Elsevier

Seminars in Cancer Biology

Volume 82, July 2022, Pages 162-175
Seminars in Cancer Biology

Cancer cell heterogeneity & plasticity in glioblastoma and brain tumors

https://doi.org/10.1016/j.semcancer.2021.02.014Get rights and content

Abstract

Brain tumors remain one of the most difficult tumors to treat and, depending on the diagnosis, have a poor prognosis. Of brain tumors, glioblastoma (GBM) is the most common malignant glioma and has a dismal prognosis, with only about 5% of patients alive five years after diagnosis. While advances in targeted therapies and immunotherapies are rapidly improving outcomes in a variety of other cancers, the standard of care for GBM has largely remained unaltered since 2005. There are many well-studied challenges that are either unique to brain tumors (i.e., blood-brain barrier and immunosuppressive environment) or amplified within GBM (i.e., tumor heterogeneity at the cellular and molecular levels, plasticity, and cancer stem cells) that make this disease particularly difficult to treat. While we touch on all these concepts, the focus of this review is to discuss the immense inter- and intra-tumoral heterogeneity and advances in our understanding of tumor cell plasticity and epigenetics in GBM. With each improvement in technology, our understanding of the complexity of tumoral heterogeneity and plasticity improves and we gain more clarity on the causes underlying previous therapeutic failures. However, these advances are unlocking new therapeutic opportunities that scientists and physicians are currently exploiting and have the potential for new breakthroughs.

Introduction

A study published in 2017 by the National Cancer Institute found across-the-board improvements in 5-year survival for patients with cancer from the 1970s to 2013 (except for cervical and uterine cancer) [1]. However, even with this improvement in survival, patients with brain tumors still have only a 30 % survival at 5 years after diagnosis, and this number is far worse for some tumors types, such as glioblastoma (GBM) in adults and diffuse intrinsic pontine glioma (DIPG) in children.

The brain and spinal cord are the most well-protected organs in the body and are evolutionarily designed to be protected from damage as much as possible throughout one’s lifetime. In addition, each region of the brain has a specific function and can cause serious physical or mental disability when perturbed. These truths lead to various challenges that are not seen in other regions of the body when attempting to treat tumors within the central nervous system (CNS). An obvious example of this is that neurosurgeons must first maneuver through the skull and, when in the parenchyma, must resect the tumor with as narrow margins as possible to limit debilitating comorbidities.

This review will discuss the many ways by which our understanding of brain tumors has evolved and prevented scientists and clinician from developing treatments that improve long-term survival. We will focus on the high levels of heterogeneity and plasticity in GBM and other brain tumors as well as discuss new therapeutic avenues that have been exposed due to the exponential growth in our understanding of these tumors.

Section snippets

Overview of brain tumors

It is estimated that between 10–20 % of patients with cancer will be diagnosed with a brain metastasis, and autopsy studies have reported a true incidence of 30–40 % in all patients with cancer [[2], [3], [4], [5]]. A total of 67–80 % of brain metastases come from either lung, breast, or melanoma primary tumors. In all three of these tumors, the presence of brain metastases portends a poor survival. The median survival of patients with brain metastasis has been observed to be 13 months [6], and

Molecular heterogeneity of glioblastoma

It has long been observed that GBM possesses extensive inter- and intra-tumoral heterogeneity. Early histological studies of GBM focused on the extent of necrosis, nuclear size, astrocytic differentiation, cell size, number of mitotic cells, distribution of cell density and vascularization [26]. Through this approach and the sampling of various locations within the tumor, scientists and pathologists were able to observe significant histological variations even within the same tumor. This

Epigenetic heterogeneity in glioblastoma

While advances in our understanding of the genetics and expression patterns of GBM have enabled vertical advancements in the field, these advances have yet to result in changes in the clinical management of GBM, and the five-year survival remains around 5% [57]. Additionally, the GLASS consortium finding that mutations at recurrence are largely random suggests that mutational evolution is not driving therapeutic resistance and plasticity in GBM. This has led many in the field to investigate

Cancer stem cells

Any discussion of GBM heterogeneity and plasticity is not complete without a discussion of the CSC population. CSCs are a population of tumor cells that are defined by their functional ability to self-renew, initiate tumors, and undergo persistent proliferation [31,67]. There is no single marker of CSCs in GBM, however the glycoprotein CD133 was initially found to mark a CSC population and is widely used for sorting tumor cells today [68,69]. Furthermore, it was discovered that those with a

Glioblastoma interaction with non-tumor cells

One fact that has been understood for many years is that the GBM microenvironment consists of more than merely tumor cells. Numerous other cell populations within the brain and the immune system contribute to the conditions that lead to tumor growth. These populations include endothelial cells, microglia, astrocytes, neurons, and immune cells. The failure of many tumor cell targeting therapies has led scientists and physicians to investigate the potential of targeting other cells within the

Conclusion

Brain tumors are among the most heterogeneous tumors to have been characterized. From early histological studies that documented regions of necrosis to more recent single-cell RNA sequencing and methylation profiling, GBM is a tumor with resounding intra- and inter-tumoral heterogeneity. In addition, heterogeneity at the level of cell type, the sex and age of the patient, and plasticity over time all add to the complexity of the disease. Unfortunately, this lack of homogeneity or single driver

Funding source

Work in the Lathia laboratory is supported by the Cleveland Clinic, Case Comprehensive Cancer Center, the American Brain Tumor Association, National Brain Tumor Society, and NIH R01 NS109742, R01 NS117104, and P01 CA245705. Adam Lauko is supported by the Ruth L. Kirschstein National Research Service Award (NRSA) Individual Fellowship F30 CA250254. Alice is supported by a summer research scholarship provided by CWRU SOURCE with additional funding provided by the Women in Science and Engineering

Declaration of Competing Interest

Manmeet Ahluwalia declares the following: grants/research support-Astrazeneca, Abbvie, BMS, Bayer, Incyte, Pharmacyclics, Novocure, Mimivax, Merck. Receipt of honoraria or consulting fees: Elsevier, Wiley, VBI, Vaccines, Bayer, Tocagen, Novocure. Shareholder: Doctible, Mimivax, Cytodyn.

Acknowledgements

We regret that we were unable to include all work on glioblastoma, cancer stem cells, cellular heterogeneity, and plasticity in this review due to spatial limitations. We would like to thank the members of the Lathia laboratory for insightful discussions. We thank Ms. Amanda Mendelsohn for illustration assistance and Dr. Erin Mulkearns-Hubert for editorial assistance. Work in the Lathia laboratory is supported by the Cleveland Clinic, Case Comprehensive Cancer Center, the American Brain Tumor

References (186)

  • H. Noushmehr et al.

    Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma

    Cancer Cell

    (2010)
  • R. Chen et al.

    A hierarchy of self-renewing tumor-initiating cell types in glioblastoma

    Cancer Cell

    (2010)
  • M.L. Suvà et al.

    Reconstructing and reprogramming the tumor-propagating potential of glioblastoma stem-like cells

    Cell

    (2014)
  • K.L. Ligon et al.

    Olig2-regulated lineage-restricted pathway controls replication competence in neural stem cells and malignant glioma

    Neuron

    (2007)
  • J.D. Lathia et al.

    Integrin alpha 6 regulates glioblastoma stem cells

    Cell Stem Cell

    (2010)
  • J. Anido et al.

    TGF-β Receptor Inhibitors Target the CD44(high)/Id1(high) Glioma-Initiating Cell Population in Human Glioblastoma

    Cancer Cell

    (2010)
  • B.C. Prager et al.

    Cancer stem cells: the architects of the tumor ecosystem

    Cell Stem Cell

    (2019)
  • R. Stupp et al.

    European Organisation for Research and Treatment of Cancer brain Tumour and Radiation Oncology Groups, National Cancer institute of Canada Clinical Trials Group, Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial

    Lancet Oncol.

    (2009)
  • G. Liu et al.

    Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma

    Mol. Cancer

    (2006)
  • N. McGranahan et al.

    Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future

    Cell

    (2017)
  • A. Jemal et al.

    Annual report to the nation on the status of Cancer, 1975–2014, featuring survival

    J. Natl. Cancer Inst.

    (2017)
  • E. Tabouret et al.

    Recent trends in epidemiology of brain metastases: an overview

    Anticancer Res.

    (2012)
  • W.A. Hall et al.

    Long-term survival with metastatic cancer to the brain

    Med. Oncol.

    (2000)
  • A.K. Percy et al.

    Neoplasms of the central nervous system. Epidemiologic considerations

    Neurology

    (1972)
  • Y. Tsukada et al.

    Central nervous system metastasis from breast carcinoma. Autopsy study

    Cancer

    (1983)
  • P.W. Sperduto et al.

    Survival in patients with brain metastases: summary report on the updated diagnosis-specific graded prognostic assessment and definition of the eligibility quotient

    J. Clin. Oncol.

    (2020)
  • P.K. Brastianos et al.

    Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets

    Cancer Discov.

    (2015)
  • D.N. Louis et al.

    The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary

    Acta Neuropathol.

    (2016)
  • J. Wang et al.

    Medulloblastoma: From Molecular Subgroups to Molecular Targeted Therapies

    Annu. Rev. Neurosci.

    (2018)
  • P.A. Northcott et al.

    The whole-genome landscape of medulloblastoma subtypes

    Nature

    (2017)
  • P.A. Northcott et al.

    Medulloblastoma

    Nat. Rev. Dis. Primers

    (2019)
  • M.D. Taylor et al.

    Molecular subgroups of medulloblastoma: the current consensus

    Acta Neuropathol.

    (2012)
  • B. Manoranjan et al.

    Wnt activation as a therapeutic strategy in medulloblastoma

    Nat. Commun.

    (2020)
  • R. Stupp et al.

    Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial

    JAMA

    (2017)
  • R. Stupp et al.

    European organisation for research and treatment of Cancer brain tumor and radiotherapy groups, national Cancer institute of canada clinical trials group, radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma

    N. Engl. J. Med.

    (2005)
  • B.G. Harder et al.

    Developments in blood-brain barrier penetrance and drug repurposing for improved treatment of glioblastoma

    Front. Oncol.

    (2018)
  • N. Colwell et al.

    Hypoxia in the glioblastoma microenvironment: shaping the phenotype of cancer stem-like cells

    Neuro Oncol.

    (2017)
  • R.C. Gimple et al.

    Glioblastoma stem cells: lessons from the tumor hierarchy in a lethal cancer

    Genes Dev.

    (2019)
  • P.C. Burger et al.

    Patient age, histologic features, and length of survival in patients with glioblastoma multiforme

    Cancer

    (1987)
  • R. Reinartz et al.

    Functional subclone profiling for prediction of treatment-induced intratumor population shifts and discovery of rational drug combinations in human glioblastoma

    Clin. Cancer Res.

    (2017)
  • S. Nobusawa et al.

    Intratumoral patterns of genomic imbalance in glioblastomas

    Brain Pathol.

    (2010)
  • A. Kumar et al.

    Deep sequencing of multiple regions of glial tumors reveals spatial heterogeneity for mutations in clinically relevant genes

    Genome Biol.

    (2014)
  • J.D. Lathia et al.

    Cancer stem cells in glioblastoma

    Genes Dev.

    (2015)
  • K. Ludwig et al.

    Molecular markers in glioma

    J. Neurooncol.

    (2017)
  • S.E. Little et al.

    Receptor tyrosine kinase genes amplified in glioblastoma exhibit a mutual exclusivity in variable proportions reflective of individual tumor heterogeneity

    Cancer Res.

    (2012)
  • N.J. Szerlip et al.

    Intratumoral heterogeneity of receptor tyrosine kinases EGFR and PDGFRA amplification in glioblastoma defines subpopulations with distinct growth factor response

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

    (2012)
  • M.E. Hegi et al.

    Pathway analysis of glioblastoma tissue after preoperative treatment with the EGFR tyrosine kinase inhibitor gefitinib--a phase II trial

    Mol. Cancer Ther.

    (2011)
  • D.A. Haas-Kogan et al.

    Epidermal growth factor receptor, protein kinase B/Akt, and glioma response to erlotinib

    J. Natl. Cancer Inst.

    (2005)
  • T. Reungwetwattana et al.

    CNS Response to Osimertinib Versus Standard Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Patients With Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer

    J. Clin. Oncol.

    (2018)
  • J.-C. Soria et al.

    Osimertinib in untreated EGFR-Mutated advanced non–Small-Cell lung Cancer

    N. Engl. J. Med.

    (2017)
  • Cited by (48)

    View all citing articles on Scopus
    View full text