuals compared with SYMP individuals. PBMCs first compared the level of GzmB, GzmK, and PFN expressed on
(∼10 3 106) derived from 10 HLA-A*02:01–positive HSV-1–seropositive gated VP11/1266–74, VP11/12220–228, and VP11/12702–710 epitope–
ASYMP individuals and from 10 HLA-A*02:01–positive HSV-1–sero-
specific CD8+ T cells from SYMP and ASYMP individuals
positive SYMP individuals were analyzed ex vivo by FACS for the fre-
quency of CD8+ T cells specific to three VP11/12 epitopes using (Fig. 4A). We found high levels of GzmB, GzmK, and PFN
HLA-A*0201 VP11/12 peptide–tetramer complexes representing each of expressed on VP11/1266–74, VP11/12220–228, and VP11/12702–710
the three high to medium peptide binders (VP11/1266–74, VP11/12220–228, epitope–specific CD8+ T cells from ASYMP individuals com-
and VP11/12702–710 epitopes), as shown in Fig. 1. (A) Representative FACS pared with VP11/12 epitope–specific CD8+ T cells from SYMP
data of the frequencies of CD8+ T cells, specific to VP11/1266–74, VP11/ individuals. This suggests a positive correlation of strong HSV-
12220–228, and VP11/12702–710 epitopes, detected in PBMCs from one specific CD8+ T cell cytotoxic responses with protection against
HLA-A*02:01–positive HSV-1–seropositive ASYMP individual (top ocular herpetic disease (Fig. 4A).
panels) and one HLA-A*02:01–positive HSV-1–seropositive SYMP indi- We next examined the ability of each of the three VP11/12
vidual (lower panels). (B) Average frequencies of PBMC-derived CD8+ epitope peptides to induce the expression of CD107a/b after
T cells, specific to VP11/1266–74, VP11/12220–228, and VP11/12702–710
in vitro stimulation of CD8+ T cells from SYMP versus ASYMP
epitopes, detected from 10 HLA-A*02:01–positive HSV-1–seropositive
ASYMP individuals compared with 10 HLA-A*02:01–positive HSV-se- individuals. CD107a and CD107b are lysosomal-associated mem-
ropositive SYMP individuals. Results are representative of two indepen- brane glycoproteins that surround the core of the lytic granules in
dent experiments in each individual. The indicated p values, calculated CTLs (1, 25). Upon TCR engagement and epitope stimulation,
using one-way ANOVA test, show statistical significance between SYMP CD107a/b are exposed on the cell membrane of CD8+ CTLs. Thus,
and ASYMP individuals. the level of CD107a/b expression on the surface of CTLs is used as
2238 VP11/12-SPECIFIC CD8+ T CELLS IN SYMPTOMATIC HERPES

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


FIGURE 3. Frequent VP11/12 epitope–specific CD44highCD62LlowCD8+ TEMs detected in ASYMP individuals compared with SYMP individuals. The
phenotype of CD8+ T cells specific to VP11/12 peptide–tetramer complexes representing each of the two immunodominant VP11/12220–228 and VP11/
12702–710 epitopes were analyzed in terms of TEMs and TCMs. (A) Representative FACS data of the frequencies of CD44highCD62LlowCD8+ TEMs and
CD44highCD62LhighCD8+ TCMs specific to VP11/12220–228 peptide–tetramer complexes from one HLA-A*02:01–positive HSV-1–seropositive ASYMP
individual and from one HLA-A*02:01–positive HSV-1–seropositive SYMP individual. (B) Average frequencies of blood-derived CD8+ T cells specific to
VP11/12220–228 peptide–tetramer complexes with either TEM or TCM phenotype analyzed from 10 ASYMP and 10 SYMP individuals. (C) Representative
data of the frequencies of CD44highCD62LlowCD8+ TEMs and CD44highCD62LhighCD8+ TCMs specific to VP11/12702–710 epitope from one ASYMP in-
dividual and one SYMP individual. (D) Average frequencies of blood-derived CD8+ T cells specific to VP11/12702–710 epitope with either TEM or TCM
phenotype analyzed from 10 ASYMP and 10 SYMP individuals. Representative FACS data of the frequencies of CD45RAlowCCR7lowCD8+ TEMs and
CD45RAlowCCR7highCD8+ TCMs, specific to (E) VP11/12220–228 epitope and (G) VP11/12702–710 epitope, analyzed in one ASYMP individual and one
SYMP individual. Average frequencies of PBMC-derived CD8+ T cells with either TEM or TCM phenotype, specific to (F) VP11/12220–228 epitope and (H)
VP11/12702–710 epitope, analyzed from 10 ASYMP and 10 SYMP individuals. Results are representative of two independent experiments in each indi-
vidual. The indicated p values, calculated using one-way ANOVA test, show statistical significance between SYMP and ASYMP individuals.

a direct assay for the human epitope-specific CTL response (25). measured against autologous target monocyte-derived DCs infec-
To assess whether VP11/12 epitope–specific CD8+ T cells display ted with HSV-1 by detecting the level of CD107a/b expression by
CTL activity, we generated fresh PBMC-derived CD8+ T cell lines FACS on gated CD8+ T cells. As shown in Fig. 4B–D, a higher
from HLA-A*02:01–positive ASYMP and HLA-A*02:01–posi- percentage of VP11/1266–74, VP11/12220–228, and VP11/12702–710
tive SYMP individuals after in vitro stimulation with individual epitope-specific CD8+ T cells from ASYMP individuals expressed
VP11/1266–74, VP11/12220–228, and VP11/12702–710 epitope pep- significant levels of CD107a/b. In contrast, fewer VP11/1266–74, VP11/
tides. The cytotoxicity of each of the three CD8+ T cell lines was 12220–228, and VP11/12702–710 epitope-specific CD8+ T cells from
The Journal of Immunology 2239

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


FIGURE 4. ASYMP individuals had a significantly higher proportion of polyfunctional HSV-1 VP11/12 epitope–specific CD8+ T cells. (A) VP11/12
epitope–primed CD8+ T cells from ASYMP individuals express a high level of lytic granules compared with CD8+ T cells from SYMP individuals. FACS
was used to determine the level of expression of GzmB, GzmK, and PFN on tetramer-gated CD8+ T cells specific to VP11/1266–74, VP11/12220–228, and
VP11/12702–710 epitopes, as described in Materials and Methods. Samples were acquired on BD LSR II, and data analysis was performed using FlowJo. The
numbers on the top of each histogram represent MFI depicting the level of expression of each cytotoxic molecule. Numbers in bold represent MFI of
ASYMP individual. (B) Representative FACS data of VP11/1266–74, VP11/12220–228, and VP11/12702–710 epitope–specific CD107a/bhighCD8+ T cells from
one ASYMP versus one SYMP individual. Average percentage (C) and average absolute number (D) of VP11/12220–228 epitope–specific CD107a/bhighCD8+
T cells from 10 ASYMP and 10 SYMP individuals. (E) Representative FACS data of VP11/1266–74, VP11/12220–228, and VP11/12702–710 epitope–specific
IFN-ghighCD8+ T cells from one ASYMP versus one SYMP individual. Average percentage (F) and average absolute number (G) of VP11/12220–228
epitope–specific IFN-ghighCD8+ T cells from 10 ASYMP and 10 SYMP individuals. (H) Summary pie charts showing the average amount of each cytokine
produced by CD8+ T cells from ASYMP (n = 10, black) and SYMP patients (n = 8, white), as detected by Luminex assay. The average frequencies of
cytokine-producing CD8+ T cells from SYMP and ASYMP individuals are shown inside each pie chart. (I) Each pie chart represents the overall mean of
CD8+ T cell functions from 10 HLA-A*02:01–positive ASYMP and 10 SYMP individuals in responses to stimulation with either SYMP or ASYMP VP11/
12 peptides. Each sector of the pie chart represents the number of CD8+ T cell functions produced.

SYMP patients expressed CD107a/b. As expected, no significant ASYMP individuals. Freshly isolated CD8+ T cells from SYMP
percentage of CD8+ T cells upregulated CD107a/b after incubation and ASYMP individuals were stimulated in vitro for 6 h with
with mock-infected or target cells (data not shown). The results individual VP11/1266–74, VP11/12220–228, and VP11/12702–710
indicate that VP11/12-specific CD8+ T cells from ASYMP indi- epitope peptides, as described in Materials and Methods. The
viduals: 1) have higher cytotoxic activity compared with CD8+ percentage and number of IFN-g+CD8+ T cells were compared
T cells from SYMP patients, 2) have cytotoxic activity against from SYMP and ASYMP individuals by intracellular FACS
HSV-1–infected cells, and 3) are able to specifically recognize staining. Significantly higher percentages of VP11/1266–74, VP11/
endogenously processed VP11/12 epitopes from HSV-1–infected 12220–228, and VP11/12702–710 epitope–specific IFN-g–producing
target cells. The results also indicate that VP11/1266–74, VP11/ CD8+ T cells were detected in ASYMP individuals, as compared
12220–228, and VP11/12702–710 are functional cytotoxic epitopes. with SYMP individuals (p , 0.01; Fig. 4E–G).
We next determined the ability of VP11/12 epitopes to stimulate The levels of IL-2, IFN-g, and TNF-a effector cytokines pro-
the production of IFN-g by CD8+ T cells from SYMP versus duced by CD8+ T cells from ASYMP versus SYMP individuals
2240 VP11/12-SPECIFIC CD8+ T CELLS IN SYMPTOMATIC HERPES

after in vitro restimulation with VP11/1266–74, VP11/12220–228, TEM cells as seen in ASYMP individuals (45); and 4) ASYMP
and VP11/12702–710 epitopes were also compared by the Luminex individuals mostly had multifunctional CD8+ T cells with si-
microbeads system. As shown in Fig. 4H, CD8+ T cells from multaneous expression of three to five functions, whereas SYMP
ASYMP patients produced high levels of IL-2, IFN-g, and TNF-a individuals had a majority of CD8+ T cells with just one or two
consistent with differentiated T cells (Fig. 4H, black portions of simultaneous functions.
pie charts). In contrast, CD8+ T cells from SYMP individuals
produced less effector cytokines consistent with less differentiated Immunization with three VP11/12 CD8+ TEM epitopes that are
T cells (Fig. 4H, white portions of pie charts). highly recognized in ASYMP individuals elicited a strong
The percentage of ASYMP and SYMP individuals who showed protective immunity in the humanized HLA-A*02:01 Tg mouse
positive significant results for one or several CD8+ T cell functions model of ocular herpes
after in vitro restimulation with each VP11/12 peptide are sum- We next set up a preclinical vaccine trial, using our established
marized in Fig. 4I. Overall, 89% of ASYMP individuals had HSV- susceptible humanized HLA-A*02:01 Tg mouse model (HLA Tg
specific CD8+ T cells with three to five functions, indicating their mice on BALB/c genetic background) (1), to evaluate whether
ability to display concurrent polyfunctional activities: 1) produc- immunization with ASYMP immunodominant CD8+ TEM epito-
tion of high levels of GzmB, GzmK, and PFN; 2) production of pes (i.e., VP11/1266–74, VP11/12220–228, and VP11/12702–710) will
high levels of CD107a/b lytic granules (cytotoxic activity); 3) confer protection against ocular herpes infection and disease.
expression of IFN-g (FACS); 4) production of high levels of IL-2, Groups of susceptible HLA Tg mice (n = 10 mice/group) were
IFN-g, and TNF-a effector cytokines (Luminex); and 5) low immunized s.c. twice, 21 d apart, with a mixture of VP11/1266–74,
levels of expression of T cell activation markers CD27 and CD28 VP11/12220–228, and VP11/12702–710 peptide epitopes together

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


(FACS). In contrast, only 37% of SYMP individuals had HSV- with an immunodominant CD4+ T cell epitope recently identified
specific CD8+ T cells with three functions, whereas the majority from VP11/12 tegument protein (VP11/12129–143, Group 1) or with
had just one or two functions, suggesting that CD8+ T cells from a mixture of VP11/1266–74, VP11/12220–228, and VP11/12702–710
SYMP patients tended to have two or less functions (p , 0.005). peptide epitopes together with a promiscuous CD4+ T cell epitope
To further study the differences in HSV-specific CD8+ T cell (PADRE, Group 2). The CD8+ and CD4+ T cell epitope peptides
function from SYMP versus ASYMP individuals, we studied the were emulsified in CpG1826 adjuvant and delivered s.c. as a mix-
expression of CD27 and CD28 on VP11/12 epitope–specific CD8+ ture. As negative control, mock-immunized mice received CpG1826
T cells. Cell-surface expression of CD27 and CD28 costimulatory adjuvant alone (Mock, Group 3). Two weeks after the second and
molecules is often associated with continuous antigenic stimula- final immunization, animals from all groups received an ocular
tion (38, 39). Increased frequencies of VP11/12220–228–specific HSV-1 challenge (2 3 105 PFU, McKrae strain) without scarifi-
CD8+CD27+CD28+ T cells were detected from SYMP individuals cation. The mice were then assessed for up to 30 d postchallenge
compared with ASYMP individuals (Fig. 5A, 5B). We also ana- for ocular herpes pathology, ocular viral titers, and survival.
lyzed the expression of PD-1, a marker of T cell exhaustion (40). As shown in Fig. 6A and 6B, on day 10 postinfection, the pa-
As shown in Fig. 5C and 5D, SYMP patients exhibited higher thology clinical scores observed in the immunized groups were
frequencies of VP11/12220–228–specific PD1+CD8+ T cells com- significantly lower compared with those recorded in the mock-
pared with ASYMP individuals, suggesting functional exhaustion immunized group (p = 0.001, for all). Furthermore, significantly
of HSV-specific effector CD8+ T cells from SYMP patients. less virus was detected on day 7 postinfection (the peak of viral
To determine the differentiation status of HSV-specific CD8+ replication) in the eye swabs of the vaccinated groups compared
T cells from SYMP versus ASYMP individuals, we compared the with the mock-immunized group (p = 0.01; Fig. 6C). Most ani-
expression patterns of eomesodermin (Eomes) and T-bet tran- mals in the vaccinated groups survived infection (80 and 60%)
scription factors ex vivo on CD8+ T cells from SYMP and compared with 0% survival in the mock-immunized group (p ,
ASYMP individuals at both mRNA (using RT-PCR) and protein 0.005; Fig. 6D).
level (using FACS). Eomes and T-bet function as a molecular Altogether, these results indicate that immunization with immu-
switch between TCM and TEM cell differentiation by driving dif- nodominant HSV-1 VP11/1266–74, VP11/12220–228, and VP11/
ferentiation of CD8+ TEMs at the expense of TCMs (30, 41–44). As 12702–710 CD8+ TEM peptide epitopes decreased ocular herpes
shown in Fig. 5E–G, a significantly higher proportion of VP11/ disease, decreased virus replication, and protected against lethal
12220–228–specific CD8+ T cells from ASYMP individuals ocular herpes in susceptible HLA Tg mice.
expressed Eomes at the RNA level (using RT-PCR; Fig. 5E). In
addition, a higher proportion of VP11/12220–228–specific CD8+ Frequent VP11/12-specific CD8+ TEMs are detected in the
T cells from ASYMP individuals were Eomeshigh (p , 0.001; cornea of HSV-1–infected “ASYMP” HLA Tg mice
Fig. 5F, 5G). Similar results were obtained when analyzing the We next determined the phenotype and function of CD8+ T cells
expression of T-bet in VP11/12 CD8+ T cells from SYMP versus from the corneas of protected (“ASYMP”) versus nonprotected
ASYMP individuals (Fig. 5H–J). (“SYMP”) HLA Tg mice that were vaccinated and HSV-1 chal-
Altogether, the results indicate that VP11/12-specific CD8+ lenged, as described earlier. For simplicity, only the extremes
T cell responses associated with control of herpes disease differ in of ASYMP and SYMP mice were used for phenotypic and
ASYMP versus SYMP individuals: 1) a stepwise loss of CD28 functional characterization of cornea-resident CD8+ T cells
and CD27 together with an increase in IFN-g and cytolytic ac- (Fig. 7). ASYMP mice were from group 1 in the experiment
tivity by VP11/12 epitope–specific CD8+ T cells from ASYMP shown in Fig. 6B that had a clinical score of 1.0. SYMP mice
individuals are both characteristics of less differentiated effector were mice from the mock group in the experiment shown in
T cells; 2) high expression of PD-1 exhaustion marker on HSV-1 Fig. 6B that had a clinical score of 5 or 4. As shown in Fig. 7A
VP11/12-specific effector CD8+ T cells from SYMP individuals and 7B, protected (“ASYMP”) mice had a significantly higher
suggests a total or partial dysfunctionality; 3) higher expression percentage of spleen-resident VP11/12220–228 epitope–specific
of Eomes and T-bet by CD8+ T cells from ASYMP individuals, CD44highCD62LlowCD8+ TEMs, as compared with nonprotected
which may favor effector-to-memory CD8+ T cell transition (“SYMP”) mice (49.6 6 3 versus 25.1 6 2%; p , 0.05). In contrast,
(E . ..M) and the formation of a higher percentage of CD8+ a significantly higher percentage of spleen-resident VP11/12220–228
The Journal of Immunology 2241

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020

FIGURE 5. ASYMP individuals had a significantly higher proportion of differentiated and functional VP11/12 epitope–specific CD27lowCD28lowPD-1low
CD8+ T cells. Representative (A) and average frequencies (B) of VP220–228 epitope–specific CD8+ T cells from 10 ASYMP and versus 10 SYMP individuals
who are CD27highCD28high. Representative (C) and average frequencies (D) of VP220–228 epitope–specific CD8+ T cells from 10 ASYMP and versus 10 SYMP
individuals who are PD-1high. (E and H) The expression patterns of T-bet and Eomes and T-bet transcription factors were analyzed, at RNA level using RT-PCR,
from total CD8+ T cells derived from either SYMP (black bar) or ASYMP individuals (white bar). Representative FACS data of the percentage of Eomes- (F)
and T-bet (I)–positive HSV-1 VP11/12220–228 epitope–specific CD8+ T cells, derived from one SYMP individual (right) and one ASYMP individual (left).
Average frequencies of the expression patterns of Eomes (G) and T-bet (J) transcription factors analyzed by FACS at the protein level from gated VP11/
12220–228–specific CD8+ TEMs derived from 10 SYMP (s) and 10 ASYMP individuals (d). Results are representative of three independent experiments in
each individual. The indicated p values, calculated using one-way ANOVA test, show statistical significance between SYMP and ASYMP individuals.

epitope-specific CD44highCD62LhighCD8+ TCMs were detected in for CD8+ T cells specific to the subdominant VP11/1266–74
“SYMP” mice, as compared with “ASYMP” mice (p , 0.01; epitope (data not shown). These results indicate that “ASYMP”
Fig. 7A, 7B). Similar results were obtained for CD8 + T cells cornea-resident VP11/12 epitope–specific CD8 + T cells with
specific to VP11/12220–228 epitope in cornea (Fig. 7C, 7D, p , 0.01) TEM phenotype are associated with protection from ocular herpes
and TG (Fig. 7E, 7F, p , 0.01). Similar results were obtained disease.
2242 VP11/12-SPECIFIC CD8+ T CELLS IN SYMPTOMATIC HERPES

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


FIGURE 6. Protective immunity against ocular herpes infection and disease induced by immunodominant VP11/12 CD8+ TEM epitopes in humanized
HLA Tg mice. Three groups of age-matched male HLA Tg mice (n = 10 each) were immunized s.c., on days 0 and 21, with a mixture of three human CD8+
TEM cell epitope peptides (VP11/1266–74, VP11/12220–228, and VP11/12702–710) delivered either with a novel CD4+ T cell epitope (VP11/12129–143)
emulsified in CpG1826 adjuvant (Group 1) or with the promiscuous CD4+ T cell PADRE epitope peptide emulsified in CpG1826 adjuvant (Group 2). CpG1826
adjuvant alone was used as control (Group 3, mock-vaccinated). Two weeks after the final immunization, all animals were challenged ocularly with 2 3 105
PFU HSV-1 (strain McKrae). The eye disease was detected and scored 2 wk after immunization as described in Materials and Methods (A and B). Virus
titrations were determined from eye swabs on day 7 postinfection, as described in Materials and Methods (C). Survival was determined in a window of 30 d
postchallenge, as described in Materials and Methods (D). Results are representative of two independent experiments. The indicated p values, calculated
using one-way ANOVA test, show statistical significance between protected (ASYMP) and nonprotected (SYMP) mice.

To further study the differences in HSV-specific CD8 + T cell Altogether, the phenotypic and functional properties of HSV-1
function from “SYMP” versus “ASYMP” mice, we compared VP11/12 epitope–specific CD8+ T cells in HLA Tg mice revealed
the level of expression of GzmB, as determined by MFI, as that: 1) the “ASYMP” mice, but not “SYMP” mice, had frequent
well as the percentage of GzmB +CD8 + T cells, on gated VP11/ protective HSV-1 VP11/12-specific CD8+ TEM cells; 2) HSV-1
12 epitope–specific CD8+ T cells. GzmB was also significantly VP11/12-specific CD8+ T cells from “ASYMP” mice displayed
higher in “ASYMP” VP11/12 epitope–specific CD8+ T cells more concurrent polyfunctional activities than CD8+ T cells from
compared with “SYMP” VP11/12 epitope–specific CD8+ T cells SYMP mice; and 3) HSV-1 VP11/1266–74, VP11/12220–228, and
(p , 0.005; Fig. 7G). In addition, a higher percentage of VP11/12 VP11/12702–710 epitopes may represent strong T cell epitope
epitope–specific CD8+ T cells expressing GzmB was also detected candidates for inclusion in immunotherapeutic and immune-
in “ASYMP” corneas compared with “SYMP” corneas, suggest- prophylactic herpes vaccines. Of note, the sequences of VP11/
ing more protective effector VP11/12-specific CD8+ T cells in 1266–74, VP11/12220–228, and VP11/12702–710 epitopes are highly
“ASYMP” corneas (p , 0.01; Fig. 7H). conserved between HSV-1 (100%) and HSV-2 strains (77–100%;
We next determined the ability of CD8+ T cells from “SYMP” Supplemental Table I). However, no significant homology exists
versus “ASYMP” mice to produce IFN-g after recall with the between the amino acid sequences of these three epitopes and the
immunizing VP11/1266–74, VP11/12220–228, and VP11/12702–710 VP11/12 amino acid sequences of Varicella zoster virus (44%) and
epitope peptides. CD8+ T cells from “SYMP” cornea and CMV (33%) strains (Supplemental Table I). The results also
“ASYMP” cornea were stimulated in vitro for 6 h with individual confirm that HLA Tg mice are a useful animal model for under-
immunodominant (VP11/12220–228 and VP11/12702–710) and sub- standing the mechanisms by which human epitope-specific CD8+
dominant (VP11/1266–74) epitope peptides, as described in Mate- T cells mediate control of herpes infection and disease, as we
rials and Methods. The percentages of IFN-g+CD8+ T cells were previously suggested (13).
compared from SYMP and ASYMP mice by intracellular FACS
staining. Significantly higher percentages (Fig. 7I, 7J) of VP11/ Discussion
1266–74, VP11/12220–228, and VP11/12702–710 epitope–specific The HSV-1–derived tegument VPs, which are located between
CD8+ T cells producing IFN-g were detected in protected the capsid and the outer viral envelope proteins, contain a large
“ASYMP” corneas, as compared with nonprotected “SYMP” number of possible protective CD8+ T cell epitopes (9); however,
corneas (p , 0.001). only a handful of epitopes have been identified so far. In this study,
The Journal of Immunology 2243

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020

FIGURE 7. The corneas of protected HSV-1–infected “ASYMP” HLA Tg mice contain frequent VP11/12-specific polyfunctional CD8+ TEMs. (A)
Representative FACS data of the frequencies of CD44highCD62LlowCD8+ TEMs and CD44highCD62LlowCD8+ TCMs specific to VP11/12220–228 peptide–
tetramer complexes detected the spleen of one protected “ASYMP” versus one nonprotected “SYMP” HLA Tg mouse. The spleen, cornea, and trigeminal
ganglia were assayed for CD8+ T cell responses on day 9 postinfection. (B) Average frequencies of VP11/12220–228 epitope–specific TCMs and TEMs
detected in the spleen of five protected ASYMP and versus five nonprotected SYMP HLA Tg mice. ASYMP mice had a clinical score of 1.0 in Fig. 6B.
SYMP mice had a score of 5 or 4 in Fig. 6B. Representative (C and E) and average frequencies (D and F) of VP11/12220–228 epitope–specific TCMs and TEMs
detected in the cornea and TG of five protected ASYMP and versus five nonprotected SYMP HLA Tg mice. (G) Representative data of level of expression of
GzmB on cornea-derived CD8+ T cells specific to VP11/66–74, VP11/12220–228, and VP11/12702–710 detected from protected “ASYMP” mice (black his-
togram) and nonprotected “SYMP” mice (white histogram). (H) Average frequencies of GzmB+CD8+ T cells specific to VP11/12220–228 detected from the
corneas of protected “ASYMP” mice (d) and nonprotected “SYMP” mice (s). (I) Representative data of the % IFN-g+CD8+ T cells specific to VP11/66–74,
VP11/12220–228, and VP11/12702–710 detected from the corneas of protected “ASYMP” mice (right) and nonprotected “SYMP” mice (left). (J) Average
frequencies of IFN-g+CD8+ T cells specific to VP11/12220–228 detected from the corneas of protected “ASYMP” mice (d) and (Figure legend continues)
2244 VP11/12-SPECIFIC CD8+ T CELLS IN SYMPTOMATIC HERPES

we identified three protective “ASYMP” human CD8+ TEMs epi- VP11/12 peptides studied in this report, only 3 (VP11/1266–74,
topes from the abundant tegument VP11/12 protein: VP11/1266–74, VP11/12220–228, and VP11/12702–710) were generated from native
VP11/12220–228, and VP11/12702–710. These new VP11/12 epitopes VP11/12 protein by natural processing. This observation illus-
displayed high-affinity binding to purified HLA-A*0201 molecules, trated the importance of experimentally evaluating whether pep-
stabilized HLA-A*0201 molecules on target cells, and recalled tides generated by computer algorithms and various HLA-peptides
CD8+ T cells in HSV-1–seropositive ASYMP individuals and in binding assays are able to functionally stimulate CD8+ T cells
“ASYMP” HLA-A*0201 Tg mice ocularly infected with HSV-1. In in vitro and in vivo.
addition, we found two major VP11/12 epitope–specific CD8+ Many immunological assays have been developed to validate
T cell subpopulations, which have remarkably different phenotype computationally predicted human CD8+ T cell epitopes, including
and function, one of which was mostly in SYMP and the other frequency of epitope-specific CD8+ TEMs versus TCMs by tetramer
mostly in ASYMP individuals. Notably, ASYMP individuals had assays, cell membrane HLA stabilization assays, CD8+ T cell
a significantly higher proportion of differentiated polyfunctional cytokine analysis, IFN-g intracellular assay, GzmB, GzmK, PFN,
VP11/12 epitope–specific CD8+ TEMs. In contrast, SYMP patients CD107a/b degranulation cytotoxic assays, CD27/CD28 activation
had significantly higher frequencies of less differentiated mono- markers, and PD-1 exhaustion marker (52–54). When used individ-
functional VP11/12 epitope–specific CD8+ TCMs. To the best of our ually, each screen is not sufficient to conclusively identify functional
knowledge, this is the first report of the following: 1) protective CD8+ T cell epitopes (52, 54–56). However, combinations of these
CD8+ T cells epitopes from the HSV-1 VP11/12 tegument protein, screens can provide strong evidence of functional epitopes (2, 54,
and 2) phenotypic and functional differences of HSV-1 VP11/12- 57). Using these multiple screens, we identified three high-affinity
specific effector and memory CD8+ T cells from ocular herpes VP11/12 epitopes that were naturally processed and able to recall

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


SYMP and ASYMP individuals. Our findings provide a framework frequent functional human CD8+ TEMs in HSV-1–seropositive
for the rational design of a herpes vaccine based on tegument ASYMP individuals. Of interest is that the VP11/12702–710 peptide
proteins that elicit long-term CD8+ T cell–protective immunity. that appeared to bind with only low affinity to HLA-A*02:01
CD8+ T cells recognize their target Ags as 8- to 10-aa-long molecules in vitro still induced strong IFN-g–producing cytotoxic
peptides, which are derived from intracellular, processed viral CD8+ TEM responses. Although the reason for this is not completely
Ags and presented by HLA class I molecules (e.g., HLA-A*0201 understood, this result highlights the importance of screening po-
molecules) expressed at the surface of infected cells (46, 47). tential peptide epitopes using multiple immunological assays.
Despite the HSV-1 84+ open reading frames encoding hundreds of Epitope-based vaccine approaches offer several potential ad-
potential HLA-compatible CD8+ T cell epitopes, only a handful of vantages over conventional protein- or DNA-based vaccine ap-
epitopes/protein drive robust CD8+ T cells in seropositive indi- proaches in terms of purity, lot-to-lot consistency, cost of production,
viduals (48–51). Tegument proteins are delivered to the cytoplasm and a high specificity in eliciting immune responses (1, 58). In
of host cells during viral entry, resulting in an immediate pro- addition, peptide-epitope technology has proved an extremely
cessing by APCs shortly postinfection (7–10). They hence con- useful strategy to avoid potentially harmful immune responses (1,
stitute an immediate target Ag to induce host T cells (7, 8). 58). Among other advantages, the epitope-based vaccines avoid
Considering the recent failures of clinical vaccine trials using the potential for “pathogenic” epitopes to inadvertently drive un-
HSV envelope glycoproteins (mainly gB and gD) (16, 17), and wanted pathogenic responses (59). Such pathogenic responses
because tegument proteins are major targets of CD8+ T cells from might contribute to exacerbation of disease, as we recently found
herpes-seropositive individuals (7–10, 19–22), it is surprising how for an HLA-A*02:01–restricted CD8+ T cell epitope from the
only a few reports have identified protective T cell epitopes on HSV-1 gK protein (60). Because our approach to epitope identi-
tegument proteins and characterized the phenotype and function fication in this study was tailored specifically for HLA-A*02:01–
of HSV-1 tegument protein–derived epitope-specific “protective” restricted epitopes, the results reported in this study do not imply
CD8+ T cells from HSV-seropositive healthy ASYMP individuals, that these are the only human CD8+ T cell epitopes present in
who appear to have acquired a “natural” protective immunity. This VP11/12. However, this study describes a model system that could
information is necessary for the successful design of an effective be applied to any HLA class I haplotype and to any other herpes
T cell–based therapeutic vaccine strategy. Rather than just using tegument, structural, or regulatory proteins to identify other SYMP
a few in vitro predictive assays to map CD8+ T cell epitopes from versus ASYMP epitopes.
HSV-1 VP11/12 protein, in this study, we have used several The choice of animal model is crucial in herpes vaccine de-
computational algorithms followed by both in vitro and in vivo velopment (reviewed in Ref. 61). A major goal of this study was to
functional immunological assays. Only peptides that conform to examine the protective efficacy of human HLA-restricted epitopes.
given sequence motifs preferentially bind with high affinity to Thus, the ideal animal model should mount HLA-restricted im-
HLA class I molecules (1), and the amino acid residues at specific mune responses specific to human epitopes, whereas at the same
positions are conserved for peptides that bind HLA-A*0201 time mimicking as many aspects of herpes infection and disease as
molecules with high affinity (1, 4). Potential HLA-A*0201– possible (62). Normal mice (e.g., BALB/c or C57BL/6) have been
binding peptides can be selected based on the presence of leucine the standard choice for most immunologists, and results from
(L), isoleucine (I), or methionine (M) as the dominant residue at these mice have yielded tremendous insights into the role of H2-
position 2, and valine (V) or a residue with an aliphatic hydro- restricted mouse T cells in protection against herpes infection and
carbon side chain at the C terminus (1). After identification of disease (4, 23, 36, 62–65). However, normal mice do not mount
antigenic VP11/12 peptides in HLA-A*0201–positive HSV-1– specific humanized T cell responses to HLA-restricted epitopes
seropositive humans, it is important to address whether each (reviewed in Ref. 61). In contrast, similar to humans, HLA Tg
peptide can be correctly processed from the native Ag and sub- mice can develop humanized T cell responses to HLA-restricted
sequently displayed on the surface of an APC. Of the 10 antigenic epitopes (66).

nonprotected “SYMP” mice (s). A total of 1 3 106 cells for each assay and the results are representative of two independent experiments. The indicated
p values, calculated using one-way ANOVA test, show statistical significance between SYMP and ASYMP mice.
The Journal of Immunology 2245

A question of practical importance is the translation of the the population size, the phenotype, and the function of CD8+
current preclinical findings for the development of an epitope- T cells obtained from peripheral blood. Although we are aware
based clinical vaccine for a genetically heterogeneous human that information gained from peripheral blood T cells may not be
population (15, 27, 58, 67–69). A universal herpes vaccine should completely reflective of tissue-resident T cells, because of the
induce protective ASYMP CD8+ T cell responses in the context of obvious ethical and practical considerations of obtaining tissue-
many different HLA alleles. This report focused on HLA-A*0201 resident CD8+ T cells (i.e., from the cornea or from trigeminal
because of its high prevalence in most populations worldwide, ganglia, the site of acute and latent infections, respectively),
regardless of race and ethnicity (61, 70–72). The high degree of our investigations were limited to human PBMC-derived CD8+
HLA polymorphism that might be a hindrance to epitope-based T cells. Under the earlier circumstances, HSV-specific CD8+
vaccines can be dealt with by the inclusion of multiple HLA- T cells from ASYMP patients had a greater frequency of poly-
restricted epitopes that are recognized in the context of diverse functional VP11/12 epitope–specific CD8+ TEMs, whereas HSV-
related HLA alleles, and by designing peptide-based vaccines with specific CD8+ T cells from SYMP individuals had more mono-
higher CD8+ T cell epitope densities. A combination of nine HLA functional VP11/12 epitope–specific CD8+ TCMs. These results
supertypes has been defined to provide an almost perfect coverage suggest that compared with SYMP individuals, at a second path-
(.99%) of the entire repertoire of HLA molecules (62). Because ogen encounter (e.g., after HSV-1 reactivation from latency),
a mixture of multiple ASYMP epitopes is likely to produce ASYMP individuals would mount faster and stronger protective
polyclonal CD8+ T cell lines (one T cell clone for each epitope) HSV-specific CD8+ TEM responses, allowing a better clearance of
and more effector T cells than a single epitope (63), we used herpes infection and disease.
multiple CD8+ T cell epitopes to induce a broader T cell response We also found a low expression of Eomes associated with high

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


(15, 63). To further increase efficacy, a multiepitope-based peptide expression of PD-1 in VP11/12-specific CD8+ T cells from SYMP
vaccine should include several CD8+ T cell epitopes from several individuals. In contrast, high expression of T-bet associated with
different glycoproteins and tegument proteins, each chosen to rep- low expression of PD-1 was recorded in VP11/12-specific CD8+
resent the HLA supertypes known to provide recognition in a large T cells from ASYMP individuals. This is consistent with differ-
proportion of the global population, regardless of race and ethnicity. entiated and functional CD8+ T cells in ASYMP individuals. It is
Focusing the immune response toward selected ASYMP epit- likely that the observed decrease in T-bet expression in SYMP
opes could be of value in the case of herpes infections, where individuals contributes to a restrain in terminal differentiation of
T cells directed against the ASYMP epitopes might have been secondary effector and memory CD8+ T cells as recently reported
inactivated and T cells specific for SYMP epitopes might have in another system (66). On one hand, T-bet and Eomes are key
escaped T cell tolerance. In addition, monitoring immunogenicity transcription factors involved in the long-term renewal of memory
after vaccination is easily done due to the fact that the epitopes used CD8+ T cells to their characteristic effector potency (30,41–44).
in the vaccine also serve as the required diagnostic reagents needed T-bet drives differentiation of CD8+ TEMs at the expense of TCMs
to follow T cell responses in the vaccines. The efficacy of peptide (43). T-bet is also overexpressed in CD8+ T cells that differenti-
epitopes immunization on different T cell memory populations has ated in the absence of CD4+ T cell help, a condition that is as-
been recently reported (64). Most notably, peptide immunotherapy sociated with defective central memory formation (43). Eomes
appeared to induce strong and long-lived tissue-resident TEMs, but enables CD8+ T cells to compete for the memory cell niche (42).
not circulating TCMs (64). This study supports our recently ad- In addition, CD8+ T cells deficient in T-bet and Eomes fail to
vanced “SYMP/ASYMP concept” by showing that immunization differentiate into functional killers, which is required for defense
with immunodominant VP11/12 epitopes that are mainly recog- against viral pathogens (30). Instead, virus-specific CD8+ T cells
nized by CD8+ TEMs from “naturally” protected ASYMP individ- lacking both T-bet and Eomes differentiate into an IL-17–secret-
uals also protected HLA Tg mice against ocular herpes infection ing autoimmune and inflammatory T cell lineage (30). The earlier
and disease. observations together with our results are reminiscent of the CD8+
Humans are not immunologically “naive” and often develop T cell fate implicated in SYMP herpes disease. On the other hand,
T cell responses that cross-react between different viruses in- T cell exhaustion has a major role in the failure to control per-
cluding members of the same (or different) family (65, 73, 74). sistent infections, likely including latent HSV-1 and HSV-2 in-
However, this is unlikely for the three VP11/12 epitopes discov- fections. High expression of inhibitory receptors, including PD-1,
ered in this study because: 1) alignment of their sequences with and the inability to sustain functional T cell responses contribute
published sequences of VP11/12 proteins from other human her- to T cell exhaustion. We found a low expression of T-bet associ-
pesviruses (e.g., VZV and CMV) did not reveal significant ho- ated with high expression of PD-1 in VP11/12-specific CD8+
mologies that might be translated to cross-reactive T cell T cells from SYMP individuals. In contrast, high expression of
responses (Supplemental Table I); and 2) the three HLA-A*02: T-bet associated with low expression of PD-1 was recorded in
01–restricted immunodominant VP11/12 epitopes reported in this VP11/12-specific CD8+ T cells from ASYMP individuals. Our
study as recognized by CD8+ T cells from HLA-A*02:01–positive results suggest a persistent antigenic stimulation in SYMP indi-
HSV-1–seropositive individuals are also recognized with the same viduals and are in agreement with a recent report showing per-
magnitude and breath by CD8+ T cells induced in “pathogen-free” sistent antigenic stimulation caused downregulation of T-bet and
HLA-A*02:01 Tg mice after HSV-1 ocular infection. These resulted in more severe exhaustion of CD8+ T cells (41). To our
results suggest that CD8+ T cells detected in humans are likely knowledge, this is the first study to show differential expression of
induced by HSV-1 VP11/12 epitopes, rather than cross-reactive transcription factors in HSV-specific human memory CD8+ T cells
epitopes from other viruses within the a, b, g herpes family. among HLA-A*02:01–positive, HSV-1–seropositive SYMP and
However, one cannot definitely exclude a cross-reactivity of these ASYMP individuals. It is likely that the concerted action of T-bet,
HSV-1 VP11/12 epitopes with epitopes from other nonherpes Eomes, and other transfection factors results in the development
pathogens outside the herpes family. of fully differentiated CD8+ TEMs in ASYMP individuals that
To gain more insight into the nature of HSV-specific CD8+ T cell migrate to inflamed tissues (i.e., eyes and sensory ganglia) after
subpopulations that are predominant in SYMP versus ASYMP Ag recognition and secrete IFN-g and/or release cytotoxic gran-
individuals, we used several immunological assays to determine ules that contain GzmB and PFN (75, 76).
2246 VP11/12-SPECIFIC CD8+ T CELLS IN SYMPTOMATIC HERPES

The underlying protective and nonprotective immune mecha- positive asymptomatic individuals and protect HLA transgenic mice against
ocular herpes. J. Immunol. 191: 5124–5138.
nisms of TEMs and TCMs in ocular herpes remain to be fully 2. Chentoufi, A. A., X. Zhang, K. Lamberth, G. Dasgupta, I. Bettahi, A. Nguyen,
elucidated. A recent study suggests that CD8+ T cells are required M. Wu, X. Zhu, A. Mohebbi, S. Buus, et al. 2008. HLA-A*0201-restricted CD8+
to eliminate virus more efficiently from the cornea but play cytotoxic T lymphocyte epitopes identified from herpes simplex virus glyco-
protein D. J. Immunol. 180: 426–437.
a minimal role in immunopathology (77). However, the TEM and 3. Zhang, X., X. Dervillez, A. A. Chentoufi, T. Badakhshan, I. Bettahi, and
TCM phenotype of these cells was not reported, and the translation L. Benmohamed. 2012. Targeting the genital tract mucosa with a lipopeptide/
of these findings to the human situation, where “SYMP” CD8+ recombinant adenovirus prime/boost vaccine induces potent and long-lasting
CD8+ T cell immunity against herpes: importance of MyD88. J. Immunol.
TCMs and “ASYMP” CD8+ TEMs may be in play, still remains to 189: 4496–4509.
be determined. Based on our present findings, we propose a 4. Samandary, S., H. Kridane-Miledi, J. S. Sandoval, Z. Choudhury, F. Langa-
Vives, D. Spencer, A. A. Chentoufi, F. A. Lemonnier, and L. BenMohamed.
mechanism build on a novel “SYMP/ASYMP concept,” in which
2014. Associations of HLA-A, HLA-B and HLA-C alleles frequency with
the corneal lesions are not a direct consequence of damage caused prevalence of herpes simplex virus infections and diseases across global pop-
by the virus or by autoreactive or bystander T cells, but rather ulations: implication for the development of an universal CD8+ T-cell epitope-
based vaccine. Hum. Immunol. 75: 715–729.
from the balance between nonprotective TCM responses specific to 5. Kuo, T., C. Wang, T. Badakhshan, S. Chilukuri, and L. BenMohamed. 2014. The
SYMP HSV-1 epitopes and protective TEM responses specific to challenges and opportunities for the development of a T-cell epitope-based
nonoverlapping ASYMP HSV-1 epitopes. Both direct and indirect herpes simplex vaccine. Vaccine 32: 6733–6745.
6. Chentoufi, A. A., L. BenMohamed, P. Van De Perre, and A. A. Ashkar. 2012.
mechanisms might be involved in TEM-mediated protection and in Immunity to ocular and genital herpes simplex viruses infections. Clin. Dev.
TCM-mediated susceptibility to recurrent herpetic disease. The Immunol. 2012: 732546.
polyfunctionality of CD8+ TEMs segregated with immunologic 7. Long, D., M. Skoberne, T. M. Gierahn, S. Larson, J. A. Price, V. Clemens,
A. E. Baccari, K. P. Cohane, D. Garvie, G. R. Siber, and J. B. Flechtner. 2014.
control of herpes infection and disease, in both ASYMP individ-

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


Identification of novel virus-specific antigens by CD4⁺ and CD8⁺ T cells from
uals and in “ASYMP” HLA Tg mice. Thus, “polyfunctionality” of asymptomatic HSV-2 seropositive and seronegative donors. Virology 464-465:
HSV-specific TEMs may be an important factor accounting for the 296–311.
8. Hosken, N., P. McGowan, A. Meier, D. M. Koelle, P. Sleath, F. Wagener,
immunologic control of herpes. Epitope-specific TEM lines from M. Elliott, K. Grabstein, C. Posavad, and L. Corey. 2006. Diversity of the CD8+
ASYMP individuals appeared to recognize naturally processed T-cell response to herpes simplex virus type 2 proteins among persons with
genital herpes. J. Virol. 80: 5509–5515.
epitopes on HSV-1–infected cells and had strong virus inhibitory 9. Dasgupta, G., A. A. Chentoufi, M. Kalantari, P. Falatoonzadeh, S. Chun,
cytotoxic activity. A formal demonstration that ASYMP CD8+ C. H. Lim, P. L. Felgner, D. H. Davies, and L. BenMohamed. 2012. Immuno-
TEMs, with these functions intact, cause immunologic control of dominant “asymptomatic” herpes simplex virus 1 and 2 protein antigens iden-
tified by probing whole-ORFome microarrays with serum antibodies from
recurrent herpes infection and disease would require passive- seropositive asymptomatic versus symptomatic individuals. J. Virol. 86: 4358–
transfer studies in humans or in experimental animals that develop 4369.
spontaneous recurrent disease (11). 10. Kalantari-Dehaghi, M., S. Chun, A. A. Chentoufi, J. Pablo, L. Liang,
G. Dasgupta, D. M. Molina, A. Jasinskas, R. Nakajima-Sasaki, J. Felgner,
In conclusion, this study provides several new insights about the et al. 2012. Discovery of potential diagnostic and vaccine antigens in herpes
specificity, phenotype, and function of HSV-1 VP11/12 epitope– simplex virus 1 and 2 by proteome-wide antibody profiling. J. Virol. 86: 4328–
specific CD8+ T cell subpopulation that segregate with immuno- 4339.
11. Chentoufi, A. A., G. Dasgupta, N. D. Christensen, J. Hu, Z. S. Choudhury,
logic control of ocular herpes infection and disease in both A. Azeem, J. V. Jester, A. B. Nesburn, S. L. Wechsler, and L. BenMohamed.
ASYMP individuals and humanized HLA-A*02:01 Tg mice. We 2010. A novel HLA (HLA-A*0201) transgenic rabbit model for preclinical
evaluation of human CD8+ T cell epitope-based vaccines against ocular herpes.
report three previously unknown HLA-A*02:01–restricted ASYMP J. Immunol. 184: 2561–2571.
epitopes derived from HSV-1 VP11/12 that tend to recall poly- 12. Langenberg, A. G., L. Corey, R. L. Ashley, W. P. Leong, and S. E. Straus, Chiron
functional CD8+ TEM responses in HLA-A*02:01–positive HSV- HSV Vaccine Study Group. 1999. A prospective study of new infections with
herpes simplex virus type 1 and type 2. N. Engl. J. Med. 341: 1432–1438.
seropositive ASYMP humans and in humanized HLA-A*02:01 Tg 13. Dasgupta, G., and L. BenMohamed. 2011. Of mice and not humans: how reliable
mice protected from ocular herpes infection and disease. Under- are animal models for evaluation of herpes CD8(+)-T cell-epitopes-based im-
standing the detailed mechanisms by which HLA-A*02:01– munotherapeutic vaccine candidates? Vaccine 29: 5824–5836.
14. Chentoufi, A. A., N. R. Binder, N. Berka, G. Durand, A. Nguyen, I. Bettahi,
restricted ASYMP CD8+ T cells are associated with protection B. Maillère, and L. BenMohamed. 2008. Asymptomatic human CD4+ cytotoxic
against disease will reveal underlying principles of immune T-cell epitopes identified from herpes simplex virus glycoprotein B. J. Virol. 82:
control of herpes, which is critical for the rational design of a 11792–11802.
15. Zhang, X., A. Issagholian, E. A. Berg, J. B. Fishman, A. B. Nesburn, and
T cell–based vaccine strategy. Overall, these findings demonstrate L. BenMohamed. 2005. Th-cytotoxic T-lymphocyte chimeric epitopes extended
quantitative and qualitative features of an effective HSV-specific by Nepsilon-palmitoyl lysines induce herpes simplex virus type 1-specific ef-
fector CD8+ Tc1 responses and protect against ocular infection. J. Virol. 79:
CD8+ T cell response that should be considered in the testing of 15289–15301.
the next generation of herpes vaccines. 16. Stanberry, L. R., S. L. Spruance, A. L. Cunningham, D. I. Bernstein, A. Mindel,
S. Sacks, S. Tyring, F. Y. Aoki, M. Slaoui, M. Denis, et al; GlaxoSmithKline
Herpes Vaccine Efficacy Study Group. 2002. Glycoprotein-D-adjuvant vaccine
Acknowledgments to prevent genital herpes. N. Engl. J. Med. 347: 1652–1661.
We thank Dale Long from the National Institutes of Health Tetramer 17. Belshe, R. B., P. A. Leone, D. I. Bernstein, A. Wald, M. J. Levin, J. T. Stapleton,
I. Gorfinkel, R. L. A. Morrow, M. G. Ewell, A. Stokes-Riner, et al; Herpevac
Facility (Emory University, Atlanta, GA) for providing the Tetramers Trial for Women. 2012. Efficacy results of a trial of a herpes simplex vaccine. N.
used in this study. We also thank Barbara Bodenhoefer, RN, from the Engl. J. Med. 366: 34–43.
University of California Irvine Institute for Clinical and Translational 18. Murphy, M. A., M. A. Bucks, K. J. O’Regan, and R. J. Courtney. 2008. The
Science for helping with blood drawing from HSV-1–seropositive SYMP HSV-1 tegument protein pUL46 associates with cellular membranes and viral
capsids. Virology 376: 279–289.
and ASYMP individuals. 19. Muller, W. J., L. Dong, A. Vilalta, B. Byrd, K. M. Wilhelm, C. L. McClurkan,
M. Margalith, C. Liu, D. Kaslow, J. Sidney, et al. 2009. Herpes simplex virus
type 2 tegument proteins contain subdominant T-cell epitopes detectable in
Disclosures BALB/c mice after DNA immunization and infection. J. Gen. Virol. 90: 1153–
The authors have no financial conflicts of interest. 1163.
20. Dervillez, X., C. Gottimukkala, K. W. Kabbara, C. Nguyen, T. Badakhshan,
S. M. Kim, A. B. Nesburn, S. L. Wechsler, and L. Benmohamed. 2012. Future of
an “Asymptomatic” T-cell Epitope-Based Therapeutic Herpes Simplex Vaccine.
References Future Virol 7: 371–378.
1. Dervillez, X., H. Qureshi, A. A. Chentoufi, A. A. Khan, E. Kritzer, D. C. Yu, 21. Koelle, D. M., Z. Liu, C. L. McClurkan, R. C. Cevallos, J. Vieira, N. A. Hosken,
O. R. Diaz, C. Gottimukkala, M. Kalantari, M. C. Villacres, et al. 2013. C. A. Meseda, D. C. Snow, A. Wald, and L. Corey. 2003. Immunodominance
Asymptomatic HLA-A*02:01-restricted epitopes from herpes simplex virus among herpes simplex virus-specific CD8 T cells expressing a tissue-specific
glycoprotein B preferentially recall polyfunctional CD8+ T cells from sero- homing receptor. Proc. Natl. Acad. Sci. USA 100: 12899–12904.
The Journal of Immunology 2247

22. Koelle, D. M., H. B. Chen, M. A. Gavin, A. Wald, W. W. Kwok, and L. Corey. 43. Intlekofer, A. M., N. Takemoto, C. Kao, A. Banerjee, F. Schambach,
2001. CD8 CTL from genital herpes simplex lesions: recognition of viral teg- J. K. Northrop, H. Shen, E. J. Wherry, and S. L. Reiner. 2007. Requirement for
ument and immediate early proteins and lysis of infected cutaneous cells. J. T-bet in the aberrant differentiation of unhelped memory CD8+ T cells. J. Exp.
Immunol. 166: 4049–4058. Med. 204: 2015–2021.
23. Oh, S., M. Terabe, C. D. Pendleton, A. Bhattacharyya, T. K. Bera, M. Epel, 44. Takemoto, N., A. M. Intlekofer, J. T. Northrup, E. J. Wherry, and S. L. Reiner.
Y. Reiter, J. Phillips, W. M. Linehan, C. Kasten-Sportes, et al. 2004. Human 2006. Cutting edge: IL-12 inversely regulates T-bet and eomesodermin expres-
CTLs to wild-type and enhanced epitopes of a novel prostate and breast tumor- sion during pathogen-induced CD8+ T cell differentiation. J. Immunol. 177:
associated protein, TARP, lyse human breast cancer cells. Cancer Res. 64: 2610– 7515–7519.
2618. 45. Joshi, N. S., and S. M. Kaech. 2008. Effector CD8 T cell development: a bal-
24. Stuber, G., G. H. Leder, W. T. Storkus, M. T. Lotze, S. Modrow, L. Székely, ancing act between memory cell potential and terminal differentiation. J.
H. Wolf, E. Klein, K. Kärre, and G. Klein. 1994. Identification of wild-type and Immunol. 180: 1309–1315.
mutant p53 peptides binding to HLA-A2 assessed by a peptide loading-deficient 46. Bernardin, F., D. Kong, L. Peddada, L. A. Baxter-Lowe, and E. Delwart. 2005.
cell line assay and a novel major histocompatibility complex class I peptide Human immunodeficiency virus mutations during the first month of infection are
binding assay. Eur. J. Immunol. 24: 765–768. preferentially found in known cytotoxic T-lymphocyte epitopes. J. Virol. 79:
25. Betts, M. R., J. M. Brenchley, D. A. Price, S. C. De Rosa, D. C. Douek, 11523–11528.
M. Roederer, and R. A. Koup. 2003. Sensitive and viable identification of 47. Wang, Q. J., X. L. Huang, G. Rappocciolo, F. J. Jenkins, W. H. Hildebrand,
antigen-specific CD8+ T cells by a flow cytometric assay for degranulation. J. Z. Fan, E. K. Thomas, and C. R. Rinaldo, Jr. 2002. Identification of an HLA
Immunol. Methods 281: 65–78. A*0201-restricted CD8(+) T-cell epitope for the glycoprotein B homolog of
26. Rubio, V., T. B. Stuge, N. Singh, M. R. Betts, J. S. Weber, M. Roederer, and human herpesvirus 8. Blood 99: 3360–3366.
P. P. Lee. 2003. Ex vivo identification, isolation and analysis of tumor-cytolytic 48. Pelte, C., G. Cherepnev, Y. Wang, C. Schoenemann, H. D. Volk, and F. Kern.
T cells. Nat. Med. 9: 1377–1382. 2004. Random screening of proteins for HLA-A*0201-binding nine-amino acid
27. BenMohamed, L., R. Krishnan, C. Auge, J. F. Primus, and D. J. Diamond. 2002. peptides is not sufficient for identifying CD8 T cell epitopes recognized in the
Intranasal administration of a synthetic lipopeptide without adjuvant induces context of HLA-A*0201. J. Immunol. 172: 6783–6789.
systemic immune responses. Immunology 106: 113–121. 49. Stock, A. T., C. M. Jones, W. R. Heath, and F. R. Carbone. 2006. CTL response
28. BenMohamed, L., R. Krishnan, J. Longmate, C. Auge, L. Low, J. Primus, and compensation for the loss of an immunodominant class I-restricted HSV-1 de-

Downloaded from http://www.jimmunol.org/ by guest on September 19, 2020


D. J. Diamond. 2000. Induction of CTL response by a minimal epitope vaccine terminant. Immunol. Cell Biol. 84: 543–550.
in HLA A*0201/DR1 transgenic mice: dependence on HLA class II restricted T 50. Storkus, W. J., and H. M. Zarour. 2000. Melanoma antigens recognised by CD8+
(H) response. Hum. Immunol. 61: 764–779. and CD4+ T cells. Forum (Genova) 10: 256–270.
29. Boucherma, R., H. Kridane-Miledi, R. Bouziat, M. Rasmussen, T. Gatard, 51. Wallace, M. E., R. Keating, W. R. Heath, and F. R. Carbone. 1999. The cytotoxic
F. Langa-Vives, B. Lemercier, A. Lim, M. Bérard, L. Benmohamed, et al. 2013. T-cell response to herpes simplex virus type 1 infection of C57BL/6 mice is
HLA-A*01:03, HLA-A*24:02, HLA-B*08:01, HLA-B*27:05, HLA-B*35:01, almost entirely directed against a single immunodominant determinant. J. Virol.
HLA-B*44:02, and HLA-C*07:01 monochain transgenic/H-2 class I null mice: 73: 7619–7626.
novel versatile preclinical models of human T cell responses. J. Immunol. 191: 52. Hertz, T., and C. Yanover. 2007. Identifying HLA supertypes by learning dis-
583–593. tance functions. Bioinformatics 23: e148–e155.
30. Intlekofer, A. M., A. Banerjee, N. Takemoto, S. M. Gordon, C. S. Dejong, 53. Botten, J., J. Alexander, V. Pasquetto, J. Sidney, P. Barrowman, J. Ting, B. Peters,
H. Shin, C. A. Hunter, E. J. Wherry, T. Lindsten, and S. L. Reiner. 2008. S. Southwood, B. Stewart, M. P. Rodriguez-Carreno, et al. 2006. Identification of
Anomalous type 17 response to viral infection by CD8+ T cells lacking T-bet protective Lassa virus epitopes that are restricted by HLA-A2. J. Virol. 80: 8351–8361.
and eomesodermin. Science 321: 408–411. 54. Sidney, J., S. Southwood, C. Moore, C. Oseroff, C. Pinilla, H. M. Grey, and
31. Zhang, X., A. A. Chentoufi, G. Dasgupta, A. B. Nesburn, M. Wu, X. Zhu, A. Sette. 2013. Measurement of MHC/peptide interactions by gel filtration or
D. Carpenter, S. L. Wechsler, S. You, and L. BenMohamed. 2009. A genital tract monoclonal antibody capture. Curr. Protoc. Immunol. Chapter 18: Unit 18.13.
peptide epitope vaccine targeting TLR-2 efficiently induces local and systemic 55. Harndahl, M., M. Rasmussen, G. Roder, I. Dalgaard Pedersen, M. Sørensen,
CD8+ T cells and protects against herpes simplex virus type 2 challenge. Mu- M. Nielsen, and S. Buus. 2012. Peptide-MHC class I stability is a better predictor
cosal Immunol. 2: 129–143. than peptide affinity of CTL immunogenicity. Eur. J. Immunol. 42: 1405–1416.
32. Benmohamed, L., A. Thomas, M. Bossus, K. Brahimi, J. Wubben, H. Gras- 56. St Leger, A. J., B. Peters, J. Sidney, A. Sette, and R. L. Hendricks. 2011. De-
Masse, and P. Druilhe. 2000. High immunogenicity in chimpanzees of peptides fining the herpes simplex virus-specific CD8+ T cell repertoire in C57BL/6 mice.
and lipopeptides derived from four new Plasmodium falciparum pre-erythrocytic J. Immunol. 186: 3927–3933.
molecules. Vaccine 18: 2843–2855. 57. Gilchuk, P., C. T. Spencer, S. B. Conant, T. Hill, J. J. Gray, X. Niu, M. Zheng,
33. Daubersies, P., A. W. Thomas, P. Millet, K. Brahimi, J. A. Langermans, J. J. Erickson, K. L. Boyd, K. J. McAfee, et al. 2013. Discovering naturally
B. Ollomo, L. BenMohamed, B. Slierendregt, W. Eling, A. Van Belkum, et al. processed antigenic determinants that confer protective T cell immunity. J. Clin.
2000. Protection against Plasmodium falciparum malaria in chimpanzees by Invest. 123: 1976–1987.
immunization with the conserved pre-erythrocytic liver-stage antigen 3. Nat. 58. BenMohamed, L., S. L. Wechsler, and A. B. Nesburn. 2002. Lipopeptide vac-
Med. 6: 1258–1263. cines—yesterday, today, and tomorrow. Lancet Infect. Dis. 2: 425–431.
34. Bottius, E., L. BenMohamed, K. Brahimi, H. Gras, J. P. Lepers, L. Raharimalala, 59. Nesburn, A. B., I. Bettahi, G. Dasgupta, A. A. Chentoufi, X. Zhang, S. You,
M. Aikawa, J. Meis, B. Slierendregt, A. Tartar, et al. 1996. A novel Plasmodium N. Morishige, A. J. Wahlert, D. J. Brown, J. V. Jester, et al. 2007. Functional
falciparum sporozoite and liver stage antigen (SALSA) defines major B, T Foxp3+ CD4+ CD25(Bright+) “natural” regulatory T cells are abundant in rabbit
helper, and CTL epitopes. J. Immunol. 156: 2874–2884. conjunctiva and suppress virus-specific CD4+ and CD8+ effector T cells during
35. McKinney, D. M., R. Skvoretz, B. D. Livingston, C. C. Wilson, M. Anders, ocular herpes infection. J. Virol. 81: 7647–7661.
R. W. Chesnut, A. Sette, M. Essex, V. Novitsky, and M. J. Newman. 2004. 60. Mott, K. R., A. A. Chentoufi, D. Carpenter, L. BenMohamed, S. L. Wechsler,
Recognition of variant HIV-1 epitopes from diverse viral subtypes by vaccine- and H. Ghiasi. 2009. The role of a glycoprotein K (gK) CD8+ T-cell epitope of
induced CTL. J. Immunol. 173: 1941–1950. herpes simplex virus on virus replication and pathogenicity. Invest. Ophthalmol.
36. Kessler, J. H., N. J. Beekman, S. A. Bres-Vloemans, P. Verdijk, P. A. van Veelen, Vis. Sci. 50: 2903–2912.
A. M. Kloosterman-Joosten, D. C. Vissers, G. J. ten Bosch, M. G. Kester, 61. Sidney, J., S. Southwood, D. L. Mann, M. A. Fernandez-Vina, M. J. Newman,
A. Sijts, et al. 2001. Efficient identification of novel HLA-A(*)0201-presented and A. Sette. 2001. Majority of peptides binding HLA-A*0201 with high affinity
cytotoxic T lymphocyte epitopes in the widely expressed tumor antigen PRAME crossreact with other A2-supertype molecules. Hum. Immunol. 62: 1200–1216.
by proteasome-mediated digestion analysis. J. Exp. Med. 193: 73–88. 62. Sette, A., M. Newman, B. Livingston, D. McKinney, J. Sidney, G. Ishioka, S. Tangri,
37. Sallusto, F., D. Lenig, R. Förster, M. Lipp, and A. Lanzavecchia. 1999. Two J. Alexander, J. Fikes, and R. Chesnut. 2002. Optimizing vaccine design for cellular
subsets of memory T lymphocytes with distinct homing potentials and effector processing, MHC binding and TCR recognition. Tissue Antigens 59: 443–451.
functions. Nature 401: 708–712. 63. Bettahi, I., A. B. Nesburn, S. Yoon, X. Zhang, A. Mohebbi, V. Sue,
38. Hikono, H., J. E. Kohlmeier, S. Takamura, S. T. Wittmer, A. D. Roberts, and A. Vanderberg, S. L. Wechsler, and L. BenMohamed. 2007. Protective immunity
D. L. Woodland. 2007. Activation phenotype, rather than central- or effector- against ocular herpes infection and disease induced by highly immunogenic self-
memory phenotype, predicts the recall efficacy of memory CD8+ T cells. J. Exp. adjuvanting glycoprotein D lipopeptide vaccines. Invest. Ophthalmol. Vis. Sci.
Med. 204: 1625–1636. 48: 4643–4653.
39. Ladell, K., M. K. Hellerstein, D. Cesar, R. Busch, D. Boban, and J. M. McCune. 64. Mackenzie, K. J., D. J. Nowakowska, M. D. Leech, A. J. McFarlane, C. Wilson,
2008. Central memory CD8+ T cells appear to have a shorter lifespan and reduced P. M. Fitch, R. A. O’Connor, S. E. Howie, J. Schwarze, and S. M. Anderton.
abundance as a function of HIV disease progression. J. Immunol. 180: 7907–7918. 2014. Effector and central memory T helper 2 cells respond differently to peptide
40. Channappanavar, R., B. S. Twardy, and S. Suvas. 2012. Blocking of PDL-1 immunotherapy. Proc. Natl. Acad. Sci. USA 111: E784–E793.
interaction enhances primary and secondary CD8 T cell response to herpes 65. Cornberg, M., S. C. Clute, L. B. Watkin, F. M. Saccoccio, S. K. Kim,
simplex virus-1 infection. PLoS ONE 7: e39757. Y. N. Naumov, M. A. Brehm, N. Aslan, R. M. Welsh, and L. K. Selin. 2010. CD8
41. Kao, C., K. J. Oestreich, M. A. Paley, A. Crawford, J. M. Angelosanto, T cell cross-reactivity networks mediate heterologous immunity in human EBV
M. A. Ali, A. M. Intlekofer, J. M. Boss, S. L. Reiner, A. S. Weinmann, and and murine vaccinia virus infections. J. Immunol. 184: 2825–2838.
E. J. Wherry. 2011. Transcription factor T-bet represses expression of the in- 66. Joshi, N. S., W. Cui, C. X. Dominguez, J. H. Chen, T. W. Hand, and S. M. Kaech.
hibitory receptor PD-1 and sustains virus-specific CD8+ T cell responses during 2011. Increased numbers of preexisting memory CD8 T cells and decreased T-
chronic infection. Nat. Immunol. 12: 663–671. bet expression can restrain terminal differentiation of secondary effector and
42. Banerjee, A., S. M. Gordon, A. M. Intlekofer, M. A. Paley, E. C. Mooney, memory CD8 T cells. J. Immunol. 187: 4068–4076.
T. Lindsten, E. J. Wherry, and S. L. Reiner. 2010. Cutting edge: the transcription 67. BenMohamed, L., Y. Belkaid, E. Loing, K. Brahimi, H. Gras-Masse, and
factor eomesodermin enables CD8+ T cells to compete for the memory cell P. Druilhe. 2002. Systemic immune responses induced by mucosal administra-
niche. J. Immunol. 185: 4988–4992. tion of lipopeptides without adjuvant. Eur. J. Immunol. 32: 2274–2281.
2248 VP11/12-SPECIFIC CD8+ T CELLS IN SYMPTOMATIC HERPES

68. Zhu, X., T. V. Ramos, H. Gras-Masse, B. E. Kaplan, and L. BenMohamed. 2004. 73. Selin, L. K., M. A. Brehm, Y. N. Naumov, M. Cornberg, S. K. Kim, S. C. Clute,
Lipopeptide epitopes extended by an Nepsilon-palmitoyl-lysine moiety increase and R. M. Welsh. 2006. Memory of mice and men: CD8+ T-cell cross-reactivity
uptake and maturation of dendritic cells through a Toll-like receptor-2 pathway and heterologous immunity. Immunol. Rev. 211: 164–181.
and trigger a Th1-dependent protective immunity. Eur. J. Immunol. 34: 3102– 74. Selin, L. K., M. Cornberg, M. A. Brehm, S. K. Kim, C. Calcagno, D. Ghersi,
3114. R. Puzone, F. Celada, and R. M. Welsh. 2004. CD8 memory T cells: cross-
69. Nesburn, A. B., I. Bettahi, X. Zhang, X. Zhu, W. Chamberlain, R. E. Afifi, reactivity and heterologous immunity. Semin. Immunol. 16: 335–347.
S. L. Wechsler, and L. BenMohamed. 2006. Topical/mucosal delivery of sub-unit 75. Khan, A. A., R. Srivastava, P. P. Lopes, C. Wang, T. T. Pham, J. Cochrane,
vaccines that stimulate the ocular mucosal immune system. Ocul. Surf. 4: 178– N. T. Thai, L. Gutierrez, and L. Benmohamed. 2014. Asymptomatic memory
187. CD8+ T cells: from development and regulation to consideration for human
70. El-Awar, N., J. H. Lee, C. Tarsitani, and P. I. Terasaki. 2007. HLA class I vaccines and immunotherapeutics. Hum. Vaccin. Immunother. 10: 945–963.
epitopes: recognition of binding sites by mAbs or eluted alloantibody confirmed 76. H€ufner, K., A. Horn, T. Derfuss, C. Glon, I. Sinicina, V. Arbusow, M. Strupp,
with single recombinant antigens. Hum. Immunol. 68: 170–180. T. Brandt, and D. Theil. 2009. Fewer latent herpes simplex virus type 1 and
71. Pekiner, F. N., E. Aytugar, G. Y. Demirel, and M. O. Borahan. 2013. HLA-A, B cytotoxic T cells occur in the ophthalmic division than in the maxillary and
(Class I) and HLA-DR, DQ (Class II) antigens in Turkish patients with recurrent mandibular divisions of the human trigeminal ganglion and nerve. J. Virol. 83:
aphthous ulceration and Behçet’s disease. Med. Princ. Pract. 22: 464–468. 3696–3703.
72. Castelli, E. C., D. S. Gil, L. C. Veiga, and J. L. de Camargo. 2005. Typing class I 77. Conrady, C. D., M. Zheng, D. U. Stone, and D. J. Carr. 2012. CD8+ T cells
HLA-A gene using a nested PCR-RFLP procedure. Braz. J. Med. Biol. Res. 38: suppress viral replication in the cornea but contribute to VEGF-C-induced
837–842. lymphatic vessel genesis. J. Immunol. 189: 425–432.

You might also like