Open access

Immunotherapy with Dendritic Cells and Newcastle Disease Virus in Glioblastoma Multiforme

Written By

Thomas Neßelhut, Dagmar Marx, Jan Neßelhut and Fred Fändrich

Submitted: 17 November 2010 Published: 23 August 2011

DOI: 10.5772/22074

From the Edited Volume

Brain Tumors - Current and Emerging Therapeutic Strategies

Edited by Ana L. Abujamra

Chapter metrics overview

3,382 Chapter Downloads

View Full Metrics

1. Introduction

Malignant gliomas are the most prevalent type of primary brain tumors in adults. The most aggressive form, glioblastoma multiforme (GBM, WHO grade IV glioma), is characterized as incurable with average survivals less than 18 months (Buckner, 2003; Curran et al., 1993; De Angelis, 2001). Standard therapy consists of surgical resection to the extent that is safely feasible, followed by radiotherapy and adjuvant chemotherapy. A meta-analysis based on 12 randomized trials has supported the use of adjuvant chemotherapy treatment as compared to radiotherapy alone. A five percent increase in survival at two years, from 15 percent to 20 percent was reported (Stewart, 2002). In this context, an oral alkylating agent, temozolomide, has demonstrated antitumor activity as a single agent in the treatment of glioblastoma patients (Newlands et al., 1997; Stupp et al., 2001, 2005; Yung et al., 2000). At a median follow-up of 28 months, temozolomide treatment prolonged the median survival from 12.1 months with radiotherapy alone to 14.6 months when administered with concomitant radiotherapy (Stupp et al., 2005). Adjuvant administration of carmustine, a nitrosourea drug, has not resulted in beneficial prolongation of survival, although some long-term survivors in the chemotherapy groups have been documented in some of the non-randomized phase III trials (Chang et al., 1983; Green et al., 1983). Despite these advances in combined radio- chemotherapy treatment strategies along with surgical resection, the prognosis of patients with malignant glioma is poor and ≥ 80% of the patients survive less than 2 years. Salvage treatments upon recurrence are palliative at best and rarely provide a significant survival benefit. The minor clinical benefit of radio-chemotherapy is supposed to be mainly based on tumour stem cells, which in analogy to normal stem cells are resistant to radio- and chemotherapy (Liu et al., 2006; Singh et al., 2003). Therefore, the development of new treatment modalities is extremely important.

It is known that defects in the immune system play a crucial role in the development of cancer as well as in the host defense against cancer. Thus, specific immune therapeutic therapy approaches have been tested for a long time. Tumor-associated antigens recognized by tumor-reactive cytotoxic T lymphocytes (CTL) have been identified during the last two decades (Janeway et al., 2004). Also in patients with GBM such tumor antigen specific CD8+ T cells have been identified in the circulating blood (Tang et al., 2005). A specific cancer immunotherapy can be based on the use of antibodies directed against specific antigens, which are expressed in tumors. However, the more recently described immunological therapy strategy for GBM with an antibody (Bevacizumab), which blocks angiogenesis by binding to the vascular endothelial growth factor also shows insufficient results (Lai et al., 2010). The more promising strategy seems to be the use of tumor specific vaccines, which induce a specific cellular immune reaction by induction of tumor specific CTL.

Advertisement

2. Dendritic cells in cancer therapy

A specific cellular immune response against tumors is based on the uptake and presentation of tumor antigens on MHC molecules by professional antigen presenting cells leading to the induction of antigen specific CTL, which can then attack the tumor. Dendritic cells (DC) are the most potent antigen presenting cells and have been shown to be able to induce an efficient and specific immune response (Banchereau & Steinman, 1998; Steinman, 1991; Whiteside & Odoux, 2004). In comparison to other antigen presenting cells, like B cells, which induce and activate only memory T cells, dendritic cells can stimulate naive T cells (Banchereau & Steinman, 1998; Steinman, 1991; Whiteside & Odoux, 2004). Thus, today one of the most promising immunological therapy approaches in treatment of various types of human cancer is a specific immunotherapy with autologous dendritic cells (Engleman, 2003; Gilboa, 2007; Nestle, 2000).

According to their phenotype and functional properties dendritic cells can be classified as immature and mature type (Banchereau & Steinman, 1998). Immature dendritic cells are able to capture and internalize particles and soluble antigens from the environment (Banchereau & Steinman, 1998). Upon being internalized, antigens are processed and split into peptides, which are then presented on the cell surface in association with MHC molecules (Banchereau & Steinman, 1998). In response to micro-environmental signals immature DC switch to a mature phenotype with T cell stimulatory capacity (Vieira et al., 2000; Whiteside & Odoux, 2004). Mature dendritic cells express higher levels of major histocompatibility complex class I and class II molecules as well as co-stimulatory molecules like CD80 and CD86 (Dietz et al., 2000; Whiteside & Odoux, 2004). During the maturation process DC lose their ability to take up and process antigens (Dietz et al., 2000; Whiteside & Odoux, 2004). Mature dendritic cells are characterised by a distinct cytokine expression profile and their increased ability to produce Interleukin 12 (IL-12) in response to environmental signals, a cytokine known to be necessary for induction of TH1-T cell response leading to production and proliferation of CTL (Whiteside & Odoux, 2004, Rouard et al., 2002).

Until now several methods have been published to generate dendritic cells in vitro from monocytes isolated from the peripheral blood of volunteers or cancer patients, called monocytes-derived dendritic cells (MoDC) in the presence of GM-CSF and IL-4 (Morse et al., 2002; Peters et al., 1996; Romani et al., 1996, Sallusto & Lanzavecchia 1994, Zhou & Tedder, 1996). Immature MoDC can be loaded either with tumor-cell-lysate, known tumor specific peptides or with specific RNA or DNA. Upon exposure to various maturation stimuli the antigen loaded immature MoDC can be matured and are able to induce a clinical antitumor response in patients with various types of tumors through induction of tumor specific CTL, which can then attack the tumor (Engleman, 2003; Gilboa, 2007; Morse et al., 2002; Nestle, 2000; Romani et al., 1996, Zhou & Tedder, 1996).

2.1. Dendritic cell based immunotherapy in patients with GBM

Although the tumor is hidden behind the blood brain barrier immune cells can be recruited into the central nervous system (Tambuyzer et al., 2009; Vauleon et al., 2010; Wilson, et al., 2010). Macrophages and dendritic cells are found in perivacscular zones, the choid plexus and the meninges (Vauleon et al., 2010). In gliomas infiltration of CD8+ as well as CD4+ T cells have been reported (Barcia et al., 2009; Mittelbronn et al, 2007). Thus, several types of immune reactions take place in primary brain tumors. In this context Ueda and coworkers have found T cells directed against two glioma antigens (IL13Ra2 and EphA2) within the peripheral blood mononuclear cells from a long surviving patient with an anaplastic astrocytoma (Ueda et al., 2007), showing that an efficient immune reaction in brain tumors is generally possible. Hoverer, the tumor itself shows variety of so-called “tumor escape mechanisms” which are able to dampen tumor antigen-specific immune responses. These include an immunosuppressive environment by secretion of immunosuppressive cytokines and factors such as transforming growth factor beta (TGFβ) (Schneider et al., 2006) and vascular endothelial growth factor (Naumov et al., 2006), which increase the number of tumor infiltrating regulatory T cells and lower the number of cytotoxic killer cells and others. Moreover, the high expression of HLA-4 on gliomas, upon binding to NKG2A7B has a negative effect on T and NK cell activity (Wischhusen et al., 2005). Thus, in GBM the immune reactions are mostly not effective as shown by the unfavorable course of the disease and the high level of recurrences. A specific immunotherapy based on ex vivo generated antigen loaded monocyte-derived dendritic cells may overcome this inhibition leading to a re-induction of an efficient immune reaction against GBM. Such immune responses require antigen recognition. Several groups demonstrated an over-expression of several antigens including a mutated form of the Epidermal Growth Factor Receptor (EGFRvIII), GP 100 and MAGE 1 (Okada et al., 2009; Saikali et al., 2007), which could be used as targets for induction of an antigen specific immune response by priming the MoDC with peptides from these antigens. However, using the whole tumor cell lysate as well as tumor RNA as antigen source may probably reduce the risk of an immune evasion due to clonal expansion of antigen-loss tumor cell variants (Jäger et al, 1996, Vauleon et al., 2010).

Several reports have demonstrated that specific cellular immunotherapy using dendritic cells is capable of inducing a clinical antitumor response within the immunologically privileged brain, confirming that the central nervous system (CNS) is accessible to this kind of immunotherapy. Table 1 gives an overview on clinical studies with dendritic cell vaccines performed for glioma patients. The published data are based on phase I and II trials as well as on case reports including in total over 200 patients with GBM and over 100 patients with anaplastic astrocytomas or other malignant brain tumors (table 1). Two studies and one case report include children or young adults (Ardon et al., 2010a; Caruso et al., 2004; De Vleeschouwer et al., 2004). The dendritic cells used in these studies were ex-vivo generated from patients blood monocytes using the described standard procedure and primed with peptides, autologous tumor cell lysates or tumor-derived RNA. In two trials dendritic cells were fused with autologous tumor cells (Kikuchi et al., 2001, 2004). Dendritic cells were further matured by exposure to various maturation stimuli (Ardon et al., 2010a, 2010b; De Vleeschouwer et al.; 2008; Kikuchi et al., 2004; Okada et al., 2007; Rutkowski et al., 2004; Yamanaka et al., 2005) or were used without further maturation signals (Caruso et al., 2004; De Vleeschouwer et al., 2004; Kikuchi et al., 2001; Liau et al., 2000, 2005; Yamanaka et al., 2003; Yu et al., 2001, 2004).

Antigen sourcetype of trialresponsesauthor
tumor cell lysate +
TLR-7 ligand
(children)
Phase I
median OS:
12.2 months
Ardon et al., 2010a
tumor cell lysatePhase I/IImedian OS:
24 months
Ardon et al., 2010b
tumor RNA
(children, young
adults)
Phase I
(relapsed brain
tumors)
1 PR, 4 SDCaruso et al., 2004
tumor cell lysate
(4-year old child)
case report
(recurrent GBM)
CR over 2 years De Vleeschouver et al. 2004
tumorc ell lysatePhase I/II
(recurrent GBM)
median OS:
9.6 months
De Vleeschouver et al. 2008
tumor cell lysatecase report
(recurrent GBM)
median OS:
11 months
Khan & Yaquin, 2006
fusion with tumor cells Phase I1 MR, 4 SD, 2 SDKikuchi et al., 2001
fusion with tumor cells Phase I
(recurrent GBM)
1 SD in GBMKikuchi et al., 2004
peptidescase report
(recurrent GBM)
no survival benefitLiau et al., 2000
tumor cell lysatePhase I
de novo vs.
recurrent GBM
median OS:
23.4 months vs.
18.3 months
Liau et al., 2005
tumor cell lysate or
irradiated tumor cells
Phase I2 PR
irradiated tumor
cells
Okada et al., 2007
tumor cell lysate Phase I
(recurrent GBM)
median OS :
10,5 months
Rutkowski et al., 2004
EGFRvIII antigenPhase Imedian OS:
22.8 months
Sampson et al., 2009
inactivated tumor
cells
Phase I12-/18-months survival: 46%/23%Walker et al., 2008
tumor cell lysate Phase I/II
vaccine + chemo
vs. vaccine alone
2-year-survival
42% vs. 8%
Wheeler et al., 2004
tumor cell lysatePhase II
(de novo vs.
recurrent GBM)
median OS:
642 vs. 430 days
Wheeler et al., 2008
tumor cell lysatePhase I/IIOS: "/> 200 weeksYamanaka et al., 2003
tumor cell lysate
+ TLR-4 ligand
Phase I/II
(recurrent GBM)
median OS: 480
(+ TLR-4 ligand)
vs. 400 days
Yamanaka et al., 2005
antigens Phase Imedian OS:
455 days
Yu et al., 2001

Table 1.

Clinical Studies for GBM treated with autologous DCAbbreviations: OS: Overall Survival; PR: Partial Remission; CR: Complete Remission; SD: Stable Disease

For all used methods of MoDC generation a clinical response was described. With the exception of one trial (Yamanaka et al., 2003), where some patients receive an intracerebral injection of dendritic cells, the vaccines were administered intradermally or subcutaneously. The reported side effects were in general low besides one case with grade IV neurotoxicity (Yamanaka et al., 2005). Such reactions are probably based on an excessive inflammatory process within the brain leading to the development of severe edema. Based on our experiences, in those cases, an immune suppressive treatment with corticosteroids has to be accompanied to modulate the highly inflammatory process, which can of course counteract the efficacy of the immune response. The reported major side effects are short term flu like symptoms like fever, headaches and chill. Thus, this treatment is well tolerable without having a negative impact on the quality of life.

Clinical responses based on radiological findings, which were performed for a part of the treated patients (n=108 patients with GBM) consisted of complete remissions (3.7 %), partial remissions (4.6 %), stable diseases (12.9 %) and mixed responses (5.5 %). In some trials a prolongation of the overall survival compared with historical data or non randomized control groups are described (Ardon et al., 2010b; De Vleeschouwer et al., 2008; Liau et al., 2005; Sampson et al., 2009; Rutkowski et al., 2004; Wheeler et al., 2008; Yamanaka et al., 2003, 2005; Yu et al., 2001, 2004). In a pilot trial published by Ardon and coworkers 8 patients with de novo diagnosed GBM received an immunotherapy with tumor cell lysate pulsed dendritic cells after surgery and radio-chemotherapy. The median overall survival was 24 months ranging from 13 to 44 months (Ardon et al., 2010b). Comparable results with a median survival of 22.8 months are reported from Sampson and coworkers using dendritic cells pulsed with the mutated EGFRvIII antigen coupled to KLH (Sampson et al., 2009). Even in patients with relapsed GBM a dendritic cell therapy is reported to be successful. De Vleeschouwer and coworkers reported a case of a 4 year old child, who received a dendritic cell therapy after surgery of recurrence. 2 years after onset of the immune therapy the child still remains in a complete remission (De Vleeschouwer et al., 2004). In a phase I/II trial performed by the same group (De Vleeschouwer et al., 2008) 56 patients were treated with dendritic cells pulsed with tumor cell lysate after surgery of recurrences. The median overall survival was 9.6 months with a 2-year survival rate of 14.8 % after onset of the therapy.

Besides these very promising results the objective clinical responses are low and a lot of patients fail to respond to the therapy. One main reason for low clinical efficacy of MoDC-based anticancer therapies could be due to low IL-12 and high IL-10 production by the MoDC favoring a TH2 rather than a TH1 immune response. In fact, a lot of clinically approved maturation protocols mature MoDC insufficiently in a TH1-polarizing direction (Hildenbrandt et al., 2007, 2008; Mailliard et al., 2004; Zobywalski et al., 2007). Hildenbrandt and coworkers found that the used cytokine cocktail with TNF-a, IL-1ß, IL-6 and PGE2 according to Jonuleit et al. (Jonuleit et al., 1997) induce the phenotypic characteristics of fully matured MoDC expressing high levels of CD83, MHC, costimulatory molecules and CCR7, but secrete high levels of IL-10 and only low levels of IL-12p70 suggesting a preference for a TH2-polarisation (Hildenbrandt et al., 2008). For this reason, improved maturation protocols are required. In this context the activation of Toll-like receptors (TLR) by adding TLR-ligands to the MoDC cultures becomes an interesting alternative to mature monocytes-derived dendritic cells.

2.1.1. Toll-like receptor agonists in maturation protocols for dendritic cells

The expression of certain Toll-like receptors (TLR), a family yet 11 identified transmembrane receptors, expressed on dendritic cells plays a crucial role in the recognition of microbial or viral infections (Medzhitov, 2001). TLR mediated activation of dendritic cells is supposed to be vital for the generation of a TH1 effector response (Schnare et al., 2001). Thus, TLR ligands, which are molecular signatures of microbial or viral infections, may signal the presence of an infection (danger signal), which leads to maturation of DC with a TH1 polarization profile (Akira et al., 2001; Medzhitov, 2001). Interestingly, the TLR 3, a cell surface receptor for double-stranded RNA as well as for the synthetic analogue poly I:C, is predominantly expressed on dendritic cells (Alexopoulou et al., 2001; Muzio et al., 2000). Bone marrow derived immature dendritic cells activated by poly I:C produce large amounts of IL-12 and up-regulate MHC as well as co-stimulatory molecules on their surface (Alexopoulou et al., 2001).

Several TLR are also expressed on monocytes and immature monocytes-derived dendritic cells (Visintin et al., 2001). Moreover, it has been shown that immature MoDC have an increase in TLR 3 expression as compared to monocytes, indicating that in MoDC this TLR may play an important role (Visintin et al., 2001). At the ASCO meeting 2007 our group have show that immature MoDC from patients with GBM when activated by adding the TLR 3 ligand poly I:C switch to a mature phenotype with up-regulation of the maturation marker CD83 and of co-stimulatory molecules. After short time exposure to IFN-γ these matured MoDC were able to produce IL-12p70 at a higher level than IL-10 which clearly favours a TH1 response (Nesselhut et al., 2007). Moreover the IL-12/IL-10 ratio seems to be correlated to clinical response. The use of poly I:C is currently tested in a phase I/II trial and recent published data show that poly I:C boosted DC are safe and have clinical anti tumor activity (Okada et al., 2010). Furthermore, the authors found a correlation between the IL-12 production level and the time to progression (Okada et al, 2010). Comparable data was seen by Yamanaka and coworkers using DC matured with a ligand to TLR 4. Patients who received TLR matured DC had longer survival than those vaccinated with immature DC (Yamanaka et al., 2005). Van Gool et al. reported an improvement of the progression free survival of glioma patients, when the skin at the injection site of the mature DC was pretreated with the TLR-7 agonist Imiquimod (van Gool et al., 2009). Concordant with findings from our group Hildebrandt et al. found that the addition of INF-γ to TLR-agonists can improve the maturation status as well as the TH1 polarization of MoDC (Hildenbrandt et al., 2008). However, most TLR-agonists are not clinically approved and therefore are not allowed to be used in clinical trials, yet. In this context it is interesting, that Hildenbrandt et al. could show that IFN-γ when added to the clinical-grade cytokine cocktail (Jonuleit et al., 1997) induces the production of lower levels of IL-10 and higher levels of IL-12p70, demonstrating the TH1-prolarizing capacity of this cytokine (Hildenbrandt et al., 2008).

2.1.2. Impact of cancer induced immune suppression on the efficacy of denditic cell therapy

During the last years several investigations show an increasing evidence of a strong association between chronic infection, inflammation and cancer development as well as tumor progression (Balkwill & Mantovani, 2001; Coussens & Werb, 2002). Inflammation is a physiological process occurring after tissue damage, infections caused by microbial or viral pathogen, chemical irritation or wounding (Philipp et al., 2004.) In fact, many of the same inflammatory mediators that are secreted by wounds are found in the tumor microenvironment. Immune cells infiltrate into the developing tumor site and establish an inflammatory tumor microenvironment (Balkwill & Mantovani, 2001; Whiteside, 2008). As wound healing is self limited cancer can be described as “wounds that do not heal” (Dvorak, 1986). Meanwhile it is well established that the tumor per se can change the immunological balance into an inflammatory microenvironment by release of certain cytokines and chemokines leading not only to the promotion of tumor growth but also to the inhibition of an efficient immune response. This may be one of the reasons for an unsuccessful immunotherapy for GBM patients based on dendritic cells (Couldwell et al., 1991, Nitta et al., 1994, Hishii et al, 1995, Weller & Fontana, 1995). An example is the intratumoral expression of TGF-ß, which can suppress an adaptive antigen specific immune response. Moreover, TGF- ß expression was shown to be a predictive factor for the clinical outcome in vaccinated patients (Liau et al., 2005).

Taken together, an efficient induction of a clinical antitumor response requires both a polarization of MoDC in a TH1 direction as well as a change of the immune suppressive tumor associated microenvironment. Beside several cytokines which promote an immune suppressive microenvironment, regulatory T cells (T-reg), which are required for maintaining immunological self tolerance (Sakaguchi, 2005), play an important role in the regulation of an immune response and thus may limit an antigen specific immune response in cancer (Borsellino et al. 2007; Deaglio et al. 2007; Mandapathil et a., 2009; Nizar et al, 2009).

Several subpopulations of T-reg have been described (Sakaguchi, 2005; Seddiki et al., 2006). Most commonly used is the CD4+, CD25 +high, CD127-/dim phenotype with intracellular expression of the transcription factor FoxP3 (Seddiki et al., 2006). More recently, a sub-population of CD39+ T-reg was described (Borsellino et al. 2007; Deaglio et al., 2007). CD39 belongs to the family of ecto-nucleoside triphosphate-diphosphatehydrolase-1 and hydrolyses extracellular ATP. This leads to a pericellular accumulation of the nucleoside adenosine, which is known to play an important role in inhibiting effector functions of activated T cells (Borsellino et al. 2007, Huang et al., 1997). The authors propose that the immune suppressive activity of T-reg is partly due to this enzymatic activity of CD39 and thus may play a crucial role in inhibition of antigen specific T cell response. Woo et al. were the first who reported an increased percentage of T-reg cells in tumor infiltrating lymphocytes in non-small-cell-lung cancer and ovarian cancer (Woo et al., 2001). Meanwhile accumulating T-reg cells are detected in the tumor microenvironment, ascites as well as in the peripheral blood of cancer patients and have been associated with poor prognosis (Beyer & Schultze 2010; Nizar et al. 2009, Wolf et al., 2003).

Our group have shown that subpopulations of T-reg characterized by a CD4+, CD25 +high as well as CD127-/dim or CD39+ expression profile can be detected in the peripheral blood of healthy donors, tumor free patients as well as patient with tumor load (Neßelhut et al., 2009). The percentage of T-reg was significantly higher in cancer patients, including patients with GBM, as compared to healthy donors. Moreover, patients with advanced tumors show higher levels of T-reg cells as compared to patients with no evidence of disease. Several other groups reported an increase of T-reg cells in GBM patients in peripheral blood as well as within the tumor microenvironment (Fecci et al., 2006a; Grauer et al., 2007; Hussain et al., 2006, El Andaloussi et al., 2006). Measurement of T-reg cells along with an immune therapy may be useful to discriminate between non- responding and responding patients. In fact, first analysis of our group show that rising T-reg numbers are partly associated with lack of clinical response to a dendritic cell therapy in patients with GBM. Thus, the elimination of excessive T-reg cell numbers seems to be a promising approach to improve the efficacy of a dendritic cell based immune therapy.

Several chemotherapeutical agents have immune stimulatory and T-reg modulating effects when used at lower dosages with reduced toxic effects (Nizar et al., 2009). Beside this option there are some reagents which target T-reg directly through recognition of CD25 (Morse et al., 2008, Fecci 2006 b) or through CTLA-4 blockade (Ribas et al., 2009). Some of these approaches are tested in association with DC therapy in cancer (Chiringelli et al, 2004, Ribas et al. 2009, Morse et al., 2008) including GBM (Fecci ate al. 2006 b, Maes et al., 2009). Interestingly, in a rat glioma model low dose temozolamide could also decrease the number of T-reg cells. (Banissi et al, 2009). In this context it is noteworthy that Wheeler et al. show that GBM patients, who responded to DC therapy, also exhibited a better response to a following chemotherapy (Wheeler et al., 2004). In the study published by Ardon et al. immunotherapy based on dendritic cells started after radio-chemotherapy (Ardon et al., 2010b). One can suppose that the chemotherapy with temozolamide has lead to a decrease of T-reg cells and thus enhanced the immunological response causing the high 24 months median survival for the treated patients. The combination of dendritic cell therapy and adjuvant chemotherapy was also tested in malignant astrocytomas (Walker et al., 2008). The 12- and 18-months survival rates were 46% and 23%, respectively. Depletion or inhibition of T-reg may thus improve the efficacy of dendritic cell based therapy.

2.1.3. Newcastle disease virus in treatment of GBM; a tool for improving DC therapy

Besides dendritic cell therapy another promising approach for the treatment of malignant brain tumors is the treatment with replication-selective viruses, also called oncolytic viruses. This is based on the fact that most tumor cells are more or less unable of an effective virus defense. This approach is also known as virotherapy. The application of viruses for cancer treatment is based on reports since the beginning of the 20th century on temporary improvement of cancer following natural viral infections or vaccinations against viral diseases. (DePace 1912). Meanwhile several replication competent viruses (mainly herpes and adenoviruses) were tested in vitro, in animal models as well as in phase I/II clinical trials for treatment of malignant brain tumors (Shah et al., 2003; Rainov & Ren, 2003; Wollmann et al., 2005). However, the viruses have to be genetically modified in a way that makes sure that they selectively infect and replicate in tumor cells. Within the viruses tested for human anticancer treatment the Newcastle Disease Virus (NDV), an enveloped poultry virus with single strained RNA as genetic material, seems to be one of the most promising candidates. NDV is not a pathogen for humans, and is absolutely harmless causing only mild flu-like symptoms or conjunctivitis in the worst of cases (Lorence et al., 2001; Reichard et al., 1992). NDV shows a natural distinct tropism for cancer cells. Cancer cells infected with NDV can be killed directly by the virus within a short time after infection, whereas normal infected cells are not lysed by NDV.

As described earlier tumor cell lysate may be the better antigen source for priming of dendritc cells because it contains the whole antigen repertoire of the tumor. However, it has to be taken in mind that most of the antigens expressed in tumors are poor inducers of immune response and are often recognized by the immune system as self antigens (Vergati et al., 2010). Opposed to this adjuvant active specific immunization based on tumor cells modified with a low pathogenic strain of the NDV has been reported to achieve sustained immune responses in patients with advanced colonic cancer and liver metastasis (Lehner et al., 1990 Schulze et al., 2009). NDV can have lytic activity on tumor cells directly as well as immune stimulating properties that affect both innate and adaptive immune responses. Infection of tumor cells with live NDV results in a potent up-regulation of cell adhesion molecules on the tumor cells surface (Lehner et al. 190; Wasburn et al., 2002). Moreover, NDV infection of tumor cells leads to an improved tumor cell / T cell interaction and an increased T cell co-stimulatory activity (Ertel et al., 1993; Termeer et al., 2000). Expression of viral proteins on the tumor cell surface and presence of virus derived pathogen-associated molecular patterns (e.g. double-stranded RNA) result in breaking of host tolerance towards the tumor in vitro (Bai et al., 2002). The T cell stimulatory action of dendritic cells pulsed with lysates of NDV infected tumor cells as well as the antitumor cytotoxicity of macrophages and monocytes is increased (Schirrmacher et al., 2000; Washburn et al., 2003; Zeng et al., 2002). Finally, NDV induces an increased production of various cytokines, e.g. Interferon-α as well as chemokines, influencing the migration, the activation status and cytotoxic activity of various immune cells (Lokuta et al., 1996; Schirrmacher 2005, Schlag et al., 1992). Clinical phase I and II studies in various tumor entities have proven the safety of active specific immunization with NDV-modified tumor cells. A detailed description of the mechanisms of action of NDV modified tumor cell vaccines and results from other studies in cancer patients were reviewed by Schirrmacher (Schirrmacher, 2005).

In malignant brain tumors case reports as well as clinical phase I/II studies have shown that treatment with intravenously applied NDV as well as with vaccines utilizing NDV modified tumor cells can induce a clinical anti-tumor response in malignant brain tumors with objective clinical responses as well as with a trend towards improvement of overall survival (Csatary & Bakacs, 1999, Csatary et al., 2004; Freeman et al., 2005; Schneider et al., 2001; Wagner et al., 2006). Recent results from our group show that a therapy with dendritic cells in combination with the NDV virotherapy may improve the clinical anti-tumor response in patients with GBM (Neßelhut et al., 2007, 2011). Patients were pre-treated with intravenously administration of NDV. Dendritic cells were primed with NDV modified tumor cells or with NDV alone in patient with tumor recurrence. When tested in vitro, NDV primed MoDC of such treated patients induce the activation of autologous CD8+ T cells with release of IFN-γ. This leads to the hypothesis that, if viral antigens are expressed on the tumor cell surface, a NDV specific dendritic cell based therapy may lead to the induction of NDV specific CD8+ T cells and thus to the induction of a specific immune response against the virus infected cancer cell (Neßelhut et al., 2011).

Advertisement

3. Conclusion

Taken together a dendritic cell therapy as well as the NDV virotherapy have shown clinical efficacy and have been proven to be safe with only minimal side-effects. Especially patients with primary resistance against temozolamide caused by the MGMT status could benefit from such alternative therapy strategies. Furthermore, new insights in the nature and biology of glioma tumor stem cells may improve an immunological concept by using tumor stem cell specific antigens as targets in dendritic cell therapy as it was recently shown (Pellegatta et al., 2006, Stupp & Hegi,, Xu et al., 2009). An immunotherapy with dendritic cells can either be used in adjuvant, curative settings to avoid or delay tumor recurrence or in palliative settings to keep the disease stable offering the patients a chance for prolonged overall survival. Based on current results it has to be supposed that patients with minor tumor burden or no evidence of disease are the best candidates for an immunotherapy.

The main reasons for a deficient anti-tumor immune response are most likely the immune suppressive tumor microenvironment and the up-regulation of immune suppressive cells. Thus, a combination with therapies, which can modulate the tumor microenvironment as well as lower the percentage of the immune suppressive T-reg cells should be considered as treatment prior to or in combination with dendritic cell based immune therapy. Additionally, glioma cells are considered to be poor antigen-presenting cells. In order to promote antigen-specific recognition and avidity of potential anti-tumor effector cells, the biology of antigens expressed by malignant glioma cells must be taken into consideration while designing protocols for programming autologous dendritic cells and defining the appropriate patient. For example, Prins and coworkers could recently shown that the gene expression profile of the individual glioma can identify a subgroup of patients, that may be more responsive to dendritic cell based immunotherapy (Prins et al., 2011). In this context the combination with NDV seems to be a promising tool to modify tumor cells in vitro as well as in vivo to a more immunogenic phenotype resulting in better immune recognition as well as in better immune response.

The encouraging results of phase I/II trials have to be confirmed in phase III studies with special emphasis on the maturation protocols for improvement of TH1-polarisation of dendritic cells, which so far are still a matter of controversial discussion. Moreover, the optimal antigen source, cell number of injected dendritic cells, frequency of vaccinations as well as the right time point for vaccination are still not fully defined. Future studies should also involve an immune monitoring of vaccinated patients. Immune monitoring methods can include quantification of CD8+ and CD4+ T cells generated in response to the antigen or measurement of the functional status of T cells analyzed by cytokine production. However, it has to be taken into account that in vitro measurement of T cell responses do not always correlate with clinical outcome (Carpentier & Meng, 2006). Thus, the best immunological monitoring strategy is still intensively debated.

Finally, clinical trials for the evaluation of immune therapeutical approaches have to be designed in a different way as for chemotherapeutical drugs. In this context the overall survival rather than the reduction of tumor burden is probably the best primary endpoint. Based on such a study design ProvengeTM, an immunological treatment based on antigen presenting cells, received the FDA approval for treatment of hormone refractory prostate cancer in spring 2010.

References

  1. 1. AkiraS.TakedaK.KaishoT.2001Toll-like receptors: critical proteins linking innate and acquired immunity. Nature Immunology, 28Aug 2001), 675680
  2. 2. AlexopoulouL.Czopik-HoltA.MedzhitovR.FlavellR.2001Recognition of double stranded RNA and activation of NF-kb by Toll-like receptor 3. Nature, 4136857Oct 2001), 732738
  3. 3. ArdonH.De VleeschouwerS.Van CalenberghF.ClaesL.KrammC. M.et al.2010aAdjuvant dendritic cell-based tumour vaccination for children with malignant brain tumours. Pediatric Blood and Cancer, 544April 2010), 519525
  4. 4. ArdonH.Van GoolS.LopesI. S.MaesW.SciotR.et al.2010bIntegration of autologous dendritic cell-based immunotherapy in the primary treatment for patients with newly diagnosed glioblastoma multiforme: a pilot study. Journal of Neurooncology, 992Sep 2010), 261272
  5. 5. BaiL.KoopmannJ.FiolaC.FournierP.SchirrmacherV.2002Dendritic cells pulsed with viral oncolysates potently stimulate autologous T cells from cancer patients. International Journal of Oncology, 214Oct 2002), 685694
  6. 6. BalkwillF.MantovaniA.2001Inflammation and cancer: back to Virchow? Lancet, 3579255Feb 2001), 539545
  7. 7. BanchereauJ.SteinmanR. M.1998Dendritic cells and the control of immunity. Nature, 3926673Mar 1998), 245252
  8. 8. BanissiC.ChiringhelliF.ChenL.CarpentierA. F.2009Treg depletion with a low-dose metronomic temozolomide regimen in a rat glioma model. Cancer Immunology Immunotherapy, 5810Oct 2009), 16271634
  9. 9. BarciaC.GomezA.Gallego-SanchezJ. M.Perez-VallésA.CastroM. G.et al.2009Infiltrating CTLs in human glioblastoma establish immunological synapses with tumorigenic cells. American Journal of Pathology, 1752Aug 2009), 786789
  10. 10. BeyerM.SchultzeJ. L.2006Regulatory T cells in cancer. Blood, 1083Aug 2006), 804811
  11. 11. BorsellinoG.KleinewietfeldM.Di MitriD.SternjakA.DiamantiniA.et al.2007Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood, 110N0. 4, (Aug 2007), 12251232
  12. 12. BucknerJ. C.2003Factors influencing survival in high-grade gliomas. Seminars in Oncology, 30Suppl. 19, 1014
  13. 13. CarpentierA. F.MengY.2006Recent advances in immunotherapy for human glioma. Current Opinion Oncology, 186Nov 2006), 631636
  14. 14. CarusoD. A.OrmeL. M.NealeA. M.RadcliffF. J.AmorG. M.et al.2004Results of a phase 1 study utilizing monocyte-derived dendritic cells pulsed with tumor RNA in children and young adults with brain cancer. Journal of Neurooncology, 63Jul 2004), 236246
  15. 15. ChangC. H.HortonJ.SchoenfeldD.SalazerO.Perez-TamayoR.et al.1983Comparison of postoperative radiotherapy and combined postoperative radiotherapy and chemotherapy in the multidisciplinary management of malignant gliomas: a joint Radiaton Therapy Oncology Group and Eastern Cooperative Oncology Group study. Cancer, 529971007
  16. 16. CouldwellW. T.Dore-DuffyP.ApuzzoM. L. J.AntelJ. P.1991Malignant glioma modulation of immune function: relative contribution of different soluble factors. Journal of Neuroimmunology, 332Aug 1991), 8996
  17. 17. CoussensL. M.WerbZ.2002Inflammation and cancer. Nature, 4206917Dec 2002), 860867
  18. 18. CsataryL. K.BakacsT.1999Newcastle disease virus vaccine (MTH-68/H) in a patient with high-grade glioblastoma. Jama, 28117May 1999), 15881589
  19. 19. CsataryL. K.GosztonyiG.SzeberenyiJ.FabianZ.LiszkaV.et al.2004MTH-68/H oncolytic viral treatment in human high-grade gliomas. Journal of Neurooncology, 671-2Mar-Apr), 8393
  20. 20. CurranW. J.Jr ScottC. B.HortonJ.NelsonJ. S.ASWeinsteinet.al1993Recursive portioning analysis of prognostic factors in three Radiation Therapy Oncology Group malignant glioma trials. Journal of National Cancer Institute, 859May 1993), 704710
  21. 21. DeaglioS.DwyerK. M.GaoFriedmanW.D.UshevaA.et al.2007Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. Journal of Experimental Medicine, 2046Jun 2007), 12571265
  22. 22. De AngelisL. M.2001Brain tumors. New England Journal of Medicine, 3442Jan 2001), 114123
  23. 23. De PaceN. G.1912Sulla scomparsa di un enorme cancro vegetante del callo dell`utero senza cura chirurgica. Ginecologia, 98288
  24. 24. De VleeschouwerS.Van CalenberghF.DemaerelP.FlamenP.RutkowskiS.et al.2004Transient local response and persistent tumor control in a child with recurrent malignant glioma: treatment with combination therapy including dendritic cell therapy: case report. Journal of Neurosurgery, 1005May 2004), 492497
  25. 25. De VleeschouwerS.FieuwsS.RutkowskiS.Van CalenberghF.Van LoonJ.et al.2008Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clinical Cancer Research, 1410May 2008), 30983104
  26. 26. DvorakH. F.1986Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. New England Journal of Medicine, 31526Dec 1986), 16501659
  27. 27. DietzA. B.BulurP. A.KnutsonG. J.MatasicR.Vuk-PavlovicS.2000Maturation of human monocyte-derived dendritic cells studied by microarray hybridization. Biochem Biophys Res Commun, 2753Sep 2000), 731738
  28. 28. El AndaloussiA.MSLesniak2006An increase in CD4+ CD25+FOXP3+ regulatory T cells in tumor-infiltrating lymphocytes of human glioblastoma multiforme. Neurooncology, 83Jul 2006), 234243
  29. 29. EnglemanE. G.2003Dendritic cell-based cancer immunotherapy. Seminars in Oncology, 303Jun 2003), 2329
  30. 30. ErtelC.MillarN. S.EmmersonP. T.SchirrmacherV.vonHoegen. P.1993Viral hemaglutinin augments peptide-specific cytotoxic T cell responses. European Journal of Immunology, 2310Oct 1993), 25922596
  31. 31. FecciP. E.MitchellD. A.WhitesidesJ. F.XieW.FriedmannA. H.et al.2006Increased regulatory T-cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Research, 666Mar 2006), 32943302
  32. 32. FecciP. E.SweeneyA. E.GrossiP. M.NairS. K.CALearnet.al2006Systemic anti-CD25 monoclonal antibody administration safely enhances immunity in murine glioma without eliminating regulatory T cells. Clinical Cancer Research, 1214Jul 2006), 42944305
  33. 33. FreemanA. I.Zackay-RonesZ.GomoriJ. M.LinetskyE.RasoolyL.et al.2005Phase I/II trial of intravenous NDV-HUJ oncolytic virus in recurrent glioblastoma multiforme.Molecular Therapy, 131Jan 2005), 221228
  34. 34. GhiringhelliF.LarmonierN.SchmittE.ParcellierA.CathelinD.et al.2004CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. European Journal of Immunology, 342Feb 2004), 336344
  35. 35. GilboaE.2007DC-based cancer vaccines. Journal of Clinical Investigation, 1175May 2007), 11951203
  36. 36. GrauerO. M.NierkensS.BenninkE.ToonenL. W.BoonL.et al.2007CD4+FoxP3+ regulatory T cells gradually accumulate in gliomas during tumor growth and efficiently suppress antiglioma immune responses in vivo. International Journal of Cancer, 1211Jul 2007), 95105
  37. 37. GreenS. B.ByarD. P.MDWalkerPistenmaaD. A.AlexanderE.et al.1983Comparisons of carmustine, procarbazine, and high-dose methylprednisolone as additions to surgery and radiotherapy for the treatment of malignant glioma. Cancer Treat Rep, 672Feb 1983), 121132
  38. 38. HildenbrandB.SauerB.KalisO.StollC.MAFreudenberget.al2007Immunotherapy of patients with hormone-refractory prostate carcinoma pre-treated with interferon-gamma and vaccinated with autologous PSA-peptide loaded dendritic cells- A pilot study. Prostate, 672Apr 2007), 500508
  39. 39. HildenbrandB.LorenzenD.SauerB.HertkornC.MAFreudenberget.al2008IFN-g enhances TH1 polarization of monocyte-derived dendritic cells matured with clinical-grade cytokines using serum-free conditions. Anticancer Research, 283AMay-Jun 2008), 14671476
  40. 40. HishiiM.NittaT.IshidaH.EbatoM.KurosuetA.al1995Human glioma derived interleukin-10 inhibits antitumor immune responses in vitro. Neurosurgery, 376Dec 1995), 11601166
  41. 41. HuangS.ApasovS.KoshubaM.SitkowskiM.1997Role of A2a extracellular adenosine receptor-mediated signaling in adenosine-mediated inhibition of T-cell activation and expansion. Blood, 904Aug 1997), 16001610
  42. 42. HussainS. F.YangD.SukiD.GrimmE.HeimbergerA. B.2006Innate immune functions of microglia isolated from human glioma patients. Journal of Translational Medicine, 4Article ID, (Jul 2006), 15
  43. 43. JägerE.RinghofferM.KarbachJ.ArandM.OeschF.KnuthA.1996Inverse relationship of melanocyte differentiation antigen expression in melanoma tissues and CD8+ cytotoxic-T-cell responses: evidence of immune selection of antigen-loss variants in vivo. International Journal of Cancer, 664May 1996), 470476
  44. 44. CAJanewayMJShlomchikTraversP.WalportM.2004Using the immune response to attack tumors, In: Immunobiology 6th edn., Janeway CA, Shlomchik MJ, Travers P, Walport J (eds), 630642Garland Science, New York,
  45. 45. JonuleitH.KuhnU.MullerG.SteinbrinkK.ParagnikL.et al.1997Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum- free conditions. European Journal of Immunology, 2712Dec 1997), 31353142
  46. 46. KikuchiT.AkasakiY.IrieM.HommaS.AbeT.OhnoT.2001Results of a phase I clinical trial of vaccination of glioma patients with fusions of dendritic and glioma cells. Cancer Immunology Immunotherapy, 507Sep 2001), 337344
  47. 47. KikuchiT.AkasakiY.AbeT.FukudaT.SaotomeH.et al.2004Vaccination of glioma patients with fusions of dendritic and glioma cells and recombinant human interleukin 12. Journal of Immunotherapy, 276Nov-Dec 2004), 452459
  48. 48. KhanJ. A.YaquinS.2006Dendritic cell therapy with improved outcome in glioma multiforme-a case report. J Zhejiang Univ Science B, 72Feb 2006), 114117
  49. 49. LaiA.TranA.NghiemphuP. L.PopeW. B.SolisO. E.et al.2010Phase II study of bevacizumab plus temozolomide during and after radiation therapy for patients with newly diagnosed glioblastoma multiforme. Journal of Clinical Oncology, 292Jan 2010), 142148
  50. 50. LehnerB.SchlagP.LiebrichW.SchirrmacherV.1990Postoperative active specific immunization in curatively resected colorectal cancer patients with a virus-modified autologous tumor cell vaccine. Cancer Immunology Immunotherapy, 323173178
  51. 51. LiauL. M.BlackK. L.MartinN. A.SykesS. N.BronsteinJ. M.et al.2000Treatment of a patient by vaccination with autologous dendritic cells pulsed with allogeneic major histocompatibility complex class I matched tumor peptides. Case report. Neurosurgical Focus, 96Dec 2000), e8
  52. 52. LiauL. M.PrinsR. M.KiertscherS. M.OdesaS. K.KremenT. J.et al.2005Dendritic cell vaccination in glioblastoma patients induces systemic and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment. Clinical Cancer Research, 1115Aug 2005), 55155525
  53. 53. LiuG.YuanX.ZengZ.TuniciP.NgH.et al.2006Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer, 5Dec 2006), 67
  54. 54. MALokutaMaherJ.NoeK. H.PithaP. M.ShinM. L.ShinH. S.1996Mechanisms of murine RANTES chemokine gene induction by Newcastle disease virus. J Biol Chem, 27123Jan 1996), 1373113738
  55. 55. LorenceR. M.MSRobertsGroeneW. S.RabinH.2001Replication-competent, oncolytic Newcastle disease virus for cancer therapy. In: Replication competent viruses for cancer therapy, 22Hernaiz-Driever, Rabkin (eds), 160182Karger, Basel
  56. 56. MaesW.RosasG.VerbinnenB.BoonL.De VleeschouwerS.CeuppensJ. L.Van GoolS. W.2009DC vaccination with anti-CD25 treatment leads to long-term immunity against experimental glioma. Neurooncology, 115Oct 2009), 529542
  57. 57. MandapathilM.HilldorferB.MJSzczepanskiCzystowskaM.SzajnikM.et al.2009Generation and accumulation of immunosuppressive adenosine by human CD4+CD25highFOXP3+ regulatory T cells. J Biol Chem, 28510Mar 2009), 71767186
  58. 58. MailliardR. B.Wankowicz-KalinskaA.CaiQ.WesaA.HilkensC. M.et al.2004A-Type-1 polarized dendritic cells: A novel immunization tool with optimized CTL-inducing activity. Cancer Research, 6417Sep 2004), 9345937
  59. 59. MedzhitovR.2001Toll-like receptors and innate immunity. Nat Rev Immunol, 12Nov 2001), 135145
  60. 60. MittelbronnM.SimonP.LöfflerC.CapperD.BunzB.et al.2007Elevated HLAE levels in human glioblastomas but not in grade I to III astrocytomas correlate with infiltrating CD8+ cells. Journal of Neuroimmunology, 1891-2Sep 2007), 5058
  61. 61. MAMorseMoscaP.ClayT. M.LyerlyH. K.2002Dendritic cell maturation in active immunotherapy strategies. Expert Opin Biol Ther, 21Jan 2002), 3543
  62. 62. MAMorseHobeikaA. C.OsadaT.SerraD.NiedzwieckiD.et al.2008Depletion of human regulatory T cells specifically enhances antigen-specific immune responses to cancer vaccines. Blood, 1123Aug 2008), 610618
  63. 63. MuzioM.BosisioD.PolentaruttiN.D´amicoG.StoppacciaroA.atal. (2000Differential expression and regulation of Toll-like receptors (TLR) in human leukocytes: selective expression of TLR3 in dendritic cells. Journal of Immunology, 1641Jun 2000), 59986004
  64. 64. NaumovG. N.BenderE.ZurakowskiD.KangS. Y.SampsonD.et al.2006A model of human tumor dormancy: an angiogenic switch from the angiogenic phenotype. Journal National Cancer Institute, 985Mar 2006), 316325
  65. 65. NeßelhutJ.NeßelhutT.ChangR.MarxD.BrockmannW.et al.2007Dendritic cell therapy in glioblastoma multiforme. Journal of Clinical Oncology 25 (18S) suppl: abstract 3065, Chicago, May-Jun 2007
  66. 66. NeßelhutJ.LorenzenD.MarxD.ChangR. Y.MatthesC.et al.2009Cellular immune suppression in cancer patients and its implication for dendritic cell therapy. Journal of Clinical Oncology 27 (15s) suppl: abstract 3028, Orlando, May-Jun 2009
  67. 67. NeßelhutJ.MarxD.LorenzenD.CillienN.GoebelW.et al.2011Improvement of dendritic cell therapy in glioblastoma multiforme WHO IV by Newcastle disease virus. Abstract 2508, Chicago, June 2011 (in progress)
  68. 68. NestleF. Q.2000Dendritic cell vaccination for cancer therapy. Oncogene, 1956Dec 2000), 66736679
  69. 69. NewlandsE. S.StevensM. F. G.WedgeS. R.WheelhouseR. T.BrockC.1997Temozolomide: a review of its discovery, chemical properties, pre-clinical development and clinical trials. Cancer Treat Rev, 231Jan 1997), 3561
  70. 70. NittaT.HishiiM.SatoK.OkumuraK.1994Selective expression of interleukin-10 gene within glioblastoma multiforme. Brain Research, 6491-2Jun 1994), 122128
  71. 71. NizarS.CopierJ.MeyerB.Bodman-SmithM.GalustianC.et al.2009T-regulatory cell modulation: the future of cancer immunotherapy? British Journal of Cancer, 10011Jun 2009), 16971703
  72. 72. OkadaH.LiebermanF. S.WalterK. A.LunsfordL. D.DSKondziolkaet.al2007Autologous glioma cell vaccine admixed with interleukin-4 gene transfected fibroblasts in the treatment of patients with malignant gliomas. Journal of Translational Medicine, 195Dec 2007), 67
  73. 73. OkadaH.KohanbashG.ZhuX.KastenhuberE. R.HojiA.et al.2009Immunotherapeutic approaches for glioma. Critical Reviews in Immunology, 291142
  74. 74. OkadaH.KalinskiP.UedaR.HojiA.KohanbahG.et al.2011Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with (alpha)-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. Journal of Clinical Oncology, 293Jan 2011), 330336
  75. 75. PellegattaS.PolianiP. L.CornoD.MenghiF.GhielmettiF.et al.2006Neurospheres enriched in cancer stem-like cells are highly effective in eliciting a dendritic cell-mediated immune response against malignant gliomas. Cancer Research, 6621Nov 2006), 1024710252
  76. 76. PetersJ. H.GieselerR.ThieleB.SteinbachF.1996Dendritic cells: from ontogenetic orphans to myelomonocytic descendants. Immunology Today, 176Jun 1996), 273
  77. 77. PhilipM.RowleyD. A.SchreiberH.2004Inflammation as a tumor promoter in cancer induction. Semin Cancer Biol, 146Dec 2004), 433439
  78. 78. PrinsR. M.SotoH.KonkankitV.OdesaS. K.EskinA.et al.2011Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clinical Cancer Research, 176Mar 2011), 16031615
  79. 79. RainovN. G.RenH.2003Oncolytic viruses for treatment of malignant brain tumors. Acta Neurochir Suppl. 88, 113123
  80. 80. ReichardK. W.LorenceR. M.CascinoC. J.MEPeeplesWalterR. J.et al.1992Newcastle disease virus selectively kills human tumor cells. J Surg Res, 525May 1992), 448453
  81. 81. RibasA.Comin-AnduixB.ChmielowskiB.JalilJ.de la RochaP.et al.2009Dendritic cell vaccination combined with CTLA4 blockade in patients with metastatic melanoma. Clinical Cancer Research, 1519Oct 2009), 62676276
  82. 82. RomaniN.ReiderD.HeuerM.et al.1996Generation of mature dendritic cells from human blood. An improved method with special regard to clinical applicability. Journal of Immunology Methods, 1962Sep 1996), 11371151
  83. 83. RouardH.MarquetJ.LéonA.MaisonP.HaiounC.et al.2002IL-12 secreting dendritic cells are required for optimum activation of human secondary lymphoid tissue T cells. Journal of Immunotherapy, 254Jul-Aug 2002), 324333
  84. 84. RutkowskiS.De VleeschouwerS.KaempgenE.WolffJ. E.KühlJ.et al.2004Surgery and adjuvant dendritic cell-based tumour vaccination for patients with relapsed malignant glioma, a feasibility study. British Journal of Cancer, 919Nov 2004), 16561662
  85. 85. SakaguchiS.2005Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self. Nature Immunology, 64Apr 2005), 345352
  86. 86. SaikaliS.AvrilT.ColletB.HamlatA.BansardJ. Y.et al.2007Expression of nine tumour antigens in a series of human glioblastoma multiforme: interest of EGFRvIII, IL-13Rα2, gp100 and TRP-2 for immunotherapy. Journal of Neurooncology, 812Jan 2007), 139148
  87. 87. SallustoF.LanzavecchiaA.1994Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. Journal of Experimental Medicine, 1794Apr 1994), 11091118
  88. 88. SampsonJ. H.ArcherG. E.MitchellD. A.HeimbergerA. B.HerndonJ. E.2ndet.al2009An epidermal growth factor receptor variant III-targeted vaccine is safe and immunogenic in patients with glioblastoma multiforme. Molecular Cancer Therapeutics, 810Oct 2009), 27732779
  89. 89. SchirrmacherV.BaiL.UmanskyV.YuL.XingY.QianZ.2000Newcastle disease virus activates macrophages for anti-tumor activity. International Journal of Oncology, 16363373
  90. 90. SchirrmacherV.2005Clinical trials of antitumor vaccination with an autologous tumor cell vaccine modified by virus infection: improvement of patient survival based on improved antitumor immune memory. Cancer Immunology Immunotherapy, 546Jun 2005), 587598
  91. 91. SchlagP.ManasterskiM.GernethT.HohenbergerP.DueckM.et al.1992Active specific immunotherapy with Newcastle-disease-virus-modified autologous tumor cells following resection of liver metastases in colorectal cancer. First evaluation of clinical response of a phase II-trial. Cancer Immunology Immunotherapy, 355325330
  92. 92. SchnareM.BartonG. M.HoltA. C.TakedaK.AkiraS.et al.2001Toll-like receptors control activation of adaptive immune responses. Nature Immunology, 210Oct 2001), 947950
  93. 93. SchneiderT.SailerM.AnsorgeS.FirschingR.ReinholdD.2006Increased concentrations of transforming growth factor beta1 and beta2 in the plasma of patients with glioblastoma.Journal of Neurooncology, 791Aug 2001), 6165
  94. 94. SchneiderT.GerhardsR.KirchesE.FirschingR.2001Preliminary results of active specific immunization with modified tumor cell vaccine in glioblastoma multiforme. Journal of Neuooncology, 531May 2001), 3946
  95. 95. SchulzeT.KemmerW.WeitzJ.WerneckeK. D.SchirrmacheretV.al2009Efficiency of adjuvant active specific immunization with Newcastle disease virus modified tumor cells in colorectal cancer patients following resection of liver metastases: results of a prospective randomized trial. Cancer Immunology Immunotherapy, 581Jan 2009), 6169
  96. 96. SeddikiN.Santner-NananB.MartinsonJ.ZaundersJ.SassonS.et al.2006Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. Journal of Experimental Medicine, 2037Jul 2006), 16931700
  97. 97. ShahA. C.BenosD.GillepsieG. Y.MarkertJ. M.2003Oncolytic viruses: clinical applications as vectors for the treatment of malignant gliomas. Journal of Neurooncology, 653Dec 2003), 20232026
  98. 98. SinghS. K.ClarkeI. D.TerasakiM.BonnV. E.HawkinsC.2003Identification of a cancer stem cell in human brain tumors. Cancer Research, 6318Sep 2003), 58215828
  99. 99. SteinmanR. M.1991The dendritic cell system and its role in immunogenicity. Annual Review of Immunology, 9271296
  100. 100. StewartL. A.2002Chemotherapy in adult high-grade glioma: a systematic review and metaanalysis of individual patient data from 12 randomised trials. Lancet, 3599311Mar 2002), 10111018
  101. 101. StuppR.GanderM.LeyvrazS.NewlandsE.2001Current and future developments in the use of temozolomide for the treatment of brain tumours. Lancet Oncology, 29Sep 2001), 552560
  102. 102. StuppR.MasonW. P.van denBent.MJWellerM.FisherB.et al.2005Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. New England Journal of Medicine, 35210Mar 2005), 987996
  103. 103. StuppR.MEHegi2007Targeting brain-tumor stem cells. Nature Biotechnology, 252Feb 2007), 193194
  104. 104. TangJ.FlomenbergP.HarshyneL.KenyonL.AndrewsD. W.2005Glioblastoma patients exhibit circulating tumor-specific CD8+ T cells. Clinical Cancer Research, 1114Jul 2005), 52925299
  105. 105. TambuyzerB. R.PonsaertsP.NouwenJ. E.2009Microglia: gatekeepers of central nervous system immunology. Journal of Leukocyte Biology, 853Mar 2009), 352370
  106. 106. TermeerC. C.SchirrmacherV.BrockerB. B.BeckerJ. C.2000Newcastle disease virus infection induces B7-1/B7-2-independent T-cell costimulatory activity in human melanoma cells. Cancer Gene Therapy, 72Feb 2000), 316323
  107. 107. UedaR.LowK. L.ZhuX.FujitaM.SasakiK.et al.2007Spontaneous immune responses against glioma-associated antigens in a long term survivor with malignant glioma. Journal of Translational Medicine, 195Dec 2007), 68
  108. 108. UedaR.FujitaM.ZhuX.SasakiK.KastenhuberE. R.et al.2009Systemic inhibition of transforming growth factor-β in glioma-bearing mice improves the therapeutic efficacy of glioma-associated antigen peptide vaccines. Clinical Cancer Research, 1521Nov 2009), 65516559
  109. 109. Van GoolS.MaesW.ArdonH.VerschuereT.Van CauterS.De VleeschouwerS.2009Dendritic cell therapy of high-grade gliomas. Brain Pathology, 194Oct 2009), 694712
  110. 110. VauleonE.AvrilT.ColletB.MosserJ.QuillienV.1 EOF18 EOF2010Overview of cellular immunotherapy for patients with glioblastoma. Clinical and Development Immunology, (Oct 2010), pii: 689171.
  111. 111. VergatiM.IntriviciC.HuenN. Y.SchlomJ.TsangK. Y.2010Strategies for cancer vaccine development. Journal of Biomedical Biotechnology, (Jul 2010), pii: 596432 EOF14 EOF
  112. 112. VieiraP. L.de JongE. C.WierengaE. A.KapsenbergM. L.KalinskiP.2000Development of Th1-inducing capacity in myeloid dendritic cells requires environmental instruction. Journal of Immunology, 1649May 2000), 45074512
  113. 113. VisintinA.MazzoniA.SpitzerJ. H.WyllieD. H.DowerS. K.SegalD. M.2001Regulation of Toll-like receptors in human monocytes and dendritic cells. Journal of Immunology, 1661Jan 2001), 249255
  114. 114. WagnerS.CsataryC. M.GosztonyiG.KochH. C.HartmannC.et al.2006Combined treatment of pediatric high-grade glioma with the oncolytic viral strain MTH-68/H and oral valproic acid. APMIS, 11410Oct 2006), 731743
  115. 115. WalkerG.LahertyR.TomlinsonF. H.ChuahT.SchmidtC.2008Results of a phase I dendritic cell vaccine trial for malignant astrocytoma: potential interaction with adjuvant chemotherapy. Journal of Clinical Neuroscience, 152Feb 2008), 114121
  116. 116. WashburnB.SchirrmacherV.2002Human tumor cell infection by Newcastle disease virus leads to upregulation of HLA and cell adhesion molecules and to induction of interferons, chemokines and finally apoptosis. International Journal of Oncology, 211Jul 2002), 8593
  117. 117. WashburnB.MAWeigandGrosse-WildeA.JankeM.StahlH.et al.2003TNF-related apoptosis-inducing ligand mediates tumoricidal activity of human monocytes stimulated by Newcastle disease virus. Journal of Immunology, 1704Feb 2003), 18141821
  118. 118. WellerM.FontanaA.1995The failure of current immunotherapy for malignant glioma. Tumor-derived TGF- β, T-cell apoptosis, and the immune privilege of the brain. Brain Research Reviews, 212Sep 1995), 128151
  119. 119. WheelerC. J.DasA.LiuG.YuJ. S.BlackK. L.2004Clinical responsiveness of glioblastoma multiforme to chemotherapy after vaccination. Clinical Cancer Research, 1016Aug 2004), 53165326
  120. 120. WheelerC. J.BlackK. L.LiuG.MazerM.ZhangX.et al.2008Vaccination elicits correlated immune and clinical responses in glioblastoma multiforme patients. Cancer Research, 6814Jul 2008), 59555964
  121. 121. WhitesideT. L.OdouxC.2004Dendritic cellbiology and cancer therapy. Cancer Immunology Immunotherapy, 533Mar 2004), 240248
  122. 122. WhitesideT. L.2008The tumor microenvironment and its role in promoting tumor growth. Oncogene, 2745Oct 2008), 59045912
  123. 123. WilsonE. H.WeningerW.CAHunter2010Trafficking of immune cells in the central nervous system. Journal of Clinical Investigation, 1205May 2010), 13681379
  124. 124. WischhusenJ.MAFrieseMittelbronnM.MeyermannR.WellerM.2005HLA-E protects glioma cells from NKG2D-mediated immune responses in vitro: implications for immune escape in vivo. J Neuropathol Exp Neurol, 646Jun 2005), 523528
  125. 125. WolfA. M.WolfD.SteurerM.GastlG.GunsiliusE.Grubeck-LoebensteinB.2003Increase of regulatory T cells in the peripheral blood of cancer patients. Clinical Cancer Research, 92Feb 2003), 606612
  126. 126. WollmannG.TattersalP.van denPol. A. N.2005Targeting human glioblastoma cells: comparison of nine viruses with oncolytic potential. Journal of Virology, 7910May 2005), 60056022
  127. 127. WooE. Y.ChuC. S.GoletzT. J.SchliengerK.YehH.et al.2001Regulatory CD4 (+) CD25 (+) T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Research, 6112Jun 2001), 47664772
  128. 128. XuQ.LiuG.YuanX.XuM.WangH.et al.2009Antigen-specific T-cell response from dendritic cell vaccination using cancer stem-like cell-associated antigens. Stem Cells, 278Aug 2009), 13741740
  129. 129. YamanakaR.AbeT.YajimaN.TsuchiyaN.HommaJ.et al.2003Vaccination of recurrent glioma patients with tumour lysate-pulsed dendritic cells elicits immune responses: results of a clinical phase I/II trial. British Journal of Cancer, 897Oct 2003), 11721179
  130. 130. YamanakaR.HommaJ.YajimaN.TsuchiyaN.SanoM.et al.2005Clinical evaluation of dendritic cell vaccination for patients with recurrent glioma: results of a clinical phase I/II trial. Clinical Cancer Research, 1111Jun 2005), 41604167
  131. 131. YuJ. S.WheelerC. J.ZeltzerP. M.YingH.FingerD. N.et al.2001Vaccination of malignant glioma patients with peptide-pulsed dendritic cells elicits systemic cytotoxicity and intracranial T-cell infiltration. Cancer Research, 613Feb 2001), 842847
  132. 132. YuJ. S.LiuG.YingH.YongW. H.BlackK. L.WheelerC. J.2004Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific cytotoxic T-cells in patients with malignant glioma.Cancer Research6414Jul 2004), 49734979
  133. 133. YungW. K.AlbrightR. E.OlsonJ.FredericksR.FinkK.et al.2000A phase II study of temozolomide vs. procarbazine in patients with glioblastoma multiforme at first relapse. Br J Cancer, 835Sep 2000), 588593
  134. 134. ZengJ.FournierP.SchirrmacherV.2002Induction of interferon-alpha and tumor necrosis factor-related apoptosis-inducing ligand in human blood mononuclear cells by hemaglutinin-neuraminidase but not F protein of Newcastle disease virus. Virology, 2971May 2002), 1930
  135. 135. ZhouL. J.TedderT. F.1996CD14+ blood monocytes can differentiate into functionally mature CD83+ dendritic cells. Proceedings of the National Academy of Sciences of the United States of America, 936Mar 1996), 25882592
  136. 136. ZobywalskiA.JavorovicM.FrankenbergerB.PohlaH.KremmerE.et al.2007Generation of clinical grade dendritic cells with capacity to produce biologically active IL-12p70. Journal of Translational Medicine, 125Apr 2007), 518

Written By

Thomas Neßelhut, Dagmar Marx, Jan Neßelhut and Fred Fändrich

Submitted: 17 November 2010 Published: 23 August 2011