Elsevier

Cellular Signalling

Volume 57, May 2019, Pages 2-9
Cellular Signalling

Review
Understanding and exploiting cell signalling convergence nodes and pathway cross-talk in malignant brain cancer

https://doi.org/10.1016/j.cellsig.2019.01.011Get rights and content

Highlights

  • The review discusses the role and interaction of MAPK and PI3K cell signalling pathways in brain cancer cells.

  • The review presents emerging evidence of the importance of the cAMP pathway in the context of cancer cell survival.

  • The review examines the possibility of targeting cell signalling hubs, exemplified by the transcription factor CREB.

  • Safer, more effective cancer treatment can be achieved by targeting specific, pivotal nodes, across multiple pathways.

Abstract

In cancer, complex intracellular and intercellular signals constantly evolve for the advantage of the tumour cells but to the disadvantage of the whole organism. Decades of intensive research have revealed the critical roles of cellular signalling pathways in regulating complex cell behaviours which influence tumour development, growth and therapeutic response, and ultimately patient outcome. Most studies have focussed on specific pathways and the resulting tumour cell function in a rather linear fashion, partly due to the available methodologies and partly due to the traditionally reductionist approach to research. Advances in cancer research, including genomic technologies have led to a deep appreciation of the complex signals and pathway interactions operating in tumour cells. In this review we examine the role and interaction of three major cell signalling pathways, PI3K, MAPK and cAMP, in regulating tumour cell functions and discuss the prospects for exploiting this knowledge to better treat difficult to treat cancers, using glioblastoma, the most common and deadly malignant brain cancer, as the example disease.

Introduction

Glioblastoma (GBM), is an aggressive and invariably lethal form of malignant brain tumour. GBM is highly heterogeneous and invasive, which accounts for the fact that it has one of the worst survival rates across all types of cancers. The highly complex nature of GBM also impacts advances toward the development of better treatments. The best current standard treatment across the globe relies on surgery, radiotherapy and a single drug, temozolomide (TMZ) [1]. The median survival time with optimal treatment is about 14 months, while the 5-year survival rate is close to or below 5% [2]. The uniformly poor prognosis in the majority of GBM patients highlights the importance of research needed to understand the molecular pathways and mechanisms which drive GBM pathology as a means to identify better and safer treatments.

GBM can occur as a primary, de novo tumour, or as a secondary tumour developing from pre-existing lower grade glioma tumours. In primary GBM, tumours develop rapidly, within several months, without evidence of pre-existing symptoms or neoplasms. Secondary GBM arises from a lower grade tumour. For example, diffuse astrocytoma, a grade II tumour, can gradually progress into malignant anaplastic astrocytoma (grade III), and ultimately into GBM (grade IV). Progression from low grade glioma (LGG) to GBM is slow, occurring over the course of several years, which is reflected in the longer median survival time of LGG patients compared to GBM patients [3].

Section snippets

GBM subtypes

In a landmark study by The Cancer Genome Atlas (TCGA) consortium involving the consolidation of clinical data together with DNA sequencing, expression analysis via microarray and RNA sequencing of hundreds of patient tumours, four GBM molecular subtypes have been identified which, in part, explain some variations in clinical the characteristics observed and patient outcomes [4].

Drug resistance

GBM is relatively resistant to therapy, with recurrent GBM being even more resistant, even to highly aggressive treatment and is the result of a complex interplay of factors. A primary reason for poor efficacy of many anticancer drugs which are used for other cancers, is inefficient drug penetrance due to the impediment of the Blood Brain Barrier (BBB) and high tumour interstitial pressure [16,17]. This is compounded by genomic instability induced by widespread alterations in genetic material

MAPK signalling

The activity of the mitogen activated protein kinase (MAPK) pathway is frequently altered in GBM. This pathway has key roles regulating cell proliferation, cell survival and metastasis (J.-Y. [22,23]). High levels of phosphorylated (activated) MAPK has been linked to poor patient survival in GBM [24].

The MAPK pathway can be activated when growth factor ligands such as epidermal growth factor (EGF) bind to their corresponding receptors, which belong to the receptor tyrosine kinase (RTK) family (

cAMP signalling

The cyclic adenosine 3′, 5′-monophosphate (cAMP) pathway regulates multiple cellular functions. Signalling typically begins with the binding of a ligand to a G-protein-coupled receptor (GPCR) [44]. This causes G-proteins to activate the enzyme adenylyl cyclase (AC) which converts adenosine triphosphate (ATP) to the second messenger cAMP (Fig. 3). Subsequently, cAMP can activate several effectors including the cAMP dependent protein kinase A (PKA). An important regulatory mechanism of this

Targeting the PI3K and MAPK pathways

The PI3K and MAPK pathways regulate GBM development and tumour growth but the precise cellular behaviours regulated by each pathway is not fully understood. Given the limited treatment strategies currently available to patients, the development of new drugs which target the PI3K and MAPK pathways could potentially improve overall patient survival and quality of life significantly. In fact, numerous preclinical studies and clinical trials have been undertaken using inhibitors of the PI3K and

The influence of signalling pathways on the GBM microenvironment

Activation of MAPK and PI3K pathways are linked to immunosuppression in GBM. Upregulation of pERK1/2 in the MAPK pathway and inactivation of PTEN in the PI3K pathway are associated with increased expression of PD-L1 on the surface of GBM cells [81,82]. PD-L1 is an immune checkpoint which can induce T-cell apoptosis when interacting with PD-1 on the T-cell surface. GBM tumours harbouring PTEN inactivating mutations are known to upregulate PD-L1 expression in monocytes through IL-10 modulation [83

Conclusions

Backed by the evidence from a vast array of research so far, it seems possible that specific targeting of three key cellular signal transduction pathways may lead to an improved clinical management of difficult to treat cancers, such as GBM. Targeting individual pathways in GBM is unlikely to provide effective tumour control, but using a combinatorial drug approach, each targeting distinct, pivotal positions or nodes in multiple pathways may lead to safer treatment and improved patient outcome.

Acknowledgements

Our laboratories are supported by grants from the CASS Foundation Australia, The Brain Foundation Australia and the Royal Melbourne Hospital Neuroscience Foundation.

References (87)

  • Audrey Carriere et al.

    ERK1/2 phosphorylate raptor to promote ras-dependent activation of mTOR complex 1 (mTORC1)

    J. Biol. Chem.

    (2011)
  • Bruno D. Fonseca et al.

    Pharmacological and genetic evaluation of proposed roles of mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK), extracellular signal-regulated kinase (ERK), and p90(RSK) in the control of mTORC1 protein signaling by phorbol esters

    J. Biol. Chem.

    (2011)
  • R.W. Butcher et al.

    Adenosine 3′,5'-phosphate in biological materials. I. Purification and properties of cyclic 3′,5'-nucleotide phosphodiesterase and use of this enzyme to characterize adenosine 3′,5'-phosphate in human urine

    J. Biol. Chem.

    (1962)
  • Rajarshi Sengupta et al.

    Treating brain tumors with PDE4 Inhibitors

    Trends Pharmacol. Sci.

    (2011)
  • Sebastian Dworkin et al.

    CREB activity modulates neural cell proliferation, midbrain-hindbrain organization and patterning in zebrafish

    Dev. Biol.

    (2007)
  • Deepa B. Shankar et al.

    The role of CREB as a proto-oncogene in hematopoiesis and in acute myeloid leukemia

    Cancer Cell

    (2005)
  • Christophe Massard et al.

    Phase Ib dose-finding study of abiraterone acetate plus buparlisib (BKM120) or dactolisib (BEZ235) in patients with castration-resistant prostate cancer

    Eur. J. Cancer

    (2017)
  • Peter J.R. Ebert et al.

    MAP kinase inhibition promotes T cell and anti-tumor activity in combination with PD-L1 checkpoint blockade

    Immunity

    (2016)
  • Paul M. Daniel et al.

    Intratumor MAPK and PI3K signaling pathway heterogeneity in glioblastoma tissue correlates with CREB signaling and distinct target gene signatures

    Exp. Mol. Pathol.

    (2018)
  • Roger Stupp et al.

    Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma

    N. Engl. J. Med.

    (2005)
  • P.D. Delgado-López et al.

    Survival in glioblastoma: a review on the impact of treatment modalities

    Clin. Transl. Oncol.

    (2016)
  • Hiroko Ohgaki et al.

    Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas

    J. Neuropathol. Exp. Neurol.

    (2005)
  • Beth K. Neilsen et al.

    Comprehensive genetic alteration profiling in primary and recurrent glioblastoma

    J. Neuro-Oncol.

    (2018)
  • Heikki Joensuu et al.

    Amplification of genes encoding KIT, PDGFRalpha and VEGFR2 receptor tyrosine kinases is frequent in glioblastoma multiforme

    J. Pathol.

    (2005)
  • Lewis C. Hou et al.

    Recurrent glioblastoma multiforme: a review of natural history and management options

    Neurosurg. Focus.

    (2006)
  • D. Koul et al.

    Suppression of matrix metalloproteinase-2 gene expression and invasion in human glioma cells by MMAC/PTEN

    Oncogene

    (2001)
  • Xing-Chen Chen et al.

    EGF stimulates glioblastoma metastasis by induction of matrix metalloproteinase-9 in an EGFR-dependent mechanism

    Oncotarget

    (2017)
  • Leon Mao et al.

    Enhancement of invadopodia activity in glioma cells by sublethal doses of irradiation and temozolomide

    J. Neurosurg.

    (2018)
  • Daisuke Hoshino et al.

    Signaling inputs to invadopodia and podosomes

    J. Cell Sci.

    (2013)
  • Kelly Harper et al.

    The hypoxic tumor microenvironment promotes invadopodia formation and metastasis through LPA1 receptor and EGFR cooperation

    Mol. Cancer Res.

    (2018)
  • Yasuharu Akasaki et al.

    Induction of a CD4+ T regulatory type 1 response by cyclooxygenase-2-overexpressing glioma

    J. Immunol.

    (2004)
  • Y. Boucher et al.

    Interstitial fluid pressure in intracranial tumours in patients and in rodents

    Br. J. Cancer

    (1997)
  • D.R. Groothuis et al.

    Regional Blood Flow and Blood-to-Tissue Transport in Five Brain Tumor Models

    (1984)
  • Mona Meyer et al.

    Single cell-derived clonal analysis of human glioblastoma links functional and genomic heterogeneity

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

    (2015)
  • Daniel V. Brown et al.

    Expression of CD133 and CD44 in glioblastoma stem cells correlates with cell proliferation, phenotype stability and intra-tumor heterogeneity

    PLoS One

    (2017)
  • K. Onda et al.

    Cerebral glioblastoma with cerebrospinal fluid dissemination: a clinicopathological study of 14 cases examined by complete autopsy

    Neurosurgery

    (1989)
  • Sheila K. Singh et al.

    Identification of a cancer stem cell in human brain tumors

    Cancer Res.

    (2003)
  • Jee-Youn Kim et al.

    The critical role of ERK in death resistance and invasiveness of hypoxia-selected glioblastoma cells

    BMC Cancer

    (2009)
  • Christian Mawrin et al.

    Prognostic relevance of MAPK expression in glioblastoma multiforme

    Int. J. Oncol.

    (2003)
  • Y. Hu et al.

    Sos1 rapidly associates with Grb2 and is hypophosphorylated when complexed with the EGF receptor after EGF stimulation

    Oncogene

    (1996)
  • N. al-Alawi et al.

    Differential regulation of cellular activities by GTPase-activating protein and NF1

    Mol. Cell. Biol.

    (1993)
  • Yutaka Hayashi et al.

    Association of EGFR gene amplification and CDKN2 (p16/MTS1) gene deletion in glioblastoma multiforme

    Brain Pathol.

    (1997)
  • Cancer Genome Atlas Research Network

    Comprehensive genomic characterization defines human glioblastoma genes and core pathways

    Nature

    (2008)
  • Cited by (0)

    1

    Equal contribution.

    View full text