Review
Means to the ends: The role of telomeres and telomere processing machinery in metastasis

https://doi.org/10.1016/j.bbcan.2016.10.005Get rights and content

Abstract

Despite significant clinical advancements, cancer remains a leading cause of mortality throughout the world due largely to the process of metastasis and the dissemination of cancer cells from their primary tumor of origin to distant secondary sites. The clinical burden imposed by metastasis is further compounded by a paucity of information regarding the factors that mediate metastatic progression. Linear chromosomes are capped by structures known as telomeres, which dictate cellular lifespan in humans by shortening progressively during successive cell divisions. Although telomere shortening occurs in nearly all somatic cells, telomeres may be elongated via two seemingly disjoint pathways: (i) telomerase-mediated extension, and (ii) homologous recombination-based alternative lengthening of telomeres (ALT). Both telomerase and ALT are activated in various human cancers, with more recent evidence implicating both pathways as potential mediators of metastasis. Here we review the known roles of telomere homeostasis in metastasis and posit a mechanism whereby metastatic activity is determined by a dynamic fluctuation between ALT and telomerase, as opposed to the mere activation of a generic telomere elongation program. Additionally, the pleiotropic nature of the telomere processing machinery makes it an attractive therapeutic target for metastasis, and as such, we also explore the therapeutic implications of our proposed mechanism.

Introduction

When considered as a single disease, cancer is one of the leading causes of global mortality, with an estimated 14.9 million new cases and 8.2 million deaths attributable to cancer each year [1]. The incidence of many cancers is increasing in both developed and developing nations due in part to the prevalence of risk factors (e.g., tobacco and obesity) in an expanding and increasingly aging population [2]. Metastasis, while comprising only a fraction of this growing clinical burden, is responsible for the overwhelming majority of cancer mortality. Indeed, although the rates of diagnosing metastatic disease are typically low in many cancers (< 10–30%; [3], [4], [5]), approximately 90% of cancer-related deaths are attributable to metastasis [6]. The underlying lethality of metastasis reflects its molecular complexity, which has greatly limited the success of therapies targeting this process in both overt disease and adjuvant settings [7], [8], [9]. Thus, there remains a significant unmet need for novel therapeutic approaches to target metastasis.

Metastasis is most accurately thought of as a cascade of systemic and cellular events undertaken by a subset of cells within the primary tumor [10], [11]. Generally speaking, metastatic cells become liberated from well-vascularized, angiogenic primary tumors and undergo intravasation to gain access to the circulation, where they persist in the blood, lymph, or bone marrow. Upon reaching their target tissue, disseminated cells extravasate and initiate growth of pre-angiogenic “micrometastases” before fully colonizing the metastatic niche upon reinstatement of angiogenesis [10]. The classical view of metastasis as the terminal stage of cancer progression suggests that a subpopulation of primary tumor cells progressively acquire genetic alterations necessary for their dissemination and colonization, and that these cells remain rare until clonally expanded within secondary organs [12]. However, recent evidence indicates that the capacity of tumor cells to metastasize is present in the earliest stages of primary tumor development [13], [14] and that these variant cells are often genetically divergent from their primary tumor counterparts and from one another [15], [16], [17], [18]. In many respects, metastases may be considered as discrete entities from their primary tumors of origin due in part to their acquisition of genomic alterations during dissemination and distant organ colonization, suggesting that distinct regulatory pathways are operant during metastasis versus those active in primary tumor development [19].

Telomeres have long been implicated in driving tumorigenesis, yet emerging evidence indicates that the established concept whereby telomeres and their homeostatic machinery serve solely as cellular “immortalizers” may be drastically oversimplified. Indeed, telomeres and telomeric proteins subserve diverse functions in many of the stages that define the metastatic cascade. Herein we examine the varying roles that telomeres play in driving the dissemination and interaction of cancer cells with the metastatic microenvironment. We also discuss the therapeutic potential of targeting telomeres as a novel means to alleviate metastatic disease.

Section snippets

Metastasis at the cellular level

The metastatic cascade is defined by the following sequence of events: (i) primary tumor angiogenesis; (ii) cancer cell migration away from the primary tumor and intravasation into the tumor vascular supply; (iii) cancer cell survival within the circulation; (iv) extravasation of circulating tumor cells at secondary organs; and (v) proliferation of disseminated tumor cells (DTCs) at these secondary sites [19]. Each of these stages is spatially and temporally regulated by a host of cancer

Telomeres: dynamic structures with dynamic functions

Telomeres function as determinants of cellular age and replicative potential [62]. Consequently, aberrant telomere length imparts cells with replicative immortality, one of the hallmarks of cancer [63]. Indeed, abnormal telomere elongation is found in nearly all human cancers, with the vast majority of these exhibiting activation of the reverse transcriptase telomerase, while the remaining cases accomplish this task via the recombination-based Alternative Lengthening of Telomeres (ALT; [64],

Telomerase: more than a telomere machine

Telomerase is an RNA-dependent DNA polymerase that is composed of two moieties: (i) a RNA component (TR) that serves as a template for telomeric DNA replication, and (ii) a protein component (TERT) that is responsible for polymerase activity. The TERT protein contains four domains, and regions of the N- and C-terminal domains are sufficient to induce cell immortalization, even in the absence of catalytic activity [101]. Indeed, alternative splice variants of TERT lacking its catalytic domain

Telomere homeostasis: determinant or consequence of metastatic progression?

Associations between cancer and mutations in various telomeric proteins are continually being discovered (Table 1). However, the extent to which these aberrations specifically influence metastatic progression remains unclear. To date, many studies that have examined the association between cancer progression and telomere homeostasis have employed descriptive readouts of telomere dynamics, primarily differences in either telomere length or telomerase expression or activity that are then

Future directions: telomere-directed therapy for metastasis

The deadly nature of metastatic disease necessitates the development of therapies that specifically target essential pathways operant during dissemination. Current efforts aimed at anti-telomerase therapy have adopted several approaches, including direct enzyme inhibition, telomere destabilization, anti-telomerase immunotherapy, and telomerase-driven suicide gene therapy [147]. Enzyme inhibitors include both small molecules and RNA template antagonists, both of which have demonstrated robust

Financial disclosures/conflicts of interest

The authors declare that they have no competing interests, nor conflicts of interest.

Transparency document

Transparency document.

Acknowledgements

Research support was provided in part by the National Institutes of Health to NJR (T32GM007250 and TL1TR000441) and WPS (CA129359, CA177069, and CA194518), and by METAvivor to WPS.

References (176)

  • F. Chiacchiera et al.

    Blocking p38/ERK crosstalk affects colorectal cancer growth by inducing apoptosis in vitro and in preclinical mouse models

    Cancer Lett.

    (2012)
  • H. Gao et al.

    The BMP inhibitor Coco reactivates breast cancer cells at lung metastatic sites

    Cell

    (2012)
  • F.H. Brembeck et al.

    Balancing cell adhesion and Wnt signaling, the key role of β-catenin

    Curr. Opin. Genet. Dev.

    (2006)
  • A. Vlad-Fiegen et al.

    The Wnt pathway destabilizes adherens junctions and promotes cell migration via β-catenin and its target gene cyclin D1

    FEBS Open Bio.

    (2012)
  • S. Malladi et al.

    Metastatic latency and immune evasion through autocrine inhibition of WNT

    Cell

    (2016)
  • D. Hanahan et al.

    Hallmarks of cancer: the next generation

    Cell

    (2011)
  • S.Y. Lin et al.

    Multiple tumor suppressor pathways negatively regulate telomerase

    Cell

    (2003)
  • R.C. O'Hagan et al.

    Telomere dysfunction provokes regional amplification and deletion in cancer genomes

    Cancer Cell

    (2002)
  • J.P. Murnane

    Telomere dysfunction and chromosome instability

    Mutat. Res.

    (2012)
  • J.D. Griffith et al.

    Mammalian telomeres end in a large duplex loop

    Cell

    (1999)
  • C.W. Greider

    Telomeres do D-loop-T-loop

    Cell

    (1999)
  • J.Z. Ye et al.

    TIN2 binds TRF1 and TRF2 simultaneously and stabilizes the TRF2 complex on telomeres

    J. Biol. Chem.

    (2004)
  • T. Davoli et al.

    Telomere-driven tetraploidization occurs in human cells undergoing crisis and promotes transformation of mouse cells

    Cancer Cell

    (2012)
  • B.J. Lamarche et al.

    The MRN complex in double-strand break repair and telomere maintenance

    FEBS Lett.

    (2010)
  • L. Wu et al.

    Pot1 deficiency initiates DNA damage checkpoint activation and aberrant homologous recombination at telomeres

    Cell

    (2006)
  • J. Yu et al.

    PML3 interacts with TRF1 and is essential for ALT-associated PML bodies assembly in U2OS cells

    Cancer Lett.

    (2010)
  • C. Fitzmaurice et al.

    The global burden of cancer 2013

    JAMA Oncol.

    (2015)
  • L.A. Torre et al.

    Global cancer statistics, 2012

    CA Cancer J. Clin.

    (2015)
  • H. Kennecke et al.

    Metastatic behavior of breast cancer subtypes

    J. Clin. Oncol.

    (2010)
  • L.J. Schouten et al.

    Incidence of brain metastases in a cohort of patients with carcinoma of the breast, colon, kidney, and lung and melanoma

    Cancer

    (2002)
  • S. Manfredi et al.

    Epidemiology and management of liver metastases from colorectal cancer

    Ann. Surg.

    (2006)
  • P. Mehlen et al.

    Metastasis: a question of life or death

    Nat. Rev. Cancer

    (2006)
  • N. Sethi et al.

    Unravelling the complexity of metastasis - molecular understanding and targeted therapies

    Nat. Rev. Cancer

    (2011)
  • L.A. Mina et al.

    Rethinking the metastatic cascade as a therapeutic target

    Nat. Rev. Clin. Oncol.

    (2011)
  • J. Massague et al.

    Metastatic colonization by circulating tumour cells

    Nature

    (2016)
  • A.F. Chambers et al.

    Dissemination and growth of cancer cells in metastatic sites

    Nat. Rev. Cancer

    (2002)
  • K. Pantel et al.

    Dissecting the metastatic cascade

    Nat. Rev. Cancer

    (2004)
  • I.J. Fidler et al.

    Metastasis results from preexisting variant cells within a malignant tumor

    Science

    (1977)
  • S. Ramaswamy et al.

    A molecular signature of metastasis in primary solid tumors

    Nat. Genet.

    (2003)
  • J.E. Talmadge

    Clonal selection of metastasis within the life history of a tumor

    Cancer Res.

    (2007)
  • L. Torres et al.

    Intratumor genomic heterogeneity in breast cancer with clonal divergence between primary carcinomas and lymph node metastases

    Breast Cancer Res. Treat.

    (2007)
  • C.A. Klein

    Selection and adaptation during metastatic cancer progression

    Nature

    (2013)
  • D.P. Tabassum et al.

    Tumorigenesis: it takes a village

    Nat. Rev. Cancer

    (2015)
  • P. Carmeliet et al.

    Angiogenesis in cancer and other diseases

    Nature

    (2000)
  • P. Carmeliet et al.

    Molecular mechanisms and clinical applications of angiogenesis

    Nature

    (2011)
  • U. Sahin et al.

    Distinct roles for ADAM10 and ADAM17 in ectodomain shedding of six EGFR ligands

    J. Cell Biol.

    (2004)
  • K.D. Cowden Dahl et al.

    Matrix metalloproteinase 9 is a mediator of epidermal growth factor-dependent E-cadherin loss in ovarian carcinoma cells

    Cancer Res.

    (2008)
  • Q. Yu et al.

    Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-β and promotes tumor invasion and angiogenesis

    Genes Dev.

    (2000)
  • D. Mu et al.

    The integrin αvβ8 mediates epithelial homeostasis through MT1-MMP-dependent activation of TGF-β1

    J. Cell Biol.

    (2002)
  • M.K. Wendt et al.

    Deconstructing the mechanisms and consequences of TGF-β-induced EMT during cancer progression

    Cell Tissue Res.

    (2012)
  • Cited by (18)

    View all citing articles on Scopus
    View full text