The factors that determine progression from monoclonal gammopathy of undetermined significance (MGUS) to multiple myeloma are unclear but may include the breakdown of immune surveillance. Cancer testis antigens (CTAgs) are expressed by the majority of myelomas and MGUS tumors and are a potential immune target. We have characterized CD4+ and CD8+ T-cell immune responses to MAGE-A1/A2/A3 in these patients. CD4+ T-cell immunity to MAGE proteins is stronger and more frequent in MGUS compared with myeloma with a predominantly CD45RACCR7 effector memory profile and cytotoxicity against MAGE-positive cell lines. In contrast CD8+ T-cell immune responses were present almost exclusively in patients with multiple myeloma, correlating with disease, with a CD45RA+CCR7 memory phenotype, localizing poorly to the bone marrow but were able to lyse myeloma cell lines in vitro. This suggests that the CD4+ CTAg-specific immune response may play a role in controlling tumor growth, whereas the efficacy of the CD8+ T-cell response appears to be limited in vivo. Despite this, patients with evidence of a CTAg-specific immune response had a 53% reduction in mortality over a median follow-up of 4 years. These findings have important implications for clinical approaches to CTAg-specific immunotherapy in patients with cancer.

Multiple myeloma (MM) is a malignant disease of bone marrow plasma cells and remains incurable with current chemotherapy regimens.1-3  Although allogeneic stem cell transplantation is associated with significant morbidity and mortality, a graft-versus-myeloma effect has clearly been demonstrated,4,5  and tumor-specific immunotherapy offers potential in the management of this disease.6 

CTAgs are a family of proteins whose expression is restricted to spermatogenic germ cells,7  and more than 50 CTAg genes exhibit mRNA expression in testis with minimal expression in adult somatic cells.8,9  CTAg expression is observed in a wide variety of malignancies including melanoma and epithelial tumors. T-cell tolerance to CTAg peptides appears incomplete and functional T-cell responses may be generated as a consequence of CTAg expression in tumor cells.10  Expression of several CTAg proteins, including MAGE and NY-ESO, has been described in malignant plasma cells from patients with MM as well as transformed cell lines.11-18  CTAg expression is detected most commonly in patients with advanced disease but is also found in a significant proportion of patients with MGUS.15  Recently, aberrant expression of CTAgs has been shown to be a negative prognostic marker for MM17,19  and to be associated with a proliferative subset of tumor cells.15  There is also a correlation in MM18  and in certain solid tumors20  between the age of the patient and the number of expressed CTAgs. The mechanisms that underlie this expression are unclear but are at least partially related to demethylation of gene promoter sequences.7 

CTAgs have been the target for several vaccination studies in melanoma with tumor regression reported in approximately 5% to 20% of cases and no reports of vaccine toxicity.21-23 

MM is characterized by immune dysfunction and there have been few reports of T-cell immunity to CTAg proteins in patients with MM.11-18  MGUS is a premalignant precursor of MM with a 1% annual risk of progression to myeloma. The factors that determine progression are uncertain but could include immune control of plasma cell proliferation. Given the potential role of CTAgs as targets for immunosurveillance and immunotherapy strategies, it is essential to understand the cellular and humoral immune responses to these proteins in MM and MGUS to maximize tumor-specific immune responses.6  Indeed, both NY-ESO-1–based immunotherapy using protein transduction of dendritic cells24  and MA9E-A3 protein immunizations25  are being developed in multiple myeloma patients.

We have previously reported CTAg-specific CD8+ T-cell responses in more than 40% of patients with MM.26  Here we have characterized the CD4+ and CD8+ T-cell immune responses to MAGE-A1, MAGE-A2, and MAGE-A3 in patients with both MGUS and MM. We show that CD4+ T-cell immunity to these proteins is seen more commonly in patients with MGUS, whereas the CD8+ T-cell immune response was present almost exclusively in patients with MM. CD4+ and CD8+ CTAg-specific T-cell clones demonstrated high avidity for CTAg peptides and were cytotoxic for target cells expressing CTAg protein.

Patient samples

Patients with MM and MGUS were recruited from specialist clinics at the University Hospital NHS Trust and Heart of England NHS Trust (Birmingham, United Kingdom). The study received approval from the local ethics committees at South Birmingham, Birmingham East, North, and Solihull, and informed written consent was obtained in accordance with the Declaration of Helsinki in all cases. Peripheral blood (20 mL) was obtained from patients at various stages of disease and mononuclear cells were purified by density centrifugation using Lymphoprep (Nycomed, Oslo, Norway). MM patients were sampled at a variety of disease stages including presentation and plateau phase (Table S1, available on the Blood website; see the Supplemental Materials link at the top of the online article). Freshly isolated lymphocytes were used in all assays, and blood samples were processed within 3 to 6 hours of collection.

Cell lines and culture conditions

Epstein-Barr virus (EBV)–transformed cell lines were prepared with prototype 1 strain B95.8 or prototype 2 strain Ag876. LCLs were cultured in medium containing 10% fetal calf serum and human leukocyte antigen (HLA) type was obtained from the Anthony Nolan Clinic (London, United Kingdom). To assess the ability of CD4 T-cell clones to recognize processed antigen, 2 LCL cell lines of different HLA types (KS114d trans M3 DR52b+ and DESA-M3 DR52a+) were transfected with a retrovirus containing the sequence coding for the fusion truncated protein invariant chain (1-80)-MAGE-A3 using established methods27  and this work was kindly carried out by S. Ottaviani (Ludwig Institute for Cancer Research, Brussels, Belgium). MM cell lines U266, JJN3, and H929, expressing MAGE, were maintained in medium containing 10% fetal calf serum.12  The HLA type of these cell lines is listed as follows28 : U266: HLA A2, 3, B7, 60(40); DR15, DR17, DR52a, DR51, DQ-6, 2 DPw-2, DPw-4. JJN2: A3, 33, (19) B7, 14; DR15, 1, 51, DQ-5, 6. H929: A3, 24, B7, 18, Cw*0701, 0702, class II–negative.

Detection of CTAg-reactive T cells using the cytokine secretion assay

Fourteen peptides derived from CTAgs MAGE-A1/A2/A3 (Alta Bioscience, Birmingham, United Kingdom) were chosen from previously described epitopes for CD8+ and CD4+ T cells restricted by a wide variety of HLA alleles (HLA class I: A*0101, A*201, A*301, A*2401, B*702, and B*3501; and HLA class II DRB1*0401, DRB1*0701, DRB1*1101, DRB1*1301, DPB1*0401, and DQB1*0601)27,29-33  (Table 1). The expression of these 5 CD4+ MAGE-specific peptides is representative of approximately 94% of the alleles expressed by the white population.31  The IFN-γ cytokine secretion assay (CSA) was used to detect functional responses, and magnetic selection with anti-PE microbeads was used to increase the sensitivity of detection and further characterize the reactive cells.34  CSA was performed according to the manufacturer's instructions (Miltenyi Biotec, Bergisch Gladbach, Germany). Briefly, freshly isolated peripheral blood mononuclear cells (PBMCs) were seeded at a cell density of 107/mL in culture media containing RPMI 1640 (Invitrogen, Cergy Pontoise, France) supplemented with 10% human serum (HD Supplies, Aylesbury, United Kingdom) and l-glutamine (Invitrogen). The cells were then left unstimulated overnight at 37°C 5% CO2. The next day peptides were added to the cultures for 3 to 6 hours at a final concentration of 10 μg/mL. DMSO and Staphylococcal enterotoxin B (1 μg/mL; Sigma-Aldrich, Poole, United Kingdom) were used as negative and positive controls, respectively. After stimulation, the cells were labeled with IFN-γ catch reagent for 5 minutes and then incubated for 45 minutes at 37°C under continuous rotation. Cells were then labeled with IFN-γ detection antibody (PE-labeled) and then were magnetically labeled, using anti-PE magnetic beads. Enrichment of IFN-γ secretion cells was performed by magnetic separation using the auto–magnetic-activated cell sorting (MACS) separation program Posseld (Miltenyi Biotech). The positively selected cells were labeled with CD4-FITC and CD8 PC5 (Beckman Coulter, High Wycombe, United Kingdom), and propidium iodide (PI; 1 μg/mL; Sigma-Aldrich) was added before analysis to allow exclusion of dead cells. Analysis was performed with a Coulter EPICS XL flow cytometer (Beckman Coulter). The frequency of cytokine-secreted antigen-specific T cells was calculated as a percentage of the total CD8+ or CD4+ T-cell pool on the basis of the number of cells detected in the positively selected fraction compared with the total number of CD8+ or CD4+ T cells within PBMCs.34  The frequency was also calculated from the absolute lymphocyte count and expressed as the number of cytokine-secreting antigen-specific T cells/mL.

Table 1

Antigenic peptides encoded by cancer testis antigens used in the cytokine secretion assay

ProteinHLA (I) restrictionHLA frequency (%) within the populationPeptide sequence
MAGE-A1    
    96-104 A3 23 SLFRAVITK 
    135-143 A24 19 NYKHCFPEI 
    161-169 A1 26 EADPTGHSY 
    289-298 B7 17 RVRFFFPSL 
MAGE-A2    
    157-166 A2 44 YLQLVFGIEV 
MAGE-A3    
    97-105 A24 19 TFPDLESEF 
    112-120 A2 44 KVAEELVHFL 
    168-176 A1/B35 26 EVDPIGHLY 
    195-203 A24 19 IMPKAGLLI 
MAGE-A1/A2/A3/A6    
    121-134 DR13 19 LLKYRAREPVTKAE 
MAGE-A3    
    149-160 DR4/7 24, 25 VIFSKASSSLQL 
    191-205 DR11 25 GDNQIMPKAGLLIIV 
    243-258 DP4/DQ-6 75, 63 KKLLTQHFVQENYLEY 
    281-295 DR11 25 TSYVKVLHHMVKISG 
ProteinHLA (I) restrictionHLA frequency (%) within the populationPeptide sequence
MAGE-A1    
    96-104 A3 23 SLFRAVITK 
    135-143 A24 19 NYKHCFPEI 
    161-169 A1 26 EADPTGHSY 
    289-298 B7 17 RVRFFFPSL 
MAGE-A2    
    157-166 A2 44 YLQLVFGIEV 
MAGE-A3    
    97-105 A24 19 TFPDLESEF 
    112-120 A2 44 KVAEELVHFL 
    168-176 A1/B35 26 EVDPIGHLY 
    195-203 A24 19 IMPKAGLLI 
MAGE-A1/A2/A3/A6    
    121-134 DR13 19 LLKYRAREPVTKAE 
MAGE-A3    
    149-160 DR4/7 24, 25 VIFSKASSSLQL 
    191-205 DR11 25 GDNQIMPKAGLLIIV 
    243-258 DP4/DQ-6 75, 63 KKLLTQHFVQENYLEY 
    281-295 DR11 25 TSYVKVLHHMVKISG 

Memory phenotype of the reactive cells

The phenotype of CTAg-specific T cells was characterized using antibodies to CCR7-FITC (R&D Systems, Abingdon, United Kingdom) and CD45RA-PC5 (BD Pharmingen, San Diego, CA). The IFN-γ cytokine secretion assay was used initially and the positively selected cells were counterstained with CCR7-FITC and CD45RA-PC5. Analysis was performed with a Coulter EPICS XL flow cytometer (Beckman Coulter).

Generation of CTAg-specific T-cell clones

After IFN-γ CSA and enrichment, cells were isolated under sterile conditions using 2 MS columns (Miltenyi Biotec) and cloned by limiting dilution assay (LDA) over irradiated (40 Gy) allogeneic PBMCs and LCLs in RPMI 1640 supplemented with 10% human serum (HD Supplies), IL-2 (100 U/mL), and PHA (5 μg/mL). IL-7 (5 ng/mL), IL-15 (2 ng/mL), and IL-21 (2 ng/mL; Peprotech, Rocky Hill, NJ) were added to established cultures.

ELISA for quantification of IFN-γ cytokine production

Cloned T cells were washed twice in RPMI and incubated in 96-well V-bottom microculture plates with HLA-matched or -mismatched LCLs or MM cell lines U266, JJN3, and H929 prepulsed for 1 hour with 5 μM peptide (or DMSO control) or transfected with MAGE-3. Supernatant was harvested onto Maxisorb (NUNC, Roskilde, Denmark) 96-well flat-bottom plates after 18 hours and assayed for IFN-γ by enzyme-linked immunosorbent assay (ELISA; Endogen, Woburn, MA) in accordance with the manufacturer's recommended protocol. For peptide titration experiments, the peptide was diluted from 10−5 M to 10−11 M and expressed as a percentage of maximum IFN-γ release.

51Cr cytotoxicity assay

After 51Cr labeling (1 hour), target cells (2500/well) were added to effector cells (25 000/well) at an E:T ratio of 10:1 in a final volume of 200 μL complete culture medium in 96-well V-bottomed plates. CD4+ T-cell clones were incubated for 12 hours and CD8+ T-cell clones for 6 hours at 37°C. Supernatants were harvested and the mean percentage of specific lysis of triplicate wells was calculated according to the formula: (experimental release − spontaneous release)/(maximal release − spontaneous release) × 100%. Targets were HLA-matched or -mismatched LCLs or MM cell lines pre-exposed for 1 hour to 5 μM epitope peptide or to an equivalent concentration of DMSO solvent.

Statistical analysis

Mann-Whitney U test was used to test the level of significance between MAGE-specific CD8 versus CD4 T-cell responses in myeloma patients and between MAGE-specific CD8+ T-cell responses in myeloma versus MGUS patients. Linear regression statistical analysis was used to compare the correlation of the frequency of the MAGE-A1–specific CD8+ T-cell response with the paraprotein level over time in patient 7. Both of these were performed using Prism 4 for Windows (GraphPad, San Diego, CA).

The survival of MM patients was studied in relation to the presence or absence of a CTAg-specific CD8+ T-cell response using the R survival package (Vienna University, Vienna, Austria; http://www.r-project.org). The hazard ratio between the 2 groups was calculated using the Cox proportional hazards model and a log-rank test was used to calculate a P value. Statistical analysis of the survival data was performed by Wenbin Wei (Cancer Research UK Institute for Cancer Studies, University of Birmingham).

MAGE-A1–, MAGE-A2–, and MAGE-A3–specific CD8+ T cells are detected more frequently in patients with MM than in patients with MGUS

We have previously reported the use of the IFN-γ CSA to detect functional CD8 T-cell responses to CTAg peptides26  and now extend this cohort to long-term follow-up of 53 patients. A CTAg-specific CD8+ T-cell response was observed in 21 of these patients, of which 19 were specific for MAGE-A1, MAGE-A2, or a MAGE-A3 peptide with a magnitude that represented 0.001% to 0.3% of the CD8+ T-cell pool (Figure 1).

Figure 1

CD8+ and CD4+ T-cell responses to MAGE-A1/A2/A3–specific peptides. Maximal levels of CD8+ and CD4+ T-cell responses to MAGE-A1/A2/A3–specific peptides detected MM and MGUS patients. n indicates the number of patients screened in each group. The y-axis illustrates the percentage of CD8+ or CD4+ T cells responding to MAGE-A1/A2/A3–specific peptides calculated from the enriched sample (“Detection of CTAg-reactive T cells using the cytokine secretion assay”). The x-axis lists the patient group and CD8+ or CD4+. Undetected responses were given a value of 0.0001%, which is below the sensitivity of the assay. The Mann-Whitney U test was used to test level of significance between CD8+ and CD4+ MAGE responses in myeloma patients and between CD8+ responses in myeloma and MGUS patients.

Figure 1

CD8+ and CD4+ T-cell responses to MAGE-A1/A2/A3–specific peptides. Maximal levels of CD8+ and CD4+ T-cell responses to MAGE-A1/A2/A3–specific peptides detected MM and MGUS patients. n indicates the number of patients screened in each group. The y-axis illustrates the percentage of CD8+ or CD4+ T cells responding to MAGE-A1/A2/A3–specific peptides calculated from the enriched sample (“Detection of CTAg-reactive T cells using the cytokine secretion assay”). The x-axis lists the patient group and CD8+ or CD4+. Undetected responses were given a value of 0.0001%, which is below the sensitivity of the assay. The Mann-Whitney U test was used to test level of significance between CD8+ and CD4+ MAGE responses in myeloma patients and between CD8+ responses in myeloma and MGUS patients.

Close modal

MAGE-A1/A2/A3 expression has also been reported in plasma cells from patients with MGUS15  but there is no information as to whether this triggers a T-cell immune response. We therefore used the IFN-γ CSA to look for evidence of a CTAg-specific CD8+ T-cell response to pools of MAGE-A1/A2/A3 peptides in a cohort of 25 MGUS patients (Table 1). There was a significant difference between the frequency of MAGE-specific CD8 T-cell responses in MGUS versus MM patients (P = .029). Only 2 responses were seen in this cohort and both were of very small magnitude, representing only 0.001% of the CD8+ T-cell pool (Figure 1). Interestingly, both of these patients also had significant MAGE-A1/A2/A3–specific CD4+ T-cell responses (Figure 1).

MAGE-specific CD4+ T-cell responses are more commonly observed in MGUS than in myeloma patients

Antigen-specific CD4+ T cells can play an important role in regulating CD8+ T-cell immune responses and may also have a direct cytotoxic activity on target cells. We therefore undertook a comprehensive analysis of the CD4+ T-cell response to cancer testis antigens using a panel of 5 T-cell epitopes from MAGE-A1/A2/A3 (Table 1).

The IFN-γ CSA was used to detect functional CD4+ T-cell responses in 32 patients with MM and 30 patients with MGUS. A CD4+ T-cell response was seen in only 1 patient with myeloma (patient 53) and represented a median value of 0.03% of the CD4+ T-cell pool. The absolute magnitude of this CD4+ CTAg-specific T-cell response peaked at 262 cells/mL with a median of 143 cells/mL. In contrast 4 of the 30 MGUS pa-tients demonstrated a CD4+ CTAg-specific T-cell response with peak and median values of 0.07% and 0.03% of the CD4+ repertoire, respectively.

These results demonstrate that MAGE-specific CD4+ T-cell responses are significantly less common than CD8+ T-cell responses in patients with myeloma (P = .011; Figure 1). However, the reverse pattern is observed in MGUS patients in whom MAGE-specific CD8+ T-cell responses are rare, although MAGE-specific CD4+ T-cell responses are more commonly seen (Figure 1).

MAGE-specific CD4+ T-cell responses are maintained in patients with stable paraproteinemia

Serial analysis was performed in all the patients who demonstrated a MAGE-specific CD4+ T-cell response. The patient with myeloma was monitored over a 2-year period and demonstrated a sustained CD4+ T-cell response that fluctuated between 0.01% and 0.06% of the CD4+ T-cell pool (Figure 2A). Importantly, this patient has remained at plateau stage of disease throughout this period and has not been on any treatment for the last 2 years.

Figure 2

Serial detection of CTAg-specific CD4+ T-cell responses in patients with MGUS and myeloma. (A) The CTAg-specific CD4+ T-cell response was determined at several time points in a patient with myeloma. (B) Similar analysis was performed in 3 patients with MGUS. The x-axis indicates the number of days from diagnosis; ----, paraprotein level; and —, MAGE-A3–specific CD4+ T-cell response from the point of analysis. Treatment regimens are shown. VAD indicates vincristine, doxorubicin, and dexamethasone; and PBSCT, peripheral blood stem cell transplantation.

Figure 2

Serial detection of CTAg-specific CD4+ T-cell responses in patients with MGUS and myeloma. (A) The CTAg-specific CD4+ T-cell response was determined at several time points in a patient with myeloma. (B) Similar analysis was performed in 3 patients with MGUS. The x-axis indicates the number of days from diagnosis; ----, paraprotein level; and —, MAGE-A3–specific CD4+ T-cell response from the point of analysis. Treatment regimens are shown. VAD indicates vincristine, doxorubicin, and dexamethasone; and PBSCT, peripheral blood stem cell transplantation.

Close modal

The 4 patients with MGUS were also studied at several time points and MAGE-specific CD4+ T-cell responses were maintained over an 18-month period. (Figure 2B). All these patients exhibited stable disease and included 1 patient (MGUS patient 3) who exhibited a stable paraprotein of more than 30 g/L33  for 9 years (initial level 28 g/L).

The magnitude of the MAGE-specific CD8+ immune response increases in patients with relapsing disease

To investigate the magnitude of the CD8+ T-cell response in relation to disease activity, we performed serial analysis of CTAg-specific CD8+ immune responses during several years of patient follow-up (Figure 3). Analysis of patient 7 started 30 months into disease and CD8+ T-cell responses remained relatively stable for the next 40 months ranging from 0.001% to 0.016% of the CD8+ T-cell pool. However, at 74 months after diagnosis the patient demonstrated disease progression, with the IgG paraprotein rising from 17 g/L to 34 g/L. At this time, the magnitude of the CTAg-specific CD8+ T-cell response also increased by more than 10-fold, rising from 0.007% to 0.06% of the T-cell pool. In addition MAGE-A1–specific CD8+ T-cell clones could be isolated during this period and demonstrated cytolytic function against target cells in vitro. The patient did not demonstrate any evidence of a clinical response and died 2 months later. Linear regression analysis demonstrated that the frequency of the MAGE-A1–specific CD8+ T-cell response correlated positively with the paraprotein level (P = .042).

Figure 3

Serial analysis of MAGE-A1289-298–specific CD8+ T-cell responses in a patient with myeloma. The magnitude of the CTAg-specific CD8+ T-cell response was determined at several time points in patient 7 who was suffering from myeloma. The x-axis indicates the number of days from diagnosis; ----, paraprotein level; and —, MAGE-A1289-298–specific CD8+ T-cell response from the point of analysis. Treatment regimens are shown. Thal indicates thalidomide; and Dex, dexamethasone.

Figure 3

Serial analysis of MAGE-A1289-298–specific CD8+ T-cell responses in a patient with myeloma. The magnitude of the CTAg-specific CD8+ T-cell response was determined at several time points in patient 7 who was suffering from myeloma. The x-axis indicates the number of days from diagnosis; ----, paraprotein level; and —, MAGE-A1289-298–specific CD8+ T-cell response from the point of analysis. Treatment regimens are shown. Thal indicates thalidomide; and Dex, dexamethasone.

Close modal

A similar pattern was seen in patient 4, who demonstrated an increase in the CTAg-specific CD8+ T-cell response before disease progression (data not shown). Dual immunohistochemistry staining of bone marrow trephines with primary antibodies to CD138 and MAGE has been previously performed and demonstrated the dual expression of MAGE within the CD138+ cells in 7 patients, including patient 4 and patient 7.26 

CD8+ and CD4+ CTAg-specific T-cell responses have a predominant effector memory phenotype

To examine the memory phenotype of the CTAg-specific immune response in vivo, cells were stimulated with CTAg peptides and costained with antibodies to CD45RA and CCR7. The CTAg-specific CD8+ T-cell response was phenotyped in 5 patients with myeloma and in all cases the population was CD45RA+ but showed no expression of the chemokine receptor CCR7. This CD45RA+CCR7 subset represents a revertant effector memory population (TEMRA) that has undergone re-expression of the CD45RA isotype due to lack of recent antigen-specific stimulation (Figure 4A).36-40  This phenotype is consistent with a previous paper reporting Melan-A and tyrosinase-specific CD8+ T cells in patients with melanoma.40 

Figure 4

Phenotypic analysis of MAGE-specific CD4+ and CD8+ responses in patients with MM and MGUS. PBMCs from patients with paraproteinemia were stimulated with MAGE peptides and IFN-γ–responsive cells were counterstained with CD45RA and CCR7. The phenotype of the MAGE-A1289-298–specific CD8+ T-cell response in MM patient 7 is shown in panel A and is a representative plot; 4 other patients all demonstrated the same phenotype. Panel B illustrates the phenotype of the MM patient 53 and panel C shows the phenotype of MGUS patient 1.

Figure 4

Phenotypic analysis of MAGE-specific CD4+ and CD8+ responses in patients with MM and MGUS. PBMCs from patients with paraproteinemia were stimulated with MAGE peptides and IFN-γ–responsive cells were counterstained with CD45RA and CCR7. The phenotype of the MAGE-A1289-298–specific CD8+ T-cell response in MM patient 7 is shown in panel A and is a representative plot; 4 other patients all demonstrated the same phenotype. Panel B illustrates the phenotype of the MM patient 53 and panel C shows the phenotype of MGUS patient 1.

Close modal

In contrast, the CD4+ CTAg-specific T-cell response in the patient with myeloma showed no expression of CD45RA and had a homogeneous CD45RACCR7 effector memory phenotype (Figure 4B). Although the majority of the CTAg-specific CD4+ T cells detected in MGUS patients were also effector memory, there was also a small population of CCR7+ central memory cells (Figure 4C).

MAGE-A1–specific CD8+ T cells do not show selective recruitment to the bone marrow of patients with myeloma

Malignant plasma cells in patients with MGUS or myeloma are located almost exclusively within bone marrow, with minimal dissemination within the bloodstream. To determine whether the CTAg-specific immune response was able to home to tumor sites, we then compared the frequency of CTAg-specific CD8+ T cells within paired samples of peripheral blood and bone marrow. Patients 4 and 7 had demonstrated a consistent MAGE-A1289-298–specific CD8+ T-cell response within blood over many months, and paired samples became available from both patients at the time of disease relapse. Despite large numbers of plasma cells within the marrow biopsies, there was no evidence of selective recruitment of CTAg-specific T cells to the marrow microenvironment. In patient 4, the frequency of this T-cell response within marrow was reduced by 50% compared with that seen in blood and represented only 0.005% of the T-cell pool (Figure 5). Analysis of patient 7 showed that the T-cell response was comparable in both compartments. The phenotype of the CTAg-specific CD8+ T-cell immune response within bone marrow was identical (CD45RA+CCR7) to that in peripheral blood (data not shown).

Figure 5

Comparative analysis of CTAg-specific CD8 T cells within blood and bone marrow of a MM patient. The IFN-γ CSA was used to detect and quantify MAGE-A1289-298–specific CD8+ T cells in blood and bone marrow samples taken at the same time point. The bar chart shows the percentage frequency of MAGE-A1289-298–specific CD8+ T cells detected in blood and bone marrow (y-axis).

Figure 5

Comparative analysis of CTAg-specific CD8 T cells within blood and bone marrow of a MM patient. The IFN-γ CSA was used to detect and quantify MAGE-A1289-298–specific CD8+ T cells in blood and bone marrow samples taken at the same time point. The bar chart shows the percentage frequency of MAGE-A1289-298–specific CD8+ T cells detected in blood and bone marrow (y-axis).

Close modal

MAGE-A3–specific CD4+ T-cell clones from patients with paraproteinemia have high avidity for peptide and recognize endogenously processed antigen

Several CD4+ T-cell clones were isolated from 3 patients who exhibited a CTAg-specific CD4+ T-cell response, and their avidity assessed by cytokine release in response to target cells loaded with variable concentrations of peptide (Figure 6Ai,Bi). The clones were able to respond to target cells loaded with peptide concentrations down to 10−7 M to 10−9 M, which is comparable to values obtained for virus-specific CD4+ populations and indicates high-avidity antigen recognition.41  Importantly, CD4+ T-cell clones were also able to recognize target cells transfected with recombinant MAGE-A3, which demonstrates their ability to respond to naturally processed MAGE-A3 epitopes (Figure 6Aii, Bii).

Figure 6

Functional analysis of CD4 and CD8 clones generated from MM and MGUS patients. (A,B) IFN-γ production and cytotoxicity properties of generated CD4+ T-cell clones (TCC) from MGUS and MM patients. (i) Peptide titration analysis of CD4+ T-cell clones from MM (Bi) and MGUS (Ai) patients. (ii) MAGE-A3–specific CD4+ T cells were stimulated with peptide-loaded or DMSO-loaded HLA-matched or -mismatched LCL target cells or target cells transfected with a recombinant MAGE-A3 protein (KS114d transfected with MAGE-A3 [DR52b+] or DESA mismatched DR52b LCL trans MAGE-A3). (Biii) T-cell clones from MM patient were screened against a panel of different LCLs to identify class II restriction of T-cell clones specific to MAGE-A3145-160. Cytolytic activity of the CD4+ T-cell clones by standard 12-hour chromium release assay at an effector:target ratio of 10:1 (Biv) and (Aiii) against peptide-loaded HLA-matched target cells or target cells transfected with MAGE-A3. (Ci,ii) IFN-γ production and cytotoxicity properties of generated CD8 TCC from MM patients 4 and 7: (a) Peptide titration analysis of CD8+ T-cell clones from MM patients 4 and 7 represented as a percentage of IFN-γ release. The line represents 50% recognition of peptide concentration. (b) MAGE-A1289-298–specific CD8+ T cells were stimulated with peptide-loaded or DMSO-loaded HLA-matched or -mismatched LCL target cells or MM cell lines U266, JJN3, and H929. (c) Cytolytic activity of the CD8+ T-cell clones by standard 6-hour chromium release assay at an effector-target ratio of 10:1 against peptide or DMSO-loaded HLA-matched target cells or MM cell lines.

Figure 6

Functional analysis of CD4 and CD8 clones generated from MM and MGUS patients. (A,B) IFN-γ production and cytotoxicity properties of generated CD4+ T-cell clones (TCC) from MGUS and MM patients. (i) Peptide titration analysis of CD4+ T-cell clones from MM (Bi) and MGUS (Ai) patients. (ii) MAGE-A3–specific CD4+ T cells were stimulated with peptide-loaded or DMSO-loaded HLA-matched or -mismatched LCL target cells or target cells transfected with a recombinant MAGE-A3 protein (KS114d transfected with MAGE-A3 [DR52b+] or DESA mismatched DR52b LCL trans MAGE-A3). (Biii) T-cell clones from MM patient were screened against a panel of different LCLs to identify class II restriction of T-cell clones specific to MAGE-A3145-160. Cytolytic activity of the CD4+ T-cell clones by standard 12-hour chromium release assay at an effector:target ratio of 10:1 (Biv) and (Aiii) against peptide-loaded HLA-matched target cells or target cells transfected with MAGE-A3. (Ci,ii) IFN-γ production and cytotoxicity properties of generated CD8 TCC from MM patients 4 and 7: (a) Peptide titration analysis of CD8+ T-cell clones from MM patients 4 and 7 represented as a percentage of IFN-γ release. The line represents 50% recognition of peptide concentration. (b) MAGE-A1289-298–specific CD8+ T cells were stimulated with peptide-loaded or DMSO-loaded HLA-matched or -mismatched LCL target cells or MM cell lines U266, JJN3, and H929. (c) Cytolytic activity of the CD8+ T-cell clones by standard 6-hour chromium release assay at an effector-target ratio of 10:1 against peptide or DMSO-loaded HLA-matched target cells or MM cell lines.

Close modal

The MAGE-A3–specific CD4+ T-cell response that was detected in the patient with myeloma was specific for peptide MAGE-A3149-160, which has previously been reported to be presented by the products of the HLA DRB1*0401 and DRB1*0701 alleles. To confirm the HLA restriction of the T-cell clones, we used several LCLs with defined HLA class II genotype as target cells for peptide presentation. Interestingly, only target cells that expressed DR52b (determined by HLA DRB3*0201) were recognized by the clones (Figure 6Biii), and no recognition was observed when peptides were presented on HLA-DR52a (determined by HLA DRB3*0101)–expressing target cells (Figure 6Bii). This specificity corresponded with the patient's own HLA-DR type (DR1, 17, 52b), which was negative for DR4 and DR7 but positive for DR52b, and illustrates that the same CD4 peptide can be restricted through several HLA DR alleles. Interestingly, the MAGE-A3191-205–specific CD4+ T-cell clones generated from the MGUS patients (Figure 6Aii) were also able to recognize peptides presented by any target cell that expressed HLA DR52b. This peptide has been reported to be restricted through HLA-DR11, but recognition of the clones from this patient proved to be independent of this allele (Figure 6Aii).

MAGE-A3–specific CD4+ T-cell clones have cytolytic function

To gain some insight into the physiologic relevance of the CTAg-specific CD4+ clones, we tested the ability of the cells to kill target cells loaded with peptide. The CD4+ clones from the patients with MGUS showed strong cytolytic activity against peptide-loaded targets and were also able to kill target cells expressing the recombinant MAGE-A3 protein (Figure 6Aiii).

Interestingly, clones from the myeloma patient were able to kill only peptide-loaded target cells (Figure 6Biv). The mechanism of cytotoxicity was investigated through flow cytometric analysis and demonstrated positive staining for granzyme but only minimal expression of perforin (data not shown).

MAGE-A1–specific CD8+ T-cell clones demonstrate high-avidity recognition of MAGE peptide and can kill myeloma tumor cells

CD8+ T-cell clones were generated from 2 patients with MM (patients 4 and 7) to allow a detailed analysis of their functional capacity and antigenic specificity. The avidity of the clones was assessed by cytokine response in the presence of limiting peptide concentrations (Figure 6Cia,Cib). MAGE-A1289-298–specific CD8+ T-cell clones from both patients were able to recognize target cells loaded with peptide concentrations down to 10−9 M to 10−11 M. The concentration that sensitized for half-maximal IFN-γ release was 10−8 M, which is comparable to values obtained for virus-specific CD8+ populations.42 

We next investigated the capacity of MAGE-A1289-298–specific CD8+ T-cell clones to recognize HLA-matched MM cell lines with endogenous expression of MAGE-A1. CD8+ T-cell clones were restricted through HLA-B7, which is also expressed by 3 MM cell lines U266, JJN3, and H929, all of which have previously been shown to express MAGE-A1.12,28  Both sets of clones were able to recognize endogenously processed antigen in all 3 cell lines as determined by IFN-γ ELISA assay (Figures 6Cib,Ciib).

The chromium release assay was then used to assess the cytotoxic capacity of the CD8+ CTAg T-cell response and confirmed that all 3 MM cell lines were killed by the T-cell clones. (Figure 6Cic,Ciic).

Patients with MM who demonstrate a CTAg-specific T-cell response have an improved clinical outcome.

The clinical significance of a CTAg-specific immune response in patients with paraproteinemia is unknown, and we therefore compared the survival rates in 52 patients in whom a CD8+ CTAg-specific T-cell response could be detected and compared it with patients who exhibited no CTAg-specific immunity (Figure 7; Table S1). The presence of a CTAg-specific immune response was associated with a 53% reduction in mortality (hazard ratio: 0.457; 95% confidence interval [CI] 0.203-1.03), although this failed to reach statistical significance (P value of log-rank test = .052).

Figure 7

Impact on survival of CTAg-specific immunity in MM patients. A Kaplan-Meier survival curve was constructed for 52 patients with myeloma who had been screened for CTAg-specific immune responses. — represents MM patients in whom a CTAg-specific T-cell response could be detected; ----, MM patients who were negative. The hazard ratio between the CTAg-positive and -negative groups is 0.457 (95% CI 0.203-1.03) with P = .052 by log-rank test.

Figure 7

Impact on survival of CTAg-specific immunity in MM patients. A Kaplan-Meier survival curve was constructed for 52 patients with myeloma who had been screened for CTAg-specific immune responses. — represents MM patients in whom a CTAg-specific T-cell response could be detected; ----, MM patients who were negative. The hazard ratio between the CTAg-positive and -negative groups is 0.457 (95% CI 0.203-1.03) with P = .052 by log-rank test.

Close modal

CTAg protein expression has been observed in a wide range of tumors and frequently induces a cellular and humoral immune response that may help to control tumor growth.10  T-cell immunity to CTAg proteins has been identified in association with several different cancers, although the frequency of CTAg-specific T cells is often of a lower magnitude than the immune response against other tumor-associated antigens such as differentiation antigens.43,44  CTAgs are expressed in many cases of myeloma and MGUS, and an immune response to these proteins may play a role in controlling disease progression.14,15,45  Indeed, there is precedent for this principle as patients with MGUS often exhibit an immune response to the embryonal antigen SOX2.46  CD8+ immune responses against CTAg proteins have been documented in patients with myeloma,14-16,18,24,26,45  but there is currently no information regarding CTAg-specific immunity in MGUS. In this study, we performed a comprehensive study of CD8+ and CD4+ T-cell immunity to cancer testis antigens in patients with paraproteinemia by long-term study of cohorts of patients with MM or MGUS.

A striking feature of the current study is that although CD8+ immune responses were seen in many patients with myeloma, they were observed most strongly in late stages of disease and CTAg-specific T-cell clones could be isolated readily from patients with terminal disease. These observations suggest that expanded CD8+ CTAg-specific immune responses have limited value in controlling tumor growth in vivo despite the potent activity of T-cell clones against tumor cells in vitro. The biologic basis for this is unclear, but potential factors have been addressed in this study. The localization of CD8+ CTAg-specific T cells to the site of tumor is clearly impaired as no selective recruitment to bone marrow was observed. In contrast, previous studies have shown that the bone marrow can act as a reservoir for T cells that have the ability to lyse malignant plasma cells.47,48  Our study focused on CTAg-specific T cells rather than whole effector T-cell populations, and the observation that CTAg-specific CD8+ T cells are not sufficient to eradicate the tumor may partly be explained by their low frequency in the tumor microenvironment. In addition, samples were taken at the time of clinical relapse, which may relate to their poor localization to marrow.

The phenotype of CD8+ CTAg-specific T cells was that of CD45RA+CCR7 resting memory cells that represent a population that has not recently undergone antigenic stimulation.39,49  This phenotype has been reported on Melan-A and tyrosinase tumor-reactive CD8+ T cells in melanoma patients40  and indicates that CTAg-specific CD8+ T cells appear to be prevented from interacting with tumor cells in vivo. This is supported by the finding that such clones revert to CD45RO expression after stimulation with antigen in vitro, and a recent study showed that MAGE-A3 vaccine-induced CD8 T cells expressed a CD45RO25  phenotype in vivo. The physiologic mechanisms behind these observations are uncertain but could potentially be related to the increase in T-regulatory cells that has been observed in MM, or a lack of appropriate costimulation.49  The lack of any protective component of the CTAg-specific CD8+ T-cell response is supported with the observation that such responses are virtually absent in MGUS patients who demonstrate stable disease. CD8+ immune responses were seen in only 2 MGUS patients and were themselves of unusually small magnitude and seen in patients who also had CTAg-specific CD4+ T-cell responses. Overall, these results suggest that the magnitude of the CTAg-specific CD8+ CD45RA+ T-cell response reflects the magnitude of tumor burden in myeloma but is not sufficient to control disease.

Many experimental models have demonstrated that effective CD8+ T-cell immunity is dependent on the presence of a CD4+ T-cell response, and we therefore undertook a comprehensive analysis of the CD4+ T-cell response to CTAg in patients at different stages of paraproteinemia using T-cell epitopes derived from MAGE-A1/A2/A3.27,30-33  Thirty-two patients with MM were screened and only a single CD4+ T-cell response was observed. Interestingly, this was present in a patient with stable plateau phase disease and was durable on 7 occasions during 21 months of observation. We then went on to study 30 patients with MGUS and found that 4 of them exhibited a CD4+ CTAg-specific T-cell response. Although present in only a minority of patients, it is important to remember that the study used only 5 CTAg peptides and, as more than 50 CTAg genes have been described, the frequency of CTAg-specific CD4+ T cells is likely to have been considerably underestimated. As well as being more common in patients with MGUS rather than myeloma, the breadth, magnitude, and phenotype of the CTAg-specific CD4+ T-cell response were also unique in this group. Responses were directed against all the 5 CD4+ peptides used in the study and demonstrated a mean frequency of 0.03% of the CD4+ T-cell pool. Finally, although the majority of CTAg-specific CD4+ cells in all patients exhibited a CD45RACCR7 effector memory phenotype, patients with MGUS also exhibited a small population of CCR7+ central memory cells, which were absent in the patient with myeloma. The functional capability of these cells was demonstrated by the observation that CTAg-specific CD4+ T-cell clones could be isolated from both myeloma and MGUS patients and demonstrated cytolytic activity against endogenously processed MAGE-A3 protein.

The current study has used a peptide approach to investigate the detection of CD4+ T-cell responses in MM and MGUS patients, and it could be argued that our results are underestimating the frequency of responses. Indeed a whole-protein approach could potentially be more comprehensive. However, the peptides chosen were all previously described and discovered using whole-protein screening, and therefore are likely to represent the most immunodominant regions of the protein. Therefore, although the cohort size is small, these data suggest that CD4+ CTAg-specific T-cell responses may play a more important role in the control of tumor progression than CD8+ immunity. Indeed, there is substantial precedent for this model in other diseases, and the presence of a CD4+ T-cell IFNγ response has been shown to be predictive of control of Papillomavirus infection in malignant cervical disease.50,51  A CD4+ immune response has been shown to play an important role in the control of plasma cell proliferation in murine models,52  and potential mechanisms could include growth suppression through cytokine release, direct cytotoxicity, or maintenance of functional CD8+ immunity.

These observations on the differential induction and function of the CD4+ and CD8+ immune response to cancer testis antigens during tumor progression have important implications for immunotherapy. If CTAg-specific CD4+ T-cell responses do indeed play a role in controlling the progression of paraproteinemia, then effort should be given to maintaining or re-establishing their presence in patients with progressive disease. In addition, although the CD8+ CTAg-specific T-cell response may have limited therapeutic value in vivo, at least in patients with advanced disease, it should be possible to activate these cells in vivo through the use of vaccination or dendritic cell therapy.53  Indeed, patients with myeloma have been shown to harbor a tumor-specific CD8+ T-cell response that can lyse primary tumor cells after stimulation with tumor cells in vitro, although the antigenic specificity of this population has not been documented.54 

Patients who developed a CTAg-specific CD8+ T-cell immune response demonstrated an improved long-term survival, but it is unclear whether this reflects a specific CTAg-specific CD8+ role in controlling tumor progression. Previous studies indicate that the expression of CTAgs by malignant plasma cells in MM is indicative of poor prognosis11,14-16,45  but did not address the prognostic role of CTAg-specific immune responses. In our study, many patients were analyzed during or after therapy and received varied treatments including immunodulatory drugs such as thalidomide, and this is clearly relevant in the detection of immune responses and interpreting our data. Further work is required to look at T-cell responses to other CTAgs, particularly MAGE-C1, which is widely expressed in myeloma, although no T-cell epitopes from MAGE-C1 have been described to date.55  Our data indicate that cancer testis antigens represent an important target for future immunotherapeutic intervention in MM.

The online version of this article contains a data supplement.

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 USC section 1734.

We are grateful to Tina McSkeane and Julie Arrazi for collection of blood samples from patient cohorts and to Rahela Khanum for her help in the laboratory.

Contribution: O.C.G. performed all the laboratory analysis and helped write the paper; G.P. supplied patient samples and patient information and helped write the paper; A.M. helped with some of the laboratory analysis; M.C. supplied patient samples and patient information; K.P. helped with experimental design and writing of the paper; and P.M. supervised the project and helped write the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Oliver C. Goodyear, Cancer Research, UK Institute for Cancer Studies, University of Birmingham, Edgebaston, Birmingham, B15 2TT, United Kingdom; e-mail: goodyeoc@bham.ac.uk.

1
Richardson
 
PG
Mitsiades
 
CS
Hideshima
 
T
Anderson
 
KC
Novel biological therapies for the treatment of multiple myeloma.
Best Pract Res Clin Haematol
2005
, vol. 
18
 (pg. 
619
-
634
)
2
Bataille
 
R
Harousseau
 
JL
Multiple myeloma.
N Engl J Med
1997
, vol. 
336
 (pg. 
1657
-
1664
)
3
Barlogie
 
B
Shaughnessy
 
J
Tricot
 
G
, et al. 
Treatment of multiple myeloma.
Blood
2004
, vol. 
103
 (pg. 
20
-
32
)
4
Crawley
 
C
Lalancette
 
M
Szydlo
 
R
, et al. 
Outcomes for reduced-intensity allogeneic transplantation for multiple myeloma: an analysis of prognostic factors from the Chronic Leukaemia Working Party of the EBMT.
Blood
2005
, vol. 
105
 (pg. 
4532
-
4539
)
5
Lokhorst
 
HM
Schattenberg
 
A
Cornelissen
 
JJ
, et al. 
Donor lymphocyte infusions for relapsed multiple myeloma after allogeneic stem-cell transplantation: predictive factors for response and long-term outcome.
J Clin Oncol
2000
, vol. 
18
 (pg. 
3031
-
3037
)
6
Pratt
 
G
Goodyear
 
O
Moss
 
P
Immunodeficiency and immunotherapy in multiple myeloma.
Br J Haematol
2007
, vol. 
138
 (pg. 
563
-
579
)
7
Simpson
 
AJ
Caballero
 
OL
Jungbluth
 
A
Chen
 
YT
Old
 
LJ
Cancer/testis antigens, gametogenesis and cancer.
Nat Rev Cancer
2005
, vol. 
5
 (pg. 
615
-
625
)
8
Old
 
LJ
Cancer is a somatic cell pregnancy [editor's view].
Cancer Immun
2007
, vol. 
7
 pg. 
19
 
9
Stevenson
 
BJ
Iseli
 
C
Panji
 
S
, et al. 
Rapid evolution of cancer/testis genes on the X chromosome.
BMC Genomics
2007
, vol. 
8
 pg. 
129
  
10
Boon
 
T
van der Bruggen
 
P
Human tumor antigens recognized by T lymphocytes.
J Exp Med
1996
, vol. 
183
 (pg. 
725
-
729
)
11
van Baren
 
N
Brasseur
 
F
Godelaine
 
D
, et al. 
Genes encoding tumor-specific antigens are expressed in human myeloma cells.
Blood
1999
, vol. 
94
 (pg. 
1156
-
1164
)
12
Pellat-Deceunynck
 
C
Mellerin
 
MP
Labarriere
 
N
, et al. 
The cancer germ-line genes MAGE-1, MAGE-3 and PRAME are commonly expressed by human myeloma cells.
Eur J Immunol
2000
, vol. 
30
 (pg. 
803
-
809
)
13
Lim
 
SH
Bumm
 
K
Chiriva-Internati
 
M
Xue
 
Y
Wang
 
Z
MAGE-C1 (CT7) gene expression in multiple myeloma: relationship to sperm protein 17.
Eur J Haematol
2001
, vol. 
67
 (pg. 
332
-
334
)
14
Dhodapkar
 
MV
Osman
 
K
Teruya-Feldstein
 
J
, et al. 
Expression of cancer/testis (CT) antigens MAGE-A1, MAGE-A3, MAGE-A4, CT-7, and NY-ESO-1 in malignant gammopathies is heterogeneous and correlates with site, stage and risk status of disease.
Cancer Immun
2003
, vol. 
3
 (pg. 
9
-
16
)
15
Jungbluth
 
AA
Ely
 
S
DiLiberto
 
M
, et al. 
The cancer-testis antigens CT7 (MAGE-C1) and MAGE-A3/6 are commonly expressed in multiple myeloma and correlate with plasma-cell proliferation.
Blood
2005
, vol. 
106
 (pg. 
167
-
174
)
16
van Rhee
 
F
Szmania
 
SM
Zhan
 
F
, et al. 
NY-ESO-1 is highly expressed in poor-prognosis multiple myeloma and induces spontaneous humoral and cellular immune responses.
Blood
2005
, vol. 
105
 (pg. 
3939
-
3944
)
17
Taylor
 
BJ
Reiman
 
T
Pittman
 
JA
, et al. 
SSX cancer testis antigens are expressed in most multiple myeloma patients: co-expression of SSX1, 2, 4, and 5 correlates with adverse prognosis and high frequencies of SSX-positive PCs.
J Immunother
2005
, vol. 
28
 (pg. 
564
-
575
)
18
Atanackovic
 
D
Arfsten
 
J
Cao
 
Y
, et al. 
Cancer-testis antigens are commonly expressed in multiple myeloma and induce systemic immunity following allogeneic stem cell transplantation.
Blood
2007
, vol. 
109
 (pg. 
1103
-
1112
)
19
Condomines
 
M
Mose
 
D
Raynaud
 
P
, et al. 
Cancer/testis genes in multiple myeloma: expression patterns and prognosis value determined by microarray analysis.
J Immunol
2007
, vol. 
178
 (pg. 
3307
-
3315
)
20
Peng
 
JR
Chen
 
HS
Mou
 
DC
, et al. 
Expression of cancer/testis (CT) antigens in Chinese hepatocellular carcinoma and its correlation with clinical parameters.
Cancer Lett
2005
, vol. 
219
 (pg. 
223
-
232
)
21
Coulie
 
PG
van der Bruggen
 
P
T-cell responses of vaccinated cancer patients.
Curr Opin Immunol
2003
, vol. 
15
 (pg. 
131
-
137
)
22
Jäger
 
E
Karbach
 
J
Gnjatic
 
S
, et al. 
Recombinant vaccinia/fowlpox NY-ESO-1 vaccines induce both humoral and cellular NY-ESO-1-specific immune responses in cancer patients.
Proc Natl Acad Sci U S A
2006
, vol. 
103
 (pg. 
14453
-
14458
)
23
Baumgaertner
 
P
Rufer
 
N
Devevre
 
E
, et al. 
Ex vivo detectable human CD8 T-cell responses to cancer-testis antigens.
Cancer Res
2006
, vol. 
66
 (pg. 
1912
-
1916
)
24
Batchu
 
RB
Moreno
 
AM
Szmania
 
SM
, et al. 
Protein transduction of dendritic cells for NY-ESO-1-based immunotherapy of myeloma.
Cancer Res
2005
, vol. 
65
 (pg. 
10041
-
10049
)
25
Szmania
 
S
Gnjatic
 
S
Tricot
 
G
, et al. 
Immunization with a recombinant MAGE-A3 protein after high-dose therapy for myeloma.
J Immunother (1997)
2007
, vol. 
30
 (pg. 
847
-
854
)
26
Goodyear
 
O
Piper
 
K
Khan
 
N
, et al. 
CD8+ T cells specific for cancer germline gene antigens are found in many patients with multiple myeloma, and their frequency correlates with disease burden.
Blood
2005
, vol. 
106
 (pg. 
4217
-
4224
)
27
Chaux
 
P
Vantomme
 
V
Stroobant
 
V
, et al. 
Identification of MAGE-3 epitopes presented by HLA-DR molecules to CD4(+) T lymphocytes.
J Exp Med
1999
, vol. 
189
 (pg. 
767
-
778
)
28
Gooding
 
RP
Bybee
 
A
Cooke
 
F
, et al. 
Phenotypic and molecular analysis of six human cell lines derived from patients with plasma cell dyscrasia.
Br J Haematol
1999
, vol. 
106
 (pg. 
669
-
681
)
29
Van Der Bruggen
 
P
Zhang
 
Y
Chaux
 
P
, et al. 
Tumor-specific shared antigenic peptides recognized by human T cells.
Immunol Rev
2002
, vol. 
188
 (pg. 
51
-
64
)
30
Manici
 
S
Sturniolo
 
T
Imro
 
MA
, et al. 
Melanoma cells present a MAGE-3 epitope to CD4(+) cytotoxic T cells in association with histocompatibility leukocyte antigen DR11.
J Exp Med
1999
, vol. 
189
 (pg. 
871
-
876
)
31
Consogno
 
G
Manici
 
S
Facchinetti
 
V
, et al. 
Identification of immunodominant regions among promiscuous HLA-DR-restricted CD4+ T-cell epitopes on the tumor antigen MAGE-3.
Blood
2003
, vol. 
101
 (pg. 
1038
-
1044
)
32
Kobayashi
 
H
Song
 
Y
Hoon
 
DS
Appella
 
E
Celis
 
E
Tumor-reactive T helper lymphocytes recognize a promiscuous MAGE-A3 epitope presented by various major histocompatibility complex class II alleles.
Cancer Res
2001
, vol. 
61
 (pg. 
4773
-
4778
)
33
Schultz
 
ES
Lethe
 
B
Cambiaso
 
CL
, et al. 
A MAGE-A3 peptide presented by HLA-DP4 is recognized on tumor cells by CD4+ cytolytic T lymphocytes.
Cancer Res
2000
, vol. 
60
 (pg. 
6272
-
6275
)
34
Campbell
 
JD
Detection and enrichment of antigen-specific CD4+ and CD8+ T cells based on cytokine secretion.
Methods
2003
, vol. 
31
 (pg. 
150
-
159
)
35
Rajkumar
 
SV
Lacy
 
MQ
Kyle
 
RA
Monoclonal gammopathy of undetermined significance and smoldering multiple myeloma.
Blood Rev
2007
, vol. 
21
 (pg. 
255
-
265
)
36
Hamann
 
D
Kostense
 
S
Wolthers
 
KC
, et al. 
Evidence that human CD8+CD45RA+CD27- cells are induced by antigen and evolve through extensive rounds of division.
Int Immunol
1999
, vol. 
11
 (pg. 
1027
-
1033
)
37
Wills
 
MR
Okecha
 
G
Weekes
 
MP
Gandhi
 
MK
Sissons
 
PJ
Carmichael
 
AJ
Identification of naive or antigen-experienced human CD8(+) T cells by expression of costimulation and chemokine receptors: analysis of the human cytomegalovirus-specific CD8(+) T cell response.
J Immunol
2002
, vol. 
168
 (pg. 
5455
-
5464
)
38
Wills
 
MR
Carmichael
 
AJ
Weekes
 
MP
, et al. 
Human virus-specific CD8+ CTL clones revert from CD45ROhigh to CD45RAhigh in vivo: CD45RAhighCD8+ T cells comprise both naive and memory cells.
J Immunol
1999
, vol. 
162
 (pg. 
7080
-
7087
)
39
Carrasco
 
J
Godelaine
 
D
Van Pel
 
A
Boon
 
T
van der Bruggen
 
P
CD45RA on human CD8 T cells is sensitive to the time elapsed since the last antigenic stimulation.
Blood
2006
, vol. 
108
 (pg. 
2897
-
2905
)
40
Valmori
 
D
Scheibenbogen
 
C
Dutoit
 
V
, et al. 
Circulating tumor-reactive CD8(+) T cells in melanoma patients contain a CD45RA(+)CCR7(-) effector subset exerting ex vivo tumor-specific cytolytic activity.
Cancer Res
2002
, vol. 
62
 (pg. 
1743
-
1750
)
41
Long
 
HM
Haigh
 
TA
Gudgeon
 
NH
, et al. 
CD4+ T-cell responses to Epstein-Barr virus (EBV) latent-cycle antigens and the recognition of EBV-transformed lymphoblastoid cell lines.
J Virol
2005
, vol. 
79
 (pg. 
4896
-
4907
)
42
Pudney
 
VA
Leese
 
AM
Rickinson
 
AB
Hislop
 
AD
CD8+ immunodominance among Epstein-Barr virus lytic cycle antigens directly reflects the efficiency of antigen presentation in lytically infected cells.
J Exp Med
2005
, vol. 
201
 (pg. 
349
-
360
)
43
Germeau
 
C
Ma
 
W
Schiavetti
 
F
, et al. 
High frequency of antitumor T cells in the blood of melanoma patients before and after vaccination with tumor antigens.
J Exp Med
2005
, vol. 
201
 (pg. 
241
-
248
)
44
Lurquin
 
C
Lethe
 
B
De Plaen
 
E
, et al. 
Contrasting frequencies of antitumor and anti-vaccine T cells in metastases of a melanoma patient vaccinated with a MAGE tumor antigen.
J Exp Med
2005
, vol. 
201
 (pg. 
249
-
257
)
45
Andrade
 
VC
Vettore
 
AL
Felix
 
RS
, et al. 
Prognostic impact of cancer/testis antigen expression in advanced stage multiple myeloma patients.
Cancer Immun
2008
, vol. 
8
 (pg. 
2
-
9
)
46
Spisek
 
R
Kukreja
 
A
Chen
 
LC
, et al. 
Frequent and specific immunity to the embryonal stem cell-associated antigen SOX2 in patients with monoclonal gammopathy.
J Exp Med
2007
, vol. 
204
 (pg. 
831
-
840
)
47
Noonan
 
K
Matsui
 
W
Serafini
 
P
, et al. 
Activated marrow-infiltrating lymphocytes effectively target plasma cells and their clonogenic precursors.
Cancer Res
2005
, vol. 
65
 (pg. 
2026
-
2034
)
48
Dhodapkar
 
MV
Krasovsky
 
J
Osman
 
K
Geller
 
MD
Vigorous premalignancy-specific effector T cell response in the bone marrow of patients with monoclonal gammopathy.
J Exp Med
2003
, vol. 
198
 (pg. 
1753
-
1757
)
49
Waller
 
EC
McKinney
 
N
Hicks
 
R
Carmichael
 
AJ
Sissons
 
JG
Wills
 
MR
Differential costimulation through CD137 (4-1BB) restores proliferation of human virus-specific “effector memory” (CD28(-) CD45RA(HI)) CD8(+) T cells.
Blood
2007
, vol. 
110
 (pg. 
4360
-
4366
)
50
Monnier-Benoit
 
S
Mauny
 
F
Riethmuller
 
D
, et al. 
Immunohistochemical analysis of CD4+ and CD8+ T-cell subsets in high risk human papillomavirus-associated pre-malignant and malignant lesions of the uterine cervix.
Gynecol Oncol
2006
, vol. 
102
 (pg. 
22
-
31
)
51
de Jong
 
A
van Poelgeest
 
MI
van der Hulst
 
JM
, et al. 
Human papillomavirus type 16-positive cervical cancer is associated with impaired CD4+ T-cell immunity against early antigens E2 and E6.
Cancer Res
2004
, vol. 
64
 (pg. 
5449
-
5455
)
52
Corthay
 
A
Lundin
 
KU
Munthe
 
LA
, et al. 
Immunotherapy in multiple myeloma: Id-specific strategies suggested by studies in animal models.
Cancer Immunol Immunother
2004
, vol. 
53
 (pg. 
759
-
769
)
53
Schuler-Thurner
 
B
Schultz
 
ES
Berger
 
TG
, et al. 
Rapid induction of tumor-specific type 1 T helper cells in metastatic melanoma patients by vaccination with mature, cryopreserved, peptide-loaded monocyte-derived dendritic cells.
J Exp Med
2002
, vol. 
195
 (pg. 
1279
-
1288
)
54
Dhodapkar
 
MV
Krasovsky
 
J
Olson
 
K
T cells from the tumor microenvironment of patients with progressive myeloma can generate strong, tumor-specific cytolytic responses to autologous, tumor-loaded dendritic cells.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 (pg. 
13009
-
13013
)
55
Curioni-Fontecedro
 
A
Knights
 
AJ
Tinguely
 
M
, et al. 
MAGE-C1/CT7 is the dominant cancer-testis antigen targeted by humoral immune responses in patients with multiple myeloma.
Leukemia
2008
, vol. 
22
 (pg. 
1646
-
1648
)
Sign in via your Institution