The molecular characterization of the CD4+ T-cell epitope repertoire on human tumor antigens would contribute to both clinical investigation and cancer immunotherapy. In particular, the identification of promiscuous epitopes would be beneficial to a large number of patients with neoplastic diseases regardless of their HLA-DR type. MAGE-3 is a tumor-specific antigen widely expressed in solid and hematologic malignancies; therefore, is an excellent candidate antigen. We used a major histocompatability complex (MHC) class II epitope prediction algorithm, the TEPITOPE software, to predict 11 sequence segments of MAGE-3 that could form promiscuous CD4+ T-cell epitopes. In binding assays, the synthetic peptides corresponding to the 11 predicted sequences bound at least 3 different HLA-DR alleles. Nine of the 11 peptides induced proliferation of CD4+ T cells from both healthy subjects and melanoma patients. Four immunodominant regions (residues 111-125, 146-160, 191-205, and 281-295), containing naturally processed epitopes, were recognized by most of the donors, in association with 3 to 4 different HLA-DR alleles, thus covering up to 94% of the alleles expressed in whites. On the contrary, the other promiscuous regions (residues 161-175 and 171-185) contained epitopes not naturally processed in vitro. The immunodominant epitopes identified will be useful in the design of peptide-based cancer vaccines and in the study of the functional state of tumor-specific CD4+ T cells in patients bearing tumors expressing MAGE-3.

Evidence for a critical role of CD4+ T cells in orchestrating the antitumor response is increasing (for reviews, see Pardoll and Topalian1 and Toes et al2). CD4+ T cells provide help for induction and maintenance of antitumor CD8+ T cells and may exert effector functions, both indirectly via macrophage and eosinophil activation, and directly when inflammatory responses produce cytokines such as interferon γ (IFN-γ) that induce expression of major histocompatability complex (MHC) class II molecules on tumor cells. In addition, IFN-γ produced by antitumor CD4+ T cells may function as an antiangiogenic factor.3,4 CD4+ T cells specific for tumor antigens have been described in humans (for reviews, see Pardoll and Topalian,1 Pawelec et al,5 and Wang6).

MAGE-3 is a tumor-specific antigen widely expressed in solid tumors such as melanoma, lung carcinoma, head and neck carcinoma, and hematologic malignancies, including T-cell leukemias and myeloma but not in normal tissues (with the exception of testis).7 The MAGE gene family comprises several related genes divided into 3 clusters, named MAGE-A, -B, and -C.7 The gene previously known as MAGE-3 belongs to the MAGE-A cluster and it has received the official name MAGE-A3. We will refer to it from now on as MAGE-3, in accordance with our previous report.8 Clinical trials in patients with metastatic neoplastic disease using the HLA-A1–restricted MAGE-3 epitope yielded some clinical results.9-13 However, the immune responses were too weak and transient to eradicate tumor cells in the majority of immunized patients. The additional use of CD4 epitopes might help the induction of a memory antitumor response and improve the clinical efficacy of vaccines.

Several allele-specific CD8 and CD4 epitopes of MAGE-3 have been identified.14 Promiscuous epitopes, that is, those able to bind to different HLA-DR alleles, are crucial to increase the number of patients eligible for peptide-based cancer vaccination. Up to now, only MAGE-3146-160 has been described by us8 and others15 as a promiscuous epitope presented in association with HLA-DR11, HLA-DR4, and HLA-DR7.

In this paper, we report the results of the analysis of the MAGE-3 tumor antigen for promiscuous CD4 epitopes by a combined approach of computational identification of candidate T-cell epitopes, followed by in vitro biologic validation analysis of the predicted sequences as forming natural epitopes recognized by CD4+ T cells.

We identified 4 immunodominant regions (residues 111-125, 146-160, 191-205, and 281-295), containing naturally processed epitopes, that were recognized by most donors in association with 3 to 4 different HLA-DR alleles. These epitopes, potentially recognized by more than 90% of whites, will be useful for both clinical investigation and cancer immunotherapy.

T-cell epitope prediction and peptide synthesis

We used the TEPITOPE algorithm16 that allows the identification of HLA class II ligands binding in a promiscuous or allele-specific mode and the effects of polymorphic residues on the specificity of HLA class II ligands to select 11 sequences of the MAGE-3 protein for peptide synthesis. The program permits the prediction and the parallel display of ligands for each of the 25 HLA-DR alleles incorporated in the software (examples are reported in Sturniolo et al16). We set the prediction threshold (ie, the percentage of best scoring natural peptides) at 5%, and we selected the sequences predicted to bind at least 40% of the 25 HLA-DR alleles, plus an additional sequence (residues 21-35) predicted to bind 16%. The MAGE-3 sequences comprising residues 141-155, 146-160, 156-170, 171-185, and 281-295, forming pool I, were already described.8 The MAGE-3 sequences 21-35, 111-125, 161-175, 191-205, 251-265, and 286-300, forming pool II, were synthesized by manual parallel synthesis as described.17 The peptide purity was verified by reverse-phase high-performance liquid chromatography and electron spray mass spectrometry. The synthetic peptides were lyophilized, reconstituted in dimethyl sulfoxide (DMSO) at 10 mg/mL, and diluted in RPMI 1640 (Gibco, Grand Island, NY) as needed.

Peptide-binding assays

Peptide interactions with detergent-solubilized HLA-DR molecules were measured using an enzyme-linked immunosorbent assay (ELISA)–based high-flux competition assay.18 HLA-DR molecules were affinity purified from human Epstein-Barr virus–transformed lymphoblastoid cell lines (LCLs) as described.19,20 The assays measured the ability of unlabeled peptides to compete with a biotinylated indicator peptide for binding to purified HLA-DR molecules.21 To determine relative binding affinity, the promiscuous HA307-319 peptide from influenza hemagglutinin21 was included in each competition assay.

Subjects and cells

Peripheral blood mononuclear cells (PBMCs) were obtained from 4 healthy subjects (donors 1, 2, 3, and 4) and 2 patients with melanoma (donors 5 and 6). Melanoma cell lines MD TC, OI TC, and GF TC were established from 2 cutaneous metastases and one lymph node metastasis, respectively, from 3 patients with melanoma, and line SK-Mel-24 was purchased from the American Type Culture Collection (Rockville, MD). MAGE-3 expression by patients' biopsies and melanoma cell lines was verified by reverse transcriptase–polymerase chain reaction (RT-PCR) and in melanoma cells also by intracytoplasmic staining (data not shown). The HLA-DR types of donors and melanoma cell lines were identified by molecular or serologic typing and are reported in the figures. Homozygous LCLs used were Bor (DR11) and Mun (DR3), established in our laboratory; Leis (DR4), kindly provided by F. Marincola (National Institutes of Health, Bethesda, MD); BM21 (DR11) purchased from ATCC; and Pitout (DR7), purchased from the European Collection of Cell Culture (ECCC; Salisbury, United Kingdom).

Propagation of CD4+ T cells

Synthetic peptides corresponding to pool I and pool II (Table1) were used to stimulate the PBMCs from the different donors. Briefly, 20 × 106 PBMCs were cultured for 7 days in RPMI 1640 (Gibco), supplemented with heat-inactivated human serum (10%), l-glutamine (2 mM), penicillin (100 U/mL), and streptomycin (50 μg/mL; Biowhittaker, Walkersville, MD), containing pool I or pool II (1 μg/mL of each peptide). The reactive blasts were isolated on a Percoll gradient,22 expanded in interleukin 2 (IL-2)–containing medium (10 U/mL; Lymphocult, Biotest Diagnostic, Dreieich, Germany), and restimulated at weekly intervals with the same amount of peptides plus irradiated (4000 rad) autologous PBMCs as antigen-presenting cells (APCs).

Table 1.

Determination of HLA-DR binding of MAGE-A3 synthetic peptides corresponding to the sequences predicted by TEPITOPE to form promiscuous epitopes

SequencesPredicted binding alleles*HLA-DRβ1 alleles
*0101*1501*0301*0401*1101*0701*0801
Pool I residues          
 141-155 GNWQYFFPVIFSKAS 68 25 >100 0.6 0.1 3.2 
 146-160 FFPVIFSKASSSLQL 66 10 0.24 1.8 0.01 1.5 
 156-170 SSLQLVFGIELMEVD 68 0.18 90 45 28 0.03 
 171-185 PIGHLYIFATCLGLS 96 0.3 0.028 2.8 0.9 0.9 0.01 1.5 
 281-295 TSYVKVLHHMVKISG 80 15 0.48 26 70 0.035 0.2 0.01 
Pool II residues          
 21-35 EALGLVGAQAPATEE 16 14 22 >100 >100 >100 25 >100 
 111-125 RKVAELVHFLLLKYR 76 >100 0.055 >100 >100 0.7 55 
 161-175 VFGIELMEVDPIGHL 76 >100 100 0.6 28 >100 10 100 
 191-205 GDNQIMPKAGLLIIV 40 >100 0.07 >100 >100 
 251-265 VQENYLEYRQVPGSD 48 >100 >100 >100 60 26 10 
 286-300 VLHHMVKISGGPHIS 88 15 0.48 >100 >100 0.2 0.01 14 
HA (307-319) PKYVKQNTLKLAT 84 0.18 2.4 0.9 0.7 0.18 3.2 
SequencesPredicted binding alleles*HLA-DRβ1 alleles
*0101*1501*0301*0401*1101*0701*0801
Pool I residues          
 141-155 GNWQYFFPVIFSKAS 68 25 >100 0.6 0.1 3.2 
 146-160 FFPVIFSKASSSLQL 66 10 0.24 1.8 0.01 1.5 
 156-170 SSLQLVFGIELMEVD 68 0.18 90 45 28 0.03 
 171-185 PIGHLYIFATCLGLS 96 0.3 0.028 2.8 0.9 0.9 0.01 1.5 
 281-295 TSYVKVLHHMVKISG 80 15 0.48 26 70 0.035 0.2 0.01 
Pool II residues          
 21-35 EALGLVGAQAPATEE 16 14 22 >100 >100 >100 25 >100 
 111-125 RKVAELVHFLLLKYR 76 >100 0.055 >100 >100 0.7 55 
 161-175 VFGIELMEVDPIGHL 76 >100 100 0.6 28 >100 10 100 
 191-205 GDNQIMPKAGLLIIV 40 >100 0.07 >100 >100 
 251-265 VQENYLEYRQVPGSD 48 >100 >100 >100 60 26 10 
 286-300 VLHHMVKISGGPHIS 88 15 0.48 >100 >100 0.2 0.01 14 
HA (307-319) PKYVKQNTLKLAT 84 0.18 2.4 0.9 0.7 0.18 3.2 

The binding data are expressed in terms of relative binding capacity (IC50 μM), calculated as concentration of competitor peptide required to inhibit 50% of the binding of an allele specific biotinylated peptide (indicator peptide).

*

Percent of alleles predicted to bind the indicated sequences, with a threshold set at 5% (100% is represented by the 25 alleles incorporated in the software). The threshold is defined as the percentage of best scoring natural peptides and comprises a range between 1% to 10%; see “Materials and methods” for details.

HA307-319 is promiscuous sequence from influenza hemagglutinin.

IC50 values higher than 100 μM are outside the sensitivity limits of the binding assay.

Flow cytometry

Cytofluorometric analyses were performed on a FACStarPlus (Becton Dickinson, Sunnyvale, CA). We used the following monoclonal antibodies (mAbs): anti-CD4–phycoerythrin (PE) and anti-CD8–fluorescein isothiocyanate (FITC; Becton Dickinson); L243 (anti–MHC class II; ATCC); 57B (originally described as an anti–MAGE-3 mAb,23 and later identified as an anti–MAGE-A4 mAb that cross-reacts with several MAGE-A proteins24) kindly provided by G. Spagnoli (Basel, Switzerland); and 20.4 (mAb that recognizes the truncated form of the human low-affinity nerve growth factor receptor, ΔNGFr; ATCC). FITC-rabbit antimouse immunoglobulin antibody (Dako, Glostrup, Denmark) was used as second-step reagent in indirect immunofluorescence stainings. For intracytoplasmic staining, melanoma cells were fixed in 3.7% paraformaldehyde, permeabilized in 0.05% NP40, and then stained as described.25 

RT-PCR analysis

Total RNA was extracted by the use of RNAzolTMB (Biotech, Houston, TX), according to the manufacturer's instructions. Single-stranded cDNA was synthesized from 2 μg total RNA, by Moloney murine leukemia virus–derived reverse transcriptase (Life Technologies, Milan, Italy), in the presence of 20 U RNasin (Promega, Madison, WI). cDNA coding for MAGE-3 was detected by PCR amplification as described.26 Samples scored positive when a band of the appropriate size was visible on an agarose gel in the presence of ethidium bromide.

Proliferation assay

CD4+ T cells and autologous irradiated PBMCs or HLA-DR–matched homozygous LCLs as APCs were diluted at a 1:10 or 1:5 ratio, respectively, and used as we described previously.8Stimulants are indicated in the figure legends, where appropriate. In inhibition experiments, mAb L243 or an isotype-matched irrelevant mAb was added at the concentrations indicated in the figure legends. After 48 hours, the cultures were pulsed for 16 hours with [3H]-thymidine (TdR; 1 μCi [37 Bq], well, Amersham, Milan, Italy). The cells were collected with a Skatron Titertek multiple harvester (Skatron, Sterling, VA) and the thymidine incorporated was measured in a liquid scintillation counter.

Selection of peptide-specific CD4+ T cells by magnetic separation

To select MAGE-3161-175–specific CD4+ T cells from donor 5 from the polyclonal cell line, we first incubated the cells with the peptide in the presence of homozygous DR4-PBMCs and then we used the magnetic activating cell sorting (MACS) cytokine secretion assay-cell enrichment and detection kit (Miltenyi Biotec, Bergisch Gladback, Germany), following the manufacturer's instructions. MAGE-3161-175–specific CD4+ T cells were then restimulated weekly in the presence of the peptide (1-5 μg/mL) and DR4-PBMCs as APCs.

Recombinant viruses and infection of LCLs

The cDNA encoding Ii.MAGE-3,27 a fusion protein between the human invariant chain (Ii) and MAGE-3, was kindly provided by P. van der Bruggen (Brussels, Belgium). The retroviral vectors M3-CSM and IiM3-CSM, which encode the ΔLNGFr, and either the full-length MAGE-3 protein or the fusion protein Ii.MAGE-3, respectively, were produced as reported previously.28 LCLs were transduced by coculture with irradiated packaging cells producing the M3-CSM or the IiM3-CSM vectors, produced by Phoenix cells (GP Nolan, Stanford, CA). The percentage of infected cells was evaluated 48 hours after transduction by flow cytometry for ΔLNGFr expression with the mAb 20.4.28 The ΔLNGFr+ cells were purified by magnetic cell sorting, using rat antimouse IgG1-coated beads (Dynabeads M-450, Dynal, Oslo, Norway).

Cytotoxicity assay

CD4+ T cells were tested for specific lytic activity in a standard 4-hour 51Cr release assay as described previously.29 The following targets were used: melanoma cells MD TC, OI TC, GF TC, and SK-Mel-24, LCL and LCL engineered to express MAGE-3 (LCL-M3). In cold-target competition assays, unlabeled target cells (cold targets) were added to CD4+ T cells (effectors) and 51Cr-labeled target cells (hot targets) at serial ratios of cold-to-hot target cells. The percentage of inhibition was calculated as follows: [(% specific lysis without cold target−% specific lysis with cold target)/(% specific lysis without cold target)]× 100.

CD4+ T-cell stimulation assay

LCL-M3, LCL-IiM3 (LCLs expressing Ii.MAGE-3), or melanoma cells were tested for their ability to induce the production of IFN-γ by peptide-specific CD4+ T cells, after 24 to 48 hours of incubation, using a standard ELISA (Biosource Europe, Nivelles, Belgium), following the manufacturer's instructions.

We selected 11 MAGE-3 sequences, based on prediction by TEPITOPE, and used synthetic peptides corresponding to the selected sequences to confirm their binding capability in competition assays, using 7 common HLA-DRB1 alleles (Table 1).

We grouped the 11 peptides into pool I and pool II, based on the results of the binding assays. Peptides forming pool I had the highest promiscuity, and peptides forming pool II bound well to at least 3 different HLA-DR alleles. We used pool I and pool II to propagate polyclonal T-cell lines from 6 donors. Total PBMCs were stimulated with pool I or pool II or both in independent cultures for 7 days, activated cells were enriched by a density gradient, expanded in the presence of IL-2, and restimulated weekly with irradiated peptide-pulsed autologous PBMCs. For 5 donors, after 2 cycles of stimulation, we obtained lines that comprised only CD4+ T cells. In the case of donor 1, a CD8+ T-cell depletion step was necessary to obtain more than 90% CD4+ T cells after 4 cycles of stimulation (data not shown). CD4+ T-cell lines from healthy donors 1, 2, and 4 and from donor/patient 5 could be propagated for several months. The lines from healthy subject 3 and donor/patient 6 could be propagated only for a few weeks and did not expand well in culture.

Repertoire of HLA-DR–restricted epitopes recognized by polyclonal CD4+ T cells propagated with pool I and pool II

We determined the epitope repertoire of the CD4+ lines from all donors by testing, after a few cycles of stimulation, their proliferative response to each single peptide forming the pools. Figure1 summarizes the results obtained. Nine of 11 peptides tested elicited proliferation of CD4+ T cells in at least 2 donors. MAGE-3111-125, MAGE-3141-155, and the overlapping MAGE-3146-160, MAGE- 3191-205, MAGE-3281-295, and the overlapping MAGE-3286-300, were strongly recognized by most of the donors, consistent with a high degree of promiscuity. MAGE-3156-170 was weakly but significantly recognized by donor 1 and donor/patient 5, and the overlapping peptide MAGE-3161-175 was significantly and strongly recognized by donor 4 and donor/patient 5. MAGE-3171-185 was strongly recognized by donor 1 and donor/patient 5. Recognition of the MAGE-3 sequences was HLA-DR restricted as demonstrated in vitro by inhibition of the proliferation of CD4+ T cells to the peptides in the presence of an anti-DR antibody in the cultures (data not shown). MAGE-321-35 and MAGE-3251-265 were never recognized.

Fig. 1.

MAGE-3 sequence segments recognized by CD4+T cells from 6 donors and their HLA-DR type.

Polyclonal CD4+ T-cell lines propagated in culture with pool I and pool II were tested with each single peptides (10 μg/mL) forming the pools in 2-day microproliferation assays. The results are representative of several assays in the case of donors 1, 2, 4, and donor/patient 5 and, at least 2 experiments, in the case of donor 3 and donor/patient 6. Peptides that elicited a strong response are indicated in black (P < .001), peptides that elicited an intermediate response are indicated in hatched segments (.001 < P < .05), and peptides that elicited a low but still significant response are indicated in white (P < .05). Responses significantly higher than the blanks (ie, the basal level of proliferation of CD4+ T cells in the presence of autologous PBMCs as APCs) were determined by unpaired, one-tailed Student t test. NT indicates not tested.

Fig. 1.

MAGE-3 sequence segments recognized by CD4+T cells from 6 donors and their HLA-DR type.

Polyclonal CD4+ T-cell lines propagated in culture with pool I and pool II were tested with each single peptides (10 μg/mL) forming the pools in 2-day microproliferation assays. The results are representative of several assays in the case of donors 1, 2, 4, and donor/patient 5 and, at least 2 experiments, in the case of donor 3 and donor/patient 6. Peptides that elicited a strong response are indicated in black (P < .001), peptides that elicited an intermediate response are indicated in hatched segments (.001 < P < .05), and peptides that elicited a low but still significant response are indicated in white (P < .05). Responses significantly higher than the blanks (ie, the basal level of proliferation of CD4+ T cells in the presence of autologous PBMCs as APCs) were determined by unpaired, one-tailed Student t test. NT indicates not tested.

Close modal

HLA-DR restriction molecules for the immunodominant sequences

To identify the HLA-DR–restricting allele for the immunodominant sequences, CD4+ T cells from donors 1 and 2 and donor/patient 5 were challenged in microproliferation assays with APCs, homozygous for each of the 2 HLA-DRB1 alleles expressed by the donor, pulsed with individual peptides (Figure2).

Fig. 2.

HLA-DR restriction of CD4+ T cells recognizing the immunodominant epitopes.

CD4+ T cells were propagated with pool II (A,C) or pool I (B,D). The donors and their HLA-DR types are indicated. The blank (ie, the basal level of proliferation of CD4+ T cells in the presence of homozygous PBMCs or LCLs as APCs) is expressed as B + PBMC or B + LCL. Alignment of overlapping sequences 111-125 and 114-127 (presented by HLA-DR1327) is reported in the box. Responses significantly higher than the blanks are indicated: **.001 < P < .05, ***P < .001 (determined by unpaired, one-tailed Student ttest).

Fig. 2.

HLA-DR restriction of CD4+ T cells recognizing the immunodominant epitopes.

CD4+ T cells were propagated with pool II (A,C) or pool I (B,D). The donors and their HLA-DR types are indicated. The blank (ie, the basal level of proliferation of CD4+ T cells in the presence of homozygous PBMCs or LCLs as APCs) is expressed as B + PBMC or B + LCL. Alignment of overlapping sequences 111-125 and 114-127 (presented by HLA-DR1327) is reported in the box. Responses significantly higher than the blanks are indicated: **.001 < P < .05, ***P < .001 (determined by unpaired, one-tailed Student ttest).

Close modal

We did not perform HLA-DR restriction on donor 4 because donor 4 is HLA-DR11 homozygous, nor on donor 3 and donor/patient 6, because CD4+ T cells did not expand in culture enough to allow further characterization. MAGE-3111-125 was recognized in association with HLA-DR1, HLA-DR4, and HLA-DR11 (Figure2A). Moreover, MAGE-3114-127, which largely overlaps MAGE-3111-125, was shown by van der Bruggen and colleagues27 to be recognized in association with HLA-DR13. MAGE-3146-160 was recognized in association with HLA-DR1, HLA-DR4, HLA-DR11, and HLA-DR7 (Figure 2B). MAGE-3191-205 was recognized in association with HLA-DR1, HLA-DR4, and HLA-DR11 alleles (Figure 2C). MAGE-3281-295was recognized in association with HLA-DR1, HLA-DR4, and HLA-DR11 (Figure 2D). The same immunodominant sequence could be recognized in different donors with either one or both of the HLA-DRB1 alleles expressed. Moreover, different donors sharing the same alleles might preferentially recognize the same epitope with different alleles, probably due to a difference in precursors' frequency.

Immunodominant sequences contain naturally processed epitopes

MAGE-3114-127, largely overlapping MAGE-3111-125, MAGE- 3146-160, and MAGE-3281-295 were already described to contain naturally processed epitopes recognized in association with HLA-DR13,27 HLA-DR4 and HLA-DR7,15 and HLA-DR11,8 respectively. MAGE-3114-127contains a natural epitope that is formed after processing through the exogenous pathway28; we could not test the natural processing pathways for MAGE-3111-125, which most likely contain the same natural epitope, because we failed to obtain CD4+ T cells recognizing exclusively that sequence. MAGE-3146-160 was formed after both endogenous and exogenous pathways.15 In the present work, we found that MAGE-3146-160 is recognized by CD4+ cytotoxic T cells (CTLs) from donor 2 (HLA-DR3, HLA-DR7) when expressed on HLA-DR7–matched melanoma cells (GF TC) and DR7-LCL engineered to express MAGE-3 (data not shown). MAGE-3281-295 was shown by us8 to contain an epitope formed after both endogenous and exogenous pathways and recognized by CD4+ CTLs.

To verify whether MAGE-3191-205 contains a natural epitope, we used polyclonal CD4+ T cells from donor 4 after 9 weeks of propagation with pool II, when the cells strongly and exclusively recognized MAGE-3191-205 (Figure3A lower panel). Recognition was HLA-DR restricted as shown in Figure 3B. DR11-LCLs were engineered to express MAGE-3 (DR11-LCL-M3) and used in cytotoxicity assays. As shown in Figure 3C, MAGE-3191-205–specific CD4+ T cells killed DR11-LCL-M3, but not wild-type DR11-LCLs. Moreover, CD4+ CTLs killed the MD TC and OI TC melanoma cells, which express MAGE-3 and HLA-DR11, but not SK-Mel 24, expressing unrelated HLA-DR (Figure 3D). In addition, we performed cold-target inhibition experiments, in which cold targets (DR11-LCLs) preincubated with peptide MAGE-3191-205 were added to the hot tumor targets. Killing of MD TC and OI TC was inhibited by addition of peptide-pulsed DR11-LCLs (up to 75% and 98%, respectively), suggesting that this peptide contains an epitope that is presented by the melanoma cells (Figure 3E-F). The percentage of inhibition of DR11-LCL (Figure 3E) or DR11-LCLs pulsed with an irrelevant peptide (Figure 3F) was comparable in the 2 cases, and up to 25% to 30%. The high cold/hot ratio (50%) required to observe a specific inhibition of MD TC killing, compared to OI TC, might be related to different binding affinity of the peptide for the 2 subtypes of DR11-LCLs (DR*1104 and DR*1101) used. In the case of MD TC, competition for MHC binding to the cold or hot targets of free peptide released from the LCLs in culture might reduce the ability of these LCLs to inhibit the killing.

Fig. 3.

Characterization of the CD4+ T-cell response to MAGE-3191-205.

CD4+ T cells were propagated from donor 4 (HLA-DR11) by weekly restimulation with pool II. (A) Proliferative responses, measured in 2-day microproliferation assays, to each single peptide forming pool II (10 μg/mL) after 4 and 9 weeks of propagation of the line. The data shown are means of triplicate determination ± SDs. The blank (ie, the basal level of proliferation of CD4+ T cells in the presence of autologous PBMCs as APCs) is expressed as B + DR11-PBMC. Responses significantly higher than the blanks are indicated: **.001 < P < .05, ***P < .001 (determined by unpaired, one-tailed Student t test). (B) Responses to pool II (5 μg/mL), in the absence or in the presence of L243 mAb or an irrelevant control mAb (0.5 μg/mL). The blank (ie, the basal level of proliferation of CD4+ T cells in the presence of autologous PBMCs as APCs) is expressed as B + APC. (C-F) Recognition of naturally processed MAGE-3 sequences by CD4+ T cells at 9 or more weeks of propagation. (C) Cytolytic activity, measured in a51Cr release assay, against wild-type DR*1104-LCL and DR*1104-LCL engineered to express MAGE-3 (DR*1104-LCL-M3). (D) Cytolytic activity against HLA-DR–matched (MD TC and OI TC) and HLA-DR–unrelated (SK-Mel 24) melanoma cells. (E-F) Cold target inhibition experiments. (E) Cold targets (MD TC, DR*1104-LCL, and DR*1104-LCL pulsed with MAGE-3191-205) were used to inhibit the lytic activity of CD4+ cytotoxic T cells against hot MD TC (effector-target [E/T] ratio 40:1). The percentage of specific lysis against MD TC in the absence of cold target was 22 ± 1.3. (F) Cold targets (DR*1101-LCL and DR*1101-LCL pulsed with MAGE-3191-205 or MAGE-321-35) were used to inhibit the lytic activity of CD4+ cytotoxic T cells against hot OI TC (E/T ratio 40:1). The percents lytic activity against OI TC and DR*1101-LCL, as negative control, are shown in black symbols. The data presented in panels C through F are representative of at least 3 experiments and are means of triplicate determinations ± SDs.

Fig. 3.

Characterization of the CD4+ T-cell response to MAGE-3191-205.

CD4+ T cells were propagated from donor 4 (HLA-DR11) by weekly restimulation with pool II. (A) Proliferative responses, measured in 2-day microproliferation assays, to each single peptide forming pool II (10 μg/mL) after 4 and 9 weeks of propagation of the line. The data shown are means of triplicate determination ± SDs. The blank (ie, the basal level of proliferation of CD4+ T cells in the presence of autologous PBMCs as APCs) is expressed as B + DR11-PBMC. Responses significantly higher than the blanks are indicated: **.001 < P < .05, ***P < .001 (determined by unpaired, one-tailed Student t test). (B) Responses to pool II (5 μg/mL), in the absence or in the presence of L243 mAb or an irrelevant control mAb (0.5 μg/mL). The blank (ie, the basal level of proliferation of CD4+ T cells in the presence of autologous PBMCs as APCs) is expressed as B + APC. (C-F) Recognition of naturally processed MAGE-3 sequences by CD4+ T cells at 9 or more weeks of propagation. (C) Cytolytic activity, measured in a51Cr release assay, against wild-type DR*1104-LCL and DR*1104-LCL engineered to express MAGE-3 (DR*1104-LCL-M3). (D) Cytolytic activity against HLA-DR–matched (MD TC and OI TC) and HLA-DR–unrelated (SK-Mel 24) melanoma cells. (E-F) Cold target inhibition experiments. (E) Cold targets (MD TC, DR*1104-LCL, and DR*1104-LCL pulsed with MAGE-3191-205) were used to inhibit the lytic activity of CD4+ cytotoxic T cells against hot MD TC (effector-target [E/T] ratio 40:1). The percentage of specific lysis against MD TC in the absence of cold target was 22 ± 1.3. (F) Cold targets (DR*1101-LCL and DR*1101-LCL pulsed with MAGE-3191-205 or MAGE-321-35) were used to inhibit the lytic activity of CD4+ cytotoxic T cells against hot OI TC (E/T ratio 40:1). The percents lytic activity against OI TC and DR*1101-LCL, as negative control, are shown in black symbols. The data presented in panels C through F are representative of at least 3 experiments and are means of triplicate determinations ± SDs.

Close modal

MAGE-3161-175 and MAGE-3171-185 contain epitopes not naturally processed in vitro

To verify whether MAGE-3161-175 contains a natural epitope, we used polyclonal CD4+ T cells from donor/patient 5, after selection of peptide MAGE-3161-175–specific CD4+ T cells by magnetic sorting (Figure 4B). CD4+ T cells did not secrete IFN-γ after challenge with melanoma cells or DR4-LCLs engineered to express MAGE-3 (DR4-LCL-M3), nor DR4-LCLs engineered to express Ii.MAGE-3 (DR4-LCL-IiM3), suggesting that MAGE-3161-175 does not contain a natural epitope produced after processing through both the endogenous and the exogenous pathways (Figure 4C).

Fig. 4.

Characterization of the CD4+ T-cell response to MAGE-3161-175.

CD4+ T cells from donor/patient 5 (HLA-DR4, HLA-DR11) were propagated by weekly restimulation with pool II. (A) Proliferative responses, measured in 2-day microproliferation assays, to each single peptide forming pool II (10 μg/mL) after 5 weeks of propagation of the line. The blank (ie, the basal level of proliferation of CD4+ T cells in the presence of homozygous PBMCs or LCLs as APCs) is expressed as B + DR11-PBMC or B + DR4-LCL. (B) Proliferative responses to each single peptide forming pool II (10 μg/mL) after magnetic selection of MAGE-3161-175–specific CD4+ T cells. (C) IFN-γ release of CD4+ T cells in the presence of melanoma cells, DR4-LCL, MAGE-3161-175–pulsed DR4-LCL, DR4-LCL-M3, and DR4-LCL-IiM3. The data shown in panels A to C are means of triplicate determinations ± SDs. Responses significantly higher than the blanks are indicated: **.001 < P < .05, ***P < .001 (determined by unpaired, one-tailed Student ttest).

Fig. 4.

Characterization of the CD4+ T-cell response to MAGE-3161-175.

CD4+ T cells from donor/patient 5 (HLA-DR4, HLA-DR11) were propagated by weekly restimulation with pool II. (A) Proliferative responses, measured in 2-day microproliferation assays, to each single peptide forming pool II (10 μg/mL) after 5 weeks of propagation of the line. The blank (ie, the basal level of proliferation of CD4+ T cells in the presence of homozygous PBMCs or LCLs as APCs) is expressed as B + DR11-PBMC or B + DR4-LCL. (B) Proliferative responses to each single peptide forming pool II (10 μg/mL) after magnetic selection of MAGE-3161-175–specific CD4+ T cells. (C) IFN-γ release of CD4+ T cells in the presence of melanoma cells, DR4-LCL, MAGE-3161-175–pulsed DR4-LCL, DR4-LCL-M3, and DR4-LCL-IiM3. The data shown in panels A to C are means of triplicate determinations ± SDs. Responses significantly higher than the blanks are indicated: **.001 < P < .05, ***P < .001 (determined by unpaired, one-tailed Student ttest).

Close modal

To characterize the recognition of peptide MAGE-3171-185, we used polyclonal CD4+ T cells from donor/patient 5, after 12 weeks of propagation in culture, when they strongly and exclusively recognized MAGE-3171-185 (Figure5B). CD4+ T cells did not recognize melanoma cells or DR4-LCL-M3, nor DR4-LCL-IiM3, suggesting that this sequence does not contain a natural epitope as well (Figure5C). To verify that DR4-LCL-M3 and DR4-LCL-IiM3 express a sufficient amount of the MAGE-3 protein to be recognized by CD4+ T cells, we tested its expression by flow cytometry. MAGE-3 intracytoplasmic staining in transduced DR4-LCLs and DR11-LCLs recognized by CD4+ T cells specific for MAGE-3191-205 (Figure 3D) showed that they contain a comparable amount of MAGE-3 (data not shown).

Fig. 5.

Characterization of the CD4+ T-cell response to MAGE-3171-185.

CD4+ T cells from donor/patient 5 (HLA-DR4, HLA-DR11) were propagated by weekly restimulation with pool I. (A-B) Proliferative responses, measured in 2-day microproliferation assays, to each single peptide forming pool I (10 μg/mL) after 3 weeks (A) and after 12 weeks (B) of propagation of the line. The blank (ie, the basal level of proliferation of CD4+ T cells in the presence of homozygous PBMCs as APCs) is expressed as B + DR11-PBMC or B + DR4-PBMC or B + DR4-LCL. (C) IFN-γ release of CD4+ T cells in the presence of melanoma cells, DR4-LCL, MAGE-3171-185–pulsed DR4-LCL, DR4-LCL-M3, and DR4-LCL-IiM3. The data shown in panels A to C are means of triplicate determinations ± SDs. Responses significantly higher than the blanks are indicated: *P < .05, **.001 < P < .05, ***P < .001 (determined by unpaired, one-tailed Student t test).

Fig. 5.

Characterization of the CD4+ T-cell response to MAGE-3171-185.

CD4+ T cells from donor/patient 5 (HLA-DR4, HLA-DR11) were propagated by weekly restimulation with pool I. (A-B) Proliferative responses, measured in 2-day microproliferation assays, to each single peptide forming pool I (10 μg/mL) after 3 weeks (A) and after 12 weeks (B) of propagation of the line. The blank (ie, the basal level of proliferation of CD4+ T cells in the presence of homozygous PBMCs as APCs) is expressed as B + DR11-PBMC or B + DR4-PBMC or B + DR4-LCL. (C) IFN-γ release of CD4+ T cells in the presence of melanoma cells, DR4-LCL, MAGE-3171-185–pulsed DR4-LCL, DR4-LCL-M3, and DR4-LCL-IiM3. The data shown in panels A to C are means of triplicate determinations ± SDs. Responses significantly higher than the blanks are indicated: *P < .05, **.001 < P < .05, ***P < .001 (determined by unpaired, one-tailed Student t test).

Close modal

In this study, we describe a successful strategy to identify promiscuous CD4 epitopes on candidate tumor antigens. Using a combined approach of computational prediction for promiscuous MHC class II epitopes and in vitro validation by biologic assays, we identified 4 immunodominant regions of MAGE-3, which contain naturally processed epitopes presented by several different HLA-DR alleles.

For most of the predicted epitopes, we found an excellent correlation between prediction and the ability of the peptides to bind promiscuously to HLA-DR molecules and to stimulate CD4+ T cells from different donors. This was especially striking for overlapping sequences MAGE-3141-155 and MAGE-3146-160, and overlapping sequences MAGE-3281-295 and MAGE-3286-300. On the contrary, MAGE-3111-125 and MAGE-3191-205 that, as predicted, bound HLA-DR*1101 but not HLA-DR*0101 and HLA-DR*0401, could strongly stimulate proliferation of CD4+ T cells in association with all the 3 HLA-DR alleles, pointing to the importance of the in vitro studies for validation of the predicted sequences. MAGE-3251-265, which was predicted to bind 48% of the alleles, did not bind well to the HLA-DR molecules that we tested (Table 1) and was never recognized by CD4+ T cells. MAGE-3251-265 partially overlaps MAGE-3243-258, which is recognized in association with HLA-DP4 but not with HLA-DR.30 MAGE-321-35, which was predicted to bind poorly, never elicited CD4+ T-cell responses. In agreement, Kobayashi et al15 also reported that the overlapping peptide sequence MAGE-322-36 could stimulate T lymphocytes from different donors in association with HLA-DR4, but the CD4+ T cells did not expand in culture.15 All the other sequences activated CD4+ T cells from at least 2 donors and did so in association with 2 to 4 different alleles (Figure1; Table 2), confirming the ability of TEPITOPE to predict promiscuous epitopes.

Table 2.

Summary of CD4 immunodominant MAGE-3 epitopes recognized by CD4+ T cells and their HLA-DR presenting alleles

HLA-DRResidues
111-125
Residues
146-160*
Residues
191-205
Residues
281-295
HLA-DR frequency
in whites, %
DR1 18 
DR4 24-26 
DR11 24-27 
DR13 114-127 NT NT NT 19 
DR7 NA NA NA 26 
HLA-DRResidues
111-125
Residues
146-160*
Residues
191-205
Residues
281-295
HLA-DR frequency
in whites, %
DR1 18 
DR4 24-26 
DR11 24-27 
DR13 114-127 NT NT NT 19 
DR7 NA NA NA 26 

NT indicates not tested; NA, not applicable because only HLA-DR7 positive donor tested.

*

Described also in Manici et al8 and Kobayashi et al.15.

Described in Manici et al.8 

Described in Chaux et al.27 

Most importantly, the immunodominant epitopes identified here are recognized in association with HLA-DR1, HLA-DR4, HLA-DR11, and HLA-DR7 that, with the addition of HLA-DR13,27 cover up to 94% of the alleles expressed in whites (Table 2). We cannot exclude that HLA-DRB3 or HLA-DRB4 molecules in addition to HLA-DRB1 can also present the identified sequences.

The immunodominant regions contain epitopes that are naturally processed from different sources of tumor antigen and by different APCs. In our present and previous studies,8 sequences MAGE-3141-160, MAGE-3191-205, and MAGE-3281-295 were recognized by cytolytic CD4+T cells after endogenous processing of MAGE-3 by LCLs and melanoma cells. Moreover, sequence MAGE-3281-295 was recognized by CD4+ T cells after processing and cross-presentation of the recombinant protein by autologous APCs.8MAGE-3141-160–reactive T-helper cells were also shown to recognize various forms of the MAGE-3 protein (tumor cell lysate, dead/apoptotic tumor cells, or recombinant protein).15MAGE- 3111-125 largely overlaps MAGE-3114-127, which is produced after processing and presentation of recombinant MAGE-3 protein by autologous dendritic cells and by melanoma cells that express an invariant chain–MAGE-3 fusion protein in the endosomal/lysosmal compartment.27 On the contrary, MAGE-3161-175–specific and MAGE-3171-185–specific CD4+ T cells did not recognize in vitro the MAGE-3 protein after processing through either the endogenous or the exogenous pathways. A possible explanation for this finding is that we failed to detect recognition of the native epitope because peptides bind to HLA molecules in a different conformation when they are loaded exogenously or when they are generated endogenously, as shown by Unanue and colleagues.31,32 In contrast with “naturally processed” epitopes that derive from processing of the whole protein antigen, “cryptic” epitopes are presented after the exposure to the antigen in peptide form, but not after processing of the whole protein antigen.33 Protease activity as well as the nature of the APCs during the processing of the protein may have a major influence on the repertoire of naturally processed or cryptic epitopes displayed for T-cell recognition.33 It is interesting that, in addition to the immunodominant epitopes, these sequences were strongly and predominantly recognized after long-term culture by CD4+ T cells from donor/patient 5, who had metastatic melanoma. We cannot completely exclude that priming of CD4+ T cells recognizing these sequences occurred in vivo as a consequence of epitope spreading during inflammatory conditions and operated by APCs not tested in our in vitro studies, leading to the formation of “cryptic” epitopes.

The use of pools of peptides for CD4+ T-cell stimulation has the advantage that competition among peptides with different binding affinity for the presenting alleles mimics in culture what occurs in vivo, where different processed peptides compete for MHC class II binding and T-cell–receptor recognition. Therefore, the finding that some peptides are predominant in our cultures may be related to their higher affinity for the MHC class II molecules, or to the presence of peptide-specific CD4+ T cells expressing T cell receptor with high affinity for the peptide/MHC class II complexes. The finding that CD4+ T cells specific for MAGE-3191-205 and MAGE-3281-295 proliferate at peptide concentrations as low as 1 to 10 ng/mL (data not shown) supports the latter possibility. The predominant recognition of different immunodominant regions in different donors may also be related to their precursor repertoires.

Animal models have shown that tumor-specific CD4+ T cells, in addition to playing a role in the induction of the antitumor immune response, have direct effector functions.1-4 The results of the present and previous studies by us and others8,33-36 strongly support a potential role of human tumor-specific CD4+ T cells as effectors of the antitumor immune response. Although the significance of the CD4+ T cells compared with CD8+ T cells in vivo remains to be clarified, lytic function of CD4+ T cells may be relevant when neoplastic cells, constitutively or as a consequence of inflammatory milieu, express MHC class II molecules. It will be interesting to verify the expansion of CD4+ CTLs ex vivo from cancer patients, as it has been shown,37 where CD4+ perforin-positive T cells were expanded in donors with chronic viral infections.

Clinical trials of vaccination with synthetic peptides corresponding to the HLA-A1–restricted MAGE-3 epitope recognized by CD8+ T cells have started in patients bearing MAGE-3+tumors.9-13 The additional use of peptides corresponding to the immunodominant CD4 epitopes identified here would increase the magnitude and life span of the antitumor immune response in these patients. Moreover, the identification of these epitopes will help in the study of the number and function of MAGE-3–specific CD4+ T cells, and the reliable monitoring of the immune responses in cancer patients before and after vaccination.

We thank Cinzia Arcelloni for high-performance liquid chromatography analyses, and Paolo Dellabona, Matteo Bellone, and Angelo Manfredi for enjoyable discussions and critical reading of the manuscript.

Prepublished online as Blood First Edition Paper, September 19, 2002; DOI 10.1182/blood-2002-03-0933.

Supported by the AIRC (Italian Association for Cancer Research), European Community (grant QLK2-CT2000-00470; M.P.P.), the MURST (Ministry of University and Scientific Research), the Ministry of Health, the National Heart, Lung, and Blood Institute (grant HL6192; B.M.C.-F.) and the National Institute of Neurological Disorders and Stroke (grant NS23919; B.M.C.-F.).

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
Pardoll
DM
Topalian
SL
The role of CD4+ T cell responses in antitumor immunity.
Curr Opin Immunol.
10
1998
588
594
2
Toes
RE
Ossendorp
F
Offringa
R
Melief
CJ
CD4 T cells and their role in antitumor immune response.
J Exp Med.
189
1999
753
756
3
Mumberg
D
Monach
PA
Wanderling
S
et al
CD4+ T cells eliminate MHC class II-negative cancer cells in vivo by indirect effects of IFN-γ.
Proc Natl Acad Sci U S A.
96
1999
8633
8639
4
Qin
Z
Blankenstein
T
CD4+ T cell-mediated tumor rejection involves inhibition of angiogenesis that is dependent on IFN-γ receptor expression by nonhematopoietic cells.
Immunity.
12
2000
677
686
5
Pawelec
G
Muller
L
Wagner
W
MHC class II-restricted tumor antigens and CD4+ T cells play a role in hematological malignancies as well as solid tumors.
Trends Immunol.
22
2001
422
423
6
Wang
R-F
The role of MHC class II-restricted tumor antigens and CD4+ T cells in antitumor immunity.
Trends Immunol.
22
2001
269
276
7
Van der Eynde
BJ
van der Bruggen
P
T cell defined tumor antigens.
Curr Opin Immunol.
9
1997
684
693
8
Manici
S
Sturniolo
T
Imro
MA
et al
Melanoma cells present a MAGE-3 epitope to CD4+ cytotoxic T cells in association with histocompatibility leukocyte antigen DR11.
J Exp Med.
189
1999
8871
876
9
Marchand
M
Weynants
P
Rankin
E
et al
Tumor regression responses in melanoma patients treated with a peptide encoded by gene MAGE-3.
Int J Cancer.
63
1995
883
885
10
Marchand
M
van Baren
N
Weynants
P
et al
Tumor regression observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1.
Int J Cancer.
80
1999
219
230
11
Nestle
FO
Alijagic
S
Gilliet
M
et al
Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells.
Nat Med.
4
1998
328
332
12
Thurner
B
Haendle
I
Roder
C
et al
Vaccination with MAGE-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma.
J Exp Med.
190
1999
1669
1678
13
Banchereau
J
Palucka
AK
Dhodapkar
M
et al
Immune and clinical responses in patients with metastatic melanoma to CD34(+) progenitor-derived dendritic cell vaccine.
Cancer Res.
61
2001
6451
6458
14
Cancer Immunity. Peptide database. Available at:http://www.cancerimmunity.org/peptidedatabase/tumorspecific.htm
15
Kobayashi
H
Song
Y
Hoon
DSB
Appella
E
Celis
E
Tumor-reactive T helper lymphocytes recognize a promiscuous MAGE-3 epitope presented by various major histocompatibility complex class II alleles.
Cancer Res.
61
2001
4773
4778
16
Sturniolo
T
Bono
E
Ding
J
et al
Generation of tissue-specific and promiscuous HLA ligand database using DNA microarrays and virtual HLA class II matrices.
Nat Biotechnol.
17
1999
555
561
17
Houghten
RA
General method for the rapid solid phase synthesis of large numbers of peptides: specificity of antigen-antibody interaction at the level of individual amino acids.
Proc Natl Acad Sci U S A.
82
1985
5131
5135
18
Raddrizzani
L
Sturniolo
T
Guenot
J
et al
Different modes of peptide interaction enable HLA-DQ and HLA-DR molecules to bind diverse peptide repertoires.
J Immunol.
159
1997
703
711
19
Cammarota
G
Scheirle
A
Takas
B
et al
Identification of a CD4 binding site on the β2 domain of HLA-DR molecules.
Nature.
356
1992
799
801
20
Sinigaglia
F
Romagnoli
P
Guttinger
M
Takas
B
Pink
JRL
Selection of T-cell epitopes and vaccine engineering.
Methods Enzymol.
203
1992
370
386
21
Roche
PA
Cresswell
P
High-affinity binding of an influenza hemagglutinin-derived peptide to purified HLA-DR.
J Immunol.
144
1990
1849
1856
22
Protti
MP
Manfredi
AA
Straub
C
Wu
X
Howard
JF
Jr
Conti-Tronconi
BM
Use of synthetic peptides to establish anti-human acetylcholine receptor CD4+ cell lines from myasthenia gravis patients.
J Immunol.
144
1990
1711
1720
23
Kocher
T
Schultz-Tjater
E
Gudat
F
Schaefer
C
Heberer
M
Spagnoli
G
Identification and intracellular location of MAGE-3 gene product.
Cancer Res.
55
1995
2236
2239
24
Landry
C
Brasseur
F
Soagnoli
GC
et al
Monoclonal antibody 57B stains tumor tissues that express gene MAGE-A4.
Int J Cancer.
86
2000
835
841
25
Imro
MA
Dellabona
P
Manici
S
et al
Human melanoma cells transfected with the B7–2 costimulatory molecule induce tumor specific CD8+ cytotoxic T lymphocytes in vitro.
Hum Gene Ther.
9
1998
1335
1344
26
Dalerba
P
Ricci
A
Russo
V
et al
High homogeneity of Mage, Bage, Gage, Tyrosinase, and Melan-A/MART-1 gene expression in clusters of multiple simultaneous metastases of human melanoma: implication for protocol design of therapeutic antigen-specific vaccination strategies.
Int J Cancer.
77
1998
200
204
27
Chaux
P
Vantomme
V
Stroobant
V
et al
Identification of MAGE-3 epitopes presented by HLA-DR molecules to CD4 (+) T lymphocytes.
J Exp Med.
189
1999
767
778
28
Fleischhauer
K
Tanzarella
S
Russo
V
et al
Functional heterogeneity of HLA-A*02 subtypes revealed by presentation of a MAGE-3-encoded peptide to cytotoxic T cell clones.
J Immunol.
159
1997
2513
2521
29
Protti
MP
Imro
MA
Manfredi
AA
et al
Particulate naturally processed peptides prime a cytotoxic response against human melanoma in vitro.
Cancer Res.
56
1996
1210
1213
30
Schultz
ES
Lethé
B
Cambiaso
CL
et al
A MAGE-3 peptide presented by HLA-DP4 is recognized on tumor cells by CD4+ cytolytic T lymphocytes.
Cancer Res.
60
2000
6272
6275
31
Viner
NJ
Nelson
CA
Deck
B
Unanue
ER
Complexes generated by the binding of free peptides to class II MHC molecules are antigenically diverse compared with those generated by intracellular processing.
J Immunol.
156
1996
2365
2386
32
Peterson
DA
Di Paolo
RJ
Kanagawa
O
Unanue
ER
Cutting edge: a single MHC anchor residue alters the conformation of a peptide-MHC complex inducing T cells that survive negative selection.
J Immunol.
166
2001
5874
5877
33
Sercarz
EE
Lehmann
PV
Amentani
A
Benichou
G
Miller
A
Moudgil
K
Dominance and crypticity of T cell antigenic determinants.
Annu Rev Immunol.
11
1993
729
766
34
Takahashi
T
Chapman
PB
Yang
SY
Hara
I
Vijayasaradhi
S
Houghton
AN
Reactivity of autologous CD4+ T lymphocytes against human melanoma.
J Immunol.
154
1995
772
779
35
Zarour
HM
Kirkwood
JM
Salvucci Kierstead
L
et al
Melan-A/MART-1 (51-73) represents an immunogenic HLA-DR4-restricted epitope recognized by melanoma-reactive CD4+ T cells.
Proc Natl Acad Sci U S A.
97
2000
400
405
36
Touloukian
CE
Leitner
WW
Topalian
SL
et al
Identification of a MHC class II-restricted human gp100 epitope using DR4-IE transgenic mice.
J Immunol.
164
2000
3535
3542
37
Appay
V
Zaunders
JJ
Papagano
L
et al
Characterization of CD4+ CTLs ex vivo.
J Immunol.
168
2002
5954
5958

Author notes

Maria Pia Protti, Laboratory of Tumor Immunology, Cancer Immunotherapy and Gene Therapy Program, DIBIT, Scientific Institute H. San Raffaele, Via Olgettina 58, 20132 Milan, Italy; e-mail: m.protti@hsr.it.

Sign in via your Institution