4,571
Views
44
CrossRef citations to date
0
Altmetric
Reviews

Clinical trials of dendritic cell-based cancer vaccines in hematologic malignancies

, &
Pages 3125-3131 | Received 11 Jun 2014, Accepted 05 Oct 2014, Published online: 27 Jan 2015

Abstract

The potential for the immune system to target hematological malignancies is demonstrated in the allogeneic transplant setting, where durable responses can be achieved. However, allogeneic transplantation is associated with significant morbidity and mortality related to graft versus host disease. Cancer immunotherapy has the capacity to direct a specific cytotoxic immune response against cancer cells, particularly residual cancer cells, in order to reduce the likelihood of disease relapse in a more targeted and tolerated manner. Ex vivo dendritic cells can be primed in various ways to present tumor associated antigen to the immune system, in the context of co-stimulatory molecules, eliciting a tumor specific cytotoxic response in patients. Several approaches to prime dendritic cells and overcome the immunosuppressive microenvironment have been evaluated in pre-clinical and early clinical trials with promising results. In this review, we summarize the clinical data evaluating dendritic cell based vaccines for the treatment of hematological malignancies.

Abbreviations

CTLA-4=

Cytotoxic T-Lymphocyte Antigen 4

PD-L1=

Programmed death-ligand 1

PD-1=

Programmed death 1

DNA=

Deoxyribonucleic acid

RNA=

Ribonucleic acid

ASCT=

Autologous Stem Cell Transplant

OS=

Overall Survival

Id=

Idiotype

IL=

Interleukin

CML=

Chronic Myeloid Leukemia

WT-1=

Wilm's tumor suppressor gene 1

AML=

Acute Myeloid Leukemia

mRNA=

mRNA

GMCSF=

Granulocyte macrophage colony-stimulating factor

MDS=

Myelodysplastic syndrome

GVHD=

Graft vs Host Disease

SCT=

Stem cell transplant

KLH=

Keyhole limpet hemocyanin

Apo-DC=

Apoptotic body loaded- dendritic cells

IFN=

Interferon

IFNg=

Interferon gamma

HLA-A*2402=

Human Leukocyte antigen A*2402

CR=

Complete response

PR=

Partial response

DC/MM=

Dendritic cell Multiple Myeloma fusion vaccine

VGPR=

Very good partial response

DC/AML=

Dendritic cell Acute Myeloid Leukemia fusion vaccine

FLT-ITD=

Fms-like Tyrosine Kinase with Internal Tandem Duplication

VEGF=

Vascular endothelial growth factor

TGFB=

Transforming growth factor β

pDCs=

Plasmacytoid Dendritic cell

TNFα=

Tumor necrosis factor α

PRR=

Pathogen recognition receptor

MHC=

Major histocompatibility complex

Introduction

While cytotoxic chemotherapy can effectively reduce a patient's tumor burden at the time of presentation, patients typically suffer significant treatment related toxicities due to the non-specific effects of these drugs.Citation1 Furthermore, patients often subsequently succumb to relapsed disease, thought to be due to residual cancer cells that are inherently resistant to traditional cytotoxic therapy.Citation2 Cancer immunotherapy has the capacity to simultaneously overcome these 2 problems, directing a specific cytotoxic immune response against cancer cells, particularly residual cancer cells, in order to reduce the likelihood of disease relapse in a more targeted and tolerated manner.Citation3

The advent of allogeneic bone marrow transplantation in the 1960s, with its potent and in some cases curative graft vs. disease effect,Citation4 further demonstrated the potential of the immune system to eliminate malignant cells. In the setting of leukemia, it has been demonstrated that the risk of disease relapse following allogeneic transplantation is inversely related to the development of graft versus host disease, demonstrating both the potency and toxicity of alloreactive lymphocytes.Citation5 Moreover, relapsed disease following allogeneic transplantation can be eradicated in a subset of patients by the infusion of donor derived lymphocytes.Citation6 However graft vs. host disease remains a major cause of morbidity and mortality following allogeneic transplantation. As such, immunotherapeutic approaches that may generate tumor specific immunity have the potential to dramatically improve outcomes of patients with hematologic malignancies.

A number of factors contribute to the ability of tumor cells to evade immune recognition. Tumor cells present antigens in the relative absence of co-stimulatory molecules such as CD80 and CD86Citation7,8 required for the activation of effector T cells, resulted in a blunted T cell response. Secondly, the immunosuppressive tumor micro-environment consisting of increased numbers of T-regulatory cells,Citation9 myeloid derived suppressor cellsCitation10and the up-regulation of the CTLA-4 and PDL-1/PD-1 negative co-stimulatory pathwaysCitation11,12 is permissive to growth and survival of malignant cells. Furthermore, malignant cells can inhibit the function of antigen presenting cells such as dendritic cells, by polarizing them to a tolerogenic phenotype.Citation13

The development of an effective cancer vaccine requires effective presentation of tumor antigen in the context of co-stimulation that is necessary for effective T cell activation, and the concurrent reversal of the immunosuppressive milleu characteristic of patients with malignancy, in order to favor the induction of long-term immunity.

Dendritic Cell Vaccines

Dendritic cells are a heterogeneous population of bone marrow derived immune cells with potent antigen presenting abilities.Citation14-16 Crucially, dendritic cells strongly express the co-stimulatory molecules required to induce primary immunity.Citation15 Ex vivo studies have demonstrated the ability of dendritic cells to interact with foreign antigens and present these to naïve CD4+ T cells, generating a clonal expansion of effector T cells.Citation16 In addition, mature dendritic cells secrete chemokines attracting B cells, resulting in the generation of a memory B cells specific to that antigen.Citation17 While dendritic cells in cancer patients are quantitativelyCitation18 and functionally deficient,Citation19 functionally potent dendritic cells can be generated from adherent peripheral blood mononuclear cells isolated from patients with malignancy by ex-vivo by culture in the presence of cytokines. As such, dendritic cells manipulated to present tumor antigen have the potential to elicit potent anti-tumor immunity.

CD14+ monocytes represent an abundant and readily accessible source of precursor cells for DC generation. Activation and maturation of DCs leads to the upregulation of co-stimulatory molecules, lymph node homing chemokines such as CCR7, and major histocompatibility complex(MHC) molecules, necessary for activation of T- and B-cells. In vivo the signal for activation and maturation comes from direct contact with pathogens or tissue endothelium, but this can be mimicked in vitro by incubation with prostaglandin E2, pathogen recognition receptor(PRR) agonists or cytokines like TNF-α.Citation20-21 The source of dendritic cells may be either autologous or allogeneic to the patient. The use of allogeneic dendritic cells has the potential to overcome the quantitative and functional deficits of dendritic cells in patients with malignancy, although is limited by dependency on the inconsistent expression of MHC class I molecules on the tumor. Studies using allogeneic dendritic cells in renal cancerCitation22,23 and B-CLLCitation24 have demonstrated the feasibility of this method.

Antigen loading

A variety of strategies for loading tumor antigens onto dendritic cells have been evaluated in clinical studies, including approaches that present individual peptides, protein, or whole tumor cell antigen in the context of the co-stimulatory machinery of the DC. Previous efforts have investigated the used of (i) peptide based vaccines,Citation25 often with an immune adjuvant,Citation26(ii) DNACitation27-29 or RNA codingCitation30,31 for a specific antigen,(iii) viral/fungal vectors expressing cancer antigensCitation32-34 or tumor apoptotic bodies.Citation35 Ex vivo data has shown varying immunogenic responses to these techniques and some of the limitations proposed have been the need for HLA matching of peptide based approaches, potentially low immunogenicity of the chosen antigen and also the possibility that tumors could develop resistance to the vaccine by down regulating the antigen in question.Citation36,37 A strategy to overcome these limitations has been the use of the whole tumor cell as a source of antigen, although there remains a risk of the induction of autoimmunity by the presentation of large volumes of self-antigen.Citation37-38

Methods for priming of DCs with whole tumor have included the whole intact tumor cell,Citation39 cell lysate,Citation40,41 apoptotic bodies,Citation42,43 microvesicles such as exosomesCitation44 and blebs, or whole cell DNA or RNA.Citation40,45 Another interesting approach has been to target antigens toward dendritic cells in vivo, by linking antigens to nanoparticles known to be captured by dendritic cells.Citation46 Our group has focused on a whole cell vaccine approach whereby patient derived tumor cells are fused to autologous, ex-vivo generated dendritic cells by co-culture in the presence of polyethylene glycol.

Considerations in administration

The route of administration influences the migration of dendritic cell based therapies to lymph nodes where they can interact with T cells. Most clinical trials have focused on subcutaneous or intradermal administration of therapies. While some pre-clinical studies have experimented with injecting DCs into the lymphatic system, ensuring that most of the DCs reach the lymph nodes, no difference in the frequency of tetramer-specific T cells could be detected between intradermal and intra lymphatic administrations.Citation47

In our group's experience of 17 patients vaccinated intradermally with a DC/myeloma fusion vaccine, 13 patients exhibited injection-site reactions after vaccination. Biopsy of these areas demonstrated a dense CD8+ mononuclear cell infiltrate suggesting recruitment and education by the DC/MM fusions at the site of vaccination. Furthermore, the presence of CD1a cells was observed in the vaccine bed, suggesting that native Langerhans cells, perhaps recruited by the presence of GM-CSF might participate in the vaccine response.Citation48

Clinical Trials Evaluating Dendritic Cell Vaccines in Hematologic Malignancies

Dendritic cell vaccines using tumor derived protein

Building on successful feasibility studiesCitation49 Timmerman et al reported the results of their phase II trial pulsing the idiotype portion of the B cell immunoglobulin with dendritic cells in patients with follicular lymphoma,Citation50 after induction of first remission, with standard of care chemotherapy. Of 35 patients treated, 65% mounted T-cell proliferative anti-idiotype responses and while complete remission was observed only in 20% of patients, long-term follow-up of patients vaccinated during first remission showed that 16 of 23(70%) remained progression free at a median of 43 months after the completion of chemotherapy. Subsequent to this, the first randomized controlled trial of dendritic cell:idiotype vaccination was reported by Schuster et al in 2009. In this study, 177 patients with untreated follicular lymphoma were randomized to receive vaccination or placebo. Median time to relapse after randomization for the experimental arm was 44.2 months versus 30.6 months for the control arm(p = 0.045; HR = 0.62).Citation51 In 2009, Lacy et al. reported a phase II trial of dendritic cell vaccination in multiple myeloma, whereby 27 patients with at least stable disease post ASCT were given 4 doses of a vaccine made by incubating dendritic cells with a serum containing autologous monoclonal protein. While there was not a difference in progression free survival between vaccine treated patients and historical controls, there was a statistically significant improvement in overall survival with vaccination,(median OS was-5.3 years in vaccine treated patients, 3.4 years in historical contols, P value of 0.02).Citation52 In 2011, Rӧllig et al. reported results of a phase II trial evaluating an idiotype pulsed dendritic cell vaccine, in conjunction with keyhole limpet hemocyanin(KLH) in patients with early stage myeloma. 9 patients with Stage-I myeloma were treated with 5 doses of vaccine administered at 4 weekly intervals. Responses were variable with Id-specific T cell proliferation was demonstrated in 5 out of 9 patients(56%) and a reduction in M protein was observable in only 3/9 patients treated.Citation53

Based on their observation that the cancer-associated protein NY-ESO-1 is highly expressed in poor-prognosis myeloma and is highly immunogenic,Citation54,55 Van Rhee et al are conducting a phase II/III clinical trial vaccinating myeloma patients, post autologous transplantation, with peptide vaccine comprising MAGE-A3 or NY-ESO-1 peptide and GM-CSF adjuvant.

Westermann et al. investigated the use of infusions of non-primed, ex-vivo generated dendritic cells in the treatment of chronic myeloid leukemia(CML).Citation56 In their phase I/II study, 10 patients with chronic phase bcr/abl+ CML, not in adequate cytogenetic response after conventional therapy, were given of 4 subcutaneous injections of increasing numbers of autologous dendritic cells on days 1, 2, 8 and 21. Their vaccination was well tolerated and 4 of 10 patients improved their cytogenetic/molecular responses, while all patients demonstrated improved T cell proliferative capacity following ex vivo stimulation, after vaccination. Cathcart et al. conducted a phase II trial whereby 14 patients with chronic phase bcr/abl+ CML were vaccinated 5 times, on days 0, 7, 21, 35, and 54 with a bcr/abl derived fusion peptide mixed with Quillaja saponaria, an immune adjuvant. 11 of 14 patients had increased CD4 derived IFN-gamma release after vaccination, with just 3 patients showing transient improvements in their cytogenetic response, as shown by PCR.Citation57 Currently, no studies have been published on vaccination with dendritic cell: peptide fusion products in leukemia, but these 2 studies demonstrate the clear potential for a dendritic cell based vaccine in CML, using the bcr/abl fusion peptide as the tumor associated antigen.

An alternative approach to introducing primed or naïve dendritic cells to patients is vaccination with a protein or peptide capable of recruiting and stimulating native dendritic cells. Several groups have conducted feasibility and phase I/II trials of peptide vaccination with peptides derived from the leukemia associated antigen WT1, in combination with immune adjuvants.Citation58,59 Oka et al vaccinated 14 patients with AML with WT1 peptide emulsified with Montanide ISA51 adjuvant at 2 weekly intervals. 9 of the 13 evaluable patients with leukemia demonstrated immunological response as defined by a 1.5x increase in WT1-specific CTLs determined by tetramer assay. Moreover, this correlated strongly with clinical responses, defined as either a reduction in blast count or WT1 expression.

Vaccines using DNA/RNA encoding tumor associated antigen

In 2013, Hobo et al. reported their phase I trial whereby 12 patients with stage I or II myeloma, in at least partial remission after autologous SCT, were treated with dendritic cell vaccines primed with KLH and electroporated with MAGE3, Survivin or B-cell maturation antigen(BCMA) mRNA. Vaccination was well tolerated with limited toxicity and they found antigen specific T cells in 3 of their 12 patients after vaccination, although M protein levels remained unchanged.Citation60

Van Tendeloo et al vaccinated 10 patients with AML with WT1 mRNA-electroporated dendritic cells, with an expansion in WT1 specific T cells demonstrated and correlating with 2 patients in partial remission, who were converted into sustained complete remission.Citation61 Van Driessche et al. have reported their phase I/II studies of a dendritic cell vaccine made by introducing mRNA encoding the Wilm's tumor(WT1) antigen to mature dendritic cells. 10 patients with AML in at least partial remission, were treated with 4 bi-weekly vaccinations. Their vaccination was well tolerated and 2 patients who were in partial remission after chemotherapy were converted into complete remission after vaccination, and this was associated with increases in WT1-specific CD8+ T cell frequencies, as demonstrated by tetramer staining.Citation62

Vaccines using apoptotic bodies derived from tumor cells

A recent area of interest has been the priming of dendritic cells with tumor associated antigen, in the form of apoptotic bodies. It has been postulated that apoptotic bodies allow uptake and processing of a larger quantity of antigen, compared to dendritic cell pinocytic vesicles taking up tumor lysate or in vitro transcription of transfected RNA. In one phase I study in CLL, the immune stimulatory molecule CD80 was expressed on a slightly higher percentage of Apo-DC in comparison to DC loaded with tumor lysate.Citation43 In a phase I study in CLL, Palma et al. vaccinated 15 patients with asymptomatic CLL with a vaccine product made by priming dendritic cells with apoptotic bodies of autologous CLL cells. The vaccine was given alone(Cohort I), or in combination with subcutaneous GM-CSF(Cohort II) or with IV low dose cyclophosphamide(Cohort III). There were no clinical responses, however 10/15 patients were designated ‘immune responders’, as defined by higher levels of IL-2, IL5, IFN and a positive anti-leukemic ex-vivo T cell proliferation assay. The strongest responses were seen in the cohort given cyclophosphamide, although the study was not powered to demonstrate superiority.Citation35

A similar approach to priming dendritic cells was investigated in the setting of acute myeloid leukemia. Kitawaki et al. treated four AML patients(with <20% blasts on bone marrow biopsy after standard of care chemotherapy), with 5 doses of a vaccine generated by pulsing dendritic cells with autologous apoptotic blasts, alongside KLH. Two of the 4 treated patients showed immune responses as determined by raised IFN levels, and the one HLA-A*2402-positive patient was shown to have had induction of CD8+T-cell responses to WT1- and human telomerase reverse transcriptase, indicating success of the dendritic cell pulsing. The two ‘responder’ patients appeared to have longer disease free remissions.Citation63

Whole tumor cell vaccines

Priming DCs with the whole tumor cell can be achieved with a lysed or intact tumor cell. In their phase I clinical trial, Hus et al vaccinated patients with early-stage B-cell chronic lymphocytic leukemia, with HLA matched allogeneic DCs primed with tumor lysate.Citation24 In their subsequent study, they switched to autologous DCs. In total, 12 patients were vaccinated up to 8 times over at bi-weekly intervals, and this was well tolerate with minor skin reactions seen at the vaccine site. An increase in CD8+ T cells specific to the leukemia-associated antigens RHAMM or fibromodulin was detected in 4 patients. A clinical response, as defined by a decrease in leukocytosis and CD19+/CD5+ leukemic cells was seen in 5 patients, and 3 patients showed a stable disease.Citation64

In 2009, DiNicola et al reported their trial in which 18 relapsed patients with an indolent non Hodgkin's lymphoma were treated with dendritic cells primed with whole, heat shocked and irradiated autologous tumor cells. Three patients showed continuous complete responses(CRs) and 3 partial responses(PRs) at 50.5 months and this was significantly associated with a reduction of circulating regulatory T cells(TRegs) and increase in natural killer(NK) cells. Additionally, in some patients, vaccination significantly boosted the IFN-gamma-producing T-cell response to autologous tumor challenge.Citation65

Our group has developed a whole cell vaccine whereby patient derived tumor cells are fused to ex-vivo generated autologous dendritic cells. In a phase I dose escalation study in myeloma,Citation39 17 patients with multiple myeloma(median of 4 prior regimens) underwent vaccination with DC/MM fusions in conjunction with 4 days of GM-CSF administered at the vaccine site. Vaccine production was successful in all patients and was well tolerated without evidence of clinically significant autoimmunity. Vaccination resulted in a mean 10 fold expansion of CD4 and CD8 myeloma specific T cells as determined by the percentage of cells expressing IFNg in response to ex vivo exposure to autologous tumor lysate. Similarly, vaccination resulted in the development of myeloma specific antibody responses. Prolonged disease stabilization was seen in a subset of patients. In a subsequent phase II trial, vaccination with DC/MM fusion cells was administered in conjunction with autologous stem cell transplantation(ASCT). The period of post-transplant lymphopoietic reconstitution was associated with the expansion of myeloma specific T cells that was further boosted following vaccination. Seventy-eight percent of patients achieved a CR or VGPR(47% CR/nCR; 31% VGPR). Thirty-one percent achieved a CR/nCR in the early post-transplant period, whereas an additional 17%(6 patients: 4 from VGPR, 2 from PR) achieved CR/nCR as best response only after day 100 post-transplant and after undergoing vaccination. The presence of late responses several months after ASCT is consistent with an impact of vaccine therapy on post-transplant residual disease.Citation48

In acute leukemia, we are conducting a clinical trial in which AML patients who are in a first or second complete remission following chemotherapy are given 3 monthly doses of DC/AML fusion cells. Interim results from 13 evaluable patients(who have received at least 2 vaccinations) reveal 9 patients(69%) in sustained remission at 28 months.Citation66

A novel approach to augment antigen presenting activity in patients with cancer has been to vaccinate them with irradiated autologous tumor cells engineered to secrete GM-CSF. It is thought that paracrine production of GM-CSF can stimulate the recruitment, maturation, and function of dendritic cells in vivo,Citation67,68 overcoming the described qualitative and quantitative deficiencies of antigen presenting cells in cancer patients. Ho et al conducted a Phase I clinical trial whereby high-risk acute myeloid leukemia or patients with MDS were immunized with autologous, irradiated, GM-CSF-secreting tumor cells early after allogeneic, nonmyeloablative HSCT.Citation67 The immunization was broadly well tolerated with one patient suffering a GVHD type skin reaction. Six long-term responders showed marked decreases in the levels of soluble NKG2D ligands, indicative of NK cell tumor specific activity, and 3 demonstrated normalization of cytotoxic lymphocyte NKG2D expression as a function of treatment.

Leukemia derived dendritic cells

One approach in overcoming quantitative and qualitative defects in dendritic cells is to generate mature dendritic cells from immature myeloid cells. In CMLCitation69,70 and AML,Citation71-73 dendritic cells can be generated from ex-vivo leukemia blasts themselves, which has the added advantage of negating the need to load tumor-associated antigen, while expressing these antigens in the context of the necessary co-stimulatory signals. Leukemia blast derived cell linesCitation74 that have been stimulated to differentiate into dendritic like cells with antigen presenting capabilities. The advantage of this technique is the cells potential to induce an immune response against the whole repertoire of leukemic antigens without the need for exogenous loading. However there are concerns over the re-infusion of leukemic dendritic cells that have been purported to be a component of tumor related immune escape.Citation75

Litzow et al subcutaneously vaccinated 6 CML patients with ex vivo leukemic derived dendritic cells, 4 of whom were also on Imatinib therapy. While they observed no clinical responses, there were increased circulating CML specific T cells following therapy.Citation76 Roddie et al vaccinated 5 AML patients in complete remission with 4 escalating doses of dendritic cell like leukemia cells. One patient developed an eczematous rash and auto-antibodies suggestive of the induction of auto-immunity. Immunological response, as demonstrated by increased INF-g production by leukemia specific T cells was demonstrated in 4 patients, although only 2 of the 5 remained in remission more than one year post vaccination.Citation77

A practical draw-back of this approach is that Leukemia derived DCs can only be generated from a minority of patients, as FLT-ITD mutations or a lack of CD14 expressed on blasts prevent the maturation to the DC phenotype.Citation78,79

Enhancing response to vaccination

A critical factor to consider in designing clinical trials evaluating dendritic cell vaccines is the immunologic milieu into which the vaccine is being administered. Tumor induced immune suppression may blunt immune response to vaccination in patients with advanced disease. Factors contributing to tumor induced immune suppression include an increased presence of regulatory T cells,Citation80 increased circulating myeloid derived suppressor cells(10) and skew of polarity of dendritic cells toward a tolerogenic phenotype.Citation81 Additionally, a variety of soluble factors such as indolamine, VEGF, TGFB and IL-10 inhibit both effector T cell function and dendritic cell maturation.Citation82 Moving forward, ongoing and future studies that incorporate vaccination in minimal disease states, following chemotherapy and following transplantation, hold promise as a means of eradicating minimal residual disease and preventing relapse. We have identified the post allograft setting as an optimal time for immunotherapy as during lymphopoietic reconstitution there is a relative depletion of immune suppressive regulatory T cells which would otherwise limit the response to vaccination.Citation83

A second approach toward enhancing response to vaccination is combining vaccination with stimulatory cytokines, immune-modulatory drugs, and immune checkpoint blockade.

While pre-clinical studies have used immune stimulating cytokines such as IL-2 in combination with various immune therapiesCitation84 in an attempt to enhance immune responses, clinical trials using dendritic cells augmented to secrete have been limited to the use of GM-CSF. In the setting of AML, Borello et.al conducted a phase II trial where 28 patients were given induction chemotherapy, followed by a single immunotherapy treatment of allogeneic immortalized K562 tumor cells that had been modified to secrete high levels of GM-CSF.Citation85 Patients primed lymphocytes were collected by plasmapheresis and then underwent autologous stem cell transplantation, receiving the primed lymphocytes at day 0, followed by 8 further immunotherapy treatments over a 6 month period. Treatment with the immunotherapy lead to a decrease in WT1 transcripts in 69% of patients after their first immune therapy, and increase in CD4+ derived IFN-gamma and granzyme. The immune therapy lead to an overall survival of 73.4% vs. 57.4% for patients who were ineligible for the immunotherapy. Of note, only 6 patients had detectable levels of GM-CSF at any one time point and importantly, the study was not designed to demonstrate the superiority of GMCSF modified K562 cells over wildtype.

Lenalidomide is a second-generation thalidomide analog used in the treatment of myeloma. Lenalidomide has potent immunomodulatory functions which have not been fully elucidated but are thought to enhance activation of T and NK cells. In addition, in a preclinical study the immunomoulatory agents Lenaliomide and Pomalidomide have been shown to have an effect on dendritic cells themselves, with increased expression of CD86 on DCs, and increased uptake of fluorescent beads, signifying increased antigen presentation.Citation86 In preclinical studies, Lenalidomide enhanced immune response to vaccination,Citation87 and a clinical trial combining Lenalidomide with vaccination is planned.

The PD1/PDL1 pathway serves as a negative checkpoint for T cell activation and CTL-mediated targeting of tumor cells. In patients with cancer, up-regulated expression of PD-1 on T cell binds PDL-1 on tumor cell, resulting in the suppression of T cell capacity to secrete stimulatory cytokines.Citation11,83 Following autologous transplantation for myeloma, T-cell expression of PD-1 was observed to return to normal levels. In our ex-vivo study, the effect of PD-1 blockade on T-cell response to DC/whole tumor fusions was investigated. The presence the anti-PD1 antibody CT-011, promoted the vaccine-induced T-cell polarization toward an activated Th1 phenotype from a Th2 cytokine profile. Interestingly, a concomitant decrease in regulatory T cells and enhanced killing in a cytotoxicity assay was observed. A clinical trial combining PDL1 blockade with DC/tumor fusion cell vaccination for patients with AML, is underway.Citation83

Conclusion

The potency of the immune system in controlling hematologic malignancies is highlighted in the allogeneic transplant setting. However, the lack of specificity of allo-reactive T cells results in graft vs. host disease. The design of dendritic cell vaccines as a means of eliciting tumor specific immune responses has the potential to result in durable responses with minimal toxicity. A major focus of research interest lies on developing strategies to broadly target the malignant clone, mitigating the potential for immune escape, while minimizing the risk of the induction of autoimmunity. Importantly, immunotherapeutic strategies that target tumor initiating cell populations have the potential to eradicate the source of disease recurrence and chemotherapy resistance.

While most studies have focused on patients with refractory disease, immunotherapeutic approaches will likely be most effective in patients with low tumor burden. Studies evaluating the combination of dendritic cell vaccines with chemotherapy, immunomodulatory drugs, and immune checkpoint inhibitors, are ongoing.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

References

  • Bishop JF, Matthews JP, Young GA, Szer J, Gillett A, Joshua D, Bradstock K, Enno A, Wolf MM, Fox R, et al. A randomized study of high-dose cytarabine in induction in acute myeloid leukemia. Blood 1996; 87(5):1710-7; PMID:8634416
  • Wiseman DH, Greystoke BF, Somervaille TC. The variety of leukemic stem cells in myeloid malignancy. Oncogene 2013; PMID:23831573
  • Parish CR. Cancer immunotherapy: the past, the present and the future. Immunol Cell Biol 2003; 81(2):106-13; PMID:12631233
  • Butturini A, Bortin MM, Gale RP. Graft-versus-leukemia following bone marrow transplantation. Bone Marrow Transplant 1987; 2(3):233-42; PMID:3332173
  • Mavroudis D, Barrett J. The graft-versus-leukemia effect. Curr Opin Hematol. 1996 Nov; 3(6):423-9; PMID:9372113
  • Chang YJ, Huang XJ. Donor lymphocyte infusions for relapse after allogeneic transplantation: when, if and for whom? Blood Rev 2013 Jan; 27(1):55-62; PMID:23261066
  • Chen L, Ashe S, Brady WA, Hellström I, Hellström KE, Ledbetter JA, McGowan P, Linsley PS. Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell 1992; 71(7):1093-102; PMID:1335364
  • Speiser DE, Miranda R, Zakarian A, Bachmann MF, McKall-Faienza K, Odermatt B, Hanahan D, Zinkernagel RM, Ohashi PS. Self antigens expressed by solid tumors do not efficiently stimulate naive or activated T cells: implications for immunotherapy. J Exp Med 1997; 186(5):645-53; PMID:9271580
  • Liyanage UK, Moore TT, Joo HG, Tanaka Y, Herrmann V, Doherty G, Drebin JA, Strasberg SM, Eberlein TJ, Goedegebuure PS, et al. Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J Immunol 2002 169(5):2756-61; PMID:12193750
  • Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012; 12(4):253-68; PMID:22437938
  • Rosenblatt J, Glotzbecker B, Mills H, Vasir B, Tzachanis D, Levine JD, Joyce RM, Wellenstein K, Keefe W, Schickler M, et al. PD-1 blockade by CT-011, anti-PD-1 antibody, enhances ex vivo T-cell responses to autologous dendritic cell/myeloma fusion vaccine. J Immunother 2011; 34(5):409-18.PMID:21577144
  • Weber J. Immune checkpoint proteins: a new therapeutic paradigm for cancer–preclinical background: CTLA-4 and PD-1 blockade. Semin Oncol 2010; 37(5):430-9.PMID:21074057
  • Ma Y, Shurin GV, Peiyuan Z, Shurin MR. Dendritic cells in the cancer microenvironment. J Cancer 2013; 4(1):36-44; PMID:23386903
  • Nencioni A, Grünebach F, Schmidt SM, Müller MR, Boy D, Patrone F, Ballestrero A, Brossart P. The use of dendritic cells in cancer immunotherapy. Crit Rev Oncol Hematol 2008; 65(3):191-9; PMID:18055210
  • Avigan D. Dendritic cells: development, function and potential use for cancer immunotherapy. Blood Rev 1999; 13(1):51-64; PMID:10225609
  • Steinman RM. The dendritic cell system and its role in immunogenicity. Annu Rev Immunol 1991; 9 :271-96; PMID:1910679
  • Dubois B, Massacrier C, Caux C. Selective attraction of naive and memory B cells by dendritic cells. J Leukoc Biol 2001; 70(4):633-41; PMID:11590201
  • Lissoni P, Vigore L, Ferranti R, Bukovec R, Meregalli S, Mandala M, Barni S, Tancini G, Fumagalli L, Giani L. et al. Circulating dendritic cells in early and advanced cancer patients: diminished percent in the metastatic disease. J Biol Regul Homeost Agents 1999 1999; 13(4):216-9; PMID:10703945
  • Gabrilovich D. Mechanisms and functional significance of tumour-induced dendritic-cell defects. Nat Rev Immunol 2004; 4(12):941-52; PMID:15573129; http://dx.doi.org/10.1038/nri1498
  • Jonuleit H, Kühn U, Müller G, Steinbrink K, Paragnik L, Schmitt E, Knop J, Enk AH. Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum-free conditions. Eur J Immunol 1997; 27(12):3135-42; PMID:9464798; http://dx.doi.org/10.1002/eji.1830271209
  • Wimmers F, Schreibelt G, Sköld AE, Figdor CG, De Vries IJ. Paradigm Shift in dendritic cell-based immunotherapy: from in vitro generated monocyte-derived DCs to naturally circulating DC subsets. Front Immunol 2014; 5 :165; PMID:24782868; http://dx.doi.org/10.3389/fimmu.2014.00165
  • Märten A, Renoth S, Heinicke T, Albers P, Pauli A, Mey U, Caspari R, Flieger D, Hanfland P, Von Ruecker A, et al. Allogeneic dendritic cells fused with tumor cells: preclinical results and outcome of a clinical phase I/II trial in patients with metastatic renal cell carcinoma. Hum Gene Ther 2003; 14(5):483-94; PMID:12691613
  • Avigan DE GD, Kantoff PW, editor Interim safety and efficacy results from a phase I/II study of vaccination with electrofused allogeneic dendritic cells/autologous tumor-derived cells in patients with stage IV renal cell carcinoma2004. J Clin Oncol ASCO Annual Meeting Proceedings.
  • Hus I, Roliński J, Tabarkiewicz J, Wojas K, Bojarska-Junak A, Greiner J, Giannopoulos K, Dmoszyńska A, Schmitt M. Allogeneic dendritic cells pulsed with tumor lysates or apoptotic bodies as immunotherapy for patients with early-stage B-cell chronic lymphocytic leukemia. Leukemia 2005; 19(9):1621-7; PMID:15990861; http://dx.doi.org/10.1038/sj.leu.2403860
  • Philip R, Alters SE, Brunette E, Ashton J, Gadea J, Yau J, Lebkowski J, Philip M. Dendritic cells loaded with MART-1 peptide or infected with adenoviral construct are functionally equivalent in the induction of tumor-specific cytotoxic T lymphocyte responses in patients with melanoma. J Immunother 2000; 23(1):168-76; PMID:10687150; http://dx.doi.org/10.1097/00002371-200001000-00020
  • Minev BR, Chavez FL, Dudouet BM, Mitchell MS. Synthetic insertion signal sequences enhance MHC class I presentation of a peptide from the melanoma antigen MART-1. Eur J Immunol 2000; 30(8):2115-24; PMID:10940901; http://dx.doi.org/10.1002/1521-4141(2000)30:8%3c2115::AID-IMMU2115%3e3.0.CO;2-J
  • Cao J, Jin Y, Li W, Zhang B, He Y, Liu H, Xia N, Wei H, Yan J. DNA vaccines targeting the encoded antigens to dendritic cells induce potent antitumor immunity in mice. BMC Immunol 2013; 14:39.PMID:23941509
  • Li D, Hua S, Fan Y, Xu S, Duan X, Liu L, Che Y, Li S, Tan Y. DNA vaccine expressing repeated carcinoembryonic antigen(CEA)(625-667) induces strong immunity in mice. Immunol Lett 2011; 135(1-2):124-8; PMID:21073899
  • Signori E, Iurescia S, Massi E, Fioretti D, Chiarella P, De Robertis M, Rinaldi M, Tonon G, Fazio VM. DNA vaccination strategies for anti-tumour effective gene therapy protocols. Cancer Immunol Immunother 2010; 59(10):1583-91; PMID:20390416; http://dx.doi.org/10.1007/s00262-010-0853-x
  • Boczkowski D, Nair SK, Snyder D, Gilboa E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J Exp Med 1996; 184(2):465-72.PMID:8760800
  • Nair SK, Morse M, Boczkowski D, Cumming RI, Vasovic L, Gilboa E, Lyerly HK. Induction of tumor-specific cytotoxic T lymphocytes in cancer patients by autologous tumor RNA-transfected dendritic cells. Ann Surg 2002; 235(4):540-9; PMID:11923611; http://dx.doi.org/10.1097/00000658-200204000-00013
  • Hamilton DH, Litzinger MT, Jales A, Huang B, Fernando RI, Hodge JW, Ardiani A, Apelian D, Schlom J, Palena C. et al. Immunological targeting of tumor cells undergoing an epithelial-mesenchymal transition via a recombinant brachyury-yeast vaccine. Oncotarget 2013; PMID:24125763
  • Litzinger MT, Foon KA, Sabzevari H, Tsang KY, Schlom J, Palena C. Chronic lymphocytic leukemia(CLL) cells genetically modified to express B7-1, ICAM-1, and LFA-3 confer APC capacity to T cells from CLL patients. Cancer Immunol Immunother 2009; 58(6):955-65; PMID:19009294; http://dx.doi.org/10.1007/s00262-008-0611-5
  • Litzinger MT, Foon KA, Tsang KY, Schlom J, Palena C. Comparative analysis of MVA-CD40L and MVA-TRICOM vectors for enhancing the immunogenicity of chronic lymphocytic leukemia(CLL) cells. Leuk Res 2010; 34(10):1351-7; PMID:20122733; http://dx.doi.org/10.1016/j.leukres.2009.12.013
  • Palma M, Hansson L, Choudhury A, Näsman-Glaser B, Eriksson I, Adamson L, Rossmann E, Widén K, Horváth R, Kokhaei P, et al. Vaccination with dendritic cells loaded with tumor apoptotic bodies(Apo-DC) in patients with chronic lymphocytic leukemia: effects of various adjuvants and definition of immune response criteria. Cancer Immunol Immunother 2012; 61(6):865-79; PMID:22086161; http://dx.doi.org/10.1007/s00262-011-1149-5
  • Avigan D, Rosenblatt J, Kufe D. Dendritic/tumor fusion cells as cancer vaccines. Semin Oncol 2012; 39(3):287-95; PMID:22595051; http://dx.doi.org/10.1053/j.seminoncol.2012.02.003
  • Koido S, Homma S, Okamoto M, Namiki Y, Takakura K, Uchiyama K, Kajihara M, Arihiro S, Imazu H, Arakawa H, et al. Fusions between dendritic cells and whole tumor cells as anticancer vaccines. Oncoimmunology 2013; 2(5):e24437; PMID:23762810; http://dx.doi.org/10.4161/onci.24437
  • Amos SM, Duong CP, Westwood JA, Ritchie DS, Junghans RP, Darcy PK, Kershaw MH. Autoimmunity associated with immunotherapy of cancer. Blood 2011; 118(3):499-509; PMID:21531979
  • Rosenblatt J, Vasir B, Uhl L, Blotta S, Macnamara C, Somaiya P, Wu Z, Joyce R, Levine JD, Dombagoda D, et al. Vaccination with dendritic cell/tumor fusion cells results in cellular and humoral antitumor immune responses in patients with multiple myeloma. Blood 2011; 117(2):393-402; PMID:21030562
  • Pan K, Zhao JJ, Wang H, Li JJ, Liang XT, Sun JC, Chen YB, Ma HQ, Liu Q, Xia JC. et al. Comparative analysis of cytotoxic T lymphocyte response induced by dendritic cells loaded with hepatocellular carcinoma -derived RNA or cell lysate. Int J Biol Sci 2010; 6(7):639-48.PMID:20975822
  • Hong S, Li H, Qian J, Yang J, Lu Y, Yi Q. Optimizing dendritic cell vaccine for immunotherapy in multiple myeloma: tumour lysates are more potent tumour antigens than idiotype protein to promote anti-tumour immunity. Clin Exp Immunol 2012; 170(2):167-77; PMID:23039887
  • Spisek R, Chevallier P, Morineau N, Milpied N, Avet-Loiseau H, Harousseau JL, Meflah K, Gregoire M. Induction of leukemia-specific cytotoxic response by cross-presentation of late-apoptotic leukemic blasts by autologous dendritic cells of nonleukemic origin. Cancer Res 2002; 62(10):2861-8; PMID:12019165
  • Kokhaei P, Choudhury A, Mahdian R, Lundin J, Moshfegh A, Osterborg A, Mellstedt H. Apoptotic tumor cells are superior to tumor cell lysate, and tumor cell RNA in induction of autologous T cell response in B-CLL. Leukemia 2004; 18(11):1810-5; PMID:15385926
  • van den Ancker W, Ruben JM, Westers TM, Wulandari D, Bontkes HJ, Hooijberg E, Stam AG, Santegoets SJ, Ossenkoppele GJ, de Gruijl T, et al. Priming of PRAME- and WT1-specific CD8(+) T cells in healthy donors but not in AML patients in complete remission: implications for immunotherapy. Oncoimmunology 2013; 2(4):e23971; PMID:23734332
  • Milazzo C, Reichardt VL, Müller MR, Grünebach F, Brossart P. Induction of myeloma-specific cytotoxic T cells using dendritic cells transfected with tumor-derived RNA. Blood 2003; 101(3):977-82; PMID:12393470
  • Cassaday RD, Sondel PM, King DM, Macklin MD, Gan J, Warner TF, Zuleger CL, Bridges AJ, Schalch HG, Kim KM, et al. A phase I study of immunization using particle-mediated epidermal delivery of genes for gp100 and GM-CSF into uninvolved skin of melanoma patients. Clin Cancer Res 2007; 13(2 Pt 1):540-9; PMID:17255276
  • Lesterhuis WJ, de Vries IJ, Schreibelt G, Lambeck AJ, Aarntzen EH, Jacobs JF, Scharenborg NM, van de Rakt MW, de Boer AJ, Croockewit S, et al. Route of administration modulates the induction of dendritic cell vaccine-induced antigen-specific T cells in advanced melanoma patients. Clin Cancer Res 2011; 17(17):5725-35; PMID:21771874
  • Rosenblatt J, Avivi I, Vasir B, Uhl L, Munshi NC, Katz T, Dey BR, Somaiya P, Mills H, Campigotto F, et al. Vaccination with dendritic cell/tumor fusions following autologous stem cell transplant induces immunologic and clinical responses in multiple myeloma patients. Clin Cancer Res 2013; 19(13):3640-8; PMID:23685836
  • Reichardt VL, Okada CY, Liso A, Benike CJ, Stockerl-Goldstein KE, Engleman EG, Blume KG, Levy R. Idiotype vaccination using dendritic cells after autologous peripheral blood stem cell transplantation for multiple myeloma–a feasibility study. Blood 1999; 93(7):2411-9; PMID:10090953
  • Timmerman JM, Czerwinski DK, Davis TA, Hsu FJ, Benike C, Hao ZM, Taidi B, Rajapaksa R, Caspar CB, Okada CY, et al. Idiotype-pulsed dendritic cell vaccination for B-cell lymphoma: clinical and immune responses in 35 patients. Blood 2002; 99(5):1517-26; PMID:11861263
  • Schuster S, editor Idiotype vaccine therapy(biovaxID) in follicular lymphoma in first complete remission: phase III clinical trial results. University of Pennsylvania 2009.
  • Lacy MQ, Mandrekar S, Dispenzieri A, Hayman S, Kumar S, Buadi F, Dingli D, Litzow M, Wettstein P, Padley D, et al. Idiotype-pulsed antigen-presenting cells following autologous transplantation for multiple myeloma may be associated with prolonged survival. Am J Hematol 2009; 84(12):799-802; PMID:19899131
  • Röllig C, Schmidt C, Bornhäuser M, Ehninger G, Schmitz M, Auffermann-Gretzinger S. Induction of cellular immune responses in patients with stage-I multiple myeloma after vaccination with autologous idiotype-pulsed dendritic cells. J Immunother 2011; 34(1):100-6; PMID:21150718
  • van Rhee F, Szmania SM, Zhan F, Gupta SK, Pomtree M, Lin P, Batchu RB, Moreno A, Spagnoli G, Shaughnessy J, et al. NY-ESO-1 is highly expressed in poor-prognosis multiple myeloma and induces spontaneous humoral and cellular immune responses. Blood 2005; 105(10):3939-44; PMID:15671442
  • Batchu RB, Moreno AM, Szmania SM, Bennett G, Spagnoli GC, Ponnazhagan S, Barlogie B, Tricot G, van Rhee F. Protein transduction of dendritic cells for NY-ESO-1-based immunotherapy of myeloma. Cancer Res 2005; 65(21):10041-9; PMID:16267030
  • Westermann J, Kopp J, van Lessen A, Hecker AC, Baskaynak G, le Coutre P, Döhner K, Döhner H, Dörken B, Pezzutto A. et al. Vaccination with autologous non-irradiated dendritic cells in patients with bcr/abl+ chronic myeloid leukaemia. Br J Haematol 2007; 137(4):297-306; PMID:17408402
  • Cathcart K, Pinilla-Ibarz J, Korontsvit T, Schwartz J, Zakhaleva V, Papadopoulos EB, Scheinberg DA. A multivalent bcr-abl fusion peptide vaccination trial in patients with chronic myeloid leukemia. Blood 2004; 103(3):1037-42; PMID:14504104
  • Uttenthal B, Martinez-Davila I, Ivey A, Craddock C, Chen F, Virchis A, Kottaridis P, Grimwade D, Khwaja A, Stauss H, et al. Wilms' Tumour 1(WT1) peptide vaccination in patients with acute myeloid leukaemia induces short-lived WT1-specific immune responses. Br J Haematol 2014; 164(3):366-75; PMID:24422723
  • Oka Y, Tsuboi A, Taguchi T, Osaki T, Kyo T, Nakajima H, Elisseeva OA, Oji Y, Kawakami M, Ikegame K, et al. Induction of WT1(wilms' tumor gene)-specific cytotoxic T lymphocytes by WT1 peptide vaccine and the resultant cancer regression. Proc Natl Acad Sci U S A 2004; 101(38):13885-90; PMID:15365188
  • Hobo W, Strobbe L, Maas F, Fredrix H, Greupink-Draaisma A, Esendam B, de Witte T, Preijers F, Levenga H, van Rees B, et al. Immunogenicity of dendritic cells pulsed with MAGE3, survivin and B-cell maturation antigen mRNA for vaccination of multiple myeloma patients. Cancer Immunol Immunother 2013; 62(8):1381-92.PMID:23728352
  • Van Tendeloo VF, Van de Velde A, Van Driessche A, Cools N, Anguille S, Ladell K, Gostick E, Vermeulen K, Pieters K, Nijs G, et al. Induction of complete and molecular remissions in acute myeloid leukemia by wilms' tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci U S A 2010; 107(31):13824-9; PMID:20631300
  • Van Driessche A, Van de Velde AL, Nijs G, Braeckman T, Stein B, De Vries JM, Berneman ZN, Van Tendeloo VF. Clinical-grade manufacturing of autologous mature mRNA-electroporated dendritic cells and safety testing in acute myeloid leukemia patients in a phase I dose-escalation clinical trial. Cytotherapy 2009; 11(5):653-68; PMID:19530029
  • Kitawaki T, Kadowaki N, Fukunaga K, Kasai Y, Maekawa T, Ohmori K, Itoh T, Shimizu A, Kuzushima K, Kondo T, et al. Cross-priming of CD8(+) T cells in vivo by dendritic cells pulsed with autologous apoptotic leukemic cells in immunotherapy for elderly patients with acute myeloid leukemia. Exp Hematol 2011 Apr; 39(4):424-33.e2; PMID:21216276
  • Hus I, Schmitt M, Tabarkiewicz J, Radej S, Wojas K, Bojarska-Junak A, Schmitt A, Giannopoulos K, Dmoszyńska A, Roliński J. et al. Vaccination of B-CLL patients with autologous dendritic cells can change the frequency of leukemia antigen-specific CD8+ T cells as well as CD4+CD25+FoxP3+ regulatory T cells toward an antileukemia response. Leukemia 2008; 22(5):1007-17; PMID:18323802
  • Di Nicola M, Zappasodi R, Carlo-Stella C, Mortarini R, Pupa SM, Magni M, Devizzi L, Matteucci P, Baldassari P, Ravagnani F, et al. Vaccination with autologous tumor-loaded dendritic cells induces clinical and immunologic responses in indolent B-cell lymphoma patients with relapsed and measurable disease: a pilot study. Blood 2009; 113(1):18-27; PMID:18809757
  • Rosenblatt J, Stone R, Uhl L, Neuberg D, Vasir B, Somaiya P, et al. editors. Clinical trial evaluating DC/AML fusion cell vaccination in AML patients who achieve a chemotherapy-induced remission. ASH Annual Meeting; 2013; New Orleans Convention Center.
  • Ho VT, Vanneman M, Kim H, Sasada T, Kang YJ, Pasek M, Cutler C, Koreth J, Alyea E, Sarantopoulos S, et al. Biologic activity of irradiated, autologous, GM-CSF-secreting leukemia cell vaccines early after allogeneic stem cell transplantation. Proc Natl Acad Sci U S A. 2009; 106(37):15825-30; PMID:19717467
  • Burkhardt UE, Hainz U, Stevenson K, Goldstein NR, Pasek M, Naito M, Wu D, Ho VT, Alonso A, Hammond NN, et al. Autologous CLL cell vaccination early after transplant induces leukemia-specific T cells. J Clin Invest 2013; 123(9):3756-65; PMID:23912587
  • Fujii S, Shimizu K, Fujimoto K, Kiyokawa T, Shimomura T, Kinoshita M, Kawano F. Analysis of a chronic myelogenous leukemia patient vaccinated with leukemic dendritic cells following autologous peripheral blood stem cell transplantation. Jpn J Cancer Res 1999; 90(10):1117-29; PMID:10595741
  • Stam AG, Santegoets SJ, Westers TM, Sombroek CC, Janssen JJ, Tillman BW, van de Loosdrecht AA, Pinedo HM, Curiel DT, Ossenkoppele GJ, et al. CD40-targeted adenoviral GM-CSF gene transfer enhances and prolongs the maturation of human CML-derived dendritic cells upon cytokine deprivation. Br J Cancer 2003; 89(7):1162-5; PMID:14520439
  • Li L, Reinhardt P, Schmitt A, Barth TF, Greiner J, Ringhoffer M, Döhner H, Wiesneth M, Schmitt M. Dendritic cells generated from acute myeloid leukemia(AML) blasts maintain the expression of immunogenic leukemia associated antigens. Cancer Immunol Immunother 2005; 54(7):685-93; PMID:15627212
  • Cignetti A, Vallario A, Roato I, Circosta P, Allione B, Casorzo L, Ghia P, Caligaris-Cappio F. Leukemia-derived immature dendritic cells differentiate into functionally competent mature dendritic cells that efficiently stimulate T cell responses. J Immunol 2004; 173(4):2855-65; PMID:15295005
  • Schmitt A, Li L, Giannopoulos K, Greiner J, Reinhardt P, Wiesneth M, Schmitt M. Quantitative expression of Toll-like receptor-2, -4, and -9 in dendritic cells generated from blasts of patients with acute myeloid leukemia. Transfusion 2008; 48(5):861-70; PMID:18208411
  • van den Ancker W, van Luijn MM, Westers TM, Bontkes HJ, Ruben JM, de Gruijl TD, Ossenkoppele GJ, van de Loosdrecht AA. Recent advances in antigen-loaded dendritic cell-based strategies for treatment of minimal residual disease in acute myeloid leukemia. Immunotherapy 2010; 2(1):69-83; PMID:20635890
  • Fujii S, Shimizu K, Koji F, Kawano F. Malignant counterpart of myeloid dendritic cell(DC) belonging to acute myelogenous leukemia(AML) exhibits a dichotomous immunoregulatory potential. J Leukoc Biol 2003; 73(1):82-90; PMID:12525565
  • Litzow MR, Dietz AB, Bulur PA, Butler GW, Gastineau DA, Hoering A, Fink SR, Letendre L, Padley DJ, Paternoster SF, et al. Testing the safety of clinical-grade mature autologous myeloid DC in a phase I clinical immunotherapy trial of CML. Cytotherapy 2006; 8(3):290-8; PMID:16793737
  • Roddie H, Klammer M, Thomas C, Thomson R, Atkinson A, Sproul A, Waterfall M, Samuel K, Yin J, Johnson P, et al. Phase I/II study of vaccination with dendritic-like leukaemia cells for the immunotherapy of acute myeloid leukaemia. Br J Haematol 2006; 133(2):152-7.PMID:16611305
  • Houtenbos I, Westers TM, Hess CJ, Waisfisz Q, Ossenkoppele GJ, van de Loosdrecht AA. Flt-3 internal tandem duplication hampers differentiation of AML blasts towards leukemic dendritic cells. Leukemia 2006; 20(10):1892-5; PMID:16932350
  • Anguille S, Willemen Y, Lion E, Smits EL, Berneman ZN. Dendritic cell vaccination in acute myeloid leukemia. Cytotherapy 2012; 14(6):647-56; PMID:22686130
  • Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ. The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology 2013; 138(2):105-15; PMID:23216602
  • Demoulin S, Herfs M, Delvenne P, Hubert P. Tumor microenvironment converts plasmacytoid dendritic cells into immunosuppressive/tolerogenic cells: insight into the molecular mechanisms. J Leukoc Biol 2013; 93(3):343-52; PMID:23136258
  • Zamarron BF, Chen W. Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci 2011; 7(5):651-8; PMID:21647333
  • Rosenblatt J AI, Vasir B, Uhl L, Katz T, Avigan D et al. editor Blockade of PD-1 in combination with dendritic cell/myeloma fusion cell vaccination following autologous stem cell transplantation. ASH 2012 2012.
  • Hardwick N, Chan L, Ingram W, Mufti G, Farzaneh F. Lytic activity against primary AML cells is stimulated in vitro by an autologous whole cell vaccine expressing IL-2 and CD80. Cancer Immunol Immunother 2010; 59(3):379-88; PMID:19711075
  • Borrello IM, Levitsky HI, Stock W, Sher D, Qin L, DeAngelo DJ, Alyea EP, Stone RM, Damon LE, Linker CA, et al. Granulocyte-macrophage colony-stimulating factor(GM-CSF)-secreting cellular immunotherapy in combination with autologous stem cell transplantation(ASCT) as postremission therapy for acute myeloid leukemia(AML). Blood 2009; 114(9):1736-45; PMID:19556425
  • Henry JY, Labarthe MC, Meyer B, Dasgupta P, Dalgleish AG, Galustian C. Enhanced cross-priming of naive CD8+ T cells by dendritic cells treated by the IMiDs® immunomodulatory compounds lenalidomide and pomalidomide. Immunology 2013; 139(3):377-85; PMID:23374145; http://dx.doi.org/10.1111/imm.12087
  • Luptakova K, Rosenblatt J, Glotzbecker B, Mills H, Stroopinsky D, Kufe T, Vasir B, Arnason J, Tzachanis D, Zwicker JI, et al. Lenalidomide enhances anti-myeloma cellular immunity. Cancer Immunol Immunother 2013; 62(1):39-49; PMID:22733396; http://dx.doi.org/10.1007/s00262-012-1308-3

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.