Elsevier

Drug Resistance Updates

Volume 15, Issues 1–2, February–April 2012, Pages 123-131
Drug Resistance Updates

Cellular senescence and cancer chemotherapy resistance

https://doi.org/10.1016/j.drup.2012.01.002Get rights and content

Abstract

Innate or acquired resistance to cancer therapeutics remains an important area of biomedical investigation that has clear ramifications for improving cancer specific death rates. Importantly, clues to key resistance mechanisms may lie in the well-orchestrated and highly conserved cellular and systemic responses to injury and stress. Many anti-neoplastic therapies typically rely on DNA damage, which engages potent DNA damage response signaling pathways that culminate in apoptosis or growth arrest at checkpoints to allow for damage repair. However, an alternative cellular response, senescence, can also be initiated when challenged with these internal/external pressures and in ideal situations acts as a self-protecting mechanism. Senescence-induction therapies are an attractive concept in that they represent a normal, highly conserved and commonly invoked tumor-suppressing response to overwhelming genotoxic stress or oncogene activation. Yet, such approaches should ensure that senescence by-pass or senescence re-emergence does not occur, as emergent cells appear to have highly drug resistant phenotypes. Further, cell non-autonomous senescence responses may contribute to therapy-resistance in certain circumstances. Here we provide an overview of mechanisms by which cellular senescence plausibly contributes to therapy resistance and concepts by which senescence responses can be influenced to improve cancer treatment outcomes.

Introduction

Cancer is one of the most prevalent diseases diagnosed in developed countries and one of the leading causes of mortality in the United States. According to the National Cancer Institute approximately 1.6 million people were diagnosed with some form of cancer in 2010 and 570,000 lives were lost to neoplastic diseases (Howlader et al., 2011). Historically cancer was treated via surgical removal with some of the earliest indications of surgical intervention dating back to ancient Egypt with depictions of breast cancer surgeries (Mansfield, 1976). While surgical removal or radiotherapy of a solid tumor can be effective in treating localized primary disease, it has limited effectiveness in situations where tumor cells have spread outside of their tissue of origin.

The post-world war one era saw the dawning of systematic, scientifically based approaches for treating cancer patients. Experiments by Goodman and Gilman using nitrogen mustard for non-Hodgkin's lymphoma are representative of these early efforts that met with success. The evolution of systemic therapeutics and the science underlying their applications has resulted in complete cures for a subset of advanced tumor types and improved survival for most others. However, the ability of tumor cells to acquire resistance to cytotoxic and cytostatic agents can drastically reduce the efficacy of current interventions. It is interesting to note that the advent of chemotherapy was paralleled by the recognition of acquired therapy resistance (Chabner and Roberts, 2005). For patients with metastatic disease, drug resistance contributes to the majority of treatment failures (Longley and Johnston, 2005, Mahon et al., 2011). Despite initial responses, many tumors will relapse and progress regardless of repeated exposures to anti-neoplastic therapeutics. This naturally begs the question as to why? In this review we discuss the role of cellular senescence as a contributing factor in therapy resistance. We provide an overview of cellular damage responses to cancer treatments with a focus on signaling pathways leading to senescence, a state generally associated with tumor suppression. We describe evidence supporting contrary roles whereby senescence can enhance responses or promote resistance to cancer-directed therapeutics, and discuss opportunities to exploit senescence in the context of clinical care.

Section snippets

Chemotherapeutics, cellular damage and damage responses

Pharmacological agents designed to induce tumor cell death and/or suppress growth fall into several classes based on mechanism of action. Whereas recent advances in anti-cancer therapeutics have focused on developing inhibitors that exploit specific oncogenic mutations that hyper-activate growth regulatory pathways, the most widely used agents are poorly selective for neoplastic cells and rely on marginal differences between benign cells and tumor cells that involve proliferation rates, DNA

The DNA Damage Response

The DNA Damage Response (DDR) is a complex developmentally conserved process which is initiated following injury to the integrity of DNA. This response is set in motion to protect cells from irreversible damage following exposure to exogenous/endogenous genotoxins, and to eliminate those cells with damage too extreme to repair fully. There are several factors that can induce the damage response ranging from UV exposure to commonly used cancer treatments such as γ-radiation and chemotherapeutics

Cellular senescence: mechanisms and outcomes

The word senescence is derived from the Latin word senex, meaning old age or advanced in age. In addition to describing an organismal state that is associated with advanced chronological age, the term senescence has also been applied to a phenotype observed in individual cells, also associated with chronological age—often measured by numbers of cell divisions. Cellular senescence is also influenced by other factors that modify DNA and the functions of other organelles to phenocopy the

The Pro: promoting tumor senescence to overcome drug resistance

It is important to recognize that divergent views exist regarding the dynamics of senescence in the clinical setting, and the ramifications of senescence as a desirable or adverse contributor to therapeutic responses. There have been an increasing number of reports indicating that initiating a senescence program in solid tumors could be a potential therapeutic treatment to overcome drug resistance. This is based on the idea that tumor cells remain prone to senescence and that they are readily

The Con: senescence and the tumor microenvironment promote resistance

The importance of the microenvironment and its role in therapy response is increasingly recognized. A patient treated with cytotoxic agents receives systemic damage impacting normal constituents of tissue and organ systems as well as the neoplastic cells. The resulting damage may elicit a response from the tumor microenvironment (TME) which potentially can have a dramatic impact upon treatment efficacy. Several studies have documented that the DDR induces a remarkable spectrum of secreted

Therapy resistance and breaking senescence

While senescence is generally considered an irreversible state of cellular arrest, the potential dangers of an emergent phenotype should be considered. If cells in the arrested state are not cleared via processes such as phagocytosis they remain in the organism harboring the potential to recapture the proliferative state, often with damaged genomes. There have been reports both supporting immune mediated attrition (Xue et al., 2007) and the continued accretion of senescent cells in vivo (Dimri

Conclusions and future directions

Innate or acquired resistance to cancer therapeutics remains an important area of biomedical investigation that has clear ramifications for improving cancer specific death rates. In this context, manipulating cellular senescence may improve therapy responses via several mechanisms. The promotion of ‘intrinsic’ tumor cell senescence is an attractive concept in that it represents a normal, highly conserved and commonly invoked tumor-suppressing response to overwhelming genotoxic stress or

Acknowledgment

This work was supported by NIH grants P50CA097186, U54CA126540 and U01CA164188.

References (114)

  • L.W. Elmore et al.

    Adriamycin-induced senescence in breast tumor cells involves functional p53 and telomere dysfunction

    J. Biol. Chem.

    (2002)
  • D.A. Gewirtz et al.

    Accelerated senescence: an emerging role in tumor cell response to chemotherapy and radiation

    Biochem. Pharmacol.

    (2008)
  • L.A. Gilbert et al.

    DNA damage-mediated induction of a chemoresistant niche

    Cell

    (2010)
  • I. Gomez-Curet et al.

    c-Myc inhibition negatively impacts lymphoma growth

    J. Pediatr. Surg.

    (2006)
  • J.W. Harper et al.

    The DNA damage response: ten years after

    Mol. Cell

    (2007)
  • L. Hayflick et al.

    The serial cultivation of human diploid cell strains

    Exp. Cell Res.

    (1961)
  • H.Y. Ho et al.

    Enhanced oxidative stress and accelerated cellular senescence in glucose-6-phosphate dehydrogenase (G6PD)-deficient human fibroblasts

    Free Radic. Biol. Med.

    (2000)
  • M.J. Huang et al.

    A small-molecule c-Myc inhibitor, 10058-F4, induces cell-cycle arrest, apoptosis, and myeloid differentiation of human acute myeloid leukemia

    Exp. Hematol.

    (2006)
  • R.J. Knox et al.

    A new cytotoxic. DNA interstrand crosslinking agent, 5-(aziridin-1-yl)-4-hydroxylamino-2-nitrobenzamide, is formed from 5-(aziridin-1-yl)-2,4-dinitrobenzamide (CB 1954) by a nitroreductase enzyme in Walker carcinoma cells

    Biochem. Pharmacol.

    (1988)
  • T. Kuilman et al.

    Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network

    Cell

    (2008)
  • N. Mailand et al.

    RNF8 ubiquitylates histones at DNA double-strand breaks and promotes assembly of repair proteins

    Cell

    (2007)
  • D.H. Nguyen et al.

    Radiation acts on the microenvironment to affect breast carcinogenesis by distinct mechanisms that decrease cancer latency and affect tumor type

    Cancer Cell

    (2011)
  • P.E. Puig et al.

    Tumor cells can escape DNA-damaging cisplatin through DNA endoreduplication and reversible polyploidy

    Cell Biol. Int.

    (2008)
  • M. Serrano et al.

    Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a

    Cell

    (1997)
  • D.N. Shelton et al.

    Microarray analysis of replicative senescence

    Curr. Biol.

    (1999)
  • R. Sidi et al.

    Induction of senescence markers after neo-adjuvant chemotherapy of malignant pleural mesothelioma and association with clinical outcome: an exploratory analysis

    Eur. J. Cancer

    (2011)
  • M. Stucki et al.

    gammaH2AX and MDC1: anchoring the DNA-damage-response machinery to broken chromosomes

    DNA Repair (Amst)

    (2006)
  • A. Alimonti et al.

    A novel type of cellular senescence that can be enhanced in mouse models and human tumor xenografts to suppress prostate tumorigenesis

    J. Clin. Invest.

    (2010)
  • M.H. Barcellos-Hoff et al.

    Radiation and the microenvironment—tumorigenesis and therapy

    Nat. Rev. Cancer

    (2005)
  • J. Bartkova et al.

    Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints

    Nature

    (2006)
  • C. Bavik et al.

    The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms

    Cancer Res.

    (2006)
  • C.M. Beausejour et al.

    Reversal of human cellular senescence: roles of the p53 and p16 pathways

    EMBO J.

    (2003)
  • M. Braig et al.

    Oncogene-induced senescence as an initial barrier in lymphoma development

    Nature

    (2005)
  • C.J. Brown et al.

    Awakening guardian angels: drugging the p53 pathway

    Nat. Rev. Cancer

    (2009)
  • J.P. Brown et al.

    Bypass of senescence after disruption of p21CIP1/WAF1 gene in normal diploid human fibroblasts

    Science

    (1997)
  • M.C. Caino et al.

    Hallmarks for senescence in carcinogenesis: novel signaling players

    Apoptosis

    (2009)
  • B.A. Chabner et al.

    Timeline: chemotherapy and the war on cancer

    Nat. Rev. Cancer

    (2005)
  • B.D. Chang et al.

    A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents

    Cancer Res.

    (1999)
  • B.D. Chang et al.

    Molecular determinants of terminal growth arrest induced in tumor cells by a chemotherapeutic agent

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

    (2002)
  • C.F. Chantrain et al.

    Bone marrow microenvironment and tumor progression

    Cancer Microenviron.

    (2008)
  • S.K. Chao et al.

    Resistance to discodermolide, a microtubule-stabilizing agent and senescence inducer, is 4E-BP1-dependent

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

    (2011)
  • J.H. Chen et al.

    DNA damage, cellular senescence and organismal ageing: causal or correlative?

    Nucleic Acids Res.

    (2007)
  • Z. Chen et al.

    Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis

    Nature

    (2005)
  • Y. Chien et al.

    Control of the senescence-associated secretory phenotype by NF-kappaB promotes senescence and enhances chemosensitivity

    Genes Dev.

    (2011)
  • L. Chin et al.

    Cooperative effects of INK4a and ras in melanoma susceptibility in vivo

    Genes Dev.

    (1997)
  • A.R. Choudhury et al.

    Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation

    Nat. Genet.

    (2007)
  • M. Collado et al.

    Senescence in tumours: evidence from mice and humans

    Nat. Rev. Cancer

    (2010)
  • J.P. Coppe et al.

    Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor

    PLoS Biol.

    (2008)
  • S. Courtois-Cox et al.

    Many roads lead to oncogene-induced senescence

    Oncogene

    (2008)
  • F. d’Adda di Fagagna et al.

    A DNA damage checkpoint response in telomere-initiated senescence

    Nature

    (2003)
  • Cited by (119)

    View all citing articles on Scopus
    View full text