Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Jul 20.
Published in final edited form as: Crit Rev Immunol. 2009;29(1):1–42. doi: 10.1615/critrevimmunol.v29.i1.10

Immunotherapeutic Approaches for Glioma

Hideho Okada 1,*, Gary Kohanbash 1, Xinmei Zhu 1, Edward R Kastenhuber 1, Aki Hoji 1, Ryo Ueda 1, Mitsugu Fujita 1
PMCID: PMC2713019  NIHMSID: NIHMS115750  PMID: 19348609

Abstract

The development of effective immunotherapy strategies for glioma requires adequate understanding of the unique immunological microenvironment in the central nervous system (CNS) and CNS tumors. Although the CNS is often considered to be an immunologically privileged site and poses unique challenges for the delivery of effector cells and molecules, recent advances in technology and discoveries in CNS immunology suggest novel mechanisms that may significantly improve the efficacy of immunotherapy against gliomas. In this review, we first summarize recent advances in the CNS and CNS tumor immunology. We address factors that may promote immune escape of gliomas. We also review advances in passive and active immunotherapy strategies for glioma, with an emphasis on lessons learned from recent early-phase clinical trials. We also discuss novel immunotherapy strategies that have been recently tested in non-CNS tumors and show great potential for application to gliomas. Finally, we discuss how each of these promising strategies can be combined to achieve clinical benefit for patients with gliomas.

Keywords: glioma, cancer vaccines, antibody, adoptive transfer, T cells, antigen-presenting cells, tumor immunity

I. INTRODUCTION

Malignant gliomas are the most common type of primary brain tumor and a major unsolved public health problem, with more than 12,000 new cases diagnosed each year in the United States.1

On the basis of the World Health Organization (WHO) classification, the four main types of gliomas are astrocytomas, oligodendrogliomas, ependymomas, and mixed gliomas (usually oligo-astrocytomas). Astrocytomas are typically classified as pilocytic (grade I), diffuse (grade II), anaplastic (grade III), or glioblastoma multiforme (GBM) (grade IV) in order of increasing anaplasia. Categorization of astrocytomas as low (I and II) or high (III and IV) grade is generally dependent on nuclear atypia, mitotic activity, microvascular proliferation, and focal necrosis. GBM is by far the most common and most malignant glial tumor. Composed of poorly differentiated neoplastic astrocytes, glioblastomas primarily affect adults, and they are located preferentially in the cerebral hemispheres.

Patients with GBM have a median survival of approximately 15 months, whereas those with anaplastic astrocytoma (AA) have a median survival of 24 to 36 months. For patients with recurrent malignant gliomas, the median time to further tumor progression, even with therapy, is only 8 weeks.2 In addition, low-grade gliomas often progress to more malignant gliomas when they recur.2 Despite extensive research, treatment options for these tumors remain limited.3 No significant advancements in the treatment of glioblastoma have occurred in the past 25 years except for chemotherapy with Temozolomide (TMZ) combined with radiotherapy, which has demonstrated a limited prolongation (approximately 3 months, compared with radiotherapy only) of patients’ survival.4,5 The primary reason that no current treatment is curative is that the tumor is beyond the reach of local control when it is first detected clinically or radiologically. Clearly, there is an unmet clinical need for further improving treatment outcomes for patients with malignant gliomas. Recent advancements in our understanding of glioma biology should be applied to the development of effective combinational strategies, including molecularly targeted immunotherapy.

Immunotherapy, on the basis of the idea of taking advantage of the body’s physiological mechanisms to defend itself, may develop as an effective and safe treatment modality for gliomas. However, the immunological microenvironment of the CNS and tumors arising in the CNS is still believed to be suboptimal for sufficient anti-tumor immune responses to mediate clinically meaningful changes in situ (reviewed in Refs. 6, 7).

In this review, we first discuss recent advances in the CNS and CNS tumor immunology, with particular attention to factors that may promote immune escape of gliomas. We also review advances in passive and active immunotherapy strategies for glioma, as well as novel immunotherapy strategies that have been recently tested in non-CNS tumors with great potential to treat gliomas. Finally, we discuss how each of these promising strategies can be combined to achieve clinical benefits for patients with glioma.

II. TUMOR IMMUNOLOGY AND THE CENTRAL NERVOUS SYSTEM

A. CNS-Immune System Interactions

For many decades, the brain has been referred to as an immune-privileged site (1) because it has been thought to lack dendritic cells (DCs) and lymphatics, (2) because of the presence of a blood brain barrier (BBB), and (3) on the basis of studies claiming a lack of allograft rejection in the brain.8 At the time, it remained a mystery as to whether “immunosurveillance,”9 the idea that thymus-dependent T cells constantly survey tissues for transformed cells, occurs in the brain. More recent work has described the CNS as “immune specialized,”10 after strong evidence of immune-CNS interactions in diseases such as multiple sclerosis (MS), experimental autoimmune encephalitis (EAE), and brain tumors. This section focuses on three key issues of CNS immunology, including how immune cells (1) are restricted from the brain, (2) can be activated against CNS antigens, and (3) traffic to and enter the brain. This discussion provides a framework to understand current treatment strategies harnessing the immune system to treat brain tumors.

1. How Immune Cells Are Restricted from the Brain

CNS cells are known to be extremely sensitive to the toxic effects of exogenous substances, making regulation of homeostasis and proper restriction of entry to the CNS crucial. To this end, the neurovasculature has evolved specialized mechanisms to control both molecular and cellular migration into (and out of) the CNS parenchyma and cerebral spinal fluid (CSF). The CNS capillary endothelial cells are termed the BBB due to their ability to restrict passive diffusion and maintain low pinocytotic activity.11 Neuroimmunologists synonymously use the term BBB to describe both the capillary and postcapillary vessels, the latter of which is the site of T-cell migration into the brain. Restricting passive diffusion is accomplished through close cell-cell interactions known as tight junctions (TJs).10 The lack of fenestrations and the tight junctions prevent paracellular transport of large hydrophilic molecules (i.e., peptides and proteins) and cells.12 Under low magnification electron microscopy (EM), TJs appear to be fusions between the plasma membrane of juxtaposed cells, demonstrating how close these cell interactions are. However, high magnification reveals that in reality, TJs are the very close contact of cell membranes13 stabilized by specific proteins.

Many extracellular proteins have been studied as TJ proteins, primarily the occludin, claudin, and junctional adhesion molecule (JAM) families. Experimental characterization of each shows that mice carrying a null mutation in the occludin gene develop normal TJs, whereas claudins have been shown to be independently sufficient for TJ formation,14 suggesting the importance of claudins in TJ formation and regulation. Additionally, i.v. injection of monoclonal antibodies (MAbs) blocking JAM into mice has been shown to inhibit leukocyte accumulation in CSF and brain parenchyma,14 presumably by blocking leukocyte transmigration at the BBB.

The BBB does not function in isolation, and current research focuses on the BBB in the context of the neurovascular unit (NVU), which is composed of the BBB, pericytes, parenchymal membrane, and astrocytic feet.15 We know that of these NVU cells, astrocytic feet cover greater than 80% of the abluminal side of the CNS vessels.14 One study demonstrated astrocytic feet as the specific site of water entry to the brain during edema, presumably through aquaporin-4 channels.16 These astrocytes also contribute to the glia limitans, creating an added layer of protection for the brain or a barrier for immune cells. A more recent study has demonstrated that during cerebral ischemia, astrocytic swelling and detachment from the NVU results in increased permeability at the BBB.17 Thus, it seems that the BBB is just one layer of CNS capillary protection and that the capillaries are bound by a system rather than a static barrier. It is therefore important that in vitro experiments examining BBB behavior account for the function of the entire NVU.

The BBB in patients with GBM appears to be compromised. Davies18 has illustrated many variations in the capillaries of GBM microenvironments.18 In particular, GBMs secrete high levels of vascular endothelial growth factor (VEGF), leading to the upregulation of aquaporin-4 and decreased occludin and claudin-1, which correlates to edema. Furthermore, EM reveals breaks in TJs and increased pinocytosis at the TJs in GBM tissues.18 Others have reported alterations, including increased fenestrations, permeability, and a decrease in BBB-associated pericytes.19 The decrease in pericytes may be caused by endothelial cytoskeletal disorganization and a lack of cell adhesion, associated with increased edema and pericyte swelling.20 These disruptions may affect immune-cell migration by allowing for GBM-specific methods of extravasation of immune cells into the parenchyma.

Despite the barriers in place to protect the brain, it is now well understood that immune cells do enter the brain, and activated T cells have been shown to cross intact BBB.21 Additionally, in GBM, the possibility exists that damage to the BBB may prove useful and may be harnessed to promote a strengthened immune response at the tumor site.

2. How Cells Are Activated to Neuroantigens

The classic paradigm of specific immune activation is achieved through antigen uptake by antigen-presenting cells (APCs), which migrate to the lymph nodes via draining lymphatics where APCs subsequently activate T cells. In the systemic immune system, DCs are considered to be the most potent APCs. In the CNS, a variety of cell populations have been postulated as primary CNS APCs, including vascular endothelial cells, smooth muscle cells, astrocytes, perivascular macrophages, choroid plexus epithelial cells, neurons, and DCs.22 Although microglia have also been proposed to be the primary resident APCs in the CNS, their capability for stimulating cellular immune responses is controversial.2332 CD11c+ cells resembling classical bone marrow-derived DCs appear to be involved in murine infectious diseases33 and autoimmune encephalitis.34 However, in the glioma microenvironment, infiltrating microglia/macrophages fail to upregulate MHC class II in response to IFN-γ.35 Recent work focusing on possible CNS APCs examined microglia, macrophages, and DCs (F4/80-CD45hiCD11c+) isolated from the peak of acute-phase EAE in mice. These cells were subsequently co-cultured with myelin proteolipid protein (PLP)139–151-specific naive CD4+ transgenic (Tg+) T cells in the presence or absence of exogenous PLP139–151 peptide. All APCs with exogenously loaded peptides induced proliferation and IL-2 production from the T cells, with DCs being the most potent. However, with only endogenously processed peptide (no added peptide), microglia failed to induce proliferation. Macrophages induce proliferation only at very high APC:T-cell ratios, and only CNS DCs induced both proliferation and IL-2 production from the CD4+ Tg+ T cells,36 demonstrating weak capabilities of microglia and macrophages as the primary CNS APCs. A recent study by Miller and colleagues37 has further demonstrated that plasmacytoid DCs (pDCs), the major population of CNS DCs in EAE, seem to have a regulatory (rather than an activating) role for T cells. In fact, depletion of pDCs during different phases of EAE leads to increased disease severity and enhanced CNS CD4+ T-cell activation.37 Thus, distinguishing between the myeloid DC and pDC role in T-cell activation is crucial.

It should be noted that antigen presentation has been hypothesized to occur through multiple mechanisms.7 Hypothesized mechanisms include the following: (1) DC uptake of antigen within the CNS, which is followed by migration and antigen presentation at lymph nodes. (2) Alternatively, antigen could drain to lymph nodes where they are then taken up by DCs processed and presented. (3) Cells that express the antigen may directly drain to lymph nodes and present their own antigen (direct presentation as opposed to cross presentation by DCs). Further studies are warranted to fully elucidate mechanisms by which cells and antigens leave the CNS and trigger systemic immune responses.

As mentioned previously, intratumorally injected DCs migrate to the cervical lymph nodes (CLNs). Furthermore, Cserr et al. have shown that, of radioactive-labeled antigen injected into various parts of rabbit brain, 18%–47% could be identified in the CLNs (as reviewed in Ref. 22). In additional studies on the progression of demyelinating brain diseases, autoantigens from brain lesions have been shown to drain to CLNs in both primate models of EAE and in human MS.38 Notably, we and others have suggested that tumor-specific T cells can be primed in CLNs in murine GBM models.3942 Surprisingly, Cserr and colleagues41 demonstrated that ovalbumin injected into the CNS parenchyma resulted in a greater humoral response than extracerebral injections of the same antigen in locations that also drain to CLNs. Presumably, there is an extra processing system or other specific drainage mechanisms in the CNS, which lead to an effective immune response.

The mechanism through which these antigens or DCs traffic to CLNs in humans is still very much unknown. In rabbit models, it has been shown by Cserr and others that the pathway of intracerebrally injected antigen is through sub-arachnoid space, along cranial nerves, crossing the cribiform plate entering the nasal mucosa and finally draining into the lymphatic system.22 While this is a mechanism in rodent models such as rabbits, the human system may be drastically different. CSF drains through the nasal mucosa in both rodents and humans. However, human CSF drains directly into venous blood through arachnoid villi and granulations, whereas in rabbits, over 50% drains into lymph nodes.43 Although less understood, antigens may also travel via a pathway similar to interstitial fluid (ISF) clearance, a pathway unique to that of CSF. Not surprisingly, CNS ISF drainage also follows a mechanism distinct from ISF drainage from the periphery, and only 10%–15% of ISF gets transported through CSF in humans. Following endogenously produced amyloid-B, which is derived from the brain of patients with Alzheimer’s disease, a mechanism could be found in humans similar to that in mice.43

It is important to note that these experiments only represent one pathway for APC activation and that alternative pathways may exist. Further investigation is necessary in order to fully understand how antigens reach APCs and finally activate T cells.

3. T-Cell Trafficking to the Brain

As discussed earlier in this section, it is not well understood how lymphocytes traffic to the glioma sites. However, one model explains how lymphocytes translocate from high-velocity circulation into the parenchyma. This is a “multistep” model, characterized by 4 stages: (1) tethering/rolling, (2) activation, (3) adhesion, and (4) transmigration.14,44 Briefly, interactions between the carbohydrates on leukocytes and adhesion molecules on endothelial cells (usually selectins) slow down the leukocytes to a rolling phase along the endothelial walls. At a reduced velocity, the leukocytes sense chemokines on the endothelial cells, and through G-protein signaling, become activated, upregulating integrins such as very late antigen (VLA)-4.45 Lymphocyte function-associated molecule (LFA) on lymphocytes allows for a stable interaction with their ligands VCAM and ICAM-1, respectively, on endothelial cells. Finally, with this tight interaction in place, cells may transmigrate into the parenchyma.

Expression of tissue-specific homing molecules directs antigen-experienced T cells to particular peripheral tissues. It is therefore essential to gain understanding of pivotal homing receptors that dictate CNS tumor homing of T cells. Calzascia et al.46 defined in vivo imprinting of distinct homing phenotypes of T cells responding to antigens expressed by tumors in intracerebral, subcutaneous, and intraperitoneal sites with efficient VLA-4-mediated brain-tropism of CD8 T cells cross-primed in CLNs. Interestingly, multiple and distinct imprinting programs could occur simultaneously in the same LN when tumors were present in more than one site. Their study supports the concept that this critical functional parameter is dictated upstream of CLNs at the site of antigen capture by cross-presenting APCs; but the identity of the LN is not paramount in determining the homing phenotype.

In our own studies with murine brain tumors, we have demonstrated that type-1 cytotoxic T lymphocytes (Tc1), but not the type-2 counterpart (Tc2), can efficiently traffic to the CNS tumor site.47 VLA-4 and CXCR3, a chemokine receptor for type-1 chemokines CXCL9–11, were uniquely upregulated on Tc1 compared with Tc2, and these receptors are critical for efficient CNS tumor homing of Tc1.45,47,48 On the basis of these original data, our current translational efforts are directed towards the development of vaccine strategies that promote expression of VLA-4 and CXCR3 on vaccine-induced effector T cells. Further information on mechanisms underlying efficient CNS tumor homing of T cells should be gained for the development of truly effective immunotherapy strategies for CNS tumors, including GBMs.

B. Interaction between the Immune System and Gliomas

Although GBMs are potentially immunogenic tumors,4951 spontaneous clearance of established tumors by endogenous immune mechanisms is negligible. As is the case in other cancers, gliomas develop diverse strategies that escape tumor-specific immunity. A comprehensive overview of glioma development and progression suggests that the process is influenced by intrinsic properties of the glioma cells, as well as by microenvironmental factors. The glioma microenvironment is composed of tumor cells, endothelial cells, intermingling glia, neurons, a variety of leukocyte subsets, extracellular matrix fibers, and soluble mediators. An intricate interplay between the cellular and extracellular components determines the success or failure of tumor progression.5254 This section outlines the role of different elements and the reciprocal interactions that exist between them, and sheds light on their potential involvement in glioma development and progression.

1. Impaired Immune Function in Patients Harboring Malignant Gliomas

Previously characterized impairments in immunity of glioma patients have included low peripheral lymphocyte counts, reduced delayed-type hyper-sensitivity reactions to recall antigens, and impaired mitogen-induced blastogenic responses by peripheral blood mononuclear cells (PBMCs).5564 The modulation and downregulation of T-cell function by immunosuppressive cytokines such as transforming growth factor-β (TGF-β) and interleukin 10 (IL-10) elaborated by malignant gliomas has been previously characterized.65 TGF-β and IL-10 are known potent inhibitors of macrophage function66 and of microglia production of proinflammatory cytokines.27,67,68 One of the most important prerequisites for the immune activation of effector T-cell responses is the presence of functional APCs that process and present tumor antigens, secrete specific cytokines and chemokines, and abundantly express costimulatory molecules essential for initiating and/or propagating tumor-directed T cells.69

2. Glioma-Derived Immunosuppressive Factors

a. Soluble Immunosuppressive Factors Derived from Gliomas

Gliomas have been shown to synthesize and secrete multiple factors that are capable of inhibiting T-cell responsiveness. These include TGF-β1, −2, and −37081; PGE277,82,83; IL-108487; and gangliosides (GANGs).8890

Transforming Growth Factor (TGF)-β

The immunosuppressive effects of TGF-β are multiple and complex. They include the inhibition of maturation and antigen presentation by DCs or other APCs, inhibition of T-cell activation, and differentiation towards effector cells (either cytotoxic cells expressing perforin or Th1 or Th2 cells).9194 Inhibiting TGF-β in vivo in experimental tumor models has produced mixed results, probably because the effects of this cytokine are not only on the immune system, but also on the tumor cell, which may be protected from Fas (CD95)-mediated apoptosis in some circumstances.95,96 A recent study with glioma-cell clones that are resistant to the cytotoxic effects of allogeneic CTLs shows that TGF-β, but none of the other immunomodulatory molecules examined, is upregulated in these resistant glioma cells, pointing to the significance of TGF-β in glioma immune-escape mechanisms.97

Prostaglandin (PG)E2

PGE2 is a product of arachadonic acid metabolism. It is produced at sites of inflammation or tissue damage, where it exerts many effects, including the enhancement of vascular permeability. PGE2 has been shown to have profound modulatory effects on T-cell activation, and gliomas synthesize PGE2. Thus, PGE2 could be associated with the suppressed T-cell function observed in patients with gliomas.77,82,83,98100 However, the concentration of PGE2 observed in glioma-cell supernatants (GCS) does not correlate with the concentration of PGE required to inhibit T-cell proliferation.83 Moreover, the T-cell unresponsiveness caused by the addition of GCS to mitogen-stimulated T-cell cultures is not altered when PGE2 inhibitors, such as naproxen or ibuprofin, are used during the generation of GCS.100 Further characterization of the roles of PGE2 in glioma-induced immunosuppression is warranted.

Interleukin (IL)-10

The expression of IL-10 mRNA increases significantly with tumor grade; 87.5% of WHO grades III–IV astrocytomas expressed IL-10 mRNA versus 4% of grade II astrocytomas.86 Gliomas exhibiting a high degree of brain invasiveness express IL-10 at higher levels than more localized gliomas.84 In addition, glioma patient-derived monocytes, but not normal donor monocytes, produce IL-10 after in vitro stimulation with T-independent B-cell mitogen Staphylococcus aureus Cowen I strain (SAC).101

Gangliosides (GANGs)

Since GANGs are produced by gliomas and they can modulate lymphocyte responsiveness, it is interesting to speculate that immunosuppression by GANGs might account for some of the dysfunctions observed in glioma patients. The brain contains the highest concentrations of GANGs,102 which are highly immunosuppressive moieties affecting both APC and T-cell function.102107 Concentrations as low as 1 mM of all brain-derived GANGs tested cause significant inhibition of normal T-lymphocyte proliferation in vitro.102 GANGs have been shown to inhibit the expression of CD4 on human and murine T cells as well as inhibit the generation of both cytotoxic T lymphocytes (CTL) and NK-cell activity105,108,109 and induce the death of T cells.110

Macrophage Chemoattractive Protein (MCP)-1 (also CCL2)

Macrophage chemoattractive protein (MCP)-1 (also CCL2) was first purified and identified in 1989 from a human glioma.111 MCP-1 is secreted by a variety of glioma cell lines and is expressed in glioblastomas.112115 MCP-1 can directly promote angiogenesis in vitro116 and may also increase the levels of angiogenic factors in vivo through the recruitment of tumor-associated macrophages.117 MCP-1 has been shown to be critical for glioma tumor-cell proliferation118 and cancer-cell metastasis119,120 and is an important determinant of tumor aggressiveness.121124 However, the MCP-1 chemokine has multifunctional activities that appear to have opposing effects on tumor biology. MCP-1 is a potent leukocyte chemoattractant and may play an important role in the host antitumor immune response. MCP-1 has been shown to promote in vitro chemotaxis of both CD8+ and CD4+ cells.125127 Levels of tumor-derived MCP-1 often correlate with macrophage infiltration of malignant tissue. Vα24-Jβ18-invariant NK T cells have also been shown to traffic to and infiltrate neuroblastoma tumors in an MCP-1-dependent manner.128 It has also been demonstrated that MCP-1 increases the efficiency by which adoptively transferred ex vivo-expanded T cells home to sites of tumor burden.114

b. Surface Immunosuppressive Molecules Expressed on Glioma Cells
Fas-FasL

Malignant brain tumors, including GBM, express Fas, a receptor that mediates apoptotic signals upon ligation to its ligand Fas-L. Fas ligation also induces expression of proinflammatory and angiogenic mediators in malignant glioma cells,129,30 which can protect tumor cells from Fas-mediated cell death or support tumor growth by promoting angiogenesis.131 Moreover, some malignant tumors also express FasL, which induces apoptotic cell death of immune cells infiltrating into tumors and evade the host immune system, known as “counterattack.”132135 Therefore, the roles of the FasL-Fas system in malignant brain tumors are complex: first, the expression of FasL by malignant tumors is a mechanism by which these cells evade the host immune system by inducing apoptosis of Fas-positive T cells that infiltrate into the tumor tissue; and second, Fas ligation on glioma cells can provoke apoptotic and/or proliferative responses.129,131 Microenvironmental factors will certainly influence the consequences of FasL expression by tumor cells; for example, tumors co-expressing both FasL and TGF-β maybe particularly well adapted to combat CTL effector mechanisms.136,137

B7-Homologue 1 (B7-H1), Also Known As Programmed Death Ligand-1 (PD-L1)

The B7 family consists of activating and inhibitory co-stimulatory molecules that positively and negatively regulate immune responses. Recent studies have shown that human and rodent cancer cells, and stromal cells and immune cells in the cancer microenvironment, upregulate expression of inhibitory B7 molecules, and that these contribute to tumor immune evasion.

B7-homologue 1 (B7-H1), a recently identified homologue of B7.1/2 (CD80/86) and also known as programmed death ligand-1 (PD-L1), has been described to exert costimulatory and immune-regulatory functions. PD-1, a receptor for B7-H1, contains an immune receptor tyrosine-based inhibitory motif, and coligation of PD-1 and the T-cell receptor leads to rapid phosphorylation of Src homology region 2-containing protein tyrosine phosphatase-2, a phosphatase suggested to attenuate T-cell receptor signaling.138 Wintterle et al.139 investigated the expression and the functional activity of B7-H1 in human glioma cells in vitro and in vivo. They found that all 12 glioma cell lines, although lacking B7.1/2 (CD80/86), constitutively expressed B7-H1 mRNA and protein. Exposure to IFN-γ strongly enhanced B7-H1 expression. Moreover, immunohistochemical analysis of malignant glioma specimens revealed strong B7-H1 expression in all 10 samples examined, whereas no B7-H1 expression could be detected on normal brain tissues.on B7-H1 expressed on glioma cells was identified as a strong inhibitor of CD4+ as well as CD8+ T-cell activation as determined by increased cytokine production (IFN-γ, IL-2, and IL-10) and expression levels of the T-cell activation marker (CD69) in the presence of a neutralizing antibody against B7-H1 (mAb 5H1). B7-H1 expression may thus significantly influence the outcome of T-cell/tumor-cell interactions and represents a novel mechanism by which glioma cells evade immune recognition and destruction.

B7-H1 expression can be induced or maintained by many cytokines,140142 of which IFN-γ is the most potent. In light of its stimulatory effect on B7-H1 expression, IFN-γ could thus be a “double-edged sword” in tumor immunity. Whereas IFN-γ could increase antigen processing and presentation by up regulating the expression of major histocompatibility complex (MHC) molecules and components of the antigen-processing machinery,143 the effects of IFN-γ on B7-H1 expression might downregulate T-cell immunity. This could explain, at least partially, why IFN-γ has not been effective as a therapeutic agent for most human cancers.

In human glioma, loss of phosphatase and tensin homolog (PTEN) and activation of the phosphatidylinositol-3-OH kinase (PI3K) pathway induce expression of the gene encoding B7-H1 increases posttranscriptionally.144 In primary GBM tissues, levels of B7-H1 protein correlated with PTEN loss, and tumor-specific T cells lysed human glioma targets expressing wild-type PTEN more effectively than those expressing mutant PTEN, demonstrating a novel mechanism linking loss of the tumor suppressor PTEN with immunoresistance, mediated in part by B7-H1.144

Receptor-Binding Cancer Antigen Expressed on SiSo Cells (RCAS1)

Receptor-binding cancer antigen expressed on SiSo cells (RCAS1), a membrane molecule strongly expressed in aggressive human cancer cells, has also been reported to participate in tumor immune escape in certain types of cancer. It acts as a ligand for a receptor present on normal peripheral lymphocytes, such as T, B, and NKu cells, and induces apoptotic cell death.145 Nakabayashi et al.146 demonstrated that RCAS1 overexpression significantly correlates with high histological grade and poor prognosis in 57 glioma cases. Furthermore, reduced infiltration and increased apoptosis of tumor-infiltrating lymphocytes (TILs) were observed in RCAS1-positive regions. Thus, RCAS1 expression in gliomas may play roles in tumor progression and tumor immune escape.

Human Leukocyte Antigen (HLA)-G

A non-classical MHC class I molecule, HLA-G, expressed on astrocytoma cells has been proposed to have immunosuppressive potential147 and to suppress NK- and T-cell immune responses, but this is controversial.148,149 Regarding astrocytomas, a proportion of astrocytoma cell lines and tumor biopsies expressed HLA-G protein, and inhibition of CD4 and CD8 T-cell responses was demonstrated in vitro, but this was only tested after incubation of cell lines with high concentrations of IFN-γ (500 U/mL), or after gene transfer of HLA-G into glioma lines.

CD70

CD70 is a CD27 ligand and a member of the tumor necrosis factor (TNF) family. Neo-plastic glial cells often express CD70 in vivo, mediating immune escape of human malignant gliomas. 150,151 CD70-positive GBM cell lines induce apoptosis in T cells.110 Of the four GBM cell lines that induced T-cell death, three highly expressed CD70. Two nonapoptogenic GBM lines (CCF3 and U138), on the other hand, had only minimally detectable CD70 expression. Neutralization of CD70 but not TNF-α or FasL is responsible for initiating T-cell death via the receptor-dependent pathway. CD70 is also expressed on activated T and B cells, with roles in regulating immune responses via interaction with CD27 expressed on lymphoid cells.152

3. Glioma-Associated Immunosuppressive Leukocytes

A large number of observations suggest that certain types of tumor microenvironmental immune cells are not innocent bystanders at brain tumor sites, and that they actively affect tumor development and progression. Inflammatory cells, primarily macrophage/microglia and regulatory T cells, may affect these processes via their ability to express a large variety of factors, including immunoregulatory cytokines. These cytokines may be secreted not only by inflammatory cells but also by the tumor cells and stroma cells, together establishing a network of factors that significantly affects brain tumors.

Macrophages/Microglia

In the CNS, macrophage/microglial cells constitute the first line of cellular defense against a variety of stressors, participating in the regulation of innate and adaptive immune responses in human and rat gliomas.29 Many human gliomas exhibit a prominent macrophage/microglia infiltrate. Glioma-infiltrating macrophages (GIMs) can account for as much as 30% of the tumor mass.153 GIMs represent the largest sub-population infiltrating human gliomas from postoperative tissue specimens of glioma patients.154

With regard to the distinction between resident microglia (CD11b+/CD45dim) and macrophages (CD11b+/CD45high), most studies have demonstrated that glioma-infiltrating CD11b+ cells are mostly CD45high macrophages.32 Indeed, it has been described that “microglia” in human gliomas appears mostly amoeboid and morphologically distinct from the resting microglia present in the intact brain.155,156

Intratumoral macrophage/microglia density is higher than in the normal brain, and the abundance of microglia correlates with the grade of malignancy. In patients with gliomas, the number of macrophages in glioblastomas (grade IV) is higher than that in grades II or III gliomas, and it is closely correlated with vascular density in the tumors.157,158

It is postulated that the defense functions of macrophage/microglia against glioma are compromised in the tumor microenvironment. These GIMs expressed substantial levels of Toll-like receptors (TLRs), which are critical components for APCs to mediate innate immune responses to any infectious or traumatic challenge and activate adaptive immune responses. However, GIMs did not appear to be stimulated to produce proinflammatory cytokines (TNF-α, IL-1, or IL-6), and in vitro, lipopolysaccharides could bind TLR-4 but could not induce GIM-mediated T-cell proliferation.157 Moreover, it has been found that these GIMs, in addition to decreased surface expression of MHC class II,23,35 lack expression of the costimulatory molecules CD86, CD80, and CD40, which are critical for T-cell activation. They are thereby unable to activate T cells properly ex vivo.154 Macrophages/microglia can release many factors, including extracellular matrix proteases and cytokines, which may directly or indirectly influence tumor migration/invasiveness and proliferation.159161 In Boyden chamber assays, glioma cell migration is stimulated by the presence of macrophage/microglia or macrophage/microglia-conditioned medium.162 It has been recently demonstrated in an organotypic brain culture that the invasive potential of glioblastoma was lower in macrophage/microglia-depleted slices, but the addition of microglial cells to the microglia-depleted slices restored the invasiveness.163 These findings suggest that macrophage/microglia in human gliomas may promote and support the invasive phenotype of these tumors.

Regulatory T Cells (Tregs)

The suppressive activity of regulatory T cells (Tregs) has been implicated as an important factor limiting immune-mediated destruction of tumor cells. An increased FoxP3+ Treg to CD4+ T cells ratio correlates with impairment of CD4+ T-cell proliferation in peripheral blood specimens obtained from patients with GBM.164 In this referenced study, in vivo depletion of Tregs led to glioma rejection in murine model systems. Other studies have shown that an immunosuppressive population of Tregs is present within the GBM microenvironment.154,165 Moreover, it has been demonstrated that Tregs are not present in normal brain tissue and were very rarely found in low-grade gliomas and oligodendrogliomas.166 The same study also observed that Treg infiltration differed significantly in the tumors according to lineage, pathology, and grade. Tregs seemed to have the highest predilection for gliomas of the astrocytic lineage (over oligodendroglioma) and specifically in the high-grade gliomas, such as GBM. In both univariate and multivariate analyses, the presence of Tregs in GBMs seemed to be prognostically neutral.166 However, in a study with a mouse GL261 glioma model,167 treatment of glioma-bearing mice with anti-CD25 mAb delayed the tumor growth and prolonged the survival of the mice, suggesting that CD4+CD25+ Treg cells play an important role in suppressing the immune response to CNS tumors.168

Furthermore, Grauer et al.169 demonstrated a time-dependent accumulation of CD4+FoxP3+ Tregs in brain tumors with a syngeneic murine glioma GL261 model. They observed that the expression of CD25, CTLA-4, GITR, and CXCR4 on intratumoral CD4+FoxP3+ Treg during tumor growth is upregulated in a time-dependent manner. They also demonstrate that treatment with anti-CD25 MAbs significantly provokes a CD4 and CD8 T-cell-dependent destruction of glioma cells. Moreover, combining Treg depletion with administration of blocking CTLA-4 mAbs further boosted glioma-specific CD4+ and CD8+ effector T cells, as well as antiglioma IgG2a antibody titers, resulting in complete tumor eradication. This study illustrated that intratumoral accumulation and activation of CD4+FoxP3+ Tregs act as a dominant immune-escape mechanism for gliomas and underline the importance of controlling tumor-infiltrating Tregs in glioma immunotherapy.169

III. IMMUNOTHERAPY APPROACHES FOR GLIOMAS

A. Passive Immunotherapy

Passive immunotherapy consists of therapeutic modalities that utilize immune-effector cells and/or a variety of molecules, including MAbs and cytokines.

The efficient use of MAbs against brain tumors presents unique challenges. First, the passage of therapeutic agents from the circulation through the BBB favors small, uncharged, lipid-soluble molecules. The uniquely large size of antibodies as macromolecules favors novel delivery strategies to administer antibodies directly to the brain tumors. Indeed, an IgG antibody has a molecular weight of approximately 150 kDa, whereas many chemo-therapeutic agents have a molecular weight on the order of 1 kDa.170 Although most small molecule drugs rely on diffusion as a mode of transport through tissue, antibodies must rely on bulk fluid flow (convection) in which antibodies flow down their pressure gradient. It has been shown that interstitial pressure in a solid tumor mass is elevated above the interstitial pressure of the surrounding normal tissue.170172 This increased pressure maybe the result of a less-developed lymphatic system, which is thereby less able to drain interstitial fluid. Also, increased cell density could be a contributing factor to this phenomenon. 171173

The binding site barrier is another challenge to antibody penetration. At equilibrium, the concentration of antibodies in normal tissue is negligible compared with the interface at the tumor site.170 For transport into the core of a tumor mass, free antibody must pass from the low concentration region of target-free normal tissue, past a boundary layer of the tumor (the binding site). This boundary layer contains a high concentration of tumor-bound antibody. To diffuse into the tumor core, the antibody would have to flow against a significant concentration gradient and would do so inefficiently, leaving the interior of a solid tumor ineffectively treated. Because of the limitations presented by the binding site barrier and the penetrance of large molecule therapeutics through brain tissue, it is an attractive option to deliver drugs in concert with a complete surgical resection—an integral part of the current standard of care.

Convection Enhanced Delivery (CED) has emerged as an attractive drug-delivery method that may be instrumental to the development of novel therapeutic modalities. CED is suitable for enhancing the delivery of both large and small molecules by physical-pressure processes. In CED, the therapeutic agent is infused at high pressure through an intracranial catheter.174 As opposed to diffusion, drug delivery using CED relies on bulk flow. A number of passive immunotherapy clinical trials have utilized CED as the method of drug delivery, many of which are discussed in the following sections. However, a major challenge with CED remains in that the bulk flow produced may be slow and insufficient to deliver IgG-based molecules throughout a typical treatment volume.174,175 Other delivery techniques, including intraventricular infusion, intratumoral and intracavitary injections, and biodegradable polymers (such as BCNU Gliadel wafers), rely on diffusion.175

1. Antibodies: Inhibition Strategies

Anti-EGFR Antibodies

An attractive candidate for IgG-based inhibition has been epidermal growth factor receptor (EGFR). EGFR is overexpressed on 40%–50% of tumors.111 EGFR overexpression is associated with increased tumor growth rate and shorter survival.176,177 EGFR, a transmembrane receptor tyrosine kinase, binds its ligands epidermal growth factor (EGF) and transforming growth factor TGF-α. EGFR activation generates gene transcription modulations, resulting in stimulated proliferation, angiogenesis, and metastasis.178 A mutation of EGFR, termed EGFR variant III (EGFRvIII), is frequently expressed in glioblastoma and enhances tumorigenicity.179

Cetuximab (Imclone, Bristol Meyers Squibb, New York, NY) has been shown to enhance the antitumor effects of chemotherapy and radiotherapy, inhibiting the EGFR pathway. EGFRvIII can also be bound by cetuximab, and it has been suggested that cetuximab has antitumor efficacy against EGFRvIII+ glioma cells.180 Cetuximab targets the extracellular domain (ECD) of EGFR.181 In 2006, Belda-Iniesta et al.181 reported three patients with recurrent EGFR-expressing GBM who responded to treatment with a single agent, cetuximab.

Nimotuzumab (h-R3, YM Biosciences, Mississauga, ON, Canada), which similarly targets the ECD of EGFR, has been used in Phase II trials.182 Ramos et al.182 observed a 17.5-month MST for GBM patients. In high-grade glioma patients, there was a 37.9% objective-response rate and stable disease in another 41.4% of patients.

A variety of other anti-EGFR antibodies, including Panitumumab, Matuzumab, and Zalutumumab, have been applied to other EGFR+ cancers (such as colorectal cancer) but have not been applied to malignant gliomas in the clinical setting at this time, to the best of our knowledge.111

Daclizumab (Zenapax, Roche, Basel, Switzerland), a MAb against IL-2R, is primarily an immunosuppressant but may inhibit the recovery of Tregs after chemotherapy-induced lymphocytopenia. Ongoing clinical trials are investigating daclizumab along with peptide/autologous lymphocyte treatment following temozolomide therapy (NCT00626015 on www.clinicaltrials.gov).

The use of antibodies as inhibitors has the advantage of being precise and specific, but the challenges of delivery and penetrance remain. As cancer biology and the mechanisms of tumor-cell proliferation and immune escape are increasingly understood, more candidates for therapeutic inhibition will be found.

2. Radioimmunotherapy

Radionucleotides conjugated to MAbs have tremendous potential for both diagnostic imaging and for targeted radiotherapy. The paradigm of radioimmunotherapy (RIT) is the targeting of tumor-associated antigens by a radioisotope-conjugated antibody, which binds at the tumor site and delivers a local, lethal dose of radioactivity to the tumor site. Cytotoxicity from radiation is the result of the ionization of an emitted particle from a disintegration reaction as it deposits energy into the cell, irreparably damaging DNA.170 It is advantageous to concentrate radioactivity at the tumor site, thus sparing normal brain tissue.183 There has been less success with bulky solid tumors, and this approach is ideally used in combination with gross total resection, with minimum residual disease. Delivery can be focused within the surgical cavity in efforts to access the tumor cells invading at the surgical margin.183

Dosimetry of RIT is challenging because the absorbed radioactive dose is dependent on many factors, including the tissue geometry and properties, and the radiopharmaceutical clearance rate of the individual glioma patient. Many algorithms are based on absorbed dose calculated by the MIRD schema, developed by the Medical Internal Radiation Dose Committee.170 This schema employs a series of three-dimensional Monte Carlo calculations applied to idealized models of tissue properties specific to the choice of radioisotope and target organ.170 Calculations of the biological effective dose (BED) have since become more advanced, taking into account the clearance rate, interaction between absorbtion in the cortex and medulla of the kidney (S value), and red marrow dosimetry.184

Two common types of radionucleotides used in RIT emit α particles, helium nuclei, or β-particles, expelled from the nucleus with the mass of an electron. There exists an inherent tradeoff in a selection of either an α-emitting radionucleotide such as 211At or a β-emitting radionucleotide such as 131I. Although the range of most β-emitters is on the order of 1 mm, many α-emitters have a range of just a few cell diameters, with a higher linear energy transfer.183 This means that the α particles released from a disintegration reaction travel a shorter distance but deposit energy more intensely along their path. This implies greater cytotoxicity and specificity and therefore a higher allowable dose to the tumor site. However, given the heterogeneity of malignant glioma, the “cross-fire” effect of longer range β-emitters may be advantageous to kill antigen-negative tumor cells. This heterogeneity also implies that a multiantigen approach may be more effective than a single-target approach.

Halogens, which are β-emitters, can be directly attached to antibodies. This is an advantage over some other radionucleotides, including 111In and 90Y, which must be linked through intermediate agents. In light of the relatively short half-lives of many of these radioisotopes, speed and simplicity of generation are essential to on-site or near-site production.

Antitenascin

Tenascinc has been perhaps the most-studied target for glioma RIT. Tenascin is a glycoprotein expressed ubiquitously on the surface of several types of tumors, including high-grade gliomas. Tenascin expression may be correlated with disease progression, with 90% of GBMs exhibiting high levels of tenascin.185

The antitenascin antibody 81C6 was developed at Duke University and has been shown to uptake in tumor tissue with high specificity.186,187 MAb 81C6 has since been used extensively in RIT studies, typically conjugated to 131I, and has shown improved survival of glioma patients over historical controls as well as a reduction in toxicity relative to stereotactic radiotherapy.188 Trials of 131I-81C6 administered to the tumor-resection cavity at the time of surgery demonstrated median survival time (MST) of 12–13 months for patients with recurrent gliomas and up to 20 months in newly diagnosed GBM patients.189192 On the basis of these studies, an optimal radiation dose of 44 Gy was chosen to balance the risk of radiation necrosis and tumor recurrence.185

The group at Duke University has also developed a 211At-labeled chimeric antitenascin therapy. The choice of an α-emitting radioisotope is thought to allow for a much greater specificity for the delivery of radiation of a tumor. Indeed, in a Phase I trial by Zalutsky et al.,185 more than 95% of the radiation was localized to the tumor resection cavity. While maintaining a 0.01 Gy exposure to normal tissue (including tenascin + liver and spleen), an average radiation dose of 2986 Gy was able to be delivered to the resection site.185 In addition, by replacing murine 81C6 with chimeric ch81C6, considerably higher in vivo stability can be achieved.185 No dose-limiting toxicity has been reported.193 The overall median survival was 57 weeks, and MST for GBM patients was 52 weeks; however, 2 of 14 patients survived for almost 3 years.193

Tumor Pretargeting

Another group from the European Institute of Oncology has used a different antibody to tenascin, BC-2 and BC-4.194 With 131I-BC-2/4, Riva et al.195 observed no significant toxicity, a MST of 16 months, and 3 of 17 patients with complete remissions.195 A follow-up report combining data from 111 patients in a Phase I/II trial showed a MST of 20 months.196

Paganelli and colleagues197,198 have developed the method of tumor pretargeting using BC-4 MAb against tenascin. In this three-step targeting approach, high-affinity avidin-biotin binding is exploited. Biotinylated antibody is injected into the patient, and on the basis of kinetic studies, unbound antibody clears the body within 48 hours.197,198 During the second step, streptavidin is injected. Biotinylated radionucleotides are administered several hours later, as the third and final step.197 The biotinylated radioisotopes bind to the equilibrated antibody-avidin complex that is theoretically bound specifically to the tumor. Since the biotinylated radioisotope is much smaller than the bulky IgG molecule that binds directly to the tumor site, this smaller effector molecule equilibrates and migrates to the tumor site much faster than directly conjugated IgG, increasing the possible radioactive dose to the tumor site without increasing the normal tissue dose. In a clinical trial utilizing 90Y and the three-step biotin-avidin method administered intravenously, 12 of 48 patients demonstrated a reduction in tumor (17% maintained reduction after 1 year), another 52% had stable disease, and three experienced complete remissions.199 In a subsequent study applying the same strategy, 37 patients with grades III/IV glioma were treated. Patients with grade III tumors had a reported progression-free survival (PFS) of 56 months and 82% (9/11) survival at 60 months. Thirty-eight percent of GBM patients survived with a MST of 33.5 months compared with 0% survival of control patients (MST = 8.5 months) treated with the standard care.197

In later studies, pretargeted RIT was delivered “locoregionally” by direct infusion into the surgically created cavity placed during a second surgery in recurrent GBM patients. The average absorbed dose to normal brain was minimal. AA patients had a MST of 19 months from the second surgery (53 months overall). GBM patients had a MST of 11.5 months from the second surgery (20 months overall). Forty-four percent of GBM patients had significant delay in tumor growth, prolonged survival, and improvement in quality of life.200 Pretargeted RIT was also examined in combination with chemotherapy (temozolomide, TMZ). Patients with recurrent GBM were once again treated with a second surgery to administer pretargeted 90Y-antitenascin every 2 months, with or without TMZ. Patients receiving RIT alone demonstrated a 5-month PFS and 17.5-month survival compared with patients who received RIT and TMZ who demonstrated a 10-month PFS and 25-month MST. No increased neurological toxicity was seen in patients treated with combination therapy versus monotherapy. A 75% objective-response rate was reported.201

DNA/Histone HI Complex

Phase I and II clinical trials have been completed with 131I-chTNT-1/B MAb (Cotara, Peregrine Pharmaceuticals, Tustin, CA). 131I-chTNT-1/B MAb is a radioiodine-conjugated MAb specific to the DNA/histone HI complex.202 This intracellular marker is only exposed to the interstitial space in the necrotic core of the tumor, where a population of tumor cells spontaneously dies and releases DNA/histone complex. The use of the β-emitting 131I radionuclide is appropriate to generate a significant amount of midrange cytotoxicity and also affects the invasive glioma cells that are not marker specific, but with a reasonable radiation range cutoff to provide an extent of protection to the surrounding normal tissue.203 In clinical trials, 131I-chTNT-1/B MAb has been administered via CED to achieve maximal tumor penetrance.202 It was reported that the majority of patients received infusions within 10% of the prescribed radioactive dose to the targeted region, and the therapy was well tolerated.202 These trials demonstrated safety and feasibility of 131I-chTNT-1/B MAb administered via CED. Data is insufficient to determine efficacy, but in 28 patients with recurrent GBM, the median time to progression and MST were 8.4 and 23 weeks, respectively, which is in line with historical controls.202 However, in a “normalized” subset of patients receiving total radioactive dose in the therapeutic window, MST was reported as 37.9 weeks (203).202 Several patients demonstrated objective responses, and 25% of recurrent GBM patients survived for greater than 12 months.203

Other Targets

Other studies have targeted the extra domain B (ED-B) of fibronectin, a marker of tumor angiogenesis204,205; the α chain of the IL-2 receptor206; and EGFR207 in the preclinical setting. Rosenkranz et al.207 have developed a 211At-conjugated internalizable ligand to EGFR. The application of 125I-anti-EGFRMAb following surgical resection and external beam radiotherapy (EBRT) was reported to demonstrate significant improvement in survival, although this study did not include a control group.208 In a small sample of patients, simultaneous 125I-anti-EGFR MAb and EBRT failed to improve survival or PFS compared with EBRT alone.209 Alternative radionucleotides, including 225Ac, 213Bi, 212Bi, 224Ra, 212Pb, and 149Tb have been investigated preclinically, as well.210,211 The chemistry of chelating radionucleotides is yet to be fully elucidated. For example, 225Ac undergoes four disintegration reactions with intermediates 221Fr, 217At, and 213Bi emitting α particles during each event.210 To develop one chelating agent to bind all intermediates of multistep disintegrations to IgG would not be feasible, and the clinical consequences of the release of intermediates has not been demonstrated.210

3. Boron Neutron Capture Therapy

Boron Neutron Capture Therapy (BNCT) is a strategy in which an agent delivers stable isotope boron-10 selectively to the tumor site. Upon exposure to an external epithermal neutron beam, the boron disintegrates into stable lithium and emits an α particle with a short path length (9 μm) and high-linear-energy transfer.212 A variety of nonimmunological boron carriers have been used with encouraging results in clinical trials. Most notably, recent clinical trials using boronophenylalanine (BPA) or sodium borocaptate (BSH) as boron carriers have reported promising results, with MST of 13–20 months.212218

The potential to use novel targeting methods, including the use of MAbs, could dramatically improve the specificity and efficacy of this therapy.212 In a preclinical study, Yang et al.219 explored the use of a mixture of boronated antibodies targeted against EGFR (cetuximab) and EGFRvIII (L8A4) to treat composite F98EGFR/F98EGFRvIII tumors in rats. It was observed that the use of a combination of targets was significantly more effective than the use of either boronated antibody alone.219 EGFR is amplified in 57% of GBMs, and EGFRvIII is amplified in 36% of GBMs.220 On the basis of this heterogeneity, it is likely that BNCT will be most effective clinically, using a combination of specific boron carriers and also in combination with other therapeutic modalities.

4. Coupled Targeted Toxins

A number of cytokine receptors have been observed to be upregulated in glioma, including IL-4R and IL-13Rα2. By harnessing these and other receptor-ligand interactions that are up-regulated in GBM, targeting of tumor cells can be achieved, and by fusing cytokines with toxins, selective cytotoxicity can be achieved.221,222

IL4-PE

Interleukin-4 receptors (IL-4R) are upregulated in glioma cells relative to normal tissue. In fact, 83% of GBM tumors and 86% of astrocytoma tumors were found to be moderately to highly positive for IL-4R in situ by Joshi et al.223 Puri et al.224 have developed a chimeric fusion protein, including domains of IL-4 and Pseudomonas exotoxin (PE), which is produced by expressing chimeric gen in E. coli and purifying the protein using inclusion bodies. Phase I studies have been executed to determine safety and tolerability.225,226 It was also reported that in one preliminary study, 6 out of 9 patients with recurrent malignant glioma demonstrated tumor necrosis after receiving IL4-PE via CED with multiple catheters, as evidenced by gadolinium-enhanced MR images. One patient remained disease free for greater than 18 months.227

IL13-PE

Interleukin-13 receptor (IL-13R) is also found to be overexpressed on a majority of glioma cell lines and resected GBM specimens.228 A mutated form of PE fused to human IL-13, named IL-13PE38QQR. or cintredekin besudotox (CB), has been developed and has been shown to effect specific cytotoxicity on glioma cell lines.228,229 CB has been studied in a number of Phase I clinical trials to investigate dosimetry and catheter positioning and is reportedly more active against glioma cell lines than IL-4-targeted toxins in vitro.229 Among Phase I trials, a total of 51 GBM patients have been treated with CB, with a MST of 42.7 weeks (55.6 weeks for the subset of patients with two or more catheters for drug delivery).230 A Phase III study compared the efficacy of CB to Gliadel wafers. PFS was longer (17.7 versus 11.4 weeks) in patients treated with CB, but there was no significant difference in the primary end point, MST, between BC and Gliadel wafers.229 Kioi et al.231 developed a strategy in which PE is conjugated to a smaller scFv anti-IL13R human antibody. Overall, IL-13-based toxins have the potential to be utilized as an adjuvant therapy for malignant glioma pending further positive clinical studies.

Transferrin-Diphtheria Toxin

Another candidate for targeted toxin therapy has been transferrin (Tf). The receptors of this molecule (TfR) are upregulated in GBM and other rapidly dividing cells.232 Tf-CRM107 is a conjugate of human transferrin and modified diphtheria toxin (DT), which inhibits protein synthesis. The subunit of DT responsible for toxin binding is mutated to generate a 200,000-fold specificity for tumor cells, while maintaining the function of the toxin.233 A Phase I study was executed to determine safety and dosing as a primary objective for Tf-CRM107 treatment of patients with progressive glioma after receiving standard care. During this trial, a greater than 50% reduction in tumor size by MRI was achieved in 9 of 15 (60%) of evaluable patients, and a MST of 58.8 weeks was observed.233 A multicenter Phase II study followed, and 44 patients with HGG who failed to respond to standard therapy were treated with 1–2 infusions of Tf-CRM107. Complete or partial responses were seen in 35% of patients, and the MST was 37 weeks.226 Phase III studies of Tf-CRM107 are ongoing (NCT0083447, NCT00088400, NCT87230).

TGFα-PE

As mentioned previously, EGFR is an overexpressed surface molecule in GBM, making it an attractive target for specific therapies. TP-38 (IVAX, Inc., Miami, FL) is a composite of the EGFR-binding ligand TGF-α and a form of PE modified to eliminate nonspecific binding. Twenty patients (19 with HGG) were treated with 1–4 μg of TP-38. The overall MST was 23 weeks after TP-38 administration. The maximum tolerated dose was not reached.234 One patient was enrolled after a variety of chemotherapeutic treatments failed to stabilize disease. She has had a remarkable response and has survived greater than 5 years from her initial diagnosis of GBM.235

B. Adoptive T-Cell Therapy

Adoptive transfer of tumor-reactive autologous CTLs may hold promise as an attractive future immunotherapeutic intervention against malignant glioma. The earlier form of this therapeutic approach was mostly used to treat malignant melanoma, in which autologous lymphocytes infiltrating tumor nodules were first isolated and expanded in vitro in the presence of IL-2 and subsequently returned to the patients.236 In glioma, ex vivo-activated lymphokine-activated killer (LAK) cells have been applied as an adjunct to surgery, often in combination with low-dose IL-2.237242 Although clinical responses have been observed in some cases, this approach relies upon innate immune effector cells (i.e., LAK cells), whose killing activity may not be tumor specific. In contrast, antigen-specific CD8+ CTLs survey the CNS parenchyma and selectively kill astrocytes that express a model antigen hemagglutinin (HA) without collaterally damaging neurons and oligodendrocytes or myelin. This was demonstrated in an elegant study using HA-specific T cells obtained from a TCR transgenic mouse line and recipient transgenic mice expressing HA in their astrocytes.243 Importantly, i.v.-injected tumor-specific CTLs have established their anti-tumor potency in syngeneic rodent models of glioma.244 Antigen-nonspecific LAK cells, in contrast, fail to eradicate tumors in most of these experimental models.245 Collectively, these studies demonstrate that CTLs have the capacity to migrate into brain parenchyma.

Since then, the approach has been vastly improved by the use of recent advances in several areas of human T-cell biology, including in vitro human T-cell culture and ex vivo genetic manipulation. Although adoptive T-cell therapy remains the experimental therapy for limited types of cancers (mainly malignant melanoma), there have been increasing attempts to widen the use of adoptive T-cell therapy to treat other types of tumors, including malignant glioma. This section focuses on recent technological advances in adoptive T-cell therapy as well as current and future adoptive T-cell therapy for malignant glioma, with an emphasis on recent perspectives from human studies because there are several excellent recent reviews on this subject246249 that dedicate large sections to detailed discussions of adoptive T-cell therapy in various mouse tumor models.

1. Fundamentals of Adoptive T-Cell Therapy

Contrary to an active or direct immunization approach, adoptive T-cell therapy involves passive infusion or transfer of autologous CTLs specific for tumor antigens to the host. There are at least two sources of the tumor-specific CTLs. One is PBMCs from the patient. These cells can be expanded in vitro through multiple cycles of antigenic stimulation, and, subsequently, cells with a monoclonal specificity to the particular tumor-associated antigen (TAA) will be generated. In theory, infusion of a clonal population of the TAA-specific CD8+ T cell to the patient results in the accumulation of TAA-specific CD8+ T cells to tumor nodules where they recognize TAA and begin exerting cytotoxic functions on tumor cells. Evidence supporting the effectiveness of this approach came from early studies of adoptive therapy using MART-1, melanoma-associated antigen specific CD8+ T cells.250 After infusion, in vitro-expanded MART-1-specific CD8+ T-cell clones from a patient with metastatic melanoma localized to melanoma and normal skin tissues where substantial destruction of melanocytes and other cells expressing MART-1 were observed. The later study using low-dose IL-2 administration, in addition to infusion of MART-1 and gp100-specific CTL clones, for refractory, metastatic melanoma showed that low-dose administration of IL-2 enhanced infiltration and localization of MART-1 and gp100-specific CD8+ T-cell clones preferentially to the tumor tissues and had favorable antitumor responses in 8 out of 10 patients out to 21 months follow-up.251

Another important source of the TAA-specific CTLs is the tumor nodule. The tumor nodule typically contains tissue-infiltrating TAA-specific CTLs that could be first polyclonally expanded in ex vivo tissue culture in the presence of IL-2 and later selected for the TAA specificity.247 The use of ex vivo-expanded TILs has been central to adoptive T-cell therapy for malignant melanoma. Infusion of these ex vivo-expanded TILs to treat an advanced stage melanoma initially met challenges, such as low response rates associated with poor persistence of infused TILs in patients.252 However, the later modification, lymphodepletion prior to the infusion of TILs, produced a vast improvement in the efficacy of TIL-based adoptive T-cell therapy for metastatic melanoma, as demonstrated by 50% objective response rate reported recently.253 The mechanism by which lymphodepletion prior to TIL infusion dramatically improves therapeutic effects is not fully understood. It has been suggested that lymphodepletion in the absence of myeloablation removes immunoregulatory elements (e.g., regulatory T cells) and increases production of lymphokines, such as IL-21, and physical spaces in secondary lymphoid organs, allowing infused TIL to expand with less competition from pre-existing lymphocytes.

2. Genetic Manipulation of TAA-Specific CD8+ T Cells Ex Vivo

Similar to other experimental tumor immunotherapies, adoptive T-cell therapy faces several technical hurdles. However, unlike other tumor immunotherapies, for adoptive therapy, a majority of such hurdles could be overcome by ex vivo genetic manipulation of T cells prior to transfer. For instance, the majority of TAAs are poorly immunogenic, and it is often difficult to raise CTLs that possess T-cell receptors (TCRs) that have sufficiently high avidity for use in adoptive therapy. One of the ways to solve this problem is to systematically search TAA-specific CD8+ T cells for clone(s) with higher TCR avidity, clone TCRα and β genes, and exogenously express the high-avidity TCR in bulk CD8+ T cells. Li et al.254 recently utilized phage display to systematically search for a high-avidity TCR against an HLA-A0201-restricted epitope in a TAA NY-ESO-1.255 Moreover, adoptively transferred high-avidity TCR transgenic T cells demonstrated sustained high levels of circulating, engineered cells at 1 year after infusion in 2 of 15 patients, both of whom demonstrated objective regression of metastatic melanoma lesions,256 suggesting the therapeutic potential of genetically engineered cells for the biologic therapy of cancer.

An interesting alternative to the expression of high-avidity TCR-transgenic T cells is to express a chimeric molecule that has antigen-binding domains of a MAb fused with a signal-transduction domain of CD3,257 namely, chimeric antigen receptors (CAR). A significant advantage of CARs over TCRs is that the antigen recognition is not restricted by the expression of certain MHC class I molecules, and, recently, CARs have been used to treat malignant gliomas,258 ovarian cancer,259 pediatric neuroblastomas,260 and renal carcinoma,261 among others. A recent promising development with a slight variation to this approach is a single-chain CAR that targets CD19262 and a single-chain chimeric TCR-IL-2 signal-transduction domain.263

An underlying theme of these modern adoptive T-cell approaches is to design a surface receptor that has defined antigen specificity and is able to trigger T-cell activation but overcomes several limitations of using transgenic TCRs. However, overexpression of high-avidity TCR or nonphysiological expression of CAR may have unwanted consequences. Frequent exposure to TAAs may render these engineered T cells functionally unresponsive, similar to the unresponsive CD8+ T cells specific for antigens derived from viruses that establish persistent infection. In fact, there is an accumulating line of evidence suggesting that functional unresponsiveness of the tumor-specific CTL is mediated by the expression of PD-1.264269 It is possible that lengthy ex vivo expansion induces high levels of PD-1 expression on a majority of TAA-specific CD8+ T cells. However, it may also be possible to overcome this problem by administering blocking anti-PD-1 or anti-PD-L1 antibodies, as reported in a case for correcting unresponsive HIV-1-specific CD8+ T cells in persistent HIV-1 infection.270 The use of humanized anti-PD-1 antibody is currently being tested in a randomized Phase I clinical trial (272).271

Another related issue is whether a majority of ex vivo-activated cells for adoptive transfer become enriched in an effector T-cell subset (TE) during ex vivo expansion. TE are generally considered to be terminally differentiated CTLs that have the highest cytotoxic capacity but lack appreciable proliferating capacity.272 A currently prevailing notion is that because these cells are terminally differentiated, they will not be able to establish a long-term persisting population of TAA-specific CTLs. Clearly, there is a significant association between clinically favorable responses and the persistence of ex vivo-expanded melanoma-specific CTL clones after infusion.273 Moreover, persisting melanoma-specific CTL clones expanded ex vivo prior to transfer appear to have relatively shorter telomere length, indicating that the higher replicating capacity of transferred CTLs is a prerequisite for their persistence.274 Interestingly, these ex vivo-expanded CTLs (CCR7CD27CD45RA+CD28) clearly indicate the TE phenotype. However, these cells appear to re-express an immature differentiation marker, CD27, after infusion. It is known that once CD8+ T cells lose expression of these markers as a part of the differentiation process, they do not re-express such markers in vivo.275 This apparent contradiction is most likely due to the fact that ex vivo expansion involves a milieu of cytokines exogenously added at nonphysiological concentrations. In fact, it has been shown that re-expression of CCR7 on CMV-specific CD8+ T cells, a prototypical TE (CD27 CD45RA+), is only induced in the presence of high levels of IL-15 after a week of antigenic stimulation.276 Furthermore, exposure to a high level of IL-2-restored CD28 expression on otherwise CD28– CMV-specific and HIV-1-specific CD8+ T cells.277 Restoration of CD27 and CD28, but not CCR7 and CD62L expression, is also seen on melanoma-specific CD8+ T cells 2 months after transfer.278 The gain of CD27 expression by transferred CTLs appears to be particularly important because CD27 is the critical molecule for long-term persistence279 and expansion280 of transferred CD8+ T cells.

3. Adoptive T-Cell Therapy for Glioblastoma: Present and Future

Although a number of clinical trials using adoptive T-cell therapy for malignant glioma are very limited when compared to clinical trials using active immunization strategies, there are currently at least three active clinical trials for GBM using adoptive T-cell transfer according to the NIH clinical trial database (www.clinicaltrials.gov). Interestingly, two of such clinical trials involve ex vivo expansion of autologous CD8+T cells that are specific for a CMV antigen (refer to Section III.C.1).

Apparently, adoptive T-cell therapy for malignant gliomas needs to incorporate recent advances in the repertoire of target antigens and the engineering of T cells. First, one of the most important features missing in current clinical trials for malignant gliomas is use of the glioma-associated antigen (GAA)-derived epitopes to generate and expand CD8+ T cells (refer to Section III.C.1). These currently include HLA-A*02-restricted IL-13Rα2,281 TRP-2,282 AIM-2,222 HER-2, gp100, MAGE-1,283 EGFRvIII,284 EphA2,285 and EphB6.286 These GAA epitopes have been preclinically validated, and a majority of these peptides have been used in various clinical trials of DC-mediated active immunization against malignant gliomas.

As the list of GAA epitopes grows, it is necessary to generate a library of human CTL clones against GAAs and test their TCR avidities from which high-avidity CTL clones will be selected and further expanded. As previously discussed, sources of CTL clones could be TILs isolated from tumor biopsies and PBMCs. Once a high-avidity CTL clone is selected and established, TCR usage can be identified and subsequently used to build a library of high-avidity TCR gene pairs from which transgenic CTLs (expressing cloned high-avidity TCRs tailored to the particular GAA epitopes) could be generated. In this context, it will be of interest to identify long-term controllers287 or active DC immunization responders who may naturally possess high-avidity CD8+ T cells specific for GAAs and characterize TCR usage of GAA-specific CTLs from such individuals. Alternatively, CTL clones could be generated from transgenic mice expressing human HLA molecules upon antigenic challenge. An advantage of this approach is that mouse T cells naturally have relatively higher avidity for epitopes derived from human antigens,288 and appear to form a functional signal-transduction complex with human CD3.289 A major caveat of this approach is that mouse proteins are naturally immunogenic; the extent to which expression of such mouse, or even humanized, TCRs affect persistence of transferred transgenic T cells in humans is unknown. Moreover, human T cells bearing transgenic high-avidity mouse TCRs may run the risk of losing effector functions and become functionally unresponsive upon tumor infiltration, as discussed previously. In fact, a recent observation shows that high-avidity mouse CTLs differentiate into effectors in the draining lymph nodes and become functionally unresponsive after tumor infiltration.290 Thus, CTLs expressing either endogenous or exogenous high-avidity TCRs may not maintain functionality in vivo.

4. Summary

Adoptive T-cell therapy may emerge as one of the most promising cancer immunotherapy modalities. Today’s adoptive T-cell therapy employs cutting edge methodologies involving ex vivo expansion as well as genetic manipulation of TAA-specific T cells prior to the transfer of such cells. Despite a promising future, adoptive T-cell therapy has to be further refined to overcome several of the major problems, including T-cell suppression by Tregs, inadequate persistence of transferred T cells, and undirected migration of T cells. Adoptive T-cell therapy for malignant gliomas is no exception to these concerns. In fact, it faces many similar and more difficult challenges. In particular, the microenvironment of malignant glioma poses a significant problem, such as the presence of the BBB and the production of a variety of immunosuppressive substances. Nevertheless, adoptive T-cell therapy is better suited to circumventing these problems than active immunotherapy because adoptive therapy allows direct ex vivo manipulations of TAA-specific CTLs to give these cells enhanced or novel functions that are otherwise impossible in vivo. Thus, it may be possible to redirect trafficking of transferred CTLs to the site of gliomas simply by exogenously expressing chemokine receptors (e.g., CXCR3)47 and integrin receptors (e.g., very late antigen [VLA]4)45,48 that dictate efficient homing of T cells to gliomas. It should also be mentioned that adoptive T-cell therapy is not limited to the transfer of CD8+ T cells. There are NK cells and TAA-specific CD4+ T cells that can be isolated, expanded, and transferred back to the autologous cancer patient. It may be possible to overcome some of these problems discussed previously by the simultaneous transfer of TAA-specific CD4+ T cells, providing necessary helper functions to TAA-specific CTLs. Historically, therapeutic values of TAA-specific CD4+ T cells in human cancer treatment have been long neglected until recently shown by a study demonstrating beneficial clinical responses to malignant melanoma by the transfer of ex vivo-expanded melanoma-specific CD4+ T cells.291 Considering recent developments, the future of adoptive T-cell therapy against malignant gliomas appears promising. With further innovation and refinement of ex vivo T-cell manipulation, adoptive T-cell therapy may become a mainstream therapeutic intervention for malignant gliomas.

C. Active Immunotherapy (Tumor Vaccines)

1. T-Cell Epitopes Derived from Glioma-Associated Antigens

There is a large body of reported studies that have demonstrated the safety and preliminary efficacy of whole glioma cell-based vaccine approaches.292295 However, the use of whole glioma cell-derived antigens, such as glioma lysate, may limit the feasibility and safety of the approach because of the cumbersome preparation procedures and theoretical concerns of autoimmune encephalitis.296 Therefore, the effectiveness and safety of T-cell-mediated immunotherapy of glioma depends on the proper selection of the targeted antigens— that is, GAAs and, more specifically, the CTL epitopes in GAAs. In addition, the use of “off-the-shelf” synthetic peptides coding GAA-derived CTL epitopes may be feasible, especially for multicenter clinical trials. Table 1 lists GAA epitopes known to elicit T-cell responses. The following section discusses selected human GAA-derived epitopes that appear to be promising on the basis of relatively restricted expression (compared with the normal brain) and biological significance in gliomas, as well as well-characterized immunogenicity. The review primarily focuses on HLA-class I-restricted CTL epitopes. However, the growing body of knowledge on other CTL and helper T-cell epitopes will expand our future options to encompass more efficient immunotherapy for glioma.

TABLE 1.

Glioma-Associated Antigens

Antigen peptide Expression in cancers Expression in normal Tissues Roles in tumorigenesis HLA restriction Refs.
IL-13R±2 Gli, CC Testis ND A*0201, A*2402 281,297,309,312
EphA2 Gli, CC, OC, Bla, GC, RCC, Pro, EC Sites of cell-to-cell contact Regulation of angiogenesis induced by tumors A*0201 285,298,315
Survivin Gli, Mel, PC, LC, CC, BC, Pro Undetectable in most differentiated normal adult tissues Apoptosis inhibition in tumor progression A*0201, A*2402,A1,A3 299,300,301,320
WT1 Gli, AML, LC, BC Kidney, bone marrow, pleura, testis, ovary, hemopoietic stem cells Cell proliferation, apoptosis inhibition A*0201,A*2402,A1 302, 303, 304, 324, 326
SOX2 Gli Neural stem cells, fetal brain, testis Proliferation of glioma-initiating cells A*0201 305, 306, 334
SOX11 Gli Fetal brain ND A*0201 306
HER2/neu Gli, BC, CC, PC, OC, RCC, Medu Ubiquitously expressed Cell proliferation, apoptosis inhibition 13 HLA class I epitopes 283,340,341,342
EGFRvIII Gli, LC, CC, OC, GC, EC, RCC, Bla, PC No Cell proliferation A*0201 284
gp100 Gli, Mel Melanocyte, retina ND 16 HLA class I epitopes 283
MAGE-1 Gli, Mel, HCC, BC, RCC Testis, placenta ND 11 HLA class I epitopes 283
TRP-2 Gli, Mel Melanocyte, retina ND A*0201 282
AIM-2 Gli, Mel, CC, BC, OC Liver, testis ND A1 283
SART-1 Gli, EC, HCC, RCC, BC Testis ND A*2402, A*2601 307, 308

Note: Gli = glioma, Mel = melanoma; CC = colorectal cancer; LC = lung cancer; GC = gastric cancer, BC = breast cancer; HCC = hepatocellular cancer; OC = ovarian cancer; PC = pancreatic cancer; EC = esophageal cancer; RCC = renal cell cancer; Pro = prostate cancer; Bla = bladder cancer; Medu = medulloblastoma; AML = acute myeloid leukemia.

IL-13Rα2

IL-13Rα2 is a membrane glyco-protein that is overexpressed by >80% of malignant gliomas but is not expressed in normal brain tissues at detectable levels or at high levels in other normal organs, except for testes.309 Although the function of IL-13Rα2 has not been fully elucidated,310,311 IL-13Rα2 has attracted significant attention as a target for glioma therapy.258 We recently found that an analogue peptide of natural IL-13Rα2345–353,281 in which the first and ninth amino acid residues, tryptophan and isoleucine, have been replaced by valine and alanine, respectively, can elicit a greater CTL response against HLA-A2+, IL-13Rα2+ glioma cells compared to the natural peptide (IL-13Rα2345–353:1A9V).312 Recently, an HLA-A24-restricted CTL epitope was identified in IL-13Rα2, expanding the target patient population for vaccines targetingIL-13Rα2.297

EphA2

EphA2 is a tyrosine kinase receptor that plays a role in carcinogenesis.313,314 In normal cells, EphA2 localizes to sites of cell-to-cell contact,315,316 where it may negatively regulate cell growth. EphA2 is frequently overexpressed and often functionally dysregulated in advanced cancers, contributing to their malignant characters.317 We have reported that EphA2883–891 can elicit an HLA-A2-restricted CTL response against glioma cell lines.285 Recent studies by us285 and others318 have revealed that a majority of malignant gliomas express high levels of EphA2. Furthermore, EphA2 mRNA overexpression was found to correlate inversely with survival in a panel of 21 GBMs.319 These findings support the idea that targeting of EphA2 by immunotherapy may provide a major impact on controlling tumor growth and prolonging patients’ survival.

Survivin

Survivin is an apoptosis inhibitor protein overexpressed in most human cancers, and inhibition of its function results in increased apoptosis.320 The induction of cytotoxic immunity against Survivin may, therefore, be an attractive strategy.321 Survivin has multiple T-cell epitopes, including Survivin96–104 as an HLA-A2-restricted CTL epitope.299,300 Moreover, vaccination of a pancreatic cancer patient with a modified Survivin epitope-peptide, Survivin96–104:2M, in which the second residue threonine was replaced by methionine, induced complete remission of liver metastasis.322 Further, vaccinations using DCs loaded with Survivin96–104 induced positive IFN-γ ELISPOT responses in four out of five patients with advanced melanoma.323 A recent immunohistochemical study demonstrated that 100% of astrocytoma specimens (n = 29; grades II–IV), but not normal brain tissues, contain Survivin-positive cells.301 The mean percentage of immunoreactive cells in each specimen was 70.0 in grade II, 81.3 in grade III, and 85.0 in grade IV. Interestingly, high-level expression of Survivin was correlated with poor prognosis in patients with grade II or III astrocytomas.301

Wilm’s Tumor (WT)1

WT1 is the product of the Wilm’s tumor gene. WT1 is a transcription factor oncogene, overexpressed in various types of leukemia and solid tumor cells.304,324 Inhibition of WT1 in leukemic cell lines led to a decrease in proliferation and an increase in apoptosis of tumor cells,325 implying that the elimination of tumor cells that overexpress WT1 may allow efficient control against tumor growth. Two 9-mer HLA-A2-binding WT1-derived peptides (WT1126–134 and WT1187–195) have been demonstrated to sensitize CD8+ antigen-specific CTLs.303 Also, HLA-24-restricted epitopes have been characterized and tested in cancer vaccine trials.326 In human astrocytomas, 48 of 51 primary GBM tissue samples showed immunohistochemical staining for the WT1 protein.327 A recent quantitative evaluation of WT1 in grades I–IV astrocytomas showed that WT1 protein was expressed in 5 of 6 low-grade (grades I–II) and in 18 of 18 high-grade (grades III–IV) astrocytomas, with a trend of higher expression levels in high-grade astrocytomas. The WT1 protein was not detected in the normal glial cells contained in the tumor specimens.302 Furthermore, a recent Phase II study of vaccinations using an HLA-A24-restricted WT1 peptide in patients with recurrent GBM has demonstrated complete or partial response in 2 of 21 (9.5%) patients and median PFS at 20 weeks.328

Sry-Related High-Mobility Group Box (SOX)

SOX is a family of transcriptional cofactors implicated in the control of diverse developmental processes and exhibits highly dynamic expression patterns during development of diverse tissues and cell types, especially during embryogenesis.329 In addition, SOX genes are amplified or up-regulated in different tumors and tumor cell lines (reviewed in Ref. 142). Indeed, SOX2, SOX5 (335),330 SOX6,331 and SOX11306 were highly expressed in glioma cell lines and a majority of glioma tissues tested. SOX5 and SOX6 induced specific IgG responses in one third of the glioma patients.330,331 Patients with GBM who showed IgG responses against SOX5 exhibited significantly better survival periods than GBM patients without SOX5 antibodies.330 DNA vaccinations with SOX6 exerted protective and therapeutic antitumor responses in the glioma-bearing mice.332 SOX2- and SOX11-derived peptides were able to elicit HLA-A2-restricted CTL responses against glioma cell lines.305,306 Their preferential expression in glioma and immunogenicity indicate that SOX proteins are attractive targets for immunotherapy. Furthermore, a recent study demonstrated that SOX2 was expressed in neural stem cells and GBM tumor-initiating cells, and it played a role in the maintenance of self-renewal capacity of neural stem cells, as well, when they acquired cancer properties,333 suggesting that the SOX2-derived CTL epitope may induce CTL responses against glioma-initiating cells, which are likely to be responsible for the malignant behavior of cancer due to resistance to chemotherapy and radiotherapy.334338

HER-2/neu

HER-2/neu is a transmembrane glycoprotein and member of the EGFR family. HER-2/neu promotes tumor-cell proliferation, migration, adhesion, and angiogenesis, and decreases apoptosis.339 HER-2/neu is widely expressed in a variety of normal tissues, but selectively overexpressed in various tumors, such as breast, ovarian, colorectal, pancreatic, and renal-cell cancers.338,340343 HER-2/neu protein expression was detected in 76% of GBM primary cell lines and an HLA-A2-restricted HER-2/neu-specific CTL clone recognized GBM cells.283 Moreover, Her-2/neu protein overexpression was found to correlate inversely with survival in a panel of 149 GBMs,344 suggesting that HER-2/neu-targeting immunotherapy for GBM may provide a major impact in controlling tumor growth and prolonging patients’ survival.

EGFR vIII

The type III variant of the EGFR mutation (EGFR vIII) is present in 30%–50% of patients with GBM. Saikali et al.345 extensively evaluated the expression of nine tumor antigens in human GBM in both mRNA and protein levels in a series of 47 GBM cases. Among nine antigens, including ALK, EGFRvIII, GALT3, gp100, IL-13R α2, MAGE-A3, NA17-A, TRP-2, and tyrosinase, it was found that EGFRvIII and IL-13R α 2 are expressed most frequently and restrictedly (vs. normal brain samples) in primary GBM cases. A peptide encoding the amino acid sequence for EGFRvIII (1–13) was shown to induce modest IFN-γ and CTL responses against HLA-A0201+ EGFRvIII-transfected U87 glioma cells.284

Squamous Cell Carcinoma Antigen Recognized by T Cells 1 (SART-1)

SART-1 was identified as a gene-coding tumor antigen recognized by a squamous cell carcinoma-specific CTL line using the expression-gene cloning methods.308 SART-1 was expressed in various cancers, including glioma, but not in normal tissues, except for testes.307 A SART-1-derived peptide induced an HLA-A24-restricted CTL response against glioma cell lines. However, there is a paucity of information in the literature regarding the roles of SART-1 in neoplastic cells and the association between SART-1 expression and the clinical features of glioma patients.

Cytomegalovirus (CMV) as a Potential Target

Cobbs et al.346 reported that a high percentage of malignant gliomas are infected with human CMV, suggesting that this virus plays an active role in glioma pathogenesis. Although there was some controversy over these observations, recent studies demonstrated the presence of CMV in GEM347,348 and even in low-grade gliomas.349 These studies suggest that the presence of human CMV in glial tumors could serve as an immunotherapeutic target in glioma. The facilitation of an immune response against viral antigens contrasts with the difficulty of immunization against self-antigens. It will be intriguing to introduce the CMV-derived epitope to multiepitope-based vaccines for glioma.

Although the list of available GAA-derived CTL epitopes is growing, it is also important to point out the possibility that immune responses to GAAs are not restricted to HLA-A2, A24, or other HLA-A alleles. Other HLA alleles, including HLA-B and HLA-C, clearly present GAA epitopes and these epitopes could potentially be more immunogenic than those presented by the HLA-A allele. There is limited but significant evidence indicative of this notion. Immune responses to HLA-B-restricted HIV-1 epitopes typically have greater magnitude than those restricted by HLA-A alleles.350 CD8+ T cells specific for HLA-B-restricted HIV-1 epitopes have greater polyfunctionalities than the ones specific for HLA-A-restricted HIV-1 epitopes.351 Clearly, new GAA epitopes will be discovered in the future, although the safety and immunogenicity of these peptides must be further evaluated concurrently in the clinical trials.

2. Active Vaccination Strategies for Gliomas

Although numerous preclinical studies in mouse models have shown the efficacy of peripheral vaccinations against intracranial gliomas (reviewed in Ref. 255), therapeutic vaccines face a substantial challenge in glioma patients because they must overcome a variety of immunoregulatory mechanisms that have already established the immune escape of tumors, as discussed in the previous section. Nevertheless, a number of clinical trials have been attempted to generate therapeutic immune responses against gliomas and have shown some positive effects. Strategies that have been most studied are categorized as follows: (1) whole glioma-cell vaccines, (2) peptide-based vaccines targeting glioma-associated antigens, and (3) DC vaccines (Tables 2 and 3).

TABLE 2.

Published Glioma Vaccine Trials

Authors Pts Diagnosis Study design Type of vaccination Outcome Indicators for better results
Holladay et al.352 15 GBM,AA Phase I Autologous irradiated glioma cells and BCG 7 PR
Plautz et al.353 10 GBM Phase I Autologous irradiated glioma cells and GM-CSF 2PR
Plautz et al.354 12 GBM,A, astrocytoma Phase I Autologous irradiated glioma cells and GM-CSF 4PR
Wood et a l.355 9 GBM,AA Pilot study Autologous irradiated glioma cells and BCG 3PR
Yu et al.356 9 GBM,AA Phase I DCs pulsed with acid-eluted peptides
Andrews et al.357 12 GBM,AA Pilot study Autologous glioma cells treated with IGF-IR antisense oligo DNA 2CR, 4PR
Kikuchi et al.358 8 GBM,AA Phase I DC-glioma fusion cells by PEG 2 PR
Schneider et al.359 11 GBM Phase I Autologous glioma cells infected by NDV
Yamanaka et al.360 10 GBM,AA Phase I/II DCs pulsed with autologous glioma-cell lysate 2 MR
Yu et al.295 14 GBM,AA Phase I DCs pulsed with autologous glioma-cell lysate
Caruso et al.361 9 Glioma Phase I DCs transfected with glioma-derived RNA 1 PR
Steiner et al.362 23 GBM Pilot study Autologous glioma cells infected by NDV 1 CR Increased CTL activity
Ruthkowski et al.363 12 Glioma Phase I DCs pulsed with autologous glioma cell lysate 1 PR
Kikuchi et al.364 15 GBM,AA Phase I DC-glioma fusion cells by PEG and IL-12 4 PR IL-12 administration
Yamanaka et al.365 24 GBM,AA Phase I/II OK-432-stimulated DCs pulsed with autologous glioma-cell lysate 1 PR, 3 MR OK-432 stimulation; a combination of intradermal and intracranial DC injections
Liau et al.292 12 GBM Phase I DCs pulsed with acid-eluted peptides 1 PR Slowly progressing tumors; tumors producing TGF-β2 at low levels
Yajima et al.366 17 GBM,AA Phase I DCs pulsed with recombinant peptides 5 PR
Sloan et al.367 19 GBM,AA Pilot study Autologous irradiated glioma cells and GM-CSF 5 PR
Ishikawa et al.368 12 GBM Pilot study Formalin-fixed autologous glioma cells 1 CR, 1 PR, 2 MR
Okada etal.293 12 GBM,AA Phase I DCs pulsed with autologous glioma-cell lysate and IL-4-transfectedfibroblasts 2 PR
Izumoto et al.328 21 GBM Phase II DCs pulsed with WT1 peptide 2 PR
De Vleeschouwer et al.369 72 HGG Phase II DCs pulsed with autologous glioma-cell lysate Younger age, total resection of tumors
Wheeler et al.294 34 GBM Phase II DCs pulsed with autologous glioma-cell lysate 3 CR, 4 PR Higher responsiveness of IFN-γ in PBMCs

Note: GBM = glioblastoma multiforme; AA = anaplastic astrocytoma; BCG = Bacille de Calmette et Guérin; PR = partial response; GM-CSF = granulocyte-macrophage colony-stimulating factor; CR = complete response; DCs = dendritic cells; PEG = pegylated; NDV = Newcastle disease virus; CTL = cytotoxic lymphocyte; HGG = high-grade glioma; MR = minor response.

TABLE 3.

Pros and Cons of Each Vaccine Approach

Pros Cons
Whole-cell vaccine Identification of glioma-associated Ags not needed
All HLA types available
Broad immune response
Limiting risk of tumor immune escape
Quantity of antigenic materials needed
Cumbersome for patient-specific vaccine preparation
Variable vaccine composition
Increased risk of autoimmunity
Peptide vaccine “Off-the-shelf” feasibility
Autologous tumor cells not needed
Relative tumor specificity
Paucity of defined antigen for human gliomas
Specificity for individual HLA types needed
Increased risk for antigen-loss variant
DC vaccine Arguably the most potent Ag-presenting cells Cost for generating high-quality DCs

Note: Ags = antigens; DCs = dendritic cells.

Whole Glioma-Cell Vaccines

Initial vaccination strategies for gliomas consisted of subcutaneous inoculations of irradiated, autologous,370 or allogeneic167,371 glioma cells. This type of vaccine has the advantage of providing a panel of multiple potential GAAs that are naturally expressed by glioma cells. Autologous glioma cells, in particular, should allow immunizations against the most relevant GAAs expressed in the patient’s tumor (i.e., tailored medicine). Potential downsides of this approach, however, include (1) cumbersome procedures and quality control (QC)/quality assurance (QA) issues associated with large-scale cultures of autologous glioma cells (Table 2) and (2) theoretical risks of autoimmune encephalomyelitis (reviewed in Ref. 370). Nevertheless, this type of vaccine strategy has been carefully examined. Schneider et al.359 and Steiner et al.362 recently reported pilot clinical trials using autologous glioma cells modified with Newcastle-Disease-Virus, which is known to serve as a vaccine adjuvant and, therefore, to improve the efficacy of glioma vaccines. More recently, Ishikawa et al.368 reported a Phase I clinical trial using formalin-fixed glioma tissues as a source of antigens. The advantage of this strategy is that formalin fixation preserves the specific antigenicity of glioma cells. These studies reported no major adverse events.

Peptide-Based Vaccines Targeting Glioma-Associated Antigens

In vaccines using synthetic peptides for shared GAA-epitopes, advantages and disadvantages are distinct from those in whole glioma-cell approaches. Although synthetic GAA peptide-based vaccines may not adequately target antigens in each patient’s tumor, these vaccines have less concern for autoimmunity and provide “off-the-shelf” feasibility. Indeed, a wide range of peptide-based vaccines have been evaluated. Yajima et al.366 reported a Phase I study of peptide-based vaccinations in patients with recurrent malignant gliomas. In this study, prior to the first vaccine, each patient’s PBMCs were evaluated in vitro for cellular and humoral responses against a panel of antigens, and peptides that induced a positive response were used for vaccinations. The regimen was well tolerated and resulted in an 89-week median survival of treated patients. However, there is little evidence that the antigens used in this study are expressed in gliomas at high levels. More recently, Izumoto et al.328 reported a Phase II clinical trial using a single WT1 peptide. In this study, they reported a median PFS of 20 weeks and a possible association between the WT1 expression levels and clinical responses. When single or oligo antigens are selected and targeted by vaccines, it also seems necessary to harness the concepts of epitope spreading to address the problems of tumor immune escape, while avoiding the augmentation of deleterious CNS autoimmune responses.372

DC Vaccines

DCs are the most potent APCs, driving the activation of T cells in response to invading microorganisms.373 The ability to culture DCs from human peripheral blood monocytes has generated significant interest in using DCs in novel cancer-vaccination strategies.374

Yu et al.356 reported a Phase I trial of vaccinations using DCs pulsed with peptides eluted from autologous glioma cells. Later, Liau et al.292 also reported a Phase I trial in patients with newly diagnosed GBM using DCs pulsed with acid-eluted glioma peptides. In this study, the authors reported the median overall survival as 23.4 months and that the benefit of the vaccine treatment was more evident in the subgroup of patients with slowly progressing tumors and in those with tumors expressing low levels of TGF-β2.

However, pulsing DCs with eluted peptides requires a large culture of autologous glioma cells and time-consuming procedures, for which QC/QA is not always feasible. To overcome this issue, glioma-cell lysate has been used to pulse DCs in a number of trials.294,295,360,363,365,375 Yamanaka et al 360,365 reported a Phase I/II study using DC pulsed with glioma lysate. Patients received either DCs matured with OK-432 or DCs without OK-432-mediated maturation. GBM patients receiving mature DCs had longer survival than those receiving DCs without OK-432-mediated maturation. Furthermore, patients receiving both intratumoral and intradermal DC administrations demonstrated longer overall survival than those with intradermal administrations alone.365 Recently, Wheeler et al.294 reported a Phase II clinical trial with lysate-pulsed DCs. IFN-γ production levels from postvaccine PBMC correlated significantly with patients’ survival and time to progression.

Fusion of glioma cells and DCs represents a unique cancer-vaccine strategy. Fused cells can effectively present antigens derived from tumor cells.376 Kikuchi et al.358,364 reported a study using DCs fused with glioma cells by polyethylene glycol, with or without concomitant administration of IL-12. Although there were no serious adverse effects, the study did not clearly demonstrate the effects of IL-12 administration on immunological and/or clinical response.364 Further trials, including the optimization of the IL-12 dosage, are needed to draw any conclusions.

Recently, a small subset of tumor cells in GBM has been identified by their unlimited self-renewal ability and remarkable in vivo tumor-forming capability.377 These brain tumor stem cells (BTSCs) or glioma stem cells (GSC), which have been isolated by the neural stem-cell marker CD133,378 are believed to play a major role in tumor growth as well as resistance against radiation335 and chemotherapy.379 However, their susceptibility to immunotherapeutic attack has not been extensively tested. Immunologic targeting of the GL261 murine glioma using DCs pulsed with lysate derived from in vitro-cultured GL261 stem cells was more efficient than DCs pulsed with lysate of conventional tumors.380 Thus, identification of antigens expressed in GSCs and development of immunotherapies targeting GSCs is a promising approach.

To date, no consensus exists on the optimal DC subtypes, the optimal conditioning and activation stimuli, the optimal route of administration, and the optimal dose and frequency of DC vaccinations.381 We recently reported a pilot vaccine study using autologous type 1-polarized DCs (αDC1s) with IL-4-transfected fibroblasts.293 This strategy is based on the previous studies demonstrating that (1) αDC1s produce high levels of IL-12 and promote type 1 adaptive immunity,382,383 and (2) local paracrine production of IL-4 in vaccine sites promotes the accumulation and maturation of IL-12-secreting tumor-infiltrating DCs.384 Because this was a pilot study with only 1 × 106 αDC1/vaccination, there were no detectable immunological or clinical responses. However, fostered by the feasibility data for production of αDC1 in vaccine trials, we recently implemented a Phase I/II trial in patients with recurrent WHO grades III or IV glioma to evaluate the safety and induction of anti-GAA immunity of a vaccine strategy that employs intranodal injections of αDC1s (1–3 × 107) loaded with 4 human GAA peptides (EphA2883–891, IL-13Rα2345. 1A9V, YKL-40201–210 and GP100209–217:M2) in combination with intramuscular injections of poly-ICLC (20 μg/kg; twice per week) (University of Pittsburgh Cancer Institute protocol #05-115). To date, we have enrolled a total 13 patients and completed 4 scheduled vaccinations per patient in 10 participants. No CTCAE Grade 3 or higher adverse events were reported. None of the patients enrolled in this trial have demonstrated clinical indices, radiological signs, or laboratory data suggestive of auto immunity. Immune responses have been evaluated in 9 participants who received at least 4 vaccinations, and 7 participants demonstrated detectable positive responses against at least one of the GAAs in ELISPOT and/or tetramer assays. As type 1 immunity is the optimal adaptive immune response45,47,48 for CNS tumor immunotherapy, αDC1-based vaccine approaches seem to hold promise.

Several pilot and Phase I/II clinical studies of active vaccination have been undertaken in patients with glioma. Despite the fact that feasibility and safety have been sufficiently documented in most studies, clinical efficacy has not yet been convincingly proven. Although some studies demonstrated improved survival of patients and objective clinical responses, the ultimate judgment for clinical activity has to be made by rigorous evaluation in randomized studies.

IV. CONCLUDING REMARKS

We reviewed recent progress in the field of brain and brain-tumor immunology. We also reported recent progress and current challenges in immunotherapeutic strategies for brain tumors. In the field of CNS and CNS-tumor immunology, in the last decade, novel immunoregulatory mechanisms, such as B7-H1, have been identified and added to the list of classical immunosuppressive molecules, including TGF-β and IL-10. It is clear that the CNS and CNS tumors are equipped with numerous and layered immunosuppressive and immune-escape mechanisms, perhaps including ones that we have not yet identified. These discoveries, however, allow us to develop strategies to overcome each of these mechanisms.

Remaining unique challenges against gliomas include relative difficulties in obtaining tumor tissues following immunotherapeutic treatments. Unlike other cancers, such as cutaneous melanoma, intracranial glioma tissues are not readily accessible following vaccine treatment. Designing neoadjuvant settings with vaccines is not always feasible because recurrent malignant gliomas, for which surgical resection is clinically indicated, typically do not allow us to wait for weeks before surgery and often require treatment with high-dose corticosteroids.

As reviewed in this article, the concept of immunotherapy has a diverse scope of strategies and target molecules. Extensive review of each field in this article has led us to identify the challenge for each strategy. Such challenges, however, may be overcome by appropriate combinations with other strategies. For example, adoptive-transfer strategies may need to be combined with appropriate adjuvants and/or vaccinations to promote long-lasting memory responses and antitumor immunosurveillance. Use of targeted-toxin therapy may facilitate the release of tumor antigens, thereby inducing T-cell immune responses against a variety of T-cell epitopes (i.e., epitope spreading). Such events maybe harnessed by concurrent vaccinations against multiple epitopes and/or the blockade of appropriate immune-escape mechanisms. However, when each of these agents are owned by separate industries with intellectual properties, such creative combinatorial strategies may not be implemented as efficiently as we would wish. Although several early-phase clinical trials demonstrated promising therapeutic outcomes, to date, clinical trials of immunotherapy for gliomas have not yet demonstrated objective proof of clinical efficacy in rigorous Phase II and III studies. The eventual success of immunotherapies for brain tumors will be dependent upon not only an in-depth understanding of immunology behind the brain and brain tumors, but also the implementation of molecularly targeted trials that address multiple layers of challenges in gliomas.

Acknowledgments

We thank Heather A. McDonald for her critical review of the manuscript. Financial support was provided to H. Okada by Grant Nos. 1R01NS055140-01, 1P01CA100327, and 2P01NS40923.

ABBREVIATIONS

AA

anaplastic astrocytoma

APCs

antigen-presenting cells

B7-H1

B7-homologue 1

BBB

blood brain barrier

BED

biological effective dose

BNCT

boron neutron capture therapy

BPA

boronophenylalanine

BTSCs

brain tumor stem cells

CAR

chimeric antigen receptor

CB

cintredekin besudotox

CED

convection-enhanced delivery

CLNs

cervical lymph nodes

CMV

cytomegalovirus

CNS

central nervous system

CSF

cerebral spinal fluid

CTL

cytotoxic T lymphocytes

DCs

dendritic cells

DT

diphtheria toxin

EAE

experimental autoimmune encephalitis

EBRT

external beam radiotherapy

ECD

extracellular domain

EGFR

epidermal growth factor receptor

EM

electron microscopy

GAA

glioma-associated antigen

GANGs

ganglio-sides

GBM

glioblastoma multiforme

GCS

glioma cell supernatants

GIM

glioma-infiltrating macrophages

GSC

glioma stem cells

HA

hemagglutinin

HLA

human leukocyte antigen

IFN

interferon

IL

interleukin

ISF

interstitial fluid

JAM

junctional adhesion molecule

LAK

lymphokine-activated killer

LFA

lymphocyte function-associated molecule

MAb

monoclonal antibody

MCP-1

macrophage chemoattractive protein

MHC

major histocompatibility complex

MS

multiple sclerosis

MST

median survival time

NK

natural killer

NVU

neurovascular unit

PBMCs

peripheral blood mononuclear cells

PD-L1

programmed death ligand-1

PE

Pseudomonas exotoxin

PFS

progression-free survival

PG

prostaglandin

PI3K

phosphatidylinositol-3-OH kinase

PLP

myelin proteolipid protein

PTEN

phosphatase and tensin homolog

RCAS1

receptor-binding cancer antigen expressed on SiSo cells

RIT

radioimmunotherapy

SAC

Staphylococcus aureus Cowen I strain

SART-1

squamous cell carcinoma antigen recognized by T cells 1

SOX

Sry-related high-mobility group box

TAA

tumor-associated antigen

TCR

T-cell receptor

TE

effector T-cell subset

Tf

transferrin

TGF

transforming growth factor

Tg+

transgenic

TILs

tumor-infiltrating lymphocytes

TJ

tight junction

TLR

Toll-like receptor

TMZ

temozolomide

TNF

tumor necrosis factor

VEGF

vascular endothelial growth factor

VLA

very late antigen

WHO

World Health Organization

WT

Wilm’s Tumor

References

  • 1.Kleihues P, Cavenee WK. Pathology and genetics of tumours of the nervous system. Lyon: International Agency for Research on Cancer; 2000. [Google Scholar]
  • 2.Ashby LS, Shapiro WR. Low-grade glioma: supratentorial astrocytoma, oligodendroglioma, and oligoastrocytoma in adults. Curr Neurol Neurosci Rep. 2004 May;4(3):211–7. doi: 10.1007/s11910-004-0041-5. [DOI] [PubMed] [Google Scholar]
  • 3.Wen PY, Kesari S. Malignant gliomas. Curr Neurol Neurosci Rep. 2004 May;4(3):218–27. doi: 10.1007/s11910-004-0042-4. [DOI] [PubMed] [Google Scholar]
  • 4.Ataman F, Poortmans P, Stupp R, Fisher B, Mirimanoff RO. Quality assurance of the EORTC 26981/22981; NCIC CE3 intergroup trial on radiotherapy with or without temozolomide for newly-diagnosed glioblastoma multiforme: the individual case review. Eur J Cancer. 2004 Jul;40(11):1724–30. doi: 10.1016/j.ejca.2004.03.026. [DOI] [PubMed] [Google Scholar]
  • 5.Stupp R, Dietrich PY, Ostermann KS, Pica A, Maillard I, Maeder P, Meuli R, Janzer R, Pizzolato G, Miralbell R, Porchet F, Regli L, de Tribolet N, Mirimanoff RO, Leyvraz S. Promising survival for patients with newly diagnosed glioblastoma multiforme treated with concomitant radiation plus temozolomide followed by adjuvant temozolomide. J Clin Oncol. 2002 Mar 1;20(5):1375–82. doi: 10.1200/JCO.2002.20.5.1375. [DOI] [PubMed] [Google Scholar]
  • 6.Prins RM, Liau LM. Cellular immunity and immunotherapy of brain tumors. Front Biosci. 2004 Sep 1;9:3124–36. doi: 10.2741/1465. [DOI] [PubMed] [Google Scholar]
  • 7.Walker PR, Calzascia T, De Tribolet N, Dietrich PY. T-cell immune responses in the brain and their relevance for cerebral malignancies. Brain Res Brain Res Rev. 2003 May;42(2):97–122. doi: 10.1016/s0165-0173(03)00141-3. [DOI] [PubMed] [Google Scholar]
  • 8.Medawar PB. Immunity to homologous grafted skin; the fate of skin homografts transplanted to the brain, to subcutaneous tissue, and to the anterior chamber of the eye. Br J Exp Pathol. 1948 Feb;29(1):58–69. [PMC free article] [PubMed] [Google Scholar]
  • 9.Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004 Aug;21(2):137–48. doi: 10.1016/j.immuni.2004.07.017. [DOI] [PubMed] [Google Scholar]
  • 10.Engelhardt B, Ransohoff RM. The ins and outs of T-lymphocyte trafficking to the CNS: anatomical sites and molecular mechanisms. Trends Immunol. 2005 Sep;26(9):485–95. doi: 10.1016/j.it.2005.07.004. [DOI] [PubMed] [Google Scholar]
  • 11.Walker PR, Calzascia T, Dietrich PY. All in the head: obstacles for immune rejection of brain tumours. Immunology. 2002 Sep;107(1):28–38. doi: 10.1046/j.1365-2567.2002.01507.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci. 2006 Jan;7(1):41–53. doi: 10.1038/nrn1824. [DOI] [PubMed] [Google Scholar]
  • 13.Bazzoni G. Endothelial tight junctions: permeable barriers of the vessel wall. Thromb Haemost. 2006 Jan;95(1):36–42. [PubMed] [Google Scholar]
  • 14.Engelhardt B. The blood-central nervous system barriers actively control immune cell entry into the central nervous system. Curr Pharm Des. 2008;14(16):1555–65. doi: 10.2174/138161208784705432. [DOI] [PubMed] [Google Scholar]
  • 15.Neuwelt E, Abbott NJ, Abrey L, Banks WA, Blakley B, Davis T, Engelhardt B, Grammas P, Nedergaard M, Nutt J, Pardridge W, Rosenberg GA, Smith Q, Drewes LR. Strategies to advance translational research into brain barriers. Lancet Neurol. 2008 Jan;7(1):84–96. doi: 10.1016/S1474-4422(07)70326-5. [DOI] [PubMed] [Google Scholar]
  • 16.Nase G, Helm PJ, Enger R, Ottersen OP. Water entry into astrocytes during brain edema formation. GLIA. 2008 Jun;56(8):895–902. doi: 10.1002/glia.20664. [DOI] [PubMed] [Google Scholar]
  • 17.Polavarapu R, Gongora MC, Yi H, Ranganthan S, Lawrence DA, Strickland D, Yepes M. Tissue-type plasminogen activator-mediated shedding of astrocytic low-density lipoprotein receptor-related protein increases the permeability of the neurovascular unit. Blood. 2007 Apr 15;109(8):3270–8. doi: 10.1182/blood-2006-08-043125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Davies DC. Blood-brain barrier breakdown in septic encephalopathy and brain tumours. J Anat. 2002 Jun;200(6):639–46. doi: 10.1046/j.1469-7580.2002.00065.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Rascher G, Fischmann A, Kroger S, Duffner F, Grote EH, Wolburg H. Extracellular matrix and the blood-brain barrier in glioblastoma multiforme: spatial segregation of tenascin and agrin. Acta Neuropathol. 2002 Jul;104(1):85–91. doi: 10.1007/s00401-002-0524-x. [DOI] [PubMed] [Google Scholar]
  • 20.Arismendi-Morillo G, Castellano A. Tumoral micro-blood vessels and vascular microenvironment in human astrocytic tumors. A transmission electron microscopy study. J Neurooncol. 2005 Jul;73(3):211–7. doi: 10.1007/s11060-004-5674-3. [DOI] [PubMed] [Google Scholar]
  • 21.Galea I, Bernardes-Silva M, Forse PA, van Rooijen N, Liblau RS, Perry VH. An antigen-specific pathway for CD8 T cells across the blood-brain barrier. J Exp Med. 2007 Sep 3;204(9):2023–30. doi: 10.1084/jem.20070064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Dunn GP, Dunn IF, Curry WT. Focus on TILs: prognostic significance of tumor infiltrating lymphocytes in human glioma. Cancer Immun. 2007;7:12. [PMC free article] [PubMed] [Google Scholar]
  • 23.Flugel A, Labeur MS, Grasbon-Frodl EM, Kreutzberg GW, Graeber MB. Microglia only weakly present glioma antigen to cytotoxic T cells. Int J Dev Neurosci. 1999 Aug;17(5–6):547–56. doi: 10.1016/s0736-5748(99)00020-9. [DOI] [PubMed] [Google Scholar]
  • 24.Krakowski ML, Owens T. The central nervous system environment controls effector CD4+ T cell cytokine profile in experimental allergic encephalomyelitis. Eur J Immunol. 1997 Nov;27(11):2840–7. doi: 10.1002/eji.1830271115. [DOI] [PubMed] [Google Scholar]
  • 25.Carson MJ, Reffly CR, Sutcliffe JG, Lo D. Mature microglia resemble immature antigen-presenting cells. GLIA. 1998 Jan;22(1):72–85. doi: 10.1002/(sici)1098-1136(199801)22:1<72::aid-glia7>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
  • 26.Perry VH. A revised view of the central nervous system microenvironment and major histocompatibility complex class II antigen presentation. [Review] [74 refs] J Neuroimmunol. 1998 Oct 1;90(2):113–21. doi: 10.1016/s0165-5728(98)00145-3. [DOI] [PubMed] [Google Scholar]
  • 27.Aloisi F. Immune function of microglia. GLIA. 2001 Nov;36(2):165–79. doi: 10.1002/glia.1106. [DOI] [PubMed] [Google Scholar]
  • 28.Olson JK, Girvin AM, Miller SD. Direct activation of innate and antigen-presenting functions of microglia following infection with Theiler’s virus. J Virol. 2001 Oct;75(20):9780–9. doi: 10.1128/JVI.75.20.9780-9789.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Badie B, Schartner J. Role of microglia in glioma biology. Microsc Res Tech. 2001 Jul 15;54(2):106–13. doi: 10.1002/jemt.1125. [DOI] [PubMed] [Google Scholar]
  • 30.Santambrogio L, Belyanskaya SL, Fischer FR, Cipriani B, Brosnan CF, Ricciardi-Castagnoli P, Stern LJ, Strominger JL, Riese R. Developmental plasticity of CNS microglia. Proc Natl Acad Sci USA. 2001 May 22;98(11):6295–300. doi: 10.1073/pnas.111152498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Becher B, Prat A, Antel JP. Brain-immune connection: immuno-regulatory properties of CNS-resident cells. GLIA. 2000 Feb 15;29(4):293–304. [PubMed] [Google Scholar]
  • 32.Ford AL, Goodsall AL, Hickey WF, Sedgwick JD. Normal adult ramified microglia separated from other central nervous system macrophages by flow cytometric sorting. Phenotypic differences defined and direct ex vivo antigen presentation to myelin basic protein-reactive CD4+ T cells compared. J Immunol. 1995 May 1;154(9):4309–21. [PubMed] [Google Scholar]
  • 33.Fischer HG, Bonifas U, Reichmann G. Phenotype and functions of brain dendritic cells emerging during chronic infection of mice with Toxoplasma gondii. J Immunol. 2000 May 1;164(9):4826–34. doi: 10.4049/jimmunol.164.9.4826. [DOI] [PubMed] [Google Scholar]
  • 34.Fischer HG, Reichmann G. Brain dendritic cells and macrophages/microglia in central nervous system inflammation. J Immunol. 2001;166:2717–26. doi: 10.4049/jimmunol.166.4.2717. [DOI] [PubMed] [Google Scholar]
  • 35.Schartner JM, Hagar AR, Van HM, Zhang L, Nadkarni N, Badie B. Impaired capacity for up-regulation of MHC class II in tumor-associated microglia. GLIA. 2005 Sep;51(4):279–85. doi: 10.1002/glia.20201. [DOI] [PubMed] [Google Scholar]
  • 36.Miller SD, McMahon EJ, Schreiner B, Bailey SL. Antigen presentation in the CNS by myeloid dendritic cells drives progression of relapsing experimental autoimmune encephalomyelitis. Ann N Y Acad Sci. 2007 Apr;1103:179–91. doi: 10.1196/annals.1394.023. [DOI] [PubMed] [Google Scholar]
  • 37.Bailey-Bucktrout SL, Caulkins SC, Goings G, Fischer JA, Dzionek A, Miller SD. Cutting edge: central nervous system plasmacytoid dendritic cells regulate the severity of relapsing experimental autoimmune encephalomyelitis. J Immunol. 2008 May 15;180(10):6457–61. doi: 10.4049/jimmunol.180.10.6457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.de Vos AF, van Meurs M, Brok HP, Boven LA, Hintzen RQ, van der Valk PV, Ravid R, Rensing S, Boon L, ‘t Hart BA, Laman JD. Transfer of central nervous system autoantigens and presentation in secondary lymphoid organs. J Immunol. 2002 Nov 15;169(10):5415–23. doi: 10.4049/jimmunol.169.10.5415. [DOI] [PubMed] [Google Scholar]
  • 39.Kuwashima N, Nishimura F, Eguchi J, Sato H, Hatano M, Tsugawa T, Sakaida T, Dusak JE, Fellows-Mayle WK, Papworth GD, Watkins SC, Gambotto A, Pollack IF, Storkus WJ, Okada H. Delivery of dendritic cells engineered to secrete IFN-{alpha} into central nervous system tumors enhances the efficacy of peripheral tumor cell vaccines: dependence on apoptotic pathways. J Immunol. 2005 Aug 15;175(4):2730–40. doi: 10.4049/jimmunol.175.4.2730. [DOI] [PubMed] [Google Scholar]
  • 40.Okada N, Mori N, Koretomo R, Okada Y, Nakayama T, Yoshie O, Mizuguchi H, Hayakawa T, Nakagawa S, Mayumi T, Fujita T, Yamamoto A. Augmentation of the migratory ability of DC-based vaccine into regional lymph nodes by efficient CCR7 gene transduction. Gene Ther. 2005 Jan;12(2):129–39. doi: 10.1038/sj.gt.3302358. [DOI] [PubMed] [Google Scholar]
  • 41.Gordon LB, Nolan SC, Cserr HF, Knopf PM, Harling-Berg CJ. Growth of P511 mastocytoma cells in BALB/c mouse brain elicits CTL response without tumor elimination. J Immunol. 1997 Aug 15;159:2399–408. [PubMed] [Google Scholar]
  • 42.Walker PR, Calzascia T, Schnuriger V, Scamuffa N, Saas P, de Tribolet N, Dietrich PY. The brain parenchyma is permissive for full antitumor CTL effector function, even in the absence of CD4 T cells. J Immunol. 2000 Sep 15;165(6):3128–35. doi: 10.4049/jimmunol.165.6.3128. [DOI] [PubMed] [Google Scholar]
  • 43.Weller RO, Djuanda E, Yow HY, Carare RO. Lymphatic drainage of the brain and the pathophysiology of neurological disease. Acta Neuropathol. 2009;117( 1):1–14. doi: 10.1007/s00401-008-0457-0. [DOI] [PubMed] [Google Scholar]
  • 44.Engelhardt B. Molecular mechanisms involved in T cell migration across the blood-brain barrier. J Neural Transm. 2006 Apr;113(4):477–85. doi: 10.1007/s00702-005-0409-y. [DOI] [PubMed] [Google Scholar]
  • 45.Sasaki K, Zhu X, Vasquez C, Nishimura F, Dusak JE, Huang J, Fujita M, Wesa A, Potter DM, Walker PR, Storkus WJ, Okada H. Preferential expression of very late antigen-4 on type 1 CTL cells plays a critical role in trafficking into central nervous system tumors. Cancer Res. 2007 Jul 1;67(13):6451–8. doi: 10.1158/0008-5472.CAN-06-3280. [DOI] [PubMed] [Google Scholar]
  • 46.Calzascia T, Masson F, Di Berardino-Besson W, Contassot E, Wilmotte R, Aurrand-Lions M, Ruegg C, Dietrich PY, Walker PR. Homing phenotypes of tumor-specific CD8 T cells are predetermined at the tumor site by crosspresenting APCs. Immunity. 2005 Feb;22(2):175–84. doi: 10.1016/j.immuni.2004.12.008. [DOI] [PubMed] [Google Scholar]
  • 47.Nishimura F, Dusak JE, Eguchi J, Zhu X, Gambotto A, Storkus WJ, Okada H. Adoptive transfer of type 1 CTL mediates effective anti-central nervous system tumor response: critical roles of IFN-inducible protein-10. Cancer Res. 2006 Apr 15;66(8):4478–87. doi: 10.1158/0008-5472.CAN-05-3825. [DOI] [PubMed] [Google Scholar]
  • 48.Zhu X, Nishimura F, Sasaki K, Fujita M, Dusak JE, Eguchi J, Fellows-Mayle W, Storkus WJ, Walker PR, Salazar AM, Okada H. Toll like receptor-3 ligand poly-ICLC promotes the efficacy of peripheral vaccinations with tumor antigen-derived peptide epitopes in murine CNS tumor models. J Transl Med. 2007;5:10. doi: 10.1186/1479-5876-5-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Fathallah-Shaykh HM, Gao W, Cho M, Herrera MA. Priming in the brain, an immunologically privileged organ, elicits anti-tumor immunity. Int J Cancer. 1998 Jan 19;75(2):266–76. doi: 10.1002/(sici)1097-0215(19980119)75:2<266::aid-ijc16>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
  • 50.Holladay FP, Choudhuri R, Heitz T, Wood GW. Generation of cytotoxic immune responses during the progression of a rat glioma. J Neurosurg. 1994 Jan;80( 1):90–6. doi: 10.3171/jns.1994.80.1.0090. [DOI] [PubMed] [Google Scholar]
  • 51.Ridley A. Immunological reactions to gliomas. Mod Trends Neurol. 1975;6:249–66. [PubMed] [Google Scholar]
  • 52.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000 Jan 7;100(1):57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
  • 53.Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002 Dec 19;420(6917):860–7. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Liotta LA, Kohn EC. The microenvironment of the tumour-host interface. Nature. 2001 May 17;411(6835):375–9. doi: 10.1038/35077241. [DOI] [PubMed] [Google Scholar]
  • 55.Brooks WH, Netsky MG, Normansell DE, Horwitz DA. Depressed cell-mediated immunity in patients with primary intracranial tumors. Characterization of a humoral immunosuppressive factor. J Exp Med. 1972 Dec 1;136(6):1631–47. doi: 10.1084/jem.136.6.1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Brooks WH, Caldwell HD, Mortara RH. Immune responses in patients with gliomas. Surg Neurol. 1974 Nov;2(6):419–23. [PubMed] [Google Scholar]
  • 57.Brooks WH, Latta RB, Mahaley MS, Roszman TL, Dudka L, Skaggs C. Immunobiology of primary intracranial tumors. Part 5: Correlation of a lymphocyte index and clinical status. J Neurosurg. 1981 Mar;54(3):331–7. doi: 10.3171/jns.1981.54.3.0331. [DOI] [PubMed] [Google Scholar]
  • 58.Brooks WH, Roszman TL, Rogers AS. Impairment of rosette-forming T lymphocytes in patients with primary intracranial tumors. Cancer. 1976 Apr;37(4):1869–73. doi: 10.1002/1097-0142(197604)37:4<1869::aid-cncr2820370435>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
  • 59.Roszman TL, Brooks WH, Elliott LH. Immunobiology of primary intracranial tumors. VI. Suppressor cell function and lectin-binding lymphocyte subpopulations in patients with cerebral tumors. Cancer. 1982 Oct 1;50(7):1273–9. doi: 10.1002/1097-0142(19821001)50:7<1273::aid-cncr2820500709>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
  • 60.Elliott LH, Brooks WH, Roszman TL. Cytokinetic basis for the impaired activation of lymphocytes from patients with primary intracranial tumors. J Immunol. 1984 Mar;132(3):1208–15. [PubMed] [Google Scholar]
  • 61.Roszman TL, Brooks WH, Steele C, Elliott LH. Pokeweed mitogen-induced immunoglobulin secretion by peripheral blood lymphocytes from patients with primary intracranial tumors. Characterization of T helper and B cell function. J Immunol. 1985 Mar;134(3):1545–50. [PubMed] [Google Scholar]
  • 62.Elliott LH, Brooks WH, Roszman TL. Activation of immunoregulatory lymphocytes obtained from patients with malignant gliomas. J Neurosurg. 1987 Aug;67(2):231–6. doi: 10.3171/jns.1987.67.2.0231. [DOI] [PubMed] [Google Scholar]
  • 63.Ashkenazi E, Deutsch M, Tirosh R, Weinreb A, Tsukerman A, Brodie C. A selective impairment of the IL-2 system in lymphocytes of patients with glioblastomas: increased level of soluble IL-2R and reduced protein tyrosine phosphorylation. Neuroimmunomodulation. 1997 Jan;4(1):49–56. doi: 10.1159/000097315. [DOI] [PubMed] [Google Scholar]
  • 64.Morford LA, Elliott LH, Carlson SL, Brooks WH, Roszman TL. T cell receptor-mediated signaling is defective in T cells obtained from patients with primary intracranial tumors. J Immunol. 1997 Nov 1;159(9):4415_25. [PubMed] [Google Scholar]
  • 65.Roussel E, Gingras MC, Grimm EA, Bruner JM, Moser RP. Predominance of a type 2 intratumoural immune response in fresh tumour-infiltrating lymphocytes from human gliomas. Clin Exp Immunol. 1996 Aug;105(2):344–52. doi: 10.1046/j.1365-2249.1996.d01-753.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Bogdan C, Nathan C. Modulation of macrophage function by transforming growth factor beta, interleukin-4, and interleukin-10. Ann N Y Acad Sci. 1993 Jun 23;685:713–39. doi: 10.1111/j.1749-6632.1993.tb35934.x. [DOI] [PubMed] [Google Scholar]
  • 67.Hu S, Chao CC, Ehrlich LC, Sheng WS, Sutton RL, Rockswold GL, Peterson PK. Inhibition of microglial cell RANTES production by IL-10 and TGF-beta. J Leukoc Biol. 1999 Jun;65(6):815–21. doi: 10.1002/jlb.65.6.815. [DOI] [PubMed] [Google Scholar]
  • 68.Vincent VA, Tilders FJ, Van Dam AM. Inhibition of endotoxin-induced nitric oxide synthase production in microglial cells by the presence of astroglial cells: a role for transforming growth factor beta. GLIA. 1997 Mar;19(3):190–8. doi: 10.1002/(sici)1098-1136(199703)19:3<190::aid-glia2>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
  • 69.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–52. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
  • 70.Fontana A, Hengartner H, de Tribolet N, Weber E. Glioblastoma cells release interleukin 1 and factors inhibiting interleukin 2-mediated effects. J Immunol. 1984 Apr;132(4):1837–44. [PubMed] [Google Scholar]
  • 71.de Martin R, Haendler B, Hofer-Warbinek R, Gaugitsch H, Wrann M, Schlusener H, Seifert JM, Bodmer S, Fontana A, Hofer E. Complementary DNA for human glioblastoma-derived T cell suppressor factor, a novel member of the transforming growth factor-beta gene family. EMBO J. 1987 Dec l;6(12):3673–7. doi: 10.1002/j.1460-2075.1987.tb02700.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wrann M, Bodmer S, de Martin R, Siepl C, Hofer-Warbinek R, Frei K, Hofer E, Fontana A. T cell suppressor factor from human glioblastoma cells is a 12.5-kd protein closely related to transforming growth factor-beta. EMBO J. 1987 Jun;6(6):1633–6. doi: 10.1002/j.1460-2075.1987.tb02411.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Siepl C, Bodmer S, Frei K, MacDonald HR, de MR, Hofer E, Fontana A. The glioblastoma-derived T cell suppressor factor/transforming growth factor-beta 2 inhibits T cell growth without affecting the interaction of interleukin 2 with its receptor. Eur J Immunol. 1988 Apr;18(4):593–600. doi: 10.1002/eji.1830180416. [DOI] [PubMed] [Google Scholar]
  • 74.Bodmer S, Huber D, Held I, Fontana A. Human glioblastoma cell derived transforming growth factor-beta 2: evidence for secretion of both high and low molecular weight biologically active forms. J Neuroimmunol. 1991 Oct;34(1):33–42. doi: 10.1016/0165-5728(91)90096-p. [DOI] [PubMed] [Google Scholar]
  • 75.Bodmer S, Strommer K, Frei K, Siepl C, de Tribolet N, Held I, Fontana A. Immunosuppression and transforming growth factor-beta in glioblastoma. Preferential production of transforming growth factor-beta 2. J Immunol. 1989 Nov 15;143(10):3222–9. [PubMed] [Google Scholar]
  • 76.Kuppner MC, Hamou MF, Sawamura Y, Bodmer S, de Tribolet N. Inhibition of lymphocyte function by glioblastoma-derived transforming growth factor beta 2. J Neurosurg. 1989 Aug;71(2):211–7. doi: 10.3171/jns.1989.71.2.0211. [DOI] [PubMed] [Google Scholar]
  • 77.Black KL, Chen K, Becker DP, Merrill JE. Inflammatory leukocytes associated with increased immunosuppression by glioblastoma. J Neurosurg. 1992 Jul;77( 1):120–6. doi: 10.3171/jns.1992.77.1.0120. [DOI] [PubMed] [Google Scholar]
  • 78.Constam DB, Philipp J, Malipiero UV, ten Dijke P, Schachner M, Fontana A. Differential expression of transforming growth factor-beta 1, -beta 2, and -beta 3 by glioblastoma cells, astrocytes, and microglia. J Immunol. 1992 Mar 1;148(5):1404–10. [PubMed] [Google Scholar]
  • 79.Olofsson A, Miyazono K, Kanzaki T, Colosetti P, Engstrom U, Heldin CH. Transforming growth factor-beta 1, -beta 2, and -beta 3 secreted by a human glioblastoma cell line. Identification of small and different forms of large latent complexes. J Biol Chem. 1992 Sep 25;267(27):19482–8. [PubMed] [Google Scholar]
  • 80.Sasaki A, Naganuma H, Satoh E, Nagasaka M, Isoe S, Nakano S, Nukui H. Secretion of transforming growth factor-beta 1 and -beta 2 by malignant glioma cells. Neurol Med Chir (Tokyo) 1995 Jul;35(7):423–30. doi: 10.2176/nmc.35.423. [DOI] [PubMed] [Google Scholar]
  • 81.Weller M, Constam DB, Malipiero U, Fontana A. Transforming growth factor-beta 2 induces apoptosis of murine T cell clones without down-regulating bcl-2 mRNA expression. Eur J Immunol. 1994 Jun;24(6):1293–300. doi: 10.1002/eji.1830240608. [DOI] [PubMed] [Google Scholar]
  • 82.Castelli MG, Chiabrando C, Fanelli R, Martelli L, Butti G, Gaetani P, Paoletti P. Prostaglandin and thromboxane synthesis by human intracranial tumors. Cancer Res. 1989 Mar 15;49(6):1505–8. [PubMed] [Google Scholar]
  • 83.Couldwell WT, Dore-Duffy P, Apuzzo ML, Antel JP. Malignant glioma modulation of immune function: relative contribution of different soluble factors. J Neuroimmunol. 1991 Aug;33(2):89–96. doi: 10.1016/0165-5728(91)90052-9. [DOI] [PubMed] [Google Scholar]
  • 84.Nitta T, Hishii M, Sato K, Okumura K. Selective expression of interleukin-10 gene within glioblastoma multiforme. Brain Res. 1994 Jun 27;649(1–2):122–8. doi: 10.1016/0006-8993(94)91055-3. [DOI] [PubMed] [Google Scholar]
  • 85.Hishii M, Nitta T, Ishida H, Ebato M, Kurosu A, Yagita H, Sato K, Okumura K. Human glioma-derived interleukin-10 inhibits antitumor immune responses in vitro. Neurosurgery. 1995 Dec;37(6):1160–6. doi: 10.1227/00006123-199512000-00016. [DOI] [PubMed] [Google Scholar]
  • 86.Huettner C, Czub S, Kerkau S, Roggendorf W, Tonn JC. Interleukin 10 is expressed in human gliomas in vivo and increases glioma cell proliferation and motility in vitro. Anticancer Res. 1997 Sep;17(5A):3217–24. [PubMed] [Google Scholar]
  • 87.Huettner C, Paulus W, Roggendorf W. Messenger RNA expression of the immunosuppressive cytokine IL-10 in human gliomas. Am J Pathol. 1995 Feb;146(2):317–22. [PMC free article] [PubMed] [Google Scholar]
  • 88.Wikstrand CJ, Fredman P, Svennerholm L, Bigner DD. Detection of glioma-associated gangliosides GM2, GD2, GD3, 3′-isoLM1 3′,6′-isoLD1 in central nervous system tumors in vitro and in vivo using epitope-defined monoclonal antibodies. Prog Brain Res. 1994;101:213–23. doi: 10.1016/s0079-6123(08)61951-2. [DOI] [PubMed] [Google Scholar]
  • 89.Fredman P, Mansson JE, Dellheden B, Bostrom K, von HH. Expression of the GM1-species, [NeuN]-GM1, in a case of human glioma. Neurochem Res. 1999 Feb;24(2):275–9. doi: 10.1023/a:1022570222876. [DOI] [PubMed] [Google Scholar]
  • 90.Kawai K, Takahashi H, Watarai S, Ishizu H, Fukai K, Tanabe Y, Nose S, Kuroda S. Occurrence of ganglioside GD3 in neoplastic astrocytes. An immunocytochemical study in humans. Virchows Arch. 1999 Mar;434(3):201–5. doi: 10.1007/s004280050328. [DOI] [PubMed] [Google Scholar]
  • 91.Smyth MJ, Strobl SL, Young HA, Ortaldo JR, Ochoa AC. Regulation of lymphokine-activated killer activity and pore-forming protein gene expression in human peripheral blood CD8+ T lymphocytes. Inhibition by transforming growth factor-beta. J Immunol. 1991 May 15;146(10):3289–97. [PubMed] [Google Scholar]
  • 92.Inge TH, McCoy KM, Susskind BM, Barrett SK, Zhao G, Bear HD. Immunomodulatory effects of transforming growth factor-beta on T lymphocytes. Induction of CD8 expression in the CTLL-2 cell line and in normal thymocytes. J Immunol. 1992 Jun 15;148(12):3847–56. [PubMed] [Google Scholar]
  • 93.Gorelik L, Flavell RA. Transforming growth factor-beta in T-cell biology. Nat Rev Immunol. 2002 Jan;2(1):46–53. doi: 10.1038/nri704. [DOI] [PubMed] [Google Scholar]
  • 94.Jachimczak P, Bogdahn U, Schneider J, Behl C, Meixensberger J, Apfel R, Dorries R, Schlingensiepen KH, Brysch W. The effect of transforming growth factor-beta 2-specific phosphorothioate-anti-sense oligodeoxynucleotides in reversing cellular immunosuppression in malignant glioma. J Neurosurg. 1993 Jun;78(6):944–51. doi: 10.3171/jns.1993.78.6.0944. [DOI] [PubMed] [Google Scholar]
  • 95.Fakhrai H, Dorigo O, Shawler DL, Lin H, Mercola D, Black KL, Royston I, Sobol RE. Eradication of established intracranial rat gliomas by transforming growth factor beta antisense gene therapy. Proc Natl Acad Sci USA. 1996 Apr;93(7):2909–14. doi: 10.1073/pnas.93.7.2909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ashley DM, Kong FM, Bigner DD, Hale LP. Endogenous expression of transforming growth factor beta1 inhibits growth and tumorigenicity and enhances Fas-mediated apoptosis in a murine high-grade glioma model. Cancer Res. 1998 Jan 15;58(2):302–9. [PubMed] [Google Scholar]
  • 97.Gomez GG, Kruse CA. Cellular and functional characterization of immunoresistant human glioma cell clones selected with alloreactive cytotoxic T lymphocytes reveals their up-regulated synthesis of biologically active TGF-beta. J Immunother. 2007 Apr;30(3):261–73. doi: 10.1097/01.cji.0000211339.81211.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Anastassiou ED, Paliogianni F, Balow JP, Yamada H, Boumpas DT. Prostaglandin E2 and other cyclic AMP-elevating agents modulate IL-2 and IL-2R alpha gene expression at multiple levels. J Immunol. 1992 May 1;148(9):2845–52. [PubMed] [Google Scholar]
  • 99.Northrop JP, Crabtree GR, Mattila PS. Negative regulation of interleukin 2 transcription by the glucocorticoid receptor. J Exp Med. 1992 May 1;175(5):1235–45. doi: 10.1084/jem.175.5.1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Roszman TL, Brooks WH, Elliott LH. Inhibition of lymphocyte responsiveness by a glial tumor cell-derived suppressive factor. J Neurosurg. 1987 Dec;67(6):874–9. doi: 10.3171/jns.1987.67.6.0874. [DOI] [PubMed] [Google Scholar]
  • 101.Zou JP, Yamamoto N, Fujii T, Takenaka H, Kobayashi M, Herrmann SH, Wolf SF, Fujiwara H, Hamaoka T. Systemic administration of rIL-12 induces complete tumor regression and protective immunity: response is correlated with a striking reversal of suppressed IFN-gamma production by anti-tumor T cells. Int Immunol. 1995 Jul;7(7):1135–45. doi: 10.1093/intimm/7.7.1135. [DOI] [PubMed] [Google Scholar]
  • 102.Ladisch S, Becker H, Ulsh L. Immunosuppression by human gangliosides: I. Relationship of carbohydrate structure to the inhibition of T cell responses. Biochim Biophys Acta. 1992 Apr 23;1125(2):180–8. doi: 10.1016/0005-2760(92)90043-u. [DOI] [PubMed] [Google Scholar]
  • 103.Whisler RL, Yates AJ. Regulation of lymphocyte responses by human gangliosides. I. Characteristics of inhibitory effects and the induction of impaired activation. J Immunol. 1980 Nov;125(5):2106–11. [PubMed] [Google Scholar]
  • 104.Ladisch S, Ulsh L, Gillard B, Wong C. Modulation of the immune response by gangliosides. Inhibition of adherent monocyte accessory function in vitro. J Clin Invest. 1984 Dec;74(6):2074–81. doi: 10.1172/JCI111631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Merritt WD, Bailey JM, Pluznik DH. Inhibition of interleukin-2-dependent cytotoxic T-lymphocyte growth by gangliosides. Cell Immunol. 1984 Nov;89(1):1–10. doi: 10.1016/0008-8749(84)90191-6. [DOI] [PubMed] [Google Scholar]
  • 106.Li R, Villacreses N, Ladisch S. Human tumor gangliosides inhibit murine immune responses in vivo. Cancer Res. 1995 Jan 15;55(2):211–4. [PubMed] [Google Scholar]
  • 107.Irani DN, Lin KI, Griffin DE. Brain-derived gangliosides regulate the cytokine production and proliferation of activated T cells. J Immunol. 1996 Nov 15;157(10):4333–40. [PubMed] [Google Scholar]
  • 108.Offner H, Thieme T, Vandenbark AA. Gangliosides induce selective modulation of CD4 from helper T lymphocytes. J Immunol. 1987 Nov 15;139(10):3295–305. [PubMed] [Google Scholar]
  • 109.Ikeda T, Nakakuma H, Shionoya H, Kawaguchi T, Yamatsu K, Takatsuki K. Ganglioside-induced inhibition of in vivo immune response in mice. Life Sci. 1992;51(11):847–51. doi: 10.1016/0024-3205(92)90612-s. [DOI] [PubMed] [Google Scholar]
  • 110.Chahlavi A, Rayman P, Richmond AL, Biswas K, Zhang R, Vogelbaum M, Tannenbaum C, Barnett G, Finke JH. Glioblastomas induce T-lymphocyte death by two distinct pathways involving gangliosides and CD70. Cancer Res. 2005 Jun 15;65(12):5428–38. doi: 10.1158/0008-5472.CAN-04-4395. [DOI] [PubMed] [Google Scholar]
  • 111.Rivera F, Vega-Villegas ME, Lopez-Brea MF, Marquez R. Current situation of Panitumumab, Matuzumab, Nimotuzumab and Zalutumumab. Acta Oncol. 2008;47(1):9–19. doi: 10.1080/02841860701704724. [DOI] [PubMed] [Google Scholar]
  • 112.Desbaillets I, Tada M, De Tribolet N, Diserens AC, Hamou MF, Van Meir EG. Human astrocytomas and glioblastomas express monocyte chemoattractant protein-1 (MCP-1) in vivo and in vitro. Int J Cancer. 1994 Jul 15;58(2):240–7. doi: 10.1002/ijc.2910580216. [DOI] [PubMed] [Google Scholar]
  • 113.Takeshima H, Kuratsu J, Takeya M, Yoshimura T, Ushio Y. Expression and localization of messenger RNA and protein for monocyte chemoattractant protein-1 in human malignant glioma. J Neurosurg. 1994 Jun;80(6):1056–62. doi: 10.3171/jns.1994.80.6.1056. [DOI] [PubMed] [Google Scholar]
  • 114.Brown CE, Vishwanath RP, Aguilar B, Starr R, Najbauer J, Aboody KS, Jensen MC. Tumor-derived chemokine MCP-1/CCL2 is sufficient for mediating tumor tropism of adoptively transferred T cells. J Immunol. 2007 Sep 1;179(5):3332–41. doi: 10.4049/jimmunol.179.5.3332. [DOI] [PubMed] [Google Scholar]
  • 115.Jordan JT, Sun W, Hussain SF, Deangulo G, Prabhu SS, Heimberger AB. Preferential migration of regulatory T cells mediated by glioma-secreted chemokines can be blocked with chemotherapy. Cancer Immunol Immunother. 2008 Jan;57(1):123–31. doi: 10.1007/s00262-007-0336-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Salcedo R, Ponce ML, Young HA, Wasserman K, Ward JM, Kleinman HK, Oppenheim JJ, Murphy WJ. Human endothelial cells express CCR2 and respond to MCP-1: direct role of MCP-1 in angiogenesis and tumor progression. Blood. 2000 Jul 1;96(1):34–40. [PubMed] [Google Scholar]
  • 117.Ueno T, Toi M, Saji H, Muta M, Bando H, Kuroi K, Koike M, Inadera H, Matsushima K. Significance of macrophage chemoattractant protein-1 in macrophage recruitment, angiogenesis, and survival in human breast cancer. Clin Cancer Res. 2000 Aug;6(8):3282–9. [PubMed] [Google Scholar]
  • 118.Huang R, Lin Y, Wang CC, Gano J, Lin B, Shi Q, Boynton A, Burke J, Huang RP. Connexin 43 suppresses human glioblastoma cell growth by down-regulation of monocyte chemotactic protein 1, as discovered using protein array technology. Cancer Res. 2002 May 15;62(10):2806–12. [PubMed] [Google Scholar]
  • 119.Wang JM, Chertov O, Proost P, Li JJ, Menton P, Xu L, Sozzani S, Mantovani A, Gong W, Schirrmacher V, Van Damme J, Oppenheim JJ. Purification and identification of chemokines potentially involved in kidney-specific metastasis by a murine lymphoma variant: induction of migration and NFkappaB activation. Int J Cancer. 1998 Mar 16;75(6):900–7. doi: 10.1002/(sici)1097-0215(19980316)75:6<900::aid-ijc13>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
  • 120.Nakashima E, Mukaida N, Kubota Y, Kuno K, Yasumoto K, Ichimura F, Nakanishi I, Miyasaka M, Matsushima K. Human MCAF gene transfer enhances the metastatic capacity of a mouse cachectic adenocarcinoma cell line in vivo. Pharm Res. 1995 Nov;12(11):1598–604. doi: 10.1023/A:1016276613684. [DOI] [PubMed] [Google Scholar]
  • 121.Lebrecht A, Grimm C, Lantzsch T, Ludwig E, Hefler L, Ulbrich E, Koelbl H. Monocyte chemoattractant protein-1 serum levels in patients with breast cancer. Tumour Biol. 2004 Jan;25(1–2):14–7. doi: 10.1159/000077718. [DOI] [PubMed] [Google Scholar]
  • 122.Kuratsu J, Yoshizato K, Yoshimura T, Leonard EJ, Takeshima H, Ushio Y. Quantitative study of monocyte chemoattractant protein-1 (MCP-1) in cerebrospinal fluid and cyst fluid from patients with malignant glioma. J Natl Cancer Inst. 1993 Nov 17;85(22):1836–9. doi: 10.1093/jnci/85.22.1836. [DOI] [PubMed] [Google Scholar]
  • 123.Amann B, Perabo FG, Wirger A, Hugenschmidt H, Schultze-Seemann W. Urinary levels of monocyte chemo-attractant protein-1 correlate with tumour stage and grade in patients with bladder cancer. Br J Urol. 1998 Jul;82(1):118–21. doi: 10.1046/j.1464-410x.1998.00675.x. [DOI] [PubMed] [Google Scholar]
  • 124.Valkovic T, Lucin K, Krstulja M, Dobi-Babic R, Jonjic N. Expression of monocyte chemotactic protein-1 in human invasive ductal breast cancer. Pathol Res Pract. 1998;194(5):335–40. doi: 10.1016/S0344-0338(98)80057-5. [DOI] [PubMed] [Google Scholar]
  • 125.Roth SJ, Carr MW, Springer TA. C-C chemokines, but not the C-X-C chemokines interleukin-8 and interferon-gamma inducible protein-10, stimulate transendothelial chemotaxis of T lymphocytes. Eur J Immunol. 1995 Dec;25(12):3482–8. doi: 10.1002/eji.1830251241. [DOI] [PubMed] [Google Scholar]
  • 126.Roth SJ, Diacovo TG, Brenner MB, Rosat JP, Buccola J, Morita CT, Springer TA. Transendothelial chemotaxis of human alpha/beta and gamma/delta T lymphocytes to chemokines. Eur J Immunol. 1998 Jan;28(1):104–13. doi: 10.1002/(SICI)1521-4141(199801)28:01<104::AID-IMMU104>3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
  • 127.Carr MW, Roth SJ, Luther E, Rose SS, Springer TA. Monocyte chemoattractant protein 1 acts as a T-lymphocyte chemoattractant. Proc Natl Acad Sci USA. 1994 Apr 26;91(9):3652–6. doi: 10.1073/pnas.91.9.3652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Metelitsa LS, Wu HW, Wang H, Yang Y, Warsi Z, Asgharzadeh S, Groshen S, Wilson SB, Seeger RC. Natural killer T cells infiltrate neuroblastomas expressing the chemokine CCL2. J Exp Med. 2004 May 3;199(9):1213–21. doi: 10.1084/jem.20031462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Choi C, Xu X, Oh JW, Lee SJ, Gillespie GY, Park H, Jo H, Benveniste EN. Fas-induced expression of chemokines in human glioma cells: involvement of extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase. Cancer Res. 2001 Apr 1;61(7):3084–91. [PubMed] [Google Scholar]
  • 130.Choi C, Gillespie GY, Van Wagoner NJ, Benveniste EN. Fas engagement increases expression of interleukin-6 in human glioma cells. J Neurooncol. 2002 Jan;56(1):13–9. doi: 10.1023/a:1014467626314. [DOI] [PubMed] [Google Scholar]
  • 131.Shinohara H, Yagita H, Ikawa Y, Oyaizu N. Fas drives cell cycle progression in glioma cells via extracellular signal-regulated kinase activation. Cancer Res. 2000 Mar 15;60(6):1766–72. [PubMed] [Google Scholar]
  • 132.Bennett MW, O’Connell J, O’Sullivan GC, Brady C, Roche D, Collins JK, Shanahan F. The Fas counterattack in vivo: apoptotic depletion of tumor-infiltrating lymphocytes associated with Fas ligand expression by human esophageal carcinoma. J Immunol. 1998 Jun 1;160(11):5669–75. [PubMed] [Google Scholar]
  • 133.Cefai D, Simeoni E, Ludunge KM, Driscoll R, von Segesser LK, Kappenberger L, Vassalli G. Multiply attenuated, self-inactivating lentiviral vectors efficiently transduce human coronary artery cells in vitro and rat arteries in vivo. J Mol Cell Cardiol. 2005 Feb;38(2):333–44. doi: 10.1016/j.yjmcc.2004.11.031. [DOI] [PubMed] [Google Scholar]
  • 134.Chouaib S, Asselin-Paturel C, Mami-Chouaib F, Caignard A, Blay JY. The host-tumor immune conflict: from immunosuppression to resistance and destruction. Immunol Today. 1997 Oct;18(10):493–7. doi: 10.1016/s0167-5699(97)01115-8. [DOI] [PubMed] [Google Scholar]
  • 135.Didenko VV, Ngo HN, Minchew C, Baskin DS. Apoptosis of T lymphocytes invading glioblastomas multiforme: a possible tumor defense mechanism. J Neurosurg. 2002 Mar;96(3):580–4. doi: 10.3171/jns.2002.96.3.0580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Chen JJ, Sun Y, Nabel GJ. Regulation of the proinflammatory effects of Fas ligand (CD95L) Science. 1998 Nov 27;282(5394):1714–7. doi: 10.1126/science.282.5394.1714. [DOI] [PubMed] [Google Scholar]
  • 137.Walker PR, Saas P, Dietrich PY. Role of Fas ligand (CD95L) in immune escape: the tumor cell strikes back. J Immunol. 1997 May 15;158(10):4521–4. [PubMed] [Google Scholar]
  • 138.Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T. PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci USA. 2001 Nov 20;98(24):13866–71. doi: 10.1073/pnas.231486598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Wintterle S, Schreiner B, Mitsdoerffer M, Schneider D, Chen L, Meyermann R, Weller M, Wiendl H. Expression of the B7-related molecule B7-H1 by glioma cells: a potential mechanism of immune paralysis. Cancer Res. 2003 Nov 1;63(21):7462–7. [PubMed] [Google Scholar]
  • 140.Curiel TJ, Wei S, Dong H, Alvarez X, Cheng P, Mottram P, Krzysiek R, Knutson KL, Daniel B, Zimmermann MC, David O, Burow M, Gordon A, Dhurandhar N, Myers L, Berggren R, Hemminki A, Alvarez RD, Emilie D, Curiel DT, Chen L, Zou W. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med. 2003 May;9(5):562–7. doi: 10.1038/nm863. [DOI] [PubMed] [Google Scholar]
  • 141.Brown JA, Dorfman DM, Ma FR, Sullivan EL, Munoz O, Wood CR, Greenfield EA, Freeman GJ. Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J Immunol. 2003 Feb 1;170(3):1257–66. doi: 10.4049/jimmunol.170.3.1257. [DOI] [PubMed] [Google Scholar]
  • 142.Dong C, Wilhelm D, Koopman P. Sox genes and cancer. Cytogenet Genome Res. 2004;105(2–4):442–7. doi: 10.1159/000078217. [DOI] [PubMed] [Google Scholar]
  • 143.Boehm U, Klamp T, Groot M, Howard JC. Cellular responses to interferon-gamma. Annu Rev Immunol. 1997;15:749–95. doi: 10.1146/annurev.immunol.15.1.749. [DOI] [PubMed] [Google Scholar]
  • 144.Parsa AT, Waldron JS, Fanner A, Crane CA, Parney IF, Barry JJ, Cachola KE, Murray JC, Tihan T, Jensen MC, Mischel PS, Stokoe D, Pieper RO. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2007 Jan;13(1):84–8. doi: 10.1038/nm1517. [DOI] [PubMed] [Google Scholar]
  • 145.Nakashima M, Sonoda K, Watanabe T. Inhibition of cell growth and induction of apoptotic cell death by the human tumor-associated antigen RCAS1. Nat Med. 1999 Aug;5(8):938–42. doi: 10.1038/11383. [DOI] [PubMed] [Google Scholar]
  • 146.Nakabayashi H, Nakashima M, Hara M, Toyonaga S, Yamada SM, Park KC, Shimizu K. Clinico-pathological significance of RCAS1 expression in gliomas: a potential mechanism of tumor immune escape. Cancer Lett. 2007 Feb 8;246(1–2):182–9. doi: 10.1016/j.canlet.2006.02.021. [DOI] [PubMed] [Google Scholar]
  • 147.Wiendl H, Mitsdoerffer M, Hofmeister V, Wischhusen J, Bornemann A, Meyermann R, Weiss EH, Melms A, Weller M. A functional role of HLA-G expression in human gliomas: an alternative strategy of immune escape. J Immunol. 2002 May 1;168(9):4772–80. doi: 10.4049/jimmunol.168.9.4772. [DOI] [PubMed] [Google Scholar]
  • 148.Braud VM, Allan DS, McMichael AJ. Functions of nonclassical MHC and non-MHC-encoded class I molecules. Curr Opin Immunol. 1999 Feb;11(1):100–8. doi: 10.1016/s0952-7915(99)80018-1. [DOI] [PubMed] [Google Scholar]
  • 149.Bainbridge D, Ellis S, Le Bouteiller P, Sargent I. HLA-G remains a mystery. Trends Immunol. 2001 Oct;22(10):548–52. doi: 10.1016/s1471-4906(01)02031-2. [DOI] [PubMed] [Google Scholar]
  • 150.Wischhusen J, Jung G, Radovanovic I, Beier C, Steinbach JP, Rimner A, Huang H, Schulz JB, Ohgaki H, Aguzzi A, Rammensee HG, Weller M. Identification of CD70-mediated apoptosis of immune effector cells as a novel immune escape pathway of human glioblastoma. Cancer Res. 2002 May 1;62(9):2592–9. [PubMed] [Google Scholar]
  • 151.Held-Feindt J, Mentlein R. CD70/CD27 ligand, a member of the TNF family, is expressed in human brain tumors. Int J Cancer. 2002 Mar 20;98(3):352–6. doi: 10.1002/ijc.10207. [DOI] [PubMed] [Google Scholar]
  • 152.Kobata T, Jacquot S, Kozlowski S, Agematsu K, Schlossman SF, Morimoto C. CD27-CD70 interactions regulate B-cell activation by T cells. Proc Natl Acad Sci USA. 1995 Nov 21;92(24):11249–53. doi: 10.1073/pnas.92.24.11249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Giometto B, Bozza F, Faresin F, Alessio L, Mingrino S, Tavolato B. Immune infiltrates and cytokines in gliomas. Acta Neurochir (Wien) 1996;138(1):50–6. doi: 10.1007/BF01411724. [DOI] [PubMed] [Google Scholar]
  • 154.Hussain SF, Yang D, Suki D, Aldape K, Grimm E, Heimberger AB. The role of human glioma-infiltrating microglia/macrophages in mediating antitumor immune responses. Neuro Oncol. 2006 Jul;8(3):261–79. doi: 10.1215/15228517-2006-008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Wierzba-Bobrowicz T, Kuchna I, Matyja E. Reaction of microglial cells in human astrocytomas (preliminary report) Folia Neuropathol. 1994;32(4):251–2. [PubMed] [Google Scholar]
  • 156.Graeber MB, Scheithauer BW, Kreutzberg GW. Microglia in brain tumors. GLIA. 2002 Nov;40(2):252–9. doi: 10.1002/glia.10147. [DOI] [PubMed] [Google Scholar]
  • 157.Nishie A, Ono M, Shono T, Fukushi J, Otsubo M, Onoue H, Ito Y, Inamura T, Ikezaki K, Fukui M, Iwaki T, Kuwano M. Macrophage infiltration and heme oxygenase-1 expression correlate with angiogenesis in human gliomas. Clin Cancer Res. 1999 May;5(5):1107–13. [PubMed] [Google Scholar]
  • 158.Nishie A, Masuda K, Otsubo M, Migita T, Tsuneyoshi M, Kohno K, Shuin T, Naito S, Ono M, Kuwano M. High expression of the Cap43 gene in infiltrating macrophages of human renal cell carcinomas. Clin Cancer Res. 2001 Jul;7(7):2145–51. [PubMed] [Google Scholar]
  • 159.Flatten M, Kretz A, Naumann U, Aulwurm S, Egashira K, Isenmann S, Weller M. Monocyte chemoattractant protein-1 increases microglial infiltration and aggressiveness of gliomas. Ann Neurol. 2003 Sep;54(3):388–92. doi: 10.1002/ana.10679. [DOI] [PubMed] [Google Scholar]
  • 160.Rao JS. Molecular mechanisms of glioma invasiveness: the role of proteases. Nat Rev Cancer. 2003 Jul;3(7):489–501. doi: 10.1038/nrc1121. [DOI] [PubMed] [Google Scholar]
  • 161.Watters JJ, Schartner JM, Badie B. Microglia function in brain tumors. J Neurosci Res. 2005 Aug 1;81(3):447–55. doi: 10.1002/jnr.20485. [DOI] [PubMed] [Google Scholar]
  • 162.Bettinger I, Thanos S, Paulus W. Microglia promote glioma migration. Acta Neuropathol. 2002 Apr;103( 4):351–5. doi: 10.1007/s00401-001-0472-x. [DOI] [PubMed] [Google Scholar]
  • 163.Markovic DS, Glass R, Synowitz M, Rooijen N, Kettenmann H. Microglia stimulate the invasiveness of glioma cells by increasing the activity of metallo-protease-2. J Neuropathol Exp Neurol. 2005 Sep;64(9):754–62. doi: 10.1097/01.jnen.0000178445.33972.a9. [DOI] [PubMed] [Google Scholar]
  • 164.Feed PE, Mitchell DA, Whitesides JF, Xie W, Friedman AH, Archer GE, Herndon JE, Bigner DD, Dranoff G, Sampson JH. Increased regulatory T-cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res. 2006 Mar 15;66(6):3294–302. doi: 10.1158/0008-5472.CAN-05-3773. [DOI] [PubMed] [Google Scholar]
  • 165.Andaloussi AE, Han Y, Lesniak MS. Progression of intracranial glioma disrupts thymic homeostasis and induces T-cell apoptosis in vivo. Cancer Immunol Immunother. 2008 Dec;57(12):1807–16. doi: 10.1007/s00262-008-0508-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Heimberger AB, Abou-Ghazal M, Reina-Ortiz C, Yang DS, Sun W, Qiao W, Hiraoka N, Fuller GN. Incidence and prognostic impact of FoxP3+ regulatory T cells in human gliomas. Clin Cancer Res. 2008 Aug 15;14(16):5166–72. doi: 10.1158/1078-0432.CCR-08-0320. [DOI] [PubMed] [Google Scholar]
  • 167.Mahaley MSJ, Bigner DD, Dudka LF, Wilds PR, Williams DH, Bouldin TW, Whitaker JN, Bynum JM. Immunobiology of primary intracranial tumors. Part 7: Active immunization of patients with anaplastic human glioma cells: a pilot study. J Neurosurg. 1983;59:201–7. doi: 10.3171/jns.1983.59.2.0201. [DOI] [PubMed] [Google Scholar]
  • 168.El AA, Han Y, Lesniak MS. Prolongation of survival following depletion of CD4+CD25+ regulatory T cells in mice with experimental brain tumors. J Neurosurg. 2006 Sep;105(3):430–7. doi: 10.3171/jns.2006.105.3.430. [DOI] [PubMed] [Google Scholar]
  • 169.Grauer OM, Nierkens S, Bennink E, Toonen LW, Boon L, Wesseling P, Sutmuller RP, Adema GJ. CD4+FoxP3+ regulatory T cells gradually accumulate in gliomas during tumor growth and efficiently suppress antiglioma immune responses in vivo. Int J Cancer. 2007 Jul 1;121(1):95–105. doi: 10.1002/ijc.22607. [DOI] [PubMed] [Google Scholar]
  • 170.DeVita VT, Hellstrom S, Rosenberg SA. Biologic therapy of cancer. 2. Philadelphia: J. B. Lippincott Company; 1995. [Google Scholar]
  • 171.Butler TP, Grantham FH, Gullino PM. Bulk transfer of fluid in the interstitial compartment of mammary tumors. Cancer Res. 1975 Nov;35(11 Pt 1):3084–8. [PubMed] [Google Scholar]
  • 172.Jain RK, Baxter LT. Mechanisms of heterogeneous distribution of monoclonal antibodies and other macro-molecules in tumors: significance of elevated interstitial pressure. Cancer Res. 1988 Dec 15;48(24 Pt 1):7022–32. [PubMed] [Google Scholar]
  • 173.Williams LE, Duda RB, Proffitt RT, Beatty BG, Beatty JD, Wong JY, Shively JE, Paxton RJ. Tumor uptake as a function of tumor mass: a mathematic model. J Nucl Med. 1988 Jan;29(1):103–9. [PubMed] [Google Scholar]
  • 174.Bobo RH, Laske DW, Akbasak A, Morrison PF, Dedrick RL, Oldfield EH. Convection-enhanced delivery of macromolecules in the brain. Proc Natl Acad Sci USA. 1994 Mar 15;91(6):2076–80. doi: 10.1073/pnas.91.6.2076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Ferguson S, Lesniak MS. Convection enhanced drug delivery of novel therapeutic agents to malignant brain tumors. Curr Drug Deliv. 2007 Apr;4(2):169–80. doi: 10.2174/156720107780362302. [DOI] [PubMed] [Google Scholar]
  • 176.Carlsson J, Ren ZP, Wester K, Sundberg AL, Heldin NE, Hesselager G, Persson M, Gedda L, Tolmachev V, Lundqvist H, Blomquist E, Nister M. Planning for intracavitary anti-EGFR radionuclide therapy of gliomas. Literature review and data on EGFR expression. J Neurooncol. 2006 Mar;77(1):33–45. doi: 10.1007/s11060-005-7410-z. [DOI] [PubMed] [Google Scholar]
  • 177.Shinojima N, Tada K, Shiraishi S, Kamiryo T, Kochi M, Nakamura H, Makino K, Saya H, Hirano H, Kuratsu J, Oka K, Ishimaru Y, Ushio Y. Prognostic value of epidermal growth factor receptor in patients with glioblastoma multiforme. Cancer Res. 2003 Oct 15;63(20):6962–70. [PubMed] [Google Scholar]
  • 178.Baselga J. The EGFR as a target for anticancer therapy—focus on cetuximab. Eur J Cancer. 2001 Sep;37( Suppl 4):516–22. doi: 10.1016/s0959-8049(01)00233-7. [DOI] [PubMed] [Google Scholar]
  • 179.Batra SK, Castelino-Prabhu S, Wikstrand CJ, Zhu X, Humphrey PA, Friedman HS, Bigner DD. Epidermal growth factor ligand-independent, unregulated, cell-transforming potential of a naturally occurring human mutant EGFRvIII gene. Cell Growth Differ. 1995 Oct;6(10):1251–9. [PubMed] [Google Scholar]
  • 180.Fukai J, Nishio K, Itakura T, Koizumi F. Antitumor activity of cetuximab against malignant glioma cells overexpressing EGFR deletion mutant variant III. Cancer Sci. 2008 Oct;99(10):2062–9. doi: 10.1111/j.1349-7006.2008.00945.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Belda-Iniesta C, Carpeno JC, Saenz EC, Gutierrez M, Perona R, Baron MG. Long term responses with cetuximab therapy in glioblastoma multiforme. Cancer Biol Ther. 2006 Aug;5(8):912–4. doi: 10.4161/cbt.5.8.3118. [DOI] [PubMed] [Google Scholar]
  • 182.Ramos TC, Figueredo J, Catala M, Gonzalez S, Selva JC, Cruz TM, Toledo C, Silva S, Pestano Y, Ramos M, Leonard I, Torres O, Marinello P, Perez R, Lage A. Treatment of high-grade glioma patients with the humanized anti-epidermal growth factor receptor (EGFR) antibody h-R3: report from a phase I/II trial. Cancer Biol Ther. 2006 Apr;5(4):375–9. doi: 10.4161/cbt.5.4.2522. [DOI] [PubMed] [Google Scholar]
  • 183.Rosenberg SA. Principles and practice of the biologic therapy of cancer. 3. Philadelphia: Lippincott Williams & Wilkins; 2000. [Google Scholar]
  • 184.Baechler S, Hobbs RF, Prideaux AR, Wahl RL, Sgouros G. Extension of the biological effective dose to the MIRD schema and possible implications in radionuclide therapy dosimetry. Med Phys. 2008 Mar;35(3):1123–34. doi: 10.1118/1.2836421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Zalutsky MR. Targeted radiotherapy of brain tumours. Br J Cancer. 2004 Apr 19;90(8):1469–73. doi: 10.1038/sj.bjc.6601771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Bourdon MA, Wikstrand CJ, Furthmayr H, Matthews TJ, Bigner DD. Human glioma-mesenchymal extracellular matrix antigen defined by monoclonal antibody. Cancer Res. 1983 Jun;43(6):2796–805. [PubMed] [Google Scholar]
  • 187.Zalutsky MR, Moseley RP, Coakham HB, Coleman RE, Bigner DD. Pharmacokinetics and tumor localization of 1311-labeled antitenascin monoclonal antibody 81C6 in patients with gliomas and other intracranial malignancies. Cancer Res. 1989 May 15;49(10):2807–13. [PubMed] [Google Scholar]
  • 188.Aarts F, Bleichrodt RP, Oyen WJ, Boerman OC. Intracavitary radioimmunotherapy to treat solid tumors. Cancer Biother Radiopharm. 2008 Feb;23(1):92–107. doi: 10.1089/cbr.2007.0412. [DOI] [PubMed] [Google Scholar]
  • 189.Bigner DD, Brown MT, Friedman AH, Coleman RE, Akabani G, Friedman HS, Thorstad WL, McLendon RE, Bigner SH, Zhao XG, Pegram CN, Wikstrand CJ, Herndon JE, Vick NA, Paleologos N, Cokgorl Provenzale JM, Zalutsky MR. Iodine-131-labeled antitenascin monoclonal antibody 81C6 treatment of patients with recurrent malignant gliomas: phase I trial results. J Clin Oncol. 1998 Jun;16(6):2202–12. doi: 10.1200/JCO.1998.16.6.2202. [DOI] [PubMed] [Google Scholar]
  • 190.Cokgor I, Akabani G, Kuan CT, Friedman HS, Friedman AH, Coleman RE, McLendon RE, Bigner SH, Zhao XG, Garcia-Turner AM, Pegram CN, Wikstrand CJ, Shafman TD, Herndon JE, Provenzale JM, Zalutsky MR, Bigner DD. Phase I trial results of iodine-131-labeled antitenascin monoclonal antibody 81C6 treatment of patients with newly diagnosed malignant gliomas. J Clin Oncol. 2000 Nov 15;18(22):3862–72. doi: 10.1200/JCO.2000.18.22.3862. [DOI] [PubMed] [Google Scholar]
  • 191.Akabani G, Reardon DA, Coleman RE, Wong TZ, Metzler SD, Bowsher JE, Barboriak DP, Provenzale JM, Greer KL, DeLong D, Friedman HS, Friedman AH, Zhao XG, Pegram CN, McLendon RE, Bigner DD, Zalutsky MR. Dosimetry and radiographic analysis of 1311-labeled antitenascin 81C6 murine monoclonal antibody in newly diagnosed patients with malignant gliomas: a phase II study. J Nucl Med. 2005 Jun;46(6):1042–51. [PubMed] [Google Scholar]
  • 192.Reardon DA, Akabani G, Coleman RE, Friedman AH, Friedman HS, Herndon JE, Cokgor I, McLendon RE, Pegram CN, Provenzale JM, Quinn JA, Rich JN, Regalado LV, Sampson JH, Shafman TD, Wikstrand CJ, Wong TZ, Zhao XG, Zalutsky MR, Bigner DD. Phase II trial of murine (131)I-labeled antitenascin monoclonal antibody 81C6 administered into surgically created resection cavities of patients with newly diagnosed malignant gliomas. J Clin Oncol. 2002 Mar 1;20(5):1389–97. doi: 10.1200/JCO.2002.20.5.1389. [DOI] [PubMed] [Google Scholar]
  • 193.Zalutsky MR, Reardon DA, Akabani G, Coleman RE, Friedman AH, Friedman HS, McLendon RE, Wong TZ, Bigner DD. Clinical experience with alpha-particle emitting 211 At: treatment of recurrent brain tumor patients with 211At-labeled chimeric antitenascin monoclonal antibody 81C6. J Nucl Med. 2008 Jan;49(1):30–8. doi: 10.2967/jnumed.107.046938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Riva P, Arista A, Franceschi G, Frattarelli M, Sturiale C, Riva N, Casi M, Rossitti R. Local treatment of malignant gliomas by direct infusion of specific monoclonal antibodies labeled with 1311: comparison of the results obtained in recurrent and newly diagnosed tumors. Cancer Res. 1995 Dec 1;55(23 Suppl):5952s–6s. [PubMed] [Google Scholar]
  • 195.Riva P, Arista A, Tison V, Sturiale C, Franceschi G, Spinelli A, Riva N, Casi M, Moscatelli G, Frattarelli M. Intralesional radioimmunotherapy of malignant gliomas. An effective treatment in recurrent tumors. Cancer. 1994 Feb 1;73(3 Suppl):1076–82. doi: 10.1002/1097-0142(19940201)73:3+<1076::aid-cncr2820731347>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
  • 196.Riva P, Franceschi G, Frattarelli M, Riva N, Guiducci G, Cremonini AM, Giuliani G, Casi M, Gentile R, Jekunen AA, Kairemo KJ. 1311 radioconjugated antibodies for the locoregional radioimmunotherapy of high-grade malignant glioma—phase I and II study. Acta Oncol. 1999;38(3):351–9. doi: 10.1080/028418699431438. [DOI] [PubMed] [Google Scholar]
  • 197.Grana C, Chinol M, Robertson C, Mazzetta C, Bartolomei M, De CC, Fiorenza M, Gatti M, Caliceti P, Paganelli G. Pretargeted adjuvant radioimmunotherapy with yttrium-90-biotin in malignant glioma patients: a pilot study. Br J Cancer. 2002 Jan 21;86(2):207–12. doi: 10.1038/sj.bjc.6600047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Cremonesi M, Ferrari M, Chinol M, Stabin MG, Grana C, Prisco G, Robertson C, Tosi G, Paganelli G. Three-step radioimmunotherapy with yttrium-90 biotin: dosimetry and pharmacokinetics in cancer patients. Eur J Nucl Med. 1999 Feb;26(2):110–20. doi: 10.1007/s002590050366. [DOI] [PubMed] [Google Scholar]
  • 199.Paganelli G, Grana C, Chinol M, Cremonesi M, De CC, de BF, Robertson C, Zurrida S, Casadio C, Zoboli S, Siccardi AG, Veronesi U. Antibody-guided three-step therapy for high grade glioma with yttrium-90 biotin. Eur J Nucl Med. 1999 Apr;26(4):348–57. doi: 10.1007/s002590050397. [DOI] [PubMed] [Google Scholar]
  • 200.Paganelli G, Bartolomei M, Ferrari M, Cremonesi M, Broggi G, Maira G, Sturiale C, Grana C, Prisco G, Gatti M, Caliceti P, Chinol M. Pre-targeted locoregional radioimmunotherapy with 90Y-biotin in glioma patients: phase I study and preliminary therapeutic results. Cancer Biother Radiopharm. 2001 Jun;16(3):227–35. doi: 10.1089/10849780152389410. [DOI] [PubMed] [Google Scholar]
  • 201.Bartolomei M, Mazzetta C, Handkiewicz-Junak D, Bodei L, Rocca P, Grana C, Maira G, Sturiale C, Villa G, Paganelli G. Combined treatment of glioblastoma patients with locoregional pre-targeted 90Y-biotin radioimmunotherapy and temozolomide. QJ Nucl Med Mol Imaging. 2004 Sep;48(3):220–8. [PubMed] [Google Scholar]
  • 202.Patel SJ, Shapiro WR, Laske DW, Jensen RL, Asher AL, Wessels BW, Carpenter SP, Shan JS. Safety and feasibility of convection-enhanced delivery of Cotara for the treatment of malignant glioma: initial experience in 51 patients. Neurosurgery. 2005 Jun;56(6):1243–52. doi: 10.1227/01.neu.0000159649.71890.30. [DOI] [PubMed] [Google Scholar]
  • 203.Shapiro WR, Carpenter SP, Roberts K, Shan JS. (131)I-chTNT-1/B mAb: tumour necrosis therapy for malignant astrocytic glioma. Expert Opin Biol Ther. 2006 May;6(5):539–45. doi: 10.1517/14712598.6.5.539. [DOI] [PubMed] [Google Scholar]
  • 204.Moosmayer D, Berndorff D, Chang CH, Sharkey RM, Rother A, Borkowski S, Rossi EA, McBride WJ, Cardillo TM, Goldenberg DM, Dinkelborg LM. Bispecific antibody pretargeting of tumor neovasculature for improved systemic radiotherapy of solid tumors. Clin Cancer Res. 2006 Sep 15;12(18):5587–95. doi: 10.1158/1078-0432.CCR-06-0210. [DOI] [PubMed] [Google Scholar]
  • 205.Spaeth N, Wyss MT, Pahnke J, Biollaz G, Trachsel E, Drandarov K, Treyer V, Weber B, Neri D, Buck A. Radioimmunotherapy targeting the extra domain B of fibronectin in C6 rat gliomas: a preliminary study about the therapeutic efficacy of iodine-131-labeled SIP(L19) Nucl Med Biol. 2006 Jul;33(5):661–6. doi: 10.1016/j.nucmedbio.2006.05.001. [DOI] [PubMed] [Google Scholar]
  • 206.Yordanov AT, Garmestani K, Zhang M, Zhang Z, Yao Z, Phillips KE, Herring B, Horak E, Beitzel MP, Schwarz UP, Gansow OA, Plascjak PS, Eckelman WC, Waldmann TA, Brechbiel MW. Preparation and in vivo evaluation of linkers for 211 At labeling of humanized anti-Tac. Nucl Med Biol. 2001 Oct;28(7):845–56. doi: 10.1016/s0969-8051(01)00257-8. [DOI] [PubMed] [Google Scholar]
  • 207.Rosenkranz AA, Vaidyanathan G, Pozzi OR, Lunin VG, Zalutsky MR, Sobolev AS. Engineered modular recombinant transporters: application of new platform for targeted radiotherapeutic agents to alpha-particle emitting 211 At. Int J Radiat Oncol Biol Phys. 2008 Sep 1;72(1):193–200. doi: 10.1016/j.ijrobp.2008.05.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Emrich JG, Brady LW, Quang TS, Class R, Miyamoto C, Black P, Rodeck U. Radioiodinated (I-125) monoclonal antibody 425 in the treatment of high grade glioma patients: ten-year synopsis of a novel treatment. Am J Clin Oncol. 2002 Dec;25(6):541–6. doi: 10.1097/00000421-200212000-00001. [DOI] [PubMed] [Google Scholar]
  • 209.Wygoda Z, Kula D, Bierzynska-Macyszyn G, Larysz D, Jarzab M, Wlaszczuk P, Bazowski P, Wojtacha M, Rudnik A, Stepien T, Kaspera W, Etmanska A, Skladowski K, Tarnawski R, Kokocinska D, Jarzab B. Use of monoclonal anti-EGFR antibody in the radioimmunotherapy of malignant gliomas in the context of EGFR expression in grade III and IV tumors. Hybridoma (Larchmt) 2006 Jun;25(3):125–32. doi: 10.1089/hyb.2006.25.125. [DOI] [PubMed] [Google Scholar]
  • 210.McDevitt MR, Ma D, Simon J, Frank RK, Scheinberg DA. Design and synthesis of 225Ac radioimmunopharmaceuticals. Appl Radiat Isot. 2002 Dec;57(6):841–7. doi: 10.1016/s0969-8043(02)00167-7. [DOI] [PubMed] [Google Scholar]
  • 211.Couturier O, Supiot S, Degraef-Mougin M, Faivre-Chauvet A, Carlier T, Chatal JF, Davodeau F, Cherel M. Cancer radioimmunotherapy with alpha-emitting nuclides. Eur J Nucl Med Mol Imaging. 2005 May;32(5):601–14. doi: 10.1007/s00259-005-1803-2. [DOI] [PubMed] [Google Scholar]
  • 212.Yamamoto T, Nakai K, Matsumura A. Boron neutron capture therapy for glioblastoma. Cancer Lett. 2008 Apr 18;262(2):143–52. doi: 10.1016/j.canlet.2008.01.021. [DOI] [PubMed] [Google Scholar]
  • 213.Yamamoto T, Matsumura A, Nakai K, Shibata Y, Endo K, Sakurai F, Kishi T, Kumada H, Yamamoto K, Torii Y. Current clinical results of the Tsukuba BNCT trial. Appl Radiat Isot. 2004 Nov;61(5):1089–93. doi: 10.1016/j.apradiso.2004.05.010. [DOI] [PubMed] [Google Scholar]
  • 214.Chanana AD, Capala J, Chadha M, Coderre JA, Diaz AZ, Elowitz EH, Iwai J, Joel DD, Liu HB, Ma R, Pendzick N, Peress NS, Shady MS, Slatkin DN, Tyson GW, Wielopolski L. Boron neutron capture therapy for glioblastoma multiforme: interim results from the phase I/II dose-escalation studies. Neurosurgery. 1999 Jun;44(6):1182–92. doi: 10.1097/00006123-199906000-00013. [DOI] [PubMed] [Google Scholar]
  • 215.Capala J, Stenstam BH, Skold K, Munck af RP, Giusti V, Persson C, Wallin E, Brun A, Franzen L, Carlsson J, Salford L, Ceberg C, Persson B, Pellettieri L, Henriksson R. Boron neutron capture therapy for glioblastoma multiforme: clinical studies in Sweden. JNeurooncol. 2003 Mar;62(1–2):135–44. doi: 10.1007/BF02699940. [DOI] [PubMed] [Google Scholar]
  • 216.Diaz AZ. Assessment of the results from the phase I/II boron neutron capture therapy trials at the Brookhaven National Laboratory from a clinician’s point of view. J Neurooncol. 2003 Mar;62(1–2):101–9. doi: 10.1007/BF02699937. [DOI] [PubMed] [Google Scholar]
  • 217.Coderre JA, Turcotte JC, Riley KJ, Binns PJ, Harling OK, Kiger WS., III Boron neutron capture therapy: cellular targeting of high linear energy transfer radiation. Technol Cancer Res Treat. 2003 Oct;2(5):355–75. doi: 10.1177/153303460300200502. [DOI] [PubMed] [Google Scholar]
  • 218.Joensuu H, Kankaanranta L, Seppala T, Auterinen I, Kallio M, Kulvik M, Laakso J, Vahatalo J, Kortesniemi M, Kotiluoto P, Seren T, Karila J, Brander A, Jarviluoma E, Ryynanen P, Paetau A, Ruokonen I, Minn H, Tenhunen M, Jaaskelainen J, Farkkila M, Savolainen S. Boron neutron capture therapy of brain tumors: clinical trials at the finnish facility using boronophenylalanine. J Neurooncol. 2003 Mar;62(1–2):123–34. doi: 10.1007/BF02699939. [DOI] [PubMed] [Google Scholar]
  • 219.Yang W, Earth RF, Wu G, Bandyopadhyaya AK, Thirumamagal BT, Tjarks W, Binns PJ, Riley K, Patel H, Coderre JA, Ciesielski MJ, Fenstermaker RA. Boronated epidermal growth factor as a delivery agent for neutron capture therapy of EGF receptor positive gliomas. Appl Radiat Isot. 2004 Nov;61(5):981–5. doi: 10.1016/j.apradiso.2004.05.071. [DOI] [PubMed] [Google Scholar]
  • 220.Pelloski CE, Ballman KV, Furth AF, Zhang L, Lin E, Sulman EP, Bhat K, McDonald JM, Yung WK, Colman H, Woo SY, Heimberger AB, Suki D, Prados MD, Chang SM, Barker FG, Buckner JC, James CD, Aldape K. Epidermal growth factor receptor variant III status defines clinically distinct subtypes of glioblastoma. J Clin Oncol. 2007 Jun 1;25(16):2288–94. doi: 10.1200/JCO.2006.08.0705. [DOI] [PubMed] [Google Scholar]
  • 221.Hall WA, Fodstad O. Immunotoxins and central nervous system neoplasia. J Neurosurg. 1992 Jan;76(1):1–12. doi: 10.3171/jns.1992.76.1.0001. [DOI] [PubMed] [Google Scholar]
  • 222.Liu G, Yu JS, Zeng G, Yin D, Xie D, Black KL, Ying H. AIM-2: a novel tumor antigen is expressed and presented by human glioma cells. J Immunother. 2004 May;27(3):220–6. doi: 10.1097/00002371-200405000-00006. [DOI] [PubMed] [Google Scholar]
  • 223.Joshi BH, Leland P, Asher A, Prayson RA, Varricchio F, Puri RK. In situ expression of interleukin-4 (IL-4) receptors in human brain tumors and cytotoxicity of a recombinant IL-4 cytotoxin in primary glioblastoma cell cultures. Cancer Res. 2001 Nov 15;61(22):8058–61. [PubMed] [Google Scholar]
  • 224.Puri RK, Hoon DS, Leland P, Snoy P, Rand RW, Pastan I, Kreitman RJ. Preclinical development of a recombinant toxin containing circularly permuted interleukin 4 and truncated Pseudomonas exotoxin for therapy of malignant astrocytoma. Cancer Res. 1996 Dec 15;56(24):5631–7. [PubMed] [Google Scholar]
  • 225.Kawakami M, Kawakami K, Puri RK. Interleukin-4-Pseudomonas exotoxin chimeric fusion protein for malignant glioma therapy. J Neurooncol. 2003 Oct;65(1):15–25. doi: 10.1023/a:1026294416718. [DOI] [PubMed] [Google Scholar]
  • 226.Weber F, Asher A, Bucholz R, Berger M, Prados M, Chang S, Bruce J, Hall W, Rainov NG, Westphal M, Warnick RE, Rand RW, Floeth F, Rommel F, Pan H, Hingorani VN, Puri RK. Safety, tolerability, and tumor response of IL4-Pseudomonas exotoxin (NBI-3001) in patients with recurrent malignant glioma. J Neurooncol. 2003 Aug;64(1–2):125–37. doi: 10.1007/BF02700027. [DOI] [PubMed] [Google Scholar]
  • 227.Rand RW, Kreitman RJ, Patronas N, Varricchio F, Pastan I, Puri RK. Intratumoral administration of recombinant circularly permuted interleukin-4-Pseudomonas exotoxin in patients with high-grade glioma. Clin Cancer Res. 2000 Jun;6(6):2157–65. [PubMed] [Google Scholar]
  • 228.Debinski W, Obiri NI, Powers SK, Pastan I, Puri RK. Human glioma cells overexpress receptors for interleukin 13 and are extremely sensitive to a novel chimeric protein composed of interleukin 13 and pseudomonas exotoxin. Clin Cancer Res. 1995 Nov;1(11):1253–8. [PubMed] [Google Scholar]
  • 229.Mut M, Sherman JH, Shaffrey ME, Schiff D. Cintredekin besudotox in treatment of malignant glioma. Expert Opin Biol Ther. 2008 Jun;8(6):805–12. doi: 10.1517/14712598.8.6.805. [DOI] [PubMed] [Google Scholar]
  • 230.Kunwar S, Chang SM, Prados MD, Berger MS, Sampson JH, Croteau D, Sherman JW, Grahn AY, Shu VS, Dul JL, Husain SR, Joshi BH, Pedain C, Puri RK. Safety of intraparenchymal convection-enhanced delivery of cintredekin besudotox in early-phase studies. Neurosurg Focus. 2006;20(4):E15. [PubMed] [Google Scholar]
  • 231.Kioi M, Seetharam S, Puri RK. Targeting IL-13 Ralpha2-positive cancer with a novel recombinant immunotoxin composed of a single-chain antibody and mutated Pseudomonas exotoxin. Mol Cancer Ther. 2008 Jun;7(6):1579–87. doi: 10.1158/1535-7163.MCT-07-2131. [DOI] [PubMed] [Google Scholar]
  • 232.Weaver M, Laske DW. Transferrin receptor ligand-targeted toxin conjugate (Tf-CRM107) for therapy of malignant gliomas. J Neurooncol. 2003 Oct;65(1):3–13. doi: 10.1023/a:1026246500788. [DOI] [PubMed] [Google Scholar]
  • 233.Laske DW, Youle RJ, Oldfield EH. Tumor regression with regional distribution of the targeted toxin TF-CRM107 in patients with malignant brain tumors. Nat Med. 1997 Dec;3(12):1362–8. doi: 10.1038/nm1297-1362. [DOI] [PubMed] [Google Scholar]
  • 234.Sampson JH, Akabani G, Archer GE, Bigner DD, Berger MS, Friedman AH, Friedman HS, Herndon JE, Kunwar S, Marcus S, McLendon RE, Paolino A, Penne K, Provenzale J, Quinn J, Reardon DA, Rich J, Stenzel T, Tourt-Uhlig S, Wikstrand C, Wong T, Williams R, Yuan F, Zalutsky MR, Pastan I. Progress report of a Phase I study of the intracerebral micro-infusion of a recombinant chimeric protein composed of transforming growth factor (TGF)-alpha and a mutated form of the Pseudomonas exotoxin termed PE-38 (TP-38) for the treatment of malignant brain tumors. J Neurooncol. 2003 Oct;65(1):27–35. doi: 10.1023/a:1026290315809. [DOI] [PubMed] [Google Scholar]
  • 235.Sampson JH, Reardon DA, Friedman AH, Friedman HS, Coleman RE, McLendon RE, Pastan I, Bigner DD. Sustained radiographic and clinical response in patient with bifrontal recurrent glioblastoma multiforme with intracerebral infusion of the recombinant targeted toxin TP-38: case study. Neuro Oncol. 2005 Jan;7(1):90–6. doi: 10.1215/S1152851703000589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, Simon P, Lotze MT, Yang JC, Seipp CA, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med. 1988 Dec 22;319(25):1676–80. doi: 10.1056/NEJM198812223192527. [DOI] [PubMed] [Google Scholar]
  • 237.Kruse CA, Cepeda L, Owens B, Johnson SD, Stears J, Lillehei KO. Treatment of recurrent glioma with intracavitary alloreactive cytotoxic T lymphocytes and interleukin-2. Cancer Immunol Immunother. 1997 Oct;45(2):77–87. doi: 10.1007/s002620050405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Lillehei KO, Mitchell DH, Johnson SD, McCleary EL, Kruse CA. Long-term follow-up of patients with recurrent malignant gliomas treated with adjuvant adoptive immunotherapy. Neurosurgery. 1991 Jan;28(1):16–23. doi: 10.1097/00006123-199101000-00003. [DOI] [PubMed] [Google Scholar]
  • 239.Hayes RL, Koslow M, Hiesiger EM, Hymes KB, Hochster HS, Moore EJ, Pierz DM, Chen DK, Budzilovich GN, Ransohoff J. Improved long term survival after intracavitary interleukin-2 and lymphokine-activated killer cells for adults with recurrent malignant glioma. Cancer. 1995 Sep 1;76(5):840–52. doi: 10.1002/1097-0142(19950901)76:5<840::aid-cncr2820760519>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
  • 240.Hayes RL, Arbit E, Odaimi M, Pannullo S, Scheff R, Kravchinskiy D, Zaroulis C. Adoptive cellular immunotherapy for the treatment of malignant gliomas. Crit Rev Oncol Hematol. 2001 Jul;39(1–2):31–42. doi: 10.1016/s1040-8428(01)00122-6. [DOI] [PubMed] [Google Scholar]
  • 241.Salmaggi A, Dufour A, Silvani A, Ciusani E, Nespolo A, Boiardi A. Immunological fluctuations during intra-thecal immunotherapy in three patients affected by CNS tumours disseminating via CSF. Int J Neurosci. 1994 Jul;77(1–2):117–25. doi: 10.3109/00207459408986024. [DOI] [PubMed] [Google Scholar]
  • 242.Barba D, Saris SC, Holder C, Rosenberg SA, Oldfield EH. Intratumoral LAK cell and interleukin-2 therapy of human gliomas. J Neurosurg. 1989 Feb;70(2):175–82. doi: 10.3171/jns.1989.70.2.0175. [DOI] [PubMed] [Google Scholar]
  • 243.Cabarrocas J, Bauer J, Piaggio E, Liblau R, Lassmann H. Effective and selective immune surveillance of the brain by MHC class I-restricted cytotoxic T lymphocytes. Eur J Immunol. 2003 May;33(5):1174–82. doi: 10.1002/eji.200323492. [DOI] [PubMed] [Google Scholar]
  • 244.Holladay FP, Heitz T, Chen YL, Chiga M, Wood GW. Successful treatment of a malignant rat glioma with cytotoxic T lymphocytes. Neurosurgery. 1992 Sep;31(3):528–33. doi: 10.1227/00006123-199209000-00015. [DOI] [PubMed] [Google Scholar]
  • 245.Holladay FP, Heitz T, Wood GW. Antitumor activity against established intracerebral gliomas exhibited by cytotoxic T lymphocytes, but not by lymphokine-activated killer cells. J Neurosurg. 1992 Nov;77(5):757–62. doi: 10.3171/jns.1992.77.5.0757. [DOI] [PubMed] [Google Scholar]
  • 246.Rosenberg SA, Dudley ME, Restifo NP. Cancer immunotherapy. N Engl J Med. 2008 Sep 4;359(10):1072. doi: 10.1056/NEJMc081511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Rosenberg SA. Overcoming obstacles to the effective immunotherapy of human cancer. Proc Natl Acad Sci U S A. 2008 Sep 2;105(35):12643–4. doi: 10.1073/pnas.0806877105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Paulos CM, Suhoski MM, Plesa G, Jiang T, Basu S, Golovina TN, Jiang S, Aqui NA, Powell DJ, Jr, Levine BL, Carroll RG, Riley JL, June CH. Adoptive immunotherapy: good habits instilled at youth have long-term benefits. Immunol Res. 2008;42(1–3):182–96. doi: 10.1007/s12026-008-8070-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Perret R, Ronchese F. Memory T cells in cancer immunotherapy: which CD8 T-cell population provides the best protection against tumours? Tissue Antigens. 2008 Sep;72(3):187–94. doi: 10.1111/j.1399-0039.2008.01088.x. [DOI] [PubMed] [Google Scholar]
  • 250.Yee C, Thompson JA, Roche P, Byrd DR, Lee PP, Piepkorn M, Kenyon K, Davis MM, Riddell SR, Greenberg PD. Melanocyte destruction after antigen-specific immunotherapy of melanoma: direct evidence of t cell-mediated vitiligo. J Exp Med. 2000 Dec 4;192(11):1637–44. doi: 10.1084/jem.192.11.1637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Yee C, Thompson JA, Byrd D, Riddell SR, Roche P, Celis E, Greenberg PD. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci U S A. 2002 Dec 10;99(25):16168–73. doi: 10.1073/pnas.242600099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Dreno B, Nguyen JM, Khammari A, Pandolfino MC, Tessier MH, Bercegeay S, Cassidanius A, Lemarre P, Billaudel S, Labarriere N, Jotereau F. Randomized trial of adoptive transfer of melanoma tumor-infiltrating lymphocytes as adjuvant therapy for stage III melanoma. Cancer Immunol Immunother. 2002 Nov;51(10):539–46. doi: 10.1007/s00262-002-0315-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, Royal RE, Kammula U, White DE, Mavroukakis SA, Rogers LJ, Gracia GJ, Jones SA, Mangiameli DP, Pelletier MM, Gea-Banacloche J, Robinson MR, Berman DM, Filie AC, Abati A, Rosenberg SA. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol. 2005 Apr 1;23(10):2346–57. doi: 10.1200/JCO.2005.00.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Li Y, Moysey R, Molloy PE, Vuidepot AL, Mahon T, Baston E, Dunn S, Liddy N, Jacob J, Jakobsen BK, Boulter JM. Directed evolution of human T-cell receptors with picomolar affinities by phage display. Nat Biotechnol. 2005 Mar;23(3):349–54. doi: 10.1038/nbt1070. [DOI] [PubMed] [Google Scholar]
  • 255.Parney IF, Farr-Jones MA, Chang LJ, Petruk KC. Human glioma immunobiology in vitro: implications for immunogene therapy. Neurosurgery. 2000 May;46(5):1169–77. doi: 10.1097/00006123-200005000-00030. [DOI] [PubMed] [Google Scholar]
  • 256.Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, Royal RE, Topalian SL, Kammula US, Restifo NP, Zheng Z, Nahvi A, de Vries CR, Rogers-Freezer LJ, Mavroukakis SA, Rosenberg SA. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314(5796):126–9. doi: 10.1126/science.1129003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989 Dec;86(24):10024–8. doi: 10.1073/pnas.86.24.10024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Kahlon KS, Brown C, Cooper LJ, Raubitschek A, Forman SJ, Jensen MC. Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolyticT cells. Cancer Res. 2004 Dec 15;64(24):9160–6. doi: 10.1158/0008-5472.CAN-04-0454. [DOI] [PubMed] [Google Scholar]
  • 259.Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, White DE, Wunderlich JR, Canevari S, Rogers-Freezer L, Chen CC, Yang JC, Rosenberg SA, Hwu P. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006 Oct 15;12(20 Pt 1):6106–15. doi: 10.1158/1078-0432.CCR-06-1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Park JR, Digiusto DL, Slovak M, Wright C, Naranjo A, Wagner J, Meechoovet HB, Bautista C, Chang WC, Ostberg JR, Jensen MC. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007 Apr;15(4):825–33. doi: 10.1038/sj.mt.6300104. [DOI] [PubMed] [Google Scholar]
  • 261.Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R, Gratama JW, Stoter G, Oosterwijk E. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol. 2006 May 1;24(13):e20–22. doi: 10.1200/JCO.2006.05.9964. [DOI] [PubMed] [Google Scholar]
  • 262.Huang X, Guo H, Kang J, Choi S, Zhou TC, Tammana S, Lees CJ, Li ZZ, Milone M, Levine BL, Tolar J, June CH, Scott MR, Wagner JE, Blazar BR, Zhou X. Sleeping Beauty transposon-mediated engineering of human primary T cells for therapy of CD19+ lymphoid malignancies. Mol Ther. 2008 Mar;16(3):580–9. doi: 10.1038/sj.mt.6300404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Wen J, Zhu X, Liu B, You L, Kong L, Lee HI, Han KP, Wong JL, Rhode PR, Wong HC. Targeting activity of a TCR/IL-2 fusion protein against established tumors. Cancer Immunol Immunother. 2008 Dec;57(12):1781–94. doi: 10.1007/s00262-008-0504-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007 Mar;8(3):239–45. doi: 10.1038/ni1443. [DOI] [PubMed] [Google Scholar]
  • 265.Kozako T, Yoshimitsu M, Fujiwara H, Masamoto I, Horai S, White Y, Akimoto M, Suzuki S, Matsushita K, Uozumi K, Tei C, Arima N. PD-1/PD-L1 expression in human T-cell leukemia virus type 1 carriers and adult T-cell leukemia/lymphoma patients. Leukemia. 2009 Feb;23(2):375–82. doi: 10.1038/leu.2008.272. [DOI] [PubMed] [Google Scholar]
  • 266.Wong RM, Scotland RR, Lau RL, Wang C, Korman AJ, Kast WM, Weber JS. Programmed death-1 blockade enhances expansion and functional capacity of human melanoma antigen-specific CTLs. Int Immunol. 2007 Oct;19(10):1223–34. doi: 10.1093/intimm/dxm091. [DOI] [PubMed] [Google Scholar]
  • 267.Katou F, Ohtani H, Watanabe Y, Nakayama T, Yoshie O, Hashimoto K. Differing phenotypes between intraepithelial and stromal lymphocytes in early-stage tongue cancer. Cancer Res. 2007 Dec 1;67(23):11195–201. doi: 10.1158/0008-5472.CAN-07-2637. [DOI] [PubMed] [Google Scholar]
  • 268.Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, Higuchi T, Yagi H, Takakura K, Minato N, Honjo T, Fujii S. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci U S A. 2007 Feb 27;104(9):3360–5. doi: 10.1073/pnas.0611533104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Nomi T, Sho M, Akahori T, Hamada K, Kubo A, Kanehiro H, Nakamura S, Enomoto K, Yagita H, Azuma M, Nakajima Y. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clin Cancer Res. 2007 Apr 1;13(7):2151–7. doi: 10.1158/1078-0432.CCR-06-2746. [DOI] [PubMed] [Google Scholar]
  • 270.Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, Mackey EW, Miller JD, Leslie AJ, DePierres C, Mncube Z, Duraiswamy J, Zhu B, Eichbaum Q, Altfeld M, Wherry EJ, Coovadia HM, Goulder PJ, Klenerman P, Ahmed R, Freeman GJ, Walker BD. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006 Sep 21;443(7109):350–4. doi: 10.1038/nature05115. [DOI] [PubMed] [Google Scholar]
  • 271.Berger R, Rotem-Yehudar R, Slama G, Landes S, Kneller A, Leiba M, Koren-Michowitz M, Shimoni A, Nagler A. Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clin Cancer Res. 2008 May 15;14(10):3044–51. doi: 10.1158/1078-0432.CCR-07-4079. [DOI] [PubMed] [Google Scholar]
  • 272.Wherry EJ, Teichgraber V, Becker TC, Masopust D, Kaech SM, Antia R, von Andrian UH, Ahmed R. Lineage relationship and protective immunity of memory CD8 T cell subsets. Nat Immunol. 2003 Mar;4(3):225–34. doi: 10.1038/ni889. [DOI] [PubMed] [Google Scholar]
  • 273.Robbins PF, Dudley ME, Wunderlich J, El-Gamil M, Li YF, Zhou J, Huang J, Powell DJ, Jr, Rosenberg SA. Cutting edge: persistence of transferred lymphocyte clonotypes correlates with cancer regression in patients receiving cell transfer therapy. J Immunol. 2004 Dec 15;173(12):7125–30. doi: 10.4049/jimmunol.173.12.7125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Zhou J, Shen X, Huang J, Hodes RJ, Rosenberg SA, Robbins PF. Telomere length of transferred lymphocytes correlates with in vivo persistence and tumor regression in melanoma patients receiving cell transfer therapy. J Immunol. 2005 Nov 15;175(10):7046–52. doi: 10.4049/jimmunol.175.10.7046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Appay V, Dunbar PR, Callan M, Klenerman P, Gillespie GM, Papagno L, Ogg GS, King A, Lechner F, Spina CA, Little S, Havlir DV, Richman DD, Gruener N, Pape G, Waters A, Easterbrook P, Salio M, Cerundolo V, McMichael AJ, Rowland-Jones SL. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nat Med. 2002 Apr;8(4):379–85. doi: 10.1038/nm0402-379. [DOI] [PubMed] [Google Scholar]
  • 276.van Leeuwen EM, van Buul JD, Remmerswaal EB, Hordijk PL, Ten BI, Van Lier RA. Functional re-expression of CCR7 on CMV-specific CD8+ T cells upon antigenic stimulation. Int Immunol. 2005 Jun;17(6):713–9. doi: 10.1093/intimm/dxh251. [DOI] [PubMed] [Google Scholar]
  • 277.Topp MS, Riddell SR, Akatsuka Y, Jensen MC, Blattman JN, Greenberg PD. Restoration of CD28 expression in CD28− CD8+ memory effector T cells reconstitutes antigen-induced IL-2 production. J Exp Med. 2003 Sep 15;198(6):947–55. doi: 10.1084/jem.20021288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Powell DJ, Jr, Dudley ME, Robbins PF, Rosenberg SA. Transition of late-stage effector T cells to CD27+ CD28+ tumor-reactive effector memory T cells in humans after adoptive cell transfer therapy. Blood. 2005 Jan 1;105(1):241–50. doi: 10.1182/blood-2004-06-2482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Ochsenbein AF, Riddell SR, Brown M, Corey L, Baerlocher GM, Lansdorp PM, Greenberg PD. CD27 expression promotes long-term survival of functional effector-memory CD8+ cytotoxic T lymphocytes in HIV-infected patients. J Exp Med. 2004 Dec 6;200(11):1407–17. doi: 10.1084/jem.20040717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Gamadia LE, van Leeuwen EM, Remmerswaal EB, Yong SL, Surachno S, Wertheim-van Dillen PM, Ten Berge IJ, Van Lier RA. The size and phenotype of virus-specific T cell populations is determined by repetitive antigenic stimulation and environmental cytokines. J Immunol. 2004 May 15;172(10):6107–14. doi: 10.4049/jimmunol.172.10.6107. [DOI] [PubMed] [Google Scholar]
  • 281.Okano F, Storkus WJ, Chambers WH, Pollack IF, Okada H. Identification of a novel HLA-A*0201 restricted cytotoxic T lymphocyte epitope in a human glioma associated antigen, interleukin-13 receptor 2 chain. Clin Cancer Res. 2002;8:2851–5. [PubMed] [Google Scholar]
  • 282.Liu G, Khong HT, Wheeler CJ, Yu JS, Black KL, Ying H. Molecular and functional analysis of tyrosinase-related protein (TRP)-2 as a cytotoxic T lymphocyte target in patients with malignant glioma. JImmunother. 2003 Jul;26(4):301–12. doi: 10.1097/00002371-200307000-00002. [DOI] [PubMed] [Google Scholar]
  • 283.Liu G, Ying H, Zeng G, Wheeler CJ, Black KL, Yu JS. HER-2, gp100, and MAGE-1 are expressed in human glioblastoma and recognized by cytotoxic T cells. Cancer Res. 2004 Jul 15;64(14):4980–6. doi: 10.1158/0008-5472.CAN-03-3504. [DOI] [PubMed] [Google Scholar]
  • 284.Wu AH, Xiao J, Anker L, Hall WA, Gregerson DS, Cavenee WK, Chen W, Low WC. Identification of EGFRvIII-derived CTL epitopes restricted by HLA A0201 for dendritic cell based immunotherapy of gliomas. J Neurooncol. 2006 Jan;76(1):23–30. doi: 10.1007/s11060-005-3280-7. [DOI] [PubMed] [Google Scholar]
  • 285.Hatano M, Eguchi J, Tatsumi T, Kuwashima N, Dusak JE, Kinch MS, Pollack IF, Hamilton RL, Storkus WJ, Okada H. EphA2 as a glioma-associated antigen: a novel target for glioma vaccines. Neoplasia. 2005 Aug;7(8):717–22. doi: 10.1593/neo.05277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Jin M, Komohara Y, Shichijo S, Yamanaka R, Nikawa J, Itoh K, Yamada A. Erythropoietin-producing hepatocyte B6 variant-derived peptides with the ability to induce glioma-reactive cytotoxic T lymphocytes in human leukocyte antigen-A2+ glioma patients. Cancer Sci. 2008 Aug;99(8):1656–62. doi: 10.1111/j.1349-7006.2008.00866.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Ueda R, Low KL, Zhu X, Fujita M, Sasaki K, Whiteside TL, Butterfield LH, Okada H. Spontaneous immune responses against glioma-associated antigens in a long term survivor with malignant glioma. J Transl Med. 2007 Dec 19;5(1):68. doi: 10.1186/1479-5876-5-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Theobald M, Biggs J, Dittmer D, Levine AJ, Sherman LA. Targeting p53 as a general tumor antigen. Proc Natl Acad Sci U S A. 1995 Dec 19;92(26):11993–7. doi: 10.1073/pnas.92.26.11993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Cohen CJ, Li YF, El-Gamil M, Robbins PF, Rosenberg SA, Morgan RA. Enhanced antitumor activity of T cells engineered to express T-cell receptors with a second disulfide bond. Cancer Res. 2007 Apr 15;67(8):3898–903. doi: 10.1158/0008-5472.CAN-06-3986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Janicki CN, Jenkinson SR, Williams NA, Morgan DJ. Loss of CTL function among high-avidity tumor-specific CD8+ T cells following tumor infiltration. Cancer Res. 2008 Apr 15;68(8):2993–3000. doi: 10.1158/0008-5472.CAN-07-5008. [DOI] [PubMed] [Google Scholar]
  • 291.Hunder NN, Wallen H, Cao J, Hendricks DW, Reilly JZ, Rodmyre R, Jungbluth A, Gnjatic S, Thompson JA, Yee C. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med. 2008 Jun 19;358(25):2698–703. doi: 10.1056/NEJMoa0800251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Liau LM, Prins RM, Kiertscher SM, Odesa SK, Kremen TJ, Giovannone AJ, Lin JW, Chute DJ, Mischel PS, Cloughesy TF, Roth MD. Dendritic cell vaccination in glioblastoma patients induces systemic and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment. Clin Cancer Res. 2005 Aug 1;11(15):5515–25. doi: 10.1158/1078-0432.CCR-05-0464. [DOI] [PubMed] [Google Scholar]
  • 293.Okada H, Lieberman FS, Walter KA, Lunsford LD, Kondziolka DS, Bejjani GK, Hamilton RL, Torres-Trejo A, Kalinski P, Cai Q, Mabold JL, Edington HD, Butterfield LH, Whiteside TL, Potter DM, Schold SC, Jr, Pollack IF. Autologous glioma cell vaccine admixed with interleukin-4 gene transfected fibroblasts in the treatment of patients with malignant gliomas. J Transl Med. 2007 Dec 19;5(1):67. doi: 10.1186/1479-5876-5-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Wheeler CJ, Black KL, Liu G, Mazer M, Zhang XX, Pepkowitz S, Goldfinger D, Ng H, Irvin D, Yu JS. Vaccination elicits correlated immune and clinical responses in glioblastoma multiforme patients. Cancer Res. 2008 Jul 15;68(14):5955–64. doi: 10.1158/0008-5472.CAN-07-5973. [DOI] [PubMed] [Google Scholar]
  • 295.Yu JS, Liu G, Ying H, Yong WH, Black KL, Wheeler CJ. Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res. 2004 Jul 15;64(14):4973–9. doi: 10.1158/0008-5472.CAN-03-3505. [DOI] [PubMed] [Google Scholar]
  • 296.Bigner DD, Pitts OM, Wikstrand CJ. Induction of lethal experimental allergic encephalomyelitis in non-human primates and guinea pigs with human glioblastoma multiforme tissue. J Neurosurg. 1981;55:32–42. doi: 10.3171/jns.1981.55.1.0032. [DOI] [PubMed] [Google Scholar]
  • 297.Shimato S, Natsume A, Wakabayashi T, Tsujimura K, Nakahara N, Ishii J, Ito M, Akatsuka Y, Kuzushima K, Yoshida J. Identification of a human leukocyte antigen-A24-restricted T-cell epitope derived from interleukin-13 receptor alpha2 chain, a glioma-associated antigen. J Neurosurg. 2008 Jul;109(1):117–22. doi: 10.3171/JNS/2008/109/7/0117. [DOI] [PubMed] [Google Scholar]
  • 298.Hatano M, Kuwashima N, Tatsumi T, Dusak JE, Nishimura F, Reilly KM, Storkus WJ, Okada H. Vaccination with EphA2-derived T cell-epitopes promotes immunity against both EphA2-expressing and EphA2-negative tumors. J Transl Med. 2004 Nov 24;2(1):40. doi: 10.1186/1479-5876-2-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Andersen MH, Pedersen LO, Becker JC, Straten PT. Identification of a cytotoxic T lymphocyte response to the apoptosis inhibitor protein survivin in cancer patients. Cancer Res. 2001 Feb 1;61(3):869–72. [PubMed] [Google Scholar]
  • 300.Andersen MH, Pedersen LO, Capeller B, Brocker EB, Becker JC, thor SP. Spontaneous cytotoxic T-cell responses against survivin-derived MHC class I-restricted T-cell epitopes in situ as well as ex vivo in cancer patients. Cancer Res. 2001 Aug 15;61(16):5964–8. [PubMed] [Google Scholar]
  • 301.Uematsu M, Ohsawa I, Aokage T, Nishimaki K, Matsumoto K, Takahashi H, Asoh S, Teramoto A, Ohta S. Prognostic significance of the immunohistochemical index of survivin in glioma: a comparative study with the MIB-1 index. J Neurooncol. 2005 May;72(3):231–8. doi: 10.1007/s11060-004-2353-3. [DOI] [PubMed] [Google Scholar]
  • 302.Oji Y, Suzuki T, Nakano Y, Maruno M, Nakatsuka S, Jomgeow T, Abeno S, Tatsumi N, Yokota A, Aoyagi S, Nakazawa T, Ito K, Kanato K, Shirakata T, Nishida S, Hosen N, kawakami M, Tsuboi A, Oka Y, Aozasa K, Yoshimine T, Sugiyama H. Overexpression of the Wilms’ tumor gene WT1 in primary astrocytic tumors. Cancer Sci. 2004 Oct;95(10):822–7. doi: 10.1111/j.1349-7006.2004.tb02188.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Oka Y, Elisseeva OA, Tsuboi A, Ogawa H, Tamaki H, Li H, Oji Y, Kim EH, Soma T, Asada M, Ueda K, Maruya E, Saji H, Kishimoto T, Udaka K, Sugiyama H. Human cytotoxic T-lymphocyte responses specific for peptides of the wild-type Wilms’ tumor gene (WT1) product. Immunogenetics. 2000 Feb;51(2):99–107. doi: 10.1007/s002510050018. [DOI] [PubMed] [Google Scholar]
  • 304.Sugiyama H. Cancer immunotherapy targeting WT1 protein. Int J Hematol. 2002 Aug;76(2):127–32. doi: 10.1007/BF02982574. [DOI] [PubMed] [Google Scholar]
  • 305.Schmitz M, Temme A, Senner V, Ebner R, Schwind S, Stevanovic S, Wehner R, Schackert G, Schackert HK, Fussel M, Bachmann M, Rieber EP, Weigle B. Identification of SOX2 as a novel glioma-associated antigen and potential target for T cell-based immunotherapy. Br J Cancer. 2007 Apr 23;96(8):1293–301. doi: 10.1038/sj.bjc.6603696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Schmitz M, Wehner R, Stevanovic S, Kiessling A, Rieger MA, Temme A, Bachmann M, Rieber EP, Weigle B. Identification of a naturally processed T cell epitope derived from the glioma-associated protein SOX11. Cancer Lett. 2007 Jan 8;245(1–2):331–6. doi: 10.1016/j.canlet.2006.01.014. [DOI] [PubMed] [Google Scholar]
  • 307.Imaizumi T, Kuramoto T, Matsunaga K, Shichijo S, Yutani S, Shigemori M, Oizumi K, Itoh K. Expression of the tumor-rejection antigen SART1 in brain tumors. Int J Cancer. 1999 Dec 10;83(6):760–4. doi: 10.1002/(sici)1097-0215(19991210)83:6<760::aid-ijc11>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
  • 308.Shichijo S, Nakao M, Imai Y, Takasu H, Kawamoto M, Niiya F, Yang D, Toh Y, Yamana H, Itoh K. A gene encoding antigenic peptides of human squamous cell carcinoma recognized by cytotoxic T lymphocytes. J Exp Med. 1998 Feb 2;187(3):277–88. doi: 10.1084/jem.187.3.277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Debinski W, Gibo DM, Hulet SW, Connor JR, Gillespie GY. Receptor for interleukin 13 is a marker and therapeutic target for human high-grade gliomas. Clin Cancer Res. 1999 May;5(5):985–90. [PubMed] [Google Scholar]
  • 310.Donaldson DD, Whitters MJ, Fitz LJ, Neben TY, Finnerty H, Henderson SL, O’Hara RM, Jr, Beier DR, Turner KJ, Wood CR, Collins M. The murine IL-13 receptor alpha 2: molecular cloning, characterization, and comparison with murine IL-13 receptor alpha 1. J Immunol. 1998 Sep 1;161(5):2317–24. [PubMed] [Google Scholar]
  • 311.Fichtner-Feigl S, Strober W, Kawakami K, Puri RK, Kitani A. IL-13 signaling through the IL-13alpha2 receptor is involved in induction of TGF-beta1 production and fibrosis. Nat Med. 2006 Jan;12(1):99–106. doi: 10.1038/nm1332. [DOI] [PubMed] [Google Scholar]
  • 312.Eguchi J, Hatano M, Nishimura F, Zhu X, Dusak JE, Sato H, Pollack IF, Storkus WJ, Okada H. Identification of interleukin-13 receptor alpha2 peptide analogues capable of inducing improved antiglioma CTL responses. Cancer Res. 2006 Jun 1;66(11):5883–91. doi: 10.1158/0008-5472.CAN-06-0363. [DOI] [PubMed] [Google Scholar]
  • 313.Dodelet VC, Pasquale EB. Eph receptors and ephrin ligands: embryogenesis to tumorigenesis. Oncogene. 2000 Nov 20;19(49):5614–9. doi: 10.1038/sj.onc.1203856. [DOI] [PubMed] [Google Scholar]
  • 314.Oba SM, Wang YJ, Song JP, Li ZY, Kobayashi K, Tsugane S, Hamada GS, Tanaka M, Sugimura H. Genomic structure and loss of heterozygosity of EPHB2 in colorectal cancer. Cancer Lett. 2001 Mar 10;164(1):97–104. doi: 10.1016/s0304-3835(00)00716-3. [DOI] [PubMed] [Google Scholar]
  • 315.Miao H, Wei BR, Peehl DM, Li Q, Alexandrou T, Schelling JR, Rhim JS, Sedor JR, Burnett E, Wang B. Activation of EphA receptor tyrosine kinase inhibits the Ras/MAPK pathway. Nat Cell Biol. 2001 May;3(5):527–30. doi: 10.1038/35074604. [DOI] [PubMed] [Google Scholar]
  • 316.Miao H, Burnett E, Kinch M, Simon E, Wang B. Activation of EphA2 kinase suppresses integrin function and causes focal-adhesion-kinase dephosphorylation. Nat Cell Biol. 2000 Feb;2(2):62–9. doi: 10.1038/35000008. [DOI] [PubMed] [Google Scholar]
  • 317.Kinch MS, Moore MB, Harpole DH., Jr Predictive value of the EphA2 receptor tyrosine kinase in lung cancer recurrence and survival. Clin Cancer Res. 2003 Feb;9(2):613–8. [PubMed] [Google Scholar]
  • 318.Wykosky J, Gibo DM, Stanton C, Debinski W. EphA2 as a novel molecular marker and target in glioblastoma multiforme. Mol Cancer Res. 2005 Oct;3(10):541–51. doi: 10.1158/1541-7786.MCR-05-0056. [DOI] [PubMed] [Google Scholar]
  • 319.Liu F, Park PJ, Lai W, Maher E, Chakravarti A, Durso L, Jiang X, Yu Y, Brosius A, Thomas M, Chin L, Brennan C, DePinho RA, Kohane I, Carroll RS, Black PM, Johnson MD. A genome-wide screen reveals functional gene clusters in the cancer genome and identifies EphA2 as a mitogen in glioblastoma. Cancer Res. 2006 Nov 15;66(22):10815–23. doi: 10.1158/0008-5472.CAN-06-1408. [DOI] [PubMed] [Google Scholar]
  • 320.Blanc-Brude OP, Yu J, Simosa H, Conte MS, Sessa WC, Altieri DC. Inhibitor of apoptosis protein survivin regulates vascular injury. Nat Med. 2002 Sep;8(9):987–94. doi: 10.1038/nm750. [DOI] [PubMed] [Google Scholar]
  • 321.Schmidt SM, Schag K, Muller MR, Weck MM, Appel S, Kanz L, Grunebach F, Brossart P. Survivin is a shared tumor-associated antigen expressed in a broad variety of malignancies and recognized by specific cytotoxic T cells. Blood. 2003 Jul 15;102(2):571–6. doi: 10.1182/blood-2002-08-2554. [DOI] [PubMed] [Google Scholar]
  • 322.Wobser M, Keikavoussi P, Kunzmann V, Weininger M, Andersen MH, Becker JC. Complete remission of liver metastasis of pancreatic cancer under vaccination with a HLA-A2 restricted peptide derived from the universal tumor antigen survivin. Cancer Immunol Immunother. 2006;55:1294–8. doi: 10.1007/s00262-005-0102-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Otto K, Andersen MH, Eggert A, Keikavoussi P, Pedersen LO, Rath JC, Bock M, Brocker EB, Straten PT, Kampgen E, Becker JC. Lack of toxicity of therapy-induced T cell responses against the universal tumour antigen survivin. Vaccine. 2005 Jan 4;23(7):884–9. doi: 10.1016/j.vaccine.2004.08.007. [DOI] [PubMed] [Google Scholar]
  • 324.Oka Y, Tsuboi A, Elisseeva OA, Udaka K, Sugiyama H. WT1 as a novel target antigen for cancer immunotherapy. Curr Cancer Drug Targets. 2002 Mar;2(1):45–54. doi: 10.2174/1568009023334088. [DOI] [PubMed] [Google Scholar]
  • 325.Glienke W, Maute L, Koehl U, Esser R, Milz E, Bergmann L. Effective treatment of leukemic cell lines with wt1 siRNA. Leukemia. 2007 Oct;21(10):2164–70. doi: 10.1038/sj.leu.2404878. [DOI] [PubMed] [Google Scholar]
  • 326.liyama T, Udaka K, Takeda S, Takeuchi T, Adachi YC, Ohtsuki Y, Tsuboi A, Nakatsuka S, Elisseeva OA, Oji Y, Kawakami M, Nakajima H, Nishida S, Shirakata T, Oka Y, Shuin T, Sugiyama H. WT1 (Wilms’ tumor 1) peptide immunotherapy for renal cell carcinoma. Microbiol Immunol. 2007;51(5):519–30. doi: 10.1111/j.1348-0421.2007.tb03940.x. [DOI] [PubMed] [Google Scholar]
  • 327.Nakahara Y, Okamoto H, Mineta T, Tabuchi K. Expression of the Wilms’ tumor gene product WT1 in glioblastomas and medulloblastomas. Brain Tumor Pathol. 2004;21(3):113–6. doi: 10.1007/BF02482185. [DOI] [PubMed] [Google Scholar]
  • 328.Izumoto S, Tsuboi A, Oka Y, Suzuki T, Hashiba T, Kagawa N, Hashimoto N, Maruno M, Elisseeva OA, Shirakata T, Kawakami M, Oji Y, Nishida S, Ohno S, Kawase I, Hatazawa J, Nakatsuka S, Aozasa K, Morita S, Sakamoto J, Sugiyama H, Yoshimine T. Phase II clinical trial of Wilms tumor 1 peptide vaccination for patients with recurrent glioblastoma multiforme. J Neurosurg. 2008;108:963–71. doi: 10.3171/JNS/2008/108/5/0963. [DOI] [PubMed] [Google Scholar]
  • 329.Wegner M. From head to toes: the multiple facets of Sox proteins. Nucleic Acids Res. 1999 Mar 15;27(6):1409–20. doi: 10.1093/nar/27.6.1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Ueda R, Yoshida K, Kawase T, Kawakami Y, Toda M. Preferential expression and frequent IgG responses of a tumor antigen, SOX5, in glioma patients. Int J Cancer. 2007 Apr 15;120(8):1704–11. doi: 10.1002/ijc.22472. [DOI] [PubMed] [Google Scholar]
  • 331.Ueda R, lizuka Y, Yoshida K, Kawase T, Kawakami Y, Toda M. Identification of a human glioma antigen, SOX6, recognized by patients’ sera. Oncogene. 2004 Feb 19;23(7):1420–7. doi: 10.1038/sj.onc.1207252. [DOI] [PubMed] [Google Scholar]
  • 332.Ueda R, Kinoshita E, Ito R, Kawase T, Kawakami Y, Toda M. Induction of protective and therapeutic anti-tumor immunity by a DNA vaccine with a glioma antigen, SOX6. Int J Cancer. 2008 May 15;122(10):2274–9. doi: 10.1002/ijc.23366. [DOI] [PubMed] [Google Scholar]
  • 333.Gangemi RM, Griffero F, Marubbi D, Perera M, Capra MC, Malatesta P, Ravetti GL, Zona GL, Daga A, Corte G. SOX2 Silencing in glioblastoma tumor initiating cells causes stop of proliferation and loss of tumorigenicity. Stem Cells. 2008 Nov 6; doi: 10.1634/stemcells.2008-0493. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 334.Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444:756–60. doi: 10.1038/nature05236. [DOI] [PubMed] [Google Scholar]
  • 335.Hambardzumyan D, Squatrito M, Holland EC. Radiation resistance and stem-like cells in brain tumors. Cancer Cell. 2006 Dec;10(6):454–6. doi: 10.1016/j.ccr.2006.11.008. [DOI] [PubMed] [Google Scholar]
  • 336.Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer. 2005 Apr;5(4):275–84. doi: 10.1038/nrc1590. [DOI] [PubMed] [Google Scholar]
  • 337.Rich JN. Cancer stem cells in radiation resistance. Cancer Res. 2007 Oct 1;67(19):8980–4. doi: 10.1158/0008-5472.CAN-07-0895. [DOI] [PubMed] [Google Scholar]
  • 338.Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE, Levin WJ, Stuart SG, Udove J, Ullrich A, et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989 May 12;244(4905):707–12. doi: 10.1126/science.2470152. [DOI] [PubMed] [Google Scholar]
  • 339.Meric-Bernstam F, Hung MC. Advances in targeting human epidermal growth factor receptor-2 signaling for cancer therapy. Clin Cancer Res. 2006 Nov 1;12(21):6326–30. doi: 10.1158/1078-0432.CCR-06-1732. [DOI] [PubMed] [Google Scholar]
  • 340.Schneider PM, Hung MC, Chiocca SM, Manning J, Zhao XY, Fang K, Roth JA. Differential expression of the c-erbB-2 gene in human small cell and non-small cell lung cancer. Cancer Res. 1989 Sep 15;49(18):4968–71. [PubMed] [Google Scholar]
  • 341.Seliger B, Rongcun Y, Atkins D, Hammers S, Huber C, Storkel S, Kiessling R. HER-2/neu is expressed in human renal cell carcinoma at heterogeneous levels independently of tumor grading and staging and can be recognized by HLA-A2.1-restricted cytotoxic T lymphocytes. Int J Cancer. 2000 Aug 1;87(3):349–59. [PubMed] [Google Scholar]
  • 342.Weiner DB, Nordberg J, Robinson R, Nowell PC, Gazdar A, Greene MI, Williams WV, Cohen JA, Kern JA. Expression of the neu gene-encoded protein (P185neu) in human non-small cell carcinomas of the lung. Cancer Res. 1990 Jan 15;50(2):421–5. [PubMed] [Google Scholar]
  • 343.Yokota J, Yamamoto T, Toyoshima K, Terada M, Sugimura T, Battifora H, Cline MJ. Amplification of c-erbB-2 oncogene in human adenocarcinomas in vivo. Lancet. 1986 Apr 5;1(8484):765–7. doi: 10.1016/s0140-6736(86)91782-4. [DOI] [PubMed] [Google Scholar]
  • 344.Kook H, Itoh H, Choi BS, Sawada N, Doi K, Hwang TJ, Kim KK, Arai H, Baik YH, Nakao K. Physiological concentration of atrial natriuretic peptide induces endothelial regeneration in vitro. Am J Physiol Heart Circ Physiol. 2003 Apr;284(4):H1388–97. doi: 10.1152/ajpheart.00414.2002. [DOI] [PubMed] [Google Scholar]
  • 345.Saikali S, Avril T, Collet B, Hamlat A, Bansard JY, Drenou B, Guegan Y, Quillien V. Expression of nine tumour antigens in a series of human glioblastoma multiforme: interest of EGFRvIII, IL-13Ralpha2, gp100 andTRP-2 for immunotherapy. J Neurooncol. 2007 Jan;81(2):139–48. doi: 10.1007/s11060-006-9220-3. [DOI] [PubMed] [Google Scholar]
  • 346.Cobbs CS, Harkins L, Samanta M, Gillespie GY, Bharara S, King PH, Nabors LB, Cobbs CG, Britt WJ. Human cytomegalovirus infection and expression in human malignant glioma. Cancer Res. 2002 Jun 15;62(12):3347–50. [PubMed] [Google Scholar]
  • 347.Prins RM, Cloughesy TF, Liau LM. Cytomegalovirus immunity after vaccination with autologous glioblastoma lysate. N Engl J Med. 2008 Jul 31;359(5):539–41. doi: 10.1056/NEJMc0804818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.Mitchell DA, Xie W, Schmittling R, Learn C, Friedman A, McLendon RE, Sampson JH. Sensitive detection of human cytomegalovims in tumors and peripheral blood of patients diagnosed with glioblastoma. Neuro Oncol. 2008 Feb;10(1):10–8. doi: 10.1215/15228517-2007-035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Scheurer ME, Bondy ML, Aldape KD, Albrecht T, El-Zein R. Detection of human cytomegalovims in different histological types of gliomas. Acta Neuropathol. 2008 Jul;116(1):79–86. doi: 10.1007/s00401-008-0359-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350.Bihl F, Frahm N, Di GL, Sidney J, John M, Yusim K, Woodberry T, Sango K, Hewitt HS, Henry L, Linde CH, Chisholm JV, III, Zaman TM, Pae E, Mallal S, Walker BD, Sette A, Korber BT, Heckerman D, Brander C. Impact of HLA-B alleles, epitope binding affinity, functional avidity, and viral coinfection on the immunodominance of virus-specific CTL responses. J Immunol. 2006 Apr 1;176(7):4094–101. doi: 10.4049/jimmunol.176.7.4094. [DOI] [PubMed] [Google Scholar]
  • 351.Harari A, Cellerai C, Enders FB, Kostler J, Codarri L, Tapia G, Boyman O, Castro E, Gaudieri S, James I, John M, Wagner R, Mallal S, Pantaleo G. Skewed association of polyfunctional antigen-specific CD8 T cell populations with HLA-B genotype. Proc Natl Acad Sci U S A. 2007 Oct 9;104(41):16233–8. doi: 10.1073/pnas.0707570104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Holladay FP, Heitz-Turner T, Bayer WL, Wood GW. Autologous tumor cell vaccination combined with adoptive cellular immunotherapy in patients with grade III/IV astrocytoma. J Neurooncol. 1996;27:179–89. doi: 10.1007/BF00177482. [DOI] [PubMed] [Google Scholar]
  • 353.Plautz GE, Barnett GH, Miller DW, Cohen BH, Prayson RA, Krauss JC, Luciano M, Kangisser DB, Shu S. Systemic T cell adoptive immunotherapy of malignant gliomas. J Neurosurg. 1998 Jul;89(1):42–51. doi: 10.3171/jns.1998.89.1.0042. [DOI] [PubMed] [Google Scholar]
  • 354.Plautz GE, Miller DW, Barnett GH, Stevens GH, Maffett S, Kim J, Cohen PA, Shu S. T cell adoptive immunotherapy of newly diagnosed gliomas. Clin Cancer Res. 2000 Jun 1;6(6):2209–18. [PubMed] [Google Scholar]
  • 355.Wood GW, Holladay FP, Turner T, Wang YY, Chiga M. A pilot study of autologous cancer cell vaccination and cellular immunotherapy using anti-CD3 stimulated lymphocytes in patients with recurrent grade III/IV astrocytoma. J Neurooncol. 2000;48:113–20. doi: 10.1023/a:1006456421177. [DOI] [PubMed] [Google Scholar]
  • 356.Yu JS, Wheeler CJ, Zeltzer PM, Ying H, Finger DN, Lee PK, Yong WH, Incardona F, Thompson RC, Riedinger MS, Zhang W, Prins RM, Black KL. Vaccination of malignant glioma patients with peptide-pulsed dendritic cells elicits systemic cytotoxicity and intracranial T-cell infiltration. Cancer Res. 2001 Feb 1;61(3):842–7. [PubMed] [Google Scholar]
  • 357.Andrews DW, Resnicoff M, Flanders AE, Kenyon L, Curtis M, Merli G, Baserga R, Iliakis G, Aiken RD. Results of a pilot study involving the use of an antisense oligodeoxynucleotide directed against the insulin-like growth factor type I receptor in malignant astrocytomas. J Clin Oncol. 2001;19:2189–200. doi: 10.1200/JCO.2001.19.8.2189. [DOI] [PubMed] [Google Scholar]
  • 358.Kikuchi T, Akasaki Y, Irie M, Homma S, Abe T, Ohno T. Results of a phase I clinical trial of vaccination of glioma patients with fusions of dendritic and glioma cells. Cancer Immunol Immunother. 2001 Sep;50(7):337–44. doi: 10.1007/s002620100205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359.Schneider T, Gerhards R, Kirches E, Firsching R. Preliminary results of active specific immunization with modified tumor cell vaccine in glioblastoma multiforme. J Neurooncol. 2001;53:39–46. doi: 10.1023/a:1011856406683. [DOI] [PubMed] [Google Scholar]
  • 360.Yamanaka R, Abe T, Yajima N, Tsuchiya N, Homma J, Kobayashi T, Narita M, Takahashi M, Tanaka R. Vaccination of recurrent glioma patients with tumour lysate-pulsed dendritic cells elicits immune responses: results of a clinical phase I/II trial. Br J Cancer. 2003 Oct 6;89(7):1172–9. doi: 10.1038/sj.bjc.6601268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 361.Caruso DA, Orme LM, Neale AM, Radcliff FJ, Amor GM, Maixner W, Downie P, Hassall TE, Tang ML, Ashley DM. Results of a phase 1 study utilizing monocyte-derived dendritic cells pulsed with tumor RNA in children and young adults with brain cancer. Neuro Oncol. 2004;6:236–46. doi: 10.1215/S1152851703000668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 362.Steiner HH, Bonsanto MM, Beckhove P, Brysch M, Geletneky K, Ahmadi R, Schuele-Freyer R, Kremer P, Ranaie G, Matejic D, Bauer H, Kiessling M, Kunze S, Schirrmacher V, Herold-Mende C. Anti-tumor vaccination of patients with glioblastoma multiforme: a pilot study to assess feasibility, safety, and clinical benefit. J Clin Oncol. 2004;22:4272–81. doi: 10.1200/JCO.2004.09.038. [DOI] [PubMed] [Google Scholar]
  • 363.Rutkowski S, De Vleeschouwer S, Kaempgen E, Wolff JE, Kuhl J, Demaerel P, Warmuth-Metz M, Flamen P, Van Calenbergh F, Plets C, Sorensen N, Opitz A, Van Gool SW. Surgery and adjuvant dendritic cell-based tumour vaccination for patients with relapsed malignant glioma, a feasibility study. Br J Cancer. 2004;91:1656–62. doi: 10.1038/sj.bjc.6602195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Kikuchi T, Akasaki Y, Abe T, Fukuda T, Saotome H, Ryan JL, Kufe DW, Ohno T. Vaccination of glioma patients with fusions of dendritic and glioma cells and recombinant human interleukin 12. J Immunother. 2004 Nov;27(6):452–9. doi: 10.1097/00002371-200411000-00005. [DOI] [PubMed] [Google Scholar]
  • 365.Yamanaka R, Homma J, Yajima N, Tsuchiya N, Sano M, Kobayashi T, Yoshida S, Abe T, Narita M, Takahashi M, Tanaka R. Clinical evaluation of dendritic cell vaccination for patients with recurrent glioma: results of a clinical phase I/II trial. Clin Cancer Res. 2005 Jun 1;11(11):4160–7. doi: 10.1158/1078-0432.CCR-05-0120. [DOI] [PubMed] [Google Scholar]
  • 366.Yajima N, Yamanaka R, Mine T, Tsuchiya N, Homma J, Sano M, Kuramoto T, Obata Y, Komatsu N, Arima Y, Yamada A, Shigemori M, Itoh K, Tanaka R. Immunologic evaluation of personalized peptide vaccination for patients with advanced malignant glioma. Clin Cancer Res. 2005 Aug 15;11(16):5900–11. doi: 10.1158/1078-0432.CCR-05-0559. [DOI] [PubMed] [Google Scholar]
  • 367.Sloan AE, Dansey R, Zamorano L, Barger G, Hamm C, Diaz F, Baynes R, Wood G. Adoptive immunotherapy in patients with recurrent malignant glioma: preliminary results of using autologous whole-tumor vaccine plus granulocyte-macrophage colony-stimulating factor and adoptive transfer of anti-CD3-activated lymphocytes. Neurosurg Focus. 2000;9:e9. doi: 10.3171/foc.2000.9.6.10. [DOI] [PubMed] [Google Scholar]
  • 368.Ishikawa E, Tsuboi K, Yamamoto T, Muroi A, Takano S, Enomoto T, Matsumura A, Ohno T. Clinical trial of autologous formalin-fixed tumor vaccine for glioblastoma multiforme patients. Cancer Sci. 2007;98:1226–33. doi: 10.1111/j.1349-7006.2007.00518.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.De Vleeschouwer S, Fieuws S, Rutkowski S, Van Calenbergh F, Van Loon J, Goffin J, Sciot R, Wilms G, Demaerel P, Warmuth-Metz M, Soerensen N, Wolff JE, Wagner S, Kaempgen E, Van Gool SW. Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clin Cancer Res. 2008 May 15;14(10):3098–104. doi: 10.1158/1078-0432.CCR-07-4875. [DOI] [PubMed] [Google Scholar]
  • 370.Wikstrand CJ, Bigner DD. Immunobiologic aspects of the brain and human gliomas. A review. Am J Pathol. 1980;98:517–68. [PMC free article] [PubMed] [Google Scholar]
  • 371.Zhang JG, Eguchi J, Kruse CA, Gomez GG, Fakhrai H, Schroter S, Ma W, Hoa N, Minev B, Delgado C, Wepsic HT, Okada H, Jadus MR. Antigenic profiling of glioma cells to generate allogeneic vaccines or dendritic cell-based therapeutics. Clin Cancer Res. 2007 Jan 15;13(2 Pt 1):566–75. doi: 10.1158/1078-0432.CCR-06-1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Vanderlugt CL, Miller SD. Epitope spreading in immune-mediated diseases: implications for immunotherapy. Nat Rev Immunol. 2002;2:85–95. doi: 10.1038/nri724. [DOI] [PubMed] [Google Scholar]
  • 373.Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K. Immunobiology of dendritic cells. Annu Rev Immunol. 2000;18:767–811. doi: 10.1146/annurev.immunol.18.1.767. [DOI] [PubMed] [Google Scholar]
  • 374.Banchereau J, Palucka AK. Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol. 2005;5:296–306. doi: 10.1038/nri1592. [DOI] [PubMed] [Google Scholar]
  • 375.De Vleeschouwer S, Fieuws S, Rutkowski S, Van Calenbergh F, Van Loon J, Goffin J, Sciot R, Wilms G, Demaerel P, Warmuth-Metz M, Soerensen N, Wolff JE, Wagner S, Kaempgen E, Van Gool SW. Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clin Cancer Res. 2008;14:3098–104. doi: 10.1158/1078-0432.CCR-07-4875. [DOI] [PubMed] [Google Scholar]
  • 376.Akasaki Y, Kikuchi T, Homma S, Abe T, Kofe D, Ohno T. Antitumor effect of immunizations with fusions of dendritic and glioma cells in a mouse brain tumor model. J Immunother. 2001;24:106–13. [PubMed] [Google Scholar]
  • 377.Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003 Sep 15;63(18):5821–8. [PubMed] [Google Scholar]
  • 378.Yuan X, Curtin J, Xiong Y, Liu G, Waschsmann-Hogiu S, Farkas DL, Black KL, Yu JS. Isolation of cancer stem cells from adult glioblastoma multiforme. Oncogene. 2004 Dec 16;23(58):9392–400. doi: 10.1038/sj.onc.1208311. [DOI] [PubMed] [Google Scholar]
  • 379.Liu G, Yuan X, Zeng Z, Tunici P, Ng H, Abdulkadir IR, Lu L, Irvin D, Black KL, Yu JS. Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer. 2006;5:67. doi: 10.1186/1476-4598-5-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 380.Pellegatta S, Poliani PL, Corno D, Menghi F, Ghielmetti F, Suarez-Merino B, Caldera V, Nava S, Ravanini M, Facchetti F, Bruzzone MG, Finocchiaro G. Neurospheres enriched in cancer stem-like cells are highly effective in eliciting a dendritic cell-mediated immune response against malignant gliomas. Cancer Res. 2006;66:10247–52. doi: 10.1158/0008-5472.CAN-06-2048. [DOI] [PubMed] [Google Scholar]
  • 381.Figdor CG, de Vries IJ, Lesterhuis WJ, Melief CJ. Dendritic cell immunotherapy: mapping the way. Nat Med. 2004;10:475–80. doi: 10.1038/nm1039. [DOI] [PubMed] [Google Scholar]
  • 382.Mailliard RB, Wankowicz-Kalinska A, Cai Q, Wesa A, Hilkens CM, Kapsenberg ML, Kirkwood JM, Storkus WJ, Kalinski P. alpha-type-1 polarized dendritic cells: a novel immunization tool with optimized CTL-inducing activity. Cancer Res. 2004 Sep 1;64(17):5934–7. doi: 10.1158/0008-5472.CAN-04-1261. [DOI] [PubMed] [Google Scholar]
  • 383.Vieira PL, de Jong EC, Wierenga EA, Kapsenberg ML, Kalinski P. Development of Th1-inducing capacity in myeloid dendritic cells requires environmental instruction. J Immunol. 2000 May 1;164(9):4507–12. doi: 10.4049/jimmunol.164.9.4507. [DOI] [PubMed] [Google Scholar]
  • 384.Eguchi J, Hiroishi K, Ishii S, Baba T, Matsumura T, Hiraide A, Okada H, Imawari M. Interleukin-4 gene transduced tumor cells promote a potent tumor-specific CTL response in cooperation with interferon-alpha transduction. Gene Ther. 2005;12(9):733–41. doi: 10.1038/sj.gt.3302401. [DOI] [PubMed] [Google Scholar]

RESOURCES