Targeting the Limitless Replicative Potential of Cancer: The Telomerase/Telomere Pathway

Page created by Bruce Newman
 
CONTINUE READING
Targeting the Limitless Replicative Potential of Cancer: The Telomerase/Telomere Pathway
Molecular Pathways

Targeting the Limitless Replicative Potential of Cancer:
The Telomerase/Telomere Pathway
Lloyd Kelland

     Abstract          The maintenance of telomeric DNA underlies the ability of tumors to possess unlimited replicative
                       potential, one of the hallmarks of cancer. Telomere length and structure are maintained by the
                       reverse transcriptase telomerase and a multiprotein telomere complex termed shelterin. Telomer-
                       ase activity is elevated in the vast majority of tumors, and telomeres are critically shortened in
                       tumors versus normal tissues, thus providing a compelling rationale to target the telomerase/
                       telomere pathway for broad-spectrum cancer therapy. This strategy is supported by a variety of
                       genetic-based target validation studies. Both telomerase inhibitors and telomere interactive mol-
                       ecules have shown stand-alone antitumor activity at nontoxic doses against a variety of human
                       tumor xenografts in mice. These translational advances have resulted in the first antitelomerase
                       agent, the oligonucleotide-based GRN163L targeting the telomerase RNA template, entering
                       clinical evaluation. Additional translational approaches, such as targeting telomeres using
                       G-quadruplex ligands, should result in antitelomere agents, such as RHPS4, entering the clinic in
                       the near future. These prototype trials will be extremely informative in determining the role of the
                       telomerase/telomere pathway in clinical oncology and, moreover, whether drugs targeting the
                       unlimited replicative potential of cancer will find a place in cancer chemotherapy.

                                                                                    angiogenesis, migration, or invasion), and ultimately whether
 Background
                                                                                    small-molecule or biological modulators provide a benefit to
   Current cancer drug development is largely focused on                            cancer patients (2). Pioneering studies done over the past 2
combating the major phenotypical features of cancer [i.e.,                          decades have shown that one of the above-described and to
uncontrolled growth (through inactivation of tumor suppres-                         date therapeutically untapped phenotypic hallmarks of cancer,
sors, activation of oncogenes, and evasion of apoptosis),                           unlimited replicative potential, is intimately related to the
unlimited replicative potential, formation of new blood vessels                     maintenance of telomeres, repetitive TTAGGG sequences of
(angiogenesis), and tissue invasion and metastatic spread;                          DNA located on the ends of human chromosomes. Whereas
ref. 1]. In recent years, this strategy has resulted in some notable                mortal cells shorten their telomeres during each round of
success stories with new approved molecularly targeted drugs                        replication due to the ‘‘end-replication problem,’’ in tumor cells
that have made a significant effect in lengthening the survival of                  telomere length is stabilized mainly via reactivation of the
cancer sufferers (e.g., imatinib in chronic myelogenous                             reverse transcriptase termed telomerase, composed of a RNA
leukemia, trastuzumab in breast cancer, and bevacizumab in                          component (hTR or hTERC) and a catalytic protein (hTERT;
colorectal cancer; ref. 2).                                                         see ref. 3 for a recent review). Moreover, the introduction of
                                                                                    hTERT into normal human cells extends their life span (4) and
 The Relevance of theTelomere Maintenance                                           hTERT (along with two oncogenes, large T and H-ras)
                                                                                    expression results in the direct tumorigenic conversion of
 Pathway to Cancer
                                                                                    normal human epithelial and fibroblast cells (5). Other studies,
  Validation of a cancer target encompasses information on the                      however, suggest that telomerase may also play a role in
prevalence and role of the target or pathway in human cancer,                       maintaining telomeres in some normal human cells (6) and
whether modulation of the target or pathway in preclinical                          may possess additional roles to telomere maintenance, such
model systems results in a robust anticancer phenotype (e.g.,                       as regulation of chromatin state and DNA damage responses,
growth inhibition, induction of apoptosis or prevention of                          as cells lacking hTERT possessed a diminished capacity for
                                                                                    DNA double-strand break repair and fragmented chromosomes
                                                                                    (7). In addition, in some tumors (particularly those of
                                                                                    neuroepithelial and mesenchymal origin), telomeres are main-
Author’s Affiliation: Head of Biology, Cancer ResearchTechnology Development
Laboratory, Wolfson Institute for Biomedical Research, University College London,
                                                                                    tained by an alternative lengthening of telomere mechanism
London, United Kingdom                                                              (8). In patients with glioblastoma multiforme tumors, alterna-
Received 2/20/07; revised 3/16/07; accepted 3/20/07.                                tive lengthening of telomere is associated with mutant TP53
Requests for reprints: Lloyd Kelland, Head of Biology, Cancer Research              and a favorable prognosis (9); characteristics of alternative
Technology Development Laboratory, Wolfson Institute for Biomedical Research,       lengthening of telomere cells are alternative lengthening of
University College London, London WC1E BBT, United Kingdom. Phone: 44-20-
76796949; E-mail: lkelland@ cancertechnology.com.
                                                                                    telomere – associated promyelocytic leukemia bodies and cir-
   F 2007 American Association for Cancer Research.                                 cular extrachromosomal telomeric DNA (telomeric t circles,
   doi:10.1158/1078-0432.CCR-07-0422                                                which require Xrcc3 and Nbs1 for their production; ref. 10).

Clin Cancer Res 2007;13(17) September 1, 2007                                   4960                                       www.aacrjournals.org
              Downloaded from clincancerres.aacrjournals.org on October 12, 2015. © 2007 American Association for
                                                      Cancer Research.
Targeting theTelomerase/Telomere Pathway

Finally, a group (six to date) of telomere-associated proteins,      resulted in growth inhibition but this occurred with a different
collectively termed shelterin, comprising TRF1 and TRF2 that         lag time dependent on this initial telomere length (15). Such
bind to double-stranded telomeric DNA, POT1 that binds the           studies supported the view that, following inhibition of
single-stranded 3¶ G-rich overhang, and three interconnecting        telomerase, telomeres needed to be gradually eroded down to
proteins (TIN2, TPP1, and RAP1), acts to shape and safeguard         a critical length before any phenotypic effect occurred. Hence,
telomeres (Fig. 1; refs. 11, 12).                                    small-molecule mimics of this effect would be best suited to the
   The interest in telomerase and telomeres in a cancer              treatment of tumors possessing very short telomeres.
therapeutics context has emerged from two main observations:            Subsequent studies, however, targeting components of
first, telomerase activity, typically using a PCR-based assay,       telomerase or telomeres/shelterin have shown that much more
telomeric repeat amplification protocol, is elevated in the great    rapid apoptosis or induction of senescence effects can occur.
majority (f85%) of tumor types in comparison with normal             For example, antisense oligonucleotide-mediated inhibition of
tissues, and second, telomeres are generally shorter in tumors       hTERT, but not hTERC, caused a rapid reduction in cell growth
than in corresponding normal tissues (see refs. 13, 14 for           and induction of apoptosis without telomere shortening in
reviews). Furthermore, several studies provide genetic valida-       human prostate cancer cells (16). Second, the introduction of
tion of telomerase as an anticancer target in that inactivation of   mutant template telomerase RNA into human tumor cells
hTR and/or hTERT by dominant-negative mutants or antisense           rapidly decreased cell viability and increased apoptosis,
strategies resulted in an inhibition of tumor cell proliferation.    possibly via a telomere uncapping mechanism and triggering
For example, transfection of a dominant-negative mutant of           a DNA damage response (17, 18). Third, ribozymes targeting
hTERT into cancer cell lines of differing initial telomere length    murine telomerase RNA induced antitumor effects, including

Fig. 1. The telomerase/telomere pathway
and main points of possible therapeutic
intervention. Pathway modulators may
result in either mainly telomere erosion
(e.g., BIBR1532) resulting in relatively
slow phenotypic anticancer effects or
predominantly telomere uncapping (e.g.,
RHPS4) producing more rapid anticancer
effects. Agents such as GRN163L probably
mainly affect telomere erosion but also seem
to induce telomere uncapping.

www.aacrjournals.org                                             4961           Clin Cancer Res 2007;13(17) September 1, 2007
                Downloaded from clincancerres.aacrjournals.org on October 12, 2015. © 2007 American Association for
                                                        Cancer Research.
Molecular Pathways

reducing melanoma tumor invasion and metastases in vivo;             derivative of GRN163, a 13-mer thio-phosphoramidate oligo-
these studies also showed that telomerase activity may control       nucleotide complementary to the RNA template of hTR (28).
the glycolytic pathway (19, 20). Finally, other genetic and          Although cell-based studies with GRN163 generally showed a
biochemical studies suggest that targeting components of             phenotypic lag of at least a few weeks between the onset of
shelterin, such as TRF2 (21) or POT1 (12, 22, 23), or exposing       exposure and growth inhibition (e.g., ref. 28), the addition of a
the telomere 3¶ overhang (24) can induce apoptosis or                5¶ lipid (palmitate) moiety in GRN163L increased potency and
telomere-initiated senescence and thereby activate a DNA             resulted in a more rapid loss of telomeres and inhibition of cell
damage checkpoint response (25).                                     growth (29). Furthermore, in vivo inhibition of tumor growth
   Because telomerase activity is relatively high in many tumors     by GRN163L has been reported in mice in A549 human lung
and telomeres are generally shorter in tumors than normal            cancer metastases treated with 15 mg/kg thrice weekly for 3
tissues, taken together with the above-described genetic-based       consecutive weeks (30). Antitumor activity, observable from 2
validation studies, provide a compelling argument to suggest         to 3 weeks of thrice-weekly dosing, has also been reported in
that the telomerase/telomere pathway is a well-validated target      mice bearing orthotopic MDA-MB-231 human breast cancer
at the preclinical level. Therefore, appropriate modulators          xenografts (31).
should be tested in patients.                                           Over the past decade, many chemical classes of G-quadruplex
                                                                     ligands have been described, the most potent of which,
    Clinical Translational Advances                                  telomestatin, inhibits telomerase in the telomeric repeat
                                                                     amplification protocol assay at low nanomolar concentrations
   As shown in Fig. 1, the burgeoning knowledge about the            (32). Other compounds have been rationally designed based
structure of telomeres and the roles of various proteins involved    on the crystal structure of parallel quadruplexes from human
in telomere maintenance provides several possible targets for        telomeric DNA (33, 34). Several reduce the growth of various
modulation. Some of these, such as modulating the interaction        cancer cell lines in vitro after 1 to 2 weeks of exposure to low
of TRF2 or POT1 with telomeres, may hold promise as cancer           micromolar concentrations, often accompanied by cellular
drugs but, as yet, have not been described. To date, two areas       senescence (34 – 37), apoptosis (32, 36), and/or chromosomal
have received major drug discovery attention: first, the targeting   fusions/anaphase bridges (32, 35, 36). Several G-quadruplex
of components of telomerase (e.g., hTR RNA template or               ligands have exhibited antitumor activity in vivo at nonacute
hTERT protein), and second, the targeting of telomeres (e.g.,        toxic doses in mice bearing various human tumor xenografts:
G-quadruplex ligands that induce the formation of guanine-           the 3,6,9-trisubstituted acridine BRACO19 in combination with
rich telomeres to stably fold into four-stranded G-quadruplex        paclitaxel against vulval carcinoma (34) or as monotherapy
structures, thereby adversely affecting telomere function).          against DU145 prostate cancer (38) or UXF1138L uterine
   Although, as described above, telomerase represents an            cancer (39), the cationic porphyrin TMPyP4 against PC3
attractive cancer target, surprisingly few potent small-molecule     prostate cancer (40), telomestatin against U937 acute myelo-
inhibitors have been discovered to date. It is unclear whether       monocytic leukemia (41), and the pentacyclic acridine RHPS4
this is because of a general lack of interest in pursuing this       against CG5 breast cancer (42). In general, antitumor effects
target within the pharmaceutical industry (perhaps because of        were apparent relatively rapidly following the onset of drug
the phenotypic lag arguments described above) or because the         treatment, typically after f10 days (34, 38 – 42). A consistent
enzyme is not particularly tractable to the main current drug        mechanism of action schema is emerging involving displace-
discovery approach using high-throughput screening of large          ment of POT1 and TRF2 from telomeres and erosion of 3¶
compound libraries resulting in few chemical hits or leads           single-stranded telomeric overhangs (G-tails) with activation
emerging. One small-molecule nonnucleoside inhibitor of              of a DNA damage response [e.g., reported with telomestatin
hTERT, BIBR1532, has been studied in preclinical models,             (32, 43, 44) and RHPS4 (42)]. Such findings suggest that the
including in vivo (26, 27). BIBR1532 inhibited telomerase in         G-quadruplex ligands can induce relatively rapid telomere
cell-free assays with an IC50 of 93 nmol/L and was shown to be       uncapping and may provide an explanation as to why
a mixed-type noncompetitive inhibitor with a drug binding site       reductions in telomere length are not always observed in cells
distinct from the sites for deoxyribonucleotides and the DNA         exposed to this class of compound. The cell-based effects
primer. Cell-based studies with BIBR1532 revealed a significant      (senescence, apoptosis, activation of a DNA damage response
phenotypic lag, typically of f100 days, between the onset of         including gH2AX phosphorylation, etc.) are also consistent
telomerase inhibition and cellular growth arrest, hence              with modulation of the currently known biological roles of
mimicking the above-described studies with dominant-negative         POT1 (23) and TRF2 (21). Finally, measurements of POT1 or
hTERT. Antitumor effects were demonstrable in vivo in mice           TRF2 displacement from telomeres or gH2AX phosphorylation
following inoculation of HT1080 fibrosarcoma cells that had          may provide sensitive and appropriate biomarkers of target
been previously exposed to the compound for 100 days in vitro        modulation in clinical trials. At least some of the G-quadruplex
(and leading to a reduction in telomere length to 1.6 kb).           ligands, such as RHPS4, are expected to enter clinical trials in
   An oligonucleotide-based molecule that acts as a telomerase       the near future.
RNA template antagonist, GRN163L, represents the most
advanced antitelomerase therapeutic. Phase I clinical trials
                                                                        Concluding Remarks
have been initiated in patients with either chronic lymphocytic
leukemia or solid tumors.1 GRN163L is a more potent                     The telomerase/telomere field has come a long way in a
                                                                     relatively short time and is now at a particularly exciting
                                                                     juncture, as the first telomerase inhibitor, GRN163L, undergoes
1
    http://www.clinicaltrials.gov/                                   early clinical development. Other small-molecule modulators

Clin Cancer Res 2007;13(17) September 1, 2007                    4962                                      www.aacrjournals.org
                 Downloaded from clincancerres.aacrjournals.org on October 12, 2015. © 2007 American Association for
                                                         Cancer Research.
Targeting theTelomerase/Telomere Pathway

of telomerase/telomere biology, notably G-quadruplex ligands                               a broad-spectrum of cancers, most likely in combination with
such as RHPS4, are in preclinical development and are                                      existing chemotherapeutic drugs. Although there is relatively
anticipated to enter the clinic in the near future. These clinical                         little to guide rational clinical combinations of antitelomerase/
studies will be extremely informative in determining whether                               telomere therapeutics with other drugs at present (and
the positive anticancer findings from various genetic-based                                although dose-limiting toxicities remain unknown), the asso-
preclinical validation studies are borne out in cancer patients.                           ciation of telomerase/telomeres with DNA repair, particularly
Furthermore, based on the high prevalence of elevated                                      double-strand break repair, pathways is suggestive of this new
telomerase and/or shortened telomeres in multiple tumor types                              class of agent being usefully combined with cytotoxics that
in comparison with normal tissue counterparts, targeting                                   induce DNA damage (e.g., platins and topoisomerase inhib-
telomerase/telomeres could be applicable to the treatment of                               itors) or ionizing radiation.

References
1. Hanahan D, Weinberg RA. The hallmarks of cancer.          tumor cell proliferation. Proc Natl Acad Sci U S A          phase bridge formation accompanied by loss of the 3¶
  Cell 2000;100:57 ^ 70.                                     2001;98:7982 ^ 7.                                           telomeric overhang in cancer cells. Oncogene 2006;
2. Benson JD, ChenY-NP, Cornell-Kennon SA, et al. Val-      18. Goldkorn A, Blackburn EH. Assembly of mutant-            25:1955 ^ 66.
  idating cancer drug targets. Nature 2006;441:451 ^ 6.      template telomerase RNA into catalytically active telo-    33. Parkinson GN, Lee MPH, Neidle S. Crystal structure
3. Blackburn EH, Greider CW, Szostak JW. Telomeres           merase ribonucleoprotein that can act on telomeres is       of parallel quadruplexes from human telomeric DNA.
  and telomerase: the path from maize, tetrahymena           required for apoptosis and cell cycle arrest in human       Nature 2002;417:876 ^ 80.
  and yeast to human cancer and aging. Nat Med               cancer cells. Cancer Res 2006;66:5763 ^ 71.                34. Gowan SM, Harrison JR, Patterson L, et al. A
  2006;12:1133 ^ 8.                                         19. Nosrati M, Li S, Bagheri S, et al. Antitumor activity    G-quadruplex-interactive potent small-molecule inhib-
4. Bodnar AG, Ouellette M, Frolkis M, et al. Extension of    of systemically delivered ribozymes targeting murine        itor of telomerase exhibiting in vitro and in vivo anti-
  life-span by introduction of telomerase into normal        telomerase RNA. Clin Cancer Res 2004;10:4983 ^ 90.          tumour activity. Mol Pharmacol 2002;61:1154 ^ 62.
  human cells. Science 1998;279:349 ^ 52.                   20. Bagheri S, Nosrati M, Li S, et al. Genes and path-      35. Incles CM, Schultes CM, Kempski H, Koehler H,
5. Hahn WC, Counter CM, Lundberg AS, Beijersbergen           ways downstream of telomerase in melanoma metas-            Kelland LR, Neidle S. A G-quadruplex telomere target-
  RL, Brooks MW, Weinberg RA. Creation of human              tasis. Proc Natl Acad Sci U S A 2006;103:11306 ^ 11.        ing agent produces p16-associated senescence and
  tumour cells with defined genetic elements. Nature        21.Van Steensel B, Smogorzewska A, de Lange T.TRF2           chromosomal fusions in human prostate cancer cells.
  1999;400:464 ^ 8.                                          protects human telomeres from end to end fusions.           Mol CancerTher 2004;3:1201 ^ 6.
6. Masutomi K, Yu EY, Khurts S, et al. Telomerase            Cell 1998;92:401 ^ 13.                                     36. Leonetti C, Amodei S, D’Angelo C, et al. Biological
  maintains telomere structure in normal human cells.       22. Zaug AJ, Podell ER, CechTR. Human POT1disrupts           activity of the G-quadruplex ligand RHPS4 (3,11-
  Cell 2003;114:241 ^ 53.                                    telomeric G-quadruplexes allowing telomerase exten-         difluoro-6,8,13-trimethyl-8H-quino[4,3,2-kl] acridi-
7. Masutomi K, Possemato R,Wong JMY, et al.The telo-         sion in vitro. Proc Natl Acad Sci U S A 2005;102:           nium methosulfate) is associated with telomere capping
  merase reverse transcriptase regulates chromatin state     10864 ^ 9.                                                  alteration. Mol Pharmacol 2004;66:1138 ^ 46.
  and DNA damage responses. Proc Natl Acad Sci              23. Churikov D, Wei C, Price CM. Vertebrate POT1            37. Cookson JC, Dai F, SmithV, et al. Pharmacodynam-
  U S A 2005;102:8222 ^ 7.                                   restricts G-overhang length and prevents activation         ics of the G-quadruplex-stabilizing telomerase inhibi-
8. Dunham MA, Neumann AA, Fasching CL, Reddel                of a telomeric DNA damage checkpoint but is dis-            tor 3,11-difluoro-6,8,13-trimethyl-8H-quino[4,3,2-kl]
  RR. Telomere maintenance by recombination in human         pensable for overhang protection. Mol Cell Biol             acridinium methosulfate (RHPS4) in vitro : activity in
  cells. Nat Genet 2000;26:447 ^ 50.                         2006;26:6971 ^ 82.                                          human tumor cells correlates with telomere length
9. Chen Y-J, Hakin-Smith V,Teo M, et al. Association of     24. Li G-Z, Eller MS, Firoozabadi R, Gilchrest BA.           and can be enhanced, or antagonized, with cytotoxic
  mutantTP53 with alternative lengthening of telomeres       Evidence that exposure of the telomere 3¶ overhang se-      agents. Mol Pharmacol 2005;68:1551 ^ 8.
  and favorable prognosis in glioma. Cancer Res 2006;        quence induces senescence. Proc Natl Acad Sci U S A        38. Kelland LR. Overcoming the immortality of tumour
  66:6473 ^ 6.                                               2003;100:527 ^ 31.                                          cells by telomere and telomerase based cancer thera-
10. Compton SA, Choi J-H, Cesare AJ, Ozgur S,               25. di Fagagna FD, Reaper PM, Clay-Farrace L, et al.         peuticsJcurrent status and future prospects. Eur J
  Griffith JD. Xrcc3 and Nbs1 are required for the pro-      A DNA damage checkpoint response in telomere-               Cancer 2005;41:971 ^ 9.
  duction of extrachromosomal telomeric circles in           initiated senescence. Nature 2003;426:194 ^ 8.             39. Burger AM, Dai F, Schultes CM, et al. The G-quad-
  human alternative lengthening of telomere cells.          26. Damm K, Hemmann U, Garin-Chesa P, et al. A               ruplex-interactive molecule BRACO-19 inhibits tumor
  Cancer Res 2007;67:1513 ^ 9.                               highly selective telomerase inhibitor limiting human        growth, consistent with telomere targeting and inter-
11. de lange T. Shelterin: the protein complex that          cancer cell proliferation. EMBO J 2001;20:6958 ^ 68.        ference with telomerase function. Cancer Res 2005;
  shapes and safeguards human telomeres. Genes Dev          27. Pascolo E, Wenz C, Lingner J, et al. Mechanism of        65:1489 ^ 96.
  2005;19:2100 ^ 10.                                         human telomerase inhibition by BIBR1532, a synthetic,      40. Grand CL, Han H, Munoz RM, et al. The cationic
12. Wang F, Podell ER, Zaug AJ, et al. The POT1-TPP1         non-nucleoside drug candidate. J Biol Chem 2002;            porphyrin TMPyP4 down-regulates c-MYC and
  telomere complex is a telomerase processivity factor.      277:15566 ^ 72.                                             human telomerase reverse transcriptase expression
  Nature 2007;445:506 ^ 10.                                 28. Akiyama M, HideshimaT, Shammas MA, et al. Effects        and inhibits tumor growth in vivo. Mol Cancer Ther
13. Shay JW. Meeting report: the role of telomeres and       of oligonucleotide N3¶-P5¶ thio-phosphoramidate             2002;1:565 ^ 73.
  telomerase in cancer. Cancer Res 2005;65:3513 ^ 7.         (GRN163) targeting telomerase RNA in human multiple        41. Tauchi T, Shin-ya K, Sashida G, et al. Telomerase inhi-
14. Shay JW, Wright WE. Telomerase therapeutics for          myeloma cells. Cancer Res 2003;63:6187 ^ 94.                bition with a novel G-quadruplex interactive agent,
  cancer: challenges and new directions. Nat Rev Drug       29. Herbert B-S, Gellert GC, Hochreiter A, et al. Lipid      telomestatin: in vitro and in vivo studies in acute
  Discov 2006;5:577 ^ 83.                                    modification of GRN163, an N3¶-P5¶ thio-phosphora-          leukemia. Oncogene 2006;25:5719 ^ 25.
15. Hahn WC, Stewart SA, Brooks MW, et al. Inhibition        midate oligonucleotide, enhances the potency of telo-      42. Salvati E, Leonetti C, Rizzo A, et al. Telomere damage
  of telomerase limits the growth of human cancer cells.     merase inhibition. Oncogene 2005;24:5262 ^ 8.               promotes antitumoral activity of the G-Quadruplex
  Nat Med 1999;5:1164 ^ 70.                                 30. Dikmen ZG, Gellert GC, Jackson S, et al. In vivo         ligand RHPS4. J Clin Invest. In press 2007.
16. Folini M, Brambilla C, Villa R, et al. Antisense         inhibition of lung cancer by GRN163L: a novel human        43. Gomez D, Wenner T, Brassart B, et al. Telomestatin-
  oligonucleotide-mediated inhibition of hTERT, but          telomerase inhibitor. Cancer Res 2005;65:7866 ^ 73.         induced telomere uncapping is modulated by POT1
  not hTERC, induces rapid cell growth decline and          31. Hochreiter AE, Xiao H, Goldblatt EM, et al. Telomer-     through G-overhang extension in HT1080 human tumor
  apoptosis in the absence of telomere shortening in         ase template antagonist GRN163L disrupts telomere           cells. J Biol Chem 2006;281:38721 ^ 9.
  human prostate cancer cells. Eur J Cancer 2005;41:         maintenance, tumor growth, and metastasis of breast        44. Gomez D, O’Donohue M-F, Wenner T, et al. The
  624 ^ 34.                                                  cancer. Clin Cancer Res 2006;12:3184 ^ 92.                  G-quadruplex ligand telomestatin inhibits POT1 bind-
17. Kim MM, Rivera MA, Botchkina IL, Shalaby R, Thor        32. Tahara H, Shin-ya K, Seimiya H,Yamada H,TsuruoT,         ing to telomeric sequences in vitro and induces GFP-
  AD, Blackburn EH. A low threshold level of expression      IdeT. G-quadruplex stabilization by telomestatin indu-      POT1 dissociation from telomeres in human cells.
  of mutant-template telomerase RNA inhibits human           cesTRF2 protein dissociation from telomeres and ana-        Cancer Res 2006;66:6908 ^ 12.

www.aacrjournals.org                                                                4963                   Clin Cancer Res 2007;13(17) September 1, 2007
                Downloaded from clincancerres.aacrjournals.org on October 12, 2015. © 2007 American Association for
                                                        Cancer Research.
Targeting the Limitless Replicative Potential of Cancer: The
Telomerase/Telomere Pathway
Lloyd Kelland

Clin Cancer Res 2007;13:4960-4963.

 Updated version        Access the most recent version of this article at:
                        http://clincancerres.aacrjournals.org/content/13/17/4960

     Cited articles     This article cites 43 articles, 26 of which you can access for free at:
                        http://clincancerres.aacrjournals.org/content/13/17/4960.full.html#ref-list-1

    Citing articles     This article has been cited by 14 HighWire-hosted articles. Access the articles at:
                        http://clincancerres.aacrjournals.org/content/13/17/4960.full.html#related-urls

      E-mail alerts     Sign up to receive free email-alerts related to this article or journal.

     Reprints and       To order reprints of this article or to subscribe to the journal, contact the AACR Publications
    Subscriptions       Department at pubs@aacr.org.

      Permissions       To request permission to re-use all or part of this article, contact the AACR Publications
                        Department at permissions@aacr.org.

           Downloaded from clincancerres.aacrjournals.org on October 12, 2015. © 2007 American Association for
                                                   Cancer Research.
You can also read