Cancer Letters

Cancer Letters

Volume 316, Issue 2, 28 March 2012, Pages 139-150
Cancer Letters

Mini-review
Understanding the role of cytokines in Glioblastoma Multiforme pathogenesis

https://doi.org/10.1016/j.canlet.2011.11.001Get rights and content

Abstract

Cytokines play a significant role in cancer diagnosis, prognosis and therapy. The immune system’s failure to recognize the malignant tumor cells and mount an effective response may be the result of tumor-associated cytokine deregulation. Glioblastoma Multiforme (GBM) has a characteristic cytokine expression pattern, and abnormalities in cytokine expression have been implicated in gliomagenesis. Within the heterogeneous GBM microenvironment, the tumor cells, normal brain cells, immune cells, and stem cells interact with each other through the complex cytokine network. This review summarizes the current understanding of the functions of key cytokines on GBM, and highlights potential therapeutic applications targeting these cytokines.

Introduction

Gliomas, including astrocytomas, oligodendrogliomas and ependymomas, are the most common type of primary brain tumor of the central nervous system in adults [1], [2], [3]. Gliomas originate from glia cells (astrocytes, oligodendrocytes, ependymal cells) or cancer stem cells, and are classified by the World Health Organization (WHO) into four grades based on malignancy (I, II, III, and IV). Patients inflicted with grade IV Glioblastoma Multiforme (GBM) have a median survival of about 15 months even after aggressive surgery, chemotherapy and radiotherapy [4], [5]. GBM is characterized by diffuse invasiveness, immunosuppression, aggressive proliferation, vascularization, and resistance to conventional radiotherapy and chemotherapy. The current standard of care for GBM involves initial surgical resection followed by 57–60 Gy of radiotherapy. Patients with newly diagnosed high grade glioma are treated concurrently with temozolomide (TMZ) and radiotherapy. Despite these efforts, recurrence is common and the prognosis remains poor [6].

Cytokines are a group of small molecules with diverse effects depending on the microenvironment. Cytokines are composed of glycoproteins and polypeptides that exert pro-inflammatory, anti-inflammatory or immunosuppressive effects. In addition to the tumor cells, immune cells, extracellular matrix, blood vessels, and cytokines constitute key parts of the GBM tumor microenvironment which can be favorable for tumor growth. In addition, these factors may play critical roles in contributing to the complexity and lethality of GBM. Part of the lethality stems from its proficiency in evading immune surveillance by creating an immunosuppressive tumor microenvironment. Glioma patients are commonly unable to produce a reaction against bacterial antigens, often possess decreased levels of circulating T cells, have lower in vitro proliferation of T-cells in response to mitogens, exhibit depressed antibody response and decreased antibody and T cell cytotoxicity [3], [7]. Following tumor resection, some of these peripheral dysfunctions can be partially reversed, implicating the tumor involvement in global immunosuppression [3].

There is an urgent need for a consensus view of glioma tumor microenvironments, which will ultimately assist in the creation of new therapies that target the microenvironment to interrupt or reverse the GBM progression. This review summarizes the current progress on the study of cytokine functions in GBM, and discusses potential therapeutic strategies targeting cytokines in GBM.

Section snippets

GBM pathogenesis involving key cytokines

During the GBM progression, certain transcription factors have cross signaling among different cytokines. Signal transducers and activators of transcription (STAT) transcription factors play a central role in neural stem cells (NSCs) and astrocytic development. STAT3 is constitutively active in GBM and its levels correlate with tumor grade. The upregulated STAT3 mediates interferon (IFN) and interleukin-6 (IL-6) families of cytokines. The main pathway is through Janus kinase (JAK) which

Tumor necrosis factor-α (TNF-α)

Many cells secrete tumor necrosis factor-α (TNF-α), primarily monocytes, macrophages, activated natural killer (NK) cells and T cells [10]. T cells and endothelial cells express TNF receptors [10], [11]. In normal brain, TNF-α is responsible for dendritic cell (DC) maturation [10]. In a tumor environment, its expression correlates with GBM tumor grade. 80% of anaplastic GBM tumors express TNF-α but only 17% of astrocytoma and oligoastrocytoma express TNF-α [11]. However, TNF-α does not have the

Transforming growth factor-beta (TGF-β)

Transforming growth factor has two classes, α and β. Little is known about TGF-α activity in GBM, except for a recent phase I/II dose-escalating clinical trial assessing TP-38 therapy in which a fusion protein composed of pseudomonas exotoxin (PE) and TGF-α were tested in 20 recurrent malignant glioma patients. The median survival time was 21.6 weeks and one patient survived more than 5 years from the initial diagnosis [10].

One of the best characterized cytokines in GBM is transforming growth

Interferons (IFN)

Interferons are actively involved in innate immunity, which induces the downstream transcription of genes related to anti-viral, anti-microbial and antitumor effects. Interferon-α (IFN-α) is one of three main IFN isoforms. The other different IFN types are sorted based on the amino acid sequence and the receptors they bind to. Type I interferon include 13 isoforms of IFN-α, the one and only form of IFN-β, and IFN-ω, IFN-ε, IFN-κ, IFN-τ, and IFN-ζ. Type II interferon only has one type which is

Interleukin-2 (IL-2)

Interleukin-2 (IL-2) is an immuno-activating cytokine. While it is produced predominantly by T cells, it also promotes in vitro proliferation of T cells including T helper cells, CTL and T regulatory cells [44], [45], [46]. The T cells from glioma patients produce and secrete less IL-2, which is necessary for proper T-cell proliferation and activation. The CD4+ T cells are mostly affected by the drop in IL-2 secretion [7]. This is accompanied with a decreased IL-1β secretion by monocytes within

Interleukin-4 (IL-4)

Interleukin-4 (IL-4) is produced by T cells, mast cells and basophils [50]. It plays important roles in regulating the maturation and proliferation of B cells, mast cells and T cells. IL-4 can induce a type 2 T helper (Th2) response along with IL-5, IL-6, IL-9, IL-10, and IL-13 [7], [12]. The Th2 cells are important fighters against larger pathogens such as parasites and allergens through humoral immune response and downregulated tumor-specific immunity. It works in conjunction with IL-13 and

Interleukin-6 (IL-6)

As part of the Th2 cell response, interleukin-6 (IL-6) along with its receptor is highly expressed both in GBM tumors and GBM cell lines [12], [58]. IL-6 is secreted by neurons, microglia, astrocytes and peripheral monocytes [1], [58]. It has pivotal roles in the induction and modulation of reactive astrogliosis, pathological inflammatory responses and neuro-protection. Under normal conditions, IL-6 plays a key role in B cell maturation and the induction of acute inflammatory responses [15].

Interleukin-8 (IL-8)

Interleukin-8 (IL-8) is a highly prevalent cytokine in GBM both in vivo and in vitro [16], [43], and is a potent mediator of angiogenesis. This activity is accomplished through inhibiting endothelial cell apoptosis while inducing production of metallomatrix proteins (MMPs). Endothelial cells produce MMPs which are required for angiogenesis [67]. This cytokine is best characterized by its role in leukocyte, specifically neutrophil chemotactic properties [58] along with its involvement in

Interleukin-10 (IL-10)

Interleukin-10 (IL-10) is produced by many cells as well as glioma cancer stem cells (gCSCs). Dziurzynski et al. found that gCSC, which were infected with cytomegalovirus (CMV), produced CMV IL-10 that modified the monocyte lineage. Monocytes, which are precursors to macrophages, exhibited an immune-suppressive, tumor-supporting phenotype because of the CMV IL-10 which negatively impacts attempts at cancer treatment. CMV IL-10 is significant because it can bind to human IL-10 receptors and

Interleukin-12 (IL-12)

Interleukin-12 (IL-12) is secreted by mature DC and macrophages and it enhances the cytotoxic immune cells like CTLs, NK cells, and DC [57]. IL-12 also plays roles in B cell IgG production and activation of neutrophils and DCs [10]. Matching increased IL-12 levels are increased IFN-γ levels, which induce downstream pro-inflammatory cytokine release. IL-12 also polarizes T cells towards Th1 response through differentiation of T cells [7], [43]. Upon administration of IL-12, murine models have

Interleukin-13 (IL-13)

Interleukin 13 (IL-13) has been shown to be overexpressed in a majority of glioma cell lines and GBM tumor tissues [29]. IL-13 toxin can induce a more potent cytotoxic effect in human GBM cells than in nude mice. When IL-13 was injected into immunodeficient mice with tumor xenografts of human glioma cells, the mice showed highly cytotoxic effects without adverse toxicity. A mutated human IL-13 was created in order to increase affinity to its receptor IL-13α2. The mutant IL-13 exhibited 50 times

Granulocyte–macrophage colony-stimulating factor (GM-CSF)

GM-CSF modulates granulocytes and macrophages in their mature forms. This glycoprotein stimulates colony formation among granulocytes and macrophages and also helps with DCs antigen presentation. GM-CSF often leads to inflammation when overexpressed by T cells, fibroblasts, endothelial cells, macrophages and stroma cells. IL-1 and IL-2 can augment GM-CSF production while other pro-inflammatory cytokines like IL-1 and TNF can induce it through NF-κB transcription factor. IL-4, IL-13 and

Vascular endothelial growth factor (VEGF)

VEGF is a well characterized cytokine that causes neovascularization and acceleration of vascular permeability [87]. Its tumorigenic effects include enhanced migration and proliferation and reversing senescence of endothelial cells [88], [89]. The mRNA level of VEGF is higher in GBM especially in hypoxic areas of the tumor. NF-κB and STAT3 transcription factors both upregulate VEGF expression in malignant glioma. VEGF can also be induced by hypoxia-inducible factor-1 (HIF-1) to promote

Chemokines

Within the family of cytokines, there is a group of cytokines that have chemotaxis functions called chemokines. Chemokines function primarily by binding to G-coupled protein receptors to induce cell trafficking, activation and differentiation. CXCL12 chemokine and its respective receptor, CXCR4, play an important role in stem cell trafficking, angiogenesis and vasculature development in glioma tumors. CXCL12, also known as SDF-1, is overexpressed in necrotic and hypoxic regions of GBM. CXCL8

Cancer stem cells

Cancer stem cells (CSCs) have a high prevalence among high grade neural primary tumors such as gliomas and medulloblastomas. CSCs, also known as tumor initiating cells (TICs), have the capability to initiate tumor growth and are resistant to conventional chemo and radiation therapies. CSCs are often found in the perivascular spaces at the invasive lining of the tumor. The perivascular region is a unique environment for brain tumor cells to thrive because of the increased angiogenesis induced by

Comprehensive therapy

Within the brain tumor microenvironment, there is a large heterogeneity and so is the variety of cytokine function (Fig. 1). Developing a comprehensive standard on cytokine function to which researchers and clinicians can agree upon is crucial towards developing effective therapies for GBM patients. A larger scope of understanding cytokine function may yield multiple cytokine-targeted therapies towards GBM which could prove more fruitful than any single therapy alone.

Current GBM therapy

Perspectives

Of the clinical trials investigating possible cytotoxin therapies against GBM, the three most promising therapeutic targets are IL4-PE, TP-38 and IL13-PE. They all have shown acceptable dosage toxicities and safety in phase I and II clinical trials in addition to effective tumor responses [112]. There are some drawbacks to the immunotherapy approach to GBM. Targeting one type of receptor has limited effectiveness due to the cross-communication that cytokines and other ligands have with each

Acknowledgements

This work was supported in part by the William and Ella Owens Medical Research Foundation, Dr. Marnie Rose Foundation (M. Li), and the Vivian L. Smith Department of Neurosurgery at the University of Texas Health Science Center at Houston, Medical School.

References (112)

  • N.J. Van Wagoner et al.

    Interleukin-6 expression and regulation in astrocytes

    J. Neuroimmunol.

    (1999)
  • T. Nitta et al.

    Selective expression of interleukin-10 gene within glioblastoma multiforme

    Brain Res.

    (1994)
  • D. Guo et al.

    Vascular endothelial cell growth factor promotes tyrosine phosphorylation of mediators of signal transduction that contain SH2 domains. Association with endothelial cell proliferation

    J. Biol. Chem.

    (1995)
  • G. Sciume et al.

    Chemokines and glioma: invasion and more

    J. Neuroimmunol.

    (2010)
  • C. Liu et al.

    Chemokine receptor CXCR3 promotes growth of glioma

    Carcinogenesis

    (2011)
  • J.D. Lathia et al.

    Deadly teamwork: neural cancer stem cells and the tumor microenvironment

    Cell Stem Cell

    (2011)
  • V. Samaras et al.

    Application of the ELISPOT method for comparative analysis of interleukin (IL)-6 and IL-10 secretion in peripheral blood of patients with astroglial tumors

    Mol. Cell Biochem.

    (2007)
  • P.S. Mischel et al.

    Targeted molecular therapy of GBM

    Brain Pathol.

    (2003)
  • R.A. Fenstermaker et al.

    Immunotherapeutic strategies for malignant glioma

    Cancer Control

    (2004)
  • B.J. Theeler et al.

    High-grade gliomas

    Curr. Treat Options Neurol.

    (2011)
  • L. Arribas Alpuente et al.

    Glioblastoma: changing expectations?

    Clin. Transl. Oncol.

    (2011)
  • J.F. Curtin et al.

    Combining cytotoxic and immune-mediated gene therapy to treat brain tumors

    Curr. Top. Med. Chem.

    (2005)
  • N. de la Iglesia et al.

    STAT3 regulation of glioblastoma pathogenesis

    Curr. Mol. Med.

    (2009)
  • G.P. Atkinson et al.

    NF-kappaB and STAT3 signaling in glioma: targets for future therapies

    Expert Rev. Neurother.

    (2010)
  • M. Maruno et al.

    Distribution of endogenous tumour necrosis factor alpha in gliomas

    J. Clin. Pathol.

    (1997)
  • C. Hao et al.

    Cytokine and cytokine receptor mRNA expression in human glioblastomas: evidence of Th1, Th2 and Th3 cytokine dysregulation

    Acta Neuropathol.

    (2002)
  • W. Roth et al.

    Chemotherapy and immunotherapy of malignant glioma: molecular mechanisms and clinical perspectives

    Cell Mol. Life Sci.

    (1999)
  • G. Reynes et al.

    Circulating markers of angiogenesis, inflammation, and coagulation in patients with glioblastoma

    J. Neuro-Oncol.

    (2011)
  • K. Tanabe et al.

    Mechanisms of tumor necrosis factor-alpha-induced interleukin-6 synthesis in glioma cells

    J. Neuroinflamm.

    (2010)
  • D.J. Brat et al.

    The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis

    Neuro-Oncol.

    (2005)
  • S. Yoshida et al.

    Involvement of interleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor alpha-dependent angiogenesis

    Mol. Cell Biol.

    (1997)
  • L.F. Fajardo et al.

    Dual role of tumor necrosis factor-alpha in angiogenesis

    Am. J. Pathol.

    (1992)
  • V. Egea et al.

    TNF-α respecifies human mesenchymal stem cells to a neural fate and promotes migration toward experimental glioma

    Cell Death Different.

    (2010)
  • S.A. Rempel et al.

    Identification and localization of the cytokine SDF1 and its receptor, CXC chemokine receptor 4, to regions of necrosis and angiogenesis in human glioblastoma

    Clin. Cancer Res.

    (2000)
  • T. Kato et al.

    p55 and p75 tumor necrosis factor receptor expression on human glioblastoma cells

    Neurol. Med. Chir. (Tokyo)

    (1995)
  • M. Tada et al.

    Cellular and cytokine responses of the human central nervous system to intracranial administration of tumor necrosis factor alpha for the treatment of malignant gliomas

    Cancer Immunol. Immunother.

    (1993)
  • C. Di et al.

    Emerging therapeutic targets and agents for glioblastoma migrating cells

    Anticancer Agents Med. Chem.

    (2010)
  • P. Hau et al.

    TGF-beta2 signaling in high-grade gliomas

    Curr. Pharm. Biotechnol.

    (2011)
  • Y. Lu et al.

    TGF-beta1 promotes motility and invasiveness of glioma cells through activation of ADAM17

    Oncol. Rep.

    (2011)
  • D. Llopiz et al.

    Peptide inhibitors of transforming growth factor-beta enhance the efficacy of antitumor immunotherapy

    Int. J. Cancer

    (2009)
  • H. Okada et al.

    Immunotherapeutic approaches for glioma

    Crit. Rev. Immunol.

    (2009)
  • O. Grauer et al.

    Toll-like receptor triggered dendritic cell maturation and IL-12 secretion are necessary to overcome T-cell inhibition by glioma-associated TGF-beta2

    J. Neuro-Oncol.

    (2007)
  • P. Hulper et al.

    Tumor localization of an anti-TGF-beta antibody and its effects on gliomas

    Int. J. Oncol.

    (2011)
  • V. Fensterl et al.

    Interferons and viral infections

    Biofactors

    (2009)
  • J. Yoshida et al.

    Human gene therapy for malignant gliomas (glioblastoma multiforme and anaplastic astrocytoma) by in vivo transduction with human interferon beta gene using cationic liposomes

    Hum. Gene Ther.

    (2004)
  • A. Natsume et al.

    IFN-beta gene therapy induces systemic antitumor immunity against malignant glioma

    J. Neuro-Oncol.

    (2000)
  • H. Okada et al.

    Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma

    J. Clin. Oncol.

    (2011)
  • A. Haque et al.

    Emerging role of combination of all-trans retinoic acid and interferon-gamma as chemoimmunotherapy in the management of human glioblastoma

    Neurochem. Res.

    (2007)
  • S. Janelidze et al.

    Immunizations with IFNgamma secreting tumor cells can eliminate fully established and invasive rat gliomas

    J. Immunother

    (2009)
  • M. Farkkila et al.

    Randomised, controlled study of intratumoral recombinant gamma-interferon treatment in newly diagnosed glioblastoma

    Br. J. Cancer

    (1994)
  • Cited by (0)

    View full text