Review
Glioblastoma: Therapeutic challenges, what lies ahead

https://doi.org/10.1016/j.bbcan.2012.05.004Get rights and content

Abstract

Glioblastoma (GBM) is one of the most aggressive human cancers. Despite current advances in multimodality therapies, such as surgery, radiotherapy and chemotherapy, the outcome for patients with high grade glioma remains fatal. The knowledge of how glioma cells develop and depend on the tumor environment might open opportunities for new therapies. There is now a growing awareness that the main limitations in understanding and successfully treating GBM might be bypassed by the identification of a distinct cell type that has defining properties of somatic stem cells, as well as cancer-initiating capacity — brain tumor stem cells, which could represent a therapeutic target. In addition, experimental studies have demonstrated that the combination of antiangiogenic therapy, based on the disruption of tumor blood vessels, with conventional chemotherapy generates encouraging results. Emerging reports have also shown that microglial cells can be used as therapeutic vectors to transport genes and/or substances to the tumor site, which opens up new perspectives for the development of GBM therapies targeting microglial cells. Finally, recent studies have shown that natural toxins can be conjugated to drugs that bind to overexpressed receptors in cancer cells, generating targeted-toxins to selectively kill cancer cells. These targeted-toxins are highly effective against radiation- and chemotherapy-resistant cancer cells, making them good candidates for clinical trials in GBM patients. In this review, we discuss recent studies that reveal new possibilities of GBM treatment taking into account cancer stem cells, angiogenesis, microglial cells and drug delivery in the development of new targeted-therapies.

Introduction

Awareness of how cancer tissue emerges and evolves is essential for designing targeted anti-cancer therapies. Cancer emerges as a result of a multistep process; each step reflects a genetic alteration that drives the progressive cell transformation into cancer. According to Hanahan and Weinberg, six essential aspects in cellular physiology drive cancer: self-sufficiency in growth signals, insensitivity to growth-inhibitory signals, evasion of programmed cell death, limitless replication, sustained angiogenesis and tissue invasion [1]. Recently, two additional hallmarks for cancer were proposed: cellular metabolism changes to support neoplastic growth and the ability to evade immunological destruction. Moreover, two neoplasia characteristics facilitate the acquisition of all hallmarks: genomic instability and inflammation [1].

Genomic instability and inflammation are potentially interconnected, since an inflammatory microenvironment can increase spontaneous mutagenesis rates by several mechanisms. For example, activated macrophages produce reactive oxygen species that, besides directly damaging DNA, induce the failure of DNA mismatch repairs [2]. Furthermore, macrophage migration inhibitory factor (MIF) can suppress transcription of the tumor-suppressor gene TP53, resulting in defective DNA-damage repair and increased genomic instability [3]. Tumor cells also take part in inflammatory processes by activating intrinsic inflammatory mechanisms in the surrounding epithelial or stromal cells, which often show genetic and epigenetic oncogenic alterations similar to those in tumor cells [2].

Understanding cancer cell development and the dependence on the tumor environment is important for new therapies and prevention. A recent study has demonstrated that the cancer-preventive efficiency of long-term daily treatment with anti-inflammatory drugs was found to prevent many solid tumors, including colon, esophageal, lung, prostate and brain cancers [4].

Heterotypic cellular communication plays an important role in tumorigenesis. This communication can occur between cancer cells and stroma, such as inflammatory macrophages [5]. The presence of non-cancer cells that contribute to progressive malignancy by creating the tumor microenvironment opens an opportunity for therapeutic intervention [1]. In addition, some degree of heterotypic cellular communication is seen among neighboring tumor cells. For example, cancer promotion can occur due to interclonal cooperation of mutated tumor cells in the same tissue. Different mutations in distinct cells in the same epithelium collaborate for cancer development, demonstrating that cellular interactions may contribute to oncogenic cooperation and that tumor heterogeneity may be achieved at early stages of tumorigenesis [6].

In fact, recent evidence has pointed to the existence of a small fraction of cells responsible for metastasis and indiscriminate growth. In some tumors, such as malignant gliomas, a population of cancer cells expressing a stem cells marker called prominin 1, also known as CD133, has been detected [7], [8]. CD133, a cell membrane glycoprotein, was first identified on a hepatoma cell surface and is generally expressed in hematopoietic stem cells [9], endothelial precursor cells [10] and neural stem cells [11]. The function of this transmembrane protein has not yet been clearly defined. It is a 120 kDa protein, with an N-terminal extracellular domain, two large extracellular loops, which can be N-glycosylated, and an intracellular C-terminus [13]. It is highly expressed and strongly glycosylated in stem and progenitor cells, such as neural stem cells [11]. CD133+ tumor cells have been mentioned as presenting an advanced ability to generate tumors in xenographs [12] and, therefore, have been called cancer stem cells (CSCs). In glioblastoma (GBM), the most aggressive primary brain tumor, CD133+ CSC colonies showed immune suppressor properties. GBM stem cells have been shown to trigger T-cell apoptosis, inhibit T-cell proliferation and activation and induce regulatory T cells [14]. Moreover, multivariate survival analyses have shown that the proportion of CD133 + cells affects the clinical outcome in glioma patients independent of tumor grade [7]. In another study, the survival of GBM patients was significantly shorter with higher levels of CD133 mRNA [8].

Although GBMs can contain both CD133+ and CD133- cell types that generate highly aggressive tumors, CD133+ cells are radio- and chemoresistant. This suggests that many cancer therapies, while killing the majority of tumor cells, may finally fail because they do not eliminate CSCs, which survive and generate new tumors. Therefore, CSCs are key elements in the recurrence of GBM, making them a desirable target for therapy.

In this review, we discuss recent advances in GBM biology related to CSC, angiogenesis, microglia and drug delivery that are relevant to the development of new targeted-therapies.

Section snippets

Glioblastoma

Tumors of the central nervous system (CNS) often display a significant histopathological heterogeneity. The World Health Organization (WHO) classifies astrocytomas according to different grades of malignancy (I to IV). Basically, these categories result from the recognition of anaplasia (nuclear atypia, cell pleomorphism, mitotic activity, endothelial hyperplasia and necrosis) through histological analysis [15]. Astrocytomas may be classified as low-grade (WHO II) or high grade (anaplastic WHO

Stem cells as a target for GBM therapy

A main limitation in understanding and successfully treating GBM might be bypassed by the identification of a defined cell that could represent a therapeutic target. Only few atypical cells within the tumor bulk might be responsible for the development and recurrence of some brain tumors. Evidence indicates that the real culprit is a distinctly transformed CNS cell type that has the defining properties of somatic stem cells, as well as cancer-initiating capacity — a brain tumor stem cell (Fig. 2

Sonic hedgehog (SHH) signaling pathway and stemness signature of cancer stem cells: a target for therapy

In normal adult brain it has been shown that SHH via activation of the transcription factor glioma-associated oncongene-one (Gli1), participates in the maintenance and proliferation of neural stem cells of the subventricular zone (SVZ) [49], [50]. However, the SHH signaling pathway has also been postulated as being involved in the development of the chemoresistant and radioresistant properties of CSCs and their self-renewal capacity [50], [51]. Gli1 was initially described as an oncogene

Targeting vascular endothelial growth factor (VEGF) signaling as an antiangiogenic therapy

Antiangiogenic therapy, initially proposed by Folkman in 1971, consists in the disruption of tumor blood vessels [60]. Taking this principle into account, several drugs are now being tested in phase I–III clinical trials on GBM patients. These include humanized antibodies against angiogenic factors or their receptors, decoy receptors, specific tyrosine kinase inhibitors and signaling pathway inhibitors (for more details see also [61], [62], [134]).

Here, we will discuss the VEGF/VEGFR signaling

Microglial cells: an alternative against tumor progression?

The ability of gliomas to escape the host immune system has limited the application of current therapies. There is clear evidence that microglial function is controlled by tumor cells, resulting in the support of tumor growth. Microglia (the resident macrophages of CNS), rather than fighting against the tumor, may contribute to its progression [88], [89]. According to these studies, the local production of soluble factors, such as monocyte chemotactic protein-1 (MCP-1), colony stimulating

New strategies for drug delivery into GBM cells

As an effort to bypass the BBB, which prevents molecules with ionic charge or larger than 500 Da to successfully reach the tumor region, investigators have studied a variety of strategies to allow the infusion of agents into surgically resected tumor cavities or to transiently disrupt the BBB. Intracerebral drug delivery can be achieved by manual injection, by the use of sub-cutaneous reservoirs and pumps, or by convection-enhanced delivery (CED), which uses small intracranial catheters with a

Delivering interference RNA to GBM cells

RNA interference is a cellular process of post-transcriptional gene silencing that is RNA-dependent, sequence specific and conserved among eukaryotes [127]. This mechanism involves a specific enzymatic machinery and is guided by two types of molecules: miRNAs and siRNAs, which are responsible for endogenous and exogenous gene expression regulation, respectively [128].

Many gene silencing studies with siRNA have been reported recently in GBM cell lines [126], [129] and in xenograft models [126],

Conclusions

The discovery of the major genetic alterations in cancers has made a major contribution to the understanding of the molecular pathways involved in oncogenesis. However, despite recent advances, further studies regarding the molecular mechanisms governing malignant gliomas are required in order to develop successful therapies based on molecular targeting. On the other hand, awareness of cancer cell interaction with their milieu is essential for designing intelligent anti-cancer therapies. In

Acknowledgements

We thank Dr. Behnam Badie for the critical reading of this manuscript. This work was supported by Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ); Instituto Nacional de Neurociência Translacional (INNT)/Instituto Nacional de Ciência e Tecnologia (INCT) — Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq); Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES); Programa de Oncobiologia da Universidade Federal do Rio de Janeiro

References (132)

  • C. Calabrese et al.

    A perivascular niche for brain tumor stem cells

    Cancer Cell

    (2007)
  • D. Cui et al.

    Gli1 is a potential target for alleviating multidrug resistance of gliomas

    J. Neurol. Sci.

    (2010)
  • V. Clement et al.

    HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity

    Curr. Biol.

    (2007)
  • T.C. Carneiro-Lobo et al.

    Ixolaris, a tissue factor inhibitor, blocks primary tumor growth and angiogenesis in a glioblastoma model

    J. Thromb. Haemost.

    (2009)
  • M. Paez-Ribes et al.

    Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis

    Cancer Cell

    (2009)
  • D. Zagzag et al.

    Vascular apoptosis and involution in gliomas precede neovascularization: a novel concept for glioma growth and angiogenesis

    Lab. Invest.

    (2000)
  • S. Kenig et al.

    Glioblastoma and endothelial cells cross-talk, mediated by SDF-1, enhances tumour invasion and endothelial proliferation by increasing expression of cathepsins B, S, and MMP-9

    Cancer Lett.

    (2010)
  • T.R. Alves et al.

    Glioblastoma cells: a heterogeneous and fatal tumor interacting with the parenchyma

    Life Sci.

    (2011)
  • M. Sawada et al.

    Brain-specific gene expression by immortalized microglial cell-mediated gene transfer in the mammalian brain

    FEBS Lett.

    (1998)
  • N.R. Bhat et al.

    Adenovirus infection induces microglial activation: involvement of mitogen-activated protein kinase pathways

    Brain Res.

    (2002)
  • G. Trinchieri

    Innate inflammation and cancer: is it time for cancer prevention?

    F1000 Med. Rep.

    (2011)
  • M. Karin et al.

    NF-kappaB: linking inflammation and immunity to cancer development and progression

    Nat. Rev. Immunol.

    (2005)
  • A. Wu et al.

    Glioma cancer stem cells induce immunosuppressive macrophages/microglia

    Neuro Oncol.

    (2010)
  • M. Wu et al.

    Interaction between Ras(V12) and scribbled clones induces tumour growth and invasion

    Nature

    (2010)
  • F. Zeppernick et al.

    Stem cell marker CD133 affects clinical outcome in glioma patients

    Clin. Cancer Res.

    (2008)
  • S.Y. Ardebili et al.

    CD133/prominin1 is prognostic for GBM patient's survival, but inversely correlated with cysteine cathepsins' expression in glioblastoma derived spheroids

    Radiol. Oncol.

    (2011)
  • N. Uchida et al.

    Direct isolation of human central nervous system stem cells

    Proc. Natl. Acad. Sci. U. S. A.

    (2000)
  • S.K. Singh et al.

    Identification of human brain tumour initiating cells

    Nature

    (2004)
  • D. Corbeil et al.

    Prominin: a story of cholesterol, plasma membrane protrusions and human pathology

    Traffic

    (2001)
  • J. Wei et al.

    Glioblastoma cancer-initiating cells inhibit T-cell proliferation and effector responses by the signal transducers and activators of transcription 3 pathway

    Mol. Cancer Ther.

    (2010)
  • D.N. Louis et al.

    The 2007 WHO classification of tumours of the central nervous system

    Acta Neuropathol.

    (2007)
  • J.T. Huse et al.

    Molecular subclassification of diffuse gliomas: seeing order in the chaos

    Glia

    (2011)
  • P. Kleihues et al.

    Primary and secondary glioblastomas: from concept to clinical diagnosis

    Neuro Oncol.

    (1999)
  • R.J. Jackson et al.

    Limitations of stereotactic biopsy in the initial management of gliomas

    Neuro Oncol.

    (2001)
  • F.A. Siebzehnrubl et al.

    The origins of glioma: E Pluribus Unum?

    Glia

    (2011)
  • F.W. Crawford et al.

    Relationship of pre-surgery metabolic and physiological MR imaging parameters to survival for patients with untreated GBM

    J. Neurooncol

    (2009)
  • O.O. Kanu et al.

    Glioblastoma multiforme: a review of therapeutic targets

    Expert Opin. Ther. Targets

    (2009)
  • P. Hydbring et al.

    Tipping the balance: Cdk2 enables Myc to suppress senescence

    Cancer Res.

    (2010)
  • E.A. Maher et al.

    Malignant glioma: genetics and biology of a grave matter

    Genes Dev.

    (2001)
  • N. Golestaneh et al.

    TGF-beta, neuronal stem cells and glioblastoma

    Oncogene

    (2005)
  • D.A. Hilton et al.

    Histological indicators of prognosis in glioblastomas: retinoblastoma protein expression and oligodendroglial differentiation indicate improved survival

    Histopathology

    (2004)
  • G.P. de Faria et al.

    Differences in the expression pattern of P-glycoprotein and MRP1 in low-grade and high-grade gliomas

    Cancer Investig.

    (2008)
  • C. Patru et al.

    CD133, CD15/SSEA-1, CD34 or side populations do not resume tumor-initiating properties of long-term cultured cancer stem cells from human malignant glio-neuronal tumors

    BMC Cancer

    (2010)
  • G. Liu et al.

    Cytotoxic T cell targeting of TRP-2 sensitizes human malignant glioma to chemotherapy

    Oncogene

    (2005)
  • M.D. Prados et al.

    Phase 1 study of erlotinib HCl alone and combined with temozolomide in patients with stable or recurrent malignant glioma

    Neuro Oncol.

    (2006)
  • M.D. Prados et al.

    Phase II study of erlotinib plus temozolomide during and after radiation therapy in patients with newly diagnosed glioblastoma multiforme or gliosarcoma

    J. Clin. Oncol.

    (2009)
  • J.N. Rich et al.

    Phase II trial of gefitinib in recurrent glioblastoma

    Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology

    (2004)
  • T.L. Yuan et al.

    PI3K pathway alterations in cancer: variations on a theme

    Oncogene

    (2008)
  • M. Groszer et al.

    Negative regulation of neural stem/progenitor cell proliferation by the Pten tumor suppressor gene in vivo

    Science

    (2001)
  • S.M. Chang et al.

    Phase II study of CCI-779 in patients with recurrent glioblastoma multiforme

    Invest. New Drugs

    (2005)
  • Cited by (134)

    • Sonodynamic therapy for a child with recurrent brainstem Glioma: A case report

      2023, Interdisciplinary Neurosurgery: Advanced Techniques and Case Management
    • CAR T-cells to treat brain tumors

      2023, Brain Research Bulletin
    • Naphthoquinones and derivatives as potential anticancer agents: An updated review

      2022, Chemico-Biological Interactions
      Citation Excerpt :

      Primary brain tumors are the most frequent and deadly of all human malignancies, with glioblastoma multiform (GBM) being the most prevalent. In part due to the high prevalence of intrinsic drug resistance, glioblastoma multiform (GBM) tumors in the central nervous system have a poor response to existing chemotherapies [75]. It was shown that polyamine (PA)-1,4-NQ conjugates have anticancer effects against two GBM cell lines: U87MG and GBM95.

    View all citing articles on Scopus
    View full text