Abstract
Glioblastoma multiforme (GBM) is a lethal cancer that responds poorly to radiotherapy and chemotherapy. Glioma cancer stem cells (gCSCs) have been shown to recapitulate the characteristic features of GBM and to mediate chemotherapy and radiation resistance. Immunotherapeutic targeting of this cell population holds therapeutic promise but must be considered in the context of the immunosuppressive properties mediated by the gCSC. Recent findings have indicated that this goal will be challenging because the gCSC can suppress both the innate and adaptive immune systems by a variety of gCSC-secreted products and cell-membrane interactions. In this review article, we will attempt to reconcile the disparate research findings regarding the potential of immune targeting of the gCSC and propose several novel solutions.
Keywords: Glioma cancer stem cells, STAT3, glioblastoma multiforme, immunotherapy
Introduction
Brain tumor immunotherapy holds unrealized therapeutic promise that must overcome the challenge of tumor-mediated immunosuppression, in which the cancer stem cell (CSC) may play a key role. The CSCs are a heterogeneous group of undifferentiated tumor cells defined by their capacity for self-renewal, multipotency, high tumorigenicity at low cell numbers, and capacity to recapitulate the phenotypic and histological characteristics of the original tumor [1]. CSCs have been identified in a wide variety of hematological and solid malignancies, including lung, breast, prostate, colon, and brain cancers [2-8], and are believed to play important roles in tumor initiation, proliferation, progression, invasion, metastasis, recurrence, and resistance to therapy. The identifying features of the CSCs isolated from human gliomas (gCSCs) [9, 10] include the formation of nonadherent neurospheres in vitro, the capacity for multilineage differentiation along astrocytic, oligodendroglial, and neuronal lines [11], and the ability to induce tumors in vivo that recapitulate the features of human malignant gliomas. The expression of stem cell markers such as CD133 [12] and nestin [13], are also commonly observed, but are not definitive determinants of “stem cells”.
The current standard of care for glioblastoma multiforme (GBM) consists of surgical resection, if feasible, temozolomide administration, and irradiation [14]--modalities that are effective against proliferating and differentiated tumor cells. The gCSCs, however, perhaps as a result of their undifferentiated and quiescent state, slow cell cycling, enhanced DNA repair capability, and expression of antiapoptosis genes [15], are highly resistant to these treatment modalities [16, 17]. This failure of current treatments to eliminate the gCSC population may explain the grim near-inevitability of recurrence and progression in patients with GBM, as the surviving gCSCs are capable of reconstituting the original tumor. Thus, it would seem that successful long-term treatment of GBM will require the elimination or suppression of gCSCs and that a strategy of specifically targeting gCSCs may hold promise as a paradigm for the development of future novel therapies for GBM.
Immunological approaches against GBM have been attempted in the past, but to date, none have been curative [18-30]. Although GBMs express tumor-associated and tumor-specific antigens [31] that are recognized by the host’s immune system [32, 33], they are capable of mediating a profound degree of immunosuppression, both systemically and within the tumor microenvironment [34-36], allowing tumor cells to escape host immune surveillance, and thwarting current attempts to use immunotherapies against GBM. Thus, to successfully employ immune therapy against gCSCs, two conditions must be met. First, the host immune system must be able to recognize and distinguish gCSCs in order to mount an effector response against them. Second, the immunosuppression mediated by the GBM and/or gCSCs must be circumvented. It is therefore necessary to define the antigens preferentially expressed within gCSCs relative to normal cells [37] and to determine the immunologic properties of gCSCs. This article will describe recent data concerning the immunological properties of gCSCs and explore potential strategies for immunotherapeutic approaches targeting gCSCs and reversing GBM-mediated immunosuppression.
The glioma cancer stem cell hypothesis
When first formulated, the glioma cancer stem cell hypothesis proposed a model in which the gCSC population within the GBM was responsible for tumor perpetuation and identifiable by the expression of stem cell markers such as CD133 [8, 38]. However, further investigation demonstrated that CD133 negative GBM cells can also give rise to tumors and can possess the hallmark features of gCSCs [39]. Instead, gCSCs are defined empirically by their capacity for self-renewal, pluripotency, tumorigenicity at low cell numbers, and recapitulation of the phenotypic features of primary GBM [40]. This clarifying distinction can serve to resolve disparate research findings between groups such as ours that have characterized these cells based on this more stringent criterion and found them to be immune suppressive [41, 42] and others that have relied only on the property of in vitro neurosphere formation and demonstrated immune eradication of the gCSC. At our present state of knowledge, neither neurosphere formation [43] nor CD133 expression, nor any other in vitro property, alone or in combination, should be viewed as necessary or sufficient for classification as a gCSC.
The stability of the gCSC phenotype also remains to date undetermined. The original gCSC hypothesis viewed the gCSC as a fixed phenotype of a specific population of cells whose eradication would entail the eventual destruction of the entire tumor. More recent evidence suggests that factors such as hypoxia can induce glioma cells to de-differentiate and acquire stem-like properties [44]. This raises the possibility that “stemness” is a dynamic property that many glioma cells may potentially adopt, depending on circumstance. If this is true, targeting gCSCs in isolation should not be considered a panacea for GBM, since even after successful eradication of gCSCs, other glioma cells may acquire gCSC properties and reconstitute a population of gCSCs.
The argument that gCSCs are resistant to immunological targeting
GBM immunosuppression and gCSCs
Immunosuppression in GBM is mediated by multiple redundant and overlapping mechanisms. Immunosuppressive cytokines are secreted into the tumor microenvironment and released into the systemic circulation. Immune cells are recruited into the tumor microenvironment and induced to assume immunosuppressive phenotypes. Immune effector cells such as primed CD8+ cytotoxic T cells and activated macrophages that enter the tumor microenvironment have their effector functions inhibited or rendered anergic, or they are induced to undergo apoptosis or to assume immunosuppressive phenotypes, resulting in a failure to eradicate the tumor, as has been extensively reviewed in [45].
The gCSCs inhibit adaptive immunity
The gCSC can inhibit adaptive immunity through both direct cell contact and by the secretion of inhibitory cytokines. We recently found that gCSCs expressed major histocompatibility complex (MHC) I and the inhibitory costimulatory molecule B7-H1 but showed minimal to no expression of MHC II, CD40, CD80, or CD86 [42]. The lack of activating costimulatory molecules, such as CD40, CD80, and CD86, and other molecules involved in antigen presentation, prevents the activation of T cells capable of targeting the gCSCs. Additionally, the gCSC surface expression of the co-stimulatory inhibitory molecule B7-H1, previously shown to be expressed in gliomas [46], mediates cell-to-cell contact-dependent immunosuppression of autologous T cells, which reduces glioma cell immunogenicity by suppressing T-cell effector cytokine production and activation [42]. The gCSCs have also been shown to express only low levels of the molecules required for effective antigen presentation, including β-molecule-required proteosome subunits, immunoproteosomes, and transporter molecules [47]. These findings are not entirely surprising since glioma-elaborated transforming growth factor- β (TGF-β) and prostaglandin E2 (PGE2) have previously been shown to down regulate MHC II expression, and antigen-processing [48].
In addition to cell-contact mediated immune suppressive mechanisms, we have shown that the secretion of galectin-3 by gCSCs induces apoptosis in both naïve and activated T cells [42]. Galectin-3 has previously been shown to function as an immune suppressor by inhibiting T cells and promoting tumor growth [49]. Moreover, the galectin-3 gene is one member of the 9-gene profile that predicts survival outcome of GBM patients [50]. Both Galectin-3 and the signal transducer and activator of transcription 3 (STAT3) have previously been shown to be expressed in GBMs [51-54] and this may be secondary to the presence of gCSCs. Or alternatively, tumor-mediated immune suppression is not likely exclusive to only the gCSC population and multiple redundant immune suppressive mechanisms are likely to be shared between CSC, astrocytes, and neural progenitor cells [55]. However, some may be unique to the gCSC population such as the recruitment of immune suppressive macrophages (M2) into the tumor microenvironment which appears to be specific to the CSC and not the bulk tumor or CD133 tumor depleted bulk GBM [30].
After exposure to gCSC-conditioned medium in vitro, T cells obtained from peripheral blood of healthy donors exhibit comprehensive depression in cellular immune function, including minimal proliferation in response to mitogen or polyclonal stimulation and substantial reduction of IL-2 and IFN-γ production [42]. This is identical to the response of PBMCs obtained from GBM patients [56]. Furthermore, incubation of healthy donor PBMCs with gCSC-conditioned supernatant medium markedly expanded the CD4+FoxP3+ Treg population [42], and this was in part mediated by gCSC-elaborated TGF-β1. This expansion of the Treg population in GBM patients has also been well documented previously in both the peripheral blood [57] and with respect to infiltrating the tumor [58, 59]. Cumulatively, these data indicate that the gCSC employs a variety of cell surface and secreted products to markedly inhibit adaptive immune responses.
The gCSCs inhibit innate immunity
Microglia are CNS-resident macrophages and constitute the dominant infiltrating immune cell population in GBM. Although activated microglia are effective mediators of innate immunity, capable of phagocytosis, cytotoxicity, antigen presentation, and promotion of inflammation [60, 61], this function is impaired within the context of malignant gliomas. For example, glioma-infiltrating microglia do not express the costimulatory molecules necessary for effective antigen presentation [58] and are unable to secrete proinflammatory cytokines [62]. Instead, they can actually support GBMs by mediating angiogenesis, proliferation, and tissue invasion [63] and become similar to other tumor-associated macrophages found in many other malignancies [64]. The factors that induce this polarization in the GBM microenvironment to an immunosuppressive and tumor-supportive phenotype (M2) have not been clearly defined.
We have recently demonstrated that gCSCs play a role in this subversion of microglial activity. In addition to chemokine C-C motif ligand 2 (CCL2), the gCSCs also produce soluble colony stimulating factor 1 (sCSF-1), TGF-β1, and macrophage inhibitory cytokine 1 (MIC-1). The gCSC-elaborated CCL2 and sCSF-1 recruit circulating monocytes into the GBM. Then the sCSF-1 and TGF-β1 polarize the monocytes towards the immunosuppressive M2 macrophage phenotype with up regulation of STAT3 and down modulation of the proinflammatory STAT1 [30, 65, 66]. The gCSC-elaborated MIC-1 also markedly inhibits phagocytosis [30] and the macrophage proinflammatory cytokine, tumor necrosis factor-α [67]. The gCSC-exposed macrophages also showed increased secretion of the immune suppressive cytokines IL-10, TGF-β1, and IL-23, and increased capacity for inhibiting T-cell proliferation [30]. Thus, the gCSCs play an important role in recruitment of monocytes to the GBM, inhibition of effector function, and polarization towards immunosuppression of innate immunity.
The gCSC tumorigenicity associates with immunosuppression but not CD133 expression
In order to ascertain whether gCSC-mediated immunosuppression associated with tumorigeneic potential and CD133 surface expression, we studied three different gCSCs that had either minimal (4%; gCSC 1), moderate (14%; gCSC 2) or high expression (79%; gCSC 3) of CD133. CD133 expression levels did not correlate with the degree of gCSC-mediated T-cell immunosuppression. For example, gCSC 2, with moderate levels of CD133 expression, exerted the strongest inhibition of T-cell proliferation (Fig. 1A), induced the highest level of Foxp3+ Tregs (Fig. 1B), and demonstrated the greatest percentage of increased T-cell apoptosis (Fig. 1C), suggesting that CD133 is not a marker for the immunosuppressive properties of gCSCs. Interestingly, we did observe that there was an association of CD133 level with gCSC-produced galectin-3 (Fig. 1D). However, we have found that the degree of immunosuppression, as reflected by the degree of inhibition of T-cell proliferation (Fig. 1E), induced Tregs (Fig. 1F), or enhanced T-cell apoptosis associated directly with gCSC tumorigenicity as measured by animal survival, whereas the expression of CD133 did not correlate with tumorigencity (Fig. 1H). These findings are consistent with those of other recent reports [39, 68].
Fig. 1. The degree of gCSC mediated immune suppression associates with in vivo tumorigenicity but not with CD133 expression.
The gCSCs (2 × 106) were cultured in neural stem cell medium for 5 days, and then the supernatant medium was harvested for testing immunosuppressive properties. Additionally, the gCSCs (5 × 104) were intracranially implanted in nude mice (6 per group). A. The percentage of inhibited T-cell proliferation does not correlate with gCSC CD133 expression levels. B. The number of induced Foxp3+ Tregs does not correlate with the gCSC CD133 expression levels. C. Increased T-cell apoptosis does not correlate with gCSC CD133 expression levels. D. The amount of gCSC-produced galectin-3 associates with CD133 expression. E. The percentage of inhibited T-cell proliferation inversely associates with the mean survival time of gCSC-implanted animals. F. The number of induced Foxp3+ Tregs inversely associates with the mean survival time of gCSC-implanted mice. G. Increased T-cell apoptosis inversely associates with the mean survival time of gCSC-implanted animals. H. The gCSC CD133 expression levels do not correlate with the mean survival of gCSC-implanted animals.
Finally, our observation that gCSCs possess immunosuppressive properties brings into question what the appropriate murine models ought to be for studying human malignancies. Previous studies by Quintana [69] demonstrated an increasing frequency of tumorigenic/stem cells after their transplantation into highly-immunocompromised mice, which increased the investigators’ ability to detect tumorigenic melanoma cells by several orders of magnitude. Use of this type of a model system would negate or minimize a potentially defining characteristic of these cells, i.e., immunosuppression. We found that the degree of immunosuppression exerted by the gCSCs directly associated with their tumorigenicity and lethality in murine model systems, suggesting that immune suppression should be considered a characteristic of gCSCs.
The gCSC immunosuppression is mediated by STAT3
STAT3 is activated by phosphorylation on the tyrosine residue in the transactivation domain in response to signaling via Janus kinase 2, which is activated by a wide variety of cytokines and growth hormones, including IL-6 and epidermal growth factor (EGF). Phosphorylated STAT3 (p-STAT3) translocates to the nucleus and regulates transcription of immunosuppressive factors such as IL-10 [70, 71] (which suppresses Th1-mediated cytotoxic immune responses and is essential for Treg function [72]), vascular endothelial growth factor (which inhibits dendritic cell maturation and activation by inhibiting costimulatory molecule expression [73]), PGE2 (which induces the immunosuppressive Th17 cells [74, 75]), and TGF-β (which induces Tregs, inhibits T-cell proliferation, and down modulates the IL-2 receptor [34]). The STAT3 pathway has been shown to be a key molecular hub in tumor-mediated immunosuppression by inhibiting macrophage activation [76, 77] and polarization to the effector M1 phenotype, while promoting polarization to the immunosuppressive M2 phenotype [78]; moreover, it can reduce the cellular cytotoxicity of natural killer cells and neutrophils, as well as the expression of MHC II, CD80, CD86, and IL-12 in dendritic cells, rendering them unable to activate T cells and to generate antitumor immunity [79].
STAT3 is activated in many cancers, including GBM [80, 81], inducing both tumorigenesis [82] and immunosuppression [83]. Recently, Sherry et al. [84] demonstrated that STAT3 is up regulated in gCSCs. The growth and self-renewal of the gCSC was dependent on this pathway. In addition, we found that gCSCs secrete a variety of products that induce p-STAT3 expression in immune cells. The gCSC-mediated inhibition of T-cell proliferation and activation, T-cell apoptosis, Treg induction, inhibition of macrophage phagocytosis, and induction of macrophage IL-10 secretion, can all be blocked by the inhibitors of p-STAT3 [30, 41]. Thus, these findings indicate that the STAT3 signaling pathway is a central regulatory hub, mediating multiple mechanisms of immunosuppression, and that gCSCs promote immunosuppression by activating pSTAT-3 (Fig. 2).
Fig. 2. Theoretical schema showing relationship between glioma cancer stem cells and the immune system.
The gCSCs express p-STAT3, which in addition to inducing gCSC proliferation and self-renewal, is also capable of inducing p-STAT3 expression in a wide variety of immune cell populations via a feed forward mechanism. This up regulation of p-STAT-3 in the immune cells down modulates effector function such as T-cell activation, proliferation, and elaboration of effector cytokine production. The gCSC secretes a variety of factors: macrophage inhibitory cytokine, which inhibits macrophage phagocytosis; transforming growth factor (TGF-β1), which induces Tregs and polarizes the macrophage to an immunosuppressive M2 state; soluble colony stimulating factor (sCSF-1), which recruits circulating monocytes and also mediates differentiation and polarization of the monocyte into M2 tumor-associated macrophages; chemokine (C-C motif) ligand 2 (CCL-2), which enhances Treg tumor infiltration; and galectin-3, which triggers T-cell apoptosis. Additionally, the cell-surface expression of the costimulatory inhibitory molecule B7-H1, which mediates immune suppression by a variety of mechanisms, and the down modulation of the major histocompatibility complex (MHC) further enhance the immunosuppressive properties of the gCSCs.
The argument that gCSCs can be targeted with immunotherapy
Vaccination strategies against gCSCs
As potentially critical mediators of tumor initiation, progression, and recurrence, as well as mediators of resistance to chemotherapy, radiation therapy, and host immune responses, gCSCs are an important therapeutic target. The existence of immunosuppressive properties in gCSCs [41, 42] raises the issue of whether these cells can be targeted successfully by immunoherapy strategies such as vaccination. Several recent studies have suggested that vaccination may, indeed, be a feasible approach [85-87]. Specifically, Pellegatta et al. [86] have demonstrated that dendritic cell immunotherapy could be used to target murine GL261 glioma cells. This highly perpetuated cell line was characterized as “stem-like” based on the ability of the cells to form neurospheres and to express nestin, but the defining characteristics of what constitutes a cancer stem cell is currently being debated. The ubiquitous characterization of many of the human glioma cell lines as “cancer stem cells” needs to be regarded cautiously and with reservation because in vitro perpetuated cell lines can be highly divergent from the in vivo parental cells in both phenotype and function.
Consistent with the finding of Pellegatta, Xu et al. [87], using the 9L cell line, demonstrated that antigen-specific T-cell responses could be generated from dendritic cell vaccinations using “cancer stem cell”-associated antigens. They found that there was an increase in tumor-associated antigens expressed on the “cancer stem-like cells” and that these antigens could be used to trigger effector responses from CD8+ T cells, based on IFN-γ production, but they did not present cytotoxicity functional data. Of note, these investigators used irradiated cell lysates in the dendritic cell vaccination, which is likely to alter the immunological properties. In a very recent study by Brown et al., antigen-specific CD8 T cells were shown to be able to directly lyse “cancer stem-cell” targets [85]. This contrasts with our finding that gCSCs are capable of inducing T-cell apoptosis [42]. It is possible that the clonotypic tumor-specific T cells used by Brown et al. may be more resistant to gCSC-mediated apoptosis than the naïve T cells we examined. Alternatively, cytolytic T cells may still render lysis of the target but then rapidly undergo their own demise, similar to a bee delivering its fatal sting. Interestingly, Brown et al. found many of their “cancer stem cells” capable of expressing MHC II, whereas we found MHC II levels to be low [42], which may also account for the discrepancy. Di Tomaso et al. [47] also found that autologous cytotoxic T-cell responses against gCSCs could be generated in vitro in at least some GBM patients, but the efficiency of these responses was low and correlated with MHC II expression on the gCSCs.
Reconciliation of disparate research findings
We have observed that passage number markedly affects the immunosuppressive properties of perpetuated gCSCs. Specifically, the secretion of immunosuppressive cytokines by gCSCs and the ability of gCSC-conditioned medium to induce M2 macrophages diminish markedly over time with multiple passages, suggesting a progressive loss of immunosuppressive capability as gCSCs are maintained in tissue-culture conditions (Fig. 3). Indeed, Pellegata et al. [86] reported that neurospheres derived from the GL261 cell line had increased expression of immune stimulatory cell surface markers such a CD80 and CD86, whereas we have found the expression of these two markers in gCSCs recently isolated from human tumor specimens to be low [42]. Because gCSCs are a heterogeneous cell population, and immunosuppressive properties provide no survival benefit in a tissue-culture environment, selective pressures may favor the loss of immunosuppressive phenotypes over time. An alternate hypothesis would be that factors specific to the in vivo environment of gCSCs that are not present in vitro are necessary for maintaining immunosuppressive phenotypes over the long term. In either case, the instability of the immunosuppressive phenotype in gCSCs may mean that animal model systems using established glioma cell lines do not accurately replicate the immune interactions pertinent to tumors arising de novo and that a vaccination strategy effective against the former may not be so with the latter. Furthermore, some of these vaccination strategies have been conducted in immunocompromised murine model systems, and this would artificially influence the complex interaction between the adaptive and innate immune system and potentially confound the data.
Fig. 3. Decrease in immunosuppressive properties in gCSCs maintained in vitro over many passages.
Comparison of early and late passage gCSCs (n = 4) showing decrease with later passages in (A) the production of TGF-β1 (p = 0.10), and sCSF-1 (p = 0.05), (B) the ability to induce macrophages (MΦ) to secrete the immunosuppressive cytokines IL-10 (p = 0.05) and IL-23 (n = 2, p = 0.05), and (C) the ability to inhibit T-cell proliferation (p = 0.008). Late passages were 15-20 passages after early passages. P-values calculated using student’s t-test.
As recurrence and persistence are hallmark features of gliomas, it is clear that the intrinsic immune system of the patient, unaided, is usually unable to eradicate the gCSCs that are the progenitors giving rise to recurrence and progression. The gCSCs may be more than capable of immunosuppression sufficient to allow escape from the host’s natural immune surveillance, but this does not necessarily mean that an immune response properly primed by an external therapeutic manipulation will not be able to overcome this immunosuppression and still successfully target and kill the gCSCs. Several clinical trials of a peptide vaccine targeting the EGF receptor variant III (EGFRvIII) have suggested therapeutic efficacy in patients with malignant glioma [21, 24]. Because EGFRvIII expression may be enriched in CD133+ gCSCs [88], EGFRvIII-targeted therapies may help glioma patients to mount a more specific immune response against gCSCs.
Mechanisms for inhibiting the immunosuppressive properties of gCSCs
Ultimately, within malignancies there exists a balance between immune stimulation by tumor antigens and immune suppression mediated by tumor cells that promotes escape from immune surveillance. Tumorigenesis and recurrence occur in a setting in which the latter overrides the former. If tumor-mediated immunosuppression can be successfully inhibited, then the balance may tipped back in favor of immune stimulation, allowing the host’s immune system to more easily eradicate the tumor, either on its own or in conjunction with immunostimulatory therapies.
A simple way to reduce tumor-mediated immunosuppression is by a gross-total resection of tumor, which eliminates the bulk of the immunosuppressive tumor microenvironment in which immunosuppressive cytokines are most concentrated, as well as most of the immunosuppressive tumor-infiltrating cells, such as Tregs, and M2-polarized macrophages/microglia. Elimination of the tumor mass effect also allows for withdrawal of immunosuppressive adjuvant treatments such as steroids. An example of the benefit of surgery in the context of immunotherapy has been shown in GBM patients without a bulky and actively progressing disease who responded better to dendritic cell based vaccines [22]. It is for this reason that many recent clinical trials of immunotherapies in GBM patients are conducted on patients who first receive a complete surgical resection of their tumor.
Clearly, therapies that target and/or reverse gCSC immunosuppression are needed. The existence of multiple overlapping and redundant mechanisms of immunosuppression suggests that focusing on any single mode of immunosuppression alone may not be effective and probably accounts for many failures of targeted immunotherapy strategies in the past. More likely, an effective strategy will need to be able to reverse multiple modes of immunosuppression simultaneously. For this reason, the STAT3 pathway garners interest. As an important regulatory hub underlying a wide range of immunosuppressive pathways in both the adaptive and innate immune systems, inhibition of p-STAT3 holds the promise of being able to simultaneously target multiple modes of immunosuppression. Furthermore, because pSTAT-3 is also directly involved in tumorigenesis and proliferation, STAT3 inhibition can also target tumor growth directly. Inhibition of STAT3 has been shown to reverse glioma-associated immunosuppression and to inhibit tumor growth [89]. Specifically, WP1066, a small molecular inhibitor of STAT3, can be administered orally, has excellent central nervous system penetration, and can activate effector T cells, induce T-cell stimulating cytokines [83], and inhibit Tregs [90, 91]. Recently, we showed that STAT3 inhibition by WP1066 and siRNA in gCSCs can diminish gCSC induction of p-STAT3 in immune cells, reduce Treg induction, restore T-cell function, and induce T-cell proliferation [41]. The pSTAT3 blockade in gCSCs also reverses gCSC-mediated inhibition of macrophage phagocytosis, gCSC-mediated induction of IL-10 secretion in macrophages, and it diminishes the degree of gCSC-mediated M2 polarization by increasing the activity of the M1 phenotype regulator, STAT1 [30].
Furthermore, STAT3 inhibitors may exert direct anti-gCSC activity. Sherry et al. [84] showed that blocking STAT3 in gCSCs leads to inhibition of growth and neurosphere formation, but did not result in apoptosis, probably because gCSCs are relatively resistant to apoptosis. In addition, loss of expression of the CSC markers, nestin and olig2, was observed upon inhibition of STAT3. These investigators suggested that STAT3 inhibition suppresses the self-renewal properties of gCSCs in culture but does not fully execute the differentiation of these cells. This would indicate that STAT3 blockade also has the potential to reverse resistance to chemotherapy and radiation therapy mediated by the gCSC.
Future directions
Studies revealing the immune properties of gCSCs are limited. A greater understanding of the immune biology of gCSCs and their interactions with the host immune system will lead to the elucidation of novel targets and new treatment regimens to enhance the efficacy of the current immunotherapeutic approaches. Detailed studies are warranted to identify new tumor-associated antigens expressed by gCSCs to be targeted for immunological approaches as well as to clarify the dependence of gCSCs on specific signaling pathways in contrast to other stem cell populations. Vaccination strategy with dendritic cells loaded with gCSCs has been shown to be effective in animal tumor model systems with different cell lines, but it will require more investigations to better understand the mechanism of effect before being translated to human studies. Therapeutic approaches targeting the key factors allowing the tumor to evade the immune system have been studied in an attempt to reverse the immunosuppression and treat the glioma. Although the STAT3 inhibitors WP1066 [41], STA-21, and S31-201 have been shown to be effective in reserving immunosuppression mediated by gCSCs, other inhibitor molecules such as JSI-124 [92], NSC 74859 [93], and AZD1480 [94] can be tested for efficacy against gCSCs. In addition to the STAT3 pathway, other signaling pathways, including Wnt, hedgehog, Notch, the HOX family, Bmi-1, PTEN, telomerase, and efflux transporters, are involved in self-renewal and differentiation of gCSCs [95-97]; thus, these molecules may also be favorable targets for suppression of gCSCs by glioma immunotherapy. In addition, treatment strategies inducing the differentiation of CSCs can make them more vulnerable to these therapies [98]. Also, recent studies have demonstrated an association between human cytomegalovirus (HCMV) infection and malignant glioma [99-101] and have shown that HCMV nucleic acids and proteins expressed in malignant gliomas promote glioma cell invasiveness [102]. The role of HCMV in malignant glioma biology and its possible interaction with gCSCs should be further investigated. Targeting HCMV could be a new treatment strategy to eliminate cancer stem cells in glioma patients. Finally, since our knowledge of the immunologic properties of gCSCs is increasing, novel approaches can continue to be identified and improved when they are administered with other immunotherapeutic modalities such as chemotherapy or radiation therapy.
In our view, targeting gCSCs holds the most promise as one component of a multi-modal treatment strategy. The aim is not simply to just eradicate the gCSC population as if it were some mythic Achilles’ heel that would bring down the whole tumor with it, but also to suppress the tumor supportive functions, such as radiation resistance, chemoresistance, and immune suppression, provided by the gCSCs, and if necessary, to prevent or reverse the adoption of gCSC phenotypes by other glioma cells. This in turn would hopefully render the remaining tumor cells more vulnerable to other concurrent treatments, leading ultimately to the successful suppression of the malignancy.
Acknowledgments
We thank Lamonne Crutcher for assistance in obtaining tissue specimens and David M. Wildrick, Ph.D., Stephanie Jenkins and Audria Patrick for editorial assistance. Funding for these studies was graciously provided by The Anthony Bullock III Foundation (ABH), the Dr Marnie Rose Foundation (ABH), the Mitchell Foundation (ABH), The University of Texas M. D. Anderson Cancer Center (ABH), and the National Institutes of Health (CA120813) (ABH).
References
- 1.Jordan CT, Guzman ML, Noble M. Cancer stem cells. N Engl J Med. 2006;355:1253–1261. doi: 10.1056/NEJMra061808. [DOI] [PubMed] [Google Scholar]
- 2.Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100:3983–3988. doi: 10.1073/pnas.0530291100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3:730–737. doi: 10.1038/nm0797-730. [DOI] [PubMed] [Google Scholar]
- 4.Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 2005;65:10946–10951. doi: 10.1158/0008-5472.CAN-05-2018. [DOI] [PubMed] [Google Scholar]
- 5.Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M, Geschwind DH, Bronner-Fraser M, Kornblum HI. Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci USA. 2003;100:15178–15183. doi: 10.1073/pnas.2036535100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kim CF, Jackson EL, Woolfenden AE, Lawrence S, Babar I, Vogel S, Crowley D, Bronson RT, Jacks T. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell. 2005;121:823–835. doi: 10.1016/j.cell.2005.03.032. [DOI] [PubMed] [Google Scholar]
- 7.O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature. 2007;445:106–110. doi: 10.1038/nature05372. [DOI] [PubMed] [Google Scholar]
- 8.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;63:5821–5828. [PubMed] [Google Scholar]
- 9.Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, Fiocco R, Foroni C, Dimeco F, Vescovi A. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res. 2004;64:7011–7021. doi: 10.1158/0008-5472.CAN-04-1364. [DOI] [PubMed] [Google Scholar]
- 10.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;23:9392–9400. doi: 10.1038/sj.onc.1208311. [DOI] [PubMed] [Google Scholar]
- 11.Sanai N, Alvarez-Buylla A, Berger MS. Neural stem cells and the origin of gliomas. N Engl J Med. 2005;353:811–822. doi: 10.1056/NEJMra043666. [DOI] [PubMed] [Google Scholar]
- 12.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;5:67–79. doi: 10.1186/1476-4598-5-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dirks PB. Brain tumor stem cells: bringing order to the chaos of brain cancer. J Clin Oncol. 2008;26:2916–2924. doi: 10.1200/JCO.2008.17.6792. [DOI] [PubMed] [Google Scholar]
- 14.Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–996. doi: 10.1056/NEJMoa043330. [DOI] [PubMed] [Google Scholar]
- 15.Deonarain MP, Kousparou CA, Epenetos AA. Antibodies targeting cancer stem cells: a new paradigm in immunotherapy? MAbs. 2009;1:12–25. doi: 10.4161/mabs.1.1.7347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.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;444:756–760. doi: 10.1038/nature05236. [DOI] [PubMed] [Google Scholar]
- 17.Murat A, Migliavacca E, Gorlia T, Lambiv WL, Shay T, Hamou MF, de Tribolet N, Regli L, Wick W, Kouwenhoven MC, Hainfellner JA, Heppner FL, Dietrich PY, Zimmer Y, Cairncross JG, Janzer RC, Domany EDM, Stupp R, Hegi ME. Stem cell-related “self-renewal” signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma. J Clin Oncol. 2008;26:3015–3024. doi: 10.1200/JCO.2007.15.7164. [DOI] [PubMed] [Google Scholar]
- 18.Butowski N, Chang SM, Junck L, DeAngelis LM, Abrey L, Fink K, Cloughesy T, Lamborn KR, Salazar AM, Prados MD. A phase II clinical trial of poly-ICLC with radiation for adult patients with newly diagnosed supratentorial glioblastoma: a North American Brain Tumor Consortium (NABTC01-05) J Neurooncol. 2009;91:175–182. doi: 10.1007/s11060-008-9693-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.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;14:3098–3104. doi: 10.1158/1078-0432.CCR-07-4875. [DOI] [PubMed] [Google Scholar]
- 20.Fakhrai H, Mantil JC, Liu L, Nicholson GL, Murphy-Satter CS, Ruppert J, Shawler DL. Phase I clinical trial of a TGF-b antisense-modified tumor cell vaccine in patients with advanced glioma. Cancer Gene Ther. 2006;13:1052–1060. doi: 10.1038/sj.cgt.7700975. [DOI] [PubMed] [Google Scholar]
- 21.Heimberger AB, Hussain SF, Aldape K, Sawaya R, Archer GA, Friedman H, Reardon D, Friedman A, Bigner D, Sampson JH. Tumor-specific peptide vaccination in newly-diagnosed patients with GBM. J Clin Oncol (Meeting Abstracts) 2006;24:2529. [Google Scholar]
- 22.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;11:5515–5525. doi: 10.1158/1078-0432.CCR-05-0464. [DOI] [PubMed] [Google Scholar]
- 23.Mitchell DA, Archer G, Bigner D, Friedman AH, Friedman HS, Reardon D, Vrendenburgh J, Herndon JE, II, McLendon RE, Sampson J. RNA-loaded dendritic cells targeting cytomegalovirus in patients with malignant glioma. Neuro Oncol. 2007;9:509. [Google Scholar]
- 24.Sampson JH, Archer GE, Bigner DD, Davis T, Friedman HS, Keler T, Mitchell DA, Reardon DA, Sawaya R, Heimberger AB. Effect of EGFRvIII-targeted vaccine (CDX-110) on immune response and TTP when given with simultaneous standard and continuous temozolomide in patients with GBM. J Clin Oncol (Meeting Abstracts) 2008;26:2011. [Google Scholar]
- 25.Sampson JH, Archer GE, Mitchell DA, Heimberger AB, Herndon JE, 2nd, Lally-Goss D, McGehee-Norman S, Paolino A, Reardon DA, Friedman AH, Friedman HS, Bigner DD. An epidermal growth factor receptor variant III-targeted vaccine is safe and immunogenic in patients with glioblastoma multiforme. Mol Cancer Ther. 2009;8:2773–2779. doi: 10.1158/1535-7163.MCT-09-0124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.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]
- 27.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;68:5955–5964. doi: 10.1158/0008-5472.CAN-07-5973. [DOI] [PubMed] [Google Scholar]
- 28.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;11:5900–5911. doi: 10.1158/1078-0432.CCR-05-0559. [DOI] [PubMed] [Google Scholar]
- 29.Yu J, Liu G, Ying H, Yong W, Black K, Wheeler CJ. Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res. 2004;64:4973–4979. doi: 10.1158/0008-5472.CAN-03-3505. [DOI] [PubMed] [Google Scholar]
- 30.Wu A, Wei J, Kong LY, Wang Y, Priebe W, Sawaya R, Heimberger A. Glioma cancer stem cells induce immunosuppressive macrophages/microglia. Neuro Oncol. doi: 10.1093/neuonc/noq082. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kurpad SN, Zhao XG, Wikstrand CJ, Batra SK, McLendon RE, Bigner DD. Tumor antigens in astrocytic gliomas. Glia. 1995;15:244–256. doi: 10.1002/glia.440150306. Review. [DOI] [PubMed] [Google Scholar]
- 32.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;75:266–276. doi: 10.1002/(sici)1097-0215(19980119)75:2<266::aid-ijc16>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
- 33.Holladay FP, Choudhuri R, Heitz T, Wood GW. Generation of cytotoxic immune responses during the progression of a rat glioma. J Neurosurg. 1994;80:90–96. doi: 10.3171/jns.1994.80.1.0090. [DOI] [PubMed] [Google Scholar]
- 34.Heimberger AB, Bigner DD, Sampson JJ. Biological principles of brain tumor immunotherapy. In: Liau LM, Becker DP, Cloughesy TF, Bigner DD, editors. Brain tumor immunotherapy. Humana Press Inc; Totowa, NJ: 2000. pp. 101–130. [Google Scholar]
- 35.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;159:4415–4425. [PubMed] [Google Scholar]
- 36.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;134:1545–1550. [PubMed] [Google Scholar]
- 37.Wu CJ. StemBook. The Stem Cell Research Community, StemBook; 2008. Dec 15, Immunologic targeting of the cancer stem cell. [DOI] [PubMed] [Google Scholar]
- 38.Stiles CD, Rowitch DH. Glioma stem cells: a midterm exam. Neuron. 2008;58:832–846. doi: 10.1016/j.neuron.2008.05.031. [DOI] [PubMed] [Google Scholar]
- 39.Beier D, Hau P, Proescholdt M, Lohmeier A, Wischhusen J, Oefner PJ, Aigner L, Brawanski A, Bogdahn U, Beier CP. CD133(+) and CD133(-) glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles. Cancer Res. 2007;67:4010–4015. doi: 10.1158/0008-5472.CAN-06-4180. [DOI] [PubMed] [Google Scholar]
- 40.Lee J, Kotliarova S, Kotliarov Y, Li A, Su Q, Donin NM, Pastorino S, Purow BW, Christopher N, Zhang W, Park JK, Fine HA. Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell. 2006;9:391–403. doi: 10.1016/j.ccr.2006.03.030. [DOI] [PubMed] [Google Scholar]
- 41.Wei J, Bar J, Kong L-Y, Wang Y, Wu A, Sharma AK, Gumin J, Henry V, Colman H, Priebe W, Sawaya R, Lang FF, Heimberger AB. Glioblastoma cancer-initiating cells inhibit T cell proliferation and effector responses by the STAT3 pathway. Mol Cancer Ther. 2010;9:67–78. doi: 10.1158/1535-7163.MCT-09-0734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wei J, Barr J, Kong L-Y, Wang Y, Wu A, Sharma AK, Gumin J, Henry V, Colman H, Sawaya R, Lang FF, Heimberger AB. Glioma associated cancer-initiating cells induce immune suppression. Clin Cancer Res. 2010;16:461–473. doi: 10.1158/1078-0432.CCR-09-1983. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 43.Woolard K, Fine HA. Glioma stem cells: better flat than round. Cell Stem Cell. 2009;4:466–467. doi: 10.1016/j.stem.2009.05.013. [DOI] [PubMed] [Google Scholar]
- 44.Heddleston JM, Li Z, Lathia JD, Bao S, Hjelmeland AB, Rich JN. Hypoxia inducible factors in cancer stem cells. Br J Cancer. 2010;102:789–795. doi: 10.1038/sj.bjc.6605551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Dey M, Hussain SF, Heimberger AB. The role of glioma microenvironment in immune modulation: potential targets for intervention. Lett Drug Des Discov. 2006;3:443–451. [Google Scholar]
- 46.Parsa AT, Waldron JS, Panner 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;13:84–88. doi: 10.1038/nm1517. [DOI] [PubMed] [Google Scholar]
- 47.Di Tomaso T, Mazzoleni S, Wang E, Sovena G, Clavenna D, Franzin A, Mortini P, Ferrone S, Doglioni C, Marincola FM, Galli R, Parmiani G, Maccalli C. Immunobiological characterization of cancer stem cells isolated from glioblastoma patients. Clin Cancer Res. 2010;16:800–813. doi: 10.1158/1078-0432.CCR-09-2730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Facoetti A, Nano R, Zelini P, Morbini P, Benericetti E, Ceroni M, Campoli M, Ferrone S. Human leukocyte antigen and antigen processing machinery component defects in astrocytic tumors. Clin Cancer Res. 2005;11:8304–8311. doi: 10.1158/1078-0432.CCR-04-2588. [DOI] [PubMed] [Google Scholar]
- 49.Peng W, Wang HY, Miyahara Y, Peng G, Wang RF. Tumor-associated galectin-3 modulates the function of tumor-reactive T cells. Cancer Res. 2008;68:7228–7236. doi: 10.1158/0008-5472.CAN-08-1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Colman H, Zhang L, Sulman EP, McDonald JM, Shooshtari NL, Rivera A, Popoff S, Nutt CL, Louis DN, Cairncross JG, Gilbert MR, Phillips HS, Mehta MP, Chakravarti A, Pelloski CE, Bhat K, Feuerstein BG, Jenkins RB, Aldape K. A multigene predictor of outcome in glioblastoma. Neuro Oncol. 2010;12:49–57. doi: 10.1093/neuonc/nop007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Cattaneo E, Magrassi L, De-Fraja C, Conti L, Di Gennaro I, Butti G, Govoni S. Variations in the levels of the JAK/STAT and ShcA proteins in human brain tumors. Anticancer Res. 1998;18:2381–2387. [PubMed] [Google Scholar]
- 52.Lo HW, Cao X, Zhu H, Ali-Osman F. Constitutively activated STAT3 frequently coexpresses with epidermal growth factor receptor in high-grade gliomas and targeting STAT3 sensitizes them to Iressa and alkylators. Clin Cancer Res. 2008;14:6042–6054. doi: 10.1158/1078-0432.CCR-07-4923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Neder L, Marie SK, Carlotti CGJ, Ballai AA, Rosemberg S, Malheiros SM, Siqueira RP, Oba-Shinjo SM, Uno M, Aguiar PH, Miura F, Chammas R, Colli BO, Silva WAJ, Zago MA. Galectin-3 as an immunohistochemical tool to distinguish pilocytic astrocytomas from diffuse astrocytomas, and glioblastomas from aanaplastic oligodendrogliomas. Brain Pathol. 2004;14:399–405. doi: 10.1111/j.1750-3639.2004.tb00083.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Strik HM, Deininger MH, Frank B, Schluesener HJ, Meyermann R. Galectin-3: cellular distribution and correlation with WHO-grade in human gliomas. J Neurooncol. 2001;53:13–20. doi: 10.1023/a:1011874800612. [DOI] [PubMed] [Google Scholar]
- 55.Einstein O, Ben-Hur T. The changing face of neural stem cell therapy in neurologic diseases. Arch Neurol. 2008;65:452–456. doi: 10.1001/archneur.65.4.452. [DOI] [PubMed] [Google Scholar]
- 56.Ausiello CM, Palma C, Maleci A, Spagnoli GC, Amici C, Antonelli G, Casciani CU, Cassone A. Cell mediated cytotoxicity and cytokine production in peripheral blood mononuclear cells of glioma patients. Eur J Cancer. 1991;27:646–650. doi: 10.1016/0277-5379(91)90235-6. [DOI] [PubMed] [Google Scholar]
- 57.Fecci PE, Mitchell DA, Whitesides JF, Xie W, Friedman AH, Archer GE, Herndon JE, 2nd, 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;66:3294–3302. doi: 10.1158/0008-5472.CAN-05-3773. [DOI] [PubMed] [Google Scholar]
- 58.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;8:261–279. doi: 10.1215/15228517-2006-008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.El Andaloussi A, Lesniak MS. CD4+ CD25+ FoxP3+ T-cell infiltration and heme oxygenase-1 expression correlate with tumor grade in human gliomas. J Neurooncol. 2007;83:145–152. doi: 10.1007/s11060-006-9314-y. [DOI] [PubMed] [Google Scholar]
- 60.Kostianovsky AM, Maier LM, Anderson RC, Bruce JN, Anderson DE. Astrocytic regulation of human monocytic/microglial activation. J Immunol. 2008;181:5425–5432. doi: 10.4049/jimmunol.181.8.5425. [DOI] [PubMed] [Google Scholar]
- 61.Streit WJ, Kreutzberg GW. Response of endogenous glial cells to motor neuron degeneration induced by toxic ricin. J Comp Neurol. 1988;268:248–263. doi: 10.1002/cne.902680209. [DOI] [PubMed] [Google Scholar]
- 62.Hussain SF, Yang D, Suki D, Grimm E, Heimberger AB. Innate immune functions of microglia isolated from human glioma patients. J Transl Med. 2006;4:15. doi: 10.1186/1479-5876-4-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Sliwa M, Markovic D, Gabrusiewicz K, Synowitz M, Glass R, Zawadzka M, Wesolowska A, Kettenmann H, Kaminska B. The invasion promoting effect of microglia on glioblastoma cells is inhibited by cyclosporin A. Brain. 2007;130:476–489. doi: 10.1093/brain/awl263. [DOI] [PubMed] [Google Scholar]
- 64.Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4:71–78. doi: 10.1038/nrc1256. [DOI] [PubMed] [Google Scholar]
- 65.Elgert KD, Alleva DG, Mullins DW. Tumor-induced immune dysfunction: the macrophage connection. J Leukoc Biol. 1998;64:275–290. doi: 10.1002/jlb.64.3.275. [DOI] [PubMed] [Google Scholar]
- 66.Mantovani A, Bottazzi B, Colotta F, Sozzani S, Ruco L. The origin and function of tumor-associated macrophages. Immunol Today. 1992;13:265–270. doi: 10.1016/0167-5699(92)90008-U. [DOI] [PubMed] [Google Scholar]
- 67.Bootcov MR, Bauskin AR, Valenzuela SM, Moore AG, Bansal M, He XY, Zhang HP, Donnellan M, Mahler S, Pryor K, Walsh BJ, Nicholson RC, Fairlie WD, Por SB, Robbins JM, Breit SN. MIC-1, a novel macrophage inhibitory cytokine, is a divergent member of the TGF-b superfamily. Proc Natl Acad Sci U S A. 1997;94:11514–11519. doi: 10.1073/pnas.94.21.11514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ogden AT, Waziri AE, Lochhead RA, Fusco D, Lopez K, Ellis JA, Kang J, Assanah M, McKhann GM, Sisti MB, McCormick PC, Canoll P, Bruce JN. Identification of A2B5+CD133- tumor-initiating cells in adult human gliomas. Neurosurgery. 2008;62:505–514. doi: 10.1227/01.neu.0000316019.28421.95. discussion 514-505. [DOI] [PubMed] [Google Scholar]
- 69.Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ. Efficient tumour formation by single human melanoma cells. Nature. 2008;456:593–598. doi: 10.1038/nature07567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Steinbrink K, Wolfl M, Jonuleit H, Knop J, Enk AH. Induction of tolerance by IL-10-treated dendritic cells. J Immunol. 1997;159:4772–4780. [PubMed] [Google Scholar]
- 71.Williams L, Bradley L, Smith A, Foxwell B. Signal transducer and activator of transcription 3 is the dominant mediator of the anti-inflammatory effects of IL-10 in human macrophages. J Immunol. 2004;172:567–576. doi: 10.4049/jimmunol.172.1.567. [DOI] [PubMed] [Google Scholar]
- 72.Asseman C, Mauze S, Leach MW, Coffman RL, Powrie F. An essential role for interleukin 10 in the function of regulatory T cells that inhibit intestinal inflammation. J Exp Med. 1999;190:995–1004. doi: 10.1084/jem.190.7.995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D, Carbone DP. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 1996;2:1096–1103. doi: 10.1038/nm1096-1096. erratum appears in Nat Med 1996 Nov;2(11):1267. [DOI] [PubMed] [Google Scholar]
- 74.Boniface K, Bak-Jensen KS, Li Y, Blumenschein WM, McGeachy MJ, McClanahan TK, McKenzie BS, Kastelein RA, Cua DJ, de Waal Malefyt R. Prostaglandin E2 regulates Th17 cell differentiation and function through cyclic AMP and EP2/EP4 receptor signaling. J Exp Med. 2009;206:535–548. doi: 10.1084/jem.20082293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Rummel C, Sachot C, Poole S, Luheshi GN. Circulating interleukin-6 induces fever through a STAT3-linked activation of COX-2 in the brain. Am J Physiol Regul Integr Comp Physiol. 2006;291:R1316–1326. doi: 10.1152/ajpregu.00301.2006. [DOI] [PubMed] [Google Scholar]
- 76.Lang R, Patel D, Morris JJ, Rutschman RL, Murray PJ. Shaping gene expression in activated and resting primary macrophages by IL-10. J Immunol. 2002;169:2253–2263. doi: 10.4049/jimmunol.169.5.2253. [DOI] [PubMed] [Google Scholar]
- 77.O’Farrell AM, Liu Y, Moore KW, Mui AL. IL-10 inhibits macrophage activation and proliferation by distinct signaling mechanisms: Evidence for Stat3-dependent and independent pathways. EMBO J. 1998;17:1006–1018. doi: 10.1093/emboj/17.4.1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Mancino A, Lawrence T. Nuclear factor-kappaB and tumor-associated macrophages. Clin Cancer Res. 2010;16:784–789. doi: 10.1158/1078-0432.CCR-09-1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kortylewski M, Kujawski M, Wang T, Wei S, Zhang S, Pilon-Thomas S, Niu G, Kay H, Mule J, Kerr WG, Jove R, Pardoll D, Yu H. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat Med. 2005;11:1314–1321. doi: 10.1038/nm1325. [DOI] [PubMed] [Google Scholar]
- 80.Abou-Ghazal M, Yang DS, Qiao W, Reina-Ortiz C, Wei J, Kong LY, Fuller GN, Hiraoka N, Priebe W, Sawaya R, Heimberger AB. The incidence, correlation with tumor-infiltrating inflammation, and prognosis of phosphorylated STAT3 expression in human gliomas. Clin Cancer Res. 2008;14:8228–8235. doi: 10.1158/1078-0432.CCR-08-1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Rahaman SO, Harbor PC, Chernova O, Barnett GH, Vogelbaum MA, Haque SJ. Inhibition of constitutively active Stat3 suppresses proliferation and induces apoptosis in glioblastoma multiforme cells. Oncogene. 2002;21:8404–8413. doi: 10.1038/sj.onc.1206047. [DOI] [PubMed] [Google Scholar]
- 82.Yu H, Jove R. The STATs of cancer--new molecular targets come of age. Nat Rev Cancer. 2004;4:97–105. doi: 10.1038/nrc1275. [DOI] [PubMed] [Google Scholar]
- 83.Hussain SF, Kong L-Y, Jordan J, Conrad C, Madden T, Fokt I, Priebe W, Heimberger AB. A novel small molecule inhibitor of signal transducers and activators of transcription 3 reverses immune tolerance in malignant glioma patients. Cancer Res. 2007;67:9630–9636. doi: 10.1158/0008-5472.CAN-07-1243. [DOI] [PubMed] [Google Scholar]
- 84.Sherry MM, Reeves A, Wu JK, Cochran BH. STAT3 is required for proliferation and maintenance of multipotency in glioblastoma stem cells. Stem Cells. 2009;27:2383–2392. doi: 10.1002/stem.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Brown CE, Starr R, Martinez C, Aguilar B, D’Apuzzo M, Todorov I, Shih CC, Badie B, Hudecek M, Riddell SR, Jensen MC. Recognition and killing of brain tumor stem-like initiating cells by CD8+ cytolytic T cells. Cancer Res. 2009;69:8886–8893. doi: 10.1158/0008-5472.CAN-09-2687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.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;66:10247–10252. doi: 10.1158/0008-5472.CAN-06-2048. [DOI] [PubMed] [Google Scholar]
- 87.Xu Q, Liu G, Yuan X, Xu M, Wang H, Ji J, Konda B, Black KL, Yu JS. Antigen-specific T-cell response from dendritic cell vaccination using cancer stem-like cell-associated antigens. Stem Cells. 2009;27:1734–1740. doi: 10.1002/stem.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Li L, Dutra A, Pak E, Labrie JE, 3rd, Gerstein RM, Pandolfi PP, Recht LD, Ross AH. EGFRvIII expression and PTEN loss synergistically induce chromosomal instability and glial tumors. Neuro Oncol. 2009;11:9–21. doi: 10.1215/15228517-2008-081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Zhang L, Alizadeh D, Van Handel M, Kortylewski M, Yu H, Badie B. Stat3 inhibition activates tumor macrophages and abrogates glioma growth in mice. Glia. 2009;57:1458–1467. doi: 10.1002/glia.20863. [DOI] [PubMed] [Google Scholar]
- 90.Kong LY, Abou-Ghazal MK, Wei J, Chakraborty A, Sun W, Qiao W, Fuller GN, Fokt I, Grimm EA, Schmittling RJ, Archer GE, Jr, Sampson JH, Priebe W, Heimberger AB. A novel inhibitor of signal transducers and activators of transcription 3 activation is efficacious against established central nervous system melanoma and inhibits regulatory T cells. Clin Cancer Res. 2008;14:5759–5768. doi: 10.1158/1078-0432.CCR-08-0377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Kong L-K, Wei J, Sharma AK, Barr J, Abou-Ghazal MK, Fokt I, Weinberg J, Rao G, Grimm E, Priebe W, Heimberger AB. A novel phosphorylated STAT3 inhibitor enhances T cell cytotoxicity against melanoma through inhibition of regulatory T cells. Cancer Immunol Immunother. 2008;58:1023–1032. doi: 10.1007/s00262-008-0618-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Turkson J, Jove R. STAT proteins: novel molecular targets for cancer drug discovery. Oncogene. 2000;19:6613–6626. doi: 10.1038/sj.onc.1204086. [DOI] [PubMed] [Google Scholar]
- 93.Lin L, Amin R, Gallicano GI, Glasgow E, Jogunoori W, Jessup JM, Zasloff M, Marshall JL, Shetty K, Johnson L, Mishra L, He AR. The STAT3 inhibitor NSC 74859 is effective in hepatocellular cancers with disrupted TGF-beta signaling. Oncogene. 2009;28:961–972. doi: 10.1038/onc.2008.448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Hedvat M, Huszar D, Herrmann A, Gozgit JM, Schroeder A, Sheehy A, Buettner R, Proia D, Kowolik CM, Xin H, Armstrong B, Bebernitz G, Weng S, Wang L, Ye M, McEachern K, Chen H, Morosini D, Bell K, Alimzhanov M, Ioannidis S, McCoon P, Cao ZA, Yu H, Jove R, Zinda M. The JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors. Cancer Cell. 2009;16:487–497. doi: 10.1016/j.ccr.2009.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Huntly BJ, Gilliland DG. Leukaemia stem cells and the evolution of cancer-stem-cell research. Nat Rev Cancer. 2005;5:311–321. doi: 10.1038/nrc1592. [DOI] [PubMed] [Google Scholar]
- 96.Lobo NA, Shimono Y, Qian D, Clarke MF. The biology of cancer stem cells. Annu Rev Cell Dev Biol. 2007;23:675–699. doi: 10.1146/annurev.cellbio.22.010305.104154. [DOI] [PubMed] [Google Scholar]
- 97.Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–111. doi: 10.1038/35102167. [DOI] [PubMed] [Google Scholar]
- 98.Hwang KC, Kim JY, Chang W, Kim DS, Lim S, Kang SM, Song BW, Ha HY, Huh YJ, Choi IG, Hwang DY, Song H, Jang Y, Chung N, Kim SH, Kim DW. Chemicals that modulate stem cell differentiation. Proc Natl Acad Sci U S A. 2008;105:7467–7471. doi: 10.1073/pnas.0802825105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Cobbs C, Harkins L, Samanta M, Gillespie G, Bharara S, King P, Nabors L, Cobbs C, Britt W. Human cytomegalovirus infection and expression in human malignant glioma. Cancer Res. 2002;62:3347–3350. [PubMed] [Google Scholar]
- 100.Mitchell DA, Xie W, Schmittling R, Learn C, Friedman A, McLendon RE, Sampson JH. Sensitive detection of human cytomegalovirus in tumors and peripheral blood of patients diagnosed with glioblastoma. Neuro Oncol. 2008;10:10–18. doi: 10.1215/15228517-2007-035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Scheurer ME, Bondy ML, Aldape KD, Albrecht T, El-Zein R. Detection of human cytomegalovirus in different histological types of gliomas. Acta Neuropathol. 2008;116:79–86. doi: 10.1007/s00401-008-0359-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Cobbs CS, Soroceanu L, Denham S, Zhang W, Britt WJ, Pieper R, Kraus MH. Human cytomegalovirus induces cellular tyrosine kinase signaling and promotes glioma cell invasiveness. J Neurooncol. 2007;85:271–280. doi: 10.1007/s11060-007-9423-2. [DOI] [PubMed] [Google Scholar]