Antigenic peptides recognized by virus-specific cytotoxic T lymphocytes (CTLs) are useful tools for studying the CTL responses exclusively among those who own the major histocompatibility complex (MHC) class I molecules that present the peptides. For widening the application, an efficient strategy to determine such epitopes in the context of a given MHC is highly desirable. A rapid and efficient strategy is presented for the determination of CTL epitopes in the context of given MHC molecules of interest through multiple screenings consisting of a computer-assisted algorithm and MHC stabilization and enzyme-linked immunospot assays. A major cytomegalovirus (CMV)–specific CTL epitope, QYDPVAALF, in the amino acid sequence of its lower matrix 65 kd phosphoprotein (pp65) presented by HLA-A*2402 molecules was identified from 83 candidate peptides. The results indicate that the CMV-specific CTL response is highly focused to pp65 in the context of HLA-A*2402. Endogenous processing and presentation was confirmed using a peptide-specific CD8+ T-cell clone as the effectors and autologous fibroblast cells infected with recombinant vaccinia virus expressing pp65 gene or CMV as antigen-presenting cells. Flow cytometric analysis of intracellular interferon-γ production revealed 0.04% to 0.27% of CD8+ T cells in peripheral blood of HLA-A24+ and CMV-seropositive donors to be specific for the peptide. The tetrameric MHC-peptide complexes specifically bound to the reactive T-cell clone and 0.79% of CD8+ T cells in peripheral blood from a seropositive donor. The peptide could be a useful reagent to study CTL responses to CMV among populations positive for HLA-A*2402.

Human cytomegalovirus (CMV) is a ubiquitous β-herpesvirus. Primary infection in healthy hosts is usually asymptomatic, and the virus persists in CD33+progenitors expressing markers of dendritic and myeloid lineage as major reservoirs of latent infection virus1,2 without any apparent clinical symptoms. However, in immunocompromised hosts, such as patients with advanced human immunodeficiency virus (HIV) infection and those who have undergone bone marrow transplantation (BMT), CMV is frequently reactivated and disease resulting from the progression of CMV infection is a major cause of infectious morbidity and mortality.3-5 

CD8+ cytotoxic T lymphocytes (CTLs) are essential to the control of CMV infection. In patients undergoing allogeneic BMT, the delayed regeneration of CMV-specific CD8+ T lymphocytes following transplantation has been correlated with CMV disease.5,6 In addition, adoptive transfer of CMV-specific CD8+ T-cell clones has led to restoration of virus-specific immunity in BMT patients with relatively low frequencies of CMV-related disease.7,8 In addition, recent data indicate that there is an inverse correlation between the CMV-specific CTL response and CMV antigenemia after renal transplantation.9 This observation underlines the potential benefit of testing for CMV-specific CTL responses in immunocompromised patients in order to monitor and control CMV infection.

Recently, we have shown that monitoring of the Epstein-Barr virus (EBV) load and EBV-specific CD8+ T-cell frequencies, determined by multiparameter flow cytometric analysis to detect rapid accumulation of interferon γ (IFN-γ) in antigen-reacting CD8+ T cells, is useful for care of EBV-related post-BMT lymphoproliferation.10-12 Antigen-presenting cells for the detection of EBV-specific CD8+ T cells are easily obtained and propagated by transforming peripheral blood B lymphocytes with a laboratory strain of EBV. However, detection of CMV-specific CD8+ T cells using the same approach is practically hampered by difficulties in obtaining the dermal fibroblast cells necessary for presenting CMV antigens to relevant CTLs.

Virus-specific CD8+ T cells recognize short peptides—8 to 10 amino acids in most cases—presented by major histocompatibility complex (MHC) class I molecules. Identification of CMV-specific CD8+ T-cell epitopes has brought advantages for studies of many aspects of the T-cell response. These peptides can directly stimulate the T cells for rapid IFN-γ production13-16 or directly stain the T cells after being assembled into fluorescent-labeled tetrameric MHC–peptide complexes.17-21 Both techniques are sufficiently sensitive and rapid for monitoring the CMV-specific CD8+ T-cell immunity in the clinical setting. In addition, such peptides have the potential of eliciting CMV-specific CTL responses in vivo when used as vaccines preceding CMV challenge.22 Staining with tetramers carrying the antigenic peptides could make it possible to sort fluorescent-labeled CTLs from peripheral blood mononuclear cells (PBMCs), and the sorted T cells could be expanded in vitro for large-scale production of reagents for adoptive transfer treatment.23 However, the antigenic peptides need to be identified in the context of HLA, and application is always restricted to patients who possess the allele. For widening the application, an efficient strategy to determine CMV-specific CD8+ T-cell epitopes in the context of a given HLA is highly desirable.

We present here a rapid and efficient strategy for determination of CTL epitopes in the context of HLA-A*2402 molecules through multiple screenings consisting of a computer-assisted algorithm, an in vitro MHC stabilization assay, and an enzyme-linked immunospot (ELISPOT) assay. In many ethnic groups, HLA-A24 is one of the most common alleles24,25 and, especially in our country, HLA-A24 is the most frequently encountered HLA class I allele and the genotype is almost exclusively A*2402.26 We here introduce an amino acid sequence from the lower matrix 65-kd phosphoprotein (pp65) of CMV, QYDPVAALF, as one of the the major CMV-specific CTL epitopes presented by HLA-A*2402 molecules.

Donors

The study design and purpose, which had been approved by the institutional review board of Aichi Cancer Center, were fully explained to all donors. Skin biopsies and peripheral blood were obtained after informed consent was confirmed. Standard serologic HLA typing was performed using peripheral blood for some donors.

Virus and cell lines

Dermal fibroblast lines were generated from skin biopsies obtained from the volunteers and propagated in Dulbecco modified Eagle medium (Gibco, Grand Island, NY) supplemented with 10% fetal calf serum (FCS), 2 mM L-glutamine, 50 U/mL penicillin, and 50 μg/mL streptomycin.

AD169 strain CMV (VR-538, American Type Culture Collection, Manassas, VA) was propagated in fibroblast cells infected at a multiplicity of infection 0.1. Whole infected cultures were harvested when a 100% cytopathic effect was evident. They were frozen, thawed once, and spun at 3000 rpm. Aliquots of the supernatant with a titer of 5 × 106 plaque-forming units per milliliter were stocked at −80°C until used.

The peptide transporter-negative B × T hybrid cell line 174 CEM.T2 (referred to as T2) was transfected with a plasmid expressing HLA-A*2402,27 a kind gift from Dr K. Itoh, Kurume University, Japan. The transfected cell line was cloned by limiting dilution and an A24+ clone screened by indirect immunofluorescence using an anti-A24 monoclonal antibody (mAb) (clone 0041HA, One Lambda, Canoga Park, CA), and antimouse fluorescein isothiocyanate (FITC)-labeled antibodies, named T2-A24, were further cultured in Iscoves modified Dulbecco medium (Gibco) supplemented with 2 mM L-glutamine, 50 U/mL penicillin, 50 μg/mL streptomycin, 5 × 10−5 M β-mercaptoethanol, 10% FCS, and 0.8 mg/mL G418 (Gibco).

Peptides

To identify the potential HLA-A24–binding peptides within CMV proteins (accession code X17403),28 a computer-based program was employed by access through the World Wide Web site BioInformatics and molecular analysis section (BIMAS) HLA peptide bonding predictions.29,30 Most peptides were synthesized with a Cleaved PepSet from Mimotope (Melbourne, Australia) dissolved in 100 μL dimethyl sulfoxide and further diluted in 40% acetonitrile, 0.1 M HEPES (pH 7.4), where necessary. The yield of each peptide was assumed to be 1 μM. HLA-A24–binding peptides RYLRDQQLL, derived from the HIV envelope protein,31 and TYGPVFMSL, derived from EBV latent membrane protein 2A,32 were synthesized (Sawady, Tokyo, Japan) and used as control peptides. The octamer QYDPVAAL of CMV pp65 used in the CTL assay was also synthesized (Sawady). Peptides used in this paper are listed in Table1.

Table 1.

Characters of synthetic oligopeptides and results with the MHC stabilization assay

Peptide no.Amino acid sequenceORF nameCharacterStart positionAmino acid lengthScore*% MFI increase
RYTAESLRL UL69 IE 259 400 227 
DYEPVPRKF   33 277 141 
VYNAPNLHTL   329 10 300 196 
AYCPFDEQSL   517 10 288 92 
DYLLHIRQQF   301 10 252 123 
TYRCGAVSDL   477 10 200 58 
VYLFSLVVL US3 IE 164 300 175 
VYDLANNTL IRS1 IE 218 240 194 
RYDDESWRPL   640 10 480 36 
10 FYQIRKPPWL   598 10 300 217 
11 VYDLANNTL TRS1 IE 218 240 136 
12 LYLALGAGF   206 180 79 
13 RYTRPTEPPL   631 10 400 17 
14 AYVCCQEYL UL36 Teg 69 300 172 
15 LYDAITENL   350 280 202 
16 CYHQLLGAL   231 240 114 
17 LYRGDVESL   156 200 155 
18 KYAESDYIF UL37 IE 353 240 177 
19 KYAESDYIFL   353 10 480 145 
20 SYRWIQRKRL   21 10 200 −20 
21 QYHLEGWFPL   262 10 200 109 
22 QYILGADPL UL123 IE 353 300 189 
23 KYTQTEEKF   107 220 142 
24 AYAQKIFKIL   248 10 336 152 
25 AYLGLSKKL UL48 Teg 433 396 146 
26 DYLRFPTRL   2026 300 104 
27 EYHGVYEHL   563 280 144 
28 TYRDRDIDL   223 240 −21 
29 TYSTLDRAL   470 240 165 
30 FYDLRDLKL   662 220 121 
31 TYAQMVKKL   880 220 212 
32 GYQKKVQQDL   1016 10 504 −29 
33 LYPEYIYTVL   1753 10 504 161 
34 TYVLVTVNSL   136 10 360 203 
35 YYEALFLYML   197 10 300 215 
36 DYGNVAFKYL   1609 10 240 47 
37 KYCDPNVLL UL56 Teg 27 480 149 
38 LYLLLYRHL   756 360 157 
39 TYNKEWPLL   735 300 202 
40 IYELCNGPL   617 300 133 
41 MYTKIKHIL   397 280 155 
42 VYHQLCRAL   320 240 148 
43 LYLEMLLKAL   72 10 432 36 
44 LYNETFGKQL   708 10 360 34 
45 SYIQEAEEAL   269 10 300 −9 
46 TYNKEWPLLL   735 10 300 238 
47 AYIYTTYLL UL55 gB 154 300 239 
48 SYENKTMQL   205 300 116 
49 AYEYVDYLF   624 210 225 
50 DYLFKRMIDL   629 10 300 −2.4 
51 IYNKPIAARF   531 10 210 169 
52 SYAYIYTTYL   152 10 200 228 
53 QYRSYKRSL UL100 gM 174 200 −16 
54 GYHLGAFFGL   268 10 240 157 
55 YYHVVDFERL   46 10 240 196 
56 KYESWLRPL UL115 gL 92 600 103 
57 YYAGLPPEL   277 264 196 
58 KYYAGLPPEL   276 10 528 172 
59 LYNAVKEFCL   253 10 300 185 
60 NYLDLSALL UL75 gH 306 432 192 
61 SYVVTNQYL   589 360 209 
62 SYLKDSDFL   291 300 174 
63 TYALVSKDL   125 280 178 
64 SYRSFSQQL   135 240 82 
65 TYGRPIRFL   45 200 169 
66 YYVFHMPRCL   88 10 300 227 
67 MYMHDSDDVL   668 10 300 133 
68 HYFTTLKQYL UL18 Env 344 10 288 182 
69 EYTRLGVWL US27 Env 31 240 91 
70 EYAKHHPKL   89 220 33 
71 IYQLFEYTRL   26 10 300 169 
72 QYLLDHNSL US28 Env 91 360 161 
73 FYVAMFASL   111 360 172 
74 LYVFVGTKF   290 231 71 
75 RYRPVKQACL   137 10 480 117 
76 SYPIILNVEL   183 10 396 183 
77 TYNCTYNRL UL4 Env 110 360 122 
78 HYRYEVANL   183 10 200 −22 
79 TYNRLTLLNL   114 10 300 59 
80 VYALPLKML UL83 Teg 100 200 151 
81 QYDPVAALF   328 168 215 
82 QYVKVYLESF   209 10 150 26 
83 QYDPVAALFF   328 10 120 185 
84 RYLRDQQLL   584 720 141 
85 TYGPVFMSL1-153   287 403 229 
Peptide no.Amino acid sequenceORF nameCharacterStart positionAmino acid lengthScore*% MFI increase
RYTAESLRL UL69 IE 259 400 227 
DYEPVPRKF   33 277 141 
VYNAPNLHTL   329 10 300 196 
AYCPFDEQSL   517 10 288 92 
DYLLHIRQQF   301 10 252 123 
TYRCGAVSDL   477 10 200 58 
VYLFSLVVL US3 IE 164 300 175 
VYDLANNTL IRS1 IE 218 240 194 
RYDDESWRPL   640 10 480 36 
10 FYQIRKPPWL   598 10 300 217 
11 VYDLANNTL TRS1 IE 218 240 136 
12 LYLALGAGF   206 180 79 
13 RYTRPTEPPL   631 10 400 17 
14 AYVCCQEYL UL36 Teg 69 300 172 
15 LYDAITENL   350 280 202 
16 CYHQLLGAL   231 240 114 
17 LYRGDVESL   156 200 155 
18 KYAESDYIF UL37 IE 353 240 177 
19 KYAESDYIFL   353 10 480 145 
20 SYRWIQRKRL   21 10 200 −20 
21 QYHLEGWFPL   262 10 200 109 
22 QYILGADPL UL123 IE 353 300 189 
23 KYTQTEEKF   107 220 142 
24 AYAQKIFKIL   248 10 336 152 
25 AYLGLSKKL UL48 Teg 433 396 146 
26 DYLRFPTRL   2026 300 104 
27 EYHGVYEHL   563 280 144 
28 TYRDRDIDL   223 240 −21 
29 TYSTLDRAL   470 240 165 
30 FYDLRDLKL   662 220 121 
31 TYAQMVKKL   880 220 212 
32 GYQKKVQQDL   1016 10 504 −29 
33 LYPEYIYTVL   1753 10 504 161 
34 TYVLVTVNSL   136 10 360 203 
35 YYEALFLYML   197 10 300 215 
36 DYGNVAFKYL   1609 10 240 47 
37 KYCDPNVLL UL56 Teg 27 480 149 
38 LYLLLYRHL   756 360 157 
39 TYNKEWPLL   735 300 202 
40 IYELCNGPL   617 300 133 
41 MYTKIKHIL   397 280 155 
42 VYHQLCRAL   320 240 148 
43 LYLEMLLKAL   72 10 432 36 
44 LYNETFGKQL   708 10 360 34 
45 SYIQEAEEAL   269 10 300 −9 
46 TYNKEWPLLL   735 10 300 238 
47 AYIYTTYLL UL55 gB 154 300 239 
48 SYENKTMQL   205 300 116 
49 AYEYVDYLF   624 210 225 
50 DYLFKRMIDL   629 10 300 −2.4 
51 IYNKPIAARF   531 10 210 169 
52 SYAYIYTTYL   152 10 200 228 
53 QYRSYKRSL UL100 gM 174 200 −16 
54 GYHLGAFFGL   268 10 240 157 
55 YYHVVDFERL   46 10 240 196 
56 KYESWLRPL UL115 gL 92 600 103 
57 YYAGLPPEL   277 264 196 
58 KYYAGLPPEL   276 10 528 172 
59 LYNAVKEFCL   253 10 300 185 
60 NYLDLSALL UL75 gH 306 432 192 
61 SYVVTNQYL   589 360 209 
62 SYLKDSDFL   291 300 174 
63 TYALVSKDL   125 280 178 
64 SYRSFSQQL   135 240 82 
65 TYGRPIRFL   45 200 169 
66 YYVFHMPRCL   88 10 300 227 
67 MYMHDSDDVL   668 10 300 133 
68 HYFTTLKQYL UL18 Env 344 10 288 182 
69 EYTRLGVWL US27 Env 31 240 91 
70 EYAKHHPKL   89 220 33 
71 IYQLFEYTRL   26 10 300 169 
72 QYLLDHNSL US28 Env 91 360 161 
73 FYVAMFASL   111 360 172 
74 LYVFVGTKF   290 231 71 
75 RYRPVKQACL   137 10 480 117 
76 SYPIILNVEL   183 10 396 183 
77 TYNCTYNRL UL4 Env 110 360 122 
78 HYRYEVANL   183 10 200 −22 
79 TYNRLTLLNL   114 10 300 59 
80 VYALPLKML UL83 Teg 100 200 151 
81 QYDPVAALF   328 168 215 
82 QYVKVYLESF   209 10 150 26 
83 QYDPVAALFF   328 10 120 185 
84 RYLRDQQLL   584 720 141 
85 TYGPVFMSL1-153   287 403 229 

ORF indicates open reading frame; IE, immediate early genes; Teg, (possible) tegument proteins; g, glycoproteins; Env, (possible) envelope proteins.

*

Estimated half-time of dissociation from the HLA-A24 allele (min).

Percent MFI increase of HLA-A*2402 molecules on T2-A24 cells. Percent MFI increase = (MFI with sample peptide − MFI without peptide)/MFI without peptide) × 100. See the MHC stabilization assay in “Materials and methods.”

Derived from the HIV envelope protein.31 

F1-153

Derived from the EBV latent membrane protein 2A.32 

MHC stabilization assay

The synthesized peptides were used in an MHC stabilization assay using T2-A24 cells as described earlier.33 Briefly, T2-A24 cells (2 × 105) were incubated with 200 μL RPMI 1640 (Sigma, St Louis, MO) containing 0.1% FCS, 5 × 10−5 M β-mercaptoethanol, and each of the peptides at a concentration of 10 μM at 26°C for 16 hours, followed by incubation at 37°C for 3 hours. After the incubation, surface HLA-A24 molecules were stained with the anti-A24 mAb and antimouse FITC-labeled antibodies. Expression was measured by FACScan (Becton Dickinson, San Jose, CA), and mean fluorescence intensity (MFI) was recorded. Percent MFI increase was calculated as follows: Percent MFI increase = (MFI with the given peptide − MFI without peptide)/(MFI without peptide) × 100.

Generation of polyclonal and clonal CMV-specific CTLs

CMV-specific polyclonal CTLs were generated as described earlier.34,35 Briefly, heparinized peripheral blood was obtained by venipuncture from a CMV-seropositive volunteer, and PBMCs were isolated by density gradient centrifugation over Ficoll-Hypaque (Pharmacia Biotech, Uppsala, Sweden). The PBMCs (1 × 107) were cultured with 5 × 105autologous fibroblast cells infected 2 hours previously with the AD169 strain of CMV at a multiplicity of infection of 5 in individual wells of a 6-well plate in a final volume of 6 mL RPMI 1640 supplemented with 2 mM L-glutamine, 50 U/mL penicillin, 50 μg/mL streptomycin, 5 × 10−5 M β-mercaptoethanol, and 10% FCS (referred to as culture medium). After 7 days, the cultures were restimulated with both autologous CMV-infected fibroblast cells and γ-irradiated (33 Gy) allogeneic PBMCs, and they were then supplemented with recombinant IL-2 (20 U/mL) 2 days thereafter. The CTL line was further stimulated weekly with autologous CMV-infected fibroblast cells, γ-irradiated allogeneic PBMCs, and IL-2.

To generate CMV-specific CTL clones, CD8+ T cells were enriched with the aid of magnetic beads (Dynal, Oslo, Norway) and plated in wells of 96-well round bottom plates at 0.3 and 1 cell per well with 5 × 104 γ-irradiated allogeneic PBMCs (33 Gy), 1 × 104 γ-irradiated EBV-transformed B-lymphoblastoid cells (55 Gy), and anti-CD3 mAb (Ortho Diagnostics, Raritan, NJ) at a concentration of 30 ng/mL.8,36 The next day, IL-2 was added to a final concentration of 50 U/mL. Fourteen days after the stimulation, T cells in positive wells were tested by ELISPOT assay for their specificity. The cloning efficiency was less than 5% in all plates.

ELISPOT assay

Flat-bottom, 96-well MultiScreen-HA plates with a nitrocellulose base (Millipore, Millipore Corporation, Bedford, MA) were coated with 10 μg/mL anti–IFN-γ mAb (R & D Systems, Minneapolis, MN) and incubated overnight at 4°C. After washing with phosphate-buffered saline (PBS), plates were blocked with the culture medium for 1 hour at 37°C. T2-A24 (5 × 104) cells were pulsed with each peptide in 100 μL RPMI 1640 with 0.1% FCS and 5 × 10−5 M β-mercaptoethanol in each well of the plates for 30 minutes at room temperature. A total of 10 000 polyclonal CD8+ T cells suspended in culture medium supplemented with additional FCS to 20% and 20 U/mL IL-2 were seeded in each well. When the spots were too many to count, 1000 CD8+ T cells were used as responder cells, and the numbers of spots were multiplied by 10 (Figure1). For testing the specificity of CMV-specific T-cell clones, indicated numbers of the T cells in figures were used as responders. The peptide concentrations described in the text and figures indicate those in the final assay volume. In some experiments, HLA-A24+ fibroblast cells were infected 2 hours previously with CMV AD169 strain, recombinant vaccinia virus expressing glycoprotein B, or pp65 of CMV (kind gifts from Dr S. R. Riddell, Fred Hutchinson Cancer Research Center, Seattle, WA) at a multiplicity of infection of 5 and used as antigen-presenting cells (10 000 cells/well). All assays were performed in duplicate. The plates were incubated in a 5% CO2 incubator at 37°C for 20 hours and extensively washed with PBS containing 0.05% Tween 20. A polyclonal rabbit anti–IFN-γ antibody (Genzyme, Cambridge, MA) was added to individual wells and left for 90 minutes at room temperature, followed by exposure to peroxidase-conjugated goat antirabbit immunoglobulin G (Genzyme) for an additional 90 minutes. For visualization of IFN-γ–specific spots, 100 μL of 0.1 M sodium acetate buffer (pH 5.0) containing 3-amino-9-ethylcarbazole (Sigma) and 0.015% H2O2 was added to each well. After 40 minutes, the reaction was stopped by washing with water and the plates were dried. Diffuse large spots were counted under a dissecting microscope.

Fig. 1.

Screening of HLA-A*2402–binding peptides from amino acid sequences of CMV proteins for CD8+ T-cell stimulation by ELISPOT assay.

A total of 10 000 polyclonal CMV-specific CD8+ T cells established from PBMCs of a CMV-seropositive and HLA-A24+donor were cocultured with 50 000 T2-A24 cells in each well in the presence of each peptide at a concentration of 10 μM. When the spots were too many to count, 1000 CD8+ T cells were used as responder cells and the numbers of spots were shown after being multiplied by 10. Only peptides that had been shown to bind to HLA-A*2402 molecules by MHC stabilization assay were tested by ELISPOT. An HLA-A24–binding peptide, RYLRDQQLL, derived from the HIV envelope protein was used as control peptide (env). In some wells, autologous fibroblast cells that had been infected 2 hours previously with CMV AD169 strain (CMV) or mock-infected (mock) were used as antigen-presenting cells (10 000 cells/well). Each bar represents the average number of spots in duplicate wells.

Fig. 1.

Screening of HLA-A*2402–binding peptides from amino acid sequences of CMV proteins for CD8+ T-cell stimulation by ELISPOT assay.

A total of 10 000 polyclonal CMV-specific CD8+ T cells established from PBMCs of a CMV-seropositive and HLA-A24+donor were cocultured with 50 000 T2-A24 cells in each well in the presence of each peptide at a concentration of 10 μM. When the spots were too many to count, 1000 CD8+ T cells were used as responder cells and the numbers of spots were shown after being multiplied by 10. Only peptides that had been shown to bind to HLA-A*2402 molecules by MHC stabilization assay were tested by ELISPOT. An HLA-A24–binding peptide, RYLRDQQLL, derived from the HIV envelope protein was used as control peptide (env). In some wells, autologous fibroblast cells that had been infected 2 hours previously with CMV AD169 strain (CMV) or mock-infected (mock) were used as antigen-presenting cells (10 000 cells/well). Each bar represents the average number of spots in duplicate wells.

Close modal

CTL assay

CTL assays were performed employing 51Cr release as previously described.10,11 Briefly,51Cr-labeled T2 or T2-A24 cells (2000 cells/well) were pulsed with each peptide in 100 μL RPMI 1640 with 0.1% FCS and 5 × 10−5 M β-mercaptoethanol in each well of a V-bottom 96-well plate for 30 minutes at room temperature. Desired numbers of CTLs suspended in 100 μL of the culture medium supplemented with additional FCS to 20% were seeded in each well. The peptide concentrations described in the text and figures indicate those in the final assay volume. Each assay was performed in triplicate. After 5 hours of incubation, the supernatants were harvested and radioactivity counted with a γ-counter. Percent specific lysis was calculated as follows: Percent specific lysis = (experimental lysis − minimum lysis) × 100/(maximum lysis − minimum lysis). Minimum lysis was obtained by incubating the target cells with the culture medium alone. Maximum lysis was obtained by exposing the target cells to 1% Nonidet P-40.

Detection of IFN-γ–producing CD8+ T cells in response to the peptide by flow cytometry

Peptide-specific CD8+ T-cell frequencies were measured as previously described with slight modifications.11,13 Briefly, 4 × 106 PBMCs were incubated with 10 μM peptides in 1 mL culture medium supplemented with 20 U/mL IL-2 in a culture tube at 37°C for 6 hours in the presence of brefeldin A (Sigma) during the last 5 hours. After the incubation, the cell suspensions were fixed with 4% paraformaldehyde in PBS for 10 minutes at room temperature. After washing with PBS, cells were permeabilized with IC Perm (BioSource International, Camarillo, CA) and stained with phycoerythrin-cyanin-5.1–labeled anti-CD8, phycoerythrin-labeled anti-CD69 (Coulter, Miami, FL) and FITC-labeled antihuman IFN-γ (Coulter) mAbs. Stained cells were analyzed by FACScan using Lysis II software. Live gating of the CD8+ subset was performed, and 50 000 events were acquired for each analysis.

Tetramer production and staining

MHC-peptide tetramers were produced as described previously.17-21,23 Briefly, BL21(DE3) pLysS (Novagen, Madison, WI) competent cells were transformed with pET11d plasmid (Novagen) encoding HLA-A*2402 heavy chain (a kind gift from Dr H. Takasu, Sumitomo Pharmaceutical, Osaka, Japan) or pET-3a plasmid (Novagen) encoding β2-microglobulin (a kind gift from Dr H. Takizawa, Otsuka Pharmaceutical, Tokushima, Japan) to produce the recombinant proteins. Expression of the HLA heavy chain was limited to the extracellular domain, and C-terminus of their domain was modified by the addition of a substrate sequence for the biotinylating enzyme BirA. Monomeric HLA-peptide complexes were folded in vitro by adding the HLA protein to β2-microglobulin in the presence of the 9-mer, QYDPVAALF. Proteins were dialyzed against water and then concentrated. After purification on gel filtration, the MHC complex was biotinylated by using recombinant BirA enzyme (Avidity, Denver, CO) and then purified by gel filtration. HLA-peptide tetramers were made by mixing the biotinylated MHC with phycoerythrin-labeled streptavidin (Molecular Probes, Eugene, OR) at a molar ratio of 4:1. Tetramers were purified by gel filtration on a Superdex 200 HR 10/30 (Amersham Pharmacia Biotech, Uppsala, Sweden) and concentrated to 3 mg/mL of total proteins and stored at 4°C until use. PBMCs (2 × 106) or CTL clones (2 × 105) were stained with the tetramer at a concentration of 0.1 mg/mL tetramers and Tricolor anti-CD8 mAb (Caltag, Burlingame, CA) at 37°C for 15 minutes. After washing twice, stained cells were fixed with 0.5% paraformaldehyde before analysis with FACScan.

Selection of potential HLA-A24–binding peptides within CMV proteins

To identify potential HLA-A24–binding peptides within amino acid sequences of immediate early (IE), tegument and envelope proteins of CMV were analyzed with a computer program designed to predict HLA-binding peptides, based on estimation of the half-time dissociation of the HLA-peptide complex by access through the World Wide Web site BioInformatics and molecular analysis section (BIMAS) HLA peptide bonding predictions.29 A total of 83 peptides with estimated half-time dissociation scores above 100 were selected (Table1). All the peptides share the HLA-A24–binding motifs as tyrosine at the second residue and phenylalanine or leucine at the ninth or tenth residue.37-39 Next, MHC stabilization assays were performed to test these peptides for HLA-A*2402 binding efficiency using T2-A24 cells. Most peptides increased the HLA-A24 expression on the cells, indicating that these peptides bound and stabilized the HLA complex on the cell surface, but peptides no. 20, 28, 32, 45, 50, and 53 gave negative values for the percent MFI (Table 1) and were excluded from further studies.

Screening of peptides antigenic for an anti-CMV polyclonal CTL line by ELISPOT assay

To identify peptides recognized by CMV-specific CTLs in the context of HLA-A*2402 molecules, the ELISPOT assay was performed using T2-A24 cells as antigen-presenting cells. We chose the assay for the screening because it can test many peptides simultaneously in a few plates using relatively small numbers of CTLs as responders. Moreover, because the sensitive nature of the assay enables 40 spots to be scored as positive, subdominant epitopes could be identified when T cells recognizing such epitopes were present at concentrations no less than 40 in 10 000 cells. As shown in Figure 1, the polyclonal CMV-specific CD8+ T cells established from PBMCs of a CMV-seropositive and HLA-A24+ donor produced significant numbers of IFN-γ spots when incubated with T2-A24 cells pulsed with CMV peptides nos. 60, 80, 81, 82, and 83. Peptide no. 60 was derived from glycoprotein H, and others were from pp65 (open reading frame name UL83). The responders produced negligible spots when incubated with mock-infected autologous fibroblast cells, while more than 1000 spots were detected with CMV-infected autologous fibroblast cells. To ascertain the nature of the peptides recognized by the CMV-specific CD8+ T-cell line, we performed ELISPOT assays with serial dilutions of the 4 peptides derived from pp65. Figure 2shows that peptide no. 81 was still recognized at a concentration of 1 pM. The end points for recognition with the others were 10 pM for peptide no. 83 and 10 nM for peptides no. 80 and 82. The 2 peptides, no. 81 and 83, that were recognized at picomolar levels share the amino acid sequence QYDPVAALF, with an additional C-terminal phenylalanine for no. 83.

Fig. 2.

Results of ELISPOT assays using serial dilutions of the 4 peptides derived from pp65.

The 2 peptides, no. 81 (●) and 83 (▪), that were recognized at picomolar levels share the amino acid sequence QYDPVAALF, with an additional C-terminal phenylalanine for no. 83. The amino acid sequences of peptides no. 80 (♦) and 82 (▴) are VYALPLKML and QYVKVYLESF, respectively. A total of 1000 polyclonal CMV-specific CD8+ T cells established from PBMCs of a CMV-seropositive and HLA-A24+ donor were cocultured with 50 000 T2-A24 cells in each well in the presence of each peptide at the indicated concentrations.

Fig. 2.

Results of ELISPOT assays using serial dilutions of the 4 peptides derived from pp65.

The 2 peptides, no. 81 (●) and 83 (▪), that were recognized at picomolar levels share the amino acid sequence QYDPVAALF, with an additional C-terminal phenylalanine for no. 83. The amino acid sequences of peptides no. 80 (♦) and 82 (▴) are VYALPLKML and QYVKVYLESF, respectively. A total of 1000 polyclonal CMV-specific CD8+ T cells established from PBMCs of a CMV-seropositive and HLA-A24+ donor were cocultured with 50 000 T2-A24 cells in each well in the presence of each peptide at the indicated concentrations.

Close modal

Recognition of the 4 pp65 peptides by single CMV-specific CD8+ T-cell clones

Notably, the sum of the number of T cells that reacted with each of the 5 peptides exceeded that reacting with CMV-infected fibroblast cells (Figure 1). This could be due to either weaker antigen presentation on the CMV-infected fibroblast cells, possibly by the mechanism of class I molecule down-regulation,40-44 or the same T-cell repertoires being reactive with some of the 5 peptides. To test these possibilities, we established T-cell clones from limiting dilution culture of the bulk CTL line stimulated with anti-CD3 mAb, appropriate feeder cells, and IL-2. The results of ELISPOT assays with 2 representative T-cell clones are shown in Figure3. Both clones recognized all 4 peptides at high concentrations. The effect of peptide dilution was the same with bulk CD8 T cells and with the T-cell clones (Figures 2 and 3). Among a total of 54 clones established, 28 clones were reactive for all 4 peptides but not for peptide no. 60—at a concentration of 10 μM—and effects of dilution on recognition were similar for all the clones tested (data not shown). Thus, we conclude that the spots in the wells containing each of the 4 pp65 peptides were produced by the same T-cell repertoires.

Fig. 3.

Recognition of the 4 peptides derived from pp65 by each CMV-specific CD8+ T-cell clone.

The results of ELISPOT assay with CMV-specific CD8+ T-cell clone no. 1 (A) and no. 3 (B) are shown. A total of 100 CD8+ T cells were cocultured with 50 000 T2-A24 cells in each well in the presence of each peptide at the indicated concentrations. The 2 peptides, no. 81 (●) and 83 (▪), that were recognized at picomolar levels share the amino acid sequence QYDPVAALF, with an additional C-terminal phenylalanine for no. 83. The amino acid sequences of peptides no. 80 (♦) and 82 (▴) are VYALPLKML and QYVKVYLESF, respectively.

Fig. 3.

Recognition of the 4 peptides derived from pp65 by each CMV-specific CD8+ T-cell clone.

The results of ELISPOT assay with CMV-specific CD8+ T-cell clone no. 1 (A) and no. 3 (B) are shown. A total of 100 CD8+ T cells were cocultured with 50 000 T2-A24 cells in each well in the presence of each peptide at the indicated concentrations. The 2 peptides, no. 81 (●) and 83 (▪), that were recognized at picomolar levels share the amino acid sequence QYDPVAALF, with an additional C-terminal phenylalanine for no. 83. The amino acid sequences of peptides no. 80 (♦) and 82 (▴) are VYALPLKML and QYVKVYLESF, respectively.

Close modal

Recognition of the nonamer QYDPVAALF at a lower concentration than the octamer or the decamer

Because no. 81 and 83, which were recognized at picomolar levels, share the amino acid sequence QYDPVAALF and amino acid sequences of the peptides no. 80 (VYALPLKML) and 82 (QYVKVYLESF) were irrelevant, QYDPVAALF should be the core sequence of the T-cell epitope. Because class I molecules usually accommodate 8 to 10 amino acids in their antigen-binding sites, the octamer QYDPVAAL was simultaneously tested in CTL assays for its recognition by CTL clone no. 1. As shown in Figure 4, it far less effectively sensitized T2-A24 cells for CTL-mediated cell lysis than the nonamer or the decamer. We did not make and test the octamer YDPVAALF because tyrosine at the second residue is essential for binding to HLA-A24 molecules.37-39 Again, only the nonamer QYDPVAALF (peptide no. 81) could sensitize the target cells for CTL-mediated cell lysis at the concentration of 1 pM. In combination with the results of ELISPOT assays (Figures 2 and 3), this indicates that the nonamer QYDPVAALF is the minimal CTL epitope.

Fig. 4.

CMV-specific CD8+ CTL clone no. 1 recognition.

Clone no 1. recognizes the nonamer QYDPVAALF (●, peptide no. 81) and the decamer QYDPVAALFF (▪, peptide no. 83) at lower concentrations than octamer QYDPVAAL (▴). ■ indicates TYGPVFMSL derived from EBV latent membrane protein 2A. CTL assays were performed using 51Cr-labeled T2-A24 cells as targets at an effector-target ratio of 5:1 in the presence of each peptide at the indicated concentrations.

Fig. 4.

CMV-specific CD8+ CTL clone no. 1 recognition.

Clone no 1. recognizes the nonamer QYDPVAALF (●, peptide no. 81) and the decamer QYDPVAALFF (▪, peptide no. 83) at lower concentrations than octamer QYDPVAAL (▴). ■ indicates TYGPVFMSL derived from EBV latent membrane protein 2A. CTL assays were performed using 51Cr-labeled T2-A24 cells as targets at an effector-target ratio of 5:1 in the presence of each peptide at the indicated concentrations.

Close modal

Recognition by the QYDPVAALF-specific CD8+ T-cell clone of the epitope in the context of HLA-A*2402 molecules

To confirm the HLA restriction, CTL assays were performed using51Cr-labeled T2 or T2-A24 cells pulsed with QYDPVAALF or the control peptide TYGPVFMSL, derived from EBV latent membrane protein 2A (Figure 5). Only T2-A24 cells pulsed with QYDPVAALF were effectively lysed by the CTL clone no. 1. The results indicate that the epitope QYDPVAALF is presented by HLA-A*2402 molecules.

Fig. 5.

Recognition by the QYDPVAALF-specific CD8+T-cell clone of the epitope in the context of the HLA-A*2402 molecules.

CTL assays were performed using 51Cr-labeled T2-A24 cells pulsed with QYDPVAALF (▪) or control EBV peptide TYGPVFMSL (▴) and using T2 cells pulsed with QYDPVAALF (■) or with the control peptide (▵) as target cells, incubated with CMV-specific CD8+ CTL clone no. 1 at the indicated effector-target ratios in the presence of 100 nM of each peptide.

Fig. 5.

Recognition by the QYDPVAALF-specific CD8+T-cell clone of the epitope in the context of the HLA-A*2402 molecules.

CTL assays were performed using 51Cr-labeled T2-A24 cells pulsed with QYDPVAALF (▪) or control EBV peptide TYGPVFMSL (▴) and using T2 cells pulsed with QYDPVAALF (■) or with the control peptide (▵) as target cells, incubated with CMV-specific CD8+ CTL clone no. 1 at the indicated effector-target ratios in the presence of 100 nM of each peptide.

Close modal

Recognition by the QYDPVAALF-specific CD8+ T-cell clone of A*2402+ fibroblast cells infected either CMV or recombinant vaccinia virus expressing pp65

A set of experiments was performed to confirm that the epitope QYDPVAALF is processed within the cells infected with CMV. For that purpose, autologous and allogeneic HLA-A24+ fibroblast cells were mock-infected or CMV-infected and used as antigen-presenting cells for ELISPOT assays using the CTL clone no. 1 as responders. Figure 6 demonstrates that the CMV-infected cells stimulated for IFN-γ production by the T cells. The T cells were cytolytic for the CMV-infected fibroblast cells (data not shown). To confirm that endogenously produced pp65 proteins were processed and presented to the QYDPVAALF-specific CD8+ T cells, autologous fibroblast cells were infected with recombinant vaccinia virus expressing pp65 or glycoprotein B of CMV. The results clearly showed only fibroblast cells infected with recombinant vaccinia virus expressing pp65 to stimulate the T cells for IFN-γ production. The data indicated that pp65 expressed within the cells is endogenously processed to yield the epitope.

Fig. 6.

Recognition by the QYDPVAALF-specific CD8+ T-cell clone of A24+ fibroblast cells infected either with CMV or recombinant vaccinia virus expressing pp65.

HLA-A24+ autologous and allogeneic fibroblast cells were mock- or CMV-infected and tested for stimulation of QYDPVAALF-specific CD8+ T-cell clone no. 1 in the ELISPOT assay. Autologous fibroblast cells infected with recombinant vaccinia virus expressing pp65 or glycoprotein B (gB) of CMV were also tested for stimulation of the T cells. A total of 200 (▪), 100 (), 50 (■), or 25 (▨) CD8+ T cells were cocultured with 10 000 fibroblast cells in each well. Each bar represents the average number of spots in duplicate wells.

Fig. 6.

Recognition by the QYDPVAALF-specific CD8+ T-cell clone of A24+ fibroblast cells infected either with CMV or recombinant vaccinia virus expressing pp65.

HLA-A24+ autologous and allogeneic fibroblast cells were mock- or CMV-infected and tested for stimulation of QYDPVAALF-specific CD8+ T-cell clone no. 1 in the ELISPOT assay. Autologous fibroblast cells infected with recombinant vaccinia virus expressing pp65 or glycoprotein B (gB) of CMV were also tested for stimulation of the T cells. A total of 200 (▪), 100 (), 50 (■), or 25 (▨) CD8+ T cells were cocultured with 10 000 fibroblast cells in each well. Each bar represents the average number of spots in duplicate wells.

Close modal

Frequencies of QYDPVAALF-specific CD8+ T cells in PBMCs of healthy CMV-seropositive donors

We examined QYDPVAALF-specific CD8+ T-cell frequencies in PBMCs of healthy HLA-A24+, CMV-seropositive, and HIV-seronegative donors using tricolor flow cytometry analysis for detection of antigen-specific IFN-γ–producing T cells.10,13 First, the phycoerythrin-cyanin-5.1–labeled anti-CD8 mAb was used for gating the population. Second, the phycoerythrin-labeled anti-CD69 mAb was employed for enhancement of precise detection of responding T cells. CD69 is up-regulated on activated T cells prior to cytokine production and thus allows more definitive clustering of the true responding fraction.10,13 As a control peptide, we used HLA-A24–binding peptide RYLRDQQLL, derived from the HIV envelope protein.31 

When PBMCs from long-term healthy HLA-A24+ and CMV-seropositive individuals (n = 5) were stimulated with peptide QYDPVAALF, 0.04% to 0.27% of total CD8+ T cells produced IFN-γ (Figure 7). The values always exceeded those with control peptide, demonstrating specific reactivity to the CMV peptide. Frequencies of IFN-γ–producing CD8+T cells in PBMCs from CMV-seronegative and HLA-A24+individuals (n = 5) after stimulation with the peptide were below 0.01% (data not shown). These data demonstrate significant populations of CD8+ T cells from long-term healthy HLA-A24+and CMV-seropositive individuals to be specific to the peptide QYDPVAALF.

Fig. 7.

Frequencies of QYDPVAALF-specific CD8+ T cells in PBMCs of healthy HLA-A24+ and CMV-seropositive donors.

PBMCs were stimulated with either the CMV peptide QYDPVAALF or the control peptide RYLRDQQLL, derived from the HIV envelope protein, for IFN-γ production. After fixation and permeabilization, the cells were stained for CD8, CD69, and IFN-γ. The CD8+ subset was gated and analyzed by flow cytometry. The frequency of IFN-γ–producing cells is shown as the percentage of the total CD8+ T cells. The donors were all seronegative for HIV.

Fig. 7.

Frequencies of QYDPVAALF-specific CD8+ T cells in PBMCs of healthy HLA-A24+ and CMV-seropositive donors.

PBMCs were stimulated with either the CMV peptide QYDPVAALF or the control peptide RYLRDQQLL, derived from the HIV envelope protein, for IFN-γ production. After fixation and permeabilization, the cells were stained for CD8, CD69, and IFN-γ. The CD8+ subset was gated and analyzed by flow cytometry. The frequency of IFN-γ–producing cells is shown as the percentage of the total CD8+ T cells. The donors were all seronegative for HIV.

Close modal

Tetramer staining

Finally, we made fluorescent-labeled tetrameric MHC-peptide complexes (tetramer) using the nonamer peptide QYDPVAALF. The tetramers specifically bound to the peptide-reactive T-cell clone but not the unreactive clone (Figure 8A,B). QYDPVAALF-specific CD8+ T cells detected by the tetramers were 0.79% in peripheral blood from the donor 5 in Figure 7and 0.0% in that from an HLA-A24+ and CMV-seronegative donor (Figure 8C,D).

Fig. 8.

HLA-A*2402–QYDPVAALF tetramers specifically bound to the relevant CD8+ T cells.

An HLA-A*2402–restricted EBV-specific CD8+ CTL clone (A), the HLA-A*2402–restricted QYDPVAALF-specific CD8+ CTL clone no. 1 (B), peripheral lymphocytes from a CMV-seronegative and HLA-A*2402+ donor (C), and those from the donor 5 in Figure 7 (D) were stained phycoerythrin-conjugated HLA-A*2402–QYDPVAALF tetrameric complex and an antibody to CD8 conjugated to Tricolor. The cells were analyzed by flow cytometry. The frequency of QYDPVAALF-specific CD8+ T cells in peripheral lymphocytes is shown as the percentage of the total CD8+ T cells (C,D).

Fig. 8.

HLA-A*2402–QYDPVAALF tetramers specifically bound to the relevant CD8+ T cells.

An HLA-A*2402–restricted EBV-specific CD8+ CTL clone (A), the HLA-A*2402–restricted QYDPVAALF-specific CD8+ CTL clone no. 1 (B), peripheral lymphocytes from a CMV-seronegative and HLA-A*2402+ donor (C), and those from the donor 5 in Figure 7 (D) were stained phycoerythrin-conjugated HLA-A*2402–QYDPVAALF tetrameric complex and an antibody to CD8 conjugated to Tricolor. The cells were analyzed by flow cytometry. The frequency of QYDPVAALF-specific CD8+ T cells in peripheral lymphocytes is shown as the percentage of the total CD8+ T cells (C,D).

Close modal

The identification of new CTL epitopes is generally time-consuming and labor-intensive and consists of (1) establishing virus-specific CTL clones; (2) testing a panel of allogeneic virus-infected target cells that share a part of MHC class I molecules with the CTLs to determine the MHC restriction; (3) transfecting or transducing cells expressing the appropriate MHC class I molecules with each of the segregated virus genes for the target of CTL; (4) making overlapping peptides, eg, 20 amino acid peptides overlapping by 10 amino acids to span a given antigen length for testing the specificity of the T cells; and, finally, (5) mapping the sequences with truncated peptides to determine the minimal epitope. We show in this article a rapid and efficient strategy to find new CTL epitopes, especially presented by MHC molecules of interest. An important step in this process is the use of a computer-based program designed to predict potential HLA-binding peptides within viral proteins.29,33,45 Analysis of amino acid sequences of various proteins of CMV AD169 strain revealed a number of potential HLA-A24–binding peptides, and most of them then functionally stabilized HLA-A*2402 molecules that had been expressed on the peptide transporter-deficient cell line T2. Next and equally important is screening of the peptides by ELISPOT assay for their recognition by bulk CMV-specific CTLs, established from PBMCs of a CMV-immune donor positive for HLA-A24 typing. The ELISPOT assay has many advantages in this regard. First, it allows results with a number of different peptides to be generated simultaneously in a few plates within 3 days. Second, the assay demands relatively small numbers of CTLs, eg, 25 to 100 000 in a well, depending on the concentration of specific T cells. Third, the sensitive and specific nature of the assay enables it to detect minor populations of antigen-specific T cells. For example, 40 spots in 100 000 CD8+ T cells in a well could be scored positive as long as the numbers of background spots were sufficiently low. Fourth, it needs no radioisotopes.

Target antigens of CMV-specific CD8+ T cells have been reported to be restricted to IE proteins,46,47glycoprotein B,34,46 and tegument proteins such as pp65.35,47,48 Thus, we first searched candidate peptides that have the HLA-A24–binding motif by a computer-assisted algorithm among proteins in the IE gene family, glycoproteins, and tegument proteins to find as many epitopes as possible. Among 83 peptides synthesized, 6 were unable to stabilize HLA-A*2402 molecules expressed on T2 cells at all and were excluded from further analysis. The ELISPOT assay using CMV-specific CD8+ T cells maintained by weekly stimulation with CMV-infected autologous fibroblast cells and IL-2 revealed that the T cells recognized almost exclusively peptides derived from pp65 (Figure 1). The results underline the observations of McLaughlin-Taylor et al,35 who showed that pp65 is a target antigen for CMV-specific CTLs in most seropositive donors tested, and the finding of Wills et al47 that 70% to 90% of CMV-specific CTL precursors are pp65-specific. It has been reported that pp65-specific CTL clones recognize CMV-infected target cells in the absence of viral gene expression.34,35 The capacity of CMV to interfere with class I MHC presentation at an early time after infection40-44 might explain why the specificity of the CTL response should be focused on structural proteins, which are relatively abundant immediately after infection, derived from input virus. The pp65 is a major constituent of the virus tegument43 and consequently available to the class I processing pathway immediately after infection. Our data confirm and extend the previous observations in the context of HLA-A*2402 molecules.

Both the bulk CMV-specific CTL line and the CD8+T-cell clones simultaneously recognized peptides no. 80 (VYALPLKML), 81 (QYDPVAALF), 82 (QYVKVYLESF), and 83 (QYDPVAALFF) at concentrations higher than 10 nM. However, only peptides no. 81 and 83, which share a core sequence QYDPVAALF, were able to sensitize T cells at concentrations lower than 100 pM (Figures 2 and 3). Actually, peptides no. 80 (VYALPLKML) and 82 (QYVKVYLESF), which were resynthesized independently, were not able to sensitize the T cells even at a concentration of 10 μM (data not shown). Thus, we reason that the recognition of peptides no. 80 and 82 could be due to cross-contamination between peptides in adjacent wells during synthesis, transfer, storage, or handling and that QYDPVAALF(F) is the T-cell epitope. Because the octamer QYDPVAAL was far less effectively recognized by the T cells (Figure 4), we concluded that the nonamer QYDPVAALF is the minimal epitope. This also indicates that phenylalanine at the C-terminal residue of the nonamer is important for binding to HLA-A24 molecules.38 We did not synthesize octamer YDPVAALF for testing because tyrosine at the second residue is reported to be critical for binding to HLA-A24 molecules.37-39 We do not know yet which is the naturally processed peptide, QYDPVAALF or QYDPVAALFF, or whether they both could be. To answer this question, peptides eluted from HLA-A*2402 molecules of CMV-infected cells should be analyzed for their sequences or molecular weights after fractionating on high-performance liquid chromatography.49 However, the minimal epitope QYDPVAALF can stimulate specific T cells for IFN-γ production (Figure 7) and was durable for making tetrameric MHC complexes (Figure 8). Thus, we did not perform experiments to determine the naturally processed sequence.

The ELISPOT assay demonstrated numbers of CD8+ T cells reacting with the minimal epitope, QYDPVAALF (no. 81), to exceed half of those reacting with autologous CMV-infected fibroblast cells (Figure1), indicating that CMV-specific CD8+ T-cell responses in the donor are highly focused on pp65 and especially the HLA-A*2402–binding peptide, QYDPVAALF. This was also confirmed by the evidence that, among 54 CD8+ T-cell clones established with anti-CD3 mAb stimulation, 28 clones turned out to be specific for the epitope. Thus, as far as the donor is concerned, HLA-A*2402 molecules are dominant presenters of CMV antigens, and QYDPVAALF is a definitely major epitope of CMV-specific CD8+ T cells. To extend this observation, we performed flow cytometric analysis for detecting IFN-γ produced within QYDPVAALF-reactive CD8+ T cells in PBMCs of HLA-A24+ and CMV-seropositive donors. The fact that 0.04% to 0.27% of peripheral CD8+ T cells of the donors specifically produced IFN-γ and the tetramers stained 0.79% of CD8+ T cells of a seropositive individual suggests that the peptide has potential to be a good reagent as a representative CTL antigen to monitor CMV-specific CD8+T-cell responses among HLA-A*2402+ individuals in various clinical settings. Although we tested only limited numbers of donors, the frequencies shown here are relatively lower than those of HLA-A*0201– and HLA-B*0701–restricted pp65-specific CD8+T cells in other populations.17-20 The reason is unclear so far and awaits further study.

Peptide no. 60, NYLDLSALL, derived from glycoprotein H was recognized by bulk CTL at the concentration of 10 μM but not by the pp65-specific CTL clones at the same concentration, indicating that it is a minor epitope presented by HLA-A*2402 molecules; however, this could not be confirmed because CTL clones that specifically recognize NYLDLSALL have not been established so far.

In conclusion, we have identified a possible major CMV-specific CTL epitope from the amino acid sequence of pp65 presented by HLA-A*2402 molecules. Minimal CMV-specific CTL epitopes so far reported are exclusively within the pp65 (Table 2). Our data are in line with the highly focused reaction of CMV-specific CTLs to pp65 in the context of HLA-A*2402 molecules, as is the case with HLA-A*020118,20,21,48 and HLA-B*0702.17The newly identified peptide QYDPVAALF could be used for detection of CMV-specific CD8+ T-cell responses as a representative CTL antigen. In addition, it might be a candidate reagent for peptide vaccines for prevention of CMV disease or expansion of CMV-specific CTLs in vitro for the purpose of adoptive transfer treatment of immunocompromised patients positive for HLA-A*2402.

Table 2.

Minimal CD8+ T-cell epitopes of the human CMV protein pp65

SequenceStart positionAmino acid lengthHLAReference no.
RPHERNGFTV 265 10 B7 47  
TPRVTGGGAM 417 10 B7 47  
NLVPMVATV 495 A2 22,47  
VLGPISGHV 14 A2 50  
MLNIPSINV 120 A2 50  
EFFWDANDIY 511 10 B12(44) 47  
IPSINVHHY 123 B35 51  
QYDPVAALF 328 A*2402 This article 
SequenceStart positionAmino acid lengthHLAReference no.
RPHERNGFTV 265 10 B7 47  
TPRVTGGGAM 417 10 B7 47  
NLVPMVATV 495 A2 22,47  
VLGPISGHV 14 A2 50  
MLNIPSINV 120 A2 50  
EFFWDANDIY 511 10 B12(44) 47  
IPSINVHHY 123 B35 51  
QYDPVAALF 328 A*2402 This article 

Longer peptides containing CTL epitopes have been reported47,51 that are not listed here.

Supported by grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science and Technology of Japan (No. 13218152) and the Japan Society for Promotion of Science (No. 12670802 and RFTF 97L00703).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Taylor-Wiedeman
J
Sissons
P
Sinclair
J
Induction of endogenous human cytomegalovirus gene expression after differentiation of monocytes from healthy carriers.
J Virol.
68
1994
1597
1604
2
Kondo
K
Xu
J
Mocarski
ES
Human cytomegalovirus latent gene expression in granulocyte-macrophage progenitors in culture and in seropositive individuals.
Proc Natl Acad Sci U S A.
93
1996
11137
11142
3
Britt
WJ
Alford
CA
Cytomegalovirus.
Fields Virology.
3rd ed.
Fields
BN
Knipe
DM
Howley
PM
et al
1996
2493
2523
Lippincott-Raven Publishers
Philadelphia, PA
4
Peters
BS
Beck
EJ
Anderson
S
et al
Cytomegalovirus infection in AIDS: patterns of disease, response to therapy and trends in survival.
J Infect.
23
1991
129
137
5
Reusser
P
Riddell
SR
Meyers
JD
Greenberg
PD
Cytotoxic T-lymphocyte response to cytomegalovirus after human allogeneic bone marrow transplantation: pattern of recovery and correlation with cytomegalovirus infection and disease.
Blood.
78
1991
1373
1380
6
Li
CR
Greenberg
PD
Gilbert
MJ
Goodrich
JM
Riddell
SR
Recovery of HLA-restricted cytomegalovirus (CMV)-specific T-cell responses after allogeneic bone marrow transplant: correlation with CMV disease and effect of ganciclovir prophylaxis.
Blood.
83
1994
1971
1979
7
Riddell
SR
Watanabe
KS
Goodrich
JM
Li
CR
Agha
ME
Greenberg
PD
Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones.
Science.
257
1992
238
241
8
Walter
EA
Greenberg
PD
Gilbert
MJ
et al
Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor.
N Engl J Med.
333
1995
1038
1044
9
Reusser
P
Cathomas
G
Attenhofer
R
Tamm
M
Thiel
G
Cytomegalovirus (CMV)-specific T cell immunity after renal transplantation mediates protection from CMV disease by limiting the systemic virus load.
J Infect Dis.
180
1999
247
253
10
Kuzushima
K
Hoshino
Y
Fujii
K
et al
Rapid determination of Epstein-Barr virus-specific CD8(+) T-cell frequencies by flow cytometry.
Blood.
94
1999
3094
3100
11
Kuzushima
K
Kimura
H
Hoshino
Y
et al
Longitudinal dynamics of Epstein-Barr virus-specific cytotoxic T lymphocytes during posttransplant lymphoproliferative disorder.
J Infect Dis.
182
2000
937
940
12
Hoshino
Y
Kimura
H
Kuzushima
K
et al
Early intervention in post-transplant lymphoproliferative disorders based on Epstein-Barr viral load.
Bone Marrow Transplant.
26
2000
199
201
13
Kern
F
Surel
IP
Brock
C
et al
T-cell epitope mapping by flow cytometry.
Nat Med.
4
1998
975
978
14
Kern
F
Khatamzas
E
Surel
I
et al
Distribution of human CMV-specific memory T cells among the CD8pos. subsets defined by CD57, CD27, and CD45 isoforms.
Eur J Immunol.
29
1999
2908
2915
15
Kern
F
Faulhaber
N
Khatamzas
E
et al
Measurement of anti-human cytomegalovirus T cell reactivity in transplant recipients and its potential clinical use: a mini-review.
Intervirology.
42
1999
322
324
16
Kern
F
Faulhaber
N
Frommel
C
et al
Analysis of CD8 T cell reactivity to cytomegalovirus using protein-spanning pools of overlapping pentadecapeptides.
Eur J Immunol.
30
2000
1676
1682
17
Singhal
S
Shaw
JC
Ainsworth
J
et al
Direct visualization and quantitation of cytomegalovirus-specific CD8+ cytotoxic T-lymphocytes in liver transplant patients.
Transplantation.
69
2000
2251
2259
18
Engstrand
M
Tournay
C
Peyrat
MA
et al
Characterization of CMVpp65-specific CD8+ T lymphocytes using MHC tetramers in kidney transplant patients and healthy participants.
Transplantation.
69
2000
2243
2250
19
Spiegel
HM
Ogg
GS
DeFalcon
E
et al
Human immunodeficiency virus type 1- and cytomegalovirus-specific cytotoxic T lymphocytes can persist at high frequency for prolonged periods in the absence of circulating peripheral CD4(+) T cells.
J Virol.
74
2000
1018
1022
20
Gillespie
GM
Wills
MR
Appay
V
et al
Functional heterogeneity and high frequencies of cytomegalovirus-specific CD8(+) T lymphocytes in healthy seropositive donors.
J Virol.
74
2000
8140
8150
21
Jin
X
Demoitie
MA
Donahoe
SM
et al
High frequency of cytomegalovirus-specific cytotoxic T-effector cells in HLA-A*0201-positive subjects during multiple viral coinfections.
J Infect Dis.
181
2000
165
175
22
Diamond
DJ
York
J
Sun
JY
Wright
CL
Forman
SJ
Development of a candidate HLA-A*0201 restricted peptide-based vaccine against human cytomegalovirus infection.
Blood.
90
1997
1751
1767
23
Yee
C
Savage
PA
Lee
PP
Davis
MM
Greenberg
PD
Isolation of high avidity melanoma-reactive CTL from heterogeneous populations using peptide-MHC tetramers.
J Immunol.
162
1999
2227
2234
24
Park
MH
Juji
T
Tokunaga
K
HLA ethnic study of Japanese and Koreans.
HLA 1991. Vol 1.
Tsuji
K
Aizawa
M
Sasazuki
T
1992
674
676
Oxford University Press
Oxford, United Kingdom
25
Imanishi
T
Akaza
T
Kimura
A
Tokunaga
K
Gojobori
T
Allele and haplotype frequencies for HLA and complement loci in various ethnic group.
HLA 1991. Vol 1.
Tsuji
K
Aizawa
M
Sasazuki
T
1992
1065
1220
Oxford University Press
Oxford, United Kingdom
26
Tokunaga
K
Ishikawa
Y
Ogawa
A
et al
Sequence-based association analysis of HLA class I and II alleles in Japanese supports conservation of common haplotypes.
Immunogenetics.
46
1997
199
205
27
Yang
D
Nakao
M
Shichijo
S
et al
Identification of a gene coding for a protein possessing shared tumor epitopes capable of inducing HLA-A24-restricted cytotoxic T lymphocytes in cancer patients.
Cancer Res.
59
1999
4056
4063
28
Chee
MS
Bankier
AT
Beck
S
et al
Analysis of the protein-coding content of the sequence of human cytomegalovirus strain AD169.
Curr Top Microbiol Immunol.
154
1990
125
169
29
Taylor R. BioInformatics and molecular analysis section (BIMAS) HLA peptide bonding predictions. Available athttp://bimas.dcrt.nih.gov/molbio/hla_bind/index.html. Accessed March 1, 2000.
30
Parker
KC
Bednarek
MA
Coligan
JE
Scheme for ranking potential HLA-A2 binding peptides based on independent binding of individual peptide side-chains.
J Immunol.
152
1994
163
175
31
Ikeda-Moore
Y
Tomiyama
H
Miwa
K
et al
Identification and characterization of multiple HLA-A24-restricted HIV-1 CTL epitopes: strong epitopes are derived from V regions of HIV-1.
J Immunol.
159
1997
6242
6252
32
Lee
SP
Tierney
RJ
Thomas
WA
Brooks
JM
Rickinson
AB
Conserved CTL epitopes within EBV latent membrane protein 2: a potential target for CTL-based tumor therapy.
J Immunol.
158
1997
3325
3334
33
Khanna
R
Burrows
SR
Nicholls
J
Poulsen
LM
Identification of cytotoxic T cell epitopes within Epstein-Barr virus (EBV) oncogene latent membrane protein 1 (LMP1): evidence for HLA A2 supertype-restricted immune recognition of EBV-infected cells by LMP1-specific cytotoxic T lymphocytes.
Eur J Immunol.
28
1998
451
458
34
Riddell
SR
Rabin
M
Geballe
AP
Britt
WJ
Greenberg
PD
Class I MHC-restricted cytotoxic T lymphocyte recognition of cells infected with human cytomegalovirus does not require endogenous viral gene expression.
J Immunol.
146
1991
2795
2804
35
McLaughlin-Taylor
E
Pande
H
Forman
SJ
et al
Identification of the major late human cytomegalovirus matrix protein pp65 as a target antigen for CD8+ virus-specific cytotoxic T lymphocytes.
J Med Virol.
43
1994
103
110
36
Riddell
SR
Greenberg
PD
The use of anti-CD3 and anti-CD28 monoclonal antibodies to clone and expand human antigen-specific T cells.
J Immunol Methods.
128
1990
189
201
37
Kondo
A
Sidney
J
Southwood
S
et al
Prominent roles of secondary anchor residues in peptide binding to HLA-A24 human class I molecules.
J Immunol.
155
1995
4307
4312
38
Kubo
RT
Sette
A
Grey
HM
et al
Definition of specific peptide motifs for four major HLA-A alleles.
J Immunol.
152
1994
3913
3924
39
Maier
R
Falk
K
Rotzschke
O
et al
Peptide motifs of HLA-A3, -A24, and -B7 molecules as determined by pool sequencing.
Immunogenetics.
40
1994
306
308
40
Gilbert
MJ
Riddell
SR
Li
CR
Greenberg
PD
Selective interference with class I major histocompatibility complex presentation of the major immediate-early protein following infection with human cytomegalovirus.
J Virol.
67
1993
3461
3469
41
Beersma
MF
Bijlmakers
MJ
Ploegh
HL
Human cytomegalovirus down-regulates HLA class I expression by reducing the stability of class I H chains.
J Immunol.
151
1993
4455
4464
42
Warren
AP
Ducroq
DH
Lehner
PJ
Borysiewicz
LK
Human cytomegalovirus-infected cells have unstable assembly of major histocompatibility complex class I complexes and are resistant to lysis by cytotoxic T lymphocytes.
J Virol.
68
1994
2822
2829
43
Spaete
RR
Gehrz
RC
Landini
MP
Human cytomegalovirus structural proteins.
J Gen Virol.
75
1994
3287
3308
44
Gilbert
MJ
Riddell
SR
Plachter
B
Greenberg
PD
Cytomegalovirus selectively blocks antigen processing and presentation of its immediate-early gene product.
Nature.
383
1996
720
702
45
Jin
X
Roberts
CG
Nixon
DF
et al
Identification of subdominant cytotoxic T lymphocyte epitopes encoded by autologous HIV type 1 sequences, using dendritic cell stimulation and computer-driven algorithm.
AIDS Res Hum Retroviruses.
16
2000
67
76
46
Borysiewicz
LK
Hickling
JK
Graham
S
et al
Human cytomegalovirus-specific cytotoxic T cells: relative frequency of stage-specific CTL recognizing the 72-kD immediate early protein and glycoprotein B expressed by recombinant vaccinia viruses.
J Exp Med.
168
1988
919
931
47
Wills
MR
Carmichael
AJ
Mynard
K
et al
The human cytotoxic T-lymphocyte (CTL) response to cytomegalovirus is dominated by structural protein pp65: frequency, specificity, and T-cell receptor usage of pp65-specific CTL.
J Virol.
70
1996
7569
7579
48
Weekes
MP
Wills
MR
Mynard
K
Carmichael
AJ
Sissons
JG
The memory cytotoxic T-lymphocyte (CTL) response to human cytomegalovirus infection contains individual peptide-specific CTL clones that have undergone extensive expansion in vivo.
J Virol.
73
1999
2099
2108
49
Kuzushima
K
Sun
R
van Bleek
GM
Vegh
Z
Nathenson
SG
The role of self peptides in the allogeneic cross-reactivity of CTLs.
J Immunol.
155
1995
594
601
50
Solache
A
Morgan
CL
Dodi
AI
et al
Identification of three HLA-A*0201-restricted cytotoxic T cell epitopes in the cytomegalovirus protein pp65 that are conserved between eight strains of the virus.
J Immunol.
163
1999
5512
5518
51
Gavin
MA
Gilbert
MJ
Riddell
SR
Greenberg
PD
Bevan
MJ
Alkali hydrolysis of recombinant proteins allows for the rapid identification of class I MHC-restricted CTL epitopes.
J Immunol.
151
1993
3971
3980

Author notes

Kiyotaka Kuzushima, Division of Virology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chigusa-ku, Nagoya 464-8681, Japan; e-mail:kkuzushi@aichi-cc.pref.aichi.jp.

Sign in via your Institution