Purpose: An increasing set of B-cell non-Hodgkin lymphomas (B-NHL) show a biased usage of IGKV3-20 and IGKV3-15 immunoglobulin genes, a feature that could be exploited for the development of ready-to-use, broadly applicable cancer vaccines.

Experimental Design: The immunogenic properties of clonal IGKV3-20 and IGKV3-15 proteins were analyzed with particular focus on their ability to elicit cross-reactive responses against molecularly related IGKV proteins expressed by different B-cell lymphoproliferative disorders.

Results: IGK+ lymphoma patients show humoral and T-cell responses to IGKV3-20 and IGKV3-15 proteins and IGKV3-specific cytotoxic T lymphocytes (CTL) can be easily induced ex vivo. IGKV3-20–specific CTLs cross-react against different IGKV3 proteins, an effect mediated by the presence of 21 shared, sometimes promiscuous, T-cell epitopes, presented by common HLA class I allele products, thus assuring a broad HLA coverage of IGKV3-based vaccines. Many natural epitope variants are carried by IGK light chains expressed by a broad spectrum of B-NHLs and we show that IGKV3-20–specific CTLs cross-react also against several of these variant epitopes. Both humoral and CTL-specific responses were induced by KLH-conjugated IGKV3-20 protein in HLA-A2-transgenic mice and coinjection of IGKV3-20–specific CTLs with IGKV3-20+ or IGKV3-15+ lymphoma cells into SCID mice totally prevented tumor growth, thus confirming the ability of these effectors to mediate efficient and cross-reactive cytotoxic responses also in vivo.

Conclusions: These results provide the rationale to exploit IGKV3 proteins as “off-the-shelf” vaccines for a large fraction of lymphoma patients. Clin Cancer Res; 18(15); 4080–91. ©2012 AACR.

This article is featured in Highlights of This Issue, p. 3999

Translational Relevance

Cancer vaccines for B-cell lymphomas targeting idiotypic proteins constitute a promising therapeutic strategy. Nevertheless, the individualized production of idiotypic vaccines is still complex, costly, and poorly applicable on a large scale. Moreover, the different manufacturing protocols currently used to produce idiotypic vaccines make it difficult to compare the results of different clinical trials. These limitations may be overcome by the exploitation of the immunogenic properties of distinct clonal immunoglobulin light chains, such as IGKV3-15 and IGKV3-20, which are broadly expressed by different B-cell lymphoproliferations. The ability of IGKV3-based recombinant vaccines to elicit cross-reactive responses against molecularly related light chains provides the rationale to activate a phase I/II study aimed at assessing the safety and efficacy of these vaccines. Achievement of these goals would make available a ready-to-use recombinant vaccine for improved consolidation therapy of low-grade B-cell lymphomas.

Therapeutic vaccines targeting B-cell non-Hodgkin lymphoma (NHL) idiotype (Id) represent a promising approach to improve the clinical control of these malignancies (1, 2). Indeed, immunization with tumor B-cell Id has been associated with induction of lymphoma-specific humoral and cellular responses, molecular remissions, and prolonged progression-free or disease-free survival in several phase I/II studies (1–8). Moreover, 1 of 3 phase III randomized trials carried out so far has shown prolonged remission duration in follicular lymphoma patients in first remission (9). Although the reasons for these diverging results are still a matter of debate, differences in the methods used for vaccine production could have accounted for the different outcomes observed. In all these studies, in fact, idiotypic immunoglobulins (Ig) were produced in vitro from each patient using different protocols, which usually imply considerable efforts and costs (1, 2). Therefore, there is the need to develop Id vaccines that can be more easily manufactured and applicable to a larger number of patients (10). Indeed, availability of broadly usable Id vaccines would considerably boost the clinical application of this therapy and allow the comparison of the results of different clinical trials.

Recent evidence suggests that the limitations related to the individualized production of Id vaccines may be overcome. In fact, distinct sets of stereotyped Igs have been identified in various B-cell malignancies, suggesting that a much higher frequency of Id sharing exists among patients than appreciated previously. In particular, the molecular characterization of Hepatitis C Virus (HCV)-related lymphomas showed that >70% of these cases preferentially use either IGKV3-20 or IGKV3-15 light chains, with a high degree of homology across different lymphomas (11–14). Similar molecular features have been also found in a proportion of non-HCV–related B-NHLs, including autoimmunity-associated lymphoproliferations (13), diffuse large B-cell lymphomas (15), marginal zone B-cell lymphomas (16, 17), follicular lymphomas (18), multiple myeloma (18), and chronic lymphocytic leukemias (18–20). On these grounds, IGKV3-based vaccines could be used in a considerable fraction of patients with different B-cell lymphoproliferations.

This study aimed at characterizing the immunogenic properties of IGKV3-20 and IGKV3-15 proteins with the final goal to provide the rationale for the use of these molecules as “shared” vaccines. We herein show that these proteins are immunogenic ex vivo and capable of eliciting humoral and T-cell responses in patients with NHL. Notably, these recombinant proteins induce specific cytotoxic T lymphocyte (CTL) responses that cross-react against related IGVK proteins expressed by different lymphomas/leukemias. The IGKV3-20 protein, in particular, can be regarded as a “string of beads” carrying multiple promiscuous class I-restricted T-cell epitopes that are naturally recognized in patients with IGKV3+ lymphomas. In addition, some of these epitopes are frequently shared across a broad spectrum of IGK light chain families used by different B-cell lymphomas/leukemias, thus supporting a broad application of these candidate vaccines.

Patients and healthy donors

Peripheral blood was collected from healthy donors and 13 patients with HCV-NHL (Supplementary Table S1). All participants provided signed informed consent, and the study was approved by local IRB. HLA typing was conducted by standard high-resolution typing.

Production of recombinant Id proteins

Sequences from 1 IGKV3-15 and 2 IGKV3-20 rearranged genes were PCR amplified from NHLs using family-specific primers (21). IGKV3-specific fragments were subcloned into pFLAG-MAC and transformed into JM109 strain Escherichia coli (21). The expressed proteins were purified by affinity column and their sequence verified by fingerprinting analysis of trypsin-digested extracts (22). Endotoxin level was <1.0 EU/μg of protein (LAL method).

Cell lines

Lymphoblastoid cell lines (LCL) were generated by in vitro transformation of B cells with the B.95.8 Epstein–Barr virus isolate. The SH9 LCL was established from peripheral blood mononucleated cells (PBMC) of a patient with Hodgkin lymphoma. The DG75 Burkitt's lymphoma cell line was obtained from DSMZ and confirmed by fingerprinting (Power Plex 1.2; Promega) in April 2011. Cell lines were cultured in RPMI-1640, containing 10% heat-inactivated FBS (Gibco), 2 mmol/L l-glutamine, 100 μg/mL streptomycin, and 100 IU/mL penicillin (Sigma-Aldrich) and maintained at 37°C in a 5% CO2 atmosphere.

Generation and characterization of IGKV3-specific CTLs

Generation of IGKV3- and peptide-specific CTLs was conducted as described (23, 24). After 3 stimulations with protein- or peptide-loaded DCs, CTL cultures were characterized with the following FITC-, PE-, ECD-, PC5-, PC7-labeled antibodies used in a 5-color combination: anti-CD3 (UCHT1, IgG1), anti-CD45RA (2H4LDH11LDB9, IgG1), anti-CD4 (13B8.2, IgG1), anti-CD8 (SFCI21ThyD3, IgG1; Beckman Coulter), anti-CCR7 (3D12, Rat, IgG2b; BD Biosciences PharMingen), anti-CD11a (HI111, IgG1), anti-CD44 (IM7, Rat IgG2b), anti-CD49d (9F10, IgG1, k) and anti-CD62L (DREG56, IgG1, k; eBioscience). Mouse FITC- and PE-IgG1 (679.1Mc; Beckman Coulter) and Rat PE-IgG2a (eBRa; eBioscience) were used as isotypic controls. Cytofluorimetric analysis was conducted with a Cytomics FC500 and the data analyzed with CXP software (Beckman Coulter). To generate peptide-specific CTL microcultures, dendritic cells (DC) were differentiated from peripheral blood monocytes of HLA-A*0201+ donors and loaded overnight with 25 μg/mL of each of 10 different IGKV3-20–derived peptides selected for HLA-A*0201 binding. The HLA-A*0201–restricted cyclin D1–derived peptide (LLNDRVLRA) and the Flu-matrix58–66 peptide served as negative and positive controls, respectively. For each peptide, 30 replicate microcultures were set-up and fed with IL-2 (60 IU/mL) and IL-7 (10 IU/mL) at day 2. All cultures were restimulated at days 7 and 14 with autologous peptide-loaded and irradiated (7 Gy) PBLs (1 × 105 cells/well) and at day 21 analyzed for cytotoxicity by a calcein-AM release assay (23). All tests were conducted in triplicate. HLA class I w6/32 mAb (10 μg/mL; Abcam) or HLA-A*0201–specific cr11.351 mAb (10 μg/mL; 25) were used to assess HLA restriction, and the anti-Epstein–Barr Virus Nuclear Antigen 1 (EBNA1) mAb (1:50; Millipore) was used as irrelevant antibody. Fluorescence intensity was measured with a SpectraFluorPlus fluorimeter (Tecan).

IFN-γ ELISPOT assays

IFN-γ-ELISPOT assay was conducted with a commercial kit (Endogen) according to manufacturer's instructions and using as stimulators (×105 cells/well) autologous monocytes pulsed with either 10 μg/mL of Id protein or 20 μg/mL peptide, and immunomagnetically purified CD8+ T lymphocytes as responders (105 cells/well). Plates were evaluated by a computer-assisted ELISPOT reader (Eli.Expert, A.EL.VIS). The number of spots in negative controls never exceeded 5 spots/well. Responses were considered significant if >5 IFN-γ–producing cells were detected.

Detection of humoral responses to IGKV3-20 in lymphoma patients

The analysis was carried out using a chemiluminescent ELISA test. Briefly, 96-well microtiter plates were coated overnight with either recombinant IGKV3-20 protein or Tetanus Toxoid (TT, Calbiochem) antigens (2 μg/mL). Sera from lymphoma patients or donors were added to the wells in triplicate and incubated for 2 hours at room temperature (RT). Plates were washed, and horseradish peroxidase-conjugated goat antihuman IgG (Sigma-Aldrich) was added. After incubation for 1 hour at RT, plates were washed, developed using chemiluminescence substrate LiteAblot (CELBIO), and read with a GENios Elisa-reader (Tecan).

T-cell epitope mapping and characterization

Identification of IGKV3-20–derived epitopes was carried out with the iTopia Epitope Discovery System (Beckman Coulter; ref. 26). Briefly, 100 9-mer peptides overlapping by 8 residues spanning the entire length of the protein (108 aa) were screened for their ability to bind 8 HLA class I alleles (A*0101, A*0201, A*0301, A*1101, A*2402, B*0702, B*0801, and B*1501) under optimal conditions (27). The percentage of binding was assessed using a fluorimeter and peptides which bound MHC molecules >20% compared with the reference positive control were selected for analysis of affinity, expressed as ED50 (the concentration at which the peptide is bound to the MHC at 50% maximum binding efficiency). Peptides were also analyzed for off-rate (complex stability) where T½ represents the time taken for a reduction from optimal binding of 50% after a shift to dissociating conditions (27).

IGKV3-20–specific humoral and T-cell responses in animal models

Six- to 8-week-old female SCID or HHD mice were used according to the national guidelines (28). HHD mice were immunized by subcutaneous (s.c.) injections of KLH-conjugated IGKV3-20 (100 μg) emulsified in complete Freund adjuvant (Sigma-Aldrich). Subsequent injections were conducted at day 14, 21, 30, and 45 with 100 μg of KLH-conjugated IGKV3-20 emulsified in incomplete Freund adjuvant. HHD sera were analyzed at day 0 and 30 by ELISA. Ten μg/mL of nonconjugated IGKV3-20 were coated onto a 96-well Maxisorp NUNC-immunoplate (Thermo Scientific) in 0.05M sodium carbonate/bicarbonate buffer pH 9.6 (Sigma-Aldrich), overnight at 4°C. After incubation for 1 hour at RT with 100 μL per well of PBS-10% FBS, plates were incubated for 1 hour at RT with sequential dilutions of mouse sera in PBS-10% FBS (from 1:50 to 1:6400). Negative control was PBS-10% FBS alone and sera at day 0 (1:50 in PBS-10% FBS). Then, plates were incubated for 1 hour at RT with secondary HRP-conjugated goat antimouse antibody (Charles River Laboratories; 0.02 μg/mL in PBS-0.05% Tween). Finally, 100 μL per well of substrate solution (prepared using SIGMA-Fast OPD tablets) were added for 5 minutes at RT in the dark. Reaction was stopped with 50 μL/well of HCl 3N (Carlo Erba), and plates analyzed at 490 nm using a VictorX4 instrument (Perkin-Elmer). Data were used to generate a titration curve. The same sera used in ELISA test were analyzed for their ability to stain DG75 cells in flow cytometry.

IGKV3-20–specific CTLs were generated from HHD mice showing humoral responses capable of staining DG75 cells cytofluorimetrically. Candidate mice were sacrificed, their splenocytes isolated and cultured (25 × 106) with 1 μmol/L IGKV3-20 protein or γ-irradiated (60 Gy) DG75 cells (1 × 105) for 4 days. Splenocytes from nonimmunized HHD mice were used as negative controls. At day 4, cells were harvested and CTL cultures positive at the cytotoxicity assay were maintained with appropriate stimulus (either 1 μmol/L protein or 1 × 105 γ-irradiated DG75), and tested every 2 weeks to assess their lytic ability. Winn assay was conducted by injecting s.c. 5 × 106 DG75 or SH9 cells/animal mixed with either RPMI or donor-derived IGKV3-20–specific CTLs (5 × 106/animal), testing 6 animals/group. Mixing was conducted by incubating cells for 5 minutes at 4°C immediately before injection, to rule out T-cell/target interaction before in vivo transfer. Tumor growth was evaluated every 2 to 3 days using a caliper (29).

Ex vivo immunogenicity of IGKV3-20 and IGKV3-15 recombinant proteins

The IGKV3-15 and IGKV3-20 proteins are expressed by a sizeable fraction of B-cell malignancies (Table 1). In particular, IGKV3-20 is used 2 to 3 times more frequently than IGKV3-15. Two representative IGKV3-15 and IGKV3-20 recombinant proteins were used to induce CTL cultures by stimulating PBMCs of healthy donors with protein-pulsed autologous DC. Specific CTL lines were easily obtained from 14 donors with either IGKV3-20 (n = 12) or IGKV3-15 (n = 4) proteins (Fig. 1A and Supplementary Table S2). Overall, almost 30% of the cells in the cultures (29.5 ± 5.9%, mean ± SD; n = 10) were CD8+ T cells, most of which (>90%) were effector memory (CCR7−CD45RA−) or terminally-differentiated (CCR7−CD45RA+) T cells (Supplementary Table S3). Notably, these cultures were also composed of a significant proportion of CD4+ T cells (62.76 ± 6.20%, n = 10). All CTL cultures generated with IGKV3-20 or IGKV3-15 showed class I-restricted cytotoxic activity against autologous LCLs pulsed with the immunizing protein (Fig. 1A).

Figure 1.

A, CTL cultures generated with IGKV3-15 or IGKV3-20 showed class I–restricted cytotoxicity against autologous LCLs pulsed with the immunizing protein. Recombinant p29 BARF1 protein was used as unrelated control. B, CTLs induced by the NHL patient-derived IGKV3-20a protein specifically kill also autologous targets loaded with an unrelated IGKV3-20b. E:T ratio = 20:1 (P < 0.02, Student t test). C, amino acid sequence alignment of the prototypic and 2 variant IGKV3-20 proteins, IGKV3-20a and IGKV3-20b. D, sequence homology of DG75 and SH9 IGKV3 proteins. E, IGKV3-20- and IGKV3-15–specific CTL cultures derived from 3 donors killed, in a class I–restricted fashion, both matched (same IGKV3) and mismatched (IGKV3-20 vs. IGKV3-15 and vice versa) targets. The anti-EBNA1 mAb was used as irrelevant antibody.

Figure 1.

A, CTL cultures generated with IGKV3-15 or IGKV3-20 showed class I–restricted cytotoxicity against autologous LCLs pulsed with the immunizing protein. Recombinant p29 BARF1 protein was used as unrelated control. B, CTLs induced by the NHL patient-derived IGKV3-20a protein specifically kill also autologous targets loaded with an unrelated IGKV3-20b. E:T ratio = 20:1 (P < 0.02, Student t test). C, amino acid sequence alignment of the prototypic and 2 variant IGKV3-20 proteins, IGKV3-20a and IGKV3-20b. D, sequence homology of DG75 and SH9 IGKV3 proteins. E, IGKV3-20- and IGKV3-15–specific CTL cultures derived from 3 donors killed, in a class I–restricted fashion, both matched (same IGKV3) and mismatched (IGKV3-20 vs. IGKV3-15 and vice versa) targets. The anti-EBNA1 mAb was used as irrelevant antibody.

Close modal
Table 1.

Distribution of IGKV3-15 and IGKV3-20 proteins among B-cell malignancies

IGKV3-15IGKV3-20IGKV3-15 or IGKV3-20IGKV3 family
B-cell malignancyN (%)N (%)N (%)N (%)Ref.
HCV-related NHL 3/21 (14.3) 12/21 (57.1) 15/21 (71.4) 16/21 (76.2) 10, 13 
Diffuse large B-cell lymphoma 4/20 (20) 1/20 (5) 5/20 (25) 7/20 (35) 10, 14 
Splenic marginal-zone lymphomas 1/31 (3.2) 3/31 (9.7) 4/31 (12.9) 5/31 (16.1) 17 
Sjögren syndrome–associated lymphoproliferations 2/7 (28.6) 4/7 (57.1) 6/7 (85.7) 6/7 (85.7) 12 
MALT lymphomas 2/18 (11.1) 7/18 (38.9) 9/18 (50) 9/18 (50) 15, 16 
Follicular lymphomas — 3/47 (6.4) — 6/47 (12.8) 17 
Multiple myeloma 4/71 (5.6) 7/71 (9.9) 11/71 (15.5) 11/71 (15.5) 17 
Chronic lymphocytic leukemias 28/614 (4.6) 72/614 (11.7) 100/614 (16.3) 139/614 (22.6) 17–19 
IGKV3-15IGKV3-20IGKV3-15 or IGKV3-20IGKV3 family
B-cell malignancyN (%)N (%)N (%)N (%)Ref.
HCV-related NHL 3/21 (14.3) 12/21 (57.1) 15/21 (71.4) 16/21 (76.2) 10, 13 
Diffuse large B-cell lymphoma 4/20 (20) 1/20 (5) 5/20 (25) 7/20 (35) 10, 14 
Splenic marginal-zone lymphomas 1/31 (3.2) 3/31 (9.7) 4/31 (12.9) 5/31 (16.1) 17 
Sjögren syndrome–associated lymphoproliferations 2/7 (28.6) 4/7 (57.1) 6/7 (85.7) 6/7 (85.7) 12 
MALT lymphomas 2/18 (11.1) 7/18 (38.9) 9/18 (50) 9/18 (50) 15, 16 
Follicular lymphomas — 3/47 (6.4) — 6/47 (12.8) 17 
Multiple myeloma 4/71 (5.6) 7/71 (9.9) 11/71 (15.5) 11/71 (15.5) 17 
Chronic lymphocytic leukemias 28/614 (4.6) 72/614 (11.7) 100/614 (16.3) 139/614 (22.6) 17–19 

Considering the derivation from antigen-experienced B cells of most lymphomas expressing IGKV3-20 or IGKV3-15, these proteins may have undergone the process of somatic hypermutation and may thus show a certain degree of interpatient variation in the amino acid sequence. This does not apparently constitute a major limitation because CTLs induced by a prototypic IGKV3-20 light chain (VK3-20a) were also able to specifically kill autologous targets loaded with idiotypic VK proteins of the same subfamily (VK3-20b, Figs. 1B and C) derived from unrelated NHL. These results were obtained in 4 donors with different HLA class I haplotypes (Fig. 1B), thus showing that the CTL responses induced by a “prototypic” IGKV3-20 protein may have a potential immunotherapeutic value also in unrelated patients carrying lymphomas showing the same VK restriction.

IGKV3 proteins induce cross-reactive CTL responses

The IGKV3-15 and IGKV3-20 proteins show a sequence homology >80% (Fig. 1D), a biochemical basis for the induction of cross-reactive responses. To address this issue directly, we verified whether IGKV3-15–specific CTLs could kill autologous targets presenting IGKV3-20, and vice versa. As shown in Fig. 1E, IGKV3-15- and IGKV3-20–specific cultures obtained from 3 donors significantly killed, in a class I–restricted fashion, also mismatched targets. Intriguingly, IGKV3-20–induced CTLs killed IGKV3-15–loaded targets with an efficiency comparable to that of matched IGKV3-15-CTLs; only in a single case, IGKV3-20-CTLs showed about a 2-fold higher killing activity against IGKV3-15 targets than matched CTLs (Fig. 1E).

Sequencing of IGKV genes expressed by a large panel of B-cell lines allowed the identification of the DG75 BL cell line (HLA-A*0201/6601, B*4101/5001) naturally expressing IGKV3-20, and the SH9 LCL (HLA-A*0201/0101, B*0702/0801) expressing the IGKV3-15 protein. Notably, both these cell lines were killed in an antigen-specific and HLA-A*02–restricted manner by IGKV3-20–specific CTLs obtained from donors single-matched for HLA-A*0201 with target cell lines (Fig. 2). These findings indicate that IGKV3-20 and IGKV3-15 are naturally processed in lymphoid cells and produce “cross-reactive” epitopes.

Figure 2.

DG75 (IGKV3-20+) and SH9 (IGKV3-15+) cells were efficiently killed in a HLA-A*0201–restricted manner (cr11.351 mAb) by IGKV3-20–specific CTLs obtained from donors single-matched for HLA-A*0201 with target cells. E:T ratio = 20:1. IGKV3 LCLs and BARF1 p29 pulsed LCL were used as negative controls (P < 0.02, Student t test).

Figure 2.

DG75 (IGKV3-20+) and SH9 (IGKV3-15+) cells were efficiently killed in a HLA-A*0201–restricted manner (cr11.351 mAb) by IGKV3-20–specific CTLs obtained from donors single-matched for HLA-A*0201 with target cells. E:T ratio = 20:1. IGKV3 LCLs and BARF1 p29 pulsed LCL were used as negative controls (P < 0.02, Student t test).

Close modal

IGKV3-specific humoral and CD8+ T-cell responses in lymphoma patients

The presence of memory T-cell responses specific for IGKV3-20 and IGKV3-15 proteins was investigated in 13 patients with IGK+ HCV-NHLs. A 48-hour ELISPOT assay disclosed low-to-moderate amounts of IFN-γ–producing T cells in the majority of patients (3-98 SFC/100.000 CD8+ T cells), whereas all donors (n = 4) showed low-level responses (<7 SFC/100.000 CD8+ T cells; Fig. 3A). Moreover, specific humoral responses to IGKV3-20 protein were also observed in most of the patients from whom serum was available (n = 11; Supplementary Fig. S1).

IGKV3-20 CTL epitope mapping and validation

Using the iTopia assay (26), we identified 21 IGKV3-20 peptides able to bind at least 1 of 8 MHC allele products. Interestingly, although most peptides (n = 18) bound only one allele, the P5, P28, and P97 peptides bound 2 to 3 different alleles (Supplementary Table S4). All peptides were low-to-medium binders, with ED50 ranging from 110 to 255 μmol/L (Supplementary Table S4) and half-times varying from 18 to 135 minutes, similarly to other Id-derived peptides (30).

The ability of HLA-A*0201–binder peptides to elicit specific T-cell responses was assessed by IFN-γ-ELISPOT with purified CD8+ T cells obtained from 3 IGK+ HCV-NHL patients and 2 healthy donors. IFN-γ–producing T cells were found in the majority of patients with lymphoma, with higher responses for most epitopes as compared with healthy donors (15–20 vs. 5–10 SFC/100,000 CD8+ T cells; Fig. 3B). In addition, epitope-specific CTLs were successfully induced ex vivo from 6 different donors using each of 10 A*0201-restricted, IGKV3-20–derived peptides (Fig. 3C). Notably, CTLs induced with P20 and P33 peptides were able to kill lymphoma cells expressing either IGKV3-20 (DG75) or IGKV3-15 (SH9) in a HLA-A2–restricted fashion (Fig. 3D).

Figure 3.

A, IFN-γ-ELISPOT showing IGKV3-15- and IGKV3-20–specific CD8+ T-cell responses in IGK+ lymphoma patients and in 4 donors. Mean values of T-cell responses to TT were used as positive control. B, CD8+ T-cell responses against HLA-A*0201–restricted IGKV3-20 epitopes in 3 patients with NHL (top) and 2 donors (bottom). FLU M158–66 peptide was used as positive control and background responses were evaluated on empty monocytes. C, induction of IGKV3-20 peptide-specific T-cell responses. Lysis of peptide-loaded T2 cells by day +21 microcultures from 6 HLA-A*0201+ donors (D1–D6). For each peptide, and each donor, 30 independent microcultures (○) were set up and tested at 20:1 E:T ratio. Microcultures generated with the Flu-M158–66 peptide were used as positive control. Results expressed as specific lysis of peptide-loaded T2 cells after subtraction of lysis of empty T2 cells. Gray area, mean ± 3SD of the spontaneous release of T2 cells. D, cytotoxic activity of CTLs induced from PBMCs of 3 donors with the P20 and P33 IGKV3-20 peptides against DG75 (IGKV3-20+) and SH9 (IGKV3-15+) cells (E:T ratio = 20:1). Lysis of an autologous LCL pulsed with IGKV3-20 is shown as a positive control. HLA-A*02 restriction was confirmed with the cr11.351 antibody.

Figure 3.

A, IFN-γ-ELISPOT showing IGKV3-15- and IGKV3-20–specific CD8+ T-cell responses in IGK+ lymphoma patients and in 4 donors. Mean values of T-cell responses to TT were used as positive control. B, CD8+ T-cell responses against HLA-A*0201–restricted IGKV3-20 epitopes in 3 patients with NHL (top) and 2 donors (bottom). FLU M158–66 peptide was used as positive control and background responses were evaluated on empty monocytes. C, induction of IGKV3-20 peptide-specific T-cell responses. Lysis of peptide-loaded T2 cells by day +21 microcultures from 6 HLA-A*0201+ donors (D1–D6). For each peptide, and each donor, 30 independent microcultures (○) were set up and tested at 20:1 E:T ratio. Microcultures generated with the Flu-M158–66 peptide were used as positive control. Results expressed as specific lysis of peptide-loaded T2 cells after subtraction of lysis of empty T2 cells. Gray area, mean ± 3SD of the spontaneous release of T2 cells. D, cytotoxic activity of CTLs induced from PBMCs of 3 donors with the P20 and P33 IGKV3-20 peptides against DG75 (IGKV3-20+) and SH9 (IGKV3-15+) cells (E:T ratio = 20:1). Lysis of an autologous LCL pulsed with IGKV3-20 is shown as a positive control. HLA-A*02 restriction was confirmed with the cr11.351 antibody.

Close modal

Cross-reactivity of CTL responses elicited by IGKV3-20–derived epitopes

The ability of IGKV3-20–specific CTLs to “cross kill” targets presenting or naturally expressing IGKV3-15 prompted us to verify whether the IGKV3-20–derived epitopes are also shared by other IGK proteins from the IGKV3 as well as from unrelated IGKV families, to better define the extent of lymphoid tumors potentially treatable with IGKV3-20–based vaccines. Sequences of T-cell epitopes were then aligned against all germ-line IGKV sequences retrieved from the ImMunoGeneTics information system® (31). As a conservative rule, only peptides showing only one amino acid change compared with the IGKV3-20 epitopes were considered. The analysis showed that most of the IGKV3-20 epitopes are conserved in several light chains of the IGKV3 family (Supplementary Table S5). Interestingly, 2 of these epitopes (P32 and P33) are shared also by several other IGKV families extensively used among B-cell tumors (Supplementary Table S5). Moreover, the HLA-A alleles restricting these peptides (A*1101 and A*2402 for P32 and A*0201 for P33) would ensure a broad population coverage.

To assess whether the cross-reactivity of IGKV3-20–specific CTLs against IGKV3-15–loaded targets was mediated by the recognition of identical or variant epitopes, 30 independent CTL microcultures, for each of 13 different peptide/donor conditions, were generated either using P20, P32, P33, P46, P47, and P48 peptides. Each peptide-specific culture was tested using both the native and the corresponding IGKV3-15–derived variant peptide epitope as target. Part of peptide-specific microcultures were able to efficiently recognize and kill T2 cells loaded not only with the same native peptide, but also with the variant epitope peptides (Supplementary Fig. S2A and S2B). More interestingly, few cultures showed cell lysis selectively to variant peptides, although no cytotoxicity against the native epitope was observed (Supplementary Fig. S2). Thus, IGKV3-20-CTLs recognize both similar and variant epitopes in IGKV3-15, further emphasizing the cross-reactivity of IGKV3-induced responses.

We also analyzed the ability of 2 representative IGKV3-20–derived A*0201 peptides to generate CTLs able to cross-react against 9 epitope variants (2 for P20 and 7 for P33) differing only for 1 amino acid and carried by IGK chains expressed by different B-cell tumors (Supplementary Table S5). Four of these “variant” peptides had a single amino acid substitution at the MHC anchor sites. As shown in Fig. 4A, T-cell lines generated from A*0201 donors with the P20 and P33 peptides specifically recognized and efficiently killed autologous LCLs pulsed with all the epitope variants as well as IGKV3-15+ and IGKV3-20+ lymphoma cell lines (Fig. 4B). Notably, in some instance, the killing of LCLs pulsed with the variant peptides was even higher than that induced by the “native” peptides (Fig. 4A and B). The extent of cytotoxic responses against variant peptides was retained at comparable levels also at low effector:target ratios (1:1) and when LCLs were pulsed with peptide concentrations as low as 1 μg/mL (not shown), thus assuring about the specificity of recognition. All the variant peptides investigated showed a similar or, more frequently, lower predicted binding affinity to A*0201 as compared with the “native” epitopes (Supplementary Table S5). These findings suggest that the IGKV3-20 protein carries natural heteroclytic versions of several CTL epitopes in the IGK proteins broadly expressed among lymphoid malignancies.

Figure 4.

A, cross-reactive CTL responses induced by CTL cultures generated from 4 donors with the HLA-A*0201 P20 and P33 IGKV3-20 peptides against 2 and 7 variants of the P20 and P33 epitopes, respectively. Lysis of autologous LCLs loaded with either native peptides (N) or recombinant IGKV3-20 is shown as well as the HLA-A*0201 restriction (cr11.351 mAb). IGKV3-20- LCLs and cyclin D1–derived peptide (Cy D1) loaded LCLs served as negative control (P < 0.02, Student t test). B, lytic activity of CTLs generated from 3 donors with native or variant HLA-A*0201 P20 and P33 peptides against lymphoma cells naturally expressing IGKV3-20 (DG75) or IGKV3-15 (SH9).

Figure 4.

A, cross-reactive CTL responses induced by CTL cultures generated from 4 donors with the HLA-A*0201 P20 and P33 IGKV3-20 peptides against 2 and 7 variants of the P20 and P33 epitopes, respectively. Lysis of autologous LCLs loaded with either native peptides (N) or recombinant IGKV3-20 is shown as well as the HLA-A*0201 restriction (cr11.351 mAb). IGKV3-20- LCLs and cyclin D1–derived peptide (Cy D1) loaded LCLs served as negative control (P < 0.02, Student t test). B, lytic activity of CTLs generated from 3 donors with native or variant HLA-A*0201 P20 and P33 peptides against lymphoma cells naturally expressing IGKV3-20 (DG75) or IGKV3-15 (SH9).

Close modal

Humoral and cellular responses to IGKV3-20 in mice

Immunization of HHD mice with KLH-conjugated IGKV3-20 yielded high titers of specific antibodies (Fig. 5A). Sera were also tested for the capacity of staining cytofluorimetrically the IGKV3-20+ DG75 cells. After a first round of 3 immunizations, DG75 cells were not recognized and additional immunizations were required to obtain a detectable staining signal (Fig. 5B). Interestingly, although ELISA Ig titers remained almost constant (not shown), indicating that antibodies to IGKV3-20 had already reached a plateau, repeated immunizations boosted detectable levels of Ig capable of recognizing naturally folded, public epitopes physiologically present on the membrane-mounted protein.

Figure 5.

A, ELISA assay carried out with unconjugated IGKV3-20 protein showing the absorbance values of sera from 4 mice (black and white circles and triangles) immunized with IGKV3-20-KLH (day 31). Black squares, mean values of absorbance of sera from the same mice on day 0. B, flow cytometry of DG-75 cells stained with different mouse sera obtained after 31 (left) and 60 days (right) from immunization. C, cytotoxic activity of CTLs obtained from HHD mice immunized with IGKV3-20 and restimulated in vitro with γ-irradiated DG75 cells against the IGKV3-20+ DG-75 cell line (black circles) or an IGKV3-20–negative B-cell line (white circles). As a control, splenocytes from nonimmunized HHD mice were similarly restimulated and used as effectors against the same target cells (black and white squares, respectively). D and E, Winn assay. SCID mice (n = 3) were injected with the DG-75 (D) or SH9 (E) cell lines alone (white circles) and premixed with IGKV3-20–specific CTLs (black circles) at day 0. The panel shows the tumor mass growth. Statistical analysis was conducted with ANOVA (repeated measures of variance analysis). Both Winn assays were repeated twice using 2 different donors for CTL generation.

Figure 5.

A, ELISA assay carried out with unconjugated IGKV3-20 protein showing the absorbance values of sera from 4 mice (black and white circles and triangles) immunized with IGKV3-20-KLH (day 31). Black squares, mean values of absorbance of sera from the same mice on day 0. B, flow cytometry of DG-75 cells stained with different mouse sera obtained after 31 (left) and 60 days (right) from immunization. C, cytotoxic activity of CTLs obtained from HHD mice immunized with IGKV3-20 and restimulated in vitro with γ-irradiated DG75 cells against the IGKV3-20+ DG-75 cell line (black circles) or an IGKV3-20–negative B-cell line (white circles). As a control, splenocytes from nonimmunized HHD mice were similarly restimulated and used as effectors against the same target cells (black and white squares, respectively). D and E, Winn assay. SCID mice (n = 3) were injected with the DG-75 (D) or SH9 (E) cell lines alone (white circles) and premixed with IGKV3-20–specific CTLs (black circles) at day 0. The panel shows the tumor mass growth. Statistical analysis was conducted with ANOVA (repeated measures of variance analysis). Both Winn assays were repeated twice using 2 different donors for CTL generation.

Close modal

Immunization-induced CTL generation was also exploited in the HHD mouse model, which allows the presentation of epitopes in the HLA-A*0201 context, and hence the generation of mouse CTLs potentially able to recognize human A*0201+ target cells. Splenocytes from IGKV3-20–immunized mice were restimulated in vitro with irradiated DG75 cells and tested 4 days later for lytic activity. As shown in Fig. 5C, these effectors showed a strong IGKV3-20–specific CTL activity. To assess whether the in vitro ability of CTL lines to target and lyse IGKV3+ cells could find an in vivo counterpart in the capacity of restraining tumor growth, SCID mice were injected in one flank with DG75 tumor cells admixed with human IGKV3-20-specific CTLs (ratio 1:1), although the opposite flank was inoculated with tumor cells alone. Control DG75 tumors developed in 100% of injected animals 8 to 9 days after inoculation, and reached the allowed maximal tumor volume in about 15 days. Conversely, at the site where DG75 cells and IGKV3-20–specific CTLs were coinjected, no growth, nor formation of tumor masses occurred over the entire observation period, thus showing the tumor growth inhibitory activity of IGKV3-20–specific CTLs (P < 0.001, n = 3; Fig. 5D). Similar findings were observed with the IGKV3-15+ SH9 cell line, thus confirming the cross-reactivity of IGKV3-20 CTL responses also in vivo (P = 0.001, n = 3; Fig. 5E).

In this study, we have extensively characterized the immunogenic properties of the homologous IGKV3-20 and IGKV3-15 proteins, which can be considered as prototypic “off-the-shelf” vaccines. In fact, these proteins are collectively expressed by a large fraction of HCV-related NHL (>70%) and by a sizeable proportion of HCV-unrelated lymphoid malignancies (Table 1). Both IGKV3 proteins were shown to easily elicit HLA class I–restricted specific CTL responses ex vivo from different donors, thus excluding the existence of “holes” in the TCR repertoire because of central tolerance. These proteins are also naturally immunogenic in vivo, as shown by the detection of specific humoral and CD8+ T-cell responses in patients with lymphoma, ruling out also the occurrence of peripheral tolerance/ignorance. Notably, the presence a significant proportion of CD4+ T lymphocytes within the cultures generated with IGKV3-20 and IGKV3-15 is of relevance in the light of the critical role of CD4+ T-cell responses in mediating the vaccine effect.

An ideal “shared” IGKV3 vaccine should elicit CTL responses effective against unrelated lymphomas expressing IGKV3 proteins. This is a relevant issue if we consider that most of NHLs potentially treatable with these vaccines derive from antigen-experienced B cells, whose Id proteins may thus carry somatic mutations possibly affecting immune recognition. Our findings indicate that CTLs induced with a prototypic IGKV3-20 may be effective also against IGKV3-20 proteins derived from different NHLs, supporting the potential immunotherapeutic value of this vaccine for unrelated patients with IGKV3-20+ lymphomas. Furthermore, we also show that CTLs induced by IGKV3-20 or IGKV3-15 elicit mutually cross-reactive responses, being able to efficiently kill target cells presenting or naturally expressing the mismatched IGKV3 protein. Considering the relatively frequent usage of IGKV3-20 and IGKV3-15 among lymphoid malignancies, these findings support a broad applicability of these vaccines. The higher prevalence of IGKV3-20 expression and its slightly higher efficiency in eliciting “cross-reactive” CTL responses indicate that IGKV3-20 is the preferred candidate for vaccination.

The immunogenicity of IGKV3-20 is supported by the identification of 21 different CTL epitopes presented by common HLA class I alleles. All these peptides were low-to-medium binders, consistently with their derivation from a self-protein and with the features of other Id-derived epitopes (30). Memory CD8+ T-cell responses to the majority of HLA-A*0201–restricted IGKV3-20 epitopes were detected in patients with HCV-NHL, indicating that these epitopes are immunogenic in vivo. Moreover, we also showed that CTLs specific for these epitopes can be generated from different A*0201+ donors. Notably, these effectors were able to kill in a HLA-A2–restricted fashion lymphoma cells expressing either IGKV3-20 or IGKV3-15, showing that these shared epitopes are efficiently generated and presented by tumor cells.

Notably, IGKV3-20 carries 3 promiscuous epitopes able to bind 2 or 3 different HLA class I allele products. Given the relatively well-established class I binding motifs for most alleles (32, 33), binding of epitopes across a broad spectrum of HLA class I alleles has been considered the exception rather than the rule (34, 35). Indeed, several CTL epitopes can be presented in the context of more than 1 HLA class I allele product, although this is largely limited to the same HLA supertype (34–37). The 3 IGKV3-20 promiscuous CTL epitopes show binding properties that extend beyond a single HLA supertype, being in this respect similar to viral epitopes (38). Therefore, a broad HLA coverage of the IGKV3-20 vaccine is assured not only by a relatively high number of CTL epitopes presented by common HLA molecules, but also by the promiscuity of distinct epitopes. Notably, IGKV3-20 promiscuous peptides are able to induce specific effectors with similar cytotoxic activity from donors with unrelated HLA alleles (Supplementary Fig. S2C and S2D), a relevant prerequisite for the induction of comparable CTL responses across different HLA backgrounds.

One of the most intriguing results of this study is the demonstration that 2 IGKV3-20–derived epitopes are fully conserved and shared by various IGKV families broadly expressed among lymphomas/leukemias. Moreover, several “variants” of the IGKV3-20–derived epitopes are carried by clonotypic IGKV proteins expressed by different lymphoid tumors. Notably, IGKV3-20–specific CTLs efficiently recognize and kill autologous targets presenting different A*0201-restricted IGKV3-20 “variant” epitopes, often with a higher efficiency as compared with that elicited against the “native” epitopes. This is consistent with the lower predicted affinity showed by some of the “variant” peptides, which carry single amino-acid substitutions at the MHC anchor sites. These findings indicate that the IGKV3-20 protein carries “natural” heteroclitic versions of several CTL epitopes shared by VK/VJ proteins broadly expressed among lymphoid malignancies. Available evidence indicates that heteroclitic peptide modifications can increase the immunogenicity of low-binding peptides, thus leading to improved ability to generate CTL responses against primary tumors (39–41). The enhanced immunogenicity of heteroclitic peptides has been elegantly shown also using analog peptides derived from native epitopes of Ig heavy chains (42). Moreover, heteroclitic peptides from various tumor antigens are now widely applied in clinical studies, both as single peptides and included in multiepitope vaccines (43). The demonstration that the IGKV3-20 protein naturally carries multiple CTL epitopes shared among various IGKV families, and that may behave as heteroclitic peptides further supports the applicability and the potential efficacy of this vaccine in a discrete proportion of IGK-expressing leukemias/lymphomas.

We also show that the IGKV3-20 protein is immunogenic in mice, being able to elicit both humoral and CTL-specific responses. Although the relevance of this model may be considered somehow modest because of the foreign nature of the IGKV3-20 protein into a mouse environment, nonetheless HHD mice proved capable of generating IGKV3-20–specific, HLA-A2–restricted CTLs that, in turn, correctly recognized and lysed the relevant human targets, thus supporting the potential role of vaccination to elicit anti-lymphoma therapeutic T-cell responses. In this regard, coinjection of human IGKV3-20–specific CTLs with IGKV3-20+ or IGKV3-15+ lymphoma cells into SCID mice totally prevented the growth of tumor cells, thus confirming that these effectors mediate efficient and cross-reactive cytotoxic responses also in vivo.

One possible limitation of the use of single-chain, recombinant IGKV3 proteins as “shared” vaccines is related to the loss of the unique B-cell epitopes generated by the association of the variable portions of the heavy and light chains. Although this may prevent the induction of therapeutically relevant anti-Id antibodies (44), single-chain Id proteins may also elicit the generation of antibodies to public determinants conserved in the native Ig. Our results support this possibility, even though repeated immunizations are probably needed to obtain antibodies able to detect the native IGKV3-20 and thus potentially able to mediate clinically relevant responses. Another potential limitation of our “off-the-shelf” vaccine is the possible deletion of a fraction of normal IGKV3+ B lymphocytes. Available evidence, however, indicate that it is unlikely that this could constitute a major concern, as phase II and III trials have convincingly shown that Id vaccines including both heavy and light chains are safe and associated with only modest toxic effects, with no evidence of immune compromise related to depletion of part of B-cell repertoire.

Our findings are consistent with the recent identification of CTL epitopes in clonal Ig light chains, mainly belonging to different IGV lambda gene families (45). Notably, the single IGKV epitope identified was a somatically mutated version of our IGKV3-20 p48 peptide (Supplementary Table S2). The demonstration that several CTL epitopes are carried by a single Id light chain such as IGKV3-20 supports a role for this protein as “off-the-shelf” vaccine for extended subsets of B-cell tumors. Besides overcoming the need for customized vaccine production, the IGKV3-20 vaccine may have the additional immunological advantages to be a carrier of a set of peptides naturally selected for heteroclitic substitutions associated with nearly optimal immunogenic features. This set of T-cell epitopes potentially covers a high proportion of IGKV-restricted B-cell tumors, as at least 1 IGKV3-20 epitope can be found in several light chains from different IGKV families. Moreover, IGKV3-20 also contains promiscuous epitopes, thus assuring a broad HLA coverage. On the whole, these data provide the rationale to activate a phase I/II study aimed at assessing the safety and efficacy of the IGKV3-20 vaccine for patients with lymphomas expressing molecularly correlated Id proteins. Achievement of these goals would make available a ready-to-use recombinant vaccine for improved consolidation therapy of low-grade B-cell lymphomas.

R. Dolcetti is a consultant/advisory board member of Areta Int. No potential conflicts of interest were disclosed by the other authors.

Conception and design: M. Guidoboni, V.D. Re, R. Mortarini, A. Rosato, A. Anichini, R. Dolcetti

Development of methodology: D. Martorelli, M. Guidoboni, V.D. Re, L. Romagnoli, R. Mortarini, D. Gasparotto, M. Mazzucato, A. Rosato, A. Anichini

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): D. Martorelli, V.D. Re, E. Muraro, R. Turrini, A. Merlo, L. Romagnoli, M. Spina, M. Mazzucato, A. Rosato

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): D. Martorelli, M. Guidoboni, V.D. Re, R. Turrini, A. Merlo, R. Mortarini, A. Rosato, A. Anichini

Writing, review, and/or revision of the manuscript: D. Martorelli, M. Guidoboni, V.D. Re, R. Turrini, A. Merlo, M. Spina, R. Mortarini, A. Carbone, A. Rosato, A. Anichini, R. Dolcetti

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): D. Martorelli, V.D. Re, E. Pasini, L. Caggiari, A. Rosato, R. Dolcetti

Study supervision: V.D. Re, A. Rosato, R. Dolcetti

The authors thank T. Ooka (Université Claude Bernard Lyon-1, France) for the kind gift of recombinant p29 BARF1 protein, A. Molla and C. Vegetti (Fondazione IRCCS, INT Milan) for technical support, and Miss S. Colussi (CRO-IRCCS, Aviano) for the editing.

The study was supported by grants from the European Community (FP6 037874), the Italian Ministry of Health (ACC-4), the Associazione Italiana per la Ricerca sul Cancro (to R. Dolcetti, A. Anichini, R. Mortarini), and the Fondazione Federica per la Vita. Elisa Pasini is a FIRC fellow.

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.

1.
Park
HJ
,
Neelapu
SS
. 
Developing idiotype vaccines for lymphoma: from preclinical studies to phase III clinical trials
.
Br J Haematol
2008
;
142
:
179
91
.
2.
Houot
R
,
Levy
R
. 
Vaccines for lymphomas: idiotype vaccines and beyond
.
Blood Rev
2009
;
23
:
137
42
.
3.
Hsu
FJ
,
Benike
C
,
Fagnoni
F
,
Liles
TM
,
Czerwinski
D
,
Taidi
B
, et al
Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells
.
Nat Med
1996
;
2
:
52
8
.
4.
Nelson
EL
,
Li
X
,
Hsu
FJ
,
Kwak
LW
,
Levy
R
,
Clayberger
C
, et al
Tumor-specific, cytotoxic T-lymphocyte response after idiotype vaccination for B-cell, non-Hodgkin's lymphoma
.
Blood
1996
;
88
:
580
9
.
5.
Bendandi
M
,
Gocke
CD
,
Kobrin
CB
,
Benko
FA
,
Sternas
LA
,
Pennington
R
, et al
Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma
.
Nat Med
1999
;
5
:
1171
7
.
6.
Timmerman
JM
,
Czerwinski
DK
,
Davis
TA
,
Hsu
FJ
,
Benike
C
,
Hao
ZM
, et al
Idiotype-pulsed dendritic cell vaccination for B-cell lymphoma: clinical and immune responses in 35 patients
.
Blood
2002
;
99
:
1517
26
.
7.
Weng
WK
,
Czerwinski
D
,
Timmerman
J
,
Hsu
FJ
,
Levy
R
. 
Clinical outcome of lymphoma patients after idiotype vaccination is correlated with humoral immune response and immunoglobulin G Fc receptor genotype
.
J Clin Oncol
2004
;
22
:
4717
24
.
8.
Inoges
S
,
Rodriguez-Calvillo
M
,
Zabalegui
N
,
Lopez-Diaz de
CA
,
Villanueva
H
,
Soria
E
, et al
Clinical benefit associated with idiotypic vaccination in patients with follicular lymphoma
.
J Natl Cancer Inst
2006
;
98
:
1292
301
.
9.
Schuster
SJ
,
Neelapu
SS
,
Gause
BL
,
Janik
JE
,
Muggia
FM
,
Gockerman
JP
, et al
Vaccination with patient-specific tumor-derived antigen in first remission improves disease-free survival in follicular lymphoma
.
J Clin Oncol
2011
;
29
:
2787
94
.
10.
Houot
R
,
Levy
R
. 
Idiotype vaccination for lymphoma: moving towards optimisation
.
Leuk Lymphoma
2009
;
50
:
1
2
.
11.
De Re
V
,
De Vita
S
,
Marzotto
A
,
Rupolo
M
,
Gloghini
A
,
Pivetta
B
, et al
Sequence analysis of the immunoglobulin antigen receptor of hepatitis C virus-associated non-Hodgkin lymphomas suggests that the malignant cells are derived from the rheumatoid factor-producing cells that occur mainly in type II cryoglobulinemia
.
Blood
2000
;
96
:
3578
84
.
12.
Chan
CH
,
Hadlock
KG
,
Foung
SK
,
Levy
S
. 
V(H)1-69 gene is preferentially used by hepatitis C virus-associated B cell lymphomas and by normal B cells responding to the E2 viral antigen
.
Blood
2001
;
97
:
1023
6
.
13.
De Re
V
,
De Vita
S
,
Gasparotto
D
,
Marzotto
A
,
Carbone
A
,
Ferraccioli
G
, et al
Salivary gland B cell lymphoproliferative disorders in Sjogren's syndrome present a restricted use of antigen receptor gene segments similar to those used by hepatitis C virus-associated non-Hodgkins's lymphomas
.
Eur J Immunol
2002
;
32
:
903
10
.
14.
Ivanovski
M
,
Silvestri
F
,
Pozzato
G
,
Anand
S
,
Mazzaro
C
,
Burrone
OR
, et al
Somatic hypermutation, clonal diversity, and preferential expression of the VH 51p1/VL kv325 immunoglobulin gene combination in hepatitis C virus-associated immunocytomas
.
Blood
1998
;
91
:
2433
42
.
15.
Kuppers
R
,
Rajewsky
K
,
Hansmann
ML
. 
Diffuse large cell lymphomas are derived from mature B cells carrying V region genes with a high load of somatic mutation and evidence of selection for antibody expression
.
Eur J Immunol
1997
;
27
:
1398
405
.
16.
Bende
RJ
,
Aarts
WM
,
Riedl
RG
,
de
JD
,
Pals
ST
,
van Noesel
CJ
. 
Among B cell non-Hodgkin's lymphomas, MALT lymphomas express a unique antibody repertoire with frequent rheumatoid factor reactivity
.
J Exp Med
2005
;
201
:
1229
41
.
17.
Craig
VJ
,
Arnold
I
,
Gerke
C
,
Huynh
MQ
,
Wundisch
T
,
Neubauer
A
, et al
Gastric MALT lymphoma B cells express polyreactive, somatically mutated immunoglobulins
.
Blood
2010
;
115
:
581
91
.
18.
Smilevska
T
,
Tsakou
E
,
Hadzidimitriou
A
,
Bikos
V
,
Stavroyianni
N
,
Laoutaris
N
, et al
Immunoglobulin kappa gene repertoire and somatic hypermutation patterns in follicular lymphoma
.
Blood Cells Mol Dis
2008
;
41
:
215
8
.
19.
Tobin
G
,
Thunberg
U
,
Karlsson
K
,
Murray
F
,
Laurell
A
,
Willander
K
, et al
Subsets with restricted immunoglobulin gene rearrangement features indicate a role for antigen selection in the development of chronic lymphocytic leukemia
.
Blood
2004
;
104
:
2879
85
.
20.
Stamatopoulos
K
,
Belessi
C
,
Hadzidimitriou
A
,
Smilevska
T
,
Kalagiakou
E
,
Hatzi
K
, et al
Immunoglobulin light chain repertoire in chronic lymphocytic leukemia
.
Blood
2005
;
106
:
3575
83
.
21.
De Re
V
,
Simula
MP
,
Pavan
A
,
Garziera
M
,
Marin
D
,
Dolcetti
R
, et al
Characterization of antibodies directed against the immunoglobulin light kappa chain variable chain region (VK) of hepatitis C virus-related type-II mixed cryoglobulinemia and B-cell proliferations
.
Ann N Y Acad Sci
2009
;
1173
:
152
60
.
22.
De Re
V
,
De Vita
S
,
Sansonno
D
,
Gasparotto
D
,
Simula
MP
,
Tucci
FA
, et al
Type II mixed cryoglobulinaemia as an oligo rather than a mono B-cell disorder: evidence from GeneScan and MALDI-TOF analyses
.
Rheumatology (Oxford)
2006
;
45
:
685
93
.
23.
Martorelli
D
,
Houali
K
,
Caggiari
L
,
Vaccher
E
,
Barzan
L
,
Franchin
G
, et al
Spontaneous T cell responses to Epstein-Barr virus-encoded BARF1 protein and derived peptides in patients with nasopharyngeal carcinoma: bases for improved immunotherapy
.
Int J Cancer
2008
;
123
:
1100
7
.
24.
Sallusto
F
,
Lanzavecchia
A
. 
The instructive role of dendritic cells on T-cell responses
.
Arthritis Res
2002
;
4
Suppl 3
:
S127
32
.
25.
Russo
C
,
Ng
AK
,
Pellegrino
MA
,
Ferrone
S
. 
The monoclonal antibody CR11-351 discriminates HLA-A2 variants identified by T cells
.
Immunogenetics
1983
;
18
:
23
35
.
26.
Wulf
M
,
Hoehn
P
,
Trinder
P
. 
Identification of human MHC class I binding peptides using the iT
.
Methods Mol Biol
2009
;
524
:
361
7
.
27.
Shingler
WH
,
Chikoti
P
,
Kingsman
SM
,
Harrop
R
. 
Identification and functional validation of MHC class I epitopes in the tumor-associated antigen 5T4
.
Int Immunol
2008
;
20
:
1057
66
.
28.
Pascolo
S
,
Bervas
N
,
Ure
JM
,
Smith
AG
,
Lemonnier
FA
,
Perarnau
B
. 
HLA-A2.1-restricted education and cytolytic activity of CD8(+) T lymphocytes from beta2 microglobulin (beta2m) HLA-A2.1 monochain transgenic H-2Db beta2m double knockout mice
.
J Exp Med
1997
;
185
:
2043
51
.
29.
Tomayko
MM
,
Reynolds
CP
. 
Determination of subcutaneous tumor size in athymic (nude) mice
.
Cancer Chemother Pharmacol
1989
;
24
:
148
54
.
30.
Trojan
A
,
Schultze
JL
,
Witzens
M
,
Vonderheide
RH
,
Ladetto
M
,
Donovan
JW
, et al
Immunoglobulin framework-derived peptides function as cytotoxic T-cell epitopes commonly expressed in B-cell malignancies
.
Nat Med
2000
;
6
:
667
72
.
31.
Katsibardi
K
,
Braoudaki
M
,
Papathanasiou
C
,
Karamolegou
K
,
Tzortzatou-Stathopoulou
F
. 
Clinical significance of productive immunoglobulin heavy chain gene rearrangements in childhood acute lymphoblastic leukemia
.
Leuk Lymphoma
2011
;
52
:
1751
7
.
32.
Falk
K
,
Rotzschke
O
,
Stevanovic
S
,
Jung
G
,
Rammensee
HG
. 
Allele-specific motifs revealed by sequencing of self-peptides eluted from MHC molecules
.
Nature
1991
;
351
:
290
6
.
33.
Matsumura
M
,
Fremont
DH
,
Peterson
PA
,
Wilson
IA
. 
Emerging principles for the recognition of peptide antigens by MHC class I molecules
.
Science
1992
;
257
:
927
34
.
34.
Sabbaj
S
,
Bansal
A
,
Ritter
GD
,
Perkins
C
,
Edwards
BH
,
Gough
E
, et al
Cross-reactive CD8+ T cell epitopes identified in US adolescent minorities
.
J Acquir Immune Defic Syndr
2003
;
33
:
426
38
.
35.
Masemola
AM
,
Mashishi
TN
,
Khoury
G
,
Bredell
H
,
Paximadis
M
,
Mathebula
T
, et al
Novel and promiscuous CTL epitopes in conserved regions of Gag targeted by individuals with early subtype C HIV type 1 infection from southern Africa
.
J Immunol
2004
;
173
:
4607
17
.
36.
Threlkeld
SC
,
Wentworth
PA
,
Kalams
SA
,
Wilkes
BM
,
Ruhl
DJ
,
Keogh
E
, et al
Degenerate and promiscuous recognition by CTL of peptides presented by the MHC class I A3-like superfamily: implications for vaccine development
.
J Immunol
1997
;
159
:
1648
57
.
37.
Burrows
SR
,
Elkington
RA
,
Miles
JJ
,
Green
KJ
,
Walker
S
,
Haryana
SM
, et al
Promiscuous CTL recognition of viral epitopes on multiple human leukocyte antigens: biological validation of the proposed HLA A24 supertype
.
J Immunol
2003
;
171
:
1407
12
.
38.
Frahm
N
,
Yusim
K
,
Suscovich
TJ
,
Adams
S
,
Sidney
J
,
Hraber
P
, et al
Extensive HLA class I allele promiscuity among viral CTL epitopes
.
Eur J Immunol
2007
;
37
:
2419
33
.
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
1991
;
147
:
1392
7
.
40.
Dyall
R
,
Bowne
WB
,
Weber
LW
,
LeMaoult
J
,
Szabo
P
,
Moroi
Y
, et al
Heteroclitic immunization induces tumor immunity
.
J Exp Med
1998
;
188
:
1553
61
.
41.
Rosenberg
SA
,
Yang
JC
,
Schwartzentruber
DJ
,
Hwu
P
,
Marincola
FM
,
Topalian
SL
, et al
Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma
.
Nat Med
1998
;
4
:
321
7
.
42.
Harig
S
,
Witzens
M
,
Krackhardt
AM
,
Trojan
A
,
Barrett
P
,
Broderick
R
, et al
Induction of cytotoxic T-cell responses against immunoglobulin V region-derived peptides modified at human leukocyte antigen-A2 binding residues
.
Blood
2001
;
98
:
2999
3005
.
43.
Rosenberg
SA
,
Yang
JC
,
Restifo
NP
. 
Cancer immunotherapy: moving beyond current vaccines
.
Nat Med
2004
;
10
:
909
15
.
44.
Ai
WZ
,
Tibshirani
R
,
Taidi
B
,
Czerwinski
D
,
Levy
R
. 
Anti-idiotype antibody response after vaccination correlates with better overall survival in follicular lymphoma
.
Blood
2009
;
113
:
5743
6
.
45.
Weng
J
,
Cha
SC
,
Matsueda
S
,
Alatrash
G
,
Popescu
MS
,
Yi
Q
, et al
Targeting human B-cell malignancies through Ig light chain-specific cytotoxic T lymphocytes
.
Clin Cancer Res
2011
;
17
:
5945
52
.