Chronic lymphocytic leukemia (CLL) is associated with a variety of immunologic disturbances. Hypogammaglobulinemia and autoimmune phenomena are both often present in this disease. In contrast, humoral or cellular antitumor responses are rarely observed. It has been previously shown that antigens detected in patients with malignant diseases can provide information regarding intracellular molecules engaged in the transformation process and can identify tumor antigens that may be useful for development of immunotherapeutic strategies. Serologic identification by recombinant expression cloning (SEREX) has been demonstrated to be a useful method to detect tumor and tumor-associated antigens in a variety of malignancies. Although this approach is complicated in CLL, we used a modified SEREX approach and identified 14 antigens (KW-1 to KW-14) using this methodology. Several clones showed a restricted expression pattern in normal tissues. Moreover, distinctive expression of splice variants and aberrant gene expression in malignant tissue were detected. In this study, 6 antigens were detected exclusively in patients with CLL. Eight antigens were detected also in lymphoma patients. Healthy donors showed antibody responses against only 3 of the identified antigens. T cells with specific cytotoxicity against peptides derived from the 2 antigens tested could be generated from healthy donors. These findings demonstrate that humoral and cellular immune responses against CLL-associated antigens can be detected. Ongoing experiments investigate their potential for the development of immunotherapeutic strategies.

Chronic lymphocytic leukemia (CLL) is the most common leukemia in Western countries. It is increasing in frequency and is being increasingly diagnosed in younger patients. CLL remains incurable using currently available therapies, and younger patients almost invariably die of their disease.1,2 There is therefore a need for better understanding of the disease pathogenesis as well as a need for novel treatment strategies. CLL is in many regards an ideal candidate to study novel immunotherapeutic approaches. The disease has a long natural course, which would allow development of immune responses against the tumor cells, and most patients with CLL are advanced in age and therefore not suitable for aggressive chemotherapeutic regimens. Recent advances in the use of donor lymphocytes following allogeneic stem cell transplantation and the use of nonmyeloablative conditioning regimens have illustrated the existence of a graft-versus-leukemia effect in this disease, demonstrating that the disease can be a target of an ongoing T-cell–mediated immune response.3 However, there are several hurdles to develop immunotherapy in CLL. Patients with CLL often suffer from a variety of immune dysregulations and immunosuppression that can be worsened by conventional therapy with purine analogs and alkylating agents. Although autologous T-cell responses can be generated against the patient's own CLL cells using immunoglobulin (Ig)–derived peptides as target antigens,4,5 Ig-derived vaccine approaches have had limited clinical application. In addition, autologous cytotoxic immune responses are weak and only generated in a minority of patients. Thus, new tumor antigens are required that might represent potential targets for an effective antitumor immune response in CLL and that might also give a broader basis for polyvalent immunization strategies to prevent the escape of antigen loss variants.6 

Ideal tumor antigen candidates should be selectively expressed in tumor tissue and detectable in most patients. However, most tumor antigens do not have these ideal characteristics and might be limited in their use by specific but low expression in tumor cells, by low antigenicity, or by broader expression in healthy tissue. Cancer-testis antigens such as NY-ESO-1 are expressed only in tumor cells and testis, making them among the more ideal candidates for immunotherapeutic approaches.7 However, clinical studies targeting these antigens are limited to date.8 Most antigens have broader expression in healthy tissue. The risk of using these antigens as targets of an immune response is induction of autoimmune phenomena. Although this risk must be taken very seriously, several animal studies have shown that there might be a difference in the susceptibility of normal and tumor tissue to the effector arms of the immune response.9-11 Antibody therapy with Her2/neu, which has a broader expression profile, has been demonstrated to increase remissions in patients with breast cancer, whereas no major autoimmune phenomena have been observed so far.12,13 

Many tumor antigens have been identified either by cellular and/or humoral responses. SEREX is an approach to detect novel tumor antigens serologically.14 This method is based on the detection of antigens within recombinantly expressed phage libraries by autologous antibodies. It has been used successfully to identify new tumor antigens in several malignancies.7,15-25 There are several obstacles to the use of SEREX in CLL. Many patients with CLL suffer from hypogammaglobulinemia and other immune defects, aggravating the detection of serologic immune responses against potential tumor antigens in these patients. Patients with CLL often have autoantibodies against normal cellular proteins, so that antigens detected by SEREX might represent normal cellular components and not be suitable for developing immunotherapeutic strategies in this disease. In addition, CLL cells express immunoglobulin (Ig). Thus, false-positive results will arise by binding the secondary antibody to Ig expressed by the transfected Escherichia coli. However, we were largely able to overcome these obstacles. Fourteen antigens were identified following recognition by autologous and allogeneic sera of CLL patients. Many of the antigens described here are expressed in a variety of normal tissues. However, some show enhanced expression in hematopoietic cells, tumor samples, and testis. All antigens have been previously described as genes; however, in 6 of them protein function has not investigated experimentally. Of note, in several cases novel splice variants have been detected. Seven of the antigens are novel, whereas 7 have been previously described, 5 of them using SEREX in other malignancies. Six antigens have been exclusively recognized by patients with CLL, whereas 8 have been identified also by patients with other lymphoma. Sera from healthy donors were reactive in some cases against only 3 antigens. This study therefore identifies tumor antigen candidates in CLL that might be useful for the development of immunotherapeutic strategies in this disease.

Patient samples

Cell and serum samples were collected with the informed consent of patients and healthy donors and after receiving institutional review board approval from Dana-Farber Cancer Institute and Brigham and Women's Hospital. All patients had diagnosis of CLL confirmed by morphology and flow cytometric analysis. CLL cells and sera were simultaneously collected by leukapheresis performed on 5 different CLL patients who had not been previously treated. These 5 patients were selected as representative of the CLL patient population. Three patients had early-stage disease, and 2 patients had more advanced diseases (Table 1). Purified CLL cells (> 95%) were freshly used to create cDNA libraries, and excess cells were cryopreserved. Additional serum samples were obtained during routine diagnostic procedures from 25 patients with CLL and 5 patients with other lymphomas (2 follicular lymphomas, 1 mantle cell lymphoma, 1 diffuse large cell lymphoma, and 1 Hodgkin lymphoma). Normal sera were collected from 14 healthy donors. All serum samples were processed in exactly the same way. Tumor cells and patient sera were isolated by Ficoll-Hypaque (Amersham, Uppsala, Sweden) density-gradient centrifugation immediately after collection. Sera were stored at −80°C until use.

Table 1.

Clinical characteristics of patients included for generation of CLL-cell cDNA libraries

PatientAge, ySexStageTime from diagnosis to collection, yPrior treatmentWhite blood count, × 109/LAutoimmune phenomenaIsolated antigens
61 Rai I/BinetA None 48 None 
45 Rai II/BinetA None 72 None 
50 Rai II/BinetA None 28 None 
46 Rai II/BinetB None 54 None 
50 Rai IV/BinetC None 466 AIHA, ITP 
PatientAge, ySexStageTime from diagnosis to collection, yPrior treatmentWhite blood count, × 109/LAutoimmune phenomenaIsolated antigens
61 Rai I/BinetA None 48 None 
45 Rai II/BinetA None 72 None 
50 Rai II/BinetA None 28 None 
46 Rai II/BinetB None 54 None 
50 Rai IV/BinetC None 466 AIHA, ITP 

Purified CLL cells were used for generation of 5 different cDNA expression libraries. Patient characteristics, clinical data, and number of isolated clones are shown. One antigen, KW-5, was selected independently from 2 different libraries.

AIHA indicates autoimmune hemolytic anemia; ITP, idiopathic thrombocytopenic purpura.

RNA extraction and polyA-RNA enrichment

Total RNA was extracted from tumor cells using standard methodology according to the manufacturer's recommendation (Trizol reagent, Life Technologies, Rockville, MD). PolyA-RNA was enriched from total RNA using the Messagemaker reagent assembly kit (Life Technologies). Quality of total RNA and polyA-RNA was assessed using formaldehyde gels.

Construction of cDNA expression libraries

The cDNA expression libraries were constructed from 5 μg polyA-RNA. First-strand cDNA synthesis was performed using an oligo(dT) primer that contained an internal XhoI restriction site. The cDNA was ligated to EcoRI adaptors and digested withXhoI before being size selected, cloned directionally into ZAP expression vector (Stratagene, La Jolla, CA), packaged into phage particles, and transfected into XL1-blue E coli. Libraries containing between 6.2 × 106 and 12.8 × 107 recombinants were amplified and used for immunoscreening.

Immunoscreening of the cDNA libraries

The cDNA libraries were screened with autologous and allogeneic sera in a modification of that previously described.24Serum was diluted 1:10, preabsorbed with transfected E colilysate, and further diluted 1:20 (final serum dilution 1:200). E coli transfected with recombinant λZAPII phages were plated on agar plates. After emergence of visible plaques, nitrocellulose filters, soaked in isopropyl β-d-thiogalactosidase (IPTG) for protein induction, were placed on plates and cultured for 3 hours. After colony lifting, filters were washed in TBST (50 mM Tris [tris(hydroxymethyl)aminomethane], 138 mM NaCl, 2.7 mM KCl, 0.05% wt/vol Tween 20, pH 8.0) and blocked overnight with 1% wt/vol nonfat dry milk in triethanolamine-buffered saline (TBS). The next day the filters were washed and then incubated with alkaline phosphatase–conjugated goat antihuman Fc secondary antibodies (Jackson ImmunoResearch Labs, West Grove, PA), and the reactive phage plaques were visualized by incubating with 5-bromo-4-chloro-3-indolyl-phosphate and nitroblue tetrazolium (Promega, Madison, WI). False-positive clones were not further processed. After prescreening, nitrocellulose membranes were washed again and then incubated with the preabsorbed sera overnight at 4°C. Serum antibodies binding to recombinant proteins expressed in lytic plaques were detected by repeated incubation with the alkaline phosphatase–conjugated goat anti–human IgG antibodies (Jackson ImmunoResearch Labs) and visualization with 5-bromo-4-chloro-3-indilyl phosphate and nitroblue tetrazolium (Promega).

Sequence analysis of the reactive clones

The clones reactive with the sera but not by incubation with secondary antibody alone were subcloned for isolation, purified, and the phagemid cDNA insert excised from the λZAP vector with an ExAssist helper phage system (Stratagene, La Jolla, CA). Plasmid DNA was prepared using the Quiaprep Spin Miniprep kit (Qiagen, Hilden, Germany). Clones were sequenced by the molecular biology core facility at the Dana Farber Cancer Institute using an ABI Prism 373 DNA sequencer (Applied Biosystems, Foster City, CA). Sequences were further analyzed for homology with known genes and proteins using public databases.

RACE-PCR for complete cloning of KW-13

To complete the 5′-sequence of clone KW-13, the RACE-PCR (rapid amplification of cDNA ends–polymerase chain reaction) approach was performed using the SMART RACE cDNA amplification kit (Clontech, Palo Alto, CA) according to the manufacturer's recommendations. The antisense primer sequence was CCGTAGCACC TGCGTGGACT CCTGAACTCG C.

RT-PCR

To evaluate the mRNA expression pattern of the cloned cDNA in normal and malignant tissues, exon-overlapping oligonucleotide primers for PCR were designed to amplify cDNA segments. All primers were commercially synthesized (Life Technologies; Invitrogen, Carlsbad, CA). Reverse transcription (RT)–PCR was performed using 30 amplification cycles in a thermal cycler (Perkin-Elmer, Shelton, CT) at primer-specific annealing temperatures. PCR products were analyzed on ethidium bromide agarose gels, and cDNA panels of healthy and malignant tissues were purchased from Clontech.

Northern blot analysis

Northern blot analysis was performed using standard techniques. Briefly, 10 μg total RNA per lane was dissolved in loading buffer containing formamide and formaldehyde, heated at 65°C, separated on a 1% agarose gel with 16% formaldehyde, and transferred to a nitrocellulose membrane. Subsequent hybridization to the32P-labeled probe (NEN, Boston, MA) and washing were performed. RNA integrity of the membranes was determined using a probe for the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) transcript, and mRNA blots from healthy and malignant tissue were purchased from Clontech.

Phage plaque assay

To investigate the antigenicity of detected clones in CLL patients or other malignancies compared with healthy donors, a phage plaque assay was performed. Phages from positive clones were mixed with nonreactive phages of the cDNA library as internal negative controls and used to transfect XL1-blue E coli. IgG antibodies in 1:200 diluted E coli absorbed sera from patients with CLL, other lymphoma, and healthy donors were detected with the immunoscreening assay as described above.

Statistics

The number of detected clones in different patients was correlated with their clinical data (therapy, presence of hypogammaglobulinemia, disease stage, presence of autoimmune phenomena, CD38 expression on CLL cells, and Ig mutational status) using the Fisher exact test. In addition, detection of clones by CLL patients compared with healthy donors was analyzed by the Fisher exact test.

Peptide prediction analysis and bioinformatics

In the case of KW-2 and KW-13, clone-specific sequences were reviewed for peptides 9 and 10 amino acids in length that could potentially bind to major histocompatibility complex (MHC) class I molecules. Two independent computer prediction analyses were used (http://bimas.dcrt.nih.gov/molbio/hla_bind andhttp://syfpeithi.bmi-heidelberg.com) to examine the binding of native peptides to HLA-A*0201 molecules. Analysis was performed for HLA-A*0201 because this MHC class I allele is expressed in approximately 50% of our patients.

Peptide synthesis

Peptides deduced from the amino acid sequence of KW-2 and KW-13 were generated by fluorenylmethoxycarbonyl (Fmoc) synthesis (Sigma-Genosys Biotechnologies, The Woodlands, TX). Purity was determined by reverse phase high-performance liquid chromatography (HPLC) and verified by mass spectral analysis. Peptides were dissolved in PBS at a concentration of 2 mg/mL and stored at −70°C until use.

Screening for peptide-specific HLA-restricted cytotoxic T cells

For screening peptide-specific cytotoxic T-lymphocyte (CTL) responses, we used a previously established system.4,26 Briefly, purified HLA-A2 plus CD8 plus T cells of healthy donors were primarily stimulated with irradiated monocyte-derived peptide-pulsed dendritic cells and restimulated weekly with irradiated, peptide-pulsed CD40-activated B cells.27The cytotoxic activity of CTL lines was assessed after 4 to 5 stimulations using peptide-pulsed CD40-activated B cells, control-peptide pulsed CD40-activated B cells, and K562 cells in chromium (51Cr)–release assays. The concentration of peptide used for pulsing of the CD-40–activated B cells resulted in maximal killing of the targets. Specific lysis is presented as the percentage of specific 51Cr release, calculated from the following formula: (E − S/T − S) × 100, where E is experimental51Cr release, S is the spontaneous 51Cr release, and T is the total 51Cr release by 2% Triton X-100.

Isolation of immunoreactive clones from CLL cell libraries by SEREX

We generated cDNA libraries from CLL cells isolated from 5 patients with different stages of CLL (Table 1). Using the sera from patients with CLL, other lymphomas, and healthy donors, we identified 24 independent clones representing 14 different genes (KW-1 to KW-14) after screening of more than 30 × 106 clones using the SEREX methodology (Table 2). All clones have high homologies to previously described genes (Table3).28-39 However, in 6 of them, the protein function has not been investigated experimentally.

Table 2.

Identification of 14 CLL-associated antigens by SEREX

ClonesAccession no.No. of selected clonesSize, bp
KW-1 AF432208/AF432209 665/1342 
KW-2 AF432215 3440  
KW-3 AF432216 2008 
KW-4 AF432217 3240  
KW-5 AF432218 2376 
KW-6 AF432219 2800  
KW-7 AF432220 3076 
KW-8 AF432221 1280  
KW-9 AF432222 2100 
KW-10 AF432210 2952  
KW-11 AF432211 1380 
KW-12 AF432212 579  
KW-13 AF432213 3435 
KW-14 AF432214 2593 
ClonesAccession no.No. of selected clonesSize, bp
KW-1 AF432208/AF432209 665/1342 
KW-2 AF432215 3440  
KW-3 AF432216 2008 
KW-4 AF432217 3240  
KW-5 AF432218 2376 
KW-6 AF432219 2800  
KW-7 AF432220 3076 
KW-8 AF432221 1280  
KW-9 AF432222 2100 
KW-10 AF432210 2952  
KW-11 AF432211 1380 
KW-12 AF432212 579  
KW-13 AF432213 3435 
KW-14 AF432214 2593 
Table 3.

CLL-associated antigens have high homologies to previously described genes

ClonesHomology to known genesAccession no.Chromosomal localizationFamily motifs/domains(Suggested) function
KW-1 KIAA1641 XM047794 2q11.2 bZIP, DUF 164 (DNA binding/transcription regulation) 
KW-2 PIPMT AY028423 8q12.1 Met-10-like protein, putative RNA-methylase Methylase 
KW-3 FosB NM006732 19q13.2 bZIP DNA binding/transcription regulation 
KW-4 ZNF268 AF317549 12q24.34 Zinc finger (DNA binding/transcription regulation) 
KW-5 SEBD4 X75314 20q13.2 RNA recognition motif (RNA binding)  
KW-6 Ikaros NM006060 7p11.2 Zinc finger DNA binding/transcription regulation 
KW-7 p75/LDEGF NM021144 9p22.3 AT-hook/PWWP DNA binding/transcription regulation/growth factor 
KW-8 CHIP NM005861 16p13.3 Tetratricopeptide repeat region Proteosomal induction 
KW-9 PYGB NM002862 20p11.21 Brain-type glycogen phosphorylase Carbohydrate phosphorylase 
KW-10 ZNF148 NM021964 3q21.2 Zinc finger DNA binding/transcription regulation 
KW-11 KIAA0336 XM017779 2q12.3 Translin, GBP, bZIP, lipoprotein, GRIP (DNA binding/transcription regulation) 
KW-12 RPL11 NM000975 1p36.11 RPL-5 ribosomal L5-C terminus Ribosomal protein 
KW-13 FMNL XM038679 17q21.31 FH1, FH2, FH3 (Formin-like protein) 
KW-14 HGRG8 NM016258 1p35.3 No known family motifs present (Chaperone) 
ClonesHomology to known genesAccession no.Chromosomal localizationFamily motifs/domains(Suggested) function
KW-1 KIAA1641 XM047794 2q11.2 bZIP, DUF 164 (DNA binding/transcription regulation) 
KW-2 PIPMT AY028423 8q12.1 Met-10-like protein, putative RNA-methylase Methylase 
KW-3 FosB NM006732 19q13.2 bZIP DNA binding/transcription regulation 
KW-4 ZNF268 AF317549 12q24.34 Zinc finger (DNA binding/transcription regulation) 
KW-5 SEBD4 X75314 20q13.2 RNA recognition motif (RNA binding)  
KW-6 Ikaros NM006060 7p11.2 Zinc finger DNA binding/transcription regulation 
KW-7 p75/LDEGF NM021144 9p22.3 AT-hook/PWWP DNA binding/transcription regulation/growth factor 
KW-8 CHIP NM005861 16p13.3 Tetratricopeptide repeat region Proteosomal induction 
KW-9 PYGB NM002862 20p11.21 Brain-type glycogen phosphorylase Carbohydrate phosphorylase 
KW-10 ZNF148 NM021964 3q21.2 Zinc finger DNA binding/transcription regulation 
KW-11 KIAA0336 XM017779 2q12.3 Translin, GBP, bZIP, lipoprotein, GRIP (DNA binding/transcription regulation) 
KW-12 RPL11 NM000975 1p36.11 RPL-5 ribosomal L5-C terminus Ribosomal protein 
KW-13 FMNL XM038679 17q21.31 FH1, FH2, FH3 (Formin-like protein) 
KW-14 HGRG8 NM016258 1p35.3 No known family motifs present (Chaperone) 

Detection of genetic diversity by novel open reading frames, splice variants, and possible mutations in detected clones

Sequence analysis of the detected clones KW-1, KW-4, KW-6, KW-11, KW-13, and KW-14 identified novel splice variants that have not previously been described (Table4).

Table 4.

Identification of novel splice variants in CLL-associated antigens

CloneGeneSuggested splice variant-specific protein alteration
KW-1 KIAA1641 Deletion of an uncharacterized protein motif (DUF164) 
KW-4 ZNF286 Exclusion of the transcription-repressing KRAB domain  
KW-6 Ikaros Loss of 10 aa at the activation site of Ikaros  
KW-11 KIAA0336 Additional exon codes for a GTP binding site  
KW-13 FMNL Full-length cloning of the gene shows 3 formin motifs (FH1, FH2, FH3) as well as 2 splice variants varying in 30 bp at the 5′ end 
KW-14 HGRG8 Additional exon with unknown protein function 
CloneGeneSuggested splice variant-specific protein alteration
KW-1 KIAA1641 Deletion of an uncharacterized protein motif (DUF164) 
KW-4 ZNF286 Exclusion of the transcription-repressing KRAB domain  
KW-6 Ikaros Loss of 10 aa at the activation site of Ikaros  
KW-11 KIAA0336 Additional exon codes for a GTP binding site  
KW-13 FMNL Full-length cloning of the gene shows 3 formin motifs (FH1, FH2, FH3) as well as 2 splice variants varying in 30 bp at the 5′ end 
KW-14 HGRG8 Additional exon with unknown protein function 

Detection of novel splice variants by sequencing positive clones detected by SEREX; in silico analysis of the protein sequence suggests alterations of the protein function.

Of note, most of these splice variants suggest alterations of protein function. KW-4 was found by sequencing of 2 independent clones. These clones represent a Kruppel-like zinc finger protein.30 Splice variant 2 represents a previously undescribed splice variant missing the Kruppel-associated box (KRAB) domain. KW-6 represents the 3′ end of Ikaros mRNA within the open reading frame (ORF) containing part of exon 6 and the whole exon 7.40 The mRNA lacks 30 amino acids (aa) of the previously described Ikaros mRNAs, resulting in a loss of 9 aa at the end of exon 6 and the first aa of exon 7, where the activation site starts. KW-11 has been previously described as KIAA0336.61The protein function of the KIAA0336 gene is not known. The aa sequence codes for many protein family motifs including several bZIP domains, suggesting a function as a transcription factor. Our clone codes for a previously undescribed splice variant, including an additional exon extending the protein at the 5′ end. This splice variant introduces a motif homologous to the C-terminal domain of the guanylate-binding protein GBP141 and gives additional homology to Translin.42 KW-13 has 99% homology to the previously described human Formin-like (FMNL) mRNA or C17ORF1, containing an FH2 domain.43 However, this mRNA contains only 1789 bp. Our clone contains 3435 bp with an open reading frame of 2976 bp. The 5′ end has been determined by RACE-PCR and shows an ORF of 3177 bp with an amino acid sequence of 1059 aa (Figure1). This protein has 85.4% homology to the mouse FRL (formin-related gene in leukocytes).38 In consensus with the mouse protein it shows an FH1 and FH3 domain in addition to the FH2 domain, suggesting similar functional properties as the mouse homolog.

Fig. 1.

Full-length amino acid sequence of the gene corresponding to KW-13.

Amino acid sequence of the gene corresponding to KW-13 with homology to the murine FRL is shown. The FH3-homolog region is double underlined, the FH1-homolog region is dot-dot-dash underlined, and the FH2-homolog region is single underlined. Peptides inducing peptide-specific cytotoxic immune responses are in squares. The methionine showing the starting sequence of the previously described human FMNL is underlined in bold.

Fig. 1.

Full-length amino acid sequence of the gene corresponding to KW-13.

Amino acid sequence of the gene corresponding to KW-13 with homology to the murine FRL is shown. The FH3-homolog region is double underlined, the FH1-homolog region is dot-dot-dash underlined, and the FH2-homolog region is single underlined. Peptides inducing peptide-specific cytotoxic immune responses are in squares. The methionine showing the starting sequence of the previously described human FMNL is underlined in bold.

Close modal

Expression of identified antigens in healthy and malignant tissue

Expression of detected antigens in healthy and malignant tissue was investigated using RT-PCR. Exon-overlapping primer pairs were constructed to avoid amplification of genomic DNA. By RT-PCR, several genes demonstrated a wide tissue expression. However, 6 antigens (KW-2, KW-3, KW-4, KW-6, KW-7, KW-13) had restricted tissue expression and could not be demonstrated in several different healthy tissues by RT-PCR. For quantitative expression we performed Northern blot analysis. KW-2 has a cancer-testis–like expression profile. It has low expression in most tissues with the exception of testis. It is highly expressed in lymphoblastic leukemia MOLT-4, Raji cells, and the melanoma cell line G361 (Figure 2). We observed low levels of expression in 3 of 6 CLL samples examined but did not detect any expression in normal tonsilar B cells (data not shown).

Fig. 2.

Northern blot analysis of gene expression for the antigen KW-2 and GAPDH.

Normal tissue (A) and tumor tissue (B).

Fig. 2.

Northern blot analysis of gene expression for the antigen KW-2 and GAPDH.

Normal tissue (A) and tumor tissue (B).

Close modal

The splice variant of KW-4, including the KRAB domain (splice variant number 1), is expressed in most normal tissues, whereas splice variant number 2, which is missing the KRAB domain, is not expressed in most normal tissue but is expressed in all CLL cases tested by RT-PCR (Figure 3). It also shows expression in healthy peripheral blood mononuclear cells by RT-PCR (data not shown).

Fig. 3.

Tissue expression of different splice variants of KW-4 by RT-PCR.

RT-PCR with splice variant–specific primers for KW-4: several normal tissues as well as 8 different CLL samples are shown.

Fig. 3.

Tissue expression of different splice variants of KW-4 by RT-PCR.

RT-PCR with splice variant–specific primers for KW-4: several normal tissues as well as 8 different CLL samples are shown.

Close modal

Northern blot experiments show expression of the larger protein in normal tissue. However, the smaller protein missing the KRAB domain is highly expressed in several tumor cell lines (Figure4).

Fig. 4.

Northern blot analysis of gene expression for the antigen KW-4 and GAPDH.

Normal tissue (A) and tumor tissue (B).

Fig. 4.

Northern blot analysis of gene expression for the antigen KW-4 and GAPDH.

Normal tissue (A) and tumor tissue (B).

Close modal

KW-13 is highly expressed in peripheral leukocytes, spleen, and thymus. In contrast to the mouse homolog, which has a much more widespread expression, it has only very low expression in normal tissue and is not expressed in colon. However, it shows high expression in several tumor cell lines, including colon carcinoma, and has high expression in CLL (Figure 5).

Fig. 5.

Northern blot analysis of gene expression for the antigen KW-13 and GAPDH.

Normal tissue (A), tumor tissue (B), and CLL cells (C).

Fig. 5.

Northern blot analysis of gene expression for the antigen KW-13 and GAPDH.

Normal tissue (A), tumor tissue (B), and CLL cells (C).

Close modal

Prevalence of antibodies against antigens identified by sera from CLL patients, patients with other lymphomas, and healthy donors

To examine whether the identified antigens were recognized exclusively by autologous sera or also by allogeneic sera from CLL patients, other lymphoma patients, or from that of healthy donors, a phage plaque assay was performed as described above. Diluted, preabsorbed sera from patients with CLL, other lymphoma, and healthy donors were examined (Table 5).

Table 5.

Allogeneic serum responses against CLL-associated antigens

SerumCloneCLLLymphomaHealthyDetection as antigen in other studies
Only CLL KW-1 2 of 12 0 of 5 0 of 12  
 KW-3 2 of 16 0 of 5 0 of 12  
 KW-4 3 of 12 0 of 5 0 of 12  
 KW-2 3 of 12 0 of 5 0 of 12 AF286340 
 KW-8 4 of 23 n.d. 0 of 12 16 
 KW-12 2 of 16 0 of 5 0 of 12 21  
CLL + lymphoma KW-6 2 of 12 1 of 5 0 of 12  
 KW-10 2 of 24 2 of 5 0 of 12  
 KW-7 3 of 23 1 of 5 0 of 12 44 
 KW-11 4 of 23 3 of 5 0 of 12 17.25 
 KW-14 3 of 16 1 of 5 0 of 12 17  
CLL + lymphoma + healthy donors KW-5 20 of 30 2 of 5 2 of 12  
 KW-9 14 of 15 5 of 5 10 of 12  
 KW-13 7 of 16 2 of 5 3 of 12  
SerumCloneCLLLymphomaHealthyDetection as antigen in other studies
Only CLL KW-1 2 of 12 0 of 5 0 of 12  
 KW-3 2 of 16 0 of 5 0 of 12  
 KW-4 3 of 12 0 of 5 0 of 12  
 KW-2 3 of 12 0 of 5 0 of 12 AF286340 
 KW-8 4 of 23 n.d. 0 of 12 16 
 KW-12 2 of 16 0 of 5 0 of 12 21  
CLL + lymphoma KW-6 2 of 12 1 of 5 0 of 12  
 KW-10 2 of 24 2 of 5 0 of 12  
 KW-7 3 of 23 1 of 5 0 of 12 44 
 KW-11 4 of 23 3 of 5 0 of 12 17.25 
 KW-14 3 of 16 1 of 5 0 of 12 17  
CLL + lymphoma + healthy donors KW-5 20 of 30 2 of 5 2 of 12  
 KW-9 14 of 15 5 of 5 10 of 12  
 KW-13 7 of 16 2 of 5 3 of 12  

Phage plaque assay using multiple sera was performed. Screening results of multiple sera from patients with CLL, other lymphoma, and healthy donors for presence of antibodies against identified clones are shown. Previous detection of CLL-associated antigens in other studies is indicated.

In our experiments, 6 antigens have been detected only by CLL patients, whereas 8 of the 14 antigens were detected also in other lymphoma patients and 3 antigens were detected by sera from a subset of healthy donors. Three of the 6 antigens identified only in CLL patients in this study (KW-2, KW-8, KW-12) as well as 3 of the other antigens (KW-7, KW-11, KW-14) have been previously identified.16,17,21,25,44,62 However, KW-1, KW-3, and KW-4 are novel antigens, so far detected only in serum from patients with CLL. KW5 and KW13 were detected in a larger number of CLL patients. Both have also been recognized in some healthy donors. However, KW-5 is significantly more likely recognized by patients with CLL than in healthy donors (P = .01). It shows enhanced expression in most CLL cells compared with normal tonsilar B cells. However, it is also expressed in most normal tissues at least at low levels (data not shown). Clinical data of the majority of patients whose sera were used for the phage plaque assay are shown in Table6. Although patients with hypogammaglobulinemia and previous therapy seemed to have fewer humoral responses against identified antigens, this did not achieve statistical significance (hypogammaglobulinemia P = .08; previous therapy P = .1).

Table 6.

Clinical data of patients with CLL detecting 0 to 7 clones

Patient seraDetected clonesRai stageHypogamma-globulinemiaAutoimmune phenomenaPrior therapyCD38, % expression on CLL cellsIgH mutationCytogenetics
Low No No Yes 13q del 
II Low No Yes n.d. Yes Normal 
II Low Yes Yes 85 Yes Normal 
Normal No No 17 Yes 13q del 
IV Normal No Yes 83 No Normal 
Low No No 12 Yes Normal 
II Low Yes No 15 No +12 
Low No No Yes Normal 
II Normal Yes Yes 39 Yes 11q del 
10 II Low Yes Yes 14 No Normal 
11 II Normal Yes Yes 79 Yes Normal 
12 IV Low No Yes 86 Yes Normal 
13 IV Normal No No n.d. Yes n.d. 
14 Normal No No 10 Yes Normal 
15 Normal No No No 13q del 
16 II Normal No No 16 Yes +12, +18 
17 II Normal No No Yes 13q del 
Patient seraDetected clonesRai stageHypogamma-globulinemiaAutoimmune phenomenaPrior therapyCD38, % expression on CLL cellsIgH mutationCytogenetics
Low No No Yes 13q del 
II Low No Yes n.d. Yes Normal 
II Low Yes Yes 85 Yes Normal 
Normal No No 17 Yes 13q del 
IV Normal No Yes 83 No Normal 
Low No No 12 Yes Normal 
II Low Yes No 15 No +12 
Low No No Yes Normal 
II Normal Yes Yes 39 Yes 11q del 
10 II Low Yes Yes 14 No Normal 
11 II Normal Yes Yes 79 Yes Normal 
12 IV Low No Yes 86 Yes Normal 
13 IV Normal No No n.d. Yes n.d. 
14 Normal No No 10 Yes Normal 
15 Normal No No No 13q del 
16 II Normal No No 16 Yes +12, +18 
17 II Normal No No Yes 13q del 

Stage, serum Ig, autoimmune phenomena, therapy prior to serum harvest, CD38 expression level on CLL cells, presence of somatic hypermutation in the Ig rearrangement, and cytogenetic abnormalities are shown.

n.d. indicates not done.

Detection of cellular responses against identified antigens

The clones KW-2, KW-4, KW-5, and KW-13 appeared most interesting for investigation as potential targets for immunotherapeutic approaches either by their expression profile or by their antigenic potential. So far we identified peptides from the proteins corresponding to KW-2 and KW-13 that could potentially bind to HLA-A2+ donors. Twelve nonamer peptides with varying affinity to HLA-A2 molecules derived from clones KW-2 and KW-13 were synthesized and used for stimulation of peripheral T cells of HLA-A2+ healthy donors. Three peptides, KW-2/2: ALARNNAEV, KW-13/2: TLLHYLVKV (Figure 6), and KW-13/5: AAPPPPPPL, were repeatedly able to induce cytotoxic T-cell responses against peptide-pulsed autologous CD40-activated B cells. In contrast, CD40-activated B cells pulsed with an irrelevant peptide or unpulsed were not lysed. However, in keeping with our previous data that CLL cells are poor targets of CTLs generated against class I peptides,5 no peptide-specific CTL lines could be generated that recognized and lysed HLA-A2+ CLL cells. However, these data demonstrate that these clones do represent antigens for both T-cell and B-cell pathways.

Fig. 6.

Cytoxicity of autologous T cells of healthy donors after stimulation with antigen-presenting cells pulsed with KW-13–derived peptide 2.

Targets are CD40-activated B cells pulsed with peptide 2 of KW-13 (diamonds), pulsed with an unrelated peptide (closed circles), unpulsed CD40-activated B cells (open circles), and unstimulated CLL cells (squares).

Fig. 6.

Cytoxicity of autologous T cells of healthy donors after stimulation with antigen-presenting cells pulsed with KW-13–derived peptide 2.

Targets are CD40-activated B cells pulsed with peptide 2 of KW-13 (diamonds), pulsed with an unrelated peptide (closed circles), unpulsed CD40-activated B cells (open circles), and unstimulated CLL cells (squares).

Close modal

CLL is in several regards a tempting but difficult target for the development of immunotherapeutic strategies. The fundamental question arises if cancer-specific tumor antigens exist in CLL and if they can be recognized by the autologous host. Following CD40 activation, CLL cells can be recognized by the autologous immune system, making it likely that tumor-associated antigens are present. Although cytotoxic T-cell responses can be generated against idiotypic peptides,4,5 cytotoxic immune responses were weak. Additional tumor-associated antigens are likely needed to give a broader basis for polyvalent immunization strategies to prevent the escape of antigen loss variants.6 

SEREX provides a powerful method to identify immunogenic tumor antigens in many malignancies.14 However, the application of SEREX has several disadvantages for its use in CLL. Hypogammaglobulinemia is common in CLL patients as well as T-cell defects, resulting in reduced T-cell help. This is also reflected by the reduced antibody response of CLL patients after standard vaccinations.45 However, we detected humoral immune responses against 14 antigens expressed in cDNA libraries from 5 CLL primary tumor samples after screening of more than 30 million clones. The relatively low number of positive clones detected in this study after extensive screening compared with studies in other malignancies likely reflects the impaired immunologic responsiveness in CLL. Four CLL patients showed immune responses against 3 or more clones, suggesting that these patients had stronger immune recognition. These patients were untreated, had no hypogammaglobulinemia, early-stage disease, and low CD38 expression on CLL cells. Although the data are certainly preliminary, it is of interest to speculate that an ongoing immune response might even contribute to the low stage of these patients. In contrast, patients recognizing 2 or fewer antigens were mostly either treated with immunosuppressive agents or had hypogammaglobulinemia and higher percentage of CD38 expression on CLL cells.

The majority of antigens were recognized only in the minority of CLL patients. Molecular heterogeneity of this disease might contribute to this fact. However, in most SEREX studies selected antigens are detected only in the minority of tested patients. In a survey by Stockert et al,46 NY-ESO-1 was recognized in 12 of 127 melanoma patients and in 8 of 15 melanoma patients positive for NY-ESO-1. Antibodies against Her2/neu have been detected in 12 of 107 patients with early-stage breast cancer.47 MAGE-3 was detected only in 2 of 127 melanoma patients.46 However, clinical responses have been reported after immunizations with MAGE-3–derived HLA-A1–restricted peptides.48 Although experiences are limited, peptide vaccination has also shown promising results in a preliminary clinical trial using NY-ESO-1–related peptides.8 In this study, even 5 of the 7 vaccinated patients who were initially NY-ESO-1 antibody–negative developed stabilization or regression of individual metastases, and both humoral and cellular responses were observed.6 

The antigens detected in this study might be tumor antigen candidates in CLL. Three antigens (KW-1, KW-3, KW-4) have been detected only in CLL patients so far. Examination of larger cohorts will be necessary to establish specificity of antibody responses against these antigens for this disease. The other antigens were also detected in sera from other lymphoma patients or patients with other malignancies according to other studies.16,17,21,25,44 These antigens might therefore be tumor associated rather than CLL associated. However, they might represent more universal targets for anticancer immunotherapeutic approaches.

Of particular interest, 4 antigens (KW-2, KW-4, KW-5, KW-13) have favorable properties in regard to antigenic potential and restricted tissue or overexpression in CLL. KW-2 has a cancer-testis–like expression profile and has been detected in 3 of 12 CLL patients. A splice variant of KW-4 has been identified that is selectively expressed in CLL compared with most normal tissues. KW-5 has a broader expression in healthy tissue. However, although it has been recognized in 2 healthy donors, it is significantly more recognized in the sera of CLL patients. The expression of KW-13 is restricted to the peripheral leukocytes, thymus, and spleen but is aberrantly expressed in several tumor cell lines and highly expressed in CLL. There are other antigens that are overexpressed in cancer cells but also expressed in normal tissue, including Her2/neu, α-fetoprotein, telomerase-catalytic subunit, G-250, Muc-1, carcinoembryogenic antigen, and p53.26,49-53 With regard to Her2/neu, no histologic evidence of lymphocyte infiltration or tissue destruction was seen in rats,54 and no autoimmune phenomena have been observed in humans after treatment with anti-Her2/neu antibody.13 

We investigated 12 peptides for their potential to induce cytotoxic T-cell responses against 2 proteins (KW-2, KW-13) that might be suitable targets for immunotherapy. We were able to generate peptide-specific CTLs against these identified antigens. However, these CTLs were not able to lyse native CLL cells. There are many possible reasons for the lack of cytotoxicity. CLL cells might not present these peptides on their surface or are not able to provide sufficient costimulation. Further studies are under way to investigate this issue, including proteasome-mediated digestion analysis. However, conflicting data are available about a number of tumor antigen–derived peptides. One of the most antigenic peptides of HER-2/neu (p369-377) did not induce cytotoxic immune responses against native tumor cells in a study by Zaks and Rosenberg,55 whereas weak cytotoxic immune responses could be demonstrated by others.56 Using the 15 amino acids containing p369-384 as a putative helper peptide for vaccination, Knutson et al were able to show induction of cytotoxic peptide-specific T cells, which were able to lyse HLA-A2+tumor cells overexpressing HER2/neu protein.57SEREX-defined antigens may represent mostly T helper antigens. Nishikawa et al demonstrated that covaccination of mice with unmutated SEREX-defined antigens in combination with tumor-specific antigens mERK2 and HER2 induced specific cytotoxicity against HER2-neu+ tumors and inhibition of metastasis after late vaccination that could not be induced by vaccination with the tumor-specific antigens alone.58 These antigens might therefore provide help for tumor-specific immune responses. Humoral- or complement-mediated responses should not be underestimated in tumor eradication and protection. In transgenic mice a humoral response is essential for complete eradication of Her2/neu-expressing tumors.59 In addition, after idiotype-specific DNA vaccination in a murine B-cell lymphoma model, transfer of hyperimmune sera protected naive animals against tumor growth, whereas in vitro stimulation of immune splenocytes with tumor cells failed to induce idiotype-specific cytotoxicity.60Thus, although no suitable peptides have been identified so far to induce cytotoxic immune responses against native CLL cells, these antigens might provide antitumor help in this disease. Further studies are ongoing in vitro and in vivo to define the potential of the antigens presented in this study for immunotherapeutic strategies.

The authors are greatly thankful to G. J. Freeman and G. Dranoff for constructive discussion of the experimental work and manuscript as well as to Donna Neuberg and David Zahrieh for statistical analysis and Peter Varney for editorial assistance.

Prepublished online as Blood First Edition Paper, May 24, 2002; DOI 10.1182/blood-2002-02-0513.

Supported by grants from the Deutsche Krebshilfe and Dr Mildred Scheel Stiftung (A.M.K. and M.W.), a grant from the Berlex Oncology foundation (F.S.H.), grant UO1CA81534 from the National Cancer Institute (J.G.G.), and support from the Grayce B. Kerr Foundation.

A.M.K. 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
Rozman
C
Montserrat
E
Chronic lymphocytic leukemia [published erratum appears in N Engl J Med. 1995;333:1515] [see comments].
N Engl J Med.
333
1995
1052
1057
2
Cheson
BD
Chronic lymphocytic leukemia and hairy-cell leukemia.
Curr Opin Hematol.
1
1994
268
277
3
Khouri
IF
Keating
M
Korbling
M
et al
Transplant-lite: induction of graft-versus-malignancy using fludarabine-based nonablative chemotherapy and allogeneic blood progenitor-cell transplantation as treatment for lymphoid malignancies.
J Clin Oncol.
16
1998
2817
2824
4
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
5
Harig
S
Witzens
M
Krackhardt
AM
et al
Induction of cytotoxic T-cell responses against immunoglobulin V region-derived peptides modified at HLA-A2 binding residues.
Blood.
98
2001
2999
3005
6
Jager
E
Chen
YT
Drijfhout
JW
et al
Simultaneous humoral and cellular immune response against cancer-testis antigen NY-ESO-1: definition of human histocompatibility leukocyte antigen (HLA)-A2-binding peptide epitopes.
J Exp Med.
187
1998
265
270
7
Chen
YT
Gure
AO
Tsang
S
et al
Identification of multiple cancer/testis antigens by allogeneic antibody screening of a melanoma cell line library.
Proc Natl Acad Sci U S A.
95
1998
6919
6923
8
Jager
E
Gnjatic
S
Nagata
Y
et al
Induction of primary NY-ESO-1 immunity: CD8+ T lymphocyte and antibody responses in peptide-vaccinated patients with NY-ESO-1+ cancers.
Proc Natl Acad Sci U S A.
97
2000
12198
12203
9
Dranoff
G
Jaffee
E
Lazenby
A
et al
Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity.
Proc Natl Acad Sci U S A.
90
1993
3539
3543
10
Boczkowski
D
Nair
SK
Snyder
D
Gilboa
E
Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo.
J Exp Med.
184
1996
465
472
11
Zitvogel
L
Regnault
A
Lozier
A
et al
Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes.
Nat Med.
4
1998
594
600
12
Slamon
DJ
Leyland-Jones
B
Shak
S
et al
Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2.
N Engl J Med.
344
2001
783
792
13
Disis
ML
Cheever
MA
HER-2/neu protein: a target for antigen-specific immunotherapy of human cancer.
Adv Cancer Res.
71
1997
343
371
14
Sahin
U
Tureci
O
Schmitt
H
et al
Human neoplasms elicit multiple specific immune responses in the autologous host.
Proc Natl Acad Sci U S A.
92
1995
11810
11813
15
Jager
E
Ringhoffer
M
Arand
M
et al
Cytolytic T cell reactivity against melanoma-associated differentiation antigens in peripheral blood of melanoma patients and healthy individuals.
Melanoma Res.
6
1996
419
425
16
Scanlan
MJ
Chen
YT
Williamson
B
et al
Characterization of human colon cancer antigens recognized by autologous antibodies.
Int J Cancer.
76
1998
652
658
17
Scanlan
MJ
Gordan
JD
Williamson
B
et al
Antigens recognized by autologous antibody in patients with renal-cell carcinoma.
Int J Cancer.
83
1999
456
464
18
Jager
D
Stockert
E
Gure
AO
et al
Identification of a tissue-specific putative transcription factor in breast tissue by serological screening of a breast cancer library.
Cancer Res.
61
2001
2055
2061
19
Gure
AO
Stockert
E
Scanlan
MJ
et al
Serological identification of embryonic neural proteins as highly immunogenic tumor antigens in small cell lung cancer.
Proc Natl Acad Sci U S A.
97
2000
4198
4203
20
Naora
H
Yang
YQ
Montz
FJ
Seidman
JD
Kurman
RJ
Roden
RB
A serologically identified tumor antigen encoded by a homeobox gene promotes growth of ovarian epithelial cells.
Proc Natl Acad Sci U S A.
98
2001
4060
4065
21
Ono
T
Sato
S
Kimura
N
et al
Serological analysis of BALB/C methylcholanthrene sarcoma Meth A by SEREX: identification of a cancer/testis antigen.
Int J Cancer.
88
2000
845
851
22
Greiner
J
Ringhoffer
M
Simikopinko
O
et al
Simultaneous expression of different immunogenic antigens in acute myeloid leukemia.
Exp Hematol.
28
2000
1413
1422
23
Ling
M
Wen
YJ
Lim
SH
Prevalence of antibodies against proteins derived from leukemia cells in patients with chronic myeloid leukemia.
Blood.
92
1998
4764
4770
24
Tureci
O
Schmitt
H
Fadle
N
Pfreundschuh
M
Sahin
U
Molecular definition of a novel human galectin which is immunogenic in patients with Hodgkin's disease.
J Biol Chem.
272
1997
6416
6422
25
Eichmuller
S
Usener
D
Dummer
R
Stein
A
Thiel
D
Schadendorf
D
Serological detection of cutaneous T-cell lymphoma-associated antigens.
Proc Natl Acad Sci U S A.
98
2001
629
634
26
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
27
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
28
Zhu
Y
Qi
C
Cao
WQ
Yeldandi
AV
Rao
MS
Reddy
JK
Cloning and characterization of PIMT, a protein with a methyltransferase domain, which interacts with and enhances nuclear receptor coactivator PRIP function.
Proc Natl Acad Sci U S A.
98
2001
10380
10385
29
Zerial
M
Toschi
L
Ryseck
RP
Schuermann
M
Muller
R
Bravo
R
The product of a novel growth factor activated gene, fos B, interacts with JUN proteins enhancing their DNA binding activity.
EMBO J.
8
1989
805
813
30
Gou
DM
Sun
Y
Gao
L
et al
Cloning and characterization of a novel Kruppel-like zinc finger gene, ZNF268, expressed in early human embryo.
Biochim Biophys Acta.
1518
2001
306
310
31
Fetka
I
Radeghieri
A
Bouwmeester
T
Expression of the RNA recognition motif-containing protein SEB-4 during Xenopus embryonic development.
Mech Dev.
94
2000
283
286
32
Georgopoulos
K
Moore
DD
Derfler
B
Ikaros, an early lymphoid-specific transcription factor and a putative mediator for T cell commitment.
Science.
258
1992
808
812
33
Ge
H
Si
Y
Roeder
RG
Isolation of cDNAs encoding novel transcription coactivators p52 and p75 reveals an alternate regulatory mechanism of transcriptional activation.
EMBO J.
17
1998
6723
6729
34
Ballinger
CA
Connell
P
Wu
Y
et al
Identification of CHIP, a novel tetratricopeptide repeat-containing protein that interacts with heat shock proteins and negatively regulates chaperone functions.
Mol Cell Biol.
19
1999
4535
4545
35
Yasuda
K
Espinosa
R
III
Davis
EM
Le Beau
MM
Bell
GI
Human somatostatin receptor genes: localization of SSTR5 to human chromosome 20p11.2.
Genomics.
17
1993
785
786
36
Tommerup
N
Vissing
H
Isolation and fine mapping of 16 novel human zinc finger-encoding cDNAs identify putative candidate genes for developmental and malignant disorders.
Genomics.
27
1995
259
264
37
Graphodatsky
AS
Vorobieva
NV
Filipenko
ML
Voronina
EV
Frengen
E
Prydz
H
Assignment of the L11 ribosomal protein gene (RPL11) to human chromosome 1p36.1→p35 by in situ hybridization.
Cytogenet Cell Genet.
84
1999
97
98
38
Yayoshi-Yamamoto
S
Taniuchi
I
Watanabe
T
FRL, a novel formin-related protein, binds to Rac and regulates cell motility and survival of macrophages.
Mol Cell Biol.
20
2000
6872
6881
39
Resendez
E
Jr
Wooden
SK
Lee
AS
Identification of highly conserved regulatory domains and protein-binding sites in the promoters of the rat and human genes encoding the stress-inducible 78-kilodalton glucose-regulated protein.
Mol Cell Biol.
8
1988
4579
4584
40
Molnar
A
Wu
P
Largespada
DA
et al
The Ikaros gene encodes a family of lymphocyte-restricted zinc finger DNA binding proteins, highly conserved in human and mouse.
J Immunol.
156
1996
585
592
41
Prakash
B
Praefcke
GJ
Renault
L
Wittinghofer
A
Herrmann
C
Structure of human guanylate-binding protein 1 representing a unique class of GTP-binding proteins.
Nature.
403
2000
567
571
42
Aoki
K
Suzuki
K
Sugano
T
et al
A novel gene, Translin, encodes a recombination hotspot binding protein associated with chromosomal translocations.
Nat Genet.
10
1995
167
174
43
Aronsson
FC
Magnusson
P
Andersson
B
et al
The NIK protein kinase and C17orf1 genes: chromosomal mapping, gene structures and mutational screening in frontotemporal dementia and parkinsonism linked to chromosome 17.
Hum Genet.
103
1998
340
345
44
Singh
DP
Ohguro
N
Kikuchi
T
et al
Lens epithelium-derived growth factor: effects on growth and survival of lens epithelial cells, keratinocytes, and fibroblasts.
Biochem Biophys Res Commun.
267
2000
373
381
45
Hartkamp
A
Mulder
AH
Rijkers
GT
van Velzen-Blad
H
Biesma
DH
Antibody responses to pneumococcal and haemophilus vaccinations in patients with B-cell chronic lymphocytic leukaemia.
Vaccine.
19
2001
1671
1677
46
Stockert
E
Jager
E
Chen
YT
et al
A survey of the humoral immune response of cancer patients to a panel of human tumor antigens.
J Exp Med.
187
1998
1349
1354
47
Disis
ML
Pupa
SM
Gralow
JR
Dittadi
R
Menard
S
Cheever
MA
High-titer HER-2/neu protein-specific antibody can be detected in patients with early-stage breast cancer.
J Clin Oncol.
15
1997
3363
3367
48
Marchand
M
van Baren
N
Weynants
P
et al
Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1.
Int J Cancer.
80
1999
219
230
49
Butterfield
LH
Koh
A
Meng
W
et al
Generation of human T-cell responses to an HLA-A2.1-restricted peptide epitope derived from α-fetoprotein.
Cancer Res.
59
1999
3134
3142
50
Vissers
JL
De Vries
IJ
Schreurs
MW
et al
The renal cell carcinoma-associated antigen G250 encodes a human leukocyte antigen (HLA)-A2.1-restricted epitope recognized by cytotoxic T lymphocytes.
Cancer Res.
59
1999
5554
5559
51
Jerome
KR
Barnd
DL
Bendt
KM
et al
Cytotoxic T-lymphocytes derived from patients with breast adenocarcinoma recognize an epitope present on the protein core of a mucin molecule preferentially expressed by malignant cells.
Cancer Res.
51
1991
2908
2916
52
Tsang
KY
Zaremba
S
Nieroda
CA
Zhu
MZ
Hamilton
JM
Schlom
J
Generation of human cytotoxic T cells specific for human carcinoembryonic antigen epitopes from patients immunized with recombinant vaccinia-CEA vaccine.
J Natl Cancer Inst.
87
1995
982
990
53
Theobald
M
Biggs
J
Dittmer
D
Levine
AJ
Sherman
LA
Targeting p53 as a general tumor antigen.
Proc Natl Acad Sci U S A.
92
1995
11993
11997
54
Disis
ML
Gralow
JR
Bernhard
H
Hand
SL
Rubin
WD
Cheever
MA
Peptide-based, but not whole protein, vaccines elicit immunity to HER-2/neu, oncogenic self-protein.
J Immunol.
156
1996
3151
3158
55
Zaks
TZ
Rosenberg
SA
Immunization with a peptide epitope (p369–377) from HER-2/neu leads to peptide-specific cytotoxic T lymphocytes that fail to recognize HER-2/neu+ tumors.
Cancer Res.
58
1998
4902
4908
56
Brossart
P
Wirths
S
Stuhler
G
Reichardt
VL
Kanz
L
Brugger
W
Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells.
Blood.
96
2000
3102
3108
57
Knutson
KL
Schiffman
K
Disis
ML
Immunization with a HER-2/neu helper peptide vaccine generates HER-2/neu CD8 T-cell immunity in cancer patients.
J Clin Invest.
107
2001
477
484
58
Nishikawa
H
Tanida
K
Ikeda
H
et al
Role of SEREX-defined immunogenic wild-type cellular molecules in the development of tumor-specific immunity.
Proc Natl Acad Sci U S A.
98
2001
14571
14576
59
Reilly
RT
Machiels
JP
Emens
LA
et al
The collaboration of both humoral and cellular HER-2/neu-targeted immune responses is required for the complete eradication of HER-2/neu-expressing tumors.
Cancer Res.
61
2001
880
883
60
Syrengelas
AD
Levy
R
DNA vaccination against the idiotype of a murine B cell lymphoma: mechanism of tumor protection.
J Immunol.
162
1999
4790
4795
61
GenBank [database online]. Bethesda, MD: National Center for Biotechnology Information; 2002. Accession no. AB002334 Revised July 5, 2002.
62
GenBank [database online]. Bethesda, MD: National Center for Biotechnology Information; 2002. Accession no. AF286340 Revised July 5, 2002.

Author notes

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

Sign in via your Institution