Multiple myeloma (MM) is an incurable plasma cell malignancy characterized by immunosuppression. In this study, we identified factors in patients' bone marrow (BM) sera inhibiting autologous anti-MM immunity and developed an ex vivo strategy for inducing MM-specific cytotoxic T lymphocytes (CTLs). We found that sera from BM of MM patients inhibited induction of dendritic cells (DCs), evidenced by both phenotype and only weak stimulation of T-cell proliferation. Anti–vascular endothelial growth factor (anti-VEGF) and/or anti–interleukin 6 (anti–IL-6) antibodies neutralized this inhibitory effect, confirming that VEGF and IL-6, at least in part, mediate immunosuppression in MM patients. To induce MM-specific CTLs ex vivo, immature DCs were generated by culture of adherent mononuclear cells in medium containing granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-4 for 5 days and then cocultured with apoptotic MM bodies in the presence of tumor necrosis factor α (TNF-α) for 3 days to induce their maturation. Autologous BM or peripheral blood mononuclear cells were stimulated weekly with these DCs, and cytotoxicity was examined against the MM cells used to pulse DCs. DCs cultured with apoptotic bodies stimulated significantly greater T-cell proliferation (stimulation index [SI] = 23.2 at a T-DC ratio of 360:1) than T cells stimulated by MM cells only (SI = 5.6), DCs only (SI = 9.3), or MM lysate–pulsed DCs (SI = 13.5). These CTLs from MM patients demonstrated specific cytotoxicity (24.7% at the effector-target [E/T] ratio of 40:1) against autologous primary MM cells. These studies therefore show that CTLs from MM patients can recognize and lyse autologous tumor cells and provide the framework for novel immunotherapy to improve patient outcome in MM.

Multiple myeloma (MM) is currently incurable with conventional chemotherapy or high-dose chemotherapy with autologous stem cell support. Recent studies show that allogeneic stem cell transplantation may achieve prolonged progression-free survival compared with autografting1  and that donor lymphocyte infusion achieves responses in patients with relapsed MM after bone marrow transplantation.2,3  These reports suggest that a clinically significant immune-mediated allogeneic graft-versus-MM effect is inducible and provide the framework for development of immune-based therapeutic options.

Dendritic cells (DCs) play a pivotal role in tumor immunity.4  We have recently reported that DCs from MM patients may function normally in vitro,5  although others have shown decreased responsiveness to CD40 activation.6  Immature DCs are characterized by high-endocytic and -macropinocytic activity and low expression of accessory signals for T-cell activation; in contrast, mature DCs have enhanced T-cell stimulatory capacity in association with down-regulation of antigen uptake.7  Immature DCs in vivo efficiently phagocytose and process antigen derived from apoptotic bodies and generate peptide epitopes for cross-presentation on human leukocyte antigen (HLA) class I, which stimulates CD8+ T cells, after their maturation.7-9  Importantly, several factors that abrogate DC function in vitro, including transforming growth factor β1 (TGF-β1), interleukin 10 (IL-10),6  vascular endothelial growth factor (VEGF),10  and IL-6,11  are also implicated in MM pathogenesis in the bone marrow (BM). We therefore investigated in this study whether serum from BM of patients with MM inhibits both induction and maturation of DCs and delineated the potential role of cytokines mediating this inhibition.

In this study we also attempted to establish an effective DC-based method for generation of autologous cytotoxic T lymphocytes (CTLs) as the framework for vaccination therapy of MM. Although many DC-based vaccination strategies have been attempted, it remains controversial as to which antigen is optimal for DC loading. A single peptide directly binding to HLA molecules on DCs or an antigen expressed on tumor cells are candidate antigens, but these approaches are limited due both to HLA restriction of patients and selectivity of expression of antigen on tumor cells. For example, we established unique HLA-unrestricted CTLs against mucin-1 (MUC1) from patients with MM,12,13  but the approach is limited since MUC1 is expressed on only 55% ± 12% MM cells.14  The specific idiotypic determinant of immunoglobulin (Ig) variable regions is an ideal target antigen to overcome these limitations, since it is uniquely expressed on the malignant B-cell clone. After a clinically successful initial report in follicular lymphoma,15  many studies of vaccination with idiotype-pulsed DCs have been performed in patients with MM.16-19  Although idiotype-specific CTLs were inducible in these studies, their frequency was less than 50% and associated clinical responses were disappointing. Moreover, approaches using single antigens may also be limited due to cross-reactivity with normal cells or by evolution and escape of antigen-negative tumor cell clones.20 

In the current study, we generated mature DCs as antigen-presenting cells (APCs), which were pulsed with MM cell apoptotic bodies versus MM cell lysates to optimize antigen-presenting function and overcome limitations of single-antigen approaches. Our study shows that DCs pulsed with MM apoptotic bodies are significantly more effective at inducing CTLs against autologous MM cells than are DCs pulsed with MM cell lysates, providing the framework for related vaccination trials in MM.

Cell lines and cell culture

Human RPMI 8226 and U266 MM cell lines, as well as natural killer (NK) cell–sensitive human leukemia cell line K562, were obtained from American Type Culture Collection (Rockville, MD). All cell lines were cultured in RPMI 1640 (Mediatech, Herndon, VA) containing 10% fetal bovine serum (FBS; Harlan, Indianapolis, IN), 2 mM L-glutamine, 100 U/mL penicillin, and 100 μg/mL streptomycin (GIBCO, Grand Island, NY).

Sera and MM cells from patients' BM

BM specimens were acquired from patients with MM refractory or resistant to conventional chemotherapy (stage II or III) after obtaining informed consent and approval from the Dana-Farber Cancer Institute institutional review board for these studies. Sera from BM of MM patients were kept at –70°C until use. IL-6, IL-10, TGF-β1, and VEGF levels in sera were measured by enzyme-linked immunosorbent assay (ELISA; R&D Systems, Minneapolis, MN) according to the manufacturer's instructions. Primary MM cells (> 90% CD138+) were purified from BM specimens using RosetteSep negative selection system (StemCell Technologies, Vancouver, BC, Canada) per the manufacturer's protocol.

Induction of DCs and CTLs

Immature DCs were induced from healthy donor peripheral blood mononuclear cells (PBMCs) or BMMCs from MM patients as previously described5  with some modification. Briefly, mononuclear cells (MNCs) separated by Ficoll-Paque (Pharmacia, Piscataway, NJ) were suspended in RPMI 1640 medium containing 5% heat-inactivated human serum type AB (Sigma, St Louis, MO) or autologous serum and then incubated in 6-well culture plates (Becton Dickinson Labware, Franklin Lakes, NJ) for 2 hours at 37°C. After removing nonadherent cells, adherent cells (> 70% CD14+) were cultured in fresh medium containing 10% heat-inactivated human AB serum or autologous serum, recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF; 1000 U/mL; Immunex, Seattle, WA), and IL-4 (10 ng/mL; R&D Systems) for 5 days. In experiments to examine the effect of patient's serum on induction of DCs, 20% serum from the BM of patients was added instead of 10% AB serum, with or without neutralizing anti–IL-6 antibodies (Abs), anti-VEGF Abs (5 μg/mL; R&D System), or normal goat IgG (5 μg/mL; Oncogene Research Products, Boston, MA) as control. Recombinant human IL-6 (20 ng/mL) or VEGF (20 ng/mL; R&D Systems) in 10% AB serum was added to define its effects on DC production. Immature DCs were cocultured for 12 to 18 hours with irradiated (30 Gy) MM cell lines (2:1) or MM patient cells (5:1) and then cultured for 3 days in the presence of tumor necrosis factor α (TNF-α;10or 20 ng/mL; R&D Systems) to induce maturation. MM cell lysates, produced by repeating freezing and thawing 3 times, were pulsed to immature DCs at the same cell equivalent ratios as apoptotic bodies. MNCs from the same donors or patients were then cocultured with mature DCs at a ratio of 10:1 to 20:1 in AIM-V medium (Invitrogen, Carlsbad, CA) and stimulated with the apoptotic body–pulsed or cell lysate–pulsed DCs weekly. Seven days after the second stimulation, cytotoxicity of the CTLs was examined as described in “Cytotoxicity assay.”

Flow cytometric analysis

Cell surface phenotype was analyzed by flow cytometry using anti-CD1a, -CD14, -CD80, and -CD83 antibodies (Beckman Coulter, Fullerton, CA) for DCs, as well as anti-CD3, -CD4, -CD8, and -CD56 antibodies (Beckman Coulter) for CTLs. Cells were washed with phosphate-buffered saline (PBS) and incubated with fluorescein isothiocyanate (FITC)–conjugated or phycoerythrin (PE)–conjugated antibody for 20 minutes on ice. After washing with cold PBS once, cells were fixed with 1% paraformaldehyde (Sigma) and analyzed using a flow cytometer (Coulter Epics XL; Beckman Coulter).

Dual staining with annexin-V–FITC and propidium iodide (PI) was used to detect apoptosis, as in our prior study.21  Briefly, 1 × 106 MM cells were irradiated (30 Gy) and resuspended in 100 μL HEPES (N-2-hydroxyethylpiperazine-N′2-ethanesulfonic acid) buffer containing annexin-V–FITC and PI (annexin-V–FLUOS Staining Kit; Roche Diagnostics, Manheim, Germany) after incubation for 0, 2, 4, 6, 8, and 12 hours. Early apoptosis is defined by annexin-V–FITC+ and PI staining and detected by dual-fluorescence flow cytometry.

Phagocytosis assay

MM cells were stained red with Vybrant Dil Cell-Labeling Solution (Molecular Probes, Eugene, OR) according to the manufacturer's protocol and then irradiated (30 Gy) using Gammacell 1000 (Atomic Energy of Canada, Mississauga, ON, Canada). Immature DCs were stained green with Vybrant DiO Cell-Labeling Solution (Molecular Probes) and then cocultured with irradiated red MM cells at a ratio of 1:1 for 0, 2, 4, 6, and 8 hours at 37°C or 4°C. Phagocytosis of apoptotic bodies by immature DCs was defined by the percentage of double-positive cells detected by dual-fluorescence flow cytometry, as described previously.22 

Autologous T-cell proliferation assay

To examine the function of DCs cocultured with apoptotic bodies as APCs, autologous T-cell proliferation was measured by 3[H] thymidine incorporation assay, as previously described.23  Briefly, T cells (> 95% CD3+), obtained from PBMCs by high-affinity negative selection using T-cell enrichment columns (R&D Systems), were incubated in triplicate in 96-well round-bottom culture plates (2 × 105 cells/well) for 5 days with irradiated (15 Gy) autologous DCs at various T-DC ratios. 3[H]thymidine (1.0 μCi [0.037 MBq]) was added to each well for the last 12 hours of 5-day cultures. Cells were then harvested onto glass filters using an automatic cell harvester (Cambridge Technology, Cambridge, MA) and counted using a MicroBeta Trilux counter (Wallac, Gaithersburg, MD).

Cytotoxicity assay

Cytotoxicity of CTLs against MM cell lines or K562 cells was assessed with 51Cr-release assay, as previously described.12  Briefly, 5 × 103 target cells labeled with Na2CrO4 (Perkin Elmer Life Sciences, Boston, MA) were cocultured with effector cells in triplicate at various effector-target ratios in 96-well round-bottom tissue culture plates (Corning Inc, Corning, NY) for 4 hours at 37°C. Supernatants were then harvested with Supernatant Collection System (Molecular Devices, Sunnyvale, CA), and radioactivity was counted using a Wizard 3 gamma counter (Wallac). Percent-specific lysis was calculated as follows: ([experimental release – spontaneous release]/[maximum release – spontaneous release]) × 100. Spontaneous release was measured by incubating target cells in the medium alone, and maximum release was obtained by adding 1% Triton X-100 (Sigma) to target cells. For target blocking assays, target cells were added after incubation with blocking antibody against HLA class I (clone W6/32; Research Diagnostics, Flanders, NJ), HLA class II (clone G46-6; BD PharMingen, San Diego, CA), or normal mouse IgG (Oncogene Research Products) at a concentration of 10 μg/mL for 30 minutes at room temperature.

Specific cytotoxicity against primary MM cells was assessed by colorimetric lactate dehydrogenase (LDH) assay (CytoTox 96 Non-Radioactive Cytotoxicity Assay; Promega, Madison, WI), per manufacturer's instruction and as described for the 51Cr-release assay.

Statistical analysis

Statistical significance of differences observed in expression of surface antigens, proliferation of autologous T cells, and cytotoxicity of CTLs was determined using the Mann-Whitney U test. The minimal level of significance was P < .05.

Effects of patients' BM serum on induction and maturation of DCs

We first investigated the effects of serum from BM of MM patients on the induction of DCs especially focusing on VEGF and IL-6, which play important roles in MM pathogenesis.24-26  Adherent cells (> 70% CD14+) from a healthy donor were cultured in the presence of GM-CSF (1000 U/mL), IL-4 (10 ng/mL), and either 20% human AB serum or BM sera from 10 MM patients for 5 days to obtain immature DCs. Cells were then cultured in the presence of TNF-α (10 ng/mL) to induce their maturation for 3 additional days. Phenotypic analysis included staining of DCs for CD14 (monocyte/DC precursor), CD1a and CD80 (immature and mature DC), and CD83 (mature DC). Sera from MM patients' BM were analyzed for VEGF, IL-6, TGF-β1, and IL-10 by ELISA. As shown in Table 1, mean concentration of IL-6 and VEGF in sera from patients' BM is higher than that of AB serum used in this experiment, and DCs cultured with MM patients' BM sera express high levels of CD14 but low levels of CD1a, CD80, and CD83. There were no differences in concentrations of TGF-β1 and IL-10 (data not shown). These results indicate that sera from BM of patients with MM, which contain high concentrations of IL-6 and VEGF, inhibit induction and maturation of DCs from adherent cells.

Table 1.

Effects of sera from MM patients' BM on phenotype of DCs



Concentration of cytokines*

% Positivity

IL-6, pg/mL
VEGF, pg/mL
CD14
CD1a
CD80
CD83
MM Pt BM   47.0   1061   31.1   22.1   17.5   9.2  
AB serum
 
<4.7
 
180
 
7.7
 
55.1
 
55.8
 
48.8
 


Concentration of cytokines*

% Positivity

IL-6, pg/mL
VEGF, pg/mL
CD14
CD1a
CD80
CD83
MM Pt BM   47.0   1061   31.1   22.1   17.5   9.2  
AB serum
 
<4.7
 
180
 
7.7
 
55.1
 
55.8
 
48.8
 
*

Results shown are the mean of BM sera from 10 MM patients in duplicate experiments. Values below the detectability by ELISA (4.7 pg/mL for IL-6 and 62.5 pg/mL for VEGF) were read as 0 to calculate the mean.

Percentages of positive cells compared with isotype-matched control antibody are shown.

To define whether this inhibitory effect in BM serum was mediated via IL-6 (a major growth and survival factor for MM cells24,25 ) and VEGF (which stimulates MM cell growth and migration26  as well as BM angiogenesis27 ) we repeated these experiments in the presence of neutralizing anti-VEGF Abs (5 μg/mL) and/or anti–IL-6 Abs (5 μg/mL). Neither Ab affected the induction and maturation of DCs in the presence of human AB serum (data not shown); however, the addition of either of these Abs reduced CD14 and increased CD1a expression on immature DCs in the presence of MM patients' BM sera (Figure 1A). Moreover, addition of these Abs increased expression of CD83 on mature DCs (Figure 1B). These results suggest that VEGF and IL-6 in MM patients' BM sera mediate, at least in part, an inhibitory effect on the induction and maturation of DCs.

Figure 1.

Anti-VEGF and anti–IL-6 Abs reduce the inhibition of induction and maturation of DCs by MM patients' BM sera. (A) Immature DCs were induced from adherent healthy donor PBMCs by culturing in the presence of GM-CSF (1000 U/mL), IL-4 (10 ng/mL), and 20% MM patients' BM sera with control Ab (□), neutralizing anti–IL-6 Abs (5 μg/mL, ▨), anti-VEGF Abs (5 μg/mL, ▦), or both anti–IL-6 and VEGF Abs (▪). Cell surface phenotype was analyzed by flow cytometry using FITC- or PE-conjugated Abs for CD14 and CD1a. Percentage positivity is relative to an isotype-matched control Ab, and values indicate the mean ± SD of experiments with 6 MM patients' BM sera. Statistically significant decreased expression of CD14 (*, P = .01; **, P = .03) and increased expression of CD1a (*, P = .01) were observed with addition of MM patients' BM sera in the presence of anti–IL-6 and/or VEGF Abs. (B) Immature DCs induced in the presence of GM-CSF, IL-4, and 10% AB serum were then cultured for 3 days in the presence of 20% MM patients' BM sera, TNF-α (10 μg/mL), and either control Abs (□), neutralizing anti–IL-6 Abs (▨), anti-VEGF Abs (▦), or both anti–IL-6 and VEGF Abs (▪). Flow cytometric analysis was done using FITC-conjugated anti-CD83 Abs to assess maturation of DCs. Values indicate the mean ± SD of results from 6 MM patients. The expression of CD83 was significantly increased (*, P = .01) with addition of anti-VEGF Abs or both anti-VEGF and IL-6 Abs. (C) T cells (> 95% CD3+) obtained from the same PBMCs used for induction of DCs were incubated in 96-well round-bottom plates (2 × 105 cells/well) for 5 days with irradiated (15 Gy) autologous DCs, generated in media containing GM-CSF, IL-4, and 20% AB sera or MM patients' BM sera, with (▪) or without (□) neutralizing anti-VEGF Abs (5 μg/mL) and anti–IL-6 Abs (5 μg/mL) at indicated T-DC ratios. 3[H]thymidine (1.0 μCi [0.037 MBq]) was added to each well for the last 12 hours of 5-day cultures. Cells were then harvested and radioactivity was counted. Values indicate the mean ± SD of results from 5 triplicate experiments. T-cell proliferation was significantly inhibited with MM patients' BM sera compared with AB serum (*, P = .01), and this effect was neutralized by anti-VEGF and anti–IL-6 Abs (**, P = .01). Mean counts per minute (cpm) of T cells without stimulators was 454 and of DCs only was less than 100.

Figure 1.

Anti-VEGF and anti–IL-6 Abs reduce the inhibition of induction and maturation of DCs by MM patients' BM sera. (A) Immature DCs were induced from adherent healthy donor PBMCs by culturing in the presence of GM-CSF (1000 U/mL), IL-4 (10 ng/mL), and 20% MM patients' BM sera with control Ab (□), neutralizing anti–IL-6 Abs (5 μg/mL, ▨), anti-VEGF Abs (5 μg/mL, ▦), or both anti–IL-6 and VEGF Abs (▪). Cell surface phenotype was analyzed by flow cytometry using FITC- or PE-conjugated Abs for CD14 and CD1a. Percentage positivity is relative to an isotype-matched control Ab, and values indicate the mean ± SD of experiments with 6 MM patients' BM sera. Statistically significant decreased expression of CD14 (*, P = .01; **, P = .03) and increased expression of CD1a (*, P = .01) were observed with addition of MM patients' BM sera in the presence of anti–IL-6 and/or VEGF Abs. (B) Immature DCs induced in the presence of GM-CSF, IL-4, and 10% AB serum were then cultured for 3 days in the presence of 20% MM patients' BM sera, TNF-α (10 μg/mL), and either control Abs (□), neutralizing anti–IL-6 Abs (▨), anti-VEGF Abs (▦), or both anti–IL-6 and VEGF Abs (▪). Flow cytometric analysis was done using FITC-conjugated anti-CD83 Abs to assess maturation of DCs. Values indicate the mean ± SD of results from 6 MM patients. The expression of CD83 was significantly increased (*, P = .01) with addition of anti-VEGF Abs or both anti-VEGF and IL-6 Abs. (C) T cells (> 95% CD3+) obtained from the same PBMCs used for induction of DCs were incubated in 96-well round-bottom plates (2 × 105 cells/well) for 5 days with irradiated (15 Gy) autologous DCs, generated in media containing GM-CSF, IL-4, and 20% AB sera or MM patients' BM sera, with (▪) or without (□) neutralizing anti-VEGF Abs (5 μg/mL) and anti–IL-6 Abs (5 μg/mL) at indicated T-DC ratios. 3[H]thymidine (1.0 μCi [0.037 MBq]) was added to each well for the last 12 hours of 5-day cultures. Cells were then harvested and radioactivity was counted. Values indicate the mean ± SD of results from 5 triplicate experiments. T-cell proliferation was significantly inhibited with MM patients' BM sera compared with AB serum (*, P = .01), and this effect was neutralized by anti-VEGF and anti–IL-6 Abs (**, P = .01). Mean counts per minute (cpm) of T cells without stimulators was 454 and of DCs only was less than 100.

Close modal

We next examined the function of these DCs, induced in the presence of MM patients' BM sera with or without anti-VEGF Abs and anti–IL-6 Abs in an autologous T-cell proliferation assay. T cells (> 95% CD3+) were cocultured in AIM-V medium with DCs derived from irradiated (15 Gy) autologous adherent cells by culture for 5 days with GM-CSF (1000 U/mL), IL-4 (10 ng/mL), and 20% MM patients' BM sera or AB serum, followed by incubation with TNF-α (10 ng/mL) for 3 days. Proliferation was measured by 3[H] thymidine incorporation assay during the last 12 hours of 5-day cultures. As shown in Figure 1C, DCs cultured in the presence of MM patients' BM sera stimulated significantly less T-cell proliferation (stimulation index [SI] = 6.8 ± 1.7 at a T-DC ratio of 30:1) than DCs cultured in the presence of AB serum (SI = 36.3 ± 1.4, *P = .01). Importantly, addition of anti-VEGF and anti–IL-6 Abs neutralized these inhibitory effects on T-cell proliferation (SI = 27.6 ± 4.2, **P = .01), reflecting corresponding changes in DC phenotype.

Effects of exogenous VEGF and IL-6 on induction and maturation of DCs

To confirm this inhibitory effect of MM patients' BM sera, we next examined the effect of exogenous VEGF and IL-6 on DC induction. Healthy donor adherent cells were cultured in 10% human AB serum with GM-CSF and IL-4 for 5 days with or without VEGF (20 ng/mL) or IL-6 (20 ng/mL). TNF-α (10 ng/mL) was then added for 3 days and flow cytometric analysis was then performed. As shown in Figure 2A, CD14 expression increased, while both CD1a and CD83 decreased, on cells cultured with VEGF or IL-6 compared with cells cultured in media alone (*, ** P = .02, *** P = .046). We similarly studied the function of DCs induced in the presence of these cytokines using an autologous T-cell proliferation assay. Importantly, DCs generated in the presence of VEGF or IL-6 only weakly stimulated autologous T cells (SI = 45.0 ± 1.6 in control versus 31.9 ± 0.9 with VEGF and 20.7 ± 1.0 with IL-6 at a T-DC ratio of 30:1, * P = .02; Figure 2B). These results show that VEGF and IL-6 inhibit the maturation and function of DCs, confirming that these cytokines can account, at least in part, for immunosuppression in MM patients. Moreover, they further support the potential utility of generating anti-MM CTLs ex vivo.

Figure 2.

Exogenous VEGF and IL-6 inhibit induction and maturation of DCs. (A) DCs were induced from adherent healthy donor PBMCs by culturing in the presence of GM-CSF (1000 U/mL), IL-4 (10 ng/mL), and 10% AB serum (□), with VEGF (20 ng/mL, ▦) or IL-6 (20 ng/mL, ▪), and TNF-α (10 μg/mL) was then added for 3 days. Cell surface expression of CD14, CD1a, and CD83 on DCs was analyzed by flow cytometry. Results indicate the mean ± SD of percent-positive cells in 4 experiments. Statistically significant increased expression of CD14 (*,P = .02) and decreased expression of CD1a (**, P = .02) and CD83 (***, P = .046) were observed with addition of IL-6 or VEGF. (B) T cells (2 × 105 cells/well) from healthy donor PBMCs were incubated for 5 days with irradiated (15 Gy) autologous DCs generated in media containing 10% AB serum, GM-CSF, and IL-4 (□) with VEGF (20 ng/mL, ▦) or IL-6 (20 ng/mL, ▪) at indicated T-DC ratios. 3[H]thymidine (1.0 μCi [0.037 MBq]) was added to each well for the last 12 hours of 5-day cultures. Cells were then harvested and radioactivity was counted. Results are representative of 3 experiments, and values indicate mean ± SD of triplicate wells. Statistically significant decreased stimulation of T cells was observed with addition of VEGF or IL-6 (*, P = .02). Mean cpm of T cells without stimulators was 424 and of DCs only was less than 100.

Figure 2.

Exogenous VEGF and IL-6 inhibit induction and maturation of DCs. (A) DCs were induced from adherent healthy donor PBMCs by culturing in the presence of GM-CSF (1000 U/mL), IL-4 (10 ng/mL), and 10% AB serum (□), with VEGF (20 ng/mL, ▦) or IL-6 (20 ng/mL, ▪), and TNF-α (10 μg/mL) was then added for 3 days. Cell surface expression of CD14, CD1a, and CD83 on DCs was analyzed by flow cytometry. Results indicate the mean ± SD of percent-positive cells in 4 experiments. Statistically significant increased expression of CD14 (*,P = .02) and decreased expression of CD1a (**, P = .02) and CD83 (***, P = .046) were observed with addition of IL-6 or VEGF. (B) T cells (2 × 105 cells/well) from healthy donor PBMCs were incubated for 5 days with irradiated (15 Gy) autologous DCs generated in media containing 10% AB serum, GM-CSF, and IL-4 (□) with VEGF (20 ng/mL, ▦) or IL-6 (20 ng/mL, ▪) at indicated T-DC ratios. 3[H]thymidine (1.0 μCi [0.037 MBq]) was added to each well for the last 12 hours of 5-day cultures. Cells were then harvested and radioactivity was counted. Results are representative of 3 experiments, and values indicate mean ± SD of triplicate wells. Statistically significant decreased stimulation of T cells was observed with addition of VEGF or IL-6 (*, P = .02). Mean cpm of T cells without stimulators was 424 and of DCs only was less than 100.

Close modal

Immature DCs phagocytose MM apoptotic bodies

We next attempted to establish CTLs that recognize and lyse MM cells ex vivo. DCs can acquire antigen from apoptotic cells and present tumor antigen on HLA class I molecules, thereby inducing antigen-specific CTL responses,8,28  and we therefore pulsed immature DCs with MM apoptotic bodies. MM cells were first irradiated (30 Gy) and stained with annexin V–FITC and PI. As shown in Figure 3A, the annexin-V–FITC+ and PI cell fraction gradually increased from 0 to 12 hours, with transition to annexin-V+/PI+ cells. Trypan blue staining showed fewer than 10% viable cells at 48 hours and no viable cells at 72 hours after irradiation (30 Gy; data not shown). We next monitored uptake of irradiation-induced apoptotic bodies by immature DCs in a phagocytosis assay. Immature DCs (CD14/+, CD1a+/, CD83) were prepared by culturing adherent mononuclear cells with GM-CSF and IL-4 for 5 days and stained green; MM cells were red-stained and irradiated. Green DCs and red MM cells were cultured at a ratio of 1:1 at either 37°C or 4°C. After 0, 2, 4, 6, and 8 hours, these cocultured cells were analyzed using flow cytometry with quantification of phagocytic uptake reflected as double-positive cells. As shown in Figure 3B, immature DCs take up irradiated MM cells within several hours at 37°C but not 4°C. Uptake of apoptotic cells was confirmed by fluorescence microscopy (data not shown). These data indicate that immature DCs effectively phagocytose MM apoptotic cells at 37°C.

Figure 3.

Immature DCs phagocytose apoptotic MM bodies. (A) U266 cells were irradiated and stained with annexin-V–FITC and PI after 0, 2, 4, 6, 8, and 12 hours incubation at 37°C. Early apoptotic cells were defined as annexin-V–FITC+ and PI using flow cytometry. Results are representative of experiments with 3 MM cell lines. (B) U266 cells labeled red with Vybrant Dil Cell-Labeling Solution were incubated 4 hours at 37°C after 30-Gy irradiation to allow apoptosis to occur and then were cocultured with immature DCs stained green with Vybrant DiO Cell-Labeling Solution at a ratio of 1:1 for 0, 2, 4, 6, and 8 hours at 37°C or 4°C. Cells were analyzed by flow cytometry and double-positive cells indicate uptake of apoptotic cells by immature DCs. Culturing at 4°C blocked phagocytosis of apoptotic bodies by immature DCs.

Figure 3.

Immature DCs phagocytose apoptotic MM bodies. (A) U266 cells were irradiated and stained with annexin-V–FITC and PI after 0, 2, 4, 6, 8, and 12 hours incubation at 37°C. Early apoptotic cells were defined as annexin-V–FITC+ and PI using flow cytometry. Results are representative of experiments with 3 MM cell lines. (B) U266 cells labeled red with Vybrant Dil Cell-Labeling Solution were incubated 4 hours at 37°C after 30-Gy irradiation to allow apoptosis to occur and then were cocultured with immature DCs stained green with Vybrant DiO Cell-Labeling Solution at a ratio of 1:1 for 0, 2, 4, 6, and 8 hours at 37°C or 4°C. Cells were analyzed by flow cytometry and double-positive cells indicate uptake of apoptotic cells by immature DCs. Culturing at 4°C blocked phagocytosis of apoptotic bodies by immature DCs.

Close modal

Induction of MM cell line–specific CTLs

We next examined the ability of DCs pulsed with MM apoptotic cells and DCs pulsed with MM cell lysates to induce MM CTLs. Immature DCs were derived from adherent cells of HLA-A2–positive healthy donors by culturing with GM-CSF, IL-4, and 10% autologous serum for 5 days and then cocultured with HLA-A2–positive U266 cells that had been irradiated 4 hours earlier at a ratio of 2:1. After overnight incubation, TNF-α (20 ng/mL) was added to media and incubated for 3 days to induce maturation of DCs (CD14, CD83+). T cells from the same donors were cocultured with these irradiated (15 Gy) mature DCs in AIM-V medium, and proliferation was measured by 3[H]thymidine incorporation for the last 12 hours of 5-day cultures. As shown in Figure 4A, DCs cocultured with MM apoptotic bodies stimulated significantly (* P = .02) greater autologous T-cell proliferation (SI = 23.0 ± 2.0 at T/DC of 360:1) than MM cell lysate–pulsed DCs (SI = 13.9 ± 1.6), DCs alone (SI = 9.3 ± 1.1), or apoptotic MM cells alone (SI = 5.6 ± 0.4). Importantly, CTLs established by 2 weekly stimulations with MM apoptotic body–pulsed DCs efficiently lysed those MM cells used as stimulators, with only weak cytotoxicity in CTLs generated using other stimulators (Figure 4B). These data indicate that MM apoptotic body–pulsed DCs induce functional CTLs against MM cells.

Figure 4.

Stimulation and induction of CTLs by DCs pulsed with apoptotic MM bodies versus cell lysates. (A) T cells (2 × 105 cells) from healthy donor PBMCs were incubated for 5 days with irradiated (15 Gy) autologous DCs cocultured with apoptotic U266 bodies (▪), U266 lysate (□), DCs alone (▨), or apoptotic U266 bodies alone (▦) at indicated T cell–to-DC (T/DC) ratios. 3[H]thymidine (1.0 μCi [0.037 MBq]) was added to each well for the last 12 hours of 5-day cultures. Cells were then harvested and radioactivity was counted. Mean cpm of T cells without stimulator was 409 and of DCs only and apoptotic U266 cells only were less than 50. Data shown indicate mean ± SD of triplicate wells and are representative of 3 experiments. Significantly greater proliferation was observed in T cells stimulated with apoptotic U266–pulsed DCs than other stimuli by Mann-Whitney U test (*, P = .02). (B) Cytotoxicity of CTLs against U266 cells was assessed with 51Cr-release assay. U266 cells (5 × 103 cells) labeled with 51Cr were cultured with CTLs induced by stimulation with DCs cocultured with apoptotic U266 bodies (▪), U266 lysate (▴), DCs alone (□), or apoptotic U266 cells alone (▵) at indicated effector-to-target (E/T) ratios in triplicate for 4 hours at 37°C. Supernatants were then harvested and radioactivity was counted. CTLs stimulated with apoptotic U266 bodies showed significantly higher specific lysis than other stimuli. Spontaneous release of target cells was less than 10%. Specific lysis of K562 cells by CTLs stimulated with DCs cocultured with apoptotic U266 bodies was maximum (14.8%) at an E/T ratio of 20:1. Results shown are mean ± SD of triplicate wells and representative of 3 experiments.

Figure 4.

Stimulation and induction of CTLs by DCs pulsed with apoptotic MM bodies versus cell lysates. (A) T cells (2 × 105 cells) from healthy donor PBMCs were incubated for 5 days with irradiated (15 Gy) autologous DCs cocultured with apoptotic U266 bodies (▪), U266 lysate (□), DCs alone (▨), or apoptotic U266 bodies alone (▦) at indicated T cell–to-DC (T/DC) ratios. 3[H]thymidine (1.0 μCi [0.037 MBq]) was added to each well for the last 12 hours of 5-day cultures. Cells were then harvested and radioactivity was counted. Mean cpm of T cells without stimulator was 409 and of DCs only and apoptotic U266 cells only were less than 50. Data shown indicate mean ± SD of triplicate wells and are representative of 3 experiments. Significantly greater proliferation was observed in T cells stimulated with apoptotic U266–pulsed DCs than other stimuli by Mann-Whitney U test (*, P = .02). (B) Cytotoxicity of CTLs against U266 cells was assessed with 51Cr-release assay. U266 cells (5 × 103 cells) labeled with 51Cr were cultured with CTLs induced by stimulation with DCs cocultured with apoptotic U266 bodies (▪), U266 lysate (▴), DCs alone (□), or apoptotic U266 cells alone (▵) at indicated effector-to-target (E/T) ratios in triplicate for 4 hours at 37°C. Supernatants were then harvested and radioactivity was counted. CTLs stimulated with apoptotic U266 bodies showed significantly higher specific lysis than other stimuli. Spontaneous release of target cells was less than 10%. Specific lysis of K562 cells by CTLs stimulated with DCs cocultured with apoptotic U266 bodies was maximum (14.8%) at an E/T ratio of 20:1. Results shown are mean ± SD of triplicate wells and representative of 3 experiments.

Close modal

Characterization of CTLs induced by MM apoptotic body–pulsed DCs

To characterize reactivity of CTLs induced by repeated stimulations with mature DCs pulsed with apoptotic MM bodies, we examined their cytotoxicity against cells other than those used for stimulation. CTLs induced in this manner from healthy donors were cocultured with 51Cr-labeled target cells for 4 hours, and radioactivity of supernatants was compared with maximal release. As shown in Figure 5A, CTLs efficiently lysed those MM cells used for pulsing DCs, but not unrelated MM cells and an NK cell–sensitive cell line. We next investigated the mechanism of recognition of target cells in a 51Cr-release assay by incubating target cells with neutralizing antibodies to HLA class I or class II. Cytotoxicity of CTLs against MM cells was significantly inhibited by anti–HLA class I Abs but not anti–HLA DR or control Abs (Figure 5B). These results indicate that cytotoxicity of CTLs is specific for target MM cells and restricted to HLA class I. Phenotypic analysis revealed that CTLs were mainly CD3+ and CD8+, with only a minor population of CD56+ cells (Figure 5C). This result confirmed that the cytotoxicity was mediated via CTLs but not NK cells.

Figure 5.

Characterization of CTLs induced by stimulation with mature DCs cocultured with apoptotic MM cells. (A) CTLs induced by stimulations with DCs cocultured with apoptotic U266 bodies showed significantly higher cytotoxicity (*, P = .02) against U266 cells (▪) than against RPMI 8226 cells (□) or K562 cells (▦), assessed with 51Cr-release assay at indicated effector-to-target (E/T) ratios for 4 hours at 37°C. Spontaneous release of target cells was less than 15%. Results shown are mean ± SD of triplicate wells and representative of 3 experiments. (B) HLA restriction of CTLs was examined using target blocking 51Cr-release assay. CTLs stimulated with apoptotic U266 body–pulsed DCs were cocultured with 51Cr-labeled U266 after incubation with blocking antibody against HLA class I (□), HLA class II (▨), control antibody (▦), or without antibody (▪). Cytotoxicity was significantly inhibited by anti–HLA class I Abs (*, P = .02). Spontaneous release of target cells was less than 15%. Results shown are mean ± SD of triplicate wells and representative of 2 experiments. (C) Cell surface phenotype of CTLs was analyzed by flow cytometry using FITC- or PE-conjugated Abs against CD3, CD8, CD4, and CD56 (filled histogram). Percentages positive are calculated relative to an isotype-matched control Ab (open histogram).

Figure 5.

Characterization of CTLs induced by stimulation with mature DCs cocultured with apoptotic MM cells. (A) CTLs induced by stimulations with DCs cocultured with apoptotic U266 bodies showed significantly higher cytotoxicity (*, P = .02) against U266 cells (▪) than against RPMI 8226 cells (□) or K562 cells (▦), assessed with 51Cr-release assay at indicated effector-to-target (E/T) ratios for 4 hours at 37°C. Spontaneous release of target cells was less than 15%. Results shown are mean ± SD of triplicate wells and representative of 3 experiments. (B) HLA restriction of CTLs was examined using target blocking 51Cr-release assay. CTLs stimulated with apoptotic U266 body–pulsed DCs were cocultured with 51Cr-labeled U266 after incubation with blocking antibody against HLA class I (□), HLA class II (▨), control antibody (▦), or without antibody (▪). Cytotoxicity was significantly inhibited by anti–HLA class I Abs (*, P = .02). Spontaneous release of target cells was less than 15%. Results shown are mean ± SD of triplicate wells and representative of 2 experiments. (C) Cell surface phenotype of CTLs was analyzed by flow cytometry using FITC- or PE-conjugated Abs against CD3, CD8, CD4, and CD56 (filled histogram). Percentages positive are calculated relative to an isotype-matched control Ab (open histogram).

Close modal

CTLs recognize and lyse autologous MM cells

We finally examined induction of CTLs that can specifically lyse autologous MM cells. Immature DCs were induced from adherent cells in BMMCs from MM patients by culturing in medium with 10% AB serum, GM-CSF, and IL-4 for 5 days. These immature DCs were cocultured overnight with irradiated MM cells (5:1) obtained from the same patient. TNF-α was then added for 3 days to induce DC maturation. BM or peripheral MNCs from the same patient were cocultured with mature DCs at a ratio of 10:1 to 20:1 in AIM-V medium. Seven days after second stimulation, cytotoxicity of CTLs against primary MM cells was analyzed using the LDH-release assay due to low 51Cr uptake by patient MM cells. As shown in Figure 6A, CTLs stimulated by mature DCs pulsed with patient MM apoptotic bodies effectively lysed autologous MM cells but not an unrelated MM cell line or NK cell–sensitive cells. In addition, CTLs from patient (Pt) 1 showed only slight cytotoxicity against RPMI 8226 cells, even though both produce λ light chain, indicating that CTLs do not have cross-reactivity with this isotype-matched cell line. Flow cytometric analysis revealed that CTLs consisted primarily of CD8+ T cells with only a small population of CD4+ T cells and CD56+ NK cells (Figure 6B). To further characterize cytotoxicity of CTLs induced from MM patients we next established CTLs from HLA-A2–positive patients by repeated stimulation with apoptotic body–pulsed DCs and examined for specific lysis in the presence or absence of neutralizing antibodies to HLA class I or class II. As shown in Figure 6C, cytotoxicity of CTLs against autologous primary MM cells was significantly inhibited by anti–HLA class I Abs but not anti–HLA DR or control Abs. These results indicate that cytotoxicity of CTLs is restricted to HLA class I. We also investigated their cytotoxicity against HLA-A2–positive irrelevant U266 cells, NK cell–sensitive K562 cells, autologous PBMCs, and primary MM cells using the LDH assay. As shown in Figure 6D, CTLs lysed autologous primary MM cells, but not U266, K562, or autologous PBMCs, showing that cytotoxicity of CTLs is specific for target MM cells.

Figure 6.

Induction and characterization of CTLs against autologous MM cells. (A) Immature DCs were induced from adherent cells in BMMCs of patients with MM (Pt 1 [λ-type] and 2 [IgA, κ]) by culturing with GM-CSF (1000 U/mL), IL-4 (10 ng/mL), and 10% AB serum, followed by coculture for 12 to 18 hours at a ratio of 5:1 with autologous MM cells incubated for 4 hours after irradiation (30 Gy) and then cultured for 3 days in the presence of TNF-α (20 ng/mL) to induce maturation. MNCs from the same MM patients were then cocultured with mature DCs at a ratio of 10:1 to 20:1 in AIM-V medium and stimulated with the apoptotic body–pulsed DCs weekly. Seven days after the second stimulation, cytotoxicity of the CTL against autologous MM cells (▪), RPMI 8226 cells (□), or K562 cells (▦) was examined using a lactate dehydrogenase assay at indicated effector-to-target (E/T) ratios. Significantly greater specific lysis of autologous MM cells was observed than of RPMI 8226 cells or K562 cells (*, P = .02). Spontaneous release of target cells was less than 20%. Results shown are mean ± SD of triplicate wells. (B) Cell surface phenotype of patient-derived CTLs was analyzed by flow cytometry using specific Abs against CD3, CD8, CD4, and CD56 (filled histogram). Percentages positive cells are calculated relative to an isotype-matched control Ab (open histogram). (C) HLA restriction of CTLs derived from patients with MM (Pt 3, nonsecretory; Pt 4, κ-type MM) was examined using target-blocking LDH assay. CTLs stimulated with apoptotic MM body–pulsed DCs were cocultured with autologous MM cells after incubation with neutralizing antibody against HLA class I (□), HLA class II (▨), control antibody (▦), or without antibody (▪). Cytotoxicity against autologous MM cells was significantly inhibited by anti–HLAclass IAbs (*, P = .02). Spontaneous release of target cells was less than 20%. Results shown are mean ± SD of triplicate wells from 2 representative patient-derived CTLs. (D) Cytotoxicity of CTLs induced by stimulation with DCs cocultured with apoptotic primary MM bodies of HLA-A2–positive patient against autologous MM cells (▪), U266 cells (▴), K562 cells (▵), or autologous PBMCs (□) was assessed with LDH assay at indicated effector-to-target (E/T) ratios for 4 hours at 37°C. CTLs induced by stimulations with DCs cocultured with primary MM bodies showed significantly greater cytotoxicity against autologous MM cells than against U226 cells, K562 cells, or autologous PBMCs. Spontaneous release of target cells was less than 20%. Results shown are mean ± SD of triplicate wells.

Figure 6.

Induction and characterization of CTLs against autologous MM cells. (A) Immature DCs were induced from adherent cells in BMMCs of patients with MM (Pt 1 [λ-type] and 2 [IgA, κ]) by culturing with GM-CSF (1000 U/mL), IL-4 (10 ng/mL), and 10% AB serum, followed by coculture for 12 to 18 hours at a ratio of 5:1 with autologous MM cells incubated for 4 hours after irradiation (30 Gy) and then cultured for 3 days in the presence of TNF-α (20 ng/mL) to induce maturation. MNCs from the same MM patients were then cocultured with mature DCs at a ratio of 10:1 to 20:1 in AIM-V medium and stimulated with the apoptotic body–pulsed DCs weekly. Seven days after the second stimulation, cytotoxicity of the CTL against autologous MM cells (▪), RPMI 8226 cells (□), or K562 cells (▦) was examined using a lactate dehydrogenase assay at indicated effector-to-target (E/T) ratios. Significantly greater specific lysis of autologous MM cells was observed than of RPMI 8226 cells or K562 cells (*, P = .02). Spontaneous release of target cells was less than 20%. Results shown are mean ± SD of triplicate wells. (B) Cell surface phenotype of patient-derived CTLs was analyzed by flow cytometry using specific Abs against CD3, CD8, CD4, and CD56 (filled histogram). Percentages positive cells are calculated relative to an isotype-matched control Ab (open histogram). (C) HLA restriction of CTLs derived from patients with MM (Pt 3, nonsecretory; Pt 4, κ-type MM) was examined using target-blocking LDH assay. CTLs stimulated with apoptotic MM body–pulsed DCs were cocultured with autologous MM cells after incubation with neutralizing antibody against HLA class I (□), HLA class II (▨), control antibody (▦), or without antibody (▪). Cytotoxicity against autologous MM cells was significantly inhibited by anti–HLAclass IAbs (*, P = .02). Spontaneous release of target cells was less than 20%. Results shown are mean ± SD of triplicate wells from 2 representative patient-derived CTLs. (D) Cytotoxicity of CTLs induced by stimulation with DCs cocultured with apoptotic primary MM bodies of HLA-A2–positive patient against autologous MM cells (▪), U266 cells (▴), K562 cells (▵), or autologous PBMCs (□) was assessed with LDH assay at indicated effector-to-target (E/T) ratios for 4 hours at 37°C. CTLs induced by stimulations with DCs cocultured with primary MM bodies showed significantly greater cytotoxicity against autologous MM cells than against U226 cells, K562 cells, or autologous PBMCs. Spontaneous release of target cells was less than 20%. Results shown are mean ± SD of triplicate wells.

Close modal

In the present study, we show that sera from BM of patients with MM inhibit both induction and maturation of DCs, mediated at least in part by VEGF and IL-6, suggesting the potential utility of generating anti-MM CTLs ex vivo. We therefore establish a strategy to induce CTLs recognizing autologous MM cells that is widely applicable and does not depend upon expression of either selective tumor antigens or patient HLA type.

We first demonstrate that VEGF and IL-6 in sera from BM of MM patients can inhibit the induction of DCs from adherent mononuclear cells, evidenced by their weak APC function as stimulators of T-cell proliferation. Our results confirm and extend previous investigations. Specifically, differentiation of CD34+ cells to DCs is inhibited by exogenous IL-6,11  or by IL-6 produced by renal cell and colon carcinoma cells,29,30  due to macrophage rather than DC differentiation.31  VEGF similarly inhibits the maturation of DCs in mice.10  IL-6 induces growth, survival, and drug resistance in MM cells24,25,32,33 ; VEGF triggers growth and migration of MM cells,26  stimulates BM angiogenesis,27  and augments IL-6 production in bone marrow stromal cells (BMSCs).34  In addition to these effects on MM cells and BMSCs in the BM microenvironment, the present results further suggest that both cytokines contribute to immune deficits characteristic of MM. Conversely, suppression of VEGF and IL-6 may enhance anti-MM immunity. For example, our studies show that thalidomide and immunomodulatory derivatives (IMiDs) act not only directly on MM cells35  but also in the BM microenvironment to inhibit up-regulation of IL-6 and VEGF secretion triggered by binding of MM cells to BMSCs.36  Importantly, these drugs augment in vitro and in vivo NK-cell activity against MM cells.23  Given that DCs directly trigger NK-cell function,37  studies are ongoing to define the effects of these agents on DC induction, maturation, and function.

We have shown that TGF-β1 is secreted by MM cells and can augment IL-6 secretion in BMSCs.38  Others have reported that TGF-β1 and IL-10 inhibit peripheral DC function in MM by down-regulating expression of CD80.6  We therefore examined the role of these cytokines in mediating the DC inhibition observed. Our results show that concentrations of TGF-β1 and IL-10 in MM patients' BM sera are equivalent to those in AB serum or in normal peripheral blood,6  0 to 20 ng/mL and 0 to 30 ng/mL, respectively. Although it remains possible that TGF-β1 and IL-10 or other humoral factors contribute to inhibiting DC function in MM, since even patients' serum with low levels of VEGF and IL-6 blocked DC maturation, generation of DCs ex vivo can overcome this inhibitory effect.

We next carried out in vitro studies to provide the framework for a vaccination therapy protocol using DCs and apoptotic tumor bodies to establish MM specific CTL ex vivo. Although DCs are the most effective APCs for vaccination strategy,4  it remains controversial as to which antigens are optimal for DC loading. DC pulsing with an HLA-binding single peptide from a tumor antigen is restricted by HLA type of patients and candidate MM antigens. Idiotype protein is uniquely produced by the malignant clone of MM cells and therefore a potential ideal antigen for vaccination. Based on clinical responses after idiotype vaccination in follicular lymphoma,15  several clinical trials of vaccination with idiotype-pulsed dendritic cells have been performed in patients with MM.16-19  In these studies, idiotype-specific CTLs are inducible in vitro; however, clinical responses after injecting idiotype-pulsed immature DCs are disappointing,19  possibly due to inhibition of DC maturation by inhibitory VEGF and IL-6. In this regard, vaccination using DCs that have matured and cannot revert to immature DCs or monocytes39  appears to be promising.40 

Compared with vaccinations using a single tumor antigen or molecule, strategies pulsing DCs with tumor cell apoptotic bodies have the potential advantage of presenting known, as well as unknown, tumor antigens. We therefore examined MM patient apoptotic body–pulsed DCs, as well as MM cell lysate–pulsed DCs, as stimulators of MM specific autologous CTLs. Uptake of apoptotic bodies by DCs is required for cross-presentation of antigens to induce HLA class I–restricted CD8+ T cells.8  We therefore pulsed apoptotic bodies to immature DCs, which phagocytose apoptotic bodies more efficiently than mature DCs.7  Apoptotic bodies, produced by irradiation of MM cells at doses (30 Gy) sufficient to prevent MM cell survival, were then cultured with DCs for 3 days. We determined that addition of apoptotic bodies to DCs were optimal at 4 hours after irradiation when the apoptotic cell population was increasing and there were few necrotic cells, which might either induce maturation of DC,41  resulting in ineffective uptake of apoptotic bodies,7  or stimulate both CD4+ and CD8+ T cells.42  We show that MM apoptotic body–pulsed DCs are more effective at triggering autologous MM specific CTLs than MM lysate–pulsed DCs. Immature DCs are known to effectively phagocytose apoptotic bodies via various receptors including phosphtidylserine receptor, vitronectin receptor,43  CD36,44  and αVβ5 integrin within a few hours after pulsing7  and then cross-present tumor antigen on their HLA class I molecule.8  In contrast, cell lysates are captured by DCs via macropinocytosis and mannose receptor and are then transferred to major histocompatibility complex (MHC) class II–rich vesicles.39  These differences may account, at least in part, for the superiority of apoptotic body–pulsed DCs in our study and are consistent with other recent studies showing superiority of apoptotic bodies versus cell lysates in promoting cross-priming of CTLs in squamous-cell carcinoma28  and pancreatic cancer,45  as well as in virus-infected cells.8 

In summary, in this study we provide the rationale for ex vivo generation of DCs, due to inhibitory factors in MM BM serum for their induction and maturation, and also develop a protocol to generate CTLs recognizing autologous MM cells using these DCs pulsed with MM apoptotic bodies ex vivo. This strategy is broadly applicable since it is not dependent upon expression of selective tumor antigens or patient HLA–type restriction, and therefore provides the framework for novel immunotherapy to improve patient outcome in MM.

Prepublished online as Blood First Edition Paper, April 24, 2003; DOI 10.1182/blood-2002-09-2828.

Supported by National Institutes of Health Grant PO-1 78378, the Doris Duke Distinguished Clinical Research Scientist Award, Myeloma Research Fund, Cure for Myeloma Fund (K.C.A.), and Multiple Myeloma Research Foundation (T. Hayashi, T. Hideshima, D.C.).

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
Reynolds C, Ratanatharathorn V, Adams P, et al. Allogeneic stem cell transplantation reduces disease progression compared to autologous transplantation in patients with multiple myeloma.
Bone Marrow Transplant.
2001
;
27
:
801
-807.
2
Alyea E, Weller E, Schlossman R, et al. T-cell-depleted allogeneic bone marrow transplantation followed by donor lymphocyte infusion in patients with multiple myeloma: induction of graft-versus-myeloma effect.
Blood.
2001
;
98
:
934
-939.
3
Lokhorst HM, Schattenberg A, Cornelissen JJ, Thomas LL, Verdonck LF. Donor leukocyte infusions are effective in relapsed multiple myeloma after allogeneic bone marrow transplantation.
Blood.
1997
;
90
:
4206
-4211.
4
Banchereau J, Steinman RM. Dendritic cells and the control of immunity.
Nature.
1998
;
392
:
245
-252.
5
Raje N, Gong J, Chauhan D, et al. Bone marrow and peripheral blood dendritic cells from patients with multiple myeloma are phenotypically and functionally normal despite the detection of Kaposi's sarcoma herpesvirus gene sequences.
Blood.
1999
;
93
:
1487
-1495.
6
Brown RD, Pope B, Murray A, et al. Dendritic cells from patients with myeloma are numerically normal but functionally defective as they fail to up-regulate CD80 (B7–1) expression after huCD40LT stimulation because of inhibition by transforming growth factor-β1 and interleukin-10.
Blood.
2001
;
98
:
2992
-2998.
7
Albert ML, Pearce SFA, Francisco LM, et al. Immature dendritic cells phagocytose apoptotic cells via αvβ5 and CD36, and cross-present antigens to cytotoxic T lymphocytes.
J Exp Med.
1998
;
188
:
1359
-1368.
8
Albert ML, Sauter B, Bhardwaj N. Dendritic cells acquire antigen from apoptotic cells and induce class I-restricted CTLs.
Nature.
1998
;
392
:
86
-89.
9
Kurts C, Cannarile M, Klebba I, Brocker T. Dendritic cells are sufficient to cross-present self-antigens to CD8 T cells in vivo.
J Immunol.
2001
;
166
:
1439
-1442.
10
Gabrilovich DI, Chen HL, Girgis KR, et al. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells.
Nat Med.
1996
;
2
:
1096
-1103.
11
Ratta M, Fagnoni F, Curti A, et al. Dendritic cells are functionally defective in multiple myeloma: the role of interleukin-6.
Blood.
2002
;
100
:
230
-237.
12
Takahashi T, Makiguchi Y, Hinoda Y, et al. Expression of MUC1 on myeloma cells and induction of HLA-unrestricted CTL against MUC1 from a multiple myeloma patient.
J Immunol.
1994
;
153
:
2102
-2109.
13
Noto H, Takahashi T, Makiguchi Y, Hayashi T, Hinoda Y, Imai K. Cytotoxic T lymphocytes derived from bone marrow mononuclear cells of multiple myeloma patients recognize an underglycosylated form of MUC1 mucin.
Int Immunol.
1997
;
9
:
791
-798.
14
Treon SP, Maimonis P, Bua D, et al. Elevated soluble MUC1 levels and decreased anti-MUC1 antibody levels in patients with multiple myeloma.
Blood.
2000
;
96
:
3147
-3153.
15
Bendandi M, Gocke CD, Kobrin CB, et al. Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma.
Nat Med.
1999
;
5
:
1171
-1177.
16
Lim SH, Bailey-Wood R. Idiotypic protein-pulsed dendritic cell vaccination in multiple myeloma.
Int J Cancer.
1999
;
83
:
215
-222.
17
Reichardt VL, Okada CY, Liso A, et al. Idiotype vaccination using dendritic cells after autologous peripheral blood stem cell transplantation for multiple myeloma: a feasibility study.
Blood.
1999
;
93
:
2411
-2419.
18
Liso A, Stockerl-Goldstein KE, Auffermann-Gretzinger S, et al. Idiotype vaccination using dendritic cells after autologous peripheral blood progenitor cell transplantation for multiple myeloma.
Biol Blood Marrow Transplant.
2000
;
6
:
621
-627.
19
Titzer S, Christensen O, Manzke O, et al. Vaccination of multiple myeloma patients with idiotype-pulsed dendritic cells: immunological and clinical aspects.
Br J Haematol.
2000
;
108
:
805
-816.
20
Thurner B, Haendle I, Roder C, et al. Vaccination with mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma.
J Exp Med.
1999
;
190
:
1669
-1678.
21
Hayashi T, Hideshima T, Akiyama M, et al. Arsenic trioxide inhibits growth of human multiple myeloma cells in the bone marrow microenvironment.
Mol Cancer Ther.
2002
;
1
:
851
-860.
22
Hess KL, Babcock GF, Askew DS, Cook-Mills JM. A novel flow cytometric method for quantifying phagocytosis of apoptotic cells.
Cytometry.
1997
;
27
:
145
-152.
23
Davies FE, Raje N, Hideshima T, et al. Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma.
Blood.
2001
;
98
:
210
-216.
24
Ogata A, Chauhan D, Teoh G, et al. Interleukin-6 triggers cell growth via the Ras-dependent mitogen-activated protein kinase cascade.
J Immunol.
1997
;
159
:
2212
-2221.
25
Hideshima T, Chauhan D, Teoh G, et al. Characterization of signaling cascades triggered by human interleukin-6 versus Kaposi's sarcoma-associated herpes virus-encoded viral interleukin 6.
Clin Cancer Res.
2000
;
6
:
1180
-1189.
26
Podar K, Tai YT, Davies FE, et al. Vascular endothelial growth factor triggers signaling cascades mediating multiple myeloma cell growth and migration.
Blood.
2001
;
98
:
428
-435.
27
D'Amato RJ, Loughnan MS, Flynn E, Folkman J. Thalidomide is an inhibitor of angiogenesis.
Proc Natl Acad Sci U S A.
1994
;
91
:
4082
-4085.
28
Hoffmann TK, Meidenbauer N, Dworacki G, Kanaya H, Whiteside TL. Generation of tumor-specific T-lymphocytes by cross-priming with human dendritic cells ingesting apoptotic tumor cells.
Cancer Res.
2000
;
60
:
3542
-3549.
29
Menetrier-Caux C, Montmain G, Dieu MC, et al. Inhibition of the differentiation of dendritic cells from CD34(+) progenitors by tumor cells: role of interleukin-6 and macrophage colony-stimulating factor.
Blood.
1998
;
92
:
4778
-4791.
30
Sombroek CC, Stam AG, Masterson AJ, et al. Prostanoids play a major role in the primary tumor-induced inhibition of dendritic cell differentiation.
J Immunol.
2002
;
168
:
4333
-4343.
31
Chomarat P, Banchereau J, Davoust J, Palucka AK. IL-6 switches the differentiation of monocytes from dendritic cells to macrophages.
Nat Immunol.
2000
;
1
:
510
-514.
32
Chauhan D, Hideshima T, Rosen S, Reed JC, Kharbanda S, Anderson KC. Apaf-1/cytochrome c-independent and Smac-dependent induction of apoptosis in multiple myeloma (MM) cells.
J Biol Chem.
2001
;
276
:
24453
-24456.
33
Hideshima T, Nakamura N, Chauhan D, Anderson KC. Biologic sequelae of interleukin-6 induced PI3-K/Akt signaling in multiple myeloma.
Oncogene.
2001
;
20
:
5991
-6000.
34
Dankbar B, Padro T, Leo R, et al. Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma.
Blood.
2000
;
95
:
2630
-2636.
35
Hideshima T, Chauhan D, Shima Y, et al. Thalidomide and its analogs overcome drug resistance of human multiple myeloma cells to conventional therapy.
Blood.
2000
;
96
:
2943
-2950.
36
Gupta D, Treon SP, Shima Y, et al. Adherence of multiple myeloma cells to bone marrow stromal cells upregulates vascular endothelial growth factor secretion: therapeutic applications.
Leukemia.
2001
;
15
:
1950
-1961.
37
Fernandez NC, Lozier A, Flament C, et al. Dendritic cells directly trigger NK cell functions: cross-talk relevant in innate anti-tumor immune responses in vivo.
Nat Med.
1999
;
5
:
405
-411.
38
Urashima M, Ogata A, Chauhan D, et al. Transforming growth factor-β1: differential effects on multiple myeloma versus normal B cells.
Blood.
1996
;
87
:
1928
-1938.
39
Sallusto F, Cella M, Danieli C, Lanzavecchia A. Dendritic cells use macropinocytosis and the mannose receptor to concentrate macromolecules in the major histocompatibility complex class II compartment: downregulation by cytokines and bacterial products.
J Exp Med.
1995
;
182
:
389
-400.
40
Yi Q, Desikan R, Barlogie B, Munshi N. Optimizing dendritic cell-based immunotherapy in multiple myeloma.
Br J Haematol.
2002
;
117
:
297
-305.
41
Sauter B, Albert ML, Francisco L, Larsson M, Somersan S, Bhardwaj N. Consequences of cell death: exposure to necrotic tumor cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells.
J Exp Med.
2000
;
191
:
423
-434.
42
Inaba K, Turley S, Yamaide F, et al. Efficient presentation of phagocytosed cellular fragments on the major histocompatibility complex class II products of dendritic cells.
J Exp Med.
1998
;
188
:
2163
-2173.
43
Fadok VA, Savill JS, Haslett C, et al. Different populations of macrophages use either the vitronectin receptor or the phosphatidylserine receptor to recognize and remove apoptotic cells.
J Immunol.
1992
;
149
:
4029
-4035.
44
Fadok VA, Warner ML, Bratton DL, Henson PM. CD36 is required for phagocytosis of apoptotic cells by human macrophages that use either a phosphatidylserine receptor or the vitronectin receptor (αvβ3).
J Immunol.
1998
;
161
:
6250
-6257.
45
Schnurr M, Scholz C, Rothenfusser S, et al. Apoptotic pancreatic tumor cells are superior to cell lysates in promoting cross-priming of cytotoxic T cells and activate NK and γδ T Cells.
Cancer Res.
2002
;
62
:
2347
-2352.
Sign in via your Institution