Elsevier

Journal of Neuroimmunology

Volume 297, 15 August 2016, Pages 132-140
Journal of Neuroimmunology

Review Article
Exploring the role of inflammation in the malignant transformation of low-grade gliomas

https://doi.org/10.1016/j.jneuroim.2016.05.019Get rights and content

Highlights

  • A mechanism by which low grade gliomas malignantly transform is proposed.

  • The mechanism relies on the aberrant signaling that results from inflammation.

  • Mutations in LGGs (e.g. IDH1) may initiate this inflammatory cascade.

  • Inflammatory cytokines polarize GIMs to the pro-tumorigenic M2 phenotype.

Abstract

Studies of inflammatory mediators have established the tumor micro-environment as a driver of oncogenesis. This inflammatory milieu often precedes cancer, however recent data also point to the ability of oncogenic changes to induce inflammatory responses that are later harnessed by the tumor to survive and proliferate. In this review, we propose that the IDH1 mutation, present in the majority of low-grade gliomas (LGGs), initiates an inflammatory cascade that is ultimately hijacked by the tumor. Glioma infiltrating macrophages and microglia (GIMs) are polarized to the M2 phenotype, subverting the host's adaptive immune response, and fostering a tumor milieu ripe for angiogenesis, migration, and metastasis. As data continue to expand the role of inflammation in low-grade gliomas, new molecular pathways may emerge as therapeutic targets that offer a window of opportunity to intervene before the malignant transformation (MT) of LGGs occurs.

Introduction

In 1863, Rudolf Virchow postulated that cancer originated at sites of chronic inflammation following his observation of leukocyte infiltration within tumors (Balkwill and Mantovani, 2001). Over the next century, these early observations were considered less important and perhaps an artifact of the host's immune response to the tumor. Recent literature (Grivennikov et al., 2010, Mantovani et al., 2008, Sowers et al., 2014, Rakoff-Nahoum, 2006) from the past decade however, points to a more complex relationship between inflammation and tumor growth. Data now support mechanisms through which an inflammatory microenvironment may drive tumorigenesis (Mantovani et al., 2008, Karin, 2006).

Models of inflammation-induced cancers of the body begin with a normal cell acquiring a mutation that confers survival advantages relative to its neighbors. The development of most cancers necessitates 4–5 mutations (Fearon and Vogelstein, 1990, Hanahan and Weinberg, 2000) in sufficiently long-lived cells that will not be eliminated before the next mutational strike or in stem cells that can transmit these mutations to subsequent generations. Reactive oxygen species (ROS) and reactive nitrogen intermediates, secreted by activated inflammatory cells or cytokines in the microenvironment, may act as the source of genomic instability/DNA damage.

Similar mechanisms have been reported in the context of gliomas, whose association with an immunosuppressive milieu is well-documented. In glioblastoma patients for example, the presence of FoxP3 + T regulatory cells – which inhibit an antitumor immune response – has been associated with a more aggressive clinical course (Sayour et al., 2015). Programmed death-ligand (PD-L1) whose function is also immunoinhibitory, dominates in the tumor milieu and is known to promote glioma infiltration (Vlahovic et al., 2015).

Particularly in the case of high-grade gliomas (i.e. brain lesions with pathologic evidence of malignancy that are mitotically active, highly invasive and proliferative, and/or contain areas of internal necrosis (Louis et al., 2007), the brain parenchyma is host to a wide variety of immune infiltrates such as macrophages, microglia, neutrophils, and eosinophils. Upon stimulation by various glioma-secreted signaling factors, glioma immune infiltrates initiate ROS and cytokine-mediated cascades of inflammation that contribute to additional DNA damage, angiogenesis, metastasis, and tumor growth/proliferation (Karin, 2006, Sowers et al., 2014). This dynamic tumor niche leads to an immunosuppressive glioma microenvironment.

As immunotherapy gains traction as a brain cancer therapeutic (Vlahovic et al., 2015), it is especially important to consider the role of inflammation in gliomagenesis. Very complex and seemingly competing mechanisms of inflammation may both suppress and stimulate a glioma's inflammatory microenvironment, the balance of which drives malignancy. Despite inflammation's dichotomous effects on gliomagenesis, the summation of evidence suggests that the net effect of an inflammatory microenvironment is immunosuppressive, a characteristic which has hampered the success of various glioma immunotherapies (Charles et al., 2011, Ha et al., 2014).

While the role of inflammation in driving the malignant progression of pre-neoplastic lesions of the body is well documented, particularly in the case of cervical carcinoma, hepatocellular carcinoma, gastric cancer, and colon cancer (Coussens and Werb, 2002, Grivennikov et al., 2010, Rakoff-Nahoum, 2006, Sowers et al., 2014, Vakkila and Lotze, 2004), a similar role has not been described at length in the context of the malignant transformation of low-grade gliomas. Notwithstanding, gliomas have been associated with an immunosuppressive niche – one that allows these immunogenic tumors to evade the host's innate immune response, and provides context for this perspective review. Additionally, emerging evidence suggests the importance of immune infiltrates in low-grade gliomas (LGGs).

In low-grade glioma patients, overall survival (OS) is associated with a number of prognostic factors, one of them being astrocytoma histology compared with oligodendroglial histology (Schomas et al., 2009). This is due in part to the increased expression of inflammation-related genes in astrocytomas compared to oligodendrogliomas (Gonda et al., 2014). In fact, specific inflammatory chemokines such as CXCL12 (Salmaggi et al., 2005) have already been associated with decreased time to tumor progression in LGG patients. These recent findings point to a potential role for immune infiltrates in the malignant transformation of low-grade gliomas.

Utilizing a well-established paradigm seen in multiple other human malignancies in which inflammatory conditions precede malignant degeneration, we hypothesize that the malignant transformation of low grade gliomas occurs within the context of a normal host defense triggering inflammation in response to genomic instability and early mutational events (Mantovani et al., 2008, Grivennikov et al., 2010, Karin, 2006).

Section snippets

Step 1: Initiation

DNA damage of varying origins can stimulate an inflammatory response thereby promoting tumorigenesis via active signaling pathways that upregulate the production of additional pro-inflammatory mediators (Mantovani et al., 2008). One such example in the context of low-grade gliomas is the genetic mutation encoding the enzyme isocitrate dehydrogenase 1 (IDH1). IDH1 is of particular interest due to its fairly well-established role in the earliest stages of gliomagenesis, even before 1p/19q

Conclusion

The extant literature suggests that inflammation may be a contributing factor to glioma progression (Sayour et al., 2015, Locatelli et al., 2013). Inflammatory cells invade the glioma niche in what begins as the host's normal immune response. However, in the tumor microenvironment, the original M1 effectors are polarized to the M2 phenotype. The orchestrators of these changes include various glioma-secreted signaling factors such as cytokines, chemokines, and growth factors. What we have

Acknowledgements

The authors gratefully acknowledge Tiffany Le for the illustration of the figures.

The author Nicole Michelson received a Johns Hopkins University Second Decade Society grant.

The author Jordina Rincon-Torroella is a grant holder for “Fundacio La Caixa” Fellowship.

References (93)

  • A. Mantovani et al.

    The origin and function of tumor-associated macrophages

    Immunol. Today

    (1992)
  • A. Mantovani et al.

    Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes

    Trends Immunol.

    (2002)
  • A. Mantovani et al.

    The chemokine system in diverse forms of macrophage activation and polarization

    Trends Immunol.

    (2004)
  • C.N. Parkhurst et al.

    Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor

    Cell

    (2013)
  • K. Tanabe et al.

    cAMP/PKA enhances interleukin-1beta-induced interleukin-6 synthesis through STAT3 in glial cells

    Cell. Signal.

    (2016)
  • D. Voehringer et al.

    Type 2 immunity reflects orchestrated recruitment of cells committed to IL-4 production

    Immunity

    (2004)
  • T. Watanabe et al.

    IDH1 mutations are early events in the development of astrocytomas and oligodendrogliomas

    Am. J. Pathol.

    (2009)
  • S. Yona et al.

    Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis

    Immunity

    (2013)
  • M. Abou-Ghazal et al.

    The incidence, correlation with tumor-infiltrating inflammation, and prognosis of phosphorylated STAT3 expression in human gliomas

    Clin. Cancer Res.

    (2008)
  • M. Aghili et al.

    Hydroxyglutaric aciduria and malignant brain tumor: a case report and literature review

    J. Neuro-Oncol.

    (2009)
  • R.L. Alterman et al.

    Colony stimulating factor-1 expression in human glioma

    Mol. Chem. Neuropathol.

    (1994)
  • D.C. Anthony et al.

    Age-related effects of interleukin-1 beta on polymorphonuclear neutrophil-dependent increases in blood-brain barrier permeability in rats

    Brain

    (1997)
  • J.P. Bach et al.

    The role of macrophage inhibitory factor in tumorigenesis and central nervous system tumors

    Cancer

    (2009)
  • T. Blank et al.

    Microglia as modulators of cognition and neuropsychiatric disorders

    Glia

    (2013)
  • N.A. Brabers et al.

    Role of the pro-inflammatory cytokines TNF-alpha and IL-1beta in HIV-associated dementia

    Eur. J. Clin. Investig.

    (2006)
  • J.G. Cairncross et al.

    Benefit from procarbazine, lomustine, and vincristine in oligodendroglial tumors is associated with mutation of IDH

    J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol.

    (2014)
  • N.A. Charles et al.

    The brain tumor microenvironment

    Glia

    (2011)
  • A. Cohen et al.

    IDH1 and IDH2 mutations in gliomas

    Curr. Neurol. Neurosci. Rep.

    (2013)
  • S.J. Coniglio et al.

    Microglial stimulation of glioblastoma invasion involves epidermal growth factor receptor (EGFR) and colony stimulating factor 1 receptor (CSF-1R) signaling

    Mol. Med.

    (2012)
  • L.M. Coussens et al.

    Inflammation and cancer

    Nature

    (2002)
  • A. D'Andrea et al.

    Interleukin 10 (IL-10) inhibits human lymphocyte interferon gamma-production by suppressing natural killer cell stimulatory factor/IL-12 synthesis in accessory cells

    J. Exp. Med.

    (1993)
  • L. Dang et al.

    Cancer-associated IDH1 mutations produce 2-hydroxyglutarate

    Nature

    (2010)
  • R. de Waal Malefyt et al.

    Interleukin 10(IL-10) inhibits cytokine synthesis by human monocytes: an autoregulatory role of IL-10 produced by monocytes

    J. Exp. Med.

    (1991)
  • P. Ding et al.

    Expression of tumor-associated macrophage in progression of human glioma

    Cell Biochem. Biophys.

    (2014)
  • J.E. Eckel-Passow et al.

    Glioma groups based on 1p/19q, IDH, and TERT promoter mutations in tumors

    N. Engl. J. Med.

    (2015)
  • B. Fogal et al.

    Interleukin-1beta: a bridge between inflammation and excitotoxicity?

    J. Neurochem.

    (2008)
  • K. Frei et al.

    Granulocyte-macrophage colony-stimulating factor (GM-CSF) production by glioblastoma cells. Despite the presence of inducing signals GM-CSF is not expressed in vivo

    J. Immunol.

    (1992)
  • M. Frontczak-Baniewicz et al.

    Long-term consequences of surgical brain injury - characteristics of the neurovascular unit and formation and demise of the glial scar in a rat model

    Folia Neuropathol.

    (2011)
  • K. Gabrusiewicz et al.

    Characteristics of the alternative phenotype of microglia/macrophages and its modulation in experimental gliomas

    PLoS ONE

    (2011)
  • F. Ginhoux et al.

    Fate mapping analysis reveals that adult microglia derive from primitive macrophages

    Science (New York, N.Y.)

    (2010)
  • D.D. Gonda et al.

    The cancer genome atlas expression profiles of low-grade gliomas

    Neurosurg. Focus.

    (2014)
  • E.T. Ha et al.

    Chronic inflammation drives glioma growth: cellular and molecular factors responsible for an immunosuppressive microenvironment

    Neuroimmunol. Neuroinflammation

    (2014)
  • L. Hamard et al.

    The brain tissue response to surgical injury and its possible contribution to glioma recurrence

    J. Neuro-Oncol.

    (2016)
  • H. Huang et al.

    Gene expression profiling of low-grade diffuse astrocytomas by cDNA arrays

    Cancer Res.

    (2000)
  • C. Huettner et al.

    Messenger RNA expression of the immunosuppressive cytokine IL-10 in human gliomas

    Am. J. Pathol.

    (1995)
  • C. Huettner et al.

    Interleukin 10 is expressed in human gliomas in vivo and increases glioma cell proliferation and motility in vitro

    Anticancer Res.

    (1997)
  • Cited by (51)

    • Toll-Like Receptors

      2022, Encyclopedia of Infection and Immunity
    • Role and value of inflammatory markers in brain tumors: A case controlled study

      2021, Annals of Medicine and Surgery
      Citation Excerpt :

      SIRS is defined as the systemic inflammatory syndrome due to an inflammatory response. SIRS can be the result of trauma, hemorrhagic shock, or caused by other ischemia, pancreatitis or immunological injury [7,8]. Excessive production of IL1, JL6, and TNFα in inflammation process hence will produce excessive cytokines.

    • MiR-18a-downregulated RORA inhibits the proliferation and tumorigenesis of glioma using the TNF-α-mediated NF-κB signaling pathway

      2020, EBioMedicine
      Citation Excerpt :

      Inflammation plays a crucial role in growth of glioma tumors. Recent studies reported that inflammation plays an important role in the occurrence, development, and prognosis of glioma [41–44]. TNF-α is a major regulator of inflammation, and is overexpressed and secreted in the tumor microenvironment [45].

    View all citing articles on Scopus
    View full text