Skip to main content
Immunology logoLink to Immunology
. 2002 Sep;107(1):28–38. doi: 10.1046/j.1365-2567.2002.01507.x

All in the head: obstacles for immune rejection of brain tumours

Paul R Walker 1, Thomas Calzascia 1, Pierre-Yves Dietrich 1
PMCID: PMC1782770  PMID: 12225360

Introduction

Tumour immunology and immunotherapy is a highly active field, a clinical testing ground for cutting edge immunological techniques and concepts. But this is after many years of fundamental advances in basic immunology. In this article we suggest that immunotherapy for brain tumours cannot be rationally advanced as rapidly as that for tumours in other sites. Our understanding of anti-tumour immune responses in the brain is sketchy and frequently extrapolated from other tissues having little in common with the central nervous system (CNS). The result is that current clinical trials are built upon shakier foundations, with the somewhat naive optimism that what is looking hopeful for other tumours will also be applicable to cerebral malignancies. But of course it is easy to criticise such well-meaning attempts to treat currently incurable cancers. In the basic and preclinical domain, brain tumour models that are readily applicable to the design of future immunotherapies are only in their infancy. The ideal transplantable tumour that reiterates the key features of a malignant primary astrocytoma (poorly immunogenic, infiltrative but non-metastatic, expressing multiple mechanisms mediating immune escape) has yet to be discovered. In the meantime, we must use individual model tumours and limit the scope of the conclusions that we make from each system. Moreover, we must overcome the significant technical difficulties encountered as we strive to preserve brain integrity, whilst implanting tumours in this unique site. Or we can look to genetic models, in which there is the development of ‘spontaneous’ brain tumours (in some cases aided by the intracerebral delivery of a viral vector) incorporating many of the genetic features and heterogeneity typical of spontaneous human cancer.1 However, these models have generally been constructed to address genetic and pathological issues and they present a significant challenge for interpretable immunological studies.

With these difficulties in mind, perhaps we need to accept that brain tumour models and clinical immunotherapies are currently in their first generation. To progress to a more successful second generation of therapies, there is a need to abandon the idea that an automatic one-way progression exists from rodent models to the clinic. We need better models to make better therapies, but how to choose and design the models can be greatly guided by data from clinical trials, if the trial design actually permits the generation of useful biological data. To date, most brain tumour immunotherapies have borrowed technologies and approaches already explored for tumours in other sites, principally melanoma. Thus, most of the now ‘classical’ tumour immunology approaches have been explored for brain tumour immunotherapy: cytokine immune enhancement, whole tumour cell vaccines, cytokine-modified tumour cell vaccines, gene therapies with immune bystander effects, dendritic cell therapies (reviewed in refs. 24). A few notable and exciting novel approaches unique to the CNS should not be passed aside, for example, exploiting the migratory properties of neural stem cells to deliver immunoactive molecules efficiently to the tumour site.5 The overall conclusions from these studies are that, depending upon the stringency of the models utilized, tumours that are pre-established in the brain are generally more difficult to eliminate than those in other sites, and may require different effector mechanisms. For example, studies in which multiple cytokines have been tested as modulators of immune responses gave different results according to the tumour model (SMA-560, B16) and site of implantation.6,7 Other attempts to create a cellular vaccine by overexpression of intercellular adhesion molecule-1 on a glioma cell line, resulted in growth inhibition of glioma cells implanted subcutaneously, but not in the CNS.8 Another recent study using a recombinant Listeria monocytogenes tumour vaccine revealed more stringent T-cell subset requirements for protection against an intracerebral challenge compared with the same tumour implanted by the subcutaneous route.9 These examples, together with the fact that no convincingly successful clinical brain tumour immunotherapy has been demonstrated to date, should force us to reassess what we understand about brain tumour immunology, rather than just brain tumour immunotherapy. It is from this perspective that we will discuss the issues pertinent to the problem by drawing from both clinical and experimental situations.

The clinical problem

Both primary brain tumours and intracranial metastases pose a serious clinical problem, although their involvement with the host immune system will presumably have followed a different evolution. Concerning primary brain neoplasms, those derived from astrocytes are the most frequent, and the anaplastic astrocytomas and glioblastomas (grade III and IV astrocytomas, according to the WHO designation) are the most lethal. Indeed, despite some advances in surgical resection, radiotherapy and chemotherapy,10 it is unlikely that long-term survival rates can be significantly extended beyond the current median survival of less than 12 months for glioblastomas.11 Furthermore, although for low-grade astrocytomas the outlook is more favourable, not all can be adequately treated and they may progress to malignant lesions.1 Malignant astrocytomas infiltrate normal tissue, which renders total surgical resection virtually impossible without extensive neurological damage. It is thus essential to consider novel treatments such as immunotherapy in the hope of attacking the residual radioresistant and chemoresistant tumour cells. Other tumours metastasizing to the brain, such as melanoma, pose similar problems,12 and for these tumours it would also be useful to propose a therapeutic option that is applicable to the CNS.

The particular requirements for immune responses against tumours in the cns – a site of immune privilege

Whilst certain criteria for acceptable anti-tumour responses are applicable to all tumours, there are other requirements that are more stringent for cerebral malignancies. Indeed, the categorization of the CNS as an immune privileged site has perhaps retarded the development of immunotherapies for brain tumours, with the temptation to anticipate ineffective immune function in the brain. However, this prejudgement of the domain has arisen from a degree of misunderstanding of immune privilege. The term originally arose to describe the results of transplantation experiments in which there was extended survival of tissues transplanted to the CNS, compared with their survival in other sites.13,14 Features of the CNS were identified (certain of which are discussed individually in the sections below) that were proposed to explain this apparent lack of immune reactivity. These included the presence of the blood–brain barrier (BBB), low major histocompatibility complex (MHC) expression in the brain parenchyma, the absence of organized lymphatic drainage and a lack of dendritic cells in the normal brain parenchyma. Nevertheless, it is now apparent that immune reactions can and do occur in the CNS: autoimmune diseases of the CNS;15 immune responses to neurotropic viruses16 and parasites;17 and, as will be discussed herein, anti-tumour responses. Immune privilege is thus a term that requires an updated definition: it should remind us that although immune responses in the brain are often qualitatively and quantitatively different to those found in other sites, they are not absent. As elegantly phrased by Fabry et al.,18 we have to consider immune responses in the CNS as having a certain ‘dialect’.

Those aspects of immune privilege that impinge on afferent immune responses are clearly of fundamental interest, but may be less critical for brain tumour immunotherapy. For most vaccine strategies, immune responses will be induced at sites remote from the tumour, with the aim that effector cells can then recirculate to mediate their anti-tumour effects in the brain. The discussions that follow will therefore concentrate on those particularities of the CNS that influence the efferent arm of anti-tumour immune responses (Fig. 1), with a bias towards those that are presumed to be T-cell-mediated.

Figure 1.

Figure 1

Overview of the obstacles encountered by effector T cells primed in the periphery as they migrate towards a tumour situated in the brain parenchyma. After extravasation through the intact or locally compromised blood–brain barrier (BBB) (1),1 primed T cells encounter CNS-resident cells such as microglia and astrocytes (2), capable of antigen presentation, tolerance induction, or cytokine-mediated immunomodulation. Constitutive immunosuppressive factors present in the brain parenchyma (3) may also impede full differentiation or expression of effector molecules. As infiltrating T cells reach the tumour bed, they will be exposed to high concentrations of tumour-secreted factors that may synergize with tumour cell surface molecules to down-regulate effector function (4). Finally, efficient immune recognition of the tumour cell may be compromised by passive mechanisms of immune escape, such as low or absent MHC molecule expression and inefficient processing and presentation of tumour-associated antigens (5). Further details and references for these mechanisms are in the text.

Effector T cells must penetrate the brain parenchyma before they can reach the tumour bed

The first requirement for an effector T cell is that it must reach its target, the tumour. The problem of adequate tumour infiltration is applicable to all solid cancers.19,20 but when the tumour is located in the brain parenchyma, the T cell must also penetrate the tight junctions between the endothelial cells of the cerebral vasculature constituting the BBB.21,22 The integrity of this barrier is maintained by cells in intimate contact with the abluminal surface of the endothelium. Pericytes, perivascular cells and particularly astrocytes are implicated, the latter cells almost totally surrounding the vessel with their foot processes.23 Whilst the brain microvessels constituting the BBB appear impermeable compared with other microvessels, the barrier is conditional and selective. There is molecular and cellular traffic in both directions, but this is tightly regulated. Activated T cells can extravasate, but their trafficking may be more limited than for other sites. For example, the relative number of activated T cells found in the brain parenchyma after intravenous adoptive transfer of labelled T cells in rats was six times less than in muscle and more than 140 times less than that found in liver, for the same weight of tissue.24 For CD4+ T cells, migration away from the perivascular space into the parenchyma is inhibited in mice depleted of macrophages, suggesting that there are essential interactions with a perivascular cell after extravasation.25 The crux of the issue for tumour rejection is whether sufficient T cells reach their target to exert their anti-tumour effect. It is very difficult to quantify this for any tumour, nevertheless, under optimized conditions in different animal models, CD8+ T-cell-dependent immune responses are able to mediate anti-tumour effects in the brain.6,9,2633 For spontaneous malignant astrocytoma in humans, the integrity of the BBB is locally compromised, and tumour-induced angiogenesis will not incorporate BBB characteristics. T-cell infiltration frequently occurs,3436 but has only occasionally been correlated with a favourable prognosis.37 However, to date, the specificity and function of astrocytoma-infiltrating lymphocytes remains to be defined.

Target antigens must permit adequate discrimination of normal and malignant tissue

If we consider the CD8+ cytotoxic T lymphocyte (CTL) as the prime effector T cell for tumour rejection, its fine specificity is of critical importance for the brain. A degree of collateral damage is considered acceptable for tumours in certain extracerebral locations38 and many defined experimental vaccines aim to induce CTLs that recognize differentiation antigens, such as Melan-A, expressed in melanoma cells and normal melanocytes.39 Nevertheless, the severity of the autoimmune component of experimental melanoma vaccines, compared with their anti-tumour efficacy is being closely monitored.40,41 For the CNS, autoimmune reactions in the brain can be very serious, because most tissue is indispensable and has limited capacity for self-renewal. Indeed, early tumour immunology studies noted that lethal allergic encephalomyelitis was induced in different species by immunization with human glioma tissue.42 It is clear that a better defined tumour vaccine will have a greater chance of avoiding such unacceptable results. However, this necessitates identifying antigens for T cells expressed by brain tumours.

For malignant astrocytomas, identification of antigens recognized by T cells is far less advanced than for melanomas. However, certain antigenic similarities may be expected between melanomas and astrocytomas because their normal tissue counterparts derive from the neuroectoderm. Most studies addressing this possibility have analysed antigen expression at the mRNA level by reverse transcription–polymerase chain reaction. An initial study detected a proportion of tumours expressing one of several melanoma-associated antigens including MAGE family members, tyrosinase, TRP-1, TRP-2, gp100 and p97.43 However, subsequent publications found a much lower proportion of tumours expressing MAGE antigens, although occasional expression of other cancer-testis antigens was noted (SSX-1, SSX-2, SSX-4, SCP-1, TS85, and MAGE and GAGE family members).44,45 Although these results underline the potential antigenicity of certain brain tumours, some of the antigens that have a relatively high frequency of expression in malignant astrocytomas may be less useful as targets for immunotherapy because of their high homology to self-antigens (e.g. tumour-expressed GAGE-3 to GAGE-6 and GAGE-8 that are homologous to normal-brain-expressed GAGE-2 and GAGE-7).45 Furthermore, the only confirmation that epitopes from any of these antigens are presented at the cell surface for recognition by T cells comes from the study of certain astrocytoma cell lines that can be recognized by MAGE-specific CTL.46 However, this result reflects MAGE-1 antigen expression in cultured cells, whereas in vivo, astrocytomas are generally found to be negative for the MAGE-1 protein.45,47 This can probably be explained by a different level of DNA methylation induced by culture, because this regulates MAGE expression.48

A further candidate astrocytoma antigen that warrants further investigation is SART1259, originally identified in epithelial cancer cells and now shown to be expressed in various brain tumour lines and biopsies, including malignant astrocytoma.49 Astrocytoma cell lines expressing high levels of HLA-A24 and SART1259 could be recognized and killed by specific CTL derived from an oesophageal cancer patient, although no evidence of autologous responses was presented. Another possibility for an astrocytoma-associated antigen expression may be from neoepitopes present in epidermal growth factor receptor variant III (EGFR-vIII), expressed in a large proportion of malignant brain tumours.50 This antigen has until recently been explored as a target for monoclonal antibodies (initially for diagnostic purposes, but more recently for therapeutic application in preclinical mouse models.51 However, the possibility that EGFR-vIII-encoded T-cell epitopes may contribute to the anti-tumour response has been investigated in mouse models.52,53 and a vaccine incorporating a peptide from EGFR-vIII is currently under clinical trial.

There must be sufficient MHC molecule expression by tumour cells for direct CTL attack

For most primary brain tumours, the issue of MHC expression is rather different to that in tumours derived from tissues of non-CNS origin, because the normal tissue counterparts of astrocytes and oligodendrocytes are essentially MHC negative or low.54,55 It is thus essential that MHC molecules are induced either during tumorigenesis or during immunotherapy-induced anti-tumour responses if classical CTL-mediated cytotoxicity is to be operational. For tumour cells of astrocytic origin, these uniformly express MHC class I molecules after in vitro culture, and can be induced to express MHC class II after interferon-γ (IFN-γ) treatment (refs 46, 5658 and our own unpublished observations). The situation in vivo is far from clear, with contradictory reports in the literature.54,55,59,60 This may be because of differences in the immunohistochemistry protocols employed60 and difficulties in obtaining optimal staining for tumours such as high-grade astrocytomas that are characterized by zones of necrosis.

The generally low MHC expression by normal tissue may actually be an advantage during immunotherapy in that it may spare normal tissue from immune attack. However, normal astrocytes show inducible MHC expression, particularly after incubation with IFN-γ in vitro.56,6165 In vivo, as for neoplastic cells, the situation is more complicated. Based on data from mouse models, some studies have demonstrated that astrocytes can be induced to express MHC class I molecules after viral infection or exposure to IFN-γ.6668 However, other authors have suggested that in the absence of degenerated neurones, the oligodendrocyte and the microglial cell are the principal MHC-expressing cells of the non-malignant brain.69,70 It is clear that this issue of MHC expression of both normal and neoplastic tissue, before and at various stages during treatment, will be important in attempts to measure both positive and any adverse effects for assessment of therapies.

Brain inflammation must be regulated

Uncontrolled inflammation in the CNS rapidly leads to a severe augmentation of intracranial pressure because of the volume limitations within the confines of the skull. This may augment and exacerbate the cerebral oedema caused by the presence of intracerebral tumour mass, compromising neurological function and causing loss of critical cells for which renewal cannot be assured. Furthermore, chronic neuroinflammation may release sequestered autoantigens and contribute to the initiation or perpetuation of autoimmune reactions. Indeed, in a recent rodent gene therapy model for glioblastoma, there was evidence that intracerebral immune responses may have directly or indirectly promoted neuroinflammation and demyelination.71

Unfortunately, inflammation is often the accompaniment to vigorous immune responses induced by successful tumour vaccines, with some evidence that this may be occurring (although without mortality) in a mouse model in which there is rapid rejection of an immunogenic tumour implanted intracerebrally.32 In patients, inflammation is often controlled by administration of steroids, a treatment incompatible with efficacious induction of immune responses. However, there may be a brief and precious window of opportunity for immunotherapy immediately postoperatively whilst tumour burden is minimal and when steroids are not required. Ideally, future immunotherapies will be tailored for the CNS to incorporate limited pro-inflammatory elements. To date, eradication of pre-established, poorly immunogenic intracerebral tumours is still such a major challenge, even in animal models, that there has been little attention paid to ways of minimizing the inflammatory component of the response. Recent advances in understanding how brain-resident cells can contribute to CNS inflammation suggest that successful future immunotherapies will take these local factors into account. For example, based on in vitro data, astrocytes are proposed to stimulate more efficiently T helper type 2 (Th2) T cells whereas activated microglial cells, the specialized macrophages of the brain, stimulate a pro-inflammatory Th1 response.65,72,73 It remains to be determined whether brain-infiltrating lymphocytes in vivo are susceptible to Th subset deviation after contact with astrocytes or microglial cells, even if they were primed in secondary lymphoid organs by other antigen-presenting cells (APCs). The wide functional plasticity of reactive (activated) astrocytes is such that various roles are possible, including a fine tuning of brain inflammation. Thus, depending upon the presence of microenvironmental factors such as IFN-γ or tumour necrosis factor (TNF) and TNF-receptor family members, astrocytes can migrate, release pro-inflammatory cytokines such as interleukin-6 (IL-6) and IL-8, or undergo apoptosis.7477 Which of these roles predominates in vivo remains to be determined. However, interesting data are emerging from transgenic mouse models in which the glial fibrillary acidic protein (GFAP)-promoter targets expression of various cytokines (IL-3, IL-6, IL-12, IFN-α and TNF) to astrocytes (reviewed in ref. 78). Although such transgenic expression may appear extreme, the levels of transgene-encoded cytokine were similar to those detected in inflamed or infected brains. With age, these mice developed inflammation and degenerative neurological disease.

Anti-tumour effector function must be retained as T cells migrate through the brain parenchyma and encounter resident cells

Although we have described that T cells are able to infiltrate the brain, can effector T cells retain their function as they penetrate the brain parenchyma and approach the tumour mass? Evidence from different models suggests that this will depend upon several factors, such as the magnitude of the induced immune response, the presence of other cell types and the particular subset of the effector cell.

For brain infiltrating T cells, even certain components of normal brain have been suggested to inhibit CTL effector function. For example, T cells otherwise capable of mediating neuropathology were inhibited by brain-derived gangliosides,79 although this was strain-specific in the mouse, and has not been confirmed in humans. For anti-tumour effector T cells, in the intracerebral P511 mastocytoma tumour model, CD8+ T cells were unable to differentiate into effector cells in the brain microenvironment,80 a finding that was subsequently attributed to a sensitivity to transforming growth factor-β (TGF-β) present in the cerebrospinal fluid and the interstitial tissue fluid.81 However, these results were in contrast to those obtained with the similar P815 mastocytoma, which had been transfected with a model antigen, CW3 (P815-CW3): brain-infiltrating lymphocytes tested ex vivo from these mice showed full cytotoxic effector function.32 One difference between these systems was that in the P815-CW3 model, this was a syngeneic response to a defined peptide antigen, whereas in the P511 model most of the experiments were performed in outbred or non-syngeneic mice.

Before the final contact with the tumour cell, the infiltrating T cell may have other encounters with putative APCs of the brain. This has been most clearly defined for MHC class II-restricted CD4+ T cells, although not as yet for cells with specificity for a tumour-expressed antigen. The microglial cell is one of the few MHC class II+ cells that are resident in the brain and is thus a prime candidate brain APC. As already mentioned, CD4+ T cells require contact with a macrophage-like perivascular cell to advance into the brain parenchyma,25 however, contact with parenchymally localized microglial cells has been suggested to lead to tolerance induction or the termination of immune responses.8285 Such effects were particularly associated with the activation state of the microglial cell.83 Whether microglial cells are implicated in antigen presentation to CD8+ T cells is not known, however, they may indirectly influence CD8+ T-cell function through their influence on CD4+ T cells. Defined antigen-specific CTL responses in the CNS have principally been studied in antiviral responses that are often strictly CD4+ T-cell-dependent, with loss of cytotoxic function and/or viability in the absence of Th cells.86,87 However, for anti-tumour immune responses, independence from CD4+ T cells has been demonstrated in several different models.6,30,32 although it is difficult to generalize because so few studies have looked at this aspect in systems that permit the analysis of specific CTL responses.

The last frontier: retention of effector cell function during the encounter with the tumour cell

The last hurdle to overcome for an anti-tumour effector cell is to function when in intimate contact with the tumour cell. It is at this stage that active mechanisms of tumour immune escape will be most potent: the concentration of soluble immunosuppressive molecules is maximal and may synergize with potential cell-mediated immunosuppressive effects.

There has been a wealth of publications over the past three decades describing immunological defects in astrocytoma patients. These include abnormal delayed hypersensitivity responses, low numbers of circulating T cells, depressed mitogen responsiveness, decreased antibody responses (probably as a result of defective CD4 T helper cell activity) and impaired T-cell cytotoxicity (reviewed by Dix et al.88). It is reasonable to assume that these defects will be at their most extreme in the vicinity of the tumour, and that they can explain why tumour-infiltrating lymphocytes (TILs) are apparently inefficacious. However, until it can be confirmed that the TILs are really specific for a tumour-expressed antigen, the link remains speculative. Nevertheless, many advances have been made in dissecting the underlying causes for this apparent hyporesponsiveness. It appears that T cells from astrocytoma patients, particularly TILs, express a defective high-affinity IL-2 receptor, or have severe defects in their signalling pathway of activation.89 Similar findings were also noted in a rat glioma model.90 This may explain, at least in part, the poor in vitro proliferative abilities of T lymphocytes infiltrating malignant astrocytoma, despite the addition of recombinant IL-2,91 their low IL-2 production after mitogen stimulation,92 and the difficulty in generating T-cell clones with MHC-restricted cytotoxicity against tumour cells.93

Immunosuppression by soluble factors

Soluble factors produced within the tumour microenvironment have long been suspected to be behind many of the above mentioned immune defects. Indeed, T lymphocytes from normal individuals exhibit similar immunological abnormalities when cultured in the presence of astrocytoma supernatant.94 The most extensively studied soluble factor is TGF-β2, originally called glioblastoma cell-derived T-cell suppressor factor, which was first identified in the supernatant of a human glioblastoma cell line that suppressed T-cell growth.9597 TGF-β2 exerts multiple and complex immunosuppressive effects, such as the inhibition of maturation and antigen presentation by dendritic cells or other APCs, inhibition of T-cell activation and differentiation towards effector cells (either cytotoxic cells expressing perforin or Th1 or Th2 cells).98100 The role of TGF-β2 in inducing immunosuppression was further demonstrated in experiments in which anti-sense TGF-β2 phosphorothioate oligonucleotides inhibited TGF-β2 secretion by glioblastoma cell lines, restoring the proliferative and cytotoxic functions of autologous lymphocytes.101 These in vitro data naturally encouraged attempts to inhibit TGF-β2-mediated immunosuppression in vivo, which has produced mixed results, probably because TGF-β2 not only influences immune reactivity, but also acts on the tumour cell directly.102,103 Other studies, using decorin, a natural inhibitor of TGF-β, suppressed the growth of C6 rat astrocytoma in vivo,104 but these experiments are also complex to interpret because decorin may also be immunostimulatory in a TGF-β2 independent fashion.105 Overall, the immunoregulatory functions of TGF-β2 warrant the attention it has received, but whether it will be feasible or advisable to inhibit this cytokine in brain tumour patients still remains uncertain.

The immunosuppressive properties of malignant astrocytoma-derived supernatants demonstrated in vitro cannot be totally accounted for by TGF-β2. A non-exhaustive list of potentially immunosuppressive molecules detected in astrocytomas or astrocytoma lines includes prostaglandin E2,88,96,106,107 gangliosides108 and IL-10,109111 all of which can demonstrate certain immunosuppressive functions in vitro. However, whether these factors are playing a major role in inhibiting anti-tumour immune responses in vivo is far less certain, either because of doubts that sufficient bioactive factor is released by tumour cells in vivo (discussed in ref. 88), or, particularly for IL-10, because in vivo function is not necessarily immunosuppressive, but may actually enhance anti-tumour responses.112,113

Immunosuppression by cell-mediated interactions

Intercellular contacts through transmembrane molecules such as Fas ligand (FasL) are also proposed to contribute to tumour immune escape. FasL belongs to the TNF family and is implicated in several biological functions through its interaction with Fas, a member of the TNF receptor/nerve growth factor receptor family. FasL–Fas interaction induces the trimerization of Fas and a subsequent complex cascade of intracellular events, potentially leading to apoptosis of Fas-positive cells, a mechanism central to immune homeostasis.114,115 Although FasL was initially thought to be mainly expressed by cells of haematopoietic origin, it was subsequently shown to be expressed by other normal and neoplastic tissues, including malignant astrocytomas that we have analysed in our laboratory.116 Expression of FasL in a series of human astrocytoma lines and biopsies was tested, as well as in the rat C6 and mouse MT539MG glioma lines, using a variety of techniques (immunoblot, flow cytometry and reverse transcription–polymerase chain reaction): the majority were positive. Functional data demonstrated that FasL+ astrocytoma cells (cell lines and also astrocytoma cells tested ex vivo) can specifically and efficiently kill Fas-transfected P815 target cells. Moreover, an early passage human astrocytoma cell line was able to induce FasL/Fas-mediated apoptosis in CD4+ and CD8+ T cell lines derived from the autologous tumour.117 It has been suggested that the use of T-cell targets is inappropriate to detect FasL-mediated function by tumour cells, because T cells can also express FasL.118 However, the kinetics of sensitivity of our T cells to FasL-mediated death are inversely correlated to their endogenous FasL expression; this peaks within the first day after re-stimulation, then rapidly diminishes and is undetectable at the time-points when T cells were tested for Fas sensitivity. Overall these data suggest that astrocytoma cells express functional FasL that can induce apoptosis in Fas+ targets and that TILs, whilst capable of undergoing apoptosis through fratricide or autocrine suicide, can also be susceptible (at least in vitro), of receiving a death signal from FasL-expressing astrocytoma cells. Other groups have independently confirmed expression of FasL by astrocytoma119121 and, moreover, apoptotic T cells were observed in the proximity of FasL-expressing astrocytoma cells.122 However, the real in vivo importance of this molecule for astrocytoma, and tumours in general, remains unclear. Indeed, tumour expression of FasL in murine models has been correlated either with enhanced tumour growth123,124 or with enhanced tumour rejection,125127 probably via augmented neutrophil recruitment. Microenvironmental factors clearly influence the consequences of FasL expression by tumour cells, for example, we hypothesized that tumours expressing both FasL and TGF-β may be particularly well adapted to combat CTL effector mechanisms,117 this principle was subsequently confirmed in an in vivo model in which a FasL-positive colon carcinoma could escape rejection if TGF-β was also present.128 These data help to explain the role of FasL in this particular model (with possible analogy to the situation in the brain), but it is likely that individual combinations of factors relevant to different tumours or models are responsible for the diverse interpretations of these issues in the literature.117,118,129,130

Recent additions to the list of astrocytoma-expressed molecules with immunosuppressive potential are HLA-G131 and CD70,132,133 although the in vivo validation of their function has yet to be reported. HLA-G is a non-classical MHC class I molecule expressed by a limited range of tissues, particularly the placenta, but also certain cancers. Roles in suppressing natural killer and T-cell immune responses have been proposed, but they are controversial.134,135 Regarding brain tumours, a proportion of astrocytoma cell lines and tumour biopsies expressed HLA-G protein. Functional data showed inhibition of CD4+ and CD8+ T-cell responses 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. For the transmembrane glycoprotein CD70, previous studies have described expression of this molecule on activated T and B cells, with roles in regulating immune responses via interaction with CD27, expressed on lymphoid cells.136 Two independent studies have now described expression of CD70 by human glioma cell lines, as well as in vivo.132,133In vitro functional tests suggested a pro-apoptotic role of CD70 when tested on peripheral blood mononuclear cells targets; this was augmented (correlating with expression levels of CD70) when tumour cells were irradiated.132 However, under certain conditions, T cells shed CD27 after stimulation by CD70+ tumour cells, indicating a possible protection mechanism.

Concluding remarks

Will the CNS present an impossible obstacle course for immune effector cells induced during any future brain tumour immunotherapy? There are sufficient encouraging results from certain models to suggest that this will not be the case, because it may not be necessary to overcome all of the mechanisms of immune escape or restrictions imposed by the CNS to achieve some clinical benefit. However, with a better understanding of immune responses in the brain, we can make rational proposals to identify key areas in which to attempt immunomodulation for future brain tumour therapies.

The fundamental importance of understanding immune relations of the brain in health and disease cannot be overstated. It can be assumed that the special features of CNS immune responses, whilst apparently an impediment to the cancer immunotherapist, have evolved to protect this most critical organ of the body. We modify the neuroimmunological balance at our peril. However, the optimal balance that has evolved for a species may not be optimal for an individual. We should therefore not be deterred from shifting the balance towards a protective immune response in patients, as long as the neuropathological risks are understood and monitored.

Acknowledgments

The work in our laboratory is partly supported by grants from OncoSuisse, the International Association for Cancer Research, the Fondation Lionel Perrier and the Ligue Genevoise Contre le Cancer.

Abbreviations

APC

antigen-presenting cell

BBB

blood–brain barrier

CNS

central nervous system

CTL

cytotoxic T lymphocyte

IFN

interferon

IL

interleukin

PG

prostaglandin

TIL

tumour-infiltrating lymphocyte

TGF

transforming growth factor

References

  • 1.Holland EC. Gliomagenesis: genetic alterations and mouse models. Nat Rev Genet. 2001;2:120–9. doi: 10.1038/35052535. [DOI] [PubMed] [Google Scholar]
  • 2.Dietrich P-Y, Walker PR, Saas P, de Tribolet N. Immunobiology of gliomas: New perspectives for therapy. Ann N Y Acad Sci. 1997;824:124–40. doi: 10.1111/j.1749-6632.1997.tb46215.x. [DOI] [PubMed] [Google Scholar]
  • 3.Soling A, Rainov NG. Dendritic cell therapy of primary brain tumors. Mol Med. 2001;7:659–67. [PMC free article] [PubMed] [Google Scholar]
  • 4.Parney IF, Hao C, Petruk KC. Glioma immunology and immunotherapy. Neurosurgery. 2000;46:778–91. doi: 10.1097/00006123-200004000-00002. [DOI] [PubMed] [Google Scholar]
  • 5.Benedetti S, Pirola B, Pollo B, et al. Gene therapy of experimental brain tumors using neural progenitor cells. Nat Med. 2000;6:447–50. doi: 10.1038/74710. [DOI] [PubMed] [Google Scholar]
  • 6.Sampson JH, Archer GE, Ashley DM, Fuchs HE, Hale LP, Dranoff G, Bigner DD. Subcutaneous vaccination with irradiated, cytokine-producing tumors cells stimulates CD8+ cell-mediated immunity against tumors located in the ‘immunologically privileged’ central nervous system. Proc Natl Acad Sci USA. 1996;93:10399–404. doi: 10.1073/pnas.93.19.10399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sampson JH, Ashley DM, Archer GE, Fuchs HE, Dranoff G, Hale LP, Bigner DD. Characterization of a spontaneous murine astrocytoma and abrogation of its tumorigenicity by cytokine secretion. Neurosurgery. 1997;41:1365–73. doi: 10.1097/00006123-199712000-00024. [DOI] [PubMed] [Google Scholar]
  • 8.Kikuchi T, Joki T, Akasaki Y, Abe T, Ohno T. Induction of antitumor immunity using intercellular adhesion molecule 1 (ICAM-1) transfection in mouse glioma cells. Cancer Lett. 1999;142:201–6. doi: 10.1016/s0304-3835(99)00169-x. [DOI] [PubMed] [Google Scholar]
  • 9.Liau LM, Jensen ER, Kremen TJ, Odesa SK, Sykes SN, Soung MC, Miller JF, Bronstein JM. Tumor immunity within the central nervous system stimulated by recombinant Listeria monocytogenes vaccination. Cancer Res. 2002;62:2287–93. [PubMed] [Google Scholar]
  • 10.Stupp R, Dietrich PY, Ostermann KS, et al. Promising survival for patients with newly diagnosed glioblastoma multiforme treated with concomitant radiation plus temozolomide followed by adjuvant temozolomide. J Clin Oncol. 2002;20:1375–82. doi: 10.1200/JCO.2002.20.5.1375. [DOI] [PubMed] [Google Scholar]
  • 11.Stewart LA. Chemotherapy in adult high-grade glioma: a systematic review and meta-analysis of individual patient data from 12 randomised trials. Lancet. 2002;359:1011–18. doi: 10.1016/s0140-6736(02)08091-1. [DOI] [PubMed] [Google Scholar]
  • 12.Arnold SM, Patchell RA. Diagnosis and management of brain metastases. Hematol Oncol Clin North Am. 2001;15:1085–107. doi: 10.1016/s0889-8588(05)70269-0. [DOI] [PubMed] [Google Scholar]
  • 13.Medewar PB. Immunity to homologous grafted skin. III. 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;29:58–69. [PMC free article] [PubMed] [Google Scholar]
  • 14.Barker CF, Billingham RE. Immunologically privileged sites. Adv Immunol. 1977;25:1–54. [PubMed] [Google Scholar]
  • 15.Hohlfeld R, Wekerle H. Immunological update on multiple sclerosis. Curr Opin Neurol. 2001;14:299–304. doi: 10.1097/00019052-200106000-00006. [DOI] [PubMed] [Google Scholar]
  • 16.Dorries R. The role of T-cell-mediated mechanisms in virus infections of the nervous system. Curr Top Microbiol Immunol. 2001;253:219–45. doi: 10.1007/978-3-662-10356-2_11. [DOI] [PubMed] [Google Scholar]
  • 17.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;164:4826–34. doi: 10.4049/jimmunol.164.9.4826. [DOI] [PubMed] [Google Scholar]
  • 18.Fabry Z, Raine CS, Hart MN. Nervous tissue as an immune compartment: the dialect of the immune response in the CNS. Immunol Today. 1994;15:218–24. doi: 10.1016/0167-5699(94)90247-X. [DOI] [PubMed] [Google Scholar]
  • 19.Nelson DJ, Mukherjee S, Bundell C, Fisher S, van Hagen D, Robinson B. Tumor progression despite efficient tumor antigen cross-presentation and effective ‘arming’ of tumor antigen-specific CTL. J Immunol. 2001;166:5557–66. doi: 10.4049/jimmunol.166.9.5557. [DOI] [PubMed] [Google Scholar]
  • 20.Ochsenbein AF, Sierro S, Odermatt B, et al. Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature. 2001;411:1058–64. doi: 10.1038/35082583. [DOI] [PubMed] [Google Scholar]
  • 21.Brightman MW, Reese TS. Junctions between intimately apposed cell membranes in the vertebrate brain. J Cell Biol. 1969;40:648–77. doi: 10.1083/jcb.40.3.648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Rubin LL, Staddon JM. The cell biology of the blood–brain barrier. Annu Rev Neurosci. 1999;22:11–28. doi: 10.1146/annurev.neuro.22.1.11. [DOI] [PubMed] [Google Scholar]
  • 23.Janzer RC, Raff MC. Astrocytes induce blood–brain barrier properties in endothelial cells. Nature. 1987;325:253–7. doi: 10.1038/325253a0. [DOI] [PubMed] [Google Scholar]
  • 24.Hickey WF. Leukocyte traffic in the central nervous system: the participants and their roles. Semin Immunol. 1999;11:125–37. doi: 10.1006/smim.1999.0168. [DOI] [PubMed] [Google Scholar]
  • 25.Tran EH, Hoekstra K, Van Rooijen N, Dijkstra CD, Owens T. Immune invasion of the central nervous system parenchyma and experimental allergic encephalomyelitis, but not leukocyte extravasation from blood, are prevented in macrophage-depleted mice. J Immunol. 1998;161:3767–75. [PubMed] [Google Scholar]
  • 26.Aoki T, Tashiro K, Miyatake S, Kinashi T, Nakano T, Oda Y, Kikuchi H, Honjo T. Expression of murine interleukin 7 in a murine glioma cell line results in reduced tumorigenicity in vivo. Proc Natl Acad Sci USA. 1992;89:3850–4. doi: 10.1073/pnas.89.9.3850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Asai A, Miyagi Y, Hashimoto H, et al. Modulation of tumor immunogenicity of rat glioma cells by s-myc expression: eradication of rat gliomas in vivo. Cell Growth Differ. 1994;5:1153–8. [PubMed] [Google Scholar]
  • 28.Resnicoff M, Sell C, Rubini M, Coppola D, Ambrose D, Baserga R, Rubin R. Rat glioblastoma cells expressing an antisense RNA to the insulin-like growth factor-1 (IGF-1) receptor are nontumorigenic and induce regression of wild-type tumors. Cancer Res. 1994;54:2218–22. [PubMed] [Google Scholar]
  • 29.Ashley DM, Sampson JH, Archer GE, Batra SK, Bigner DD, Hale LP. A genetically modified allogeneic cellular vaccine generates MHC class I-restricted cytotoxic responses against tumor-associated antigens and protects against CNS tumors in vivo. J Neuroimmunol. 1997;78:34–46. doi: 10.1016/s0165-5728(97)00080-5. [DOI] [PubMed] [Google Scholar]
  • 30.Okada H, Tahara H, Shurin MR, et al. Bone marrow-derived dendritic cells pulsed with a tumor-specific peptide elicit effective anti-tumor immunity against intracranial neoplasms. Int J Cancer. 1998;78:196–201. doi: 10.1002/(sici)1097-0215(19981005)78:2<196::aid-ijc13>3.0.co;2-9. [DOI] [PubMed] [Google Scholar]
  • 31.Graf MR, Jadus MR, Hiserodt JC, Wepsic HT, Granger GA. Development of systemic immunity to glioblastoma multiforme using tumor cells genetically engineered to express the membrane-associated isoform of macrophage colony-stimulating factor. J Immunol. 1999;163:5544–51. [PubMed] [Google Scholar]
  • 32.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;165:3128–35. doi: 10.4049/jimmunol.165.6.3128. [DOI] [PubMed] [Google Scholar]
  • 33.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]
  • 34.Rossi ML, Hughes JT, Esiri MM, Coakham HB, Brownell D. Immunohistological study of mononuclear cell infiltrate in malignant gliomas. Acta Neuropathol. 1987;74:269–77. doi: 10.1007/BF00688191. [DOI] [PubMed] [Google Scholar]
  • 35.Stavrou D, Anzil AP, Weidenbach W, Rodt H. Immunofluorescence study of lymphocytic infiltration in gliomas. Identification of T-lymphocytes. J Neurol Sci. 1977;33:275–82. doi: 10.1016/0022-510x(77)90200-3. [DOI] [PubMed] [Google Scholar]
  • 36.Perrin G, Schnuriger V, Quiquerez A-L, et al. Astrocytoma infiltrating lymphocytes include major T cell clonal expansions confined to the CD8 subset. Int Immunol. 1999;11:1337–49. doi: 10.1093/intimm/11.8.1337. [DOI] [PubMed] [Google Scholar]
  • 37.Brooks WH, Markesbery WR, Gupta GD, Roszman TL. Relationship of lymphocyte invasion and survival of brain tumor patients. Ann Neurol. 1978;4:219–24. doi: 10.1002/ana.410040305. [DOI] [PubMed] [Google Scholar]
  • 38.Pardoll DM. Inducing autoimmune disease to treat cancer. Proc Natl Acad Sci USA. 1999;96:5340–2. doi: 10.1073/pnas.96.10.5340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Renkvist N, Castelli C, Robbins PF, Parmiani G. A listing of human tumor antigens recognized by T cells. Cancer Immunol Immunother. 2001;50:3–15. doi: 10.1007/s002620000169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.van Elsas A, Sutmuller RP, Hurwitz AA, et al. Elucidating the autoimmune and antitumor effector mechanisms of a treatment based on cytotoxic T lymphocyte antigen-4 blockade in combination with a B16 melanoma vaccine. Comparison of prophylaxis and therapy. J Exp Med. 2001;194:481–90. doi: 10.1084/jem.194.4.481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Banchereau J, Palucka AK, Dhodapkar M, et al. Immune and clinical responses in patients with metastatic melanoma to CD34+ progenitor-derived dendritic cell vaccine. Cancer Res. 2001;61:6451–8. [PubMed] [Google Scholar]
  • 42.Bigner DD, Pitts OM, Wikstrand CJ. Induction of lethal experimental allergic encephalomyelitis in nonhuman 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]
  • 43.Chi DD, Merchant RE, Rand R, Conrad AJ, Garrison D, Turner R, Morton DL, Hoon DS. Molecular detection of tumor-associated antigens shared by human cutaneous melanomas and gliomas. Am J Pathol. 1997;150:2143–52. [PMC free article] [PubMed] [Google Scholar]
  • 44.Sahin U, Koslowski M, Tureci O, et al. Expression of cancer testis genes in human brain tumors. Clin Cancer Res. 2000;6:3916–22. [PubMed] [Google Scholar]
  • 45.Scarcella DL, Chow CW, Gonzales MF, Economou C, Brasseur F, Ashley DM. Expression of MAGE and GAGE in high-grade brain tumors: a potential target for specific immunotherapy and diagnostic markers. Clin Cancer Res. 1999;5:335–41. [PubMed] [Google Scholar]
  • 46.Rimoldi D, Romero P, Carrel S. The human melanoma antigen-encoding gene, MAGE-1, is expressed by other tumour cells of neuroectodermal origin such as glioblastomas and neuroblastomas. Int J Cancer. 1993;54:527–8. doi: 10.1002/ijc.2910540329. [DOI] [PubMed] [Google Scholar]
  • 47.De Smet C, Lurquin C, Van der Bruggen P, De Plaen E, Brasseur F, Boon T. Sequence and expression pattern of the human MAGE2 gene. Immunogenetics. 1994;39:121–9. doi: 10.1007/BF00188615. [DOI] [PubMed] [Google Scholar]
  • 48.De Smet C, Courtois SJ, Faraoni I, Lurquin C, Szikora JP, De Backer O, Boon T. Involvement of two Ets binding sites in the transcriptional activation of the MAGE1 gene. Immunogenetics. 1995;42:282–90. doi: 10.1007/BF00176446. [DOI] [PubMed] [Google Scholar]
  • 49.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;83:760–4. doi: 10.1002/(sici)1097-0215(19991210)83:6<760::aid-ijc11>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
  • 50.Libermann TA, Nusbaum HR, Razon N, et al. Amplification, enhanced expression and possible rearrangement of EGF receptor gene in primary human brain tumours of glial origin. Nature. 1985;313:144–7. doi: 10.1038/313144a0. [DOI] [PubMed] [Google Scholar]
  • 51.Sampson JH, Crotty LE, Lee S, et al. Unarmed, tumor-specific monoclonal antibody effectively treats brain tumors. Proc Natl Acad Sci USA. 2000;97:7503–8. doi: 10.1073/pnas.130166597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Moscatello DK, Ramirez G, Wong AJ. A naturally occurring mutant human epidermal growth factor receptor as a target for peptide vaccine immunotherapy of tumors. Cancer Res. 1997;57:1419–24. [PubMed] [Google Scholar]
  • 53.Heimberger AB, Archer GE, Crotty LE, McLendon RE, Friedman AH, Friedman HS, Bigner DD, Sampson JH. Dendritic cells pulsed with a tumor-specific peptide induce long-lasting immunity and are effective against murine intracerebral melanoma. Neurosurgery. 2002;50:158–64. doi: 10.1097/00006123-200201000-00024. [DOI] [PubMed] [Google Scholar]
  • 54.Lampson LA, Hickey WF. Monoclonal antibody analysis of MHC expression in human brain biopsies: tissue ranging from ‘histologically normal’ to that showing different levels of glial tumor involvement. J Immunol. 1986;136:4054–62. [PubMed] [Google Scholar]
  • 55.Natali PG, Bigotti A, Nicotra MR, Viora M, Manfredi D, Ferrone S. Distribution of human Class I (HLA-A,B,C) histocompatibility antigens in normal and malignant tissues of nonlymphoid origin. Cancer Res. 1984;44:4679–87. [PubMed] [Google Scholar]
  • 56.Dhib-Jalbut S, Kufta CV, Flerlage M, Shimojo N, McFarland HF. Adult human glial cells can present target antigens to HLA-restricted cytotoxic T-cells. J Neuroimmunol. 1990;29:203–11. doi: 10.1016/0165-5728(90)90163-H. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bruner JM, Langford LA, Fuller GN. Neuropathology, cell biology, and newer diagnostic methods. Curr Opin Oncol. 1993;5:441–9. doi: 10.1097/00001622-199305000-00002. [DOI] [PubMed] [Google Scholar]
  • 58.Parney IF, Farr-Jones MA, Chang LJ, Petruk KC. Human glioma immunobiology in vitro: implications for immunogene therapy. Neurosurgery. 2000;46:1169–77. doi: 10.1097/00006123-200005000-00030. [DOI] [PubMed] [Google Scholar]
  • 59.Saito T, Tanaka R, Yoshida S, Washiyama K, Kumanishi T. Immunohistochemical analysis of tumor-infiltrating lymphocytes and major histocompatibility antigens in human gliomas and metastatic brain tumors. Surg Neurol. 1988;29:435–42. doi: 10.1016/0090-3019(88)90137-1. [DOI] [PubMed] [Google Scholar]
  • 60.Facoetti A, Capelli E, Nano R. HLA class I molecules expression: evaluation of different immunocytochemical methods in malignant lesions. Anticancer Res. 2001;21:2435–40. [PubMed] [Google Scholar]
  • 61.Wong GH, Bartlett PF, Clark-Lewis I, Battye F, Schrader JW. Inducible expression of H-2 and Ia antigens on brain cells. Nature. 1984;310:688–91. doi: 10.1038/310688a0. [DOI] [PubMed] [Google Scholar]
  • 62.Weber F, Meinl E, Aloisi F, Nevinny-Stickel C, Albert E, Wekerle H, Hohlfeld R. Human astrocytes are only partially competent antigen presenting cells. Possible implications for lesion development in multiple sclerosis. Brain. 1994;117:59–69. doi: 10.1093/brain/117.1.59. [DOI] [PubMed] [Google Scholar]
  • 63.Massa PT, Ozato K, McFarlin DE. Cell type-specific regulation of major histocompatibility complex (MHC) class I gene expression in astrocytes, oligodendrocytes, and neurons. Glia. 1993;8:201–7. doi: 10.1002/glia.440080307. [DOI] [PubMed] [Google Scholar]
  • 64.Soos JM, Morrow J, Ashley TA, Szente BE, Bikoff EK, Zamvil SS. Astrocytes express elements of the class II endocytic pathway and process central nervous system autoantigen for presentation to encephalitogenic T cells. J Immunol. 1998;161:5959–66. [PubMed] [Google Scholar]
  • 65.Aloisi F, Ria F, Penna G, Adorini L. Microglia are more efficient than astrocytes in antigen processing and Th1 but not Th2 cell activation. J Immunol. 1998;160:4671–80. [PubMed] [Google Scholar]
  • 66.Christian AY, Barna M, Bi Z, Reiss CS. Host immune response to vesicular stomatitis virus infection of the central nervous system in C57BL/6 mice. Viral Immunol. 1996;9:195–205. doi: 10.1089/vim.1996.9.195. [DOI] [PubMed] [Google Scholar]
  • 67.Shrikant P, Benveniste EN. The central nervous system as an immunocompetent organ. Role of glial cells in antigen presentation. J Immunol. 1996;157:1819–22. [PubMed] [Google Scholar]
  • 68.Jarosinski KW, Massa PT. Interferon regulatory factor-1 is required for interferon-gamma-induced MHC class I genes in astrocytes. J Neuroimmunol. 2002;122:74–84. doi: 10.1016/s0165-5728(01)00467-2. [DOI] [PubMed] [Google Scholar]
  • 69.Horwitz MS, Evans CF, Klier FG, Oldstone MB. Detailed in vivo analysis of interferon-gamma induced major histocompatibility complex expression in the the central nervous system: astrocytes fail to express major histocompatibility complex class I and II molecules. Lab Invest. 1999;79:235–42. [PubMed] [Google Scholar]
  • 70.Redwine JM, Buchmeier MJ, Evans CF. In vivo expression of major histocompatibility complex molecules on oligodendrocytes and neurons during viral infection. Am J Pathol. 2001;159:1219–24. doi: 10.1016/S0002-9440(10)62507-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Dewey RA, Morrissey G, Cowsill CM, et al. Chronic brain inflammation and persistent herpes simplex virus 1 thymidine kinase expression in survivors of syngeneic glioma treated by adenovirus-mediated gene therapy: implications for clinical trials. Nat Med. 1999;5:1256–63. doi: 10.1038/15207. [DOI] [PubMed] [Google Scholar]
  • 72.Aloisi F, Ria F, Columba-Cabezas S, Hess H, Penna G, Adorini L. Relative efficiency of microglia, astrocytes, dendritic cells and B cells in naive CD4+ T cell priming and Th1/Th2 cell restimulation. Eur J Immunol. 1999;29:2705–14. doi: 10.1002/(SICI)1521-4141(199909)29:09<2705::AID-IMMU2705>3.0.CO;2-1. [DOI] [PubMed] [Google Scholar]
  • 73.Aloisi F, Ria F, Adorini L. Regulation of T-cell responses by CNS antigen-presenting cells: different roles for microglia and astrocytes. Immunol Today. 2000;21:141–7. doi: 10.1016/s0167-5699(99)01512-1. [DOI] [PubMed] [Google Scholar]
  • 74.Saas P, Boucraut J, Quiquerez A-L, et al. CD95 (Fas/Apo-1) as a receptor governing astrocyte apoptotic or inflammatory responses: a key role in brain inflammation. J Immunol. 1999;162:2326–33. [PubMed] [Google Scholar]
  • 75.Saas P, Boucraut J, Walker PR, Quiquerez AL, Billot M, Desplat-Jego S, Chicheportiche Y, Dietrich PY. TWEAK stimulation of astrocytes and the proinflammatory consequences. Glia. 2000;32:102–7. [PubMed] [Google Scholar]
  • 76.Lee SJ, Zhou T, Choi C, Wang Z, Benveniste EN. Differential regulation and function of Fas expression on glial cells. J Immunol. 2000;164:1277–85. doi: 10.4049/jimmunol.164.3.1277. [DOI] [PubMed] [Google Scholar]
  • 77.Van Wagoner NJ, Benveniste EN. Interleukin-6 expression and regulation in astrocytes. J Neuroimmunol. 1999;100:124–39. doi: 10.1016/s0165-5728(99)00187-3. [DOI] [PubMed] [Google Scholar]
  • 78.Campbell IL. Cytokine-mediated inflammation and signaling in the intact central nervous system. Prog Brain Res. 2001;132:481–98. doi: 10.1016/S0079-6123(01)32097-6. [DOI] [PubMed] [Google Scholar]
  • 79.Irani DN. The susceptibility of mice to immune-mediated neurologic disease correlates with the degree to which their lymphocytes resist the effects of brain-derived gangliosides. J Immunol. 1998;161:2746–52. [PubMed] [Google Scholar]
  • 80.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 tumour elimination. J Immunol. 1997;159:2399–408. [PubMed] [Google Scholar]
  • 81.Gordon LB, Nolan SC, Ksander BR, Knopf PM, Harling-Berg CJ. Normal cerebrospinal fluid suppresses the in vitro development of cytotoxic T cells: role of the brain microenvironment in CNS immune regulation. J Neuroimmunol. 1998;88:77–84. doi: 10.1016/s0165-5728(98)00077-0. [DOI] [PubMed] [Google Scholar]
  • 82.Brabb T, von Dassow P, Ordonez N, Schnabel B, Duke B, Goverman J. In situ tolerance within the central nervous system as a mechanism for preventing autoimmunity. J Exp Med. 2000;192:871–80. doi: 10.1084/jem.192.6.871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Matyszak MK, Denis-Donini S, Citterio S, Longhi R, Granucci F, Ricciardi-Castagnoli P. Microglia induce myelin basic protein-specific T cell energy or T cell activation, according to their state of activation. Eur J Immunol. 1999;29:3063–76. doi: 10.1002/(SICI)1521-4141(199910)29:10<3063::AID-IMMU3063>3.0.CO;2-G. [DOI] [PubMed] [Google Scholar]
  • 84.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;154:4309–21. [PubMed] [Google Scholar]
  • 85.Ford AL, Foulcher E, Lemckert FA, Sedgwick JD. Microglia induce CD4 T lymphocyte final effector function and death. J Exp Med. 1996;184:1737–45. doi: 10.1084/jem.184.5.1737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Stohlman SA, Bergmann CC, Lin MT, Cua DJ, Hinton DR. CTL effector function within the central nervous system requires CD4+ T cells. J Immunol. 1998;160:2896–904. [PubMed] [Google Scholar]
  • 87.Zajac AJ, Murali-Krishna K, Blattman JN, Ahmed R. Therapeutic vaccination against chronic viral infection: the importance of cooperation between CD4+ and CD8+ T cells. Curr Opin Immunol. 1998;10:444–9. doi: 10.1016/s0952-7915(98)80119-2. [DOI] [PubMed] [Google Scholar]
  • 88.Dix AR, Brooks WH, Roszman TL, Morford LA. Immune defects observed in patients with primary malignant brain tumors. J Neuroimmunol. 1999;100:216–32. doi: 10.1016/s0165-5728(99)00203-9. [DOI] [PubMed] [Google Scholar]
  • 89.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;159:4415–25. [PubMed] [Google Scholar]
  • 90.Prins RM, Graf MR, Merchant RE. Cytotoxic T cells infiltrating a glioma express an aberrant phenotype that is associated with decreased function and apoptosis. Cancer Immunol Immunother. 2001;50:285–92. doi: 10.1007/s002620100202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Elliott LH, Brooks WH, Roszman TL. Activation of immunoregulatory lymphocytes obtained from patients with malignant gliomas. J Neurosurg. 1987;67:231–6. doi: 10.3171/jns.1987.67.2.0231. [DOI] [PubMed] [Google Scholar]
  • 92.Sawamura Y, Hosokawa M, Kuppner MC, Kobayashi H, Aida T, Abe H, de Tribolet N. Antitumor activity and surface phenotypes of human glioma-infiltrating lymphocytes after in vitro expansion in the presence of interleukin 2. Cancer Res. 1989;49:1843–9. [PubMed] [Google Scholar]
  • 93.Miyatake S, Kikuchi H, Iwasaki K, Yamashita J, Li ZY, Namba Y, Hanaoka M. Specific cytotoxic activity of T lymphocyte clones derived from a patient with gliosarcoma. J Neurosurg. 1988;69:751–9. doi: 10.3171/jns.1988.69.5.0751. [DOI] [PubMed] [Google Scholar]
  • 94.Elliott LH, Brooks WH, Roszman T. Suppression of high affinity IL-2 receptors on mitogen activated lymphocytes by glioma-derived suppressor factor. J Neurooncol. 1992;14:1–7. doi: 10.1007/BF00170940. [DOI] [PubMed] [Google Scholar]
  • 95.Fontana A, Hengartner H, de Tribolet N, Weber E. Glioblastoma cells release interleukin 1 and factors inhibiting interleukin 2-mediated effects. J Immunol. 1984;132:1837–44. [PubMed] [Google Scholar]
  • 96.Couldwell WT, Yong VW, Dore-Duffy P, Freedman MS, Antel JP. Production of soluble autocrine inhibitory factors by human glioma cell lines. J Neurol Sci. 1992;110:178–85. doi: 10.1016/0022-510x(92)90026-h. [DOI] [PubMed] [Google Scholar]
  • 97.Bodmer S, Strommer K, Frei K, Siepl C, de Tribolet N, Heid I, Fontana A. Immunosuppression and transforming growth factor-β in glioblastoma. Preferential production of transforming growth factor-β2. J Immunol. 1989;143:3222–9. [PubMed] [Google Scholar]
  • 98.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-β. J Immunol. 1991;146:3289–97. [PubMed] [Google Scholar]
  • 99.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;148:3847–56. [PubMed] [Google Scholar]
  • 100.Gorelik L, Flavell RA. Transforming growth factor-beta in T-cell biology. Nature Rev Immunol. 2002;2:46–53. doi: 10.1038/nri704. [DOI] [PubMed] [Google Scholar]
  • 101.Jachimczak P, Bogdahn U, Schneider J, et al. The effect of transforming growth factor-β2-specific phosphorothioate-anti-sense oligodeoxynucleotides in reversing cellular immunosuppression in malignant glioma. J Neurosurg. 1993;78:944–51. doi: 10.3171/jns.1993.78.6.0944. [DOI] [PubMed] [Google Scholar]
  • 102.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 β antisense gene therapy. Proc Natl Acad Sci USA. 1996;93:2909–14. doi: 10.1073/pnas.93.7.2909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.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;58:302–9. [PubMed] [Google Scholar]
  • 104.Stander M, Naumann U, Dumitrescu L, Heneka M, Loschmann P, Gulbins E, Dichgans J, Weller M. Decorin gene transfer-mediated suppression of TGF-beta synthesis abrogates experimental malignant glioma growth in vivo. Gene Ther. 1998;5:1187–94. doi: 10.1038/sj.gt.3300709. [DOI] [PubMed] [Google Scholar]
  • 105.Münz C, Naumann U, Grimmel C, Rammensee HG, Weller M. TGF-β-independent induction of immunogenicity by decorin gene transfer in human malignant glioma cells. Eur J Immunol. 1999;29:1032–40. doi: 10.1002/(SICI)1521-4141(199903)29:03<1032::AID-IMMU1032>3.0.CO;2-W. [DOI] [PubMed] [Google Scholar]
  • 106.Fontana A, Kristensen F, Dubs R, Gemsa D, Weber E. Production of prostaglandin E and an interleukin-1 like factor by cultured astrocytes and C6 glioma cells. J Immunol. 1982;129:2413–19. [PubMed] [Google Scholar]
  • 107.Sawamura Y, Diserens AC, de Tribolet N. In vitro prostaglandin E2 production by glioblastoma cells and its effect on interleukin-2 activation of oncolytic lymphocytes. J Neurooncol. 1990;9:125–30. doi: 10.1007/BF02427832. [DOI] [PubMed] [Google Scholar]
  • 108.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]
  • 109.Nitta T, Hishii M, Sato K, Okumura K. Selective expression of interleukin-10 gene within glioblastoma multiforme. Brain Res. 1994;649:122–8. doi: 10.1016/0006-8993(94)91055-3. [DOI] [PubMed] [Google Scholar]
  • 110.Merlo A, Juretic A, Zuber M, et al. Cytokine gene expression in primary brain tumours, metastases and meningiomas suggests specific transcription patterns. Eur J Cancer. 1993;29A:2118–25. doi: 10.1016/0959-8049(93)90046-i. [DOI] [PubMed] [Google Scholar]
  • 111.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;37:1160–7. doi: 10.1227/00006123-199512000-00016. [DOI] [PubMed] [Google Scholar]
  • 112.Berman RM, Suzuki T, Tahara H, Robbins PD, Narula SK, Lotze MT. Systemic administration of cellular IL-10 induces an effective, specific, and long-lived immune response against established tumors in mice. J Immunol. 1996;157:231–8. [PubMed] [Google Scholar]
  • 113.Segal BM, Glass DD, Shevach EM. Cutting edge: IL-10-producing CD4+ T cells mediate tumor rejection. J Immunol. 2002;168:1–4. doi: 10.4049/jimmunol.168.1.1. [DOI] [PubMed] [Google Scholar]
  • 114.Nagata S, Golstein P. The Fas death factor. Science. 1995;267:1449–56. doi: 10.1126/science.7533326. [DOI] [PubMed] [Google Scholar]
  • 115.Lynch DH, Ramsdell F, Alderson MR. Fas and FasL in the homeostatic regulation of immune responses. Immunol Today. 1995;16:569–74. doi: 10.1016/0167-5699(95)80079-4. [DOI] [PubMed] [Google Scholar]
  • 116.Saas P, Walker PR, Hahne M, et al. Fas Ligand expression by astrocytoma in vivo: maintaining immune privilege in the brain. J Clin Invest. 1997;99:1173–8. doi: 10.1172/JCI119273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Walker PR, Saas P, Dietrich P-Y. The role of Fas ligand (CD95L) in immune escape: the tumor cell strikes back. J Immunol. 1997;158:4521–4. [PubMed] [Google Scholar]
  • 118.Restifo NP. Not so fas: Re-evaluating the mechanisms of immune privilege and tumor escape. Nat Med. 2000;6:493–5. doi: 10.1038/74955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Gratas C, Tohma Y, Van Meir EG, et al. Fas ligand expression in glioblastoma cell lines and primary astrocytic brain tumors. Brain Pathol. 1997;7:863–9. doi: 10.1111/j.1750-3639.1997.tb00889.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Favre N, Bonnotte B, Droin N, Fromentin A, Solary E, Martin F. Fas (CD95) ligand expression by tumor cell variants can be unrelated to their capacity to induce tolerance or immune rejection. Int J Cancer. 1999;82:359–67. doi: 10.1002/(sici)1097-0215(19990730)82:3<359::aid-ijc9>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
  • 121.Frankel B, Longo SL, Ryken TC. Human astrocytomas co-expressing Fas and Fas ligand also produce TGFbeta2 and Bcl-2. J Neurooncol. 1999;44:205–12. doi: 10.1023/a:1006311231189. [DOI] [PubMed] [Google Scholar]
  • 122.Didenko VV, Ngo HN, Minchew C, Baskin DS. Apoptosis of T lymphocytes invading glioblastomas multiforme: a possible tumor defense mechanism. J Neurosurg. 2002;96:580–4. doi: 10.3171/jns.2002.96.3.0580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Hahne M, Rimoldi D, Schröter M, et al. Melanoma cell expression of Fas (Apo-1/CD95) ligand: Implications for tumor immune escape. Science. 1996;274:1363–6. doi: 10.1126/science.274.5291.1363. [DOI] [PubMed] [Google Scholar]
  • 124.Arai H, Chan SY, Bishop DK, Nabel GJ. Inhibition of the alloantibody response by CD95 ligand. Nat Med. 1997;3:843–8. doi: 10.1038/nm0897-843. [DOI] [PubMed] [Google Scholar]
  • 125.Seino K-I, Kayagaki N, Okumura K, Yagita H. Antitumor effect of locally produced CD95 ligand. Nat Med. 1997;3:165–70. doi: 10.1038/nm0297-165. [DOI] [PubMed] [Google Scholar]
  • 126.Seino K-I, Kayagaki N, Tsukada N, Fukao K, Yagita H, Okumura K. Transplantation of CD95 ligand-expressing grafts. Influence of transplantation site and difficulty in protecting allo- and xenografts. Transplantation. 1997;64:1050–4. doi: 10.1097/00007890-199710150-00018. [DOI] [PubMed] [Google Scholar]
  • 127.Arai H, Gordon D, Nabel EG, Nabel GJ. Gene transfer of Fas ligand induces tumor regression in vivo. Proc Natl Acad Sci USA. 1997;94:13862–7. doi: 10.1073/pnas.94.25.13862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Chen JJ, Sun Y, Nabel GJ. Regulation of the proinflammatory effects of Fas ligand (CD95L) Science. 1998;282:1714–17. doi: 10.1126/science.282.5394.1714. [DOI] [PubMed] [Google Scholar]
  • 129.Walker PR, Saas P, Dietrich PY. Tumor expression of Fas ligand (CD95L) and the consequences. Curr Opin Immunol. 1998;10:564–72. doi: 10.1016/s0952-7915(98)80225-2. [DOI] [PubMed] [Google Scholar]
  • 130.O'Connell J. Fas ligand and the fate of antitumour cytotoxic T lymphocytes. Immunology. 2002;105:263–6. doi: 10.1046/j.1365-2567.2002.01379.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Wiendl H, Mitsdoerffer M, Hofmeister V, et al. A functional role of HLA-G expression in human gliomas: an alternative strategy of immune escape. J Immunol. 2002;168:4772–80. doi: 10.4049/jimmunol.168.9.4772. [DOI] [PubMed] [Google Scholar]
  • 132.Wischhusen J, Jung G, Radovanovic I, et al. Identification of CD70-mediated apoptosis of immune effector cells as a novel immune escape pathway of human glioblastoma. Cancer Res. 2002;62:2592–9. [PubMed] [Google Scholar]
  • 133.Held-Feindt J, Mentlein R. CD70/CD27 ligand, a member of the TNF family, is expressed in human brain tumors. Int J Cancer. 2002;98:352–6. doi: 10.1002/ijc.10207. [DOI] [PubMed] [Google Scholar]
  • 134.Braud VM, Allan DS, McMichael AJ. Functions of nonclassical MHC and non-MHC-encoded class I molecules. Curr Opin Immunol. 1999;11:100–8. doi: 10.1016/s0952-7915(99)80018-1. [DOI] [PubMed] [Google Scholar]
  • 135.Bainbridge D, Ellis S, Le Bouteiller P, Sargent I. HLA-G remains a mystery. Trends Immunol. 2001;22:548–52. doi: 10.1016/s1471-4906(01)02031-2. [DOI] [PubMed] [Google Scholar]
  • 136.Jacquot S. CD27/CD70 interactions regulate T dependent B cell differentiation. Immunol Res. 2000;21:23–30. doi: 10.1385/IR:21:1:23. [DOI] [PubMed] [Google Scholar]

Articles from Immunology are provided here courtesy of British Society for Immunology

RESOURCES