Sie sind auf Seite 1von 8

MINIREVIEW

Human telomeric G-quadruplex: The current status


of telomeric G-quadruplexes as therapeutic targets
in human cancer
Stephen Neidle
Cancer Research UK Biomolecular Structure Group, University of London, UK

Keywords The 3-ends of human chromosomal DNA terminate in short single-


acridine; anticancer; drug; drug-like; in vivo; stranded guanine-rich tandem-repeat sequences. In cancer cells, these are
medicinal chemistry; pharmacology;
associated with the telomere-maintenance enzyme telomerase together with
quadruplex; telomerase; telomere
the end-binding protein hPOT1. Small molecules that can compete with
Correspondence these proteins and induce the single-stranded DNA to form quadruplex
Stephen Neidle, Cancer Research UK ligand complexes are, in effect, able to expose these 3-ends, which results
Biomolecular Structure Group, The School in the activation of a DNA damage response and selective inhibition of cell
of Pharmacy, University of London, growth. Several of these G-quadruplex binding molecules have shown
29-39 Brunswick Square, London promising anticancer activity in tumour xenograft models, which indicate
WC1N 1AX, UK
that the approach may be applicable to the treatment of a wide range of
Fax: +44 207 753 5970
Tel: +44 207 753 5969
human cancers. This minireview summarizes the available data on these
E-mail: stephen.neidle@pharmacy.ac.uk compounds and the challenges posed for drug discovery.

(Received 25 June 2009, revised 5 October


2009, accepted 6 October 2009)

doi:10.1111/j.1742-4658.2009.07463.x

Introduction
Human telomeres comprise tandem repeats of the DNA damage signal, producing responses analogous
DNA motif (TTAGGG) together with associated telo- to those of other mediators of telomere damage [12].
meric proteins [13], as well as other more transiently The biological function of induced telomeric quadru-
associated DNA-repair and damage-response proteins plexes remains to be fully claried; an end-protective
such as Ku [4]. The terminal 150250 nucleotides at role has been suggested, there is evidence of functional
the extreme 3-ends of telomeres are single-stranded interactions involving poly(ADP-ribose) polymerase-1
[5], but are protected from higher order aggregation by [13] and in ciliates at least, quadruplex structures are
binding to multiple repeats of a single-stranded DNA involved in telomerase recruitment [14,15]. However,
binding protein (hPOT1 in humans), which in turn to date, there is no direct evidence of a role for telo-
interacts with other proteins in the core telomere meric G-quadruplexes in the functioning of telomeres
complex, notably TPP1, to regulate telomerase action in normal human cells.
in cancer cells, and thereby maintain telomere length Telomerase is overexpressed in  8085% of cancer
[68]. Loss of hPOT1 deprotects telomeres and initiates cells and primary tumours [16,17] and maintains
DNA damage-response mediated cell death. Small telomere length homeostatis (acting as a tumour
molecules that stabilize the single strand into higher promoter). Telomere shortening in the absence of sig-
order (G-quadruplex) structures compete with hPOT1 nicant telomerase expression appears to be a tumour
and also initiate this response [911]. Thus, quadruplex suppressor mechanism [3]. Telomeres in telomerase-
formation at the single-strand overhang may itself be a negative somatic cells are gradually shortened as a

1118 FEBS Journal 277 (2010) 11181125 2009 The Author Journal compilation 2009 FEBS
S. Neidle G-quadruplexes as cancer drug targets

consequence of the end-replication effect, and once subsequently [18,21,22], although relatively few have
telomeric DNA is at a critically short length, cells been evaluated in cell-based assays, or even with reli-
enter p53 and Rb-dependent replicative senescence, able in vitro telomerase assays [23,24]. The majority of
and ultimately apoptosis. The catalytic subunit of telo- G-quadruplex ligands contain a polycyclic heteroaro-
merase (hTERT in humans) has reverse transcriptase matic core, although it is clear that this is not an
enzymatic activity and synthesizes TTAGGG repeats essential requirement for quadruplex binding. Several
on to the end of the 3 single-stranded overhang. Inhi- effective quadruplex-binding ligands do not have this
bition of hTERT by siRNA, antisense or small-mole- feature. The cyclic polyamine telomestatin (Fig. 1) was
cule inhibitors selectively inhibits cancer cell growth the rst such compound [25] to show both high quad-
and strongly suggests that induction of telomere short- ruplex afnity and telomerase inhibitory potency.
ening is a viable therapeutic strategy [18]. More recent reports have demonstrated that nonconju-
Folding the single-stranded telomeric DNA substrate gated compounds that are synthetically more accessible
of telomerase into a four-stranded quadruplex struc- than telomestatin can have potency against telomerase
ture inhibits the enzymes catalytic activity [19] because and quadruplex selectivity [2629].
it ensures that the 3-end is inaccessible to hybridize
with the telomerase RNA template, the essential rst
Telomeric quadruplex ligands possible
step in the catalytic cycle. The induction of quadruplex
mechanisms of action
stabilization and telomerase inhibition by a quadru-
plex-binding small molecule was rst demonstrated The classic model of telomerase inhibition and conse-
using a disubstituted anthraquinone derivative [20]. quent telomere attrition leading to senescence and
Many quadruplex-binding ligands have been reported apoptosis requires that cells with a mean telomere

Fig. 1. Structures of quadruplex-binding


ligands.

FEBS Journal 277 (2010) 11181125 2009 The Author Journal compilation 2009 FEBS 1119
G-quadruplexes as cancer drug targets S. Neidle

length of 5 kb, a 24 h cell-doubling time and a sub- teristic DNA damage foci using an antibody to the
sequent loss of  100 nucleotides per round of repli- damage response protein cH2AX [36], or by a signi-
cation would reach critical telomere shortening in cant population of cells undergoing end-to-end fusions
 4050 days [30,31]. This was indeed the observation in metaphase [37]. Such changes are analogous to
in dominant-negative telomerase transfection experi- those produced when the telomeric protein TRF2 is
ments, but would be therapeutically challenging for knocked out. This response is a consequence of the
human cancer treatment. Initial ndings using G-quad- displacement of bound proteins from the single-
ruplex ligands showed very different behaviour, with stranded overhang, chiey hPOT1, as well as possible
senescence occurring within 710 days after cells were uncapping of telomerase from the ends. There are
rst treated, and little evidence of concomitant telo- likely to be multiple mechanisms involved, some of
mere shortening [11,18,32]. This has subsequently been which at least have cross-talk between them (Fig. 2).
shown to be characteristic of the G-quadruplex ligand For example, hPOT1 interacts with the telomeric pro-
class as a whole, and the observations of on-target tein Tpp1 and facilitates telomere length regulation
in vivo activity within clinically useful timescales are by telomerase, and hPOT1 displacement disregulates
encouraging signs that signicant single-agent clinical telomerase function [7,8]. Also, although the classic
utility may be eventually achievable with appropriate telomerase inhibition model does not appear to be fol-
compounds. lowed by G-quadruplex-binding agents, cancer cells
The quadruplex-binding acridine ligands BRACO-19 generally have marked telomere length heterogeneity,
and RHPS4 (Fig. 1), in common with telomestatin, with some having extremely short (< 1 kb) telomeres.
induce rapid replicative senescence in cancer cells and It has been suggested that these cells are not only
activate the same DNA damage response that follows sensitive to senescence, but also that their viability is
DNA double-strand breaks. This involves in particular critical to the cell population overall [38,39], although
ATM, p16INK4a kinase and p53 pathways [3235] it is not clear to what extent telomere shortening,
which can be visualized by the appearance of charac- initially considered to be an essential marker of

Fig. 2. Schematic of mechanism of action of the telomeric quadruplex ligand BRACO-19.

1120 FEBS Journal 277 (2010) 11181125 2009 The Author Journal compilation 2009 FEBS
S. Neidle G-quadruplexes as cancer drug targets

telomerase inhibition, is relevant to the short-term no experimental structural data as yet on folded
effects of telomeric G-quadruplex ligands. Q-FISH telomeric DNA sequences containing eight or twelve
studies have shown that telomestatin is localized at TTAGGG repeats (i.e. with two or three consecutive
telomeres during replication and importantly, that telo- quadruplexes), which may be more representative of
mere replication is unaffected in mouse embryonic the totality of the single-stranded overhang, and
broblast (i.e. untransformed) cell lines [40]. which may be important for these ligands being able
Validation of a telomeric quadruplex mode of action to differentiate telomeric quadruplexes from others in
involves evidence from a number of assays. The most the genome.
important are: (a) high-afnity in vitro telomeric quad- It has long been realized that therapeutically effec-
ruplex binding, with a Ka value of at least 106 m)1; (b) tive quadruplex-binding ligands should have minimal
a low level of binding to duplex DNA, with a Ka value duplex DNA afnity (and therefore more generalized
at least 102 less than for telomeric quadruplexes; (c) toxicity), and assays for duplex:quadruplex selectivity
selective inhibition of cell growth, with normal human are routinely performed in many laboratories. The
cell lines being relatively unaffected; (d) senescence; (e) structural requirements for selectivity have not yet
inhibition of telomerase activity in cells; (f) competitive been fully claried, but mostly involve those steric fea-
inhibition of hPOT1 binding in cells; and (g) evidence tures that are incompatible with the dimensions of a
of telomere uncapping in cells from hTERT. double helix. A large number of genomic DNA and
RNA G-quadruplexes may also be drug targets [48
53], many of which are involved in proliferation. It is
G-quadruplex ligands as drugs
plausible that G-quadruplex-binding molecules even
In vivo activity in xenograft cancer models has been with relatively modest selectivity between various
reported to date for few telomeric quadruplex ligands, G-quadruplexes, may still have utility in cancer
notably the trisubstituted acridine compound BRACO- therapeutics, provided they have low toxicity to
19 [32], the polycyclic compound RHSP4 [34,35] and normal cells. Of greater practical importance is that
telomestatin [41] (Fig. 1). The telomeric DNA single- future G-quadruplex ligands are developed with regard
strand overhang is a target for all these compounds, as to their ability to be used as drugs, so that they have:
judged by the observations of hPOT1 and hTERT (a) effective and selective tumour uptake and penetra-
uncapping. To date, none of these molecules has pro- tion, (b) acceptable pharmacokinetic characteristics
gressed beyond the experimental stage into clinical and metabolism, and (c) a signicant therapeutic
trial, probably in part because these compounds are window.
insufciently drug-like. Little data is publicly available The features common to most current quadruplex
on their ADME pharmacokinetic properties. ligands, of several cationic charges and large hydro-
To date, the development of small molecules as phobic surface area, do aid cellular uptake (probably
G-quadruplex binders has been largely based on poly- by active transport mechanisms), but may also enable
cyclic planar aromatic compounds with at least one a high background of nonspecic binding to cellular
substituent terminating in a cationic group [20,21]. components, and are not consistent with oral bio-avail-
Normally two such substituents are required. The ability (although this in itself may not be an important
rationale for the planar moiety has been that this goal). The three positive charges on the BRACO-19
would stack effectively onto planar G-quartets, which molecule are probably a factor in the inability of this
has been conrmed by several crystallographic and compound to penetrate larger tumours in both the
NMR studies of G-quadruplexligand complexes [42 UXF1138L and A431 xenograft models [32,54]
47]. There is no evidence from these studies of classic (Table 1). Compound AS1410 was devised [55] to have
intercalation between G-quartets and all analyses con- increased hydrophobicity compared with its parent
cur in nding that ligands stack onto a terminal compound BRACO-19 as a result of modications to
G-quartet of a quadruplex. Substituents are normally the substituents at the 9-position. This resulted in an
short acyclic chains, such as -(CH2)3- with a terminal increase in plasma half-life from 1 to 2 h.
cationic nitrogen-containing group such as diethyl- The limited in vivo data available (Table 1) suggest
amine, pyrrolidine or piperidine. Structure-based drug that telomeric quadruplex ligands may be useful for
discovery does have these few structures as starting the treatment of solid tumours; to date there is very
points [4247], although these also indicate that the little data on haematological cancers. Notable ndings
exibility of the TTA loops is ligand dependent, and include that of single-agent activity for RHSP4 in a
therefore structural information for a given class metastatic melanoma model, as well as in a melanoma
of ligand would be highly desirable. Also, there are line resistant to the platinum drug DDP [56]. RHPS4

FEBS Journal 277 (2010) 11181125 2009 The Author Journal compilation 2009 FEBS 1121
G-quadruplexes as cancer drug targets S. Neidle

Table 1. Selected in vivo data on quadruplex-binding ligands. Tumour responses have been estimated from survival curves and other avail-
able data. Single-agent studies. i.p., intraperitoneal; i.v., intravenous.

Days to
Mean initial Dosage complete
G4 ligand Xenograft model tumour size (mgkg)1) Tumour response response Ref

TMPyP4 MX-1 mammary tumor 100 mga 10, 20; i.p. Survival increase from 60 57
45% to 75%
TMPyP4 PC-3 human prostate 60 mg 40; i.p. 60% tumour shrinkage 18 57
carcinoma
Telomestatin U937 human lymphoma 1395 mm3 15 80% tumour shrinkage 21 41
BRACO-19 UXF1138L human uterine 68 mm3 2; i.p. 96% tumour shrinkage 28 32
carcinoma + some complete
remissions
BRACO-19 A431 human epithelial 1080 mm3 2; i.p. Not significant 54
carcinoma
Quarfloxin MDA-MB-231 human > 125 mm3 6.25, 15.5; i.v. 50% tumour shrinkage 37 58
breast cancer
Quarfloxin MIA PaCa-2 human > 125 mm3 5; i.v. 59% tumour shrinkage 35 58
pancreatic cancer
RHPS4 UXF1138L human uterine 5 5 mm 5; oral 30% tumour shrinkage 28 33
carcinoma
RHPS4 M14, LP, LM melanoma 300350 mg 10; i.p. 4051% tumour weight 15 56
reduction
RHPS4b CG5 breast carcinoma 300 mg 15; i.v. 75% tumour shrinkage 30 35
a b
Animals were initially treated with cyclophosphamide to minimize tumour burden. RHPS4 was reported to have an antitumour effect in a
number of other tumour types in this study.

appears able to penetrate signicant tumour masses telomere level should not be ruled out. In vivo data on
(Table 1), in accord with its single net positive charge the recently described quadruplex-binding uoroquino-
combined with the relatively small size of this mole- lone derivative Quaroxin (CX-3543) is included. It is
cule. currently in clinical trials so its pharmacological prole
Data on two other quadruplex-binding ligands have has relevance to other quadruplex ligands. This agent
also been included. The porphyrin compound was initially suggested to be targeting a c-myc pro-
TMPyP4, which does bind with high afnity to a wide moter quadruplex, but is now believed to function by
range of quadruplex nucleic acids, albeit with low selectively disrupting nucleolin rDNA quadruplex
selectivity, has been reported to show anticancer activ- complexes [58]. It does not show the cellular behaviour
ity in MX-1 mammary tumours and PC-3 human pros- characteristic of a telomere targeting agent.
tate carcinomas [57]. Although quadruplexes in the It is encouraging for future clinical applications that
promoter region of the c-myc oncogene have been several G-quadruplex ligands show in vivo synergistic
suggested as a target for this compound, it is also an activity (Table 2) with conventional cytotoxic agents,
established telomerase inhibitor, so action at the such as cis-platinum, taxol and camptothecin deriva-

Table 2. In vivo studies of quadruplex-binding ligands in combination with established anticancer drugs. Tumour responses have been
estimated from survival curves and other available data. Studies in combination with established anticancer drugs.

Initial tumour Dosage


G4 ligand Xenograft model size (mgkg)1) Drug 2 Tumour response Ref

BRACO-19 A431 epidermal carcinoma 1080 mm3 2 Paclitaxel 68% tumour shrinkage 54
AS1410 A549 lung carcinoma 10 mm3 1 Cis-platinum  75% tumour shrinkage 59
RHPS4 UXF1138L human uterine 5 5 mm 5 Taxol Complete remissions 33
carcinoma
RHPS4a HCT116, HT29 colorectal 300350 mg 10 Irinotecan 80% tumour weight reduction 56
carcinomas
a
A number of other combinations, with a range of anticancer drugs, were also reported in this study.

1122 FEBS Journal 277 (2010) 11181125 2009 The Author Journal compilation 2009 FEBS
S. Neidle G-quadruplexes as cancer drug targets

tives [33,54,56,59], although the detailed mechanism of ligand telomestatin inhibits POT1 binding to telomeric
this effect remains to be established. The order in sequences in vitro and induces GFPPOT1 dissociation
which the drugs are administered appears to be an from telomeres in human cells. Cancer Res 66,
important determinant of whether a particular combi- 69086912.
nation is synergistic or antagonistic. It is also possible 10 Gomez D, Wenner T, Brassart B, Douarre C, ODon-
that quadruplex-binding ligands can have multiple ohue MF, El KV, Shin-Ya K, Morjani H, Trentesaux
quadruplex targets, which could confer therapeutic C & Riou J-F (2006) Telomestatin-induced telomere
advantage. Dual targeting has been reported for a uncapping is modulated by POT1 through G-overhang
extension in HT1080 human tumor cells. J Biol Chem
substituted naphthalene diimide, which interacts with
281, 3872138729.
quadruplexes in the promoter region of the c-kit onco-
11 Gunaratnam M, Greciano O, Martins C, Reszka AP,
gene that is disregulated in gastrointestinal cancer cells
Schultes CM, Morjani H, Riou JF & Neidle S (2007)
(inhibiting c-kit expression), and telomeric quadruplexes.
Mechanism of acridine-based telomerase inhibition
The inhibition of c-kit expression and telomerase activ-
and telomere shortening. Biochem Pharmacol 74, 679
ity take place at the ligand concentrations required to 689.
halt cell growth and proliferation [60]. 12 dAdda di Fagagna F & Reaper PM (2003) A DNA
damage checkpoint response in telomere-initiated senes-
Acknowledgement cence. Nature 426, 194198.
13 Soldatenkov VA, Vetcher AA, Duka T & Ladame S
I am grateful to Cancer Research UK for Programme (2008) First evidence of a functional interaction between
Grant support and a Professorial Fellowship, and to DNA quadruplexes and poly(ADP-ribose) polymerase-
my colleagues for their input to the work described in 1. ACS Chem Biol 3, 214219.
the references. 14 Paeschke K, Juranek S, Simonsson T, Hempel A,
Rhodes D & Lipps HJ (2008) Telomerase recruitment
by the telomere end binding protein-b facilitates
References G-quadruplex DNA unfolding in ciliates. Nat Struct
1 Moyzis RK, Buckingham JM, Cram LS, Dani M, Mol Biol 15, 598604.
Deaven LL, Jones MD, Meyne J, Ratliff RL & Wu 15 Lipps HJ & Rhodes D (2009) G-quadruplex structures:
JRA (1988) Highly conserved repetitive DNA sequence, in vivo evidence and function. Trends Cell Biol 19, 414
(TTAGGG)n, present at the telomeres of human chro- 422.
mosomes. Proc Natl Acad Sci USA 85, 66226626. 16 Kim NW, Piatyszek MA, Prowse KR, Harley CB, West
2 Blackburn EH (2001) Switching and signaling at the MD, Ho PL, Coviello GM, Wright WE, Weinrich SL
telomere. Cell 106, 661673. & Shay JW (1994) Specic association of human telo-
3 Stewart SA & Weinberg RA (2006) Telomeres: cancer merase activity with immortal cells and cancer. Science
to human aging. Annu Rev Cell Dev Biol 22, 531557. 266, 20112015.
4 dAdda di Fagagna F, Teo SH & Jackson SP (2004) 17 Shay JW & Wright WE (2006) Telomerase therapeutics
Functional links between telomeres and proteins of the for cancer: challenges and new directions. Nat Rev Drug
DNA-damage response. Genes Dev 18, 17811799. Discov 5, 577584.
5 Wright WE, Tesmer VM, Huffman KE, Levene SD & 18 de Cian A, Lacroix L, Douarre C, Temime-Smaali N,
Shay JW (1977) Normal human chromosomes have Trentesaux C, Riou J-F & Mergny J-L (2008)
long G-rich telomeric overhangs at one end. Genes Dev Targeting telomeres and telomerase. Biochimie 90,
11, 28012809. 131155.
6 Zaug AJ, Podell ER & Cech TR (2005) Human POT1 19 Zahler AM, Williamson JR, Cech TR & Prescott DM
disrupts telomeric G-quadruplexes allowing telomerase (1991) Inhibition of telomerase by G-quartet DNA
extension in vitro. Proc Natl Acad Sci USA 102, 10864 structures. Nature 350, 718720.
10869. 20 Sun D, Thompson B, Cathers BE, Salazar M, Kerwin
7 Denchi EL & de Lange T (2007) Protection of telo- SM, Trent JO, Jenkins TC, Neidle S & Hurley LH
meres through independent control of ATM and ATR (1997) Inhibition of human telomerase by a G-quadru-
by TRF2 and POT1. Nature 448, 10681071. plex-interactive compound. J Med Chem 40, 21132116.
8 Miyoshi T, Kanoh J, Saito M & Ishikawa F (2008) 21 Monchaud D & Teulade-Fichou MP (2008) A hitch-
Fission yeast Pot1Tpp1 protects telomeres and hikers guide to G-quadruplex ligands. Org Biomol
regulates telomere length. Science 320, 13411344. Chem 6, 627636.
9 Gomez D, ODonohue MF, Wenner T, Douarre C, 22 Ou T, Lu Y, Tan J, Huang Z, Wong K & Gu L (2008)
Macadre J, Koebel P, Giraud-Panis MJ, Kaplan H, G-quadruplexes: targets in anticancer drug design.
Kolkes A, Shin-Ya K et al. (2006) The G-quadruplex ChemMedChem 3, 690713.

FEBS Journal 277 (2010) 11181125 2009 The Author Journal compilation 2009 FEBS 1123
G-quadruplexes as cancer drug targets S. Neidle

23 De Cian A, Cristofari G, Reichenbach P, De Lemos E, 35 Salvati E, Leonetti C, Rizzo A, Scarsella M, Mottolese


Monchaud D, Teulade-Fichou MP, Shin-Ya K, Lacroix L, M, Galati R, Sperduti I, Stevens MF, DIncalci M,
Lingner J & Mergny JL (2007) Re-evaluation of telomerase Blasco M et al. (2007) Telomere damage induced by the
inhibition by quadruplex ligands and their mechanisms of G-quadruplex ligand RHPS4 has an antitumor effect.
action. Proc Natl Acad Sci USA 104, 1734717352. J Clin Invest 117, 32363247.
24 Reed J, Gunaratnam M, Beltran M, Reszka AP, Vilar 36 Rodriquez R, Muller S, Yeoman JA, Trentesaux C,
R & Neidle S (2008) TRAP-LIG, a modied telomere Riou J-F & Balasubramanian S (2008) A novel small
repeat amplication protocol assay to quantitate telo- molecule that alters shelterin integrity and triggers a
merase inhibition by small molecules. Anal Biochem DNA-damage response at telomeres. J Am Chem Soc
380, 99105. 130, 1575815759.
25 Kim MY, Vankayalapati H, Shin-Ya K, Wierzba K & 37 Incles CM, Schultes CM, Kempski H, Koehler H, Kel-
Hurley LH (2002) Telomestatin, a potent telomerase land LR & Neidle S (2004) A G-quadruplex telomere
inhibitor that interacts quite specically with the human targeting agent produces p16-associated senescence and
telomeric intramolecular G-quadruplex. J Am Chem Soc chromosomal fusions in human prostate cancer cells.
124, 20982099. Mol Cancer Ther 3, 12011206.
26 Moorhouse AD, Santos AM, Gunaratnam M, Moore 38 Hemann MT, Strong MA, Hao LY & Greider CW
M, Neidle S & Moses JE (2006) Stabilization of G- (2001) The shortest telomere, not average telomere
quadruplex DNA by highly selective ligands via click length, is critical for cell viability and chromosome
chemistry. J Am Chem Soc 128, 1597215973. stability. Cell 107, 6777.
27 Drewe WC, Nanjunda R, Gunaratnam M, Beltran M, 39 Londono-Vallejo JA (2008) Telomere instability and
Parkinson GN, Reszka AP, Wilson WD & Neidle S cancer. Biochimie 90, 7382.
(2008) Rational design of substituted diarylureas: a 40 Arnoult N, Shin-Ya K & Londono-Vallejo JA (2008)
scaffold for binding to G-quadruplex motifs. J Med Studying telomere replication by Q-CO-FISH: the effect
Chem 51, 77517767. of telomestatin, a potent G-quadruplex ligand. Cytoge-
28 Rahman KM, Reszka AP, Gunaratnam M, Haider SM, net Genome Res 122, 229236.
Howard PW, Fox KR, Neidle S & Thurston DE (2009) 41 Tauchi T, Shin-ya K, Sashida G, Sumi M, Okabe S,
Biaryl polyamides as a new class of DNA quadruplex- Ohyashiki JH & Ohyashiki K (2006) Telomerase inhibi-
binding ligands. Chem Commun 40974099. tion with a novel G-quadruplex-interactive agent, telo-
29 Dash J, Shirude PS & Balasubramanian S (2008) mestatin: in vitro and in vivo studies in acute leukemia.
G-quadruplex recognition by bis-indole carboxamides. Oncogene 25, 57195725.
Chem Commun 30553057. 42 Fedoroff OY, Salazar M, Han H, Chemeris VV,
30 Kelland LR (2007) Targeting the limitless replicative Kerwin SM & Hurley LH (1998) NMR-based model of
potential of cancer: the telomerase telomere pathway. a telomerase-inhibiting compound bound to G-quad-
Clin Cancer Res 13, 49604963. ruplex DNA. Biochemistry 37, 1236712374.
31 Oganesian L & Bryan TM (2007) Physiological rele- 43 Haider SM, Parkinson GN & Neidle S (2003) Structure
vance of telomeric G-quadruplex formation: a potential of a G-quadruplexligand complex. J Mol Biol 326,
drug target. BioEssays 29, 155165. 117125.
32 Burger AM, Dai F, Schultes CM, Reszka AP, Moore 44 Gavathiotis E, Heald RA, Stevens MF & Searle MS
MJ, Double JA & Neidle S (2005) The G-quadruplex- (2003) Drug recognition and stabilisation of the paral-
interactive molecule BRACO-19 inhibits tumor growth, lel-stranded DNA quadruplex d(TTAGGGT)4 contain-
consistent with telomere targeting and interference with ing the human telomeric repeat. J Mol Biol 334, 2536.
telomerase function. Cancer Res 65, 14891496. 45 Campbell NH, Parkinson GN, Reszka AP & Neidle S
33 Phatak P, Cookson JC, Dai F, Smith V, Gartenhaus (2008) Structural basis of DNA quadruplex recognition
RB, Stevens MF & Burger AM (2007) Telomere uncap- by an acridine drug. J Am Chem Soc 130, 67226724.
ping by the G-quadruplex ligand RHPS4 inhibits clono- 46 Parkinson GN, Cuenca F & Neidle S (2008) Topology
genic tumour cell growth in vitro and in vivo consistent conservation and loop exibility in quadruplexdrug
with a cancer stem cell targeting mechanism. Br J recognition: crystal structures of inter- and intramolecu-
Cancer 96, 12231233. lar telomeric DNA quadruplexdrug complexes.
34 Tauchi T, Shin-Ya K, Sashida G, Sumi M, Nakajima J Mol Biol 381, 11451156.
A, Shimamoto T, Ohyashiki JH & Ohyashiki K 47 Campbell NH, Patel M, Tofa AB, Ghosh R, Parkinson
(2003) Activity of a novel G-quadruplex-interactive GN & Neidle S (2009) Selectivity in ligand recognition
telomerase inhibitor, telomestatin (SOT-095), against of G-quadruplex loops. Biochemistry 48, 16751680.
human leukemia cells: involvement of ATM-dependent 48 Huppert JL & Balasubramanian S (2007) G-quadru-
DNA damage response pathways. Oncogene 22, 5338 plexes in promoters throughout the human genome.
5347. Nucleic Acids Res 35, 406413.

1124 FEBS Journal 277 (2010) 11181125 2009 The Author Journal compilation 2009 FEBS
S. Neidle G-quadruplexes as cancer drug targets

49 Todd AK, Johnston M & Neidle S (2005) Highly preva- pounds as G-quadruplex DNA telomere targeting
lent putative quadruplex sequence motifs in human agents. Bioorg Med Chem Lett 17, 22932298.
DNA. Nucleic Acids Res 33, 29012907. 56 Leonetti C, Scarsella M, Riggio G, Rizzo A, Salvati E,
50 Huppert JL & Balasubramanian S (2005) Prevalence of DIncalci M, Staszewsky L, Frapolli R, Stevens MF,
quadruplex sequences in the human genome. Nucleic Stoppacciaro A et al. (2008) G-quadruplex ligand
Acids Res 33, 29082916. RHPS4 potentiates the antitumor activity of camptot-
51 Verma A, Halder K, Halder R, Yadav VK, Rawal P, hecins in preclinical models of solid tumors. Clin Cancer
Thakur RK, Mohd F, Sharma A & Chowdhury S Res 14, 72847291.
(2008) Genome-wide computational and expression 57 Grand CL, Han H, Munoz RM, Weitman S, Von
analyses reveal G-quadruplex DNA motifs as conserved Hoff DD, Hurley LH & Bearss DJ (2002) The cat-
cis-regulatory elements in human and related species. ionic porphyrin TMPyP4 down-regulates c-myc and
J Med Chem 51, 56415649. human telomerase reverse transcriptase expression and
52 Siddiqui-Jain A, Grand CL, Bearss DJ & Hurley LH inhibits tumor growth in vivo. Mol Cancer Ther 1,
(2002) Direct evidence for a G-quadruplex in a pro- 565573.
moter region and its targeting with a small molecule to 58 Drygin D, Siddiqui-Jain A, OBrien S, Schwaebe M,
repress c-MYC transcription. Proc Natl Acad Sci USA Lin A, Bliesath J, Ho CB, Proftt C, Trent K, Whitten
99, 1159311598. JP et al. (2009) Anticancer activity of CX-3543: a direct
53 Qin Y & Hurley LH (2008) Structures, folding patterns, inhibitor of rRNA biogenesis. Cancer Res, doi:10.1158/
and functions of intramolecular DNA G-quadruplexes 0008-5472.CAN-09-1304.
found in eukaryotic promoter regions. Biochimie 90, 59 Gunaratnam M, Green C, Moreira JB, Moorhouse AD,
11491171. Kelland LR, Moses JE & Neidle S (2009) G-quadruplex
54 Gowan SM, Harrison JR, Patterson L, Valenti M, compounds and cis-platin act synergistically to inhibit
Read MA, Neidle S & Kelland LR (2002) A G-quadru- cancer cell growth in vitro and in vivo. Biochem Phar-
plex-interactive potent small-molecule inhibitor of telo- macol 78, 115122.
merase exhibiting in vitro and in vivo antitumor activity. 60 Gunaratnam M, Swank S, Haider SM, Galesa K, Res-
Mol Pharmacol 61, 11541162. zka AP, Beltran M, Cuenca F, Fletcher JA & Neidle S
55 Martins C, Gunaratnam M, Stuart J, Makwana V, (2009) Targeting human gastrointestinal stromal tumor
Greciano O, Reszka AP, Kelland LR & Neidle S (2007) cells with a quadruplex-binding small molecule. J Med
Structure-based design of benzylamino-acridine com- Chem 52, 37743783.

FEBS Journal 277 (2010) 11181125 2009 The Author Journal compilation 2009 FEBS 1125

Das könnte Ihnen auch gefallen