It is generally considered that MHC class I-restricted antigens are recognized by CD8+ T cells, whereas MHC class II-restricted antigens are recognized by CD4+ T cells. In the present study, we report an MHC class I-restricted CD4+ T cell isolated from the tumor infiltrating lymphocytes (TILs) of a patient with metastatic melanoma. TIL 1383 I recognized HLA-A2+ melanoma cell lines but not autologous transformed B cells or fibroblasts. The antigen recognized by TIL 1383 I was tyrosinase, and the epitope was the 368–376 peptide. Antibody blocking assays confirmed that TIL 1383 I was MHC class I restricted, and the CD4 and CD8 coreceptors did not contribute significantly to antigen recognition. TIL 1383 I was weakly cytolytic and secreted cytokines in a pattern consistent with it being a Th1 cell. The avidity of TIL 1383 I for peptide pulsed targets is 10–100-fold lower than most melanoma-reactive CD8+ T cell clones. These CD4+ T cells may represent a relatively rare population of T cells that express a T-cell receptor capable of cross-reacting with an MHC class I/peptide complex with sufficient affinity to allow triggering in the absence of the CD4 coreceptor.

T cells can mediate the recognition and elimination of virus-infected cells, tissue allografts, cells expressing normal self antigens, and tumor cells in humans and in animal models. Mature T cells develop in the thymus from bone marrow-derived progenitor cells (reviewed in Refs. 1 and 2). T-cell antigens are recognized in association with MHC molecules, resulting in MHC-restricted antigen recognition (3). T cells expressing the CD8 coreceptor on their cell surface recognize 8–10 amino acid peptide fragments bound to MHC class I molecules and thus are MHC class I restricted (4, 5). CD8+ T cells are generally CTLs and commonly represent the effector arm of T cell-mediated immunity. T cells expressing the CD4 coreceptor on their cell surface recognize 10–15 amino acid peptide fragments bound to MHC class II molecules and thus are MHC class II restricted (4, 5). CD4+ T cells are generally helper T cells that secrete cytokines that initiate and augment the function of CTL and B cells.

Broad cross-reactivity exists among T cells in the periphery and in the thymus, suggesting that epitope mimicry plays an important part in shaping the T-cell repertoire (6, 7). The sequence and relative abundance of individual peptides presented by the thymic epithelium can have dramatic effects on T-cell development (8, 9). Individual peptides are capable of positively selecting a diverse T-cell repertoire in fetal thymic organ cultures (10). In the periphery, individual T-cell clones can cross-react with alloantigens (11) and a broad spectrum of peptides in the context of self MHC (12, 13, 14). These studies suggest that cross-reactivity is an important mechanism for generating and maintaining T-cell immunity.

In the present study, we describe an HLA-A2-restricted, melanoma-reactive CD4+ TIL2 culture from melanoma patient 1383. Transient transfection experiments identified the antigen that was recognized by TIL 1383 I as tyrosinase. The epitope recognized by TIL 1383 I was a 9 amino acid peptide corresponding to amino acid residues 368–376 from tyrosinase. TIL 1383 I was weakly cytolytic and secreted cytokines in a pattern consistent with T helper 1 (Th1) cells that produce IFN-γ, IL-2, GM-CSF, and TNF-α but not IL-4 or IL-10. The MHC class I restricted antigen recognition by CD4+ T cells supports the notion that epitope mimicry exists in the immune response by T cells.

Tumor Cell Lines.

All melanoma, immortalized fibroblasts (E6E7 transformed) and EBV B-cell lines used in this study were established from surgical specimens (TILs, melanoma, and fibroblasts) or peripheral blood mononuclear cells (EBV B) from melanoma patients undergoing immunotherapy in the Surgery Branch, National Cancer Institute, as described previously (15, 16, 17). Prostate cancer lines 1542 CP3TX (HLA-A2) and 1550 CPTX (HLA-A2+) were kindly provided by Dr. Suzanne L. Topalian (Surgery Branch, National Cancer Institute, NIH, Bethesda, MD) and were established from patients undergoing radical prostatectomies at the National Cancer Institute as described previously (17). Renal cell carcinoma lines UOK 181 (HLA-A2) and UOK 131 (HLA-A2+) were kindly provided by Dr. W. Marston Linnehan (Urology Branch, National Cancer Institute, NIH, Bethesda, MD) and were established from patients undergoing radical nephrectomy at the National Cancer Institute as described previously (18). Esophageal carcinoma cell lines HCE-4 (HLA-A2) and SKGT-5 (HLA-A2+) were kindly provided by Dr. David S. Schrump (Surgery Branch, National Cancer Institute; Ref. 19). Colon carcinoma cell lines LOVO (HLA-A2) and SW480 (HLA-A2+) lines, breast carcinoma cell lines MCF7 (HLA-A2+) and MDA-MB-231 (HLA-A2+), and ovarian carcinoma cell line SKOV3 (HLA-A2) were obtained from American Type Culture Collection (Rockville, MD). 624–28 MEL (HLA-A2) and 624–38 MEL (HLA-A2+) were obtained by limiting dilution cloning of 624 MEL (20). 888 A2 MEL, 397 A2 MEL, and SKOV3 A2 were obtained by transfecting 888 MEL, 397 MEL, and SKOV3, respectively, with a cDNA encoding HLA-A2 that was cloned into the pcDNA 3 eukaryotic expression vector (In Vitrogen, Carlsbad, CA). COS-7 cells that were used for transient transfection assays and T2 cells that were used for peptide recognition assays have been described elsewhere (21, 22). All tumor cell lines except the prostate cell lines were maintained in CM that consisted of RPMI 1640 (Biofluids, Rockville, MD) supplemented with 10% heat-inactivated fetal bovine serum (Life Technologies, Inc., Rockville, MD) and 1% penicillin-streptomycin-glutamine (Life Technologies). The prostate cell lines were maintained in Keratinocyte-SFM (Life Technologies) supplemented with 5% heat-inactivated fetal bovine serum, 1% penicillin-streptomycin-glutamine, 10 mm HEPES (Biofluids, Rockville, MD), and 25 mm bovine pituitary extract (Life Technologies).

Generation of TIL Lines.

All TILs were established from surgical specimens obtained from patients with metastatic melanoma as described (15). Viable cells (1 × 106; tumor and lymphocytes) from patient 1383 were plated in each of 12 wells of a 24-well plate in a total of 2 ml of TIL medium that consisted of AIM V medium (Life Technologies) supplemented with 5% heat-inactivated pooled AB human serum (Sigma Chemical Co., St. Louis, MO), 1% penicillin-streptomycin-glutamine, and 6000 IU/ml recombinant human IL-2 (Chiron, Emeryville, CA). All TIL cultures were expanded and maintained at densities between 0.5–2.0 × 106 cells/ml. TIL 1235 and TIL 1520 are HLA-A2 restricted CD8+ TIL cultures that recognize the MART-1:27–35 and gp100:209–217 peptide epitopes, respectively (22, 23). TIL 888 is an HLA-A24-restricted CD8+ TIL culture that recognizes tyrosinase (24). TIL 1558 is an HLA-DR1-restricted CD4+ TIL culture that recognizes a mutated triosephosphate isomerase that is expressed only by its autologous 1558 MEL (25).

Plasmids.

The cloning of the HLA-A2.1 (22), MART-1 (22), gp100 (26), tyrosinase (24), TRP-2 (27), and NY Eso-1 (28) cDNAs has been described elsewhere. All cDNAs were subcloned into the eukaryotic expression vector pcDNA 3 (In Vitrogen).

Synthetic Peptides.

Peptides were synthesized by a solid phase method using an AMS 422 multiple peptide synthesizer (Gilson Co., Worthington, OH) as described previously (29). The amino acid sequences of the MART-1, gp100, and tyrosinase peptides used in this study are as follows: MART-1:27–35, AAGIGILTV (22); gp100:209–217, ITDQVPFSV (30); tyrosinase:1–9, MLLAVLYCL (31); tyrosinase:368–376 370 D, YMDGTMSQV (32); and tyrosinase:368–376 370 N, YMNGTMSQV (31). The amino acid sequence of the hepatitis B virus core protein:18–27 23Y peptide (HBV:18–27 23Y) that was used for peptide binding assays was FLPSDYFPSV (33). All peptides were dissolved in DMSO at 2 mg/ml.

Peptide Binding to HLA-A2.

The binding of peptides to HLA-A2 was measured using a live cell competitive binding assay as described, except T2 cells were substituted for EBV-transformed B cells (33). Values obtained represented the IC50 for each peptide, which is the concentration of peptide that inhibits 50% of the binding of 125I-labeled hepatitis B virus core protein residues 18–27 modified with a tyrosine at P6 (HBV:18–27 23Y) to HLA-A2 molecules on T2 cells. HBV:18–27 23Y was radiolabeled with 125I using chloramine T, and labeled peptide was separated from the unincorporated 125I using a Sephadex G-10 (Sigma) size exclusion column as described (33). Binding of 125I-labeled HBV:18–27 23Y peptide to T2 cells was measured in the presence of 10, 1, 0.1, 0.01, and 0.001 mg/ml unlabeled test peptide. The maximum binding and maximum inhibition values for each assay were determined by incubating T2 cells with 125I-labeled HBV:18–27 23Y peptide alone or with an excess (50 mg/ml) of unlabeled HBV:18–27 23Y peptide. Each determination was performed in triplicate, and their average was used to calculate the percentage of inhibition as follows:

The ln(% inhibition) was plotted versus the ln(peptide concentration). Using regression analysis, the IC50 was determined as the peptide concentration that inhibited 50% of the binding of the 125I-labeled HBV:18–27 23Y peptide.

Synthetic Oligonucleotides.

Oligonucleotide primers were purchased from Life Technologies. The sequences of the human TCR BV subfamily-specific primers have been described elsewhere (34). Human TCR BV subfamily-specific primers were designed based on the genomic TCR BV sequences reported by Rowen et al.(35). Each primer was designed such that there was no more than a single base mismatch with any member of the subfamily. The official nomenclature proposed by the International Union of Immunological Societies subcommittee on nomenclature has been used to identify all TCR gene segments described (36).

RNA Isolation and PCR.

Total cellular RNA was isolated from 5 × 106 TIL 1383 I cells using Trizol (Life Technologies, Inc., Gaithersburg, MD), and cDNA was synthesized using oligo dT(12–18) and Superscript II reverse transcriptase (Life Technologies, Inc., Gaithersburg, MD) as described (34). Each TCR BV subfamily-specific forward primer was used in conjunction with a BC reverse primer to amplify the TCR BV transcripts present in the TIL culture. PCR reactions consisted of 200 μm deoxynucleotide triphosphate (Oncor, Gaithersburg, MD), 400 nm of both forward and reverse primers, and 1 unit of Taq polymerase (Oncor, Gaithersburg, MD) in a 50-μl reaction. Amplifications were performed in a Perkin-Elmer 9600 DNA thermocycler (Norwalk, CT) under the following conditions: 35 cycles of 92°C denaturation for 30 s, 60°C annealing for 30 s, and 72°C extension for 1 min. PCR products were separated using ethidium bromide-stained agarose gels and electronically imaged using an Eagle Eye II Still Video System (Stratagene, La Jolla, CA).

DNA Sequence Analysis.

The DNA sequence analysis of the TCRBV12 band from TIL 1383 I was performed using the BV12a primer and Dye Terminator Cycle Sequencing kits (Perkin-Elmer/ABI, Foster City, CA). Sequences were determined using an ABI Prism 310 Genetic Analyzer (Perkin-Elmer/ABI). The resulting sequences were analyzed using the Genetics Computer Group, Inc. software package (37).

Immunofluorescence and Cell Sorting.

Cell surface expression of TCRBV12, CD3, CD4, and CD8 molecules on TIL 1383 I was measured by immunofluorescence using FITC-conjugated anti-TCR BV12 (Immunotech, Westbrook, ME), PE-conjugated anti-CD3 (Becton Dickinson, San Jose, CA), FITC- or PE-conjugated anti-CD4 (Becton Dickinson), and FITC- or PE-conjugated anti-CD8 mAbs (Becton Dickinson). For analysis, the relative log fluorescence of 104 live cells was measured using a FACScan flow cytometer (Becton Dickinson). For cell sorting, the TCR BV12 mAb was dialyzed against PBS to remove the sodium azide and filter sterilized. Cells (2 × 107) cells from a 54-day-old TIL 1383 I culture were stained with FITC-conjugated anti-TCR BV12 mAb. BV12-expressing cells were collected using a Coulter Elite flow cytometer (Coulter, Miami, FL).

Tumor Antigen Recognition Assays.

The reactivity of TIL 1383 I was measured in cytokine release assays as described (38). Briefly, TIL 1383 I cells were cocultured overnight with stimulator cells in a 1:1 ratio (the actual number of stimulators and responders used are indicated in the table legends) in RPMI 1640 supplemented with 10% heat-inactivated pooled human AB serum and 1% penicillin-streptomycin-glutamine in a total volume of 200 ml in 96-well U-bottomed plates. Supernatants were harvested, and the amount of cytokine release was measured by ELISA. IFN-γ, TNF-α, IL-4, and IL-10 ELISA kits were purchased from Endogen (Woburn, MA). GM-CSF and IL-2 ELISA kits were purchased from R&D Systems (Minneapolis, MN). The stimulators used in these assays included a panel of HLA-A2+ and HLA-A2 tumors, EBV-transformed B cells, fibroblast lines, and peptide-pulsed T2 cells. Peptide-pulsed T2 cells were prepared by incubating 1 × 106 T2 cells/ml with peptide for 2 h at 37°C.

Transient Transfection Assays.

COS-7 cells were transiently transfected using Lipofectamine Plus (Life Technologies, Rockville, MD) as described (39). Briefly, 5 × 104 COS-7 cells/well in 96-well, flat-bottomed plates were incubated in 40 ml of serum-free DMEM medium containing 200 ng of pcDNA 3 containing the cloned tumor-associated antigen cDNA (150 ng) and HLA-A2.1 (50 ng) cDNA at 37°C in a humidified CO2 incubator (5% CO2) for 3 h. Then, 160 ml of DMEM medium supplemented with 10% heat-inactivated fetal bovine serum and 1% penicillin-streptomycin-glutamine was added. After 2 days, 100 ml of medium were removed, and 2.5 × 104 T cells in 100 ml of CM were added to each well. The amount of IFN-γ was measured by ELISA as described above.

Antibody Inhibition Assays.

The ability of anti-MHC, anti-CD4, and anti-CD8 mAbs to inhibit antigen recognition by TIL 1383 I was measured in cytokine release assays as described (38). Briefly, 1 × 104 T cells and 1 × 104 tumor cells were cocultured in RPMI 1640 supplemented with 10% heat-inactivated pooled AB serum and 1% penicillin-streptomycin-glutamine in a total volume of 200 ml in 96-well U-bottomed plates for 24 h. For the MHC blocking assays, the tumor cells were incubated with IVA12 and/or W6/32 mAb (final concentration, 20 mg/ml) for 30 min at room temperature prior to the addition of the T cells. For the CD4/CD8 blocking assays, the T cells were incubated with anti-CD4 (final concentration, 2.5 mg/ml) or anti-CD8 (final concentration, 1.25 mg/ml) mAb for 30 min at room temperature prior to the addition of the tumor cells. The amount of IFN-γ released was measured by ELISA as described above.

51Cr Release Assays.

The ability of TIL 1383 I to lyse HLA-A2+, tyrosinase+ target cells was measured in 51Cr release assays as described (15). Briefly, 106 melanoma or peptide-pulsed T2 cells were labeled for 1 h at 37°C with 200 mCi of 51Cr (Amersham, Arlington Heights, IL) in CM. Labeled target cells (5 × 103) were incubated with 4 × 105 (80:1), 1 × 105 (20:1), 2.5 × 104 (5:1), and 6.25 × 103 (1.25:1) effector cells for 4 h at 37°C in 200 ml of CM. Supernatants were harvested and counted using an Wallac 1470 Wizard automatic gamma counter (Wallac, Gaithersburg, MD). Total and spontaneous 51Cr release by each target was determined by incubating 5 × 103 labeled target cells in 2% SDS or CM, respectively, for 4 h at 37°C. Each point represented the average of triplicate wells, and percentage of specific lysis was calculated as follows:

Twelve TIL cultures from patient 1383 were established from an enzymatically digested melanoma lesion by culturing the cells in medium containing 6000 IU/ml of recombinant human IL-2. Each culture was expanded separately and tested on day 17 for its reactivity against the autologous 1383 MEL, an HLA-A2+ and an HLA-A2 melanoma cell line. One culture, designated TIL 1383 I, appeared to be HLA-A2 restricted and melanoma reactive in the initial screen (data not shown). TIL 1383 I was expanded, and its reactivity was determined using a more extensive panel of melanoma and nonmelanoma cell lines. As shown in Table 1, TIL 1383 I secreted significant amounts of IFN-γ (at least twice background and >100 pg/ml) when stimulated with 9 of the 11 HLA-A2+ melanomas but with none of the five HLA-A2 melanomas (Table 1). TIL 1383 I did not recognize any of the HLA-A2+ or HLA-A2 EBV transformed B-cell, fibroblast, prostate carcinoma, ovarian carcinoma, renal cell carcinoma, colon carcinoma, esophageal carcinoma, and breast carcinoma cell lines. These results indicated that TIL 1383 I recognized a shared melanoma antigen in the context of HLA-A2.

TIL 1383 I was then tested for its ability to recognize transfectants expressing a panel of melanoma antigens described previously containing known HLA-A2-restricted epitopes. As shown in Table 2, TIL 1383 I recognized COS cells that had been transiently transfected with the HLA-A2 and tyrosinase cDNAs. COS cells transfected with HLA-A2 and MART-1 were only recognized by TIL 1235, and COS cells transfected with HLA-A2 and gp100 were only recognized by TIL 1520. None of the TILs recognized COS cells transfected with HLA-A2 alone, HLA-A2 and TRP2, or HLA-A2 and Eso I. Recognition of the tumor cell controls in this assay was consistent with each of the TILs being HLA-A2 restricted and melanoma reactive. Therefore, the melanoma-associated antigen recognized by TIL 1383 I was tyrosinase.

Two HLA-A2-restricted tyrosinase epitopes consisting of amino acid positions 1–9 and 368–376 (designated T9-1 and T9-368, respectively) have been described previously (31). To determine whether one of these two HLA-A2-restricted tyrosinase epitopes were recognized by TIL 1383 I, T2 cells pulsed with either T9-1 or T9-368 were assayed for their ability to stimulate IFN-γ release from TIL 1383 I. The T9-368 peptide undergoes a posttranslational modification that changes the N at position 370, which is encoded by the tyrosinase gene to a D, which is found on the surface of melanoma cells (32). As shown in Table 3, TIL 1383 I recognized T2 cells pulsed with the T9-368 peptide with either N or D at position 370. TIL 1383 I recognized the HLA-A2+ melanoma lines but not T9-1, M9-27, or G9-209 pulsed T2 cells, 1383 fibroblasts, or 888 MEL. TIL 1235 recognized M9-27, TIL 1520 recognized G9-209, and TIL 620 recognized both M9-27 and G9-209 as expected. TIL 888, which is known to recognize tyrosinase in the context of HLA-A24, recognized the 888 MEL lines but did not recognize any of the HLA-A2-restricted peptides pulsed on T2 cells. Therefore, the previously described tyrosinase epitope T9-368 was recognized by TIL 1383 I, with both the 370N and the 370D forms of the peptide being recognized efficiently.

The TCR BV genes used by TIL 1383 I was determined by reverse transcription-PCR using TCR BV subfamily-specific primers as described (34). TIL 1383 I was oligoclonal because we could only detect BV12, BV14, and BV24 transcripts, with BV12 being the predominant subfamily used (data not shown). DNA sequence analysis of the BV12 band found only a single productive rearrangement, indicating that the BV12 subfamily was clonally expanded in TIL 1383 I (data not shown). To confirm that BV12+ T cells were the predominant T-cell clonotype in this culture, the expression of BV12, CD3, CD4, and CD8 on TIL 1383 I was measured by immunofluorescence. Essentially 100% of the T cells in TIL 1383 I expressed both CD3 and CD4 (Fig. 1,c), with ∼99% of the CD4+ T cells expressing BV12 (Fig. 1,d). Less than 1% of the TIL 1383 I cells were CD8+ (Fig. 1,e) or CD8+ and BV12+ (Fig. 1,f). These rare events were attributable to nonspecific staining by the anti-CD8 mAb because there were no CD4 T cells in TIL 1383 I (Fig. 1, c and d), and we were unable to detect CD8a chain transcripts by reverse transcription-PCR (data not shown). We wanted to obtain a pure population of BV12+, CD4+ T cells from TIL 1383 I to determine whether these were the tyrosinase-reactive cells in the TIL 1383 I culture. Because attempts to clone TIL 1383 I in limiting dilution were unsuccessful, we stained a TIL 1383 I culture with an anti-BV12 mAb and used the cell sorter to further enrich the population of BV12+ T cells. After two sorts, we were able to generate a TIL 1383 I culture that was 100% BV12+ T cells (Fig. 2,c) from a culture that was 98% BV12+ T cells (Fig. 2,b). The sorted cells were expanded and tested for expression of BV12, CD4, and CD8 and for their ability to recognize antigen. After 2 weeks of culture, 100% of sorted cells remained CD4+ and BV12+ (data not shown). When tested for their antigen recognition, the sorted cells specifically recognized T9-368 pulsed T2 cells, and the amount of IFN-γ released was equivalent to the amount released by the unsorted cells (Table 4). These results indicated that the HLA-A2-restricted recognition of tyrosinase by TIL 1383 I was by a BV12-expressing, CD4+ T cell.

Human T cells differ from murine T cells in that activated human T cells express MHC class II molecules and can process and present class II-restricted epitopes to each other. Therefore, it is possible that the tyrosinase peptide could be shed from the tumor cells and presented by the TIL 1383 I cells to each other in the context of MHC class II rather than the MHC class I. To demonstrate that the T9-368 peptide was directly presented to TIL 1383 I cells by HLA-A2 on the tumor cells or T2 cells, we compared cytokine secretion by TIL 1383 I when stimulated with peptide-pulsed T2 cells to T cells stimulated with free peptide. TIL 1383 I secreted significant amounts of IFN-γ when stimulated with T2 cells pulsed with 1.0, 0.1, or 0.01 mg/ml of T9-368 370D peptide (Fig. 3,a) and 1.0 or 0.1 mg/ml of T9-368 370N peptide (Fig. 3,b). In contrast, TIL 1383 I did not secrete IFN-γ when stimulated with the T9-368 370D or 370N peptide alone. The CD8+ T cell control TIL 1520 cells also secreted IFN-γ when stimulated with G9-209 pulsed T2 cells (Fig. 3 c). TIL 1520 cells secreted a low but significant amount of IFN-γ when stimulated with free peptide (200 pg/ml with 1.0 mg/ml peptide). When the ability of these peptides to bind to HLA-A2 molecules was measured in a competition binding assay, the IC50 for G9-209, T9-368 370D, and T9-368 370N was measured to be 74, 702, and 249 mm, respectively. One possible explanation for the ability of free G9-209 peptide to stimulate TIL 1520 could be that the binding affinity of G9-209 was high enough to allow a small amount of peptide to be bound to the HLA-A2 molecules on the surface of the T cells. The TIL 1520 cells could then stimulate each other to secrete IFN-γ. In contrast, TIL 1383 I cells were unable to stimulate each other when cultured with free peptide, possibly because the low binding affinity of the T9-368 peptides did not allow efficient binding of peptide to the HLA-A2 molecules on their cell surface. These results indicate that the IFN-γ released by TIL 1383 I could only be attributable to the recognition of T9-368/HLA-A2 complexes on the tumor cells.

The MHC class I-restricted recognition of tyrosinase by TIL 1383 I was further demonstrated in mAb blocking assays. As shown in Table 5, anti-MHC class I mAb W6/32 inhibited TIL 1383 I recognition of 1383 MEL (35%; 184.8 to 120.4 pg/ml), 1558 MEL (46%; 220.7 to 119.1 pg/ml), and T9-368 pulsed T2 cells (61%; 1146.9 to 451.7 pg/ml). In contrast, anti-MHC class II mAb IVA12 had little effect on TIL 1383 I recognition of melanoma cells (1383 MEL: 9%, 184.8 to 167.3 pg/ml; 1558 MEL: 0%, 234.2 to 220.7 pg/ml) or peptide-pulsed T2 cells (17%; 1146.9 to 955.7 pg/ml). W6/32 inhibited MHC class I restricted IFN-γ release by control of the CD8+ T cell lines TIL 1520 or TIL 1235, and IVA12 inhibited class II-restricted IFN-γ release by the control of CD4+ T-cell line TIL 1558, as expected (Table 5). The ability of W6/32 to inhibit recognition of HLA-A2+ tyrosinase+ melanoma cells and peptide-pulsed targets by TIL 1383 I is a further demonstration of its MHC class I-restricted reactivity.

The contribution of the CD4 coreceptor to antigen recognition by TIL 1383 I was determined by blocking cytokine release with anti-CD4 mAb. The amount of IFN-γ released by TIL 1383 I when stimulated with 1558 MEL was reduced by 15% (705.4 to 604.9 pg/ml) and 23% (705.4 to 546.5 pg/ml) in the presence of anti-CD4 mAb and anti-CD8 mAb, respectively (Table 5). Anti-CD4 mAb inhibited MHC class II-restricted recognition of 1558 MEL by CD4+ TIL 1558 (44% inhibition; 1651.0 to 930.0 pg/ml) but had no effect on MHC class I-restricted recognition of 1558 MEL by TIL 1520 (4% inhibition; 507.2 to 489.5 pg/ml). Anti-CD8 mAb inhibited MHC class I-restricted recognition of 1558 MEL by CD8+ TIL 1520 (46% inhibition; 507.2 to 276.0 pg/ml) but had minimal effects on MHC class II-restricted recognition of 1558 MEL by TIL 1558 (17% inhibition; 1651.0 to 1363.0 pg/ml). These results indicate that the CD4 coreceptor is not required for antigen recognition by TIL 1383 I.

The pattern of cytokine release from TIL 1383 I after antigen stimulation is shown in Table 6. TIL 1383 I secreted IFN-γ, GM-CSF, TNF-α, and IL-2 but not IL-4 or IL-10 when stimulated with either the autologous 1383 MEL, 1558 MEL, or OKT3. By comparison, CD8+ TIL 1520 secreted only IFN-γ and GM-CSF when stimulated with 1383 MEL or 1558 MEL and IFN-γ, GM-CSF, and TNF-α when stimulated with OKT3. In this experiment, 1383 fibroblast-stimulated TIL 1383 I secreted <20% of the amount of GM-CSF secreted by TIL 1383 I stimulated with tumor cells. Fibroblasts alone secreted small amounts of TNF-α and GM-CSF. These results indicate that the pattern of cytokine secretion by TIL 1383 I was consistent with a Th1 phenotype, despite being an MHC class I-restricted CD4+ T cell.

The ability of TIL 1383 I to lyse HLA-A2+ melanoma cells and T9-368 pulsed T2 cells was tested in 4-h 51Cr release assays. As shown in Fig. 4,a, TIL 1383 I weakly lysed 888 A2 MEL and T9–368 370D pulsed T2 cells but failed to lyse autologous 1383 MEL, 1383 fibroblasts, and M9–27 pulsed T2 cells. TIL 888, which recognizes tyrosinase in the context of HLA-A24, efficiently lysed only the HLA-A24+ melanoma lines 888 MEL and 888 A2 MEL (Fig. 4,b). TIL 1235 efficiently lysed 888 A2 MEL and M9-27-pulsed T2 cells but not 1383 MEL, 1383 fibroblasts, and T9-368 pulsed T2 cells (Fig. 4 c). The failure of 1383 MEL to be lysed by TIL 1235 suggested that 1383 MEL may be resistant to cell-mediated lysis. Therefore, despite being a CD4+ T cell, TIL 1383 I could weakly lyse some of the HLA-A2+, tyrosinase+ targets.

Classically, CD4+ T cells recognize antigens presented by MHC class II molecules, whereas CD8+ T cells recognize antigens presented by MHC class I molecules (3, 4, 5). Recently, several examples of MHC class I-restricted CD4+ T cells have been reported (40, 41, 42, 43, 44, 45, 46). However, the existence of MHC class I-restricted antigen recognition by CD4+ T cells has remained controversial because no single study has been able to exclude the presence of contaminating CD8+ T cells in the culture, determine the MHC class I restriction element, and identify the nominal antigen recognized by the T cells. In this study, we described an MHC class I-restricted CD4+ T cell isolated from the infiltrating lymphocytes of a human melanoma lesion. Immunofluorescence analysis indicated that 100% of the cells in the bulk TIL 1383 I culture and cells sorted for high expression of BV12 were CD3+CD4+ T cells with no evidence of contaminating CD8+ T cells (Figs. 1 and 2). The reactivity of TIL 1383 I with a broad panel of tumor targets indicated that the antigen recognized by TIL 1383 I was melanoma associated and HLA-A2 restricted (Table 1). The antigen recognized by TIL 1383 I was tyrosinase, with the 368–376 peptide being the antigenic peptide (Tables 2 and 3). Identification of the antigenic peptide enabled us to conclusively demonstrate that a ligand for the TCR expressed by the CD3+CD4+BV12+ T cell in TIL 1383 I was the T9-368 peptide derived from tyrosinase bound to HLA-A2 (Fig. 3).

Current models of MHC restriction, antigen specificity, and T-cell development require that T cells are positively selected by peptide/MHC molecules expressed on the host thymic epithelial cells (47, 48). From these models, there are several potential ways an MHC class I-restricted CD4+ could have developed. One possibility is that TIL 1383 I was positively selected by T9–368/HLA-A2 complexes expressed on the thymic epithelium. Peptides presented by MHC class I molecules are generally derived from proteins encoded endogenously by the antigen-presenting cell, implying that all of the peptides involved in the positive selection of CD8+ T cells must be derived from proteins encoded by the thymus. The expression of tyrosinase is limited to cells of the melanocyte/melanoma lineage (49). Unless the T9-368 peptide was shed by melanocytes and exchanged with peptides on MHC class I molecules in the thymus, T9-368/HLA-A2 complexes should not exist in the thymus and would therefore be unavailable to positively select TIL 1383 I. However, we cannot exclude the formal possibility that TIL 1383 I was positively selected by some cross-reactive, thymus-derived peptide presented by HLA-A2. It is also possible that TIL 1383 I could be functionally a CD4+ CD8+ T cell that carries a mutant CD8 chain that fails to bind our anti-CD8 mAb, making TIL 1383 I appear to be a CD4+CD8 T cell. However, the model that is most consistent with the current theories of positive selection is that TIL 1383 I was positively selected by some undefined peptide in association with an MHC class II molecule in this patient, and the HLA-A2-restricted recognition of the T9-368 peptide is merely an unusual cross-reaction.

Cross-reactivity is commonly observed among mAbs but has also been attributed to T cells (6, 7, 12, 13, 14). In the immune response to human melanoma, epitope mimicry was directly investigated by determining the ability of MART-1-reactive CTLs to recognize a panel of naturally occurring peptide mimics derived from human self antigens and pathogens (12). Many of these peptides were recognized by the MART-1-reactive CTL clones, and their pattern of peptide recognition (which peptides were recognized and the magnitude of the response) was unique to each T-cell clone. It is, therefore, possible that epitope mimics of the T9-368 peptide exist that can be recognized by TIL 1383 I or that the T9-368 peptide is itself a mimic of some other unknown peptide.

We believe that the variation in the way each T-cell clone recognizes antigen is reflected by the variation in the TCR V genes used by the T-cell clone. Epitope mimicry should then lead to TCR diversity in the immune response to a single antigenic peptide. Despite reports of restricted TCR V gene usage by melanoma reactive T cells (50), we have found TCR β chain usage by melanoma-reactive T cells to be quite diverse (34, 51, 52). In one patient, as many as 10 distinct TCR clonotypes using seven different TCR BV genes were used to recognize the same gp100:209–217/HLA-A2 complex.3 These observations are consistent with the hypothesis that the vast TCR diversity we observe among melanoma-reactive T cells may be attributable to the broad cross-reactivity and redundancy of the immune system.

If epitope mimicry can lead to CD4+ T cells that recognize antigen in the context of MHC class I molecules, then T cells should be more common than they appear. There are probably two main reasons why there are very few reported examples of class I-restricted CD4+ T cells: (a) most class I-restricted T cells are isolated based on their ability to lyse 51Cr-labeled targets. Because most CD4+ T cells are not cytolytic, class I-restricted CD4+ T cells would be generally overlooked in lysis assays. In fact, had we used lysis assays as our initial screen, TIL 1383 I would have been classified as nonreactive and discarded because it failed to lyse the autologous melanoma (Fig. 4); and (b) most T cells require the CD4 or CD8 coreceptors to respond to antigen stimulation (53, 54). CD4 binds to the b2 domain of MHC class II molecules (55), and CD8 binds to the a3 domain of MHC class I molecules (56). The function of these coreceptors is to stabilize the TCR/peptide/MHC complex (57, 58) and enhance T-cell signaling by augmenting CD3z and ZAP70 phosphorylation (59). However, there is T-cell to T-cell variation in the requirement for CD8 for T-cell function (60). Those T cells whose function is CD8 dependent have low- to intermediate-affinity TCRs, and those T cells whose function is CD8 independent have high-affinity TCRs (61, 62). CD4 and CD8 independent clones are relatively rare, suggesting that T cells bearing high-affinity TCRs are relatively rare. Our inability to block T9-368 peptide or tumor cell recognition with anti-CD4 mAb, together with incomplete blocking of IFN-γ secretion by anti-MHC class I mAb, strongly suggests that the BV12 TCR expressed by TIL 1383 I must have sufficient affinity for the T9-368/HLA-A2 complex that it can activate the T cell in the absence of the CD4 coreceptor. Despite having a high-affinity TCR, TIL 1383 I has relatively low avidity compared with other class I-restricted T cells (recognized 10 nm peptide versus ≤ 1 nm peptide; Fig. 3).

TIL 1383 I may prove to be a useful reagent for addressing basic questions of T-cell development and immunological tolerance. We and others have shown that the transfer of low-affinity, MHC class I-restricted TCRs with apparent low affinity into cells that lack the CD8 coreceptor result in cells with low avidity (63, 64, 65). Coexpression of CD8 increased the avidity of the T cells, resulting in enhanced antigen recognition (64, 65). It is interesting to note that the reactivity of TIL 1383 I is similar to the reactivity of T9-368-reactive CD8+ T cells. In both cases, the T cells had higher avidity for the T9-368 370D peptide, which is expressed on the surface of melanoma cells than the T9-368 370N peptide, which is encoded by the tyrosinase gene (32). Therefore, the only apparent difference between T9-368-reactive CD8+ T cells and TIL 1383 I is the high-affinity TCR expressed by TIL 1383 I. Because high avidity T cells appear to be more effective in treating virus-infected mice than low avidity T cells (66), expressing this high-affinity TCR in normal CD8+ T cells should result in high avidity T cells that may be more effective in eliminating metastatic disease. Another possible benefit of this high-affinity TCR would be to transfer this MHC class I reactivity to normal CD4+ T cells. Given that TIL 1383 I recognized tumor cells without any contribution from the CD4 coreceptor, we would predict peripheral blood lymphocyte-derived CD4+ T cells bearing this TCR would also recognize HLA-A2+, tyrosinase+ melanoma cells. The ability of tumor-reactive CD4+ T cells to mediate tumor regression could be evaluated in a greater number of patients because HLA-A2 is expressed by ∼50% of all melanoma patients.

Fig. 1.

Cell surface phenotype of TIL 1383 I. The percentage of TIL 1383 I cells expressing CD3+, CD4+, CD8+, and BV12+ was determined by immunofluorescence staining, followed by flow cytometry analysis. Each histogram represents the fluorescence profiles of at least 5 × 103 viable cells.

Fig. 1.

Cell surface phenotype of TIL 1383 I. The percentage of TIL 1383 I cells expressing CD3+, CD4+, CD8+, and BV12+ was determined by immunofluorescence staining, followed by flow cytometry analysis. Each histogram represents the fluorescence profiles of at least 5 × 103 viable cells.

Close modal
Fig. 2.

Purification of BV12+ T cells from TIL 1383 I. A day 52 TIL 1383 I culture was stained with anti-BV12-FITC mAb and sorted for high and uniform expression of BV12+ T cells. Each histogram contains the fluorescence profiles of 104 live unstained (a), BV12 stained but not sorted (b), or BV12 stained and twice sorted cells (c).

Fig. 2.

Purification of BV12+ T cells from TIL 1383 I. A day 52 TIL 1383 I culture was stained with anti-BV12-FITC mAb and sorted for high and uniform expression of BV12+ T cells. Each histogram contains the fluorescence profiles of 104 live unstained (a), BV12 stained but not sorted (b), or BV12 stained and twice sorted cells (c).

Close modal
Fig. 3.

IFN-γ secretion by TIL 1383 I requires T9-368 peptide presented by HLA-A2 molecules. TIL 1383 I cells (a and b) or TIL 1520 cells (c) were stimulated with various concentrations of peptide alone (▪) or peptide-pulsed T2 cells (□. The peptide used to stimulate the TIL cells was T9-368 370D (a), T9-368 370N (b), and G9-209 (c).

Fig. 3.

IFN-γ secretion by TIL 1383 I requires T9-368 peptide presented by HLA-A2 molecules. TIL 1383 I cells (a and b) or TIL 1520 cells (c) were stimulated with various concentrations of peptide alone (▪) or peptide-pulsed T2 cells (□. The peptide used to stimulate the TIL cells was T9-368 370D (a), T9-368 370N (b), and G9-209 (c).

Close modal
Fig. 4.

Lysis of HLA-A2+ melanoma cells by TIL 1383 I. Lysis of melanoma and peptide-pulsed T2 cells by TIL 1383 I (a), TIL 888 (b), and TIL 1235 (c) was measured in standard 4-h 51Cr release assays. Targets included 1383 MEL (•), 1383 fibroblasts (×), 888 MEL (+), 888 A2 MEL (▪), T9-368 370D (♦), and M9-27 (▴) pulsed T2 cells.

Fig. 4.

Lysis of HLA-A2+ melanoma cells by TIL 1383 I. Lysis of melanoma and peptide-pulsed T2 cells by TIL 1383 I (a), TIL 888 (b), and TIL 1235 (c) was measured in standard 4-h 51Cr release assays. Targets included 1383 MEL (•), 1383 fibroblasts (×), 888 MEL (+), 888 A2 MEL (▪), T9-368 370D (♦), and M9-27 (▴) pulsed T2 cells.

Close modal

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

2

The abbreviations used are: TIL, tumor infiltrating lymphocyte; IL, interleukin; GM-CSF, granulocyte/macrophage-colony stimulating factor; TNF, tumor necrosis factor; CM, complete medium; TCR, T-cell receptor; PE, phycoerythrin; mAb, monoclonal antibody.

3

Unpublished observations.

Table 1

HLA-A2-restricted recognition of melanoma cell lines by TIL 1383 I

StimulatorsaHistologyHLA-A2NoneRespondersa (pg/ml IFN-γ release)
TIL 1235bTIL 1520bTIL 1383 I
None NA N/Ac 0.0 0.0 0.5 1.3 
397 MEL Melanoma − 0.0 2.1 0.7 0.3 
624-28 MEL Melanoma − 18.4 0.9 3.5 0.0 
888 MEL Melanoma − 0.0 1.7 1.3 3.2 
1011 MEL Melanoma − 0.0 0.0 0.3 0.0 
1402 MEL Melanoma − 0.0 0.0 0.0 10.2 
397 A2 MEL Melanoma 0.0 396.4              d 516.4 313.6 
624-38 MEL Melanoma 0.0 328.6 190.7 207.5 
888 A2 MEL Melanoma 0.0 892.0 1362.9 659.7 
1088 MEL Melanoma 0.0 476.4 723.8 450.0 
1199 MEL Melanoma 0.0 223.4 636.4 338.1 
1300 MEL Melanoma 49.7 398.1 913.5 359.3 
1363 MEL Melanoma 0.0 22.9 3.2 36.3 
1383 MEL Melanoma 0.0 144.2 879.1 314.4 
1558 MEL Melanoma 0.0 102.2 155.1 113.7 
SK23 Melanoma 0.0 381.9 780.3 368.2 
F002 MEL Melanoma 0.0 60.0 2.1 64.5 
1088 EBV B cell 0.0 1.1 5.6 0.0 
1363 EBV B cell 1.7 0.1 21.8 0.0 
1088 Fibro Fibroblast 1.7 3.2 2.1 5.8 
1383 Fibro Fibroblast 0.0 2.8 1.1 0.0 
1542 CP3TX Prostate − 0.0 0.0 0.0 0.0 
1550 CPTX Prostate 0.0 0.0 1.9 6.6 
SKOV3 Ovarian − 0.0 1.9 0.7 0.0 
SKOV3 A2 Ovarian 0.0 1.3 14.5 0.0 
UOK 181 Renal − 0.0 0.5 0.9 0.0 
UOK 131 Renal 0.0 0.9 0.0 0.0 
LOVO Colon − 1.3 0.3 0.5 0.0 
SW480 Colon 0.0 6.8 4.4 5.4 
HCE-4 Esophageal − 0.7 1.5 1.5 1.1 
SKGT5 Esophageal 0.0 4.6 10.7 0.0 
MDA-MB-435S Breast − 0.0 0.7 0.7 0.0 
MDA-MB-436 Breast − 0.0 1.5 2.3 3.9 
T47D Breast − 3.5 1.1 0.9 23.2 
MCF7 Breast 0.0 0.0 0.0 0.0 
MDA-MB-231 Breast 0.0 3.5 67.5 0.1 
SKBR3 Breast 0.0 0.0 0.0 0.0 
StimulatorsaHistologyHLA-A2NoneRespondersa (pg/ml IFN-γ release)
TIL 1235bTIL 1520bTIL 1383 I
None NA N/Ac 0.0 0.0 0.5 1.3 
397 MEL Melanoma − 0.0 2.1 0.7 0.3 
624-28 MEL Melanoma − 18.4 0.9 3.5 0.0 
888 MEL Melanoma − 0.0 1.7 1.3 3.2 
1011 MEL Melanoma − 0.0 0.0 0.3 0.0 
1402 MEL Melanoma − 0.0 0.0 0.0 10.2 
397 A2 MEL Melanoma 0.0 396.4              d 516.4 313.6 
624-38 MEL Melanoma 0.0 328.6 190.7 207.5 
888 A2 MEL Melanoma 0.0 892.0 1362.9 659.7 
1088 MEL Melanoma 0.0 476.4 723.8 450.0 
1199 MEL Melanoma 0.0 223.4 636.4 338.1 
1300 MEL Melanoma 49.7 398.1 913.5 359.3 
1363 MEL Melanoma 0.0 22.9 3.2 36.3 
1383 MEL Melanoma 0.0 144.2 879.1 314.4 
1558 MEL Melanoma 0.0 102.2 155.1 113.7 
SK23 Melanoma 0.0 381.9 780.3 368.2 
F002 MEL Melanoma 0.0 60.0 2.1 64.5 
1088 EBV B cell 0.0 1.1 5.6 0.0 
1363 EBV B cell 1.7 0.1 21.8 0.0 
1088 Fibro Fibroblast 1.7 3.2 2.1 5.8 
1383 Fibro Fibroblast 0.0 2.8 1.1 0.0 
1542 CP3TX Prostate − 0.0 0.0 0.0 0.0 
1550 CPTX Prostate 0.0 0.0 1.9 6.6 
SKOV3 Ovarian − 0.0 1.9 0.7 0.0 
SKOV3 A2 Ovarian 0.0 1.3 14.5 0.0 
UOK 181 Renal − 0.0 0.5 0.9 0.0 
UOK 131 Renal 0.0 0.9 0.0 0.0 
LOVO Colon − 1.3 0.3 0.5 0.0 
SW480 Colon 0.0 6.8 4.4 5.4 
HCE-4 Esophageal − 0.7 1.5 1.5 1.1 
SKGT5 Esophageal 0.0 4.6 10.7 0.0 
MDA-MB-435S Breast − 0.0 0.7 0.7 0.0 
MDA-MB-436 Breast − 0.0 1.5 2.3 3.9 
T47D Breast − 3.5 1.1 0.9 23.2 
MCF7 Breast 0.0 0.0 0.0 0.0 
MDA-MB-231 Breast 0.0 3.5 67.5 0.1 
SKBR3 Breast 0.0 0.0 0.0 0.0 
a

5 × 104 stimulators and 5 × 104 responders were cocultured in each well.

b

TIL 1235 is HLA-A2 restricted and MART-1 reactive, and TIL 1520 is HLA-A2 restricted and gp100 reactive.

c

NA, not applicable.

d

Underlined values are at least twice background and >100 pg/ml IFNC-γ released.

Table 2

HLA-A2-restricted recognition of tyrosinase by TIL 1383 I

StimulatorsaDNAbHLA-A2Respondersa (pg/ml IFN-γ released)
NoneTIL 1235cTIL 1520cTIL 1383 I
None None NAd 0.6 0.6 4.2 0.4 
COS None − 0.7 0.5 1.7 0.0 
COS A2 0.8 0.9 2.8 0.1 
COS A2 + MART-1 1.1 761.0              a 4.1 0.1 
COS A2 + gp100 0.9 1.0 1068.0 0.1 
COS A2 + TRP2 0.7 0.9 4.7 0.1 
COS A2 + Eso I 0.6 1.4 3.6 0.0 
COS A2 + tyrosinase 0.7 1.0 2.5 497.7 
1383 MEL None 0.5 41.7 701.2 1202.0 
624-28 MEL None − 1.7 1.5 4.3 0.1 
624-38 MEL None 0.5 141.8 706.9 903.5 
888 MEL None − 1.3 5.5 13.0 2.3 
888 A2 MEL None 0.5 749.6 1250.0 1433.6 
888 Fibro None 1.4 0.9 4.7 0.7 
1088 MEL None 1.2 870.3 1012.0 1137.6 
1088 Fibro None 0.4 0.3 6.6 0.4 
StimulatorsaDNAbHLA-A2Respondersa (pg/ml IFN-γ released)
NoneTIL 1235cTIL 1520cTIL 1383 I
None None NAd 0.6 0.6 4.2 0.4 
COS None − 0.7 0.5 1.7 0.0 
COS A2 0.8 0.9 2.8 0.1 
COS A2 + MART-1 1.1 761.0              a 4.1 0.1 
COS A2 + gp100 0.9 1.0 1068.0 0.1 
COS A2 + TRP2 0.7 0.9 4.7 0.1 
COS A2 + Eso I 0.6 1.4 3.6 0.0 
COS A2 + tyrosinase 0.7 1.0 2.5 497.7 
1383 MEL None 0.5 41.7 701.2 1202.0 
624-28 MEL None − 1.7 1.5 4.3 0.1 
624-38 MEL None 0.5 141.8 706.9 903.5 
888 MEL None − 1.3 5.5 13.0 2.3 
888 A2 MEL None 0.5 749.6 1250.0 1433.6 
888 Fibro None 1.4 0.9 4.7 0.7 
1088 MEL None 1.2 870.3 1012.0 1137.6 
1088 Fibro None 0.4 0.3 6.6 0.4 
a

5 × 104 stimulators and 5 × 104 responders were cocultured in each well.

b

COS cells were transfected with a total of 200 ng of plasmid DNA.

c

TIL 1235 is HLA-A2 restricted and MART-1 reactive, and TIL 1520 is HLA-A2 restricted and gp100 reactive.

d

NA, not applicable.

e

Underlined values were at least twice background and >100 pg/ml IFN-γ released.

Table 3

Recognition of the tyrosinase:368376 peptide by TIL 1383 I

StimulatorsaPeptideNoneRespondersa (pg/ml IFN-γ released)TIL 1383 I
TIL 1235bTIL 1520bTIL 620bTIL 888b
None None 0.0 2.0 2.6 2.3 33.4 0.9 
T2 M9-27 0.0 1504.8              c 13.9 227.3 25.8 3.0 
T2 G9-209 2.3 3.7 >2000.0 >2000.0 31.0 8.8 
T2 T9-1 0.6 3.0 15.2 3.3 23.5 6.3 
T2 T9-368 370D 0.9 4.0 17.2 0.9 23.8 >2000.0 
T2 T9-368 370N 0.9 6.2 34.7 4.4 24.6 >2000.0 
1383 MEL None 0.3 200.0 393.3 129.6 18.0 1474.1 
1383 Fibro None 0.0 2.6 3.3 0.3 22.3 3.7 
888 MEL None 3.0 7.0 5.3 8.6 890.9 15.4 
888 A2 MEL None 2.3 1840.6 >2000.0 >2000.0 738.1 >2000.0 
StimulatorsaPeptideNoneRespondersa (pg/ml IFN-γ released)TIL 1383 I
TIL 1235bTIL 1520bTIL 620bTIL 888b
None None 0.0 2.0 2.6 2.3 33.4 0.9 
T2 M9-27 0.0 1504.8              c 13.9 227.3 25.8 3.0 
T2 G9-209 2.3 3.7 >2000.0 >2000.0 31.0 8.8 
T2 T9-1 0.6 3.0 15.2 3.3 23.5 6.3 
T2 T9-368 370D 0.9 4.0 17.2 0.9 23.8 >2000.0 
T2 T9-368 370N 0.9 6.2 34.7 4.4 24.6 >2000.0 
1383 MEL None 0.3 200.0 393.3 129.6 18.0 1474.1 
1383 Fibro None 0.0 2.6 3.3 0.3 22.3 3.7 
888 MEL None 3.0 7.0 5.3 8.6 890.9 15.4 
888 A2 MEL None 2.3 1840.6 >2000.0 >2000.0 738.1 >2000.0 
a

2.5 × 104 stimulators and 2.5 × 104 responders were cocultured in each well.

b

TIL 1235 is HLA-A2 restricted and MART-1 reactive, TIL 1520 is HLA-A2 restricted and gp100 reactive, TIL 620 is HLA-A2 restricted and MART-1 and gp100 reactive, and TIL 888 is HLA-A24 restricted and tyrosinase reactive.

c

Underlined values are at least twice background and >100 pg/ml IFN-γ released.

Table 4

Recognition of the tyrosinase:368–376 peptide by BV12 sorted TIL 1383 I

StimulatorsaNoneRespondersa (pg/ml IFN-γ released)TIL 1383 I BV12 sortedc
TIL 1235bTIL 1520bTIL 1383 I unsorted
None 0.0 0.0 0.0 0.0 0.0 
T2 alone 0.0 0.0 0.0 0.0 0.0 
T2 + M9-27 0.0 178.0              d 25.1 0.0 0.0 
T2 + G9-209 0.0 0.0 186.5 0.0 0.0 
T2 + T9-368 370D 0.0 0.0 0.0 389.4 650.8 
StimulatorsaNoneRespondersa (pg/ml IFN-γ released)TIL 1383 I BV12 sortedc
TIL 1235bTIL 1520bTIL 1383 I unsorted
None 0.0 0.0 0.0 0.0 0.0 
T2 alone 0.0 0.0 0.0 0.0 0.0 
T2 + M9-27 0.0 178.0              d 25.1 0.0 0.0 
T2 + G9-209 0.0 0.0 186.5 0.0 0.0 
T2 + T9-368 370D 0.0 0.0 0.0 389.4 650.8 
a

104 stimulators and 104 responders were cocultured in each well.

b

TIL 1235 is HLA-A2 restricted and MART-1 reactive, and TIL 1520 is HLA-A2 restricted and gp100 reactive.

c

Cells were sorted for BV12 expression and were 100% CD4+ and >99.9% BV12+.

d

Underlined values were at least twice background and >100 pg/ml IFN-γ released.

Table 5

Antibody inhibition of IFN-γ release by TIL 1383 I

RespondersaStimulatorsaIFN-γ released (pg/ml) in the presence of mAb
A.MediumW6/32bIVA12bW6/32 + IVA12b
TIL 1383I 1383 MEL 184.8 120.4 (35%)c 167.3 (9%) 95.4 (48%) 
TIL 138I 1558 MEL 220.7 119.1 (46%) 234.2 (0%) 95.2 (57%) 
TIL 1520d 1383 MEL 444.1 120.7 (73%) 658.0 (0%) 117.1 (74%) 
TIL 1520 1558 MEL 398.6 84.7 (79%) 275.7 (31%) 72.9 (82%) 
TIL 1558 1383 MEL 37.2 30.7 36.3 28.5 
TIL 1558 1558 MEL 640.7 571.9 (11%) 317.5 (50%) 225.6 (65%) 
RespondersaStimulatorsaIFN-γ released (pg/ml) in the presence of mAb
A.MediumW6/32bIVA12bW6/32 + IVA12b
TIL 1383I 1383 MEL 184.8 120.4 (35%)c 167.3 (9%) 95.4 (48%) 
TIL 138I 1558 MEL 220.7 119.1 (46%) 234.2 (0%) 95.2 (57%) 
TIL 1520d 1383 MEL 444.1 120.7 (73%) 658.0 (0%) 117.1 (74%) 
TIL 1520 1558 MEL 398.6 84.7 (79%) 275.7 (31%) 72.9 (82%) 
TIL 1558 1383 MEL 37.2 30.7 36.3 28.5 
TIL 1558 1558 MEL 640.7 571.9 (11%) 317.5 (50%) 225.6 (65%) 
B.MediumW6/32bIVA12b
TIL 1383I T2 + M9-27 29.9 49.8 35.3 
TIL 1383I T2 + T9-368 370D 1146.9 451.7 (61%) 955.7 (17%) 
TIL 1235d T2 + M9-27 978.9 117.3 (88%) 897.9 (8%) 
TIL 1235 T2 + T9-368 370D 8.8 0.0 0.0 
B.MediumW6/32bIVA12b
TIL 1383I T2 + M9-27 29.9 49.8 35.3 
TIL 1383I T2 + T9-368 370D 1146.9 451.7 (61%) 955.7 (17%) 
TIL 1235d T2 + M9-27 978.9 117.3 (88%) 897.9 (8%) 
TIL 1235 T2 + T9-368 370D 8.8 0.0 0.0 
C.MediumAnti-CD4eAnti-CD8e
TIL 1383I 1558 MEL 705.4 604.9 (15%) 546.5 (23%) 
TIL 1520 1558 MEL 507.2 489.5 (4%) 276.0 (46%) 
TIL 1558d 1558 MEL 1651.0 930.0 (44%) 1363.0 (17%) 
C.MediumAnti-CD4eAnti-CD8e
TIL 1383I 1558 MEL 705.4 604.9 (15%) 546.5 (23%) 
TIL 1520 1558 MEL 507.2 489.5 (4%) 276.0 (46%) 
TIL 1558d 1558 MEL 1651.0 930.0 (44%) 1363.0 (17%) 
a

104 stimulators and 104 responders were cocultured in each well.

b

Stimulator cells were preincubated with W6/32 and IVA12 mAb (final concentration, 20 μg/ml) prior to the addition of T cells.

c

Values in parentheses were percentage of inhibition relative to medium alone.

d

TIL 1235 is HLA-A2 restricted and MART-1 reactive, TIL 1520 is HLA-A2 restricted and gp100 reactive, and TIL 1558 reacted only with its autologous 1558 MEL.

e

Responder cells were preincubated with anti-CD4 (final concentration, 2.5 μg/ml) and anti-CD8 (final concentration, 1.25 μg/ml) mAbs prior to the addition of stimulator cells.

Table 6

Cytokine release profile of TIL 1383 I

RespondersaStimulatorsCytokine releaseda (pg/ml)
IFN-γGM-CSFTNF-αIL-2IL-4IL-10
None None 0.0 1.1 0.0 0.0 0.0 0.0 
None OKT3 7.3 0.0 0.0 0.0 0.0 0.0 
None 1383 MEL 12.0 0.0 0.0 0.0 0.1 0.0 
None 1383 Fibro 1.4 47.0 614.2              b 0.0 0.0 0.0 
None 1558 MEL 9.4 0.0 0.0 0.0 0.0 0.0 
TIL 1520c None 18.7 0.0 0.0 0.0 0.3 0.0 
TIL 1520 OKT3 1821.2 842.1 364.0 0.0 0.5 0.0 
TIL 1520 1383 MEL 517.8 165.4 47.8 0.0 0.0 0.0 
TIL 1520 1383 Fibro 9.0 87.2 0.0 0.0 0.0 0.0 
TIL 1520 1558 MEL 640.9 216.1 43.3 0.0 0.0 0.0 
TIL 1383I None 4.4 61.8 0.0 0.0 0.0 0.0 
TIL 1383I OKT3 1327.4 1522.9 651.2 196.1 4.2 0.0 
TIL 1383I 1383 MEL >2000.0 1586.8 1324.4 >2000.0 13.5 13.1 
TIL 1383I 1383 Fibro 5.3 272.4 0.2 0.0 0.0 0.0 
TIL 1383I 1558 MEL 1926.2 1422.5 558.4 874.5 7.5 0.0 
RespondersaStimulatorsCytokine releaseda (pg/ml)
IFN-γGM-CSFTNF-αIL-2IL-4IL-10
None None 0.0 1.1 0.0 0.0 0.0 0.0 
None OKT3 7.3 0.0 0.0 0.0 0.0 0.0 
None 1383 MEL 12.0 0.0 0.0 0.0 0.1 0.0 
None 1383 Fibro 1.4 47.0 614.2              b 0.0 0.0 0.0 
None 1558 MEL 9.4 0.0 0.0 0.0 0.0 0.0 
TIL 1520c None 18.7 0.0 0.0 0.0 0.3 0.0 
TIL 1520 OKT3 1821.2 842.1 364.0 0.0 0.5 0.0 
TIL 1520 1383 MEL 517.8 165.4 47.8 0.0 0.0 0.0 
TIL 1520 1383 Fibro 9.0 87.2 0.0 0.0 0.0 0.0 
TIL 1520 1558 MEL 640.9 216.1 43.3 0.0 0.0 0.0 
TIL 1383I None 4.4 61.8 0.0 0.0 0.0 0.0 
TIL 1383I OKT3 1327.4 1522.9 651.2 196.1 4.2 0.0 
TIL 1383I 1383 MEL >2000.0 1586.8 1324.4 >2000.0 13.5 13.1 
TIL 1383I 1383 Fibro 5.3 272.4 0.2 0.0 0.0 0.0 
TIL 1383I 1558 MEL 1926.2 1422.5 558.4 874.5 7.5 0.0 
a

5 × 104 stimulators and 5 × 104 responders were cocultured in each well.

b

Underlined values are at least twice background and >100 pg/ml IFN-γ released.

c

TIL 1520 is HLA-A2 restricted and gp100 reactive.

We thank Drs. John R. Wunderlich, Suzanne L. Topalian, Timothy Clay, and Alfred Singer for thoughtful suggestions and critical reading of the manuscript. We also thank Pat Koen and Arnold Mixon for valuable assistance with flow cytometry.

1
Rothenberg E. V. The development of functionally responsive T cells.
Adv. Immunol.
,
51
:
85
-214,  
1992
.
2
Aguila H. L., Akashi K., Domen J., Gandy K. L., Lagasse E., Mebius R. E., Morrison S. J., Shizuru J., Strober S., Uchida N., Wright D. E., Weissman I. L. From stem cells to lymphocytes: biology and transplantation.
Immunol. Rev.
,
157
:
13
-40,  
1997
.
3
Zinkernagel R. M., Doherty P. C. The discovery of MHC restriction.
Immunol. Today
,
18
:
14
-17,  
1997
.
4
Sprent J., Schaefer M., Lo D., Korngold R. Functions of purified L3T4+ and Lyt-2+ cells in vitro and in vivo.
Immunol. Rev.
,
91
:
195
-,  
1986
.
5
Swain S. L. T cell subsets and the recognition of MHC class.
Immunol. Rev.
,
74
:
129
-142,  
1983
.
6
Wucherpfennig K. W., Strominger J. L. Molecular mimicry in T cell-mediated autoimmunity: viral peptides activate human T cell clones specific for myelin basic protein.
Cell
,
80
:
695
-705,  
1995
.
7
Oldstone M. B. Molecular mimicry as a mechanism for the cause and a probe uncovering etiologic agent(s) of autoimmune disease.
Curr. Top. Microbiol. Immunol.
,
145
:
127
-135,  
1989
.
8
Ashton-Rickardt P. G., Van Kaer L., Schumacher T. N., Ploegh H. L., Tonegawa S. Peptide contributes to the specificity of positive selection of CD8+ T cells in the thymus.
Cell
,
73
:
1041
-1049,  
1993
.
9
Hogquist K. A., Bevan M. J. The nature of the peptide/MHC ligand involved in positive selection.
Semin. Immunol.
,
8
:
63
-68,  
1996
.
10
Hogquist K. A., Gavin M. A., Bevan M. J. Positive selection of CD8+ T cells induced by major histocompatibility complex binding peptides in fetal thymic organ culture.
J. Exp. Med.
,
177
:
1469
-1473,  
1993
.
11
Wilson D. B., Blyth J. L., Nowell P. C. Quantitative studies on the mixed lymphocyte interaction in rats. III. Kinetics of the response.
J. Exp. Med.
,
128
:
1157
-1181,  
1968
.
12
Loftus D. J., Castelli C., Clay T. M., Squarcina P., Marincola F. M., Nishimura M. I., Parmiani G., Appella E., Rivoltini L. Identification of epitope mimics recognized by CTL reactive to the melanoma/melanocyte-derived peptide MART-1(27–35).
J. Exp. Med.
,
184
:
647
-657,  
1996
.
13
Hiemstra H. S., van Veelen P. A., Schloot N. C., Geluk A., van Meijgaarden K. E., Willemen S. J., Leunissen J. A., Benckhuijsen W. E., Amons R., de Vries R. R., Roep B. O., Ottenhoff T. H., Drijfhout J. W. Definition of natural T cell antigens with mimicry epitopes obtained from dedicated synthetic peptide libraries.
J. Immunol.
,
161
:
4078
-4082,  
1998
.
14
Evavold B. D., Sloan-Lancaster J., Wilson K. J., Rothbard J. B., Allen P. M. Specific T cell recognition of minimally homologous peptides: evidence for multiple endogenous ligands.
Immunity
,
2
:
655
-663,  
1995
.
15
Topalian S. L., Solomon D., Rosenberg S. A. Tumor-specific cytolysis by lymphocytes infiltrating human melanomas.
J. Immunol.
,
142
:
3714
-3725,  
1989
.
16
Topalian S. L., Rivoltini L., Mancini M., Ng J., Hartzman R. J., Rosenberg S. A. Melanoma-specific CD4+ T lymphocytes recognize human melanoma antigens processed and presented by Epstein-Barr virus-transformed B cells.
Int. J. Cancer
,
58
:
69
-79,  
1994
.
17
Bright R. K., Vocke C. D., Emmert-Buck M. R., Duray P. H., Solomon D., Fetsch P., Rhim J. S., Linehan W. M., Topalian S. L. Generation and genetic characterization of immortal human prostate epithelial cell lines derived from primary cancer specimens.
Cancer Res.
,
57
:
995
-1002,  
1997
.
18
Anglard P., Trahan E., Liu S., Latif F., Merino M. J., Lerman M. I., Zbar B., Linehan W. M. Molecular and cellular characterization of human renal cell carcinoma cell lines.
Cancer Res.
,
52
:
348
-356,  
1992
.
19
Schrump D. S., Chen G. A., Consuli U., Jin X., Roth J. A. Inhibition of esophageal cancer proliferation by adenovirally mediated delivery of p16INK4.
Cancer Gene Ther.
,
3
:
357
-364,  
1996
.
20
Rivoltini L., Barracchini K. C., Viggiano V., Kawakami Y., Smith A., Mixon A., Restifo N. P., Topalian S. L., Simonis T. B., Rosenberg S. A. Quantitative correlation between HLA class I allele expression and recognition of melanoma cells by antigen-specific cytotoxic T lymphocytes.
Cancer Res.
,
55
:
3149
-3157,  
1995
.
21
Kawakami Y., Eliyahu S., Delgado C. H., Robbins P. F., Rivoltini L., Topalian S. L., Miki T., Rosenberg S. A. Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor.
Proc. Natl. Acad. Sci. USA
,
91
:
3515
-3519,  
1994
.
22
Kawakami Y., Eliyahu S., Sakaguchi K., Robbins P. F., Rivoltini L., Yannelli J. R., Appella E., Rosenberg S. A. Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes.
J. Exp. Med.
,
180
:
347
-352,  
1994
.
23
Clay T. M., Custer M. C., McKee M. D., Parkhurst M., Robbins P. F., Kerstann K., Wunderlich J., Rosenberg S. A., Nishimura M. I. Changes in the fine specificity of gp100 (209–217) reactive T cells in patients following vaccination with a peptide modified at an HLA-A2.1 anchor residue.
J. Immunol.
,
162
:
1749
-1755,  
1999
.
24
Robbins P. F., El-Gamil M., Kawakami Y., Stevens E., Yannelli J. R., Rosenberg S. A. Recognition of tyrosinase by tumor-infiltrating lymphocytes from a patient responding to immunotherapy.
Cancer Res.
,
54
:
3124
-3126,  
1994
.
25
Pieper R., Christian R. E., Gonzales M. I., Nishimura M. I., Gupta G., Settlage R. E., Shabanowita J., Rosenberg S. A., Hunt D. F., Topalian S. L. Biochemical identification of a mutated human melanoma antigen recognized by CD4+ T cells.
J. Exp. Med.
,
189
:
757
-765,  
1998
.
26
Kawakami Y., Eliyahu S., Delgado C. H., Robbins P. F., Sakaguchi K., Appella E., Yannelli J. R., Adema G. J., Miki T., Rosenberg S. A. Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated with in vivo tumor rejection.
Proc. Natl. Acad. Sci. USA
,
91
:
6458
-6464,  
1994
.
27
Wang R. F., Appella E., Kawakami Y., Kang X., Rosenberg S. A. Identification of TRP-2 as a human tumor antigen recognized by cytotoxic T lymphocytes.
J. Exp. Med.
,
184
:
2207
-2216,  
1996
.
28
Wang R. F., Johnston S. L., Zeng G., Topalian S. L., Schwartzentruber D. J., Rosenberg S. A. A breast and melanoma-shared tumor antigen: T cell responses to antigenic peptides translated from different open reading frames.
J. Immunol.
,
161
:
3598
-3606,  
1998
.
29
Parkhurst M. R., Salgaller M. L., Southwood S., Robbins P. F., Sette A., Rosenberg S. A., Kawakami Y. Improved induction of melanoma-reactive CTL with peptides from the melanoma antigen gp100 modified at HLA-A*0201-binding residues.
J. Immunol.
,
157
:
2539
-2548,  
1996
.
30
Kawakami Y., Eliyahu S., Jennings C., Sakaguchi K., Kang X., Southwood S., Robbins P. F., Sette A., Appella E., Rosenberg S. A. Recognition of multiple epitopes in the human melanoma antigen gp100 by tumor-infiltrating T lymphocytes associated with in vivo tumor regression.
J. Immunol.
,
154
:
3961
-3968,  
1995
.
31
Wolfel T., Van Pel A., Brichard V., Schneider J., Seliger B., Meyer zum Buschenfelde K. H., Boon T. Two tyrosinase nonapeptides recognized on HLA-A2 melanomas by autologous cytolytic T lymphocytes.
Eur. J. Immunol.
,
24
:
759
-764,  
1994
.
32
Skipper J. C., Hendrickson R. C., Gulden P. H., Brichard V., Van Pel A., Chen Y., Shabanowitz J., Wolfel T., Slingluff C. L., Boon T., Hunt D. F., Engelhard V. H. An HLA-A2-restricted tyrosinase antigen on melanoma cells results from posttranslational modification and suggests a novel pathway for processing of membrane proteins.
J. Exp. Med.
,
183
:
527
-534,  
1996
.
33
del Guercio M. F., Sidney J., Hermanson G., Perez C., Grey H. M., Kubo R. T., Sette A. Binding of a peptide antigen to multiple HLA alleles allows definition of an A2-like supertype.
J. Immunol.
,
154
:
685
-693,  
1992
.
34
Nishimura M. I., Kawakami Y., Charmley P., O’Neil B., Shilyansky J., Yannelli J. R., Rosenberg S. A., Hood L. T-cell receptor repertoire in tumor-infiltrating lymphocytes. Analysis of melanoma-specific long-term lines.
J. Immunother.
,
16
:
85
-94,  
1994
.
35
Rowen L., Koop B. F., Hood L. The complete 685-kilobase DNA sequence of the human beta T cell receptor locus.
Science (Washington DC)
,
272
:
1755
-1762,  
1996
.
36
WHO Nomenclature for T-cell receptor (TCR) gene segments of the immune system. WHO-IUIS Nomenclature Sub-Committee on TCR Designation.
Immunogenetics
,
42
:
451
-453,  
1995
.
37
Deveraux J., Haeberli P., Smithies O. A comprehensive set of sequence analysis programs for the VAX.
Nucleic Acids Res.
,
12
:
387
-395,  
1984
.
38
Schwartzentruber D. J., Topalian S. L., Mancini M., Rosenberg S. A. Specific release of granulocyte-macrophage colony-stimulating factor, tumor necrosis factor-α, and IFN-γ by human tumor- infiltrating lymphocytes after autologous tumor stimulation.
J. Immunol.
,
146
:
3674
-3681,  
1991
.
39
Robbins P. F., El-Gamil M., Li Y. F., Topalian S. L., Rivoltini L., Sakaguchi K., Appella E., Kawakami Y., Rosenberg S. A. Cloning of a new gene encoding an antigen recognized by melanoma- specific HLA-A24-restricted tumor-infiltrating lymphocytes.
J. Immunol.
,
154
:
5944
-5950,  
1995
.
40
Darrow T. L., Abdel-Wahab Z., Quinn-Allen M. A., Seigler H. F. Recognition and lysis of human melanoma by a CD3+, CD4+, CD8-T-cell clone restricted by HLA-A2.
Cell. Immunol.
,
172
:
52
-59,  
1996
.
41
Jacob L., Somasundaram R., Smith W., Monos D., Basak S., Marincola F., Pereira S., Herlyn D. Cytotoxic T-cell clone against rectal carcinoma induced by stimulation of a patient’s peripheral blood mononuclear cells with autologous cultured tumor cells.
Int. J. Cancer
,
71
:
325
-332,  
1997
.
42
LeMay L. G., Kan-Mitchell J., Goedegebuure P., Harel W., Mitchell M. S. Detection of melanoma-reactive CD4+ HLA-class I-restricted cytotoxic T cell clones with long-term assay and pretreatment of targets with interferon-gamma.
Cancer Immunol. Immunother.
,
37
:
187
-194,  
1993
.
43
Kharkevitch D. D., Seito D., Balch G. C., Maeda T., Balch C. M., Itoh K. Characterization of autologous tumor-specific T-helper 2 cells in tumor-infiltrating lymphocytes from a patient with metastatic melanoma.
Int. J. Cancer
,
58
:
317
-323,  
1994
.
44
Hayashi Y., Hoon D. S., Park M. S., Terasaki P. I., Foshag L. J., Morton D. L. Induction of CD4+ cytotoxic T cells by sensitization with allogeneic melanomas bearing shared or cross-reactive HLA-A.
Cell. Immunol.
,
139
:
411
-425,  
1992
.
45
Wang P., Vanky F., Klein E. MHC class-I-restricted auto-tumor-specific CD4+CD8 T-cell clones established from autologous mixed lymphocyte-tumor-cell culture (MLTC).
Int. J. Cancer
,
51
:
962
-967,  
1992
.
46
Meadows L., Wang W., den Haan J. M., Blokland E., Reinhardus C., Drijfhout J. W., Shabanowitz J., Pierce R., Agulnik A. I., Bishop C. E., Hunt D. F., Goulmy E., Engelhard V. H. The HLA-A*0201-restricted H-Y antigen contains a posttranslationally modified cysteine that significantly affects T cell recognition.
Immunity
,
6
:
273
-281,  
1997
.
47
Fink P. J., Bevan M. J. H-2 antigens of the thymus determine lymphocyte specificity.
J. Exp. Med.
,
148
:
766
-775,  
1978
.
48
Ashton-Rickardt P. G., Van Kaer L., Schumacher T. N. M., Ploegh H. L., Tonegawa S. Repertoire-determining role of peptide in the positive selection of CD8+ T cells.
Immunol. Rev.
,
135
:
157
-182,  
1993
.
49
Bouchard B., Fuller B. B., Vijayasaradhi S., Houghton A. N. Induction of pigmentation in mouse fibroblasts by expression of human tyrosinase cDNA.
J. Exp. Med.
,
169
:
2029
-2042,  
1989
.
50
Sensi M., Parmiani G. Analysis of TCR usage in human tumors: a new tool for assessing tumor-specific immune responses.
Immunol. Today
,
16
:
588
-595,  
1995
.
51
Shilyansky J., Nishimura M. I., Yannelli J. R., Kawakami Y., Jacknin L. S., Charmley P., Rosenberg S. A. T-cell receptor usage by melanoma-specific clonal and highly oligoclonal tumor-infiltrating lymphocyte lines.
Proc. Natl. Acad. Sci. USA
,
91
:
2829
-2833,  
1994
.
52
Cole D. J., Weil D. P., Shamamian P., Rivoltini L., Kawakami Y., Topalian S., Jennings C., Eliyahu S., Rosenberg S. A., Nishimura M. I. Identification of MART-1-specific T-cell receptors: T cells utilizing distinct T-cell receptor variable and joining regions recognize the same tumor epitope.
Cancer Res.
,
54
:
5265
-5268,  
1994
.
53
Shinohara N., Sachs D. H. Mouse alloantibodies capable of blocking cytotoxic T-cell function. I. Relationship between the antigen reactive with blocking antibodies and the Lyt-2 locus.
J. Exp. Med.
,
150
:
432
-444,  
1979
.
54
Robey E., Ramsdell F., Elliott J., Raulet D., Kioussis D., Axel R., Fowlkes B. J. Expression of CD4 in transgenic mice alters the specificity of CD8 cells for allogeneic major histocompatibility complex.
Proc. Natl. Acad. Sci. USA
,
88
:
608
-612,  
1991
.
55
Cammarota G., Scheirle A., Takacs B., Doran D. M., Knorr R., Bannwarth W., Guardiola J., Sinigaglia F. Identification of a CD4 binding site on the β2 domain of HLA-DR molecules.
Nature (Lond.)
,
356
:
799
-801,  
1992
.
56
Salter R. D., Benjamin R. J., Wesley P. K., Buxton S. E., Garrett T. P., Clayberger C., Krensky A. M., Norment A. M., Littman D. R., Parham P. A binding site for the T-cell co-receptor CD8 on the α3 domain of HLA-A2.
Nature (Lond.)
,
345
:
41
-46,  
1990
.
57
Garcia K. C., Scott C. A., Brunmark A., Carbone F. R., Peterson P. A., Wilson I. A., Teyton L. CD8 enhances formation of stable T-cell receptor/MHC class I molecule complexes.
Nature (Lond.)
,
384
:
577
-581,  
1996
.
58
Doyle C., Strominger J. L. Interaction between CD4 and class II MHC molecules mediates cell adhesion.
Nature (Lond.)
,
330
:
256
-259,  
1987
.
59
Veillette A., Bookman M. A., Horak E. M., Bolen J. B. The CD4 and CD8 T cell surface antigens are associated with the internal membrane tyrosine-protein kinase p56lck.
Cell
,
55
:
301
-308,  
1988
.
60
MacDonald H. R., Glasebrook A. L., Acuto O., Kelso A., Bron C., Cerottini J. C. Heterogeneity of inhibition of cytolytic T lymphocyte clones by monoclonal anti-Lyt-2/3 antibodies: parallel effects on cytolysis, proliferation and lymphokine secretion.
Adv. Exp. Med. Biol.
,
146
:
533
-545,  
1982
.
61
Malissen B., Rebai N., Liabeuf A., Mawas C. Human cytotoxic T cell structures associated with expression of cytolysis. I. Analysis at the clonal cell level of the cytolysis-inhibiting effect of 7 monoclonal antibodies.
Eur. J. Immunol.
,
12
:
739
-747,  
1982
.
62
Shimonkevitz R., Luescher B., Cerottini J. C., MacDonald H. R. Clonal analysis of cytolytic T lymphocyte-mediated lysis of target cells with inducible antigen expression: correlation between antigen density and requirement for Lyt-2/3 function.
J. Immunol.
,
135
:
892
-899,  
1985
.
63
Cole D. J., Weil D. P., Shilyansky J., Custer M., Kawakami Y., Rosenberg S. A., Nishimura M. I. Characterization of the functional specificity of a cloned T-cell receptor heterodimer recognizing the MART-1 melanoma antigen.
Cancer Res.
,
55
:
748
-752,  
1995
.
64
Gabert J., Langlet C., Zamoyska R., Parnes J. R., Schmitt-Verhulst A. M., Malissen B. Reconstitution of MHC class I specificity by transfer of the T cell receptor and Lyt-2 genes.
Cell
,
50
:
545
-554,  
1987
.
65
Clay T. M., Custer M. C., Sachs J. A., Rosenberg S. A., Nishimura M. I. Efficient transfer of a tumor antigen-reactive TcR to human peripheral blood lymphocytes confers anti-tumor reactivity.
J. Immunol.
,
163
:
507
-513,  
1999
.
66
Alexander-Miller M. A., Leggatt G. R., Berzofsky J. A. Selective expansion of high- or low-avidity cytotoxic T lymphocytes and efficacy for adoptive immunotherapy.
Proc. Natl. Acad. Sci. USA
,
93
:
4102
-4107,  
1996
.