Chapter 4 - Telomeres, Telomerase, And Cancer

Embed Size (px)

Citation preview

  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    1/21

    Chapter 4: Telomeres, Telomerase, and Cancer

    Telomeres, Telomerase, and Cancer: Introduction

    Maintenance of most adult organ systems requires extensive cell renewal, typified most

    strikingly by the replacement of the intestinal lining on a weekly basis and the production of

    trillions of new blood cells daily. Yet a lifetime of factors including continual telomere erosion,

    errors in DNA replication, intrinsic and carcinogen-induced somatic mutations, cancer-

    relevant germline variants, and epigenetic insults conspire to endow cells with the large

    number of changes needed for malignant transformation. How is it that replicating tissues,

    showered with myriad cancer-relevant somatic alterations, resist malignant transformation?

    That these mutations are indeed present in normal human tissues is reflected by the

    remarkable observations that roughly 1% of neonatal cord blood collections contain

    significant numbers of myeloid clones harboring oncogenic fusions such as the AML1-ETO

    fusion associated with acute leukemia,1and that approximately one-third of adults possess

    the IgH-BCL2translocation associated with follicular lymphomagenesis.2As the prevalence

    of these cancers is far lower in the general population, these observations imply that potent

    tumor suppressor mechanisms must be operating to constrain the growth and survival of

    these aspiring cancer cells.

    The most prominent biologic manifestations of an activated tumor suppressor response are

    apoptosis (cell death) and senescence (permanent cell cycle arrest). These biologic processes

    are linked to powerful checkpoint effector molecules involving the p16INK4a-Rb pathway, the

    ARF-p53 pathway, and specialized chromosomal DNA ends termed telomeres. These genetic

    elements comprise powerful tumor suppressor barriers and act cooperatively to eliminate or

    to place a limit on the replicative lifespan of rogue would-be cells. The importance of

    apoptosis in preventing cancer is further discussed in Chapter7.The focus of this chapter will

    be on the role of telomere dynamics and associated telomere-related cellular checkpoint

    processes, particularly senescence, in the regulation of neoplastic transformation. A

    significant body of clinical and translational science now supports such a role for telomeres

    and cellular senescence, and in this chapter, we present rapidly increasing clinical data

    pointing to the relevance of these processes in human disease, particularly cancer. Indeed, it

    is worth noting that the Nobel Prize for physiology or medicine in 2009 was awarded to

    Blackburn, Greider, and Szostak for their pioneering and seminal work in telomere biology

    that advanced our present understanding of aging and cancer.

    Telomeres and Telomerase

    Telomere dysfunction is a principal tumor suppressor mechanism manifesting most

    prominently as apoptosis and senescence. At the same time, when accompanied by

    functional p53 loss, the genome-destabilizing impact of telomere dysfunction can cause

    widespread mutations that propel normal cells toward malignant transformation. Thus, the

    telomere-based anticancer mechanism can actually fuel tumorigenesis in certain contexts.

    The knowledge of the basic biology of telomeres and telomerase has yielded fundamental

    insights into both cancer prevention and cancer promotion. The powerful and complex impact

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762879#8762879http://lwwoncology.com/Textbook/Content.aspx?aid=8762879#8762879http://lwwoncology.com/Textbook/Content.aspx?aid=8762879#8762879http://lwwoncology.com/Textbook/Content.aspx?aid=8762880#8762880http://lwwoncology.com/Textbook/Content.aspx?aid=8762880#8762880http://lwwoncology.com/Textbook/Content.aspx?aid=8762880#8762880http://lwwoncology.com/Textbook/Content.aspx?aid=ch7http://lwwoncology.com/Textbook/Content.aspx?aid=ch7http://lwwoncology.com/Textbook/Content.aspx?aid=ch7http://lwwoncology.com/Textbook/Content.aspx?aid=ch7http://lwwoncology.com/Textbook/Content.aspx?aid=8762880#8762880http://lwwoncology.com/Textbook/Content.aspx?aid=8762879#8762879
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    2/21

    of telomere dynamics in model organisms and humans reflects the crucial role of telomere

    function in processes of genomic instability, organ homeostasis, chronic diseases, aging, and

    tumorigenesis. With respect to tumorigenesis, the study of telomeres in the mouse has

    provided insight into how advancing age in humans fuels the development of epithelial

    cancers as well as how chronic inflammation and degeneration may engender increasedcancer risk in affected organs. These advances in the basic understanding of telomere

    maintenance are now being translated into clinically relevant applications that may have an

    impact on the diagnosis and management of a broad spectrum of cancers as well as aging,

    age-related disorders, and degenerative conditions. The important role of telomere biology in

    aging and degenerative diseases has been reviewed elsewhere.3,4

    Telomeres

    Telomeres are specialized nucleoprotein complexes at the ends of linear chromosomes

    consisting of long arrays of double-stranded TTAGGG repeats, a G-rich 3 single-strand

    overhang, and associated telomeric repeat binding57(Fig.4.1). The work of Muller andMcClintock in the 1930s led to the concept that telomeres function to cap chromosomal

    termini and prevent end-to-end recombination, thereby maintaining chromosomal integrity.

    Subsequent work has substantiated this model across the animal and plant kingdom,

    underscoring the critical roles served by the telomere complex.

    Figure 4.1. Human telomere structure.

    A: Human telomeres form telomere loop (T loop) and displacement loop (D loop) secondary

    structures. Long stretches of telomeric repeats create a loop-back structure (T loop),

    completed by the invasion of the single GT-rich 3 overhang into the double-stranded DNA

    molecule (D loop), thus protecting the chromosome terminus. B: In human cells, double-

    stranded telomeric repeats are bound directly by two proteins, TRF1 (TTAGG repeat binding

    factor 1) and TRF2. Cell culture studies have suggested that the main function of TRF1 is to

    regulate telomere length, whereas TRF2 functions to protect telomeres from activating

    nonhomologous end-joining (NHEJ) and other DNA repair or DNA damage responsepathways. TRF2 also interacts with the human Rap1 protein (hRap). Biochemical studies also

    suggest that the formation of the T loop depends on TRF2. Another protein, POT1 (protection

    of telomere 1), has been shown to bind to the single-stranded human telomeric 3 overhang.

    Two shelterin proteins, TIN2 and TPP1, connect POT1 to TRF1 and TRF2. POT1 has been

    proposed to interact with TRF1 complexes to regulate telomere length. Thus, there is

    significant interplay between telomeric binding proteins and the formation of the

    secondary/tertiary structures that protect the ends of chromosomes.

    Telomere structure and function have been studied extensively in mammals. Although the

    overall structural features of telomeres are preserved among different mammalian

    organisms, lengths can vary considerably from species to species: for example, 5 to 15 kb

    for humans versus 20 to 80 kb for the laboratory mouse. On the structural level, electron

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762881#8762881http://lwwoncology.com/Textbook/Content.aspx?aid=8762881#8762881http://lwwoncology.com/Textbook/Content.aspx?aid=8762882#8762882http://lwwoncology.com/Textbook/Content.aspx?aid=8762882#8762882http://lwwoncology.com/Textbook/Content.aspx?aid=8762882#8762882http://lwwoncology.com/Textbook/Content.aspx?aid=8762883#8762883http://lwwoncology.com/Textbook/Content.aspx?aid=8762883#8762883http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762827%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762827%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762827%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762827%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762827%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762827%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762827%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762827%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762883#8762883http://lwwoncology.com/Textbook/Content.aspx?aid=8762883#8762883http://lwwoncology.com/Textbook/Content.aspx?aid=8762882#8762882http://lwwoncology.com/Textbook/Content.aspx?aid=8762881#8762881
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    3/21

    microscopy and other studies have shown that telomeres form complex secondary and

    tertiary structures via DNA-DNA interactions between the telomeric repeats, DNA-protein

    interactions between the telomeric DNA and the telomeric repeat binding proteins (shelterins

    or telosomes8,9), and protein-protein interactions between the telomeric repeat binding

    proteins themselves and other associated proteins (Fig.4.1). The formation of this well-documented, higher-order DNA-protein complex has provided a working model of how the

    telomere functions as a capping structure, preventing the ends of linear chromosomal DNA

    from being recognized as either a DNA double-strand break (DSB) or DNA single-strand

    break, thereby avoiding activation of the DNA damage response and the formation of

    chromosomal end-to-end fusions through the DNA repair machinery.7

    Many proteins involved in DNA DSB repair, including nonhomologous end-joining and

    homologous recombination processes, have been found to be physically associated with the

    telomeres.710These findings have fueled speculation that DSB repair proteins provide a

    protective role at the telomere; for example, by sequestering the telomere end from the DNAdamage surveillance/repair machinery. Experimental support for this hypothesis has

    emerged from the mouse, in which germ line inactivation of various repair proteins (e.g., Ku

    and DNA-PK) results in reduced telomere length or loss of capping function, or both, leading

    to increased end-to-end fusions.11Correspondingly, in cultured human cells, experimental

    disruption of telomere-binding proteins results in the unraveling of higher-order

    nucleoprotein structure and telomere localization of DNA DSB surveillance/repair proteins

    (e.g., 53BP1, gamma-H2AX, Rad17, ATM, and Mre11), establishing that dysfunctional

    telomeres can indeed serve as substrates for the classic DNA repair machinery.12Recently,

    elegant in vitroand mouse genetic experiments have shown that subunits of the shelterin

    complex actively repress the ATM and ATR DNA damage signaling pathways.7,13

    A further understanding of the molecular mechanisms governing the repression versus

    activation of the DNA DSB surveillance/repair apparatus at the telomere could lead to the

    development of novel cancer therapeutic options. For example, the design of agents that can

    uncap telomeres while preserving the DNA damage checkpoint response yet neutralize the

    actual DNA damage repair process would be ideal because they would produce unrepaired

    DSBs and elicit cell-cycle arrest or apoptosis responses. Lastly, in the near future, agents

    designed to uncap the telomeres could be used in combination with conventional

    chemotherapeutic agents that create DSB for cancer treatment, thereby simultaneously

    targeting these intertwined pathways.

    Telomerase

    Conventional DNA polymerases operating in the S phase of the cell cycle require an RNA

    primer for reverse-strand synthesis, resulting in incomplete DNA replication of telomeres

    during each cell division. The solution to this end-replication problem is the telomere-

    synthesizing telomerase enzyme, a specialized ribonucleoprotein complex with reverse

    transcriptase activity. The functional telomerase holoenzyme is a large multisubunit complex

    that includes an essential telomerase RNA (hTERC) component serving as a template for the

    addition of telomere repeats and a telomerase reverse transcriptase (hTERT) catalytic

    subunit.14In some normal human somatic cells, telomerase levels are insufficient to

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762886#8762886http://lwwoncology.com/Textbook/Content.aspx?aid=8762886#8762886http://lwwoncology.com/Textbook/Content.aspx?aid=8762887#8762887http://lwwoncology.com/Textbook/Content.aspx?aid=8762887#8762887http://lwwoncology.com/Textbook/Content.aspx?aid=8762887#8762887http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762827%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762827%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762827%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762888#8762888http://lwwoncology.com/Textbook/Content.aspx?aid=8762888#8762888http://lwwoncology.com/Textbook/Content.aspx?aid=8762889#8762889http://lwwoncology.com/Textbook/Content.aspx?aid=8762889#8762889http://lwwoncology.com/Textbook/Content.aspx?aid=8762889#8762889http://lwwoncology.com/Textbook/Content.aspx?aid=8762890#8762890http://lwwoncology.com/Textbook/Content.aspx?aid=8762890#8762890http://lwwoncology.com/Textbook/Content.aspx?aid=8762890#8762890http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762891#8762891http://lwwoncology.com/Textbook/Content.aspx?aid=8762891#8762891http://lwwoncology.com/Textbook/Content.aspx?aid=8762891#8762891http://lwwoncology.com/Textbook/Content.aspx?aid=8762892#8762892http://lwwoncology.com/Textbook/Content.aspx?aid=8762892#8762892http://lwwoncology.com/Textbook/Content.aspx?aid=8762892#8762892http://lwwoncology.com/Textbook/Content.aspx?aid=8762892#8762892http://lwwoncology.com/Textbook/Content.aspx?aid=8762891#8762891http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762890#8762890http://lwwoncology.com/Textbook/Content.aspx?aid=8762889#8762889http://lwwoncology.com/Textbook/Content.aspx?aid=8762888#8762888http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://lwwoncology.com/Textbook/Content.aspx?aid=8762885#8762885http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762827%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762887#8762887http://lwwoncology.com/Textbook/Content.aspx?aid=8762886#8762886
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    4/21

    maintain telomere length, resulting in progressive attrition with each cell division. This forms

    the basis for the theory that the metered loss of telomeres can serve as a cellular mitotic

    clock that ultimately limits the number of cell divisions and cellular lifespan. In support of

    this view, shortening of telomere length with aging can be demonstrated in human peripheral

    blood cells,1517and the rate of shortening can be associated with conditions of increasedhematopoietic stem cell turnover (e.g., in paroxysmal nocturnal hemoglobinuria).18

    Many normal somatic human cells and differentiated tissues express readily detectable levels

    of the hTERC component. In contrast, hTERT expression and activity are more restricted

    because of stringent regulation on the levels of transcriptional initiation, alternative RNA

    processing, posttranslational modification, and subcellular localization. With the identification

    of an increasing number of TERT-associated proteins, it is likely that additional regulatory

    mechanisms will surface, such as those governing the accessibility of the telomerase

    holoenzyme onto the telomere end.19Here again, a more complete elucidation of these

    regulatory mechanisms may provide additional therapeutic strategies that can preferentiallytarget telomerase-mediated telomere maintenance in cancer cells. Indeed, the development

    of such selective strategies may become paramount and more challenging as recent studies

    have revealed low telomerase levels in cycling somatic human cells that were previously

    thought to have no telomerase activity.20Eradication of residual telomerase function in

    these primary cells alters the maintenance of the 3 single-strand telomeric overhang without

    changing the rate of overall telomere shortening, resulting in diminished proliferation rates

    and overall reduction in proliferative capacity. These studies support an additional protective

    function of telomerase at the telomeres21and raise concerns that generalized

    antitelomerase therapy could lead to the immediate uncapping of telomeres in normal cells,

    thus limiting the use of antitelomerase therapy in cancer patients.

    Lastly, in addition to forming the telomerase holoenzyme complex with TERC, TERT was

    recently shown to be able to interact with the RNA component of mitochondrial RNA

    processing endoribonuclease (RMRP). This distinct TERT/RMRP ribonucleoprotein complex

    has RNA-dependent RNA polymerase activity and produces double-stranded RNAs that can

    be processed into small interfering RNAs.22Also, there is compelling experimental evidence

    that TERT can interact and engage the Wnt signaling pathway.23,24These results suggest

    that TERT contributes to cell physiology independently of its ability to elongate telomeres, a

    fact that further complicates efforts to specifically target telomerase enzymatic activity as an

    anticancer therapy.

    Senescence

    Primary human cells, even when cultured under optimal conditions, will eventually encounter

    a cell division barrier, termed cellular senescence,triggered by critically shortened

    telomeres. Senescence is a specific cell biologic phenotype composed of a permanent and

    durable growth arrest, alterations in cellular morphology, expression of characteristic

    markers of senescence such as senescence-associated (SA) -galactosidase activity, and

    alterations of chromatin structure to a growth-repressive state.25Induction of senescence is

    intimately associated with p16INK4a

    and p53 activation, and when induced in vitroas a resultof telomere dysfunction, this barrier is termed the Hayflick limit(M1) in honor of the

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762893#8762893http://lwwoncology.com/Textbook/Content.aspx?aid=8762893#8762893http://lwwoncology.com/Textbook/Content.aspx?aid=8762895#8762895http://lwwoncology.com/Textbook/Content.aspx?aid=8762895#8762895http://lwwoncology.com/Textbook/Content.aspx?aid=8762895#8762895http://lwwoncology.com/Textbook/Content.aspx?aid=8762896#8762896http://lwwoncology.com/Textbook/Content.aspx?aid=8762896#8762896http://lwwoncology.com/Textbook/Content.aspx?aid=8762896#8762896http://lwwoncology.com/Textbook/Content.aspx?aid=8762897#8762897http://lwwoncology.com/Textbook/Content.aspx?aid=8762897#8762897http://lwwoncology.com/Textbook/Content.aspx?aid=8762897#8762897http://lwwoncology.com/Textbook/Content.aspx?aid=8762898#8762898http://lwwoncology.com/Textbook/Content.aspx?aid=8762898#8762898http://lwwoncology.com/Textbook/Content.aspx?aid=8762898#8762898http://lwwoncology.com/Textbook/Content.aspx?aid=8762899#8762899http://lwwoncology.com/Textbook/Content.aspx?aid=8762899#8762899http://lwwoncology.com/Textbook/Content.aspx?aid=8762899#8762899http://lwwoncology.com/Textbook/Content.aspx?aid=8762900#8762900http://lwwoncology.com/Textbook/Content.aspx?aid=8762900#8762900http://lwwoncology.com/Textbook/Content.aspx?aid=8762900#8762900http://lwwoncology.com/Textbook/Content.aspx?aid=8762901#8762901http://lwwoncology.com/Textbook/Content.aspx?aid=8762901#8762901http://lwwoncology.com/Textbook/Content.aspx?aid=8762902#8762902http://lwwoncology.com/Textbook/Content.aspx?aid=8762902#8762902http://lwwoncology.com/Textbook/Content.aspx?aid=8762903#8762903http://lwwoncology.com/Textbook/Content.aspx?aid=8762903#8762903http://lwwoncology.com/Textbook/Content.aspx?aid=8762903#8762903http://lwwoncology.com/Textbook/Content.aspx?aid=8762903#8762903http://lwwoncology.com/Textbook/Content.aspx?aid=8762902#8762902http://lwwoncology.com/Textbook/Content.aspx?aid=8762901#8762901http://lwwoncology.com/Textbook/Content.aspx?aid=8762900#8762900http://lwwoncology.com/Textbook/Content.aspx?aid=8762899#8762899http://lwwoncology.com/Textbook/Content.aspx?aid=8762898#8762898http://lwwoncology.com/Textbook/Content.aspx?aid=8762897#8762897http://lwwoncology.com/Textbook/Content.aspx?aid=8762896#8762896http://lwwoncology.com/Textbook/Content.aspx?aid=8762895#8762895http://lwwoncology.com/Textbook/Content.aspx?aid=8762893#8762893http://lwwoncology.com/Textbook/Content.aspx?aid=8762893#8762893
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    5/21

    discoverer of senescence.26Because loss of p16INK4a-RB and/or p53 pathway function in

    primary human cells permits additional cell divisions beyond the Hayflick limit, these

    pathways appear to be involved in the activation of this senescence program brought about

    by the shortened telomere signal.

    Beyond the connection with telomeres, cellular senescence appears to be a general

    anticancer mechanism, induced by a variety of oncogenic stresses. In addition to telomere

    erosion or structural uncapping (see later discussion), senescence is also induced by forms of

    DNA damage, oxidative stress, suboptimal growth conditions, and activation of certain

    oncogenes (reviewed in refs. 8 and 13). Senescence requires activation of the Rb and/or p53

    protein; and expression of their regulators such as p16INK4a, p21CIP, and ARF (Fig.4.2).27

    30An important form of senescence is induced by p53, which has several antiproliferative

    activities including stimulation of the expression of p21CIP, a cyclin-dependent kinase

    inhibitor. These inhibit progression through the cell cycle by inhibiting cyclin-dependent

    kinases that phosphorylate and thereby inactivate Rb and related proteins p107 andp130.31The activation of p53 is predominantly effected by specific posttranslational

    modifications and its stabilization, which are prompted by the same stimuli that induce its

    expression, including telomere dysfunction, DNA damage, and oncogene activation (reviewed

    in refs. 18 through 20), as well as inappropriate cell cycle entry.32,33A major sensor of

    oncogene activation and inappropriate cell cycle entry is ARF (also designated p14ARFin the

    human or p19ARFin the mouse), which binds to and blocks MDM2-mediated degradation of

    p53.3336

    Figure 4.2. The INK4a/ARF/INK4blocus (also calledCDKN2aand CDKN2b) and

    downstream pathways.The locus contains three open reading frames encoding the ARF, p15INK4b, and p16INK4atumor

    suppressor proteins. p16INK4aand p15INK4binhibit the activity of the proliferative kinases

    CDK4/6, which phosphorylate RB and related proteins p107 and p130.

    Therefore, INK4expression induces RB-family hypophosphorylation, which in turn represses

    E2F-regulated transcription and cell-cycle arrest. ARF inhibits the MDM2-mediated

    degradation of p53; and p53 stabilization in turn induces a number of targets including many

    proteins involved in cell-cycle arrest or apoptosis. The entire locus spans a mere 35 kb in the

    human genome, and inactivation of all three genes by a single genetic deletion is common in

    many human and murine cancers.

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762904#8762904http://lwwoncology.com/Textbook/Content.aspx?aid=8762904#8762904http://lwwoncology.com/Textbook/Content.aspx?aid=8762904#8762904http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762838%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762905#8762905http://lwwoncology.com/Textbook/Content.aspx?aid=8762905#8762905http://lwwoncology.com/Textbook/Content.aspx?aid=8762905#8762905http://lwwoncology.com/Textbook/Content.aspx?aid=8762908#8762908http://lwwoncology.com/Textbook/Content.aspx?aid=8762908#8762908http://lwwoncology.com/Textbook/Content.aspx?aid=8762909#8762909http://lwwoncology.com/Textbook/Content.aspx?aid=8762909#8762909http://lwwoncology.com/Textbook/Content.aspx?aid=8762909#8762909http://lwwoncology.com/Textbook/Content.aspx?aid=8762910#8762910http://lwwoncology.com/Textbook/Content.aspx?aid=8762910#8762910http://lwwoncology.com/Textbook/Content.aspx?aid=8762911#8762911http://lwwoncology.com/Textbook/Content.aspx?aid=8762911#8762911http://lwwoncology.com/Textbook/Content.aspx?aid=8762911#8762911http://lwwoncology.com/Textbook/Content.aspx?aid=8762911#8762911http://lwwoncology.com/Textbook/Content.aspx?aid=8762911#8762911http://lwwoncology.com/Textbook/Content.aspx?aid=8762911#8762911http://lwwoncology.com/Textbook/Content.aspx?aid=8762914#8762914http://lwwoncology.com/Textbook/Content.aspx?aid=8762914#8762914http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762838%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762914#8762914http://lwwoncology.com/Textbook/Content.aspx?aid=8762911#8762911http://lwwoncology.com/Textbook/Content.aspx?aid=8762911#8762911http://lwwoncology.com/Textbook/Content.aspx?aid=8762911#8762911http://lwwoncology.com/Textbook/Content.aspx?aid=8762910#8762910http://lwwoncology.com/Textbook/Content.aspx?aid=8762909#8762909http://lwwoncology.com/Textbook/Content.aspx?aid=8762908#8762908http://lwwoncology.com/Textbook/Content.aspx?aid=8762905#8762905http://lwwoncology.com/Textbook/Content.aspx?aid=8762905#8762905http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762838%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762904#8762904
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    6/21

    Another prominent molecular correlate of senescence is up-regulation of the cyclin-

    dependent kinase inhibitor, p16INK4a, which increases markedly in senescent cells on passage

    in culture or advancing age in tissues.37Correspondingly, ectopic expression of p16INK4ais

    sufficient to induce senescence in some cell types,38and senescence can be delayed or

    prevented in some cell types by p16

    INK4a

    silencing or neutralization by antisense or siRNA.3943The regulation of p16INK4ais not as well understood as that of p53, although it appears to

    be induced by several stimuli, including oncogene activation and growth in

    culture.37Activation of p53 (and hence p21CIP1) and/or accumulating levels of p16INK4ais able

    to produce Rb-family member protein hypophosphorylation and activation, which leads to

    repression of cell-cycle progression,27,30enabling initiation of the senescence process.

    Recent data have suggested that Rb may be of particular importance in the promotion of

    senescence compared with its related family members p107 and p130, likely explaining the

    frequent inactivation of Rb in human cancers relative to the other Rb-family members.44

    Senescence as a Cancer Prevention MechanismSeveral lines of evidence have suggested an important role for senescence in the prevention

    of cancerin vivo. It is important to note that the field has been limited by the lack of

    robust in vivobiomarkers of senescence. Although (SA)--galactosidase and

    p16INK4aexpression have been used as markers of in vivosenescence, both have certain

    limitations and neither can be considered unequivocal proof of senescent state in vivo. These

    technical shortcomings notwithstanding, a growth arrest important for the prevention of

    tumorigenesis with characteristic features of senescence (p16INK4aexpression and (SA)--

    galactosidase expression) has been noted in several murine and human in vivotumor

    systems, and we believe the data suggest bona fide senescence occurs in the intact

    organism.

    The lines of evidence for senescence as a tumor suppressor mechanism are quite strong.

    First, the aforementioned minimal residual disease data showing frequent oncogenic

    translocations and other mutagenic events demonstrate a constant need for tumor

    suppression, even in young animals. Additionally, several of the initially described tumor

    suppressor proteins that are mutated in familial cancer syndromes (e.g., p16INK4a, p53, Rb)

    are intimately involved in the induction of senescence. Mice lacking p16INK4aor p53 are prone

    to spontaneous cancers,4547and mice with severe compromise of the senescence pathway

    due to combined p16INK4aand p53 inactivation die of cancer, often harboring multiple

    synchronous primary tumors, with a median age of 8 weeks (compared with a normal murine

    lifespan of more than 100 weeks).19Importantly, mice and humans with impaired

    p16INK4aand/or p53 function develop with only modest phenotypic alterations other than an

    age-dependent increase in cancer and an increased susceptibility to cancer following

    carcinogen exposure. Several groups have demonstrated a senescencelike growth arrest in

    murine and human tissues in association with somatic oncogenic events.4854For example,

    some forms of benign cutaneous nevi appear to be collections of senescent melanocytes,

    suggesting that these common benign neoplasms would transform into melanomas were it

    not for the successful interdiction of this process by the senescence tumor suppressor

    mechanism. In aggregate, these data establish that senescence-promoting molecules are

    critical to the prevention of mammalian cancer with advancing age.

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915http://lwwoncology.com/Textbook/Content.aspx?aid=8762916#8762916http://lwwoncology.com/Textbook/Content.aspx?aid=8762916#8762916http://lwwoncology.com/Textbook/Content.aspx?aid=8762916#8762916http://lwwoncology.com/Textbook/Content.aspx?aid=8762917#8762917http://lwwoncology.com/Textbook/Content.aspx?aid=8762917#8762917http://lwwoncology.com/Textbook/Content.aspx?aid=8762917#8762917http://lwwoncology.com/Textbook/Content.aspx?aid=8762921#8762921http://lwwoncology.com/Textbook/Content.aspx?aid=8762921#8762921http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915http://lwwoncology.com/Textbook/Content.aspx?aid=8762905#8762905http://lwwoncology.com/Textbook/Content.aspx?aid=8762905#8762905http://lwwoncology.com/Textbook/Content.aspx?aid=8762908#8762908http://lwwoncology.com/Textbook/Content.aspx?aid=8762908#8762908http://lwwoncology.com/Textbook/Content.aspx?aid=8762908#8762908http://lwwoncology.com/Textbook/Content.aspx?aid=8762922#8762922http://lwwoncology.com/Textbook/Content.aspx?aid=8762922#8762922http://lwwoncology.com/Textbook/Content.aspx?aid=8762922#8762922http://lwwoncology.com/Textbook/Content.aspx?aid=8762923#8762923http://lwwoncology.com/Textbook/Content.aspx?aid=8762923#8762923http://lwwoncology.com/Textbook/Content.aspx?aid=8762923#8762923http://lwwoncology.com/Textbook/Content.aspx?aid=8762925#8762925http://lwwoncology.com/Textbook/Content.aspx?aid=8762925#8762925http://lwwoncology.com/Textbook/Content.aspx?aid=8762897#8762897http://lwwoncology.com/Textbook/Content.aspx?aid=8762897#8762897http://lwwoncology.com/Textbook/Content.aspx?aid=8762897#8762897http://lwwoncology.com/Textbook/Content.aspx?aid=8762926#8762926http://lwwoncology.com/Textbook/Content.aspx?aid=8762926#8762926http://lwwoncology.com/Textbook/Content.aspx?aid=8762932#8762932http://lwwoncology.com/Textbook/Content.aspx?aid=8762932#8762932http://lwwoncology.com/Textbook/Content.aspx?aid=8762932#8762932http://lwwoncology.com/Textbook/Content.aspx?aid=8762932#8762932http://lwwoncology.com/Textbook/Content.aspx?aid=8762926#8762926http://lwwoncology.com/Textbook/Content.aspx?aid=8762926#8762926http://lwwoncology.com/Textbook/Content.aspx?aid=8762897#8762897http://lwwoncology.com/Textbook/Content.aspx?aid=8762925#8762925http://lwwoncology.com/Textbook/Content.aspx?aid=8762923#8762923http://lwwoncology.com/Textbook/Content.aspx?aid=8762923#8762923http://lwwoncology.com/Textbook/Content.aspx?aid=8762922#8762922http://lwwoncology.com/Textbook/Content.aspx?aid=8762908#8762908http://lwwoncology.com/Textbook/Content.aspx?aid=8762905#8762905http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915http://lwwoncology.com/Textbook/Content.aspx?aid=8762921#8762921http://lwwoncology.com/Textbook/Content.aspx?aid=8762917#8762917http://lwwoncology.com/Textbook/Content.aspx?aid=8762917#8762917http://lwwoncology.com/Textbook/Content.aspx?aid=8762916#8762916http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    7/21

    Lastly, although the concept of tumor maintenance is becoming well established with

    regard to oncogene-activation,55a similarly important role for the persistent inactivation of

    the senescence checkpoint has been established in cancer. Several groups have established

    in genetically engineered murine models, for example, that persistent p53 inactivation is

    required for tumor maintenance.51,52,56Therefore, just as the finding that tumors in murinemodels require persistent RAS activation presaged the successful development of therapeutic

    compounds such as epidermal growth factor receptor inhibitors that target pathways

    required for tumor maintenance in vivo,similarly, these data support the notion that

    reactivation of senescence-promoting mechanisms such as p53 could be of therapeutic

    benefit in some cancers. In fact, it is likely that certain chemotherapeutics exert their

    therapeutic effects through the promotion of senescence by activating p53 and related

    senescence-inducing pathways.57

    Dysfunction of self-renewing somatic stem cell compartments has also been suggested to

    play a role in organismal aging.25,58In this model, the activation of p16INK4a

    and p53 inresponse to cellular stresses including telomere dysfunction causes a decline in tissue-

    regenerative capacity (Fig 4.3). This model is supported by murine studies5963and makes

    several predictions relevant to human disease. For example, this hypothesis suggests that

    heritable differences in regulation of the senescence-promoting machinery should alter

    individual susceptibility to human age-associated diseases, a concept that has been

    supported by a plethora of recent genomewide association studies.64With regard to

    oncology, this model predicts that some agents and ionizing radiation used to treat cancer,

    for example, by inducing DNA damage, also can potentially induce senescence of important

    self-renewing cells of nonmalignant tissues such as the bone marrow. Studies in irradiated or

    chemotherapy-treated mice support such a role of senescence in the long-term

    hematopoietic toxicities of these therapeutic approaches.65,66Likewise, somatic attrition of

    regenerative self-renewing cells as a result of senescence activation may place an increased

    replicative demand on the remaining functional cells of a given tissue, which may increase

    the rate of telomere dysfunction and speed transformation in other stem cells of a tissue in a

    cell nonautonomous manner (see later discussion).

    In aggregate, these genetic and in vivodata support the view that senescence prevents

    cancer in the intact organism on a near-daily basis and that reactivation of this mechanism in

    fully established cancers can effect dramatically beneficial responses, but also has the

    potential to produce long-term toxicity in nonmalignant tissues. It stands to reason that an

    improved understanding of the molecular basis of senescence could lead to therapeutic

    approaches designed to beneficially reawaken this potent tumor suppressor mechanism.

    The INK4a/ARF/INK4bSenescence-Promoting Locus

    Senescence is intimately associated with activation of the INK4a/ARFlocus (also known

    as CDKN2a). This locus possesses an unusual gene structure that dually encodes p16INK4aand

    ARF (or p14ARFin humans and p19ARFin mice) in nonoverlapping open reading frames

    (Fig.4.2). The locus also harbors the neighboring CDKN2bgene, which encodes p15INK4b, a

    protein highly related to p16INK4athat also activates Rb, which is located a short physical

    distance (10 kb) from the first exon of ARF. In addition to the links of p15INK4b/p16INK4aand

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762933#8762933http://lwwoncology.com/Textbook/Content.aspx?aid=8762933#8762933http://lwwoncology.com/Textbook/Content.aspx?aid=8762933#8762933http://lwwoncology.com/Textbook/Content.aspx?aid=8762929#8762929http://lwwoncology.com/Textbook/Content.aspx?aid=8762929#8762929http://lwwoncology.com/Textbook/Content.aspx?aid=8762930#8762930http://lwwoncology.com/Textbook/Content.aspx?aid=8762930#8762930http://lwwoncology.com/Textbook/Content.aspx?aid=8762934#8762934http://lwwoncology.com/Textbook/Content.aspx?aid=8762934#8762934http://lwwoncology.com/Textbook/Content.aspx?aid=8762935#8762935http://lwwoncology.com/Textbook/Content.aspx?aid=8762935#8762935http://lwwoncology.com/Textbook/Content.aspx?aid=8762935#8762935http://lwwoncology.com/Textbook/Content.aspx?aid=8762903#8762903http://lwwoncology.com/Textbook/Content.aspx?aid=8762903#8762903http://lwwoncology.com/Textbook/Content.aspx?aid=8762936#8762936http://lwwoncology.com/Textbook/Content.aspx?aid=8762936#8762936http://lwwoncology.com/Textbook/Content.aspx?aid=8762936#8762936http://lwwoncology.com/Textbook/Content.aspx?aid=8762937#8762937http://lwwoncology.com/Textbook/Content.aspx?aid=8762937#8762937http://lwwoncology.com/Textbook/Content.aspx?aid=8762941#8762941http://lwwoncology.com/Textbook/Content.aspx?aid=8762941#8762941http://lwwoncology.com/Textbook/Content.aspx?aid=8762941#8762941http://lwwoncology.com/Textbook/Content.aspx?aid=8762942#8762942http://lwwoncology.com/Textbook/Content.aspx?aid=8762942#8762942http://lwwoncology.com/Textbook/Content.aspx?aid=8762942#8762942http://lwwoncology.com/Textbook/Content.aspx?aid=8762943#8762943http://lwwoncology.com/Textbook/Content.aspx?aid=8762943#8762943http://lwwoncology.com/Textbook/Content.aspx?aid=8762944#8762944http://lwwoncology.com/Textbook/Content.aspx?aid=8762944#8762944http://lwwoncology.com/Textbook/Content.aspx?aid=8762944#8762944http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762838%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762838%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762944#8762944http://lwwoncology.com/Textbook/Content.aspx?aid=8762943#8762943http://lwwoncology.com/Textbook/Content.aspx?aid=8762942#8762942http://lwwoncology.com/Textbook/Content.aspx?aid=8762941#8762941http://lwwoncology.com/Textbook/Content.aspx?aid=8762937#8762937http://lwwoncology.com/Textbook/Content.aspx?aid=8762937#8762937http://lwwoncology.com/Textbook/Content.aspx?aid=8762936#8762936http://lwwoncology.com/Textbook/Content.aspx?aid=8762903#8762903http://lwwoncology.com/Textbook/Content.aspx?aid=8762935#8762935http://lwwoncology.com/Textbook/Content.aspx?aid=8762934#8762934http://lwwoncology.com/Textbook/Content.aspx?aid=8762930#8762930http://lwwoncology.com/Textbook/Content.aspx?aid=8762929#8762929http://lwwoncology.com/Textbook/Content.aspx?aid=8762933#8762933
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    8/21

    ARF to Rb and p53 pathways, respectively, data showing that these proteins play prominent

    roles in the prevention of human cancer are strong. Activation of the locus in response to

    stimuli, which may be both independent and dependent on telomere dysfunction, is thought

    to promote tumor suppression through induction of senescence.

    As the INK4a/ARF/INK4blocus at chromosome 9p21 is the most frequent site of single copy

    or homozygous deletion in human cancers,67,68extensive analysis of this cytogenetic region

    has been performed. As somatic deletions in cancer frequently abrogate expression of all

    three INK4a/ARF/INK4bproteins (p15INK4b, p16INK4a, and ARF), debate has focused on which

    member or members of the locus represents the principal tumor suppressor activity located

    at human chromosome 9p21. A substantial body of human and murine data has now

    unequivocally shown that all three proteins are human tumor suppressors.37For example,

    p15INK4bappears mainly important in the suppression of hematopoietic malignancies, whereas

    p16INK4aand ARF appear to play more general anticancer roles in several tumor types.

    Elegant murine studies69have further shown that these tumor suppressor genes can playback-up roles to each other, suggesting that combined inactivation of the locus is more

    oncogenic than deletion of any single member. Therefore, although the human and murine

    genetic data considered as a whole establish that the INK4a/ARF/INK4blocus encodes three

    major human tumor suppressor proteins, their relative and combinatorial importance in a

    particular tumor type is a subject of ongoing study.

    Crisis, Telomerase Reactivation, and Alternative Lengthening of Telomeres

    Under circumstances of extended cell divisions beyond the Hayflick limit with inactivation of

    the p16INK4aand p53 pathway, progressive telomere erosion ultimately leads to loss of

    telomere capping function, resulting in increasing chromosomal instability. This leads to

    progressive loss of cell viability and proliferative capacity across the cell population,

    ultimately resulting in cellular crisis. The cellular phenotypes of massive cell death and

    growth arrest are likely by-products of DNA damage checkpoint responses and rampant

    chromosomal instability with associated loss of essential genetic material. Emergence from

    crisis is a rare event in human cell culture and requires restoration of telomere function

    either by up-regulation of telomerase activity or activation of the alternative lengthening of

    telomeres (ALT) mechanism.70The restoration of functional telomeres serves to quell DNA

    damage signaling and high levels of chromosomal instability, thereby enhancing the viability

    of cells with procancer genotypes. Finally, the extent to which normal tissues experience

    telomere-associated Hayflick and crisis transitions continues to be an area of ongoing

    investigation. Nevertheless, although clear evidence of the presence of these events is still

    lacking, strong support is mounting for telomere-based crisis, particularly during early stages

    of neoplastic development.

    Transcriptional up-regulation of the TERTgene seems to be a key rate-limiting step in

    telomerase reactivation, whereas the telomerase-independent ALT pathway appears to be

    executed via a poorly understood process involving activation of the homologous

    recombination pathway.71,72The analysis of pathways regulating TERTgene transcription

    has forged links to well-known oncoproteins and tumor suppressors including Myc, Mad, and

    Menin, among others, demonstrating the capacity of these proteins to engage the TERTgene

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762945#8762945http://lwwoncology.com/Textbook/Content.aspx?aid=8762945#8762945http://lwwoncology.com/Textbook/Content.aspx?aid=8762946#8762946http://lwwoncology.com/Textbook/Content.aspx?aid=8762946#8762946http://lwwoncology.com/Textbook/Content.aspx?aid=8762946#8762946http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915http://lwwoncology.com/Textbook/Content.aspx?aid=8762947#8762947http://lwwoncology.com/Textbook/Content.aspx?aid=8762947#8762947http://lwwoncology.com/Textbook/Content.aspx?aid=8762947#8762947http://lwwoncology.com/Textbook/Content.aspx?aid=8762948#8762948http://lwwoncology.com/Textbook/Content.aspx?aid=8762948#8762948http://lwwoncology.com/Textbook/Content.aspx?aid=8762948#8762948http://lwwoncology.com/Textbook/Content.aspx?aid=8762949#8762949http://lwwoncology.com/Textbook/Content.aspx?aid=8762949#8762949http://lwwoncology.com/Textbook/Content.aspx?aid=8762950#8762950http://lwwoncology.com/Textbook/Content.aspx?aid=8762950#8762950http://lwwoncology.com/Textbook/Content.aspx?aid=8762950#8762950http://lwwoncology.com/Textbook/Content.aspx?aid=8762950#8762950http://lwwoncology.com/Textbook/Content.aspx?aid=8762949#8762949http://lwwoncology.com/Textbook/Content.aspx?aid=8762948#8762948http://lwwoncology.com/Textbook/Content.aspx?aid=8762947#8762947http://lwwoncology.com/Textbook/Content.aspx?aid=8762915#8762915http://lwwoncology.com/Textbook/Content.aspx?aid=8762946#8762946http://lwwoncology.com/Textbook/Content.aspx?aid=8762945#8762945
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    9/21

    promoter directly.7375In contrast, the enigmatic ALT process has been variously

    associated with p53 deficiency and with tumors of mesenchymal origin.76

    Studies in yeast have also shown that ALT is enhanced in mismatch repair-deficient cells,

    owing to increased homologous recombination between chromosomes. The rare use of ALT

    by epithelial-derived tumors, coupled with functional comparisons of telomerase versus ALT-

    mediated telomere maintenance, has shown that ALT may not be as biologically robust in

    advancing malignancy, a finding that diminishes the theoretical concern that ALT may

    provide a robust resistance mechanism to antitelomerase therapy in advanced

    malignancy.77The idea that ALT may be a less effective telomere maintenance mechanism

    derives additional support from studies in human cell culture and the mouse revealing that

    telomerase per se is needed for full malignant transformation, including metastatic

    potential.78The fundamental mechanistic differences between ALT and telomerase

    reactivation in telomere maintenance may provide an explanation for the report of more

    favorable clinical outcomes for ALT-positive compared with telomerase-positiveglioblastomas,79although analysis of 71 human osteosarcoma cases failed to show a more

    favorable clinical outcome for the ALT-positive subset.80However, it should be noted that, in

    the latter, the absence of any telomere maintenance mechanism was more associated with

    improved survival than stage or response to chemotherapy, further emphasizing the general

    importance of telomere maintenance in cancer.

    Telomere Maintenance and Cancer

    Robust telomerase activity is observed in more than 80% of all human cancers,81a profile

    consistent with its role in promoting malignant progression. However, another side to the

    telomerase-cancer connection has emerged from mouse models and correlative data in

    staged human tumors. These data have indicated that a lack of telomerase and associated

    telomere attrition during the early stages of neoplastic growth provides a potent mutator

    mechanism that enables would-be cancer cells to achieve the high threshold of cancer-

    promoting changes required to traverse the benign to malignant transition.

    Indeed, telomeres of human cancer cells are often significantly shorter than their normal

    tissue counterparts, suggesting that telomere attrition has occurred during the life history of

    these cancer cells, apparently at very early phases of the transformation process when

    telomerase activity is low. The subsequent reactivation of telomerase restores telomere

    function, albeit at a shorter set length. Thus, although reactivation of telomerase is critical to

    the emergence of immortal human cells, this preceding and transient period of telomere

    shortening and dysfunction promotes the carcinogenic process through the generation of

    chromosomal rearrangements. These chromosomal rearrangements are brought about

    through breakage-fusion-bridge (BFB) cycles (Fig.4.4). A DSB created by these BFB cycles is

    now known to provide a nidus for amplification and/or deletion at the site of breakage for the

    resulting daughter cells. The broken chromosome may become fused to another

    chromosome, generating a second dicentric chromosome and perpetuating the BFB cycle.

    The accumulation of wholesale genetic changes via aneuploidy, nonreciprocal translocations,

    amplifications, and deletions by the BFB cycles coupled with the reactivation of telomerase

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762951#8762951http://lwwoncology.com/Textbook/Content.aspx?aid=8762951#8762951http://lwwoncology.com/Textbook/Content.aspx?aid=8762953#8762953http://lwwoncology.com/Textbook/Content.aspx?aid=8762953#8762953http://lwwoncology.com/Textbook/Content.aspx?aid=8762953#8762953http://lwwoncology.com/Textbook/Content.aspx?aid=8762954#8762954http://lwwoncology.com/Textbook/Content.aspx?aid=8762954#8762954http://lwwoncology.com/Textbook/Content.aspx?aid=8762954#8762954http://lwwoncology.com/Textbook/Content.aspx?aid=8762955#8762955http://lwwoncology.com/Textbook/Content.aspx?aid=8762955#8762955http://lwwoncology.com/Textbook/Content.aspx?aid=8762955#8762955http://lwwoncology.com/Textbook/Content.aspx?aid=8762956#8762956http://lwwoncology.com/Textbook/Content.aspx?aid=8762956#8762956http://lwwoncology.com/Textbook/Content.aspx?aid=8762956#8762956http://lwwoncology.com/Textbook/Content.aspx?aid=8762957#8762957http://lwwoncology.com/Textbook/Content.aspx?aid=8762957#8762957http://lwwoncology.com/Textbook/Content.aspx?aid=8762957#8762957http://lwwoncology.com/Textbook/Content.aspx?aid=8762958#8762958http://lwwoncology.com/Textbook/Content.aspx?aid=8762958#8762958http://lwwoncology.com/Textbook/Content.aspx?aid=8762958#8762958http://lwwoncology.com/Textbook/Content.aspx?aid=8762959#8762959http://lwwoncology.com/Textbook/Content.aspx?aid=8762959#8762959http://lwwoncology.com/Textbook/Content.aspx?aid=8762959#8762959http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762857%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762857%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762857%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762857%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762959#8762959http://lwwoncology.com/Textbook/Content.aspx?aid=8762958#8762958http://lwwoncology.com/Textbook/Content.aspx?aid=8762957#8762957http://lwwoncology.com/Textbook/Content.aspx?aid=8762956#8762956http://lwwoncology.com/Textbook/Content.aspx?aid=8762955#8762955http://lwwoncology.com/Textbook/Content.aspx?aid=8762954#8762954http://lwwoncology.com/Textbook/Content.aspx?aid=8762953#8762953http://lwwoncology.com/Textbook/Content.aspx?aid=8762951#8762951http://lwwoncology.com/Textbook/Content.aspx?aid=8762951#8762951
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    10/21

    enables rare cells incurring a threshold number of carcinogenic changes needed to initiate

    the transformation process.

    Figure 4.3. Senescence and aging.

    Activation of p53- and/or p16INK4a-mediated senescence pathways in stem cell compartments

    in response to DNA damage, telomere dysfunction, or other unknown stimuli leads to

    attrition of tissue-specific stem cells (e.g., hematopoietic stem cell and pancreatic -cells)

    with attendant compromise of organ function and aging.

    Figure 4.4. Dysfunctional telomere-induced genomic instability model of epithelial

    carcinogenesis.

    Continuous epithelial turnover during aging coupled with somatic mutations inactivating

    checkpoint responses is thought to lead to critical telomere erosion, resulting in telomere

    uncapping and the initiation of breakage-fusion-bridge (BFB) cycles. The double-strand

    breaks created by the BFB cycles are nidi for amplifications and deletions for the resulting

    daughter cells. The broken chromosome may become fused to another chromosome,

    generating a second dicentric chromosome and perpetuating the BFB cycle. This facilitation

    of the accumulation of genetic changes (via aneuploidy, nonreciprocal translocations,

    amplifications, and deletions) by the BFB cycles coupled with the reactivation of telomerase

    enables cells to emerge from crisis and proceed to malignancy.

    Although at first glance the cancer-promoting effects of telomere-based crisis seem to

    counter the established role of telomerase activation in cancer progression, this mechanism

    is less paradoxical if one considers that many early-stage cancers deactivate pathways

    essential for telomere checkpoint responses, thus increasing the survival and proliferation of

    cells experiencing increasing chromosomal instability.75,82This hypothesis of episodic

    instability derives additional support from genetic studies in the mouse showing that

    http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762856%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762856%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762857%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762857%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762857%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762953#8762953http://lwwoncology.com/Textbook/Content.aspx?aid=8762953#8762953http://lwwoncology.com/Textbook/Content.aspx?aid=8762960#8762960http://lwwoncology.com/Textbook/Content.aspx?aid=8762960#8762960http://lwwoncology.com/Textbook/Content.aspx?aid=8762960#8762960http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762857%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762856%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762857%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762856%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762960#8762960http://lwwoncology.com/Textbook/Content.aspx?aid=8762953#8762953http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762857%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762857%27);http://windowreference%28%27reference%27%2C%27/Popup/Index.aspx?aID=8762856%27);
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    11/21

    telomere-based crisis coupled with loss of the p53-dependent DNA damage response can act

    cooperatively to effect malignant transformation. In humans, the accumulation of oncogenic

    lesions during normal aging or accelerated accumulation of DNA damage (e.g.,

    environmental carcinogen exposure or oxidative damage) may deactivate the telomere

    checkpoint response, accelerate telomere attrition, and drive the affected premalignant cellsinto crisis. It is the rare transformed cell that emerges from this process, often with

    reactivated telomerase. Thus, telomeric shortening can be viewed as a barrier to cancer

    development in the presence of intact checkpoint response and as a facilitator for numerous

    genetic changes necessary for the emergence of nascent cancer cells in the absence of the

    checkpoint response pathways.

    It has also been suggested that telomere dysfunction can be oncogenic in a cell

    nonautonomous process.83Murine data in the hematopoietic system suggest that telomere

    dysfunction leads to stem cell dysfunction.84,85Therefore, telomere dysfunction could

    induce premature loss of stem cells (as described in Fig.4.3), which might induce acompensatory hyperproliferation of remaining functional stem cells. This increased

    proliferative drive might facilitate mutagenesis in the remaining functional self-renewing

    cells, and in turn select for clones with damaged genomes, in particular those harboring

    defects in the senesce-promoting machinery.

    Several recent lines of evidence have suggested that the oncogenic effects of telomere

    dysfunction are an important determinant of susceptibility to human cancer. For example,

    several human kindreds have been identified with congenital telomerase deficiency

    syndromes due to inactivating mutations of TERTor other members of the shelterin

    complex.4Such patients exhibit age-associated pathologies such as bone marrow failure andpulmonary fibrosis, but also appear to be at increased risk for several cancers including acute

    myelogenous leukemia and cutaneous carcinomas.4,83,8688Likewise, human genomewide

    association studies of large human cohorts have pointed to sequence variants in the

    chromosome 5p15.33 locus as a susceptibility locus for many types of cancer, including

    tumors of the skin, lung, bladder, prostate, and cervix.89The single nucleotide

    polymorphisms associated with these cancers are near to both the CLPTM1L(cisplatin

    resistance-related protein CRRP9) gene and the TERTgene. It is unclear whether one or both

    of these genes are responsible for the association as there is limited functional biological

    validation. These human data suggest that telomere dysfunction could be oncogenic, and

    that hypomorphic alleles of TERT could contribute to human cancer susceptibility on a

    population basis.

    Telomere-Induced Chromosomal Instability

    The study of senescence and telomeres has provided some insights into the link between

    advancing age and increased cancer risk. In humans, there is a dramatic escalation in cancer

    risk between the ages of 40 and 80, resulting primarily from a marked increase in epithelial

    malignancies such as carcinomas of the breast, lung, colon, and prostate. A conventional

    view is that the cancer-prone phenotype of older humans reflects the combined effects of

    cumulative mutational load, decreased DNA repair capabilities, increased epigenetic gene

    silencing, and altered hormonal and stromal milieus. Although these factors are almost

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762961#8762961http://lwwoncology.com/Textbook/Content.aspx?aid=8762961#8762961http://lwwoncology.com/Textbook/Content.aspx?aid=8762961#8762961http://lwwoncology.com/Textbook/Content.aspx?aid=8762962#8762962http://lwwoncology.com/Textbook/Content.aspx?aid=8762962#8762962http://lwwoncology.com/Textbook/Content.aspx?aid=8762963#8762963http://lwwoncology.com/Textbook/Content.aspx?aid=8762963#8762963http://lwwoncology.com/Textbook/Content.aspx?aid=8762963#8762963http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762856%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762856%27);http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762856%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762882#8762882http://lwwoncology.com/Textbook/Content.aspx?aid=8762882#8762882http://lwwoncology.com/Textbook/Content.aspx?aid=8762882#8762882http://lwwoncology.com/Textbook/Content.aspx?aid=8762882#8762882http://lwwoncology.com/Textbook/Content.aspx?aid=8762882#8762882http://lwwoncology.com/Textbook/Content.aspx?aid=8762961#8762961http://lwwoncology.com/Textbook/Content.aspx?aid=8762961#8762961http://lwwoncology.com/Textbook/Content.aspx?aid=8762964#8762964http://lwwoncology.com/Textbook/Content.aspx?aid=8762964#8762964http://lwwoncology.com/Textbook/Content.aspx?aid=8762966#8762966http://lwwoncology.com/Textbook/Content.aspx?aid=8762966#8762966http://lwwoncology.com/Textbook/Content.aspx?aid=8762966#8762966http://lwwoncology.com/Textbook/Content.aspx?aid=8762967#8762967http://lwwoncology.com/Textbook/Content.aspx?aid=8762967#8762967http://lwwoncology.com/Textbook/Content.aspx?aid=8762967#8762967http://lwwoncology.com/Textbook/Content.aspx?aid=8762967#8762967http://lwwoncology.com/Textbook/Content.aspx?aid=8762966#8762966http://lwwoncology.com/Textbook/Content.aspx?aid=8762964#8762964http://lwwoncology.com/Textbook/Content.aspx?aid=8762964#8762964http://lwwoncology.com/Textbook/Content.aspx?aid=8762961#8762961http://lwwoncology.com/Textbook/Content.aspx?aid=8762882#8762882http://lwwoncology.com/Textbook/Content.aspx?aid=8762882#8762882http://windowreference%28%27reference%27%2C%27/Popup/index.aspx?aID=8762856%27);http://lwwoncology.com/Textbook/Content.aspx?aid=8762963#8762963http://lwwoncology.com/Textbook/Content.aspx?aid=8762962#8762962http://lwwoncology.com/Textbook/Content.aspx?aid=8762961#8762961
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    12/21

    certain to contribute to increasing cancer incidence in aged humans, it is less evident why

    such processes would spur the preferential development of epithelial cancers. Moreover,

    these mechanisms do not readily explain one of the cardinal features of adult epithelial

    carcinomasnamely, a radically altered genome typified by marked aneuploidy and complex

    nonreciprocal chromosomal translocations.

    The study of telomere dynamics in normal and neoplastic cells of the mouse has provided a

    potential explanation for the observed tumor spectrum and associated cytogenetic profiles in

    aged humans. InTerc p53compound mutant mice, the presence of telomere dysfunction

    results in a dramatic shift in the tumor spectrum toward epithelial cancers, including those of

    the lung, colon, and skin.90Moreover, in contrast to the largely normal cytogenetic profiles

    of cancers arising in mice with intact telomeres, the cancers generated in the Terc

    p53compound mutant mice had highly complex cytogenetic profiles with a striking

    resemblance to human epithelial cancer genomes.

    In attempting to assign relevance of these murine studies to humans, it is worth considering

    that the typical adult cancer, an epithelial carcinoma, derives from a compartment that has

    undergone continued renewal throughout the human lifespan. Against this backdrop of

    physiologic cell turnover, combined with the occasional pro-proliferative oncogenic mutation,

    telomere lengths would shorten in self-renewing progenitor cells of these epithelial tissues. If

    somatic mutations also neutralize Rb/p16INK4a/p53-dependent senescence checkpoints,

    continued growth beyond the Hayflick limit further drives telomere erosion and loss of the

    capping function, culminating in cellular crisis with attendant genomic instability. In this

    manner, telomere-based crisis provides the means to generate many additional mutations

    required to reach the early stages of malignant transformation. The subsequent reactivationof telomerase in transformed clones would serve to stabilize the genome to a level

    compatible with cell viability, allowing these initiated neoplasms to mature further.91It is

    unclear whether additional somatic mutations, beyond telomerase activation, would be

    needed to produce a fully malignant phenotype that includes invasive and metastatic

    potential. Thus, a transient period of explosive chromosomal instability before telomerase

    reactivation appears to be required for the stochastic acquisition of the relatively high

    number of mutations thought to be required for adult epithelial carcinogenesis. Another line

    of support is the fact that a proportion of early-stage epithelial cancers are hardwired for

    lethal metastatic progression, suggesting that many cancers acquire a full profile of genome

    change early in their life history.

    The episodic instability model of epithelial carcinogenesis fits well with current knowledge

    regarding the timing of telomerase activation and evolving genomic changes during various

    stages of human carcinoma development, particularly those of the breast, esophagus, and

    colon. Comparative genome hybridization has demonstrated that dysplastic human breast,

    esophageal, and colon lesions sustain widespread gains and losses of regions of

    chromosomes early in their development, often well before these tissues exhibit carcinoma in

    situor invasive growth.9294The ploidy changes detected by comparative genome

    hybridization appear to correlate tightly with the presence of complex chromosomal

    rearrangements, and these markers of genomic instability are evident in the stages of

    advanced dysplasia of these tissues (e.g., ductal carcinoma in situ, Barretts esophagus). As

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762968#8762968http://lwwoncology.com/Textbook/Content.aspx?aid=8762968#8762968http://lwwoncology.com/Textbook/Content.aspx?aid=8762968#8762968http://lwwoncology.com/Textbook/Content.aspx?aid=8762969#8762969http://lwwoncology.com/Textbook/Content.aspx?aid=8762969#8762969http://lwwoncology.com/Textbook/Content.aspx?aid=8762969#8762969http://lwwoncology.com/Textbook/Content.aspx?aid=8762970#8762970http://lwwoncology.com/Textbook/Content.aspx?aid=8762970#8762970http://lwwoncology.com/Textbook/Content.aspx?aid=8762972#8762972http://lwwoncology.com/Textbook/Content.aspx?aid=8762972#8762972http://lwwoncology.com/Textbook/Content.aspx?aid=8762972#8762972http://lwwoncology.com/Textbook/Content.aspx?aid=8762972#8762972http://lwwoncology.com/Textbook/Content.aspx?aid=8762970#8762970http://lwwoncology.com/Textbook/Content.aspx?aid=8762970#8762970http://lwwoncology.com/Textbook/Content.aspx?aid=8762969#8762969http://lwwoncology.com/Textbook/Content.aspx?aid=8762968#8762968
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    13/21

    these cancers progress through invasive and metastatic stages, genomic instability

    continues, apparently at a moderate rate, but further mutations would be predicted to derive

    from nontelomere-based mechanisms. Correspondingly, the measurement of telomerase

    activity in adenomatous polyps and colorectal cancers has established that telomerase

    activity is low or undetectable in small and intermediate-sized polyps, reflecting less intacttelomere function. In contrast, telomerase increases markedly in large adenomas and

    colorectal carcinomas, reflecting stabilization of telomere function.95Therefore, it appears

    that widespread and severe chromosomal instability is present early on during human

    tumorigenesis at a time when telomerase activity is low.

    Additional support for this episodic instability model derives from the documentation of

    anaphase bridging (a correlate of telomere-based crisis) in evolving human colorectal

    cancers and in genomically unstable pancreatic cancers.96,97This suggests that the DSB-

    induced conditions (including but not limited to telomere dysfunction), coupled with

    mutations that allow survival in the face of a DSB, could provide amplification/deletionmechanisms across the genome. Biologic selection forces would in turn lead to the

    emergence of clones with the amplifications and deletions that target cancer-relevant loci.

    Studies in the telomerase mutant mouse have begun to provide mechanistic insight into how

    BFB leads to cancer-relevant changes. In particular, telomerase-p53 compound mutant mice

    with telomere dysfunction have increased end-to-end fusions, and the ensuing BFB process

    is associated with chromosomal regional gains and losses that appear linked to nonreciprocal

    translocations.55,75

    In future human studies, it will be important to document telomere attrition in renewing

    epithelial stem cells and to perform a simultaneous comparison of telomere status, telo-merase activity, and chromosomal instability in the same tumor samples, particularly during

    the earliest stages of human epithelial cell transformation. Defining the temporal point at

    which telomerase is reactivated in the genesis and progression of the different cancers may

    also lead to the development of biomarkers for diagnosis, prognosis, and outcomes

    prediction. Such studies are needed to more firmly establish a causal link between telomere

    dysfunction and early chromosomal instability in human neoplasms.

    Telomere Dynamics, Inflammatory Diseases, and Cancer

    The telomere dysfunction-induced genomic instability model also suggests some

    unanticipated opportunities for the therapies of other human diseases. For example, this

    model provides a potential explanation for the high cancer incidence associated with diseases

    characterized by chronic cell destruction and renewal as well as inflammation. One of the

    most notable examples of this tight link is the high incidence of hepatocellular carcinoma in

    late-stage cirrhotic livers. Cirrhosis is the phenotypic end point of prolonged cycles of

    hepatocyte destruction and regeneration, and cirrhotic livers show a documented reduction

    in telomere length over time. Humans with congenital telomerase deficiency may be

    predisposed to fibrotic liver disease, including cirrhosis.87Mouse models involving the

    telomerase-deficient mouse have shown that critical reductions in telomere length and

    function can accelerate the development of cirrhosis and hepatocellular carcinoma in chronic

    liver injury experiments.98100Another example of a telomere-based pathogenic

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762973#8762973http://lwwoncology.com/Textbook/Content.aspx?aid=8762973#8762973http://lwwoncology.com/Textbook/Content.aspx?aid=8762973#8762973http://lwwoncology.com/Textbook/Content.aspx?aid=8762974#8762974http://lwwoncology.com/Textbook/Content.aspx?aid=8762974#8762974http://lwwoncology.com/Textbook/Content.aspx?aid=8762975#8762975http://lwwoncology.com/Textbook/Content.aspx?aid=8762975#8762975http://lwwoncology.com/Textbook/Content.aspx?aid=8762975#8762975http://lwwoncology.com/Textbook/Content.aspx?aid=8762933#8762933http://lwwoncology.com/Textbook/Content.aspx?aid=8762933#8762933http://lwwoncology.com/Textbook/Content.aspx?aid=8762953#8762953http://lwwoncology.com/Textbook/Content.aspx?aid=8762953#8762953http://lwwoncology.com/Textbook/Content.aspx?aid=8762953#8762953http://lwwoncology.com/Textbook/Content.aspx?aid=8762965#8762965http://lwwoncology.com/Textbook/Content.aspx?aid=8762965#8762965http://lwwoncology.com/Textbook/Content.aspx?aid=8762965#8762965http://lwwoncology.com/Textbook/Content.aspx?aid=8762976#8762976http://lwwoncology.com/Textbook/Content.aspx?aid=8762976#8762976http://lwwoncology.com/Textbook/Content.aspx?aid=8762976#8762976http://lwwoncology.com/Textbook/Content.aspx?aid=8762978#8762978http://lwwoncology.com/Textbook/Content.aspx?aid=8762978#8762978http://lwwoncology.com/Textbook/Content.aspx?aid=8762976#8762976http://lwwoncology.com/Textbook/Content.aspx?aid=8762976#8762976http://lwwoncology.com/Textbook/Content.aspx?aid=8762965#8762965http://lwwoncology.com/Textbook/Content.aspx?aid=8762953#8762953http://lwwoncology.com/Textbook/Content.aspx?aid=8762933#8762933http://lwwoncology.com/Textbook/Content.aspx?aid=8762975#8762975http://lwwoncology.com/Textbook/Content.aspx?aid=8762974#8762974http://lwwoncology.com/Textbook/Content.aspx?aid=8762973#8762973
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    14/21

    relationship between chronic tissue turnover, telomere-based crisis, and increased cancer

    risk is ulcerative colitis, a condition characterized by rapid cell turnover and oxidative injury

    to the intestines, and a high incidence of intestinal dysplasia or cancer.97In addition to the

    progressive telomere attrition resulting from the cell turnover, accelerated telomere attrition

    might occur via increased oxidative stress and from the altered inflammatorymicroenvironment milieu. Together, such observations suggest the intriguing possibility that

    early somatic reconstitution of telomerase could attenuate telomere attrition and

    paradoxically reduce the occurrence of cancers in these high-turnover disease states, a

    theory that requires additional preclinical studies. In addition, serial analyses of telomere

    length from these tissues may provide prognostic information regarding the rising risk of

    cancer development. Thus, progress in our understanding of telomere biology has

    mechanistically connected diverse fields in medicine involving chronic inflammatory diseases,

    degenerative diseases, geriatrics, and oncology.

    Telomerase and Telomere Maintenance As Therapeutic TargetsSome evidence supports the view that telomerase-mediated telomere maintenance

    represents a near-universal therapeutic target for cancer. Indeed, cell culturebased studies

    of human cancer cells have established that inhibition of telomerase culminates in cell death

    after extended cell divisions. The past few years have witnessed intense efforts to design

    therapeutic strategies capable of targeting telomere structure and the telomerase

    holoenzyme function.14,101,102Unfortunately, most of these compounds and agents are

    still in preclinical and early clinical development and thus their safety and efficacy profiles in

    human patients are not fully known.

    Presently, the only clinically advanced telomerase-related cancer treatment strategy is

    immunotherapy, targeting immune recognition and the destruction of cells that express telo-

    merase. Immune responses, specifically cytotoxic T-cell responses, have been generated

    against peptide sequences of the hTERT protein, and it has been demonstrated that these

    cytotoxic T cells are capable of selectively lysing target cells that express TERT peptides

    presented on the cell surface in the context of major histocompatibility complex class I

    molecules. There have been several promising completed phase 1/2 trials using peptides

    from telomerase as vaccines.103,104A large randomized phase 3 trial comparing

    gemcitabine alone versus gemcitabine with a telomerase peptide vaccine (GV1001) showed

    no difference in survival benefit in the first 360 enrolled patients, and the trial was stopped.

    A second large 1,110 pancreatic cancer patient trial comparing gemcitabine/capecitabine

    combination therapy with concurrent and sequential gemcitabine/capecitabine therapy with

    GV1001 is still ongoing. Lastly, other TERT-based immune approaches, such as infusion of

    patients primed antigen-presenting dendritic cellsex vivowith TERT mRNA, are also

    currently in early clinical trials.105

    As for the ongoing design of rational clinical trials of telomere-based therapeutics, such

    efforts will be informed by the considerable body of knowledge accumulated in telomere

    biology. Experience with the telomerase mutant mouse model and human cell culture

    systems should serve to guide the design of human clinical trials. These studies suggest that

    inhibitors of telomerase activity might be expected to exhibit a long lag time and might

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762975#8762975http://lwwoncology.com/Textbook/Content.aspx?aid=8762975#8762975http://lwwoncology.com/Textbook/Content.aspx?aid=8762975#8762975http://lwwoncology.com/Textbook/Content.aspx?aid=8762892#8762892http://lwwoncology.com/Textbook/Content.aspx?aid=8762892#8762892http://lwwoncology.com/Textbook/Content.aspx?aid=8762979#8762979http://lwwoncology.com/Textbook/Content.aspx?aid=8762979#8762979http://lwwoncology.com/Textbook/Content.aspx?aid=8762980#8762980http://lwwoncology.com/Textbook/Content.aspx?aid=8762980#8762980http://lwwoncology.com/Textbook/Content.aspx?aid=8762980#8762980http://lwwoncology.com/Textbook/Content.aspx?aid=8762981#8762981http://lwwoncology.com/Textbook/Content.aspx?aid=8762981#8762981http://lwwoncology.com/Textbook/Content.aspx?aid=8762982#8762982http://lwwoncology.com/Textbook/Content.aspx?aid=8762982#8762982http://lwwoncology.com/Textbook/Content.aspx?aid=8762982#8762982http://lwwoncology.com/Textbook/Content.aspx?aid=8762983#8762983http://lwwoncology.com/Textbook/Content.aspx?aid=8762983#8762983http://lwwoncology.com/Textbook/Content.aspx?aid=8762983#8762983http://lwwoncology.com/Textbook/Content.aspx?aid=8762983#8762983http://lwwoncology.com/Textbook/Content.aspx?aid=8762982#8762982http://lwwoncology.com/Textbook/Content.aspx?aid=8762981#8762981http://lwwoncology.com/Textbook/Content.aspx?aid=8762980#8762980http://lwwoncology.com/Textbook/Content.aspx?aid=8762979#8762979http://lwwoncology.com/Textbook/Content.aspx?aid=8762892#8762892http://lwwoncology.com/Textbook/Content.aspx?aid=8762975#8762975
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    15/21

    promote malignancy in some circumstances, but also may be particularly useful in the

    setting of minimal residual disease after the administration of standard chemotherapeutic

    agents and surgery. In addition, pharmacodynamic assays capable of assessing inhibition of

    telomerase activity in individual patients are needed. Moreover, given that the activation of

    senescence-promoting mechanisms such as p53 and p16

    INK4a

    has been associated with aging-like pathologies in several tissues,5963some caution is warranted regarding the toxicity

    resulting from the induction of premature senescence in nondiseased tissues. This potential

    is underlined by evidence of germ cell defects, defects in proliferative homeostasis of certain

    tissues, and an increased rate of spontaneous malignancy in mice with telomere dysfunction,

    suggesting that clinical trials of such agents will need to be actively monitoring patients for

    these sequelae.

    Furthermore, it seems prudent that the genetic profile of tumors enlisted into clinical trials

    should be determined to assess the integrity of p53. This caution relates to mouse models

    showing that the combination of p53 deficiency and telomere dysfunction drives greatergenomic instability and thus potential for emergence of therapeutic resistance. In contrast,

    when p53 responses are intact, critical telomere shortening should induce p53-dependent

    senescence and apoptosis. The final answers to these safety questions reside in the analyses

    of current and future clinical trials with humans.

    Conversely, the telomerase-deficient mouse model has also informed that cells and animals

    with telomere dysfunction are more sensitive to ionizing radiation and DNA DSB

    chemotherapeutic agents; thus, telomerase activity inhibitors may be more effective when

    paired with radiation or certain classes of chemotherapy that produce DSBs, as they might

    produce synergistic cytogenetic catastrophe. Again, however, particular care is warrantedhere as the combination of increased DNA damage with reduced capacity for normal repair

    may also produce marked increases in the toxicity of chemoradiotherapy.

    Recent years have witnessed significant progress in the telomere biology field that is now

    maturing into new opportunities for improved diagnostics and novel therapeutic applications

    in human diseases, including cancer. Discoveries in telomere biology, rewarded with the

    2009 Nobel Prize, have provided new mechanistic insights into the pathogenesis of human

    cancer and of inherited and acquired degenerative disorders. The role of telomere

    dysfunction driving episodic genomic instability in epithelial cancersfirst seen in the

    telomerase-deficient mousehas now been substantiated in the study of several humancancer types, with further support from genomewide association studies and kindreds with

    congenital telomerase deficiency. The pivotal role of telomere attrition in the pathogenesis of

    cancer and tissue aging provides potential avenues for the development of cancer risk

    biomarkers, diagnostics, and rationally designed therapeutics.

    References

    1. Mori H, Colman SM, Xiao Z, et al. Chromosome translocations and covert leukemicclones are generated during normal fetal development. Proc Natl Acad Sci U S

    A.2002;4:4.

    http://lwwoncology.com/Textbook/Content.aspx?aid=8762937#8762937http://lwwoncology.com/Textbook/Content.aspx?aid=8762937#8762937http://lwwoncology.com/Textbook/Content.aspx?aid=8762937#8762937http://lwwoncology.com/Textbook/Content.aspx?aid=8762941#8762941http://lwwoncology.com/Textbook/Content.aspx?aid=8762941#8762941http://lwwoncology.com/Textbook/Content.aspx?aid=8762941#8762941http://lwwoncology.com/Textbook/Content.aspx?aid=8762937#8762937http://lwwoncology.com/Textbook/Content.aspx?aid=8762937#8762937
  • 7/22/2019 Chapter 4 - Telomeres, Telomerase, And Cancer

    16/21

    2. Liu Y, Hernandez AM, Shibata D, Cortopassi GA. BCL2 translocation frequency riseswith age in humans. Proc Natl Acad Sci U S A.1994;91:8910.[PMID: 8090743]

    3. Sahin E, Depinho RA. Linking functional decline of telomeres, mitochondria and stemcells during ageing. Nature.2010;464:520.[PMID: 20336134]

    4.

    Calado RT, Young NS. Telomere diseases. N Engl J Med.2009;361:2353.[PMID:20007561]

    5. de Lange T. Protection of mammalian telomeres. Oncogene.2002;21:532.[PMID:11850778]

    6. OSullivan RJ, Karlseder J. Telomeres: protecting chromosomes against genomeinstability. Nature Rev.2010;11:171.[PMID: 21438178]

    7. de Lange T. How telomeres solve the end-protectionproblem. Science.2009;326:948.[PMID: 19965504]

    8. de Lange T. Shelterin: the protein complex that shapes and safeguards humantelomeres. Genes Dev.2005;19:2100.[PMID: 16166375]

    9. Szilard RK, Durocher D. Telomere protection: an act of God. CurrBiol.2006;16:R544.[PMID: 16860732]

    10. Zhu XD, Kuster B, Mann M, Petrini JH, de Lange T. Cell-cycle-regulated associationof RAD50/MRE11/NBS1 with TRF2 and human telomeres. Nat

    Genet.2000;25:347.[PMID: 10888888]

    11. Goytisolo FA, Blasco MA. Many ways to telomere dysfunction: in vivo studies usingmouse models.Oncogene. 2002;21:584.[PMID: 11850783]

    12. Takai H, Smogorzewska A, de Lange T. DNA damage foci at dysfunctionaltelomeres. Curr Biol.2003;13:1549.[PMID: 12956959]

    13. Deng Y, Chan SS, Chang S. Telomere dysfunction and tumour suppression: thesenescence connection. Nat Rev Cancer.2008;8:450.[PMID: 18500246]

    14. Cong YS, Wright WE, Shay JW. Human telomerase and its regulation. Microbiol MolBiol Rev.2002;66:407.[PMID: 12208997]

    15. Rufer N, Brummendorf TH, Kolvraa S, et al. Telomere fluorescence measurements ingranulocytes and T lymphocyte subsets point to a high turnover of hematopoietic

    stem cells and memory T cells in early childhood.J Exp Med.1999;190:157.[PMID:

    10432279]

    16. Valdes AM, Andrew T, Gardner JP, et al. Obesity, cigarette smoking, and telomerelength in women. Lancet.2005;366:662.[PMID: 16112303]

    17. Vaziri H, Schachter F, Uchida I, et al. Loss of telomeric DNA during aging of normaland trisomy 21 human lymphocytes.Am J Hum Genet.1993;52:661.[PMID:

    8460632]

    18. Baerlocher GM, Sloand EM, Young NS, Lansdorp PM. Telomere length in paroxysmalnocturnal hemoglobinuria correlates with clone size. Exp

    Hematol.2007;35:1777.[PMID: 17697745]

    19. Artandi SE, DePinho RA. Telomeres and telomerase incancer. Carcinogenesis.2010;31:9.[PMID: 19887512]

    20. Masutomi K, Yu EY, Khurts S, et al. Telomerase maintains telomere structure innormal human cells. Cell.2003;114:241.[PMID: 12887925]

    http://www.ncbi.nlm.nih.gov/pubmed/8090743?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/8090743?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/8090743?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20336134?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20336134?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20336134?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20007561?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20007561?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20007561?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/20007561?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11850778?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11850778?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11850778?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11850778?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21438178?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21438178?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/21438178?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19965504?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19965504?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/19965504?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16166375?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16166375?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16166375?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16860732?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16860732?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16860732?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/10888888?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/10888888?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/10888888?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11850783?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11850783?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/11850783?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12956959?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12956959?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12956959?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/18500246?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/18500246?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/18500246?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12208997?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12208997?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/12208997?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/10432279?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/10432279?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/10432279?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/10432279?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16112303?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16112303?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/16112303?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubmed/8460632?dopt=Abstracthttp://www.ncbi.nlm.nih.gov/pubme