Intranodal Administration of Peptide-Pulsed Mature Dendritic Cell Vaccines Results in Superior CD8+ T-Cell Function in Melanoma Patients

2003 ◽  
Vol 21 (20) ◽  
pp. 3826-3835 ◽  
Author(s):  
Isabelle Bedrosian ◽  
Rosemarie Mick ◽  
Shuwen Xu ◽  
Harvey Nisenbaum ◽  
Mark Faries ◽  
...  

Purpose: We evaluated the feasibility, safety, and immunogenicity of mature, peptide-pulsed dendritic cell (DC) vaccines administered by different routes. Patients and Methods: We performed a randomized, phase I, dose-escalation study in 27 patients with metastatic melanoma receiving four autologous peptide-pulsed DC vaccinations. Patients were randomly assigned to an intravenous (IV), intranodal (IN), or intradermal (ID) route of administration (ROA). For each route, primary end points were dose-limiting toxicity, maximum-tolerated dose, and T-cell sensitization. Sensitization was evaluated through tetramer staining, in vitro peptide recognition assays, and delayed-type hypersensitivity (DTH) responses. Results: Twenty-two (81.5%) of 27 patients completed all four vaccinations. Vaccinations were well tolerated; a few patients exhibited grade 1 to 2 toxicities including rash, fever, and injection site reaction. All routes of administration induced comparable increases in tetramer-staining CD8+ T cells (five of seven IV, four of seven IN, and four of six ID patients). However, the IN route induced significantly higher rates for de novo development of CD8+ T cells that respond by cytokine secretion to peptide-pulsed targets (six [85.7%] of seven IN patients v two [33%] of six ID patients v none [0%] of six IV patients; P = .005) and de novo DTH (seven [87.5%] of eight IN patients v two [33.3%] of six ID patients v one [14.3%] of seven IV patients; P = .01) compared with other routes. Conclusion: Administration of this peptide-pulsed mature DC vaccine by IN, IV, or ID routes is feasible and safe. IN administration seems to result in superior T-cell sensitization as measured by de novo target-cell recognition and DTH priming, indicating that IN may be the preferred ROA for mature DC vaccines.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 571-571
Author(s):  
Sarah Herby ◽  
Matthew Milliron ◽  
Crystal Mackall ◽  
Terry Fry

Abstract Background: Although allogeneic graft versus tumor responses represent the most potent form of immunotherapy, recurrent malignancy remains a major cause of post-transplant mortality. Thus, strategies to enhance GVT are needed to improve outcomes. Dendritic cell (DC) vaccines can potently enhance anti-tumor immunity in the autologous setting. The impact of alloreactivity on DC vaccine responses is not known, but the potency of the GVT effect suggests that mild alloreactivity might enhance vaccine responses to third party antigens. Methods: To determine the effect of a minor histocompatibility mismatch on DC vaccine responses, lethally irradiated female C3H.SWxC57BL/6 and C3H.SW recipients were transplanted with T cell depleted (TCD) C3H.SW bone marrow (BM). On days 14 and 28, recipients received donor-type lymph node cells containing mature T cells (DLI, 1 x 10^6, 5 x 10^6, or 10 x 10^6 IV). To immunize against the male antigenic complex (HY), BM-derived, anti-CD40 activated donor strain male DC were administered IP at the same time as the DLI. At Day 35, HY specific immune responses were measured using interferon γ ELISPOT against the dominant (UTY) and subdominant (SMCY) class I epitopes and dominant class II epitope (DBY). In some experiments, rhIL-7 and rhIL-15 were administered (5 mcg/day IP) from day 14 to 35. Results: In syngeneic recipients, robust HY responses were observed at all DLI doses, consistent with rapid T cell reconstitution via the thymus. Allogeneic recipients demonstrated mild weight loss and decreased splenic cellularity as the DLI dose was increased, consistent with the development of mild GVHD. However, even recipients receiving the highest DLI dose recovered completely and had none of the major physical symptoms of GVHD. Increasing DLI dose from 1 x 10^6 to 10 x 10^6 paradoxically led to a marked decrease in the median total number of T cells/spleen responding to UTY, SMCY, and DBY (9196 at 1 x 10^6 vs 897 at 10 x 10^6, p = .01; 15662 vs 2854, p= .02; 10257 vs 4227, p = .02). To test whether the presence of alloreactive T cells were necessary for this effect, C3H.SW minor antigen reactive T cells were deleted from B6-derived DLI. TCD B6 BM was transplanted into C3H.SWxC57BL/6 mice and at 5 weeks LN cells were collected and transferred as DLI. Strikingly, transfer of 10 x 10^6 tolerized DLI did not result in a loss of HY vaccine responses when compared to transfer of non-tolerized DLI (UTY, 3888 vs 21075, p= .05; SMCY, 7135 vs 21126, p = .018; DBY, 7117 vs 29201, p= .006). Although IL-7 and IL-15 can potently enhance DC vaccine responses in the syngeneic setting, these cytokines did not improve vaccine responses in allogeneic recipients. Conclusions: Using a minor mismatched allogeneic transplant model, we have shown that mild GVHD can diminish the ability to respond to third party antigens expressed on activated DC vaccines. This suggests that the potency of GVT does not result from the alloreactive milieu, but rather from the nature of the antigens targeted. The mechanisms involved in the suppression of DC vaccine responses in allogeneic recipients are currently under study. These findings have important implications for the use of tumor antigen expressing vaccines as a modality to improve the graft versus tumor effect.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3020-3020
Author(s):  
Shayela Suvarna ◽  
Emily K.E. McCracken ◽  
Gowthami M. Arepally

Abstract Heparin-Induced Thrombocytopenia (HIT) is a drug-dependent immune disorder caused by autoantibodies to Platelet Factor 4 (PF4) and heparin. The immune basis of HIT is poorly understood. Recent studies describing transient antibody responses and absence of immunologic memory in HIT suggest that PF4/heparin autoantibodies may develop independently of T cell help. To investigate the cellular basis of the HIT immune response, we have developed a murine autoimmune model in which anti-murine PF4/heparin (anti-mP+H) arise de novo. Cohorts of BALB/c mice were immunized daily either intravenously (IV, n=10) or intraperitoneally (IP, n=10) for five days with complexes of murine (m) PF4/heparin (IV, n=5 or IP, n=5), heparin alone (IV, n= 2) or buffer (IV, n=3 or IP, n=5). Mice were screened for anti-mP+H for four weeks after immunization using a murine PF4/heparin ELISA. Peak antibody responses to antigen were seen at 11–15 days in 2/5 mice injected with IV mP+H (Day 11, mouse IV P+H #0 peak A450nm =0.34±0.01; Day15 mouse IV P+H #2 peak A450nm =0.69±0.01), and at days 22–25 in 2/5 mice injected by IP route (Day 22, mouse IP P+H #0 peak A450nm =0.37±0.01; Day 25 mouse IP P+H #2 peak A450nm =0.78±0.02). Anti-mP+H were not detected in mice injected with heparin alone or buffer alone at any time point (peak maximum IV A450nm= 0.1±0.001, Control #2; mouse IP P+H Control #2 A450nm =0.04±0.002). Serologically, murine autoantibodies were similar to anti-human (h) PF4/heparin. Anti-mP+H reactivity was specific for murine antigen (mouse IV P+H #2 A450nm=0.65±0.06), and was reduced with antigen in the presence of excess heparin (A450nm=0.38±0.01). Minimal reactivity was seen with wells coated with hP+H (A450nm=0.09±0.005), albumin (mouse IV P+H #2 A450nm=0.15±0.03), or PBS alone (mouse IV P+H #2 A450nm =0.16±0.01). Similar to human HIT antibodies, anti-mP+H were of IgG1 subclass. To determine if T cells are required for development of anti-mP+H, mice lacking T cell function (BIG:BALB/c-Nu, n=10) were injected IV with mP+H daily for five days. Unlike euthymic mice, nude mice did not manifest any antibody responses to IV injections of mP+H. In summary, we have developed a novel murine autoimmune model of anti-PF4/heparin that recapitulates many salient features of the human immune syndrome. Using this murine model, we demonstrate that T cells are essential for development PF4/heparin autoantibodies. Studies are currently underway to delineate mechanisms of T cell regulation and peripheral tolerance in HIT.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 957-957
Author(s):  
Christina Lutz-Nicoladoni ◽  
Patrizia Stoizner ◽  
Magdalena Pircher ◽  
Stephanie Wallner ◽  
Anna Maria Wolf ◽  
...  

Abstract Abstract 957 Introduction: Various approaches to induce immunological rejection of tumors including transfer of autologous tumor infiltrating lymhocytes (TIL) after ex vivo clonal expansion or application of ex vivo transduced antigen specific T cell (TCR) transgenic T cells have been elaborated. In general, adoptive T cell transfer (ATC) has been combined with lympho-depleting agents (e.g. cyclophosphamide). However, the therapeutic efficacy of these cancer immunotherapy approaches is limited due to insufficient in vivo activation, expansion and survival of transferred effector immune cells, which is mainly due to suppressive mileu signals and immune evasion mechanisms induced by TGF-β. The E3 ubiquitin ligase Cbl-b is a key regulator of T cell activation and is assumed to confer TGF-β resistance. Thus we performed a proof-of-concept study evaluating Cbl-b targeting as “intracellular adjuvant” strategy to improve ATC for cancer immunotherapy. Material and Methods: We first tested the in vitro sensitivity of CTL towards TGF-β mediated immuno-suppressive cues and then in vivo evaluated the anti-tumor reactivity of cblb-deficient cytotoxic T lymphocytes (CTL) in murine tumor models alone or in combination with a dendritic cell (DC) vaccine. Results: Cblb-deficient CTL are hyper-responsive to TCR/CD28-stimulation in vitro and protected from the negative cues induced by TGF-β as determined by quantification fo IFN-g secretion and quantification of their proliferative capacity. Unexpectedly, adoptive transfer of polyclonal, non TCR-transgenic cblb-deficient CD8+ CTL, however, is not sufficient to reject B16ova or EG7 tumors in vivo, which is in clear contrast to previous reports using lymphopenic animals receiving adoptively transferred TCR-transgenic T cells. Thus, we next evaluated in vivo re-activation of adoptively transferred cblb-deficient T cells by a DC vaccine (i.e. SIINFEKL-pulsed DC). In strict contrast to ATC monotherapy, this approach now markedly delays tumor outgrowth and significantly increase survival rates, which is paralleled by an increased CTL infiltration rate to the tumor site and an enrichment of ova-specific and IFN-g-secreting CTL in the draining lymph nodes. Moreover, compared to wild-type CTL, cblb-deficient mice vaccinated with the DC vaccine show an increased cytolytic activity in vivo. Conclusions: In summary, we provide experimental evidence that genetic inactivation of cblb in polyclonal, non-TCR transgenic adoptively transferred CTL might serve as a novel “adjuvant approach”, suitable to augment the effectiveness of anti-cancer immunotherapies using ATC in immune-competent recipients. Disclosures: No relevant conflicts of interest to declare.


2018 ◽  
Author(s):  
Bidesh Mahata ◽  
Jhuma Pramanik ◽  
Louise van der Weyden ◽  
Krzysztof Polanski ◽  
Gozde Kar ◽  
...  

ABSTRACTTumors subvert immune cell function to evade immune responses, yet the complex mechanisms driving immune evasion remain poorly understood. Here we show that tumors induce de novo steroidogenesis in T lymphocytes to evade anti-tumor immunity. Using a novel transgenic steroidogenesis-reporter mouse line we identify and characterize de novo steroidogenic immune cells. Genetic ablation of T cell steroidogenesis restricts primary tumor growth and metastatic dissemination in mouse models. Steroidogenic T cells dysregulate anti-tumor immunity, and inhibition of the steroidogenesis pathway was sufficient to restore anti-tumor immunity. This study demonstrates T cell de novo steroidogenesis as a mechanism of anti-tumor immunosuppression and a potential druggable target.


Vaccines ◽  
2020 ◽  
Vol 8 (1) ◽  
pp. 25 ◽  
Author(s):  
Caroline Boudousquié ◽  
Valérie Boand ◽  
Emilie Lingre ◽  
Laeticia Dutoit ◽  
Klara Balint ◽  
...  

With the emergence of immune checkpoint inhibitors and adoptive T-cell therapies, there is a considerable interest in using personalized autologous dendritic cell (DC) vaccines in combination with T cell-targeting immunotherapies to potentially maximize the therapeutic impact of DC vaccines. Here, we describe the development and optimization of a Good Manufacturing Practice (GMP)-compliant manufacturing process based on tumor lysate as a tumor antigen source for the production of an oxidized tumor cell lysate loaded DC (OC-DC) vaccine. The manufacturing process required one day for lysate preparation and six days for OC-DC vaccine production. Tumor lysate production was standardized based on an optimal tumor digestion protocol and the immunogenicity was improved through oxidation using hypochloric acid prior to freeze-thaw cycles resulting in the oxidized tumor cell lysate (OC-L). Next, monocytes were selected using the CliniMACS prodigy closed system and were placed in culture in cell factories in the presence of IL-4 and GM-CSF. Immature DCs were loaded with OC-L and matured using MPLA-IFNγ. After assessing the functionality of the OC-DC cells (IL12p70 secretion and COSTIM assay), the OC-DC vaccine was cryopreserved in multiple doses for single use. Finally, the stability of the formulated doses was tested and validated. We believe this GMP-compliant DC vaccine manufacturing process will facilitate access of patients to personalized DC vaccines, and allow for multi-center clinical trials.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 6535-6535
Author(s):  
Robert John Walsh ◽  
Emily Pauline Nickles ◽  
Yating Li ◽  
Bhushan Dharmadhikari ◽  
Mickey Koh ◽  
...  

6535 Background: Epstein-Barr virus (EBV) is associated with non-keratinising (NK) NPC, a disease prevalent in Southeast Asia, and provides a potential target for dendritic cell (DC) vaccine therapy. CD137 ligand (CD137L) expressed on antigen presenting cells costimulates CD137 expressing T cells upon receptor/ligand interaction. CD137L signalling differentiates monocytes to CD137L-DC, a novel type of DC, which are more potent than classical DC in stimulating autologous T cells. Here, we explore the safety and efficacy of autologous CD137L-DC pulsed with EBV peptides spanning Epstein Barr nuclear antigen 1, latent membrane protein 1 (LMP1) and LMP2 (CD137L-DC-EBV-VAX) in patients with locally recurrent or metastatic NPC. Methods: In this single centre, phase I study, eligible patients (pts) with locally recurrent or metastatic NK-NPC and clinical benefit (CB) from their prior treatment (stable disease [SD], partial [PR] or complete response[CR]), underwent apheresis to isolate monocytes which were differentiated to CD137L-DC through CD137L agonist exposure. CD137L-DC were pulsed with EBV antigens during maturation to obtain CD137L-DC-EBV-VAX which was administered intradermally every 2 weeks (w) for up to 7 injections following site preconditioning with Tetanus and Diphtheria vaccine. Results: 14 pts were enrolled of which 2 progressed rapidly and did not begin treatment. Mean age was 58 years. Median lines of prior treatment for metastatic NPC was 1 (range 1-6), the most common being cisplatin and gemcitabine. 9 pts received 7 vaccine doses (range 2-7) with a mean administered cell count of 23.9x106. CB was seen in 5 cases (42%) with 1 PR and 4 SD beyond 1 year. Median progression free survival (mPFS) was 26w (95% CI, 23-43). The lowest PFS (8w) was in a pt with 6 prior lines of treatment including a checkpoint inhibitor. Mean pretreatment neutrophil: lymphocyte ratio (NLR) was 3.4 and a value of less than 3 was associated with prolonged mPFS (42 vs 14w, p = 0.01). Enzyme linked immune absorbent spot (ELISPOT) analysis in 5 pts with CB showed a rise in interferon-γ secreting peripheral T cells prior to the 3rd vaccine versus baseline. Treatment was well tolerated with only 4 cases of grade 1 related adverse events reported, most commonly injection site reaction (3pts). Conclusions: CD137L-DC-EBV-VAX is safe and exhibits promising efficacy when administered following CB from chemotherapy. A rise in activated peripheral blood mononuclear cells after 2 vaccinations in selected patients showing benefit suggests immunological correlates with efficacy. Clinical trial information: NCT03282617 .


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii39-ii39
Author(s):  
Oleg Yegorov ◽  
Changlin Yang ◽  
Anjelika Dechkovskaia ◽  
Maryam Rahman ◽  
Ashley Ghiaseddin ◽  
...  

Abstract BACKGROUND The application of single cell sequencing as a novel immune monitoring platform can be used to identify the molecular mechanisms of immune response to dendritic cell- based vaccines, trace the cell types and states involved, and uncover novel biomarkers for immunotherapy. We applied single-cell RNA Seq analysis of longitudinal peripheral blood mononuclear cells (PBMCs) in patients with newly-diagnosed GBM enrolled on the ATTAC II clinical trial (FDA IND BB-16530; Clinicaltrials.gov # NCT02465268) who experienced a sustained radiographic response to autologous CMV pp65-LAMP RNA-pulsed DC vaccines plus GM-CSF and tetanus-diphtheria booster administered during adjuvant cycles of dose-intensified temozolomide. METHODS We constructed 5’ gene expression libraries and T cell receptor enriched libraries for 10x Genomics single-cell 5’ and VDJ sequencing, generated from PBMCs collected prior to and during patient immunization using dendritic cells loaded with messenger ribonucleic acid encoding the human cytomegalovirus (CMV) matrix protein pp65 conjugated with the lysosomal associated membrane protein (LAMP) sequence. RESULTS Overall, we sequenced a total of 189,808 single-cell transcriptomes from 5 patients. We leveraged these transcriptome-wide features to distinguish 15 peripheral immune cell subtypes in tested PBMCs. Analysis revealed dynamic changes in immune cell subsets over the course of first three vaccines, including increases in cytotoxic CD8 T cells, CD4 T cells, and NK cell subsets. Increased markers of T cell activation were observed during vaccination. Surprisingly, we observed a very high-level frequency of natural killer T (NKT) cells in the patient with a complete durable response compared to other patients. After three DC vaccines, the level of NKT cells in PBMC of this patient increased up to 10%. CONCLUSIONS These results emphasize the importance of subset specific profiling to achieve higher resolution in monitoring immune responses compared with bulk expression profiling in patients receiving immunotherapeutic treatment.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1954-1954
Author(s):  
Caroline J Duncan ◽  
Peter R. E. Johnson ◽  
Huw Roddie

Abstract Dendritic cell (DC) vaccines in leukaemia show promise as a novel treatment modality aimed at reducing relapse rates by augmenting immune responses when patients are in a state of minimal residual disease. DC vaccination studies to date have not been as successful as hoped and this may be because of insufficient cytotoxic T cell (CTL) responses. Immunomodulatory agents such as 4-IBB ligand, and cytokines such as IL2, IL6 and GM-CSF have been reported to improve vaccine therapy however these agents have to be tolerated clinically and this limits the use of some preparations. Our study aims to generate a more efficient DC vaccine process by using Lenalidomide, a thalidomide analogue and immunomodulatory agent which is a powerful potentiator of CTLs and natural killer cells. It has the advantage of clinical tolerability and is already in use for treating haematological malignancies. In this in vitro study DCs were generated from monocytes of eight patients in remission following chemotherapy for acute myeloid leukaemia (AML) or chronic myeloid leukaemia (CML) with standard cytokine protocols. The immature DCs were loaded with autologous leukaemia cells from the patients’ presentation samples. The presentation leukaemia cells were treated with UVB irradiation to induce apoptosis. DCs were matured with TNF alpha for two days then co-cultured with autologous T cells for one week with or without the addition of Lenalidomide at ten micromolar. A control group of unstimulated T cells were kept in culture for the same time period. The T cells from the three groups were harvested and their cytoxicity assessed in an Interferon Gamma (IFNγ) ELISPOT assay where the stimulators used were the unmodified blasts. Results in all eight patients show the leukaemia specific CTL responses (as measured by IFNγ release) were markedly improved in the DC/irradiated blasts groups compared with the unstimulated T cells. In six out of eight patients there was a further, significant improvement in the groups where Lenalidomide was added (see Figure 1). Figure Figure These results show promise for the use Lenalidomide as an agent to optimise CTL responses to DC based Immunotherapy for myeloid leukaemia and should be investigated further in a clinical trial.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A296-A296
Author(s):  
Frauke Schnorfeil ◽  
Christiane Geiger ◽  
Iris Bigalke ◽  
Dag Josefsen ◽  
Yngvar Floisand ◽  
...  

BackgroundA Phase I/II dendritic cell (DC) vaccine trial was completed in 20 patients with acute myeloid leukemia (AML) in complete remission or CRi after chemotherapy who were ineligible for hematopoietic stem cell transplantation (NCT02405338). The DC vaccines were designed to delay disease progression by mobilizing natural killer (NK) cells through secretion of IL-12(p70) and activating T cells by stimulation with WT-1 and PRAME, two prominent antigens in AML. DC vaccination was carried out in weeks 1, 2, 3, 4, 6 and monthly thereafter for 2 years. Two questions were prominent at the trial start. First, could mature DCs (mDCs) be efficiently prepared to accommodate the vaccine regimen, including use of separate DC-fractions for each antigen. Second, could suitable quality DC vaccines be generated from patients with myeloid disease, since all had received intensive chemotherapy, impairing hematopoiesis, such that several patients showed extended times for monocyte recovery in peripheral blood before being able to undergo apheresis for production.MethodsImmune monitoring tools were used to assess DC vaccines: multi-color flow cytometry for surface and intracellular protein staining, dual-color ELISpot for secretion of IL-10/IL-12, and chemokine-directed trans-well migration.ResultsAdequate regeneration of monocytes occurred post-chemotherapy in all patients, allowing production of sufficient numbers of cryopreserved vaccine cells (2.5 or 5.0 × 106 mDCs/antigen/ampule) to be completed. In 15/20 patients one batch was sufficient to cover all vaccinations, while 5 patients with lower initial monocyte counts required an additional production.Phenotypic and functional parameters of patient DC vaccines were compared to cells of a healthy control (HC). Patient mDCs expressed CD83, CD40, CD80, CD86 and HLA-DR at frequencies/levels comparable to the HC. Both DC-fractions displayed intracellular protein antigen expression in most cells. Polarized secretion of IL-12(p70) without IL-10 was seen with few exceptions. Furthermore, mDCs displayed chemokine-directed migration. Detection of delayed type hypersensitivity responses post-vaccination at six weeks indicated the DC vaccines were active in vivo in all patients.ConclusionsDC vaccine production feasibility was clearly fulfilled and high quality mDCs were generated for every patient. Quantity and quality of DC vaccines did not differ in the patient groups that relapsed or remained in remission, nor in patients who succumbed to disease during the trial. DC vaccines were remarkably consistent, although originating from patients differing in age, AML subtype and receiving varied amounts of standard chemotherapy regimens.Ethics ApprovalThe study was approved by the responsible Norwegian ethics committee, approval number 2014/1677.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3466-3466
Author(s):  
Qing Yi ◽  
Siqing Wang ◽  
Michele Wezeman ◽  
Jianfei Qian ◽  
Jing Yang

Abstract Idiotype (Id) protein secreted by myeloma cells is the best-characterized tumor-specific antigen and has been used in clinical vaccination trials administered either as Id-keyhole limpet hemocyanin (KLH) protein vaccines or as Id-pulsed dendritic cell (DC) vaccines. In this study, we used the 5TGM1 myeloma murine model to compare the efficacy of Id-KLH (protein vaccines) versus DCs pulsed with Id-KLH (DC vaccines), two formulas commonly used in clinical trials, to induce tumor-specific immunity, to protect mice from developing myeloma, and to treat myeloma-bearing mice. Vaccinations consisted of three weekly, subcutaneous injections of the protein conjugates (100 μg/injection) or Id-KLH-pulsed, bone marrow-derived mature DCs (106 cells/injection). Following each vaccination, GM-CSF (200 ng/day/mouse) was injected subcutaneously (adjacent to the vaccination sites) for three consecutive days. We found that, although the protein vaccines induced significantly higher titers of specific antibodies, DC vaccines were superior at inducing tumor-specific, cellular immune responses, evident by increased IFN-γ production, T cell proliferation, and cytotoxic T cell activities against the tumor cells. As prophylactic treatments, protein and DC vaccines were equally efficient at protecting mice from subsequent tumor challenge. However, only DC vaccines induced therapeutic immunity in tumor-bearing mice: DC vaccinations not only retarded tumor growth but also eradicated established tumors in more than 50% of mice, which was associated with an induction of potent, tumor-specific type-1 (IFN-γ) and type-2 (IL-4) T-cell responses. DC-vaccinated mice also showed a more pronounced increase in the percentages of CD8+ T cells in their spleens after in vitro stimulation with irradiated myeloma cells and increased tumor-specific cytotoxicity in enriched CD8+ T cells over the whole splenic cells, suggesting that CD8+ T cells play a major role in killing tumor cells in vivo. Furthermore, surviving mice were also protected from rechallenge with the myeloma cells, indicating that memory T cells existed and were functional. Thus, our results show that Id-based DC vaccines may be a favorable vaccine for immunotherapy in MM. Further studies are required to optimize DC vaccination protocols to achieve therapeutic efficacy in clinical trials.


Sign in / Sign up

Export Citation Format

Share Document