Cytotoxic T-lymphocyte (CTL) responses can be generated against peptides derived from the immunoglobulin (Ig) V region in some but not all patients. The main reason for this appears to be the low peptide-binding affinity of Ig-derived peptides to major histocompatibility complex (MHC) class I molecules and their resulting low immunogenicity. This might be improved by conservative amino acid modifications at the MHC-binding residues of the peptides (heteroclitic peptides). In this study, it was found that in 18 Ig-derived peptides, that heteroclitic peptides from the Ig gene with improved binding to human leukocyte antigen (HLA)-A*0201 can be used to improve CTL responses. Amino acid substitution substantially increased predicted binding affinity, and there was a strong correlation between predicted and actual binding to HLA-A*0201. CTLs generated against the heteroclitic peptide had not only enhanced cytotoxicity against the heteroclitic peptide but also increased killing of antigen-presenting cells pulsed with the native peptide. Surprisingly, no difference was observed in the frequency of T cells detected by MHC class I peptide tetramers after stimulation with the heteroclitic peptide compared with the native peptide. CTLs generated against heteroclitic peptides could kill patients' tumor cells, showing that Ig-derived peptides can be presented by the tumor cell and that the failure to mount an immune response (among other reasons) likely results from the low immunogenicity of the native Ig-derived peptide. These results suggest that heteroclitic Ig-derived peptides can enhance immunogenicity, thereby eliciting immune responses, and that they might be useful tools for enhancing immunotherapy approaches to treating B-cell malignant diseases.

Most B-cell malignant diseases are characterized by clonal expansion of a single B cell. The B-cell–antigen receptor, the immunoglobulin (Ig), is a clonal marker containing tumor-specific epitopes, or idiotypes, that can function as targets for T-cell–mediated immune responses.1-4 In murine lymphoma models, humoral and cellular mechanisms against idiotypes are effective in inducing tumor regression in tumor-bearing hosts.4-8 In clinical trials, vaccination with patient-specific Ig has shown efficacy, with predominantly humoral immune responses reported.9,10 Several studies indicated that vaccination with idiotype elicits CD4+ and CD8+ responses in vitro and in vivo.11-14 However, analysis of major histocompatibility complex (MHC) class I–restricted CD8+T-cell responses in the patients did not find the Ig-derived epitopes recognized by tumor-specific T cells.15,16 Previously, we and others showed that CD8+ T-cell responses against Ig-derived peptides can be generated and that such cells are capable of killing the primary tumor cells from which the Ig was derived.17,18 

CD8+ T lymphocytes recognize small peptides (9-10 amino acids in length) presented on the surface of antigen-presenting cells (APCs) by MHC class I molecules. Several tumor antigens have been identified on the basis of gene cloning or sequencing of the naturally presented peptide eluted from MHC class I molecules.19-21MHC class I–restricted antigenic peptides have the potential to be candidates for tumor vaccines and have been tested in animal models and humans. However, results obtained with such peptide vaccines have been variable.22 Successful immunization was obtained after vaccination with peptides derived from a mutated connexin 37 gap–junction protein expressed on murine carcinoma, with regression of established metastases.23 Protective immune responses were obtained against viruses after peptide vaccination in one study24 but, in other models, peptide vaccines had limited or negative results.25-27 

One reason for the limited immunogenicity of some tumor-derived peptides is weak binding of the peptide to the MHC molecule. The requirements for peptides to bind to human leukocyte antigen (HLA) molecules and to elicit cytotoxic T-lymphocyte (CTL) responses have been studied extensively.28-32 The strength of CTL responses depends on the binding affinity of the target peptide to HLA class I, the stability of the peptide–HLA complex, and the avidity of T-cell receptor (TCR) binding for the peptide complex.33-35 Most virus-derived immunodominant peptides have high binding affinity or peptide–HLA complex stability.33 Peptides with such characteristics can be predicted on the basis of well-established binding motifs for the most common HLA alleles.29,32 This technique has been applied to the identification of CTL epitopes derived from melanoma antigen (MAGE) 3,36 proteinase 3,37 and telomerase.38 However, using this approach in a study with Ig V region–derived peptides, we found that only 14 of 794 peptides showed strong binding to HLA-A*0201, the most common HLA class I allele. More than 90% of the analyzed peptides showed absent or only weak MHC binding.17 Therefore, one likely reason for the weakness of CTL responses against the Ig of the tumor cell is the low immunogenicity of Ig-derived MHC class I–restricted peptides.

One strategy to increase MHC binding and immunogenicity of low-binding peptides is to modify the peptide at the MHC-binding amino acid residues while leaving the T-cell–recognition residues intact.39 The resultant “heteroclitic” peptides were previously shown to lead to improved induction of CTL responses against melanoma in murine models40 and trials in humans.41 We here report that heteroclitic peptides from the Ig V region with improved binding to HLA-A*0201 can be used to induce a CTL response. Importantly, heteroclitic peptides elicited T-cell responses against not only the altered peptide but also against the native peptide from which they were derived, and they also increased the efficacy of killing of primary leukemic cells. This strategy might not only increase the pool of peptides that can be used to generate T-cell responses against B-cell malignant diseases but may also be used to increase the efficiency of tumor-cell killing.

Patients and healthy donors

Peripheral blood and tumor samples were collected from healthy donors and patients at Dana-Farber Cancer Institute and cryopreserved. Ig gene rearrangements from tumor cells were sequenced from patients with a variety of B-cell malignant diseases as previously described.17 All tumor samples used to test killing were obtained from patients with previously untreated chronic lymphocytic leukemia (CLL). All specimens were obtained after informed consent and approval by our institutional review board were provided.

Peptide-prediction analysis and bioinformatics

Ig-deduced protein sequences were reviewed for peptides 9 and 10 amino acids long that could possibly bind to MHC class I molecules. Two independent computer prediction analyses were used (http://bimas.dcrt.nih.gov/molbio/hla_bind andhttp://134.2.96.221/Scripts/MHCServer.dll/EpPredict.htm) to examine the binding of native peptides to HLA-A*0201 molecules. The analysis used HLA-A*0201 because this MHC class I allele is expressed in approximately 50% of our patients. Heteroclitic peptides were designed in which conservative amino acid substitutions of the MHC-binding residues were expected to enhance the affinity toward the MHC class I allele, as predicted by using the computer algorithms.

Peptide synthesis

All native and heteroclitic peptides were generated by fluorenylmethoxycarbonyl synthesis (Sigma-Genosys Biotechnologies, The Woodlands, Texas). Purity was determined by reverse-phase high-performance liquid chromatography and verified by mass spectral analysis. The peptides that were synthesized and assayed are shown in Table 1. Peptides were dissolved in phosphate-buffered saline at a concentration of 2 mg/mL and stored at −70°C until use. A peptide derived from MAGE 3 was used as a positive control.

Table 1.

Immunoglobulin-derived synthetic native and heteroclitic peptides

NamePositionSequenceParker scoreRammensee scoreFICTLs induced/
donors tested
FR9 FR3 TLFLQMNSL 181 24 0.2 3/6 
FR9H  KLFLQMNSL 636 25 0.2 1/1 
FR13 FR2 CVLLCEIWV 123 16 Not available Not tested 
FR13H  KVLLCEIWV 1935 23 0.6 2/2 
FR15 FR3 NQFSLKLSSV 60 17 0.9 1/6 
FR15H  NQFSLKLSSV 591 27 2.0 1/3 
FR16 FR3 SLQPEDFAT 43 19 1.3 3/7 
FR16H κ SLQPEDFAV 403 25 2.8 2/4 
FR17 FR3 YLQMNSLRA 22 17 0.3 2/7 
FR17H  YLQMNSLRV 320 23 1.8 3/3 
FR18 FR2 QAPGKGLEWV 20 0.4 0/5 
FR18H  QLPGKGLEWV 431 26 1.3 2/2 
FR21 FR2 APGKGLEWV 18 0.1 2/6 
FR21H  ALGKGLEWV 431 28 2.7 2/3 
FR23 FR3 STAYMELSSL 23 0.6 2/6 
FR23H  SLAYMELSSL 49 29 1.4 2/3 
FR24 FR2 FPGKGLVWV 26 19 0.2 0/8 
FR24H  FLGKGLVWV 4047 29 2.3 1/4 
FR25 FR3 AVYLQMNSL 14 20 0.1 0/8 
FR25H  AVYLQMNSV 512 26 2.4 1/4 
FR26 FR3 TLYLQMDNL 77 21 0.6 0/5 
FR26H  KLYLQMDNV 878 22 1.3 2/4 
FR27 FR3 SVIAADTAV 19 0.1 0/2 
FR27H  KVIAADTAV 243 24 0.9 1/2 
FR28 FR3 NQFTLKLTSV 60 17 0.5 0/2 
FR28H  NLFTLKLTSV 2071 28 0/2 
FR29 FR3 NQFSLKLSSV 60 17 0.3 0/2 
FR29H  KLFSLKLSSV 2071 28 0.8 0/2 
FR30 FR3 TLYLQMNSL 157 24 0.1 0/2 
FR30H  KLYLQMNSV 1792 25 0.5 1/2 
FR31 FR3 AVYYCARDLV 10 16 0.7 0/2 
FR31H  KVYYCARDLV 382 21 0.09 1/2 
CDR16 CDRI IVSDNCMSWV 537 17 0.5 1/5 
CDR16H  ILSDNCMSWV 6132 23 1.8 1/4 
CDR17 CDRII KGLEWISGYI 17 0.6 0/2 
CDR17H  KLLEWISGYV 6072 29 1.3 1/2 
MAGE aa 271 FLWGPRALV 2655 27 2.6 12/12 
NamePositionSequenceParker scoreRammensee scoreFICTLs induced/
donors tested
FR9 FR3 TLFLQMNSL 181 24 0.2 3/6 
FR9H  KLFLQMNSL 636 25 0.2 1/1 
FR13 FR2 CVLLCEIWV 123 16 Not available Not tested 
FR13H  KVLLCEIWV 1935 23 0.6 2/2 
FR15 FR3 NQFSLKLSSV 60 17 0.9 1/6 
FR15H  NQFSLKLSSV 591 27 2.0 1/3 
FR16 FR3 SLQPEDFAT 43 19 1.3 3/7 
FR16H κ SLQPEDFAV 403 25 2.8 2/4 
FR17 FR3 YLQMNSLRA 22 17 0.3 2/7 
FR17H  YLQMNSLRV 320 23 1.8 3/3 
FR18 FR2 QAPGKGLEWV 20 0.4 0/5 
FR18H  QLPGKGLEWV 431 26 1.3 2/2 
FR21 FR2 APGKGLEWV 18 0.1 2/6 
FR21H  ALGKGLEWV 431 28 2.7 2/3 
FR23 FR3 STAYMELSSL 23 0.6 2/6 
FR23H  SLAYMELSSL 49 29 1.4 2/3 
FR24 FR2 FPGKGLVWV 26 19 0.2 0/8 
FR24H  FLGKGLVWV 4047 29 2.3 1/4 
FR25 FR3 AVYLQMNSL 14 20 0.1 0/8 
FR25H  AVYLQMNSV 512 26 2.4 1/4 
FR26 FR3 TLYLQMDNL 77 21 0.6 0/5 
FR26H  KLYLQMDNV 878 22 1.3 2/4 
FR27 FR3 SVIAADTAV 19 0.1 0/2 
FR27H  KVIAADTAV 243 24 0.9 1/2 
FR28 FR3 NQFTLKLTSV 60 17 0.5 0/2 
FR28H  NLFTLKLTSV 2071 28 0/2 
FR29 FR3 NQFSLKLSSV 60 17 0.3 0/2 
FR29H  KLFSLKLSSV 2071 28 0.8 0/2 
FR30 FR3 TLYLQMNSL 157 24 0.1 0/2 
FR30H  KLYLQMNSV 1792 25 0.5 1/2 
FR31 FR3 AVYYCARDLV 10 16 0.7 0/2 
FR31H  KVYYCARDLV 382 21 0.09 1/2 
CDR16 CDRI IVSDNCMSWV 537 17 0.5 1/5 
CDR16H  ILSDNCMSWV 6132 23 1.8 1/4 
CDR17 CDRII KGLEWISGYI 17 0.6 0/2 
CDR17H  KLLEWISGYV 6072 29 1.3 1/2 
MAGE aa 271 FLWGPRALV 2655 27 2.6 12/12 

Shown are the name, position, sequence (single-letter abbreviations for amino acids) with heteroclitic changes (in boldface and underlined), calculated score of predicted half-life (Parker et al29 and Rammensee et al31) in arbitrary units, fluorescence index (FI) of binding to human HLA-A*0201, and number of cytotoxic T-lymphocyte (CTL) lines generated from healthy donors that elicited specific killing of peptide-pulsed, CD40-activated B cells for all peptides tested.

FR indicates framework region; CDR, complementarity-determining region; and MAGE, melanoma antigen.

Selection of heteroclitic peptides

Eighteen Ig-derived peptides from patients with B-cell malignant diseases were chosen on the basis of low binding to HLA-A*0201 or low frequency of generation of CTL lines generated against APCs pulsed with these peptides.17 The native and heteroclitic versions of the peptides were synthesized. One native peptide (framework region [FR] 13) could not be synthesized because of its high hydrophobicity; therefore, 35 Ig V region–derived peptides were tested. Sixteen of the peptides were derived from the FRs, and 2 were from the complementarity-determining regions (CDRs). All peptides were screened with the computer prediction analysis for their binding affinity.

To generate the modified or heteroclitic peptides, single and double amino acid substitutions were made at the HLA A*0201 binding motif or anchor residues. Alterations were made to amino acids at position 1 or 2 (or both) and 9 or 10 (substitution with lysine at position 1, leucine at position 2, and valine at position 9 or 10), with the intention of not altering the T-cell–recognition region of the peptide. Heteroclitic peptides were selected for synthesis when the substitutions most markedly increased computer scores for predicted binding compared with the native peptides, with the intention of increasing binding affinity most efficiently (Table 1).

Evaluation of binding affinity of Ig-derived (native and heteroclitic) peptides to HLA-A*020

To determine whether predicted peptides could bind to HLA-A2, we used a cellular peptide-binding assay employing the transporter associated with antigen-processing–deficient cell line T2 (American Type Culture Collection, Manassas, VA). For this and subsequent analyses, a total of 17 peptides and 18 modified peptides (Table 1) were synthesized. T2 cells were pulsed with 50 μg/mL peptide and 5 μg/mL β-2 microglobulin (Sigma, St Louis, MO) for 18 hours at 37°C. HLA-A*0201 expression was then measured by using monoclonal antibody (mAb) BB7.2 (ATCC) followed by incubation with fluorescein isothiocyanate (FITC)–conjugated F(ab′)2goat antimouse Ig (Zymed, San Francisco, CA). Fluorescence index (FI) was calculated as the mean fluorescence intensity (MFI) of HLA-A*0201 on T2 cells as determined by fluorescence-activated cell-sorting (FACS) analysis, according to the following formula: FI = (MFI [T2 cells + peptide]/MFI [T2 cells without peptide]) −1.

Screening for peptide-specific HLA-restricted CTLs

For screening peptide-specific CTL responses, we used a previously established system.17,38 Briefly, purified HLA-A2–positive CD8+ T cells from healthy donors were primarily stimulated with irradiated monocyte-derived, peptide-pulsed dendritic cells and restimulated weekly with irradiated, peptide-pulsed, CD40-activated B cells.42 After 4 stimulations, more than 90% of cells in each culture were CD3+ lymphocytes, of which more than 90% were CD8+ and 0.6% to 5.4% were CD4+. Cultures were less than 0.6% CD14+ and 1.9% to 4.6% CD56+. The cytotoxic activity of CTL lines was assessed after 4 to 5 stimulations by using peptide-pulsed and unpulsed CD40-activated B cells or malignant B cells in chromium 51 (51Cr)–release assays. The concentration of peptide used for pulsing the CD-40 activated B cells resulted in maximal killing of the targets. When CLL cells were used as targets, the B-cell population was obtained by negative depletion using anti–T cell, natural killer cell, and monocyte mAbs and magnetic bead depletion. In each case, more than 95% of resulting cells coexpressed CD19 and CD5. Specific lysis was calculated as the percentage of specific 51Cr release by using the following formula: (E − S/T − S) × 100, where E is experimental 51Cr release, S is the spontaneous51Cr release, and T is the total 51Cr release by 2% Triton X-100. The purity of cell populations was determined by dual-color FACS using directly conjugated mAbs against CD3, CD4, CD8, CD19, CD20, CD56, and CD14 (Dako, Carpinteria, CA). Results for specific killing were assessed for each T-cell line generated and were compared with those observed with autologous T-cell lines generated against unpulsed APCs. MHC restriction of lytic activity was tested against HLA-A2–negative tumors or by blockade of HLA-A2 using mAb BB7.2 and the isotype control mAb B5.

Tetramer synthesis and staining

Purified HLA-A*0201 peptide monomeric complexes were obtained from ProImmune (Oxford, United Kingdom). To obtain tetramers, streptavidin-phycoerythrin (PE) conjugate (Sigma) was added at a 1:4 molar ratio. Tetramers were assembled for the native peptides FR16, FR17, and FR23, as well as their heteroclitic counterparts. CD8+ T-cell lines were generated as described above. T cells were stained with PE-labeled tetramer and CD8 FITC-labeled mAb (Dako) for 30 minutes on ice. After washing, stained cells were analyzed by flow cytometry (Beckman Coulter, Miami, FL).

Generation of native and modified Ig-derived peptides

We previously showed that CTLs can be generated against peptides derived from clonal Ig in B-cell malignancies in a peptide-specific and MHC-restricted way.17 A limitation of this approach was the low binding affinity of most peptides from the clonal Ig sequences: it was not always possible to generate CTLs, especially against these low-affinity peptides. We therefore examined whether alteration to the MHC-binding sequences would increase the binding affinity of altered Ig-derived peptides and whether CTLs could then be generated against these altered peptides that would also have specificity for the native peptide. Therefore, we assessed Ig-derived peptides from 70 patients with B-cell malignant diseases in whom the Ig region had been sequenced and who expressed HLA-A*0201. The peptides chosen for further examination had previously been found by us to have low binding to HLA-A*0201 and a low frequency of generation of CTL lines generated against APCs pulsed with these peptides.17 A surprising finding of this study was that the CDR3 region contained few peptides with predicted binding potential and it was difficult to find peptides from this region for which the native peptide would likely be presented by the tumor cell as a target for T cells generated against the heteroclitic peptides.

Seventeen native peptides (one native FR-derived peptide could not be synthesized because of its hydrophobicity) and 18 heteroclitic peptides were included in this study. Their sequences and positions in the Ig region are shown in Table 1. Sixteen of these sequences were derived from the FRs. As was previously reported,17 many of these FR-derived peptides are shared by multiple patients and are therefore not unique tumor-associated antigens. Two peptides were derived from the CDRs. One FR-derived peptide (FR16) was from the κ light chain; all other peptides were from the Ig heavy-chain V region. Eleven peptides were nonamer and 7 were decamer oligopeptides.

The predicted score for binding of the native peptides was examined by using 2 separate computer algorithms (Table 1). Alterations to the MHC-binding peptides were examined in these algorithms to increase predicted binding to HLA-A*0201. These modifications consisted of substitutions of lysine at position 1, leucine at position 2, or valine at position 9. The sequences of both the native and heteroclitic peptides are shown in Table 1 with the amino acid substitutions in boldface and underlined. The predicted binding for the heteroclitic peptides was increased in all cases except FR9, for which the predicted binding increased in one algorithm but not the other.

Binding affinity of native and modified Ig-derived peptides

We examined whether the heteroclitic peptides had increased affinity to HLA-A*0201 by using the T2 binding assay. The FI for the native and heteroclitic peptides was calculated. Representative results demonstrating the binding of the native and heteroclitic FR17 peptides are shown in Figure 1, and results obtained for all peptides tested are shown in Table 1. We observed a significant increase in binding of the heteroclitic peptides compared with the native peptide (P < .0001 on paired ttesting). The FI did not increase in only one case (FR9), and the predicted binding affinity was also not increased in one of the computer algorithms for this peptide.

Fig. 1.

T2 binding assay.

Expression of MHC class I molecule on T2 cells without peptide after addition of the native peptide FR17 (FI = 0.6) and enhanced expression after addition of the heteroclitic peptide FR17H (FI = 1.3).

Fig. 1.

T2 binding assay.

Expression of MHC class I molecule on T2 cells without peptide after addition of the native peptide FR17 (FI = 0.6) and enhanced expression after addition of the heteroclitic peptide FR17H (FI = 1.3).

Close modal

Peptide-specific CTL reactivity against targets pulsed with heteroclitic peptides

We next attempted to generate T-cell lines against the native and heteroclitic peptides and determine whether the resulting T cells were capable of inducing specific lysis of autologous peptide-pulsed APCs. Representative results are shown in Figure2 for T-cell lines generated against native and heteroclitic peptide CDR16. Figure 2A shows results for the T-cell line generated against the native CDR16 peptide, which induced only weak killing of target cells pulsed with either native or heteroclitic peptide. Figure 2B shows results obtained by using a T-cell line generated against the heteroclitic peptide CDR16H. For 8 of the peptide pairs for which no cytotoxicity was observed with any of the T-cell lines generated against the native peptide, T-cell lines could be generated against the heteroclitic peptide (Table 1). In other cases, we were able to generate T-cell lines more readily and show more specific lysis with the lines generated against the heteroclitic peptide. Table 1 shows results obtained for all donors tested and the number of donors in whom CTLs with specificity for the peptide-pulsed, CD40-activated B cells (measured by chromium-release assays) could be generated. CTLs could not be generated against either the native or heteroclitic peptide in only 2 cases (FR28 and FR29).

Fig. 2.

Specificity of killing of CD40-activated B cells pulsed with native and heteroclitic peptides.

(A) Killing by T-cell lines generated against native peptide CDR16 of unpulsed CD40-activated B cells and CD40-activated B cells pulsed with heteroclitic peptide, native peptide, or control peptide (FR9) at the effector-to-target ratios shown. (B) Killing by T-cell lines generated against heteroclitic peptide CDR16H of unpulsed CD40-activated B cells and CD40-activated B cells pulsed with heteroclitic peptide, native peptide, or control peptide (FR9) at effector-to-target ratios shown. Results are mean ± SD values from experiments done in triplicate with samples from a single donor and are representative of results from experiments done with samples from 5 different donors.

Fig. 2.

Specificity of killing of CD40-activated B cells pulsed with native and heteroclitic peptides.

(A) Killing by T-cell lines generated against native peptide CDR16 of unpulsed CD40-activated B cells and CD40-activated B cells pulsed with heteroclitic peptide, native peptide, or control peptide (FR9) at the effector-to-target ratios shown. (B) Killing by T-cell lines generated against heteroclitic peptide CDR16H of unpulsed CD40-activated B cells and CD40-activated B cells pulsed with heteroclitic peptide, native peptide, or control peptide (FR9) at effector-to-target ratios shown. Results are mean ± SD values from experiments done in triplicate with samples from a single donor and are representative of results from experiments done with samples from 5 different donors.

Close modal

Results pertaining to specific lysis of CD40-actived B cells pulsed with the native compared with the heteroclitic peptide by autologous T-cell lines generated against each of the remaining peptides are shown in Figure 3. The findings represent killing of the peptide-pulsed, CD40-activated B cells subtracted from that of the unpulsed CD40-activated B cells. In all cases, killing of the unpulsed CD40-activated B cells was 5% or less. Although there was a correlation between predicted binding, FI, and ability to induce CTL responses, this was not present in all cases (Table 1). Thus, the algorithms are still incomplete and it is necessary to use a functional assay, such as the one described here, to confirm that changes result in improved binding and generation of CTL responses.

Fig. 3.

Killing of CD40-activated B cells pulsed with native and heteroclitic peptides.

Cytotoxicity of T-cell lines generated against native and heteroclitic peptides, with killing of CD40-activated autologous B cells pulsed with the peptide at an effector-to-target ratio of 30:1. Results are mean ± SD values from experiments done in triplicate with samples from up to 8 different donors.

Fig. 3.

Killing of CD40-activated B cells pulsed with native and heteroclitic peptides.

Cytotoxicity of T-cell lines generated against native and heteroclitic peptides, with killing of CD40-activated autologous B cells pulsed with the peptide at an effector-to-target ratio of 30:1. Results are mean ± SD values from experiments done in triplicate with samples from up to 8 different donors.

Close modal

Heteroclitic CTLs lyse target cells bearing the native peptide

Our most important goal was to determine whether T-cell lines generated against the heteroclitic peptide were also capable of killing cells expressing the native peptide that would be expressed by the tumor cell. To examine this question, we generated T-cell lines against the native and heteroclitic peptides and examined killing by these T-cell lines of CD40-activated B cells pulsed with the original native peptide. In 7 of the 14 matched-pair analyses done, T-cell lines generated against the heteroclitic peptide were also able to kill the CD40-activated B cells pulsed with the native peptide. In all cases, killing with the T-cell line generated against the heteroclitic peptide was greater than that by T-cell lines generated against the native peptide itself (Figure 2B and Figure 4). It was not surprising that, in some cases, the T-cell lines were not able to kill the CD40-activated B cells pulsed with the native peptide, since the binding affinity of the native peptide for the MHC class I molecule was low. Under these circumstances, it is also likely that the native peptide would not be processed and presented by the tumor cell with sufficient efficiency to present a target of an immune response.

Fig. 4.

T cells generated against heteroclitic peptide kill targets pulsed with native peptide.

Killing with T-cell lines generated against native or heteroclitic peptide of CD40-activated autologous B cells pulsed with native peptide at an effector-to-target ratio of 30:1. Results are mean ± SD values from experiments done in triplicate with samples from 3 different donors.

Fig. 4.

T cells generated against heteroclitic peptide kill targets pulsed with native peptide.

Killing with T-cell lines generated against native or heteroclitic peptide of CD40-activated autologous B cells pulsed with native peptide at an effector-to-target ratio of 30:1. Results are mean ± SD values from experiments done in triplicate with samples from 3 different donors.

Close modal

Peptide-specific CTLs can kill CLL cells

To examine whether T-cell lines generated against the heteroclitic peptide could kill not only CD40-activated normal B cells pulsed with the native peptide but also the tumor cell from which the Ig sequence was obtained, cytotoxicity assays were done by using the primary tumor cells as targets. All tumor samples tested as targets of cytotoxicity were obtained from patients with CLL who had not previously been treated. In each case, non-B cells were depleted by negative depletion, and more than 95% of the resulting cells were CLL cells as indicated by coexpression of CD19 and CD5. In 7 cases (Figure5A), we found that T-cell lines generated against the heteroclitic peptide were more efficient at killing CLL cells than were T-cell lines generated against the native peptide. This finding shows that low-affinity peptides that are ineffective in generating an immune response can still be targets of a T-cell–mediated immune response generated against an altered peptide. Efficient killing required a high ratio of effector-to-target (Figure5B).

Fig. 5.

Killing of CLL cells by T-cell lines generated against native and heteroclitic peptides.

(A) Killing of CLL cells with a T-cell line generated against native or heteroclitic peptide with rearranged Ig gene containing the sequence for the FR9, FR16, FR17, FR18, FR23, FR26, and CDR17 peptides. Results shown are at an effector-to-target ratio of 30:1 and are the mean ± SD values from experiments done in triplicate with samples from 3 different donors. (B) Killing of CLL cells by a T-cell line generated from autologous T cells against FR17 native and heteroclitic peptides at the effector-to-target ratios shown. (C) Killing of CLL cells occurs in an MHC-restricted manner and is inhibited by addition of anti–HLA-A2 mAb at a concentration of 5 μg/mL.

Fig. 5.

Killing of CLL cells by T-cell lines generated against native and heteroclitic peptides.

(A) Killing of CLL cells with a T-cell line generated against native or heteroclitic peptide with rearranged Ig gene containing the sequence for the FR9, FR16, FR17, FR18, FR23, FR26, and CDR17 peptides. Results shown are at an effector-to-target ratio of 30:1 and are the mean ± SD values from experiments done in triplicate with samples from 3 different donors. (B) Killing of CLL cells by a T-cell line generated from autologous T cells against FR17 native and heteroclitic peptides at the effector-to-target ratios shown. (C) Killing of CLL cells occurs in an MHC-restricted manner and is inhibited by addition of anti–HLA-A2 mAb at a concentration of 5 μg/mL.

Close modal

All results reported here are for killing of nonactivated CLL cells. However, we have also observed more efficient killing of CD40-activated CLL cells compared with nonactivated tumor cells.17In addition, we found increased killing of tumor cells when they were pulsed with exogenous peptide,17 suggesting that mechanisms that lead to increased antigen presentation might make these tumor cells more susceptible to CTL-mediated killing. Killing occurred in an MHC class I–specific manner and was inhibited by addition of anti–HLA-2 mAb (Figure 5C).

Enumeration of antigen-specific T cells by using HLA class I tetramers against native and heteroclitic peptides

After 4 stimulations of the T-cell lines with peptide-pulsed autologous APCs, more than 90% of the cells were CD8+ T cells, for each of the peptides studied. Staining with the HLA-A*0201 peptide tetramers for the native peptides FR16, FR17, and FR23, as well as their heteroclitic counterparts, was done for T-cell lines generated from healthy HLA-A*0201 donors against the native peptide and heteroclitic counterparts. Results are shown in Table2. We observed no peptide-specific T cells in any of the samples from healthy donors. After 4 to 5 stimulations using APCs pulsed with the native or heteroclitic peptides, specific T cells were observed in each of the resulting T-cell lines. In each case, T cells generated against the heteroclitic peptide were also detected by the tetramer for the native peptide and vice versa, demonstrating that the T-cell–recognition region of the peptide was likely not altered by the amino acid substitutions at the anchor residues. In one donor, we observed the expected higher frequency of specific cells after stimulation with the heteroclitic peptide (17.3%) compared with that observed in the donor after stimulation with the native peptide (1.46%). However, surprisingly, overall we observed no difference in the frequency of T cells observed after stimulation for the heteroclitic peptide compared with the native peptide (P = .3).

Table 2.

Tetramer staining of T-cell lines generated against native and heteroclitic peptides

TetramerHealthy
donors
T cells generated
against native
peptide
T cells generated
against heteroclitic
peptide
Negative control 0.04 0.30 0.06  
FR16 0.02 1.57 1.38 
FR16H 0.06 2.34 9.96  
Negative control 0.06 0.14 0.03  
FR17 0.04 2.07 2.61 
FR17H 0.03 3.52 2.79  
Negative control 0.02 0.04 0.04  
FR23 0.02 1.19 2.46 
FR23H 0.02 1.95 2.13 
TetramerHealthy
donors
T cells generated
against native
peptide
T cells generated
against heteroclitic
peptide
Negative control 0.04 0.30 0.06  
FR16 0.02 1.57 1.38 
FR16H 0.06 2.34 9.96  
Negative control 0.06 0.14 0.03  
FR17 0.04 2.07 2.61 
FR17H 0.03 3.52 2.79  
Negative control 0.02 0.04 0.04  
FR23 0.02 1.19 2.46 
FR23H 0.02 1.95 2.13 

Shown is enumeration of peptide-specific T cells by tetramer staining of T cells from healthy donors and of T-cell lines generated against the framework region (FR) 16, FR17, and FR23 native and heteroclitic peptides. Shown is the frequency of CD8+ T cells generated against FR16, FR17, and FR23 and their heteroclitic counterparts that stain with specific tetramer, the heteroclitic tetramer, and a negative control tetramer.

The idiotype is the best-characterized tumor antigen expressed in B-cell malignant diseases. It can function as a target for T-cell–mediated1-4 and humoral immune responses.4-10 However, in many cases, these responses do not lead to a sustained clinical response. Furthermore, technical limitations in synthesizing the full Ig protein and the need to produce an individual molecule for each patient have hampered more widespread use of this approach. In addition, only a small number of Ig-derived peptides appear to have the ability to induce a specific CD8+ CTL response.17 In an attempt to overcome this limitation and increase the pool of immunogenic Ig-derived peptides, we generated peptides with conservative modifications of the HLA-binding residues of the peptides. We examined this approach in 18 peptides with weak binding to HLA-A*0201. The resulting heteroclitic peptides showed improved binding to MHC class I molecules and improved killing of peptide-pulsed target cells. T-cell lines generated against the heteroclitic peptide killed not only target cells pulsed with that peptide but also showed increased killing of target cells pulsed with the native peptide. Most important, these T-cell lines also killed primary tumor cells that presumably express only the native peptide.

Sixteen of the 18 peptides we examined were derived from the FR. We reasoned that there was no advantage in raising T cells against heteroclitic peptides if the native peptide had extremely low binding affinities and would therefore be unlikely to be presented by the tumor cell itself. The CDR3 contained very few peptides with predicted binding potential. It is possible that there might be a selection bias against rearrangements that lead to expression of peptides from the CDRs with high binding affinity, since the B cells would be depleted by T-cell responses against such determinants. A possible advantage of FR-derived peptides is that they are not limited to a single patient. We previously found that some peptides examined are expressed in up to 20% of patients with B-cell malignant diseases.17 Indeed, 34 of the 70 patients examined shared at least one peptide. A major concern about use of this approach is some normal B cells will also express targeted FR-derived peptides. Clinical trials with the anti-CD20 antibody rituximab in which virtually all B cells are eliminated have not revealed severe side effects caused by B-cell deficiency.43,44 However, there are clearly differences between induction of an active immune response with a vaccination approach and loss of B cells after passive administration of an antibody. An ongoing active immune response against the FR could lead to loss of all normal B cells expressing that peptide sequence and this could theoretically result in a defect in the B-cell repertoire. Ongoing studies are addressing this issue in murine models in vivo.

We observed a difference between killing of malignant B cells and normal B cells. This could represent differential killing of malignant compared with normal B cells by changes in processing of Ig-derived peptides. However, we believe that it more likely indicates that normal B cells that share the FR peptides would be killed; however, because most normal B cells do not express that particular FR peptide, the observed killing did not increase above background levels. In addition, although the FR peptides are shared by malignant and normal B cells, we did not observe detectable T cells with specificity for these peptides in samples from any healthy donor on MHC class I–peptide tetramer staining. The mechanisms by which normal activated B cells, which are effective APCs, do not elicit such responses are not known. Certainly, T cells capable of reacting against these peptides exist in the repertoire and can be expanded in vitro by using the system described here. In vivo, tolerance might be induced against these FRs, and it is possible that the in vitro system we used activates cross-reacting T cells. Indeed, the relatively low specific lysis observed in vitro might be due to the fact that tolerance must be overcome.

Heteroclitic peptides were shown to improve induction of CTL responses against MAGEs in both murine models40 and trials in humans.41 Introduction of conservative modifications at particular amino acid positions in the peptide resulted in an increase in low HLA-A*0201-binding affinity and enhanced recruitment of T-cell repertoire against peptides with nondominant anchor residues.28,31,45,46 In the current study, use of heteroclitic peptides enhanced binding and cytotoxicity. The finding that the frequency of specific T cells was not enhanced against heteroclitic compared with native peptides was therefore surprising. The observation that CTLs could be enumerated after immunization with even weak Ig-derived peptides is encouraging, however, but suggests that the frequency of T cells generated might not be as important as the avidity of the TCR of the resulting T cells for the target peptide and the ability of the tumor cell to present the target peptide.

Our experiments show that by changing amino acids of anchor residues, HLA-peptide binding can be improved, as predicted by computer algorithms and confirmed by T2 binding. CTLs raised against heteroclitic peptides can kill target cells bearing the native peptide, and lysis of primary CLL cells can be improved by generating T cells against heteroclitic peptides. We therefore conclude that MHC class I–restricted heteroclitic peptides can enlarge the pool of immunogenic peptides in B-cell malignant diseases and enhance the immunogenicity of these peptides. Whether these findings have implications for immunotherapy approaches in these diseases—for use of heteroclitic peptides as vaccines or incorporation of changes in the structure of the whole Ig constructs to enhance MHC class I binding of Ig-derived peptides and enhance cytotoxicity—will be clarified by the results of ongoing in vivo murine experiments to determine whether such increased cytotoxicity also depletes the normal B-cell repertoire.

Supported by grants CA78378 and CA81534 from the National Cancer Institute to J.G.G. and by the Grace B. Kerr Foundation. M.W. and A.M.K. were supported by grants from the Deutsche Krebshilfe and Dr Mildred Scheel Stiftung.

S.H. and M.W. contributed equally to this study.

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
Janeway
CA
Jr
Sakato
N
Eisen
HN
Recognition of immunoglobulin idiotypes by thymus-derived lymphocytes.
Proc Natl Acad Sci U S A.
72
1975
2357
2360
2
Bogen
B
Malissen
B
Haas
W
Idiotope-specific T cell clones that recognize syngeneic immunoglobulin fragments in the context of class II molecules.
Eur J Immunol.
16
1986
1373
1378
3
Campbell
MJ
Carroll
W
Kon
S
et al
Idiotype vaccination against murine B cell lymphoma: humoral and cellular responses elicited by tumor-derived immunoglobulin M and its molecular subunits.
J Immunol.
139
1987
2825
2833
4
George
AJ
Tutt
AL
Stevenson
FK
Anti-idiotypic mechanisms involved in suppression of a mouse B cell lymphoma, BCL1.
J Immunol.
138
1987
628
634
5
Lauritzsen
GF
Weiss
S
Bogen
B
Anti-tumour activity of idiotype-specific, MHC-restricted Th1 and Th2 clones in vitro and in vivo.
Scand J Immunol.
37
1993
77
85
6
Syrengelas
AD
Chen
TT
Levy
R
DNA immunization induces protective immunity against B-cell lymphoma.
Nat Med.
2
1996
1038
1041
7
King
CA
Spellerberg
MB
Zhu
D
et al
DNA vaccines with single-chain Fv fused to fragment C of tetanus toxin induce protective immunity against lymphoma and myeloma.
Nat Med.
4
1998
1281
1286
8
Tani
K
Murphy
WJ
Chertov
O
et al
Defensins act as potent adjuvants that promote cellular and humoral immune responses in mice to a lymphoma idiotype and carrier antigens.
Int Immunol.
12
2000
691
700
9
Kwak
LW
Campbell
MJ
Czerwinski
DK
Hart
S
Miller
RA
Levy
R
Induction of immune responses in patients with B-cell lymphoma against the surface-immunoglobulin idiotype expressed by their tumors.
N Engl J Med.
327
1992
1209
1215
10
Hsu
FJ
Caspar
CB
Czerwinski
D
et al
Tumor-specific idiotype vaccines in the treatment of patients with B-cell lymphoma—long-term results of a clinical trial.
Blood.
89
1997
3129
3135
11
Wen
YJ
Lim
SH
T cells recognize the VH complementarity-determining region 3 of the idiotypic protein of B cell non-Hodgkin's lymphoma.
Eur J Immunol.
27
1997
1043
1047
12
Wen
YJ
Ling
M
Lim
SH
Immunogenicity and cross-reactivity with idiotypic IgA of VH CDR3 peptide in multiple myeloma.
Br J Haematol.
100
1998
464
468
13
Wen
YJ
Ling
M
Bailey-Wood
R
Lim
SH
Idiotypic protein-pulsed adherent peripheral blood mononuclear cell-derived dendritic cells prime immune system in multiple myeloma.
Clin Cancer Res.
4
1998
957
962
14
Fagerberg
J
Yi
Q
Gigliotti
D
et al
T-cell-epitope mapping of the idiotypic monoclonal IgG heavy and light chains in multiple myeloma.
Int J Cancer.
80
1999
671
680
15
Bendandi
M
Gocke
CD
Kobrin
CB
et al
Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma.
Nat Med.
5
1999
1171
1177
16
Osterroth
F
Garbe
A
Fisch
P
Veelken
H
Stimulation of cytotoxic T cells against idiotype immunoglobulin of malignant lymphoma with protein-pulsed or idiotype-transduced dendritic cells.
Blood.
95
2000
1342
1349
17
Trojan
A
Schultze
JL
Witzens
M
et al
Immunoglobulin framework-derived peptides function as cytotoxic T-cell epitopes commonly expressed in B-cell malignancies.
Nat Med.
6
2000
667
672
18
Wen
YJ
Barlogie
B
Yi
Q
Idiotype-specific cytotoxic T lymphocytes in multiple myeloma: evidence for their capacity to lyse autologous primary tumor cells.
Blood.
97
2001
1750
1755
19
van der Bruggen
P
Traversari
C
Chomez
P
et al
A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma.
Science.
254
1991
1643
1647
20
Traversari
C
van der Bruggen
P
Luescher
IF
et al
A nonapeptide encoded by human gene MAGE-1 is recognized on HLA-A1 by cytolytic T lymphocytes directed against tumor antigen MZ2-E.
J Exp Med.
176
1992
1453
1457
21
Huang
AY
Gulden
PH
Woods
AS
et al
The immunodominant major histocompatibility complex class I-restricted antigen of a murine colon tumor derives from an endogenous retroviral gene product.
Proc Natl Acad Sci U S A.
93
1996
9730
9735
22
Melief
CJ
Offringa
R
Toes
RE
Kast
WM
Peptide-based cancer vaccines.
Curr Opin Immunol.
8
1996
651
657
23
Mandelboim
O
Vadai
E
Fridkin
M
et al
Regression of established murine carcinoma metastases following vaccination with tumour-associated antigen peptides.
Nat Med.
1
1995
1179
1183
24
Schulz
M
Zinkernagel
RM
Hengartner
H
Peptide-induced antiviral protection by cytotoxic T cells.
Proc Natl Acad Sci U S A.
88
1991
991
993
25
Marchand
M
Weynants
P
Rankin
E
et al
Tumor regression responses in melanoma patients treated with a peptide encoded by gene MAGE-3 [letter].
Int J Cancer.
63
1995
883
885
26
Jaeger
E
Bernhard
H
Romero
P
et al
Generation of cytotoxic T-cell responses with synthetic melanoma-associated peptides in vivo: implications for tumor vaccines with melanoma-associated antigens.
Int J Cancer.
66
1996
162
169
27
Hu
X
Chakraborty
NG
Sporn
JR
Kurtzman
SH
Ergin
MT
Mukherji
B
Enhancement of cytolytic T lymphocyte precursor frequency in melanoma patients following immunization with the MAGE-1 peptide loaded antigen presenting cell-based vaccine.
Cancer Res.
56
1996
2479
2483
28
Parker
KC
Bednarek
MA
Hull
LK
et al
Sequence motifs important for peptide binding to the human MHC class I molecule, HLA-A2.
J Immunol.
149
1992
3580
3587
29
Parker
KC
Bednarek
MA
Coligan
JE
Scheme for ranking potential HLA-A2 binding peptides based on independent binding of individual peptide side chains.
J Immunol.
152
1994
163
175
30
Rammensee
HG
Falk
K
Rotzschke
O
Peptides naturally presented by MHC class I molecules.
Annu Rev Immunol.
11
1993
213
244
31
Rammensee
HG
Friede
T
Stevanoviic
S
MHC ligands and peptide motifs: first listing.
Immunogenetics.
41
1995
178
228
32
Rammensee
H
Bachmann
J
Emmerich
NP
Bachor
OA
Stevanovic
S
SYFPEITHI: database for MHC ligands and peptide motifs.
Immunogenetics.
50
1999
213
219
33
Sette
A
Vitiello
A
Reherman
B
et al
The relationship between class I binding affinity and immunogenicity of potential cytotoxic T cell epitopes.
J Immunol.
153
1994
5586
5592
34
Van der Burg
SH
Visseren
MJ
Brandt
RM
Kast
WM
Melief
CJ
Immunogenicity of peptides bound to MHC class I molecules depends on the MHC-peptide complex stability.
J Immunol.
156
1996
3308
3314
35
Savage
PA
Boniface
JJ
Davis
MM
A kinetic basis for T cell receptor repertoire selection during an immune response.
Immunity.
10
1999
485
492
36
Nijman
HW
Houbiers
JG
Vierboom
MP
et al
Identification of peptide sequences that potentially trigger HLA-A2.1-restricted cytotoxic T lymphocytes.
Eur J Immunol.
23
1993
1215
1219
37
Molldrem
J
Dermime
S
Parker
K
et al
Targeted T-cell therapy for human leukemia: cytotoxic T lymphocytes specific for a peptide derived from proteinase 3 preferentially lyse human myeloid leukemia cells.
Blood.
88
1996
2450
2457
38
Vonderheide
RH
Hahn
WC
Schultze
JL
Nadler
LM
The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes.
Immunity.
10
1999
673
679
39
Muller
D
Pederson
K
Murray
R
Frelinger
JA
A single amino acid substitution in an MHC class I molecule allows heteroclitic recognition by lymphocytic choriomeningitis virus-specific cytotoxic T lymphocytes.
J Immunol.
147
1991
1392
1397
40
Dyall
R
Bowne
WB
Weber
LW
et al
Heteroclitic immunization induces tumor immunity.
J Exp Med.
188
1998
1553
1561
41
Rosenberg
SA
Yang
JC
Schwartzentruber
DJ
et al
Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma.
Nat Med.
4
1998
321
327
42
Schultze
JL
Michalak
S
Seamon
MJ
et al
CD40-activated human B cells: an alternative source of highly efficient antigen presenting cells to generate autologous antigen-specific T cells for adoptive immunotherapy.
J Clin Invest.
100
1997
2757
2765
43
Maloney
DG
Grillo-Lopez
AJ
White
CA
et al
IDEC-C2B8 (rituximab) anti-CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin's lymphoma.
Blood.
90
1997
2188
2195
44
Davis
TA
Grillo-Lopez
AJ
White
CA
et al
Rituximab anti-CD20 monoclonal antibody therapy in non-Hodgkin's lymphoma: safety and efficacy of re-treatment.
J Clin Oncol.
18
2000
3135
3143
45
Hunt
DF
Henderson
RA
Shabanowitz
J
et al
Characterization of peptides bound to the class I MHC molecule HLA-A2.1 by mass spectrometry.
Science.
255
1992
1261
1263
46
Tourdot
S
Oukka
M
Manuguerra
JC
et al
Chimeric peptides: a new approach to enhancing the immunogenicity of peptides with low MHC class I affinity: application in antiviral vaccination.
J Immunol.
159
1997
2391
2398

Author notes

John G. Gribben, Dana-Farber Cancer Institute, 44 Binney St, Boston, MA 02115; e-mail: john_gribben@dfci.harvard.edu.

Sign in via your Institution