401
|
Long S, Argyle DJ, Gault EA, Nasir L. Inhibition of telomerase in canine cancer cells following telomestatin treatment. Vet Comp Oncol 2007; 5:99-107. [DOI: 10.1111/j.1476-5829.2006.00118.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
402
|
Hounsou C, Guittat L, Monchaud D, Jourdan M, Saettel N, Mergny JL, Teulade-Fichou MP. G-Quadruplex Recognition by Quinacridines: a SAR, NMR, and Biological Study. ChemMedChem 2007; 2:655-66. [PMID: 17385760 DOI: 10.1002/cmdc.200600286] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The synthesis of a novel group of quinacridine-based ligands (MMQs) is described along with an evaluation of their G-quadruplex binding properties. A set of biophysical assays was applied to characterize their interaction with DNA quadruplexes: FRET-melting experiments and equilibrium microdialysis were used to evaluate their quadruplex affinity and their ability to discriminate quadruplexes across a broad panel of DNA structures. All data collected support the proposed model of interaction of these compounds with G-quadruplexes, which is furthermore confirmed by a solution structure determined by 2D NMR experiments. Finally, the activity of the MMQ series against tumor cell growth is reported, and the data support the potential of quadruplex-interactive compounds for use in anticancer approaches.
Collapse
Affiliation(s)
- Candide Hounsou
- Laboratoire de Chimie des Interactions Moléculaires, CNRS UPR 285, Collège de France, 11, place Marcellin Berthelot, 75005 Paris, France
| | | | | | | | | | | | | |
Collapse
|
403
|
Goto Y, Hagihara S, Hagihara M, Nakatani K. Small-Molecule Binding to the Nonquadruplex Form of the Human Telomeric Sequence. Chembiochem 2007; 8:723-6. [PMID: 17393547 DOI: 10.1002/cbic.200600564] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yuki Goto
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | | | | | | |
Collapse
|
404
|
Franceschin M, Pascucci E, Alvino A, D'Ambrosio D, Bianco A, Ortaggi G, Savino M. New highly hydrosoluble and not self-aggregated perylene derivatives with three and four polar side-chains as G-quadruplex telomere targeting agents and telomerase inhibitors. Bioorg Med Chem Lett 2007; 17:2515-22. [PMID: 17317176 DOI: 10.1016/j.bmcl.2007.02.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 02/07/2007] [Accepted: 02/07/2007] [Indexed: 10/23/2022]
Abstract
Four new perylene derivatives with three and four basic side-chains are reported here as G-quadruplex interactive compounds. The new perylene derivatives are readily soluble in water and not self-aggregated, in contrast to what happens with the previously reported two side-chain perylene derivatives. All four compounds are able to induce the G-quadruplex and to inhibit 50% of telomerase activity at about 5 microM concentration, showing a similar efficiency with respect to each other. Molecular modelling studies are presented to try to explain these findings.
Collapse
Affiliation(s)
- Marco Franceschin
- Dipartimento di Chimica, Università di Roma La Sapienza, Piazzale A. Moro 5, 00185 Roma, Italy.
| | | | | | | | | | | | | |
Collapse
|
405
|
Phatak P, Cookson JC, Dai F, Smith V, Gartenhaus RB, Stevens MFG, Burger AM. Telomere uncapping by the G-quadruplex ligand RHPS4 inhibits clonogenic tumour cell growth in vitro and in vivo consistent with a cancer stem cell targeting mechanism. Br J Cancer 2007; 96:1223-33. [PMID: 17406367 PMCID: PMC2360152 DOI: 10.1038/sj.bjc.6603691] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Revised: 02/14/2007] [Accepted: 02/16/2007] [Indexed: 01/19/2023] Open
Abstract
The pentacyclic acridinium methosulfate salt RHPS4 induces the 3'single-stranded guanine-rich telomeric overhang to fold into a G-quadruplex structure. Stabilisation of the latter is incompatible with an attachment of telomerase to the telomere and thus G-quadruplex ligands can effectively inhibit both the catalytic and capping functions of telomerase. In this study, we examined mechanisms underlying telomere uncapping by RHPS4 in uterus carcinoma cells (UXF1138L) with short telomeres and compared the susceptibility of bulk and clonogenic cancer cells to the G-quadruplex ligand. We show that treatment of UXF1138L cells with RHPS4 leads to the displacement of the telomerase catalytic subunit (hTERT) from the nucleus, induction of telomere-initiated DNA-damage signalling and chromosome fusions. We further report that RHPS4 is more potent against cancer cells that grow as colonies in soft agar than cells growing as monolayers. Human cord blood and HEK293T embryonic kidney cell colony forming units, however, were more resistant to RHPS4. RHPS4-treated UXF1138L xenografts had a decreased clonogenicity, showed loss of nuclear hTERT expression and an induction of mitotic abnormalities compared with controls. Although single-agent RHPS4 had limited in vivo efficacy, a combination of RHPS4 with the mitotic spindle poison Taxol caused tumour remissions and further enhancement of telomere dysfunction.
Collapse
Affiliation(s)
- P Phatak
- Department of Pharmacology and Experimental Therapeutics, Marlene and Stewart Greenebaum Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | |
Collapse
|
406
|
Martins C, Gunaratnam M, Stuart J, Makwana V, Greciano O, Reszka AP, Kelland LR, Neidle S. Structure-based design of benzylamino-acridine compounds as G-quadruplex DNA telomere targeting agents. Bioorg Med Chem Lett 2007; 17:2293-8. [PMID: 17276687 DOI: 10.1016/j.bmcl.2007.01.056] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2006] [Revised: 01/16/2007] [Accepted: 01/18/2007] [Indexed: 01/22/2023]
Abstract
The design, synthesis, biophysical and biochemical evaluation is presented of a new series of benzylamino-substituted acridines as G-quadruplex binding telomerase inhibitors. Replacement of the previously reported anilino substituents by benzylamino groups results in enhanced quadruplex interaction, and for one compound, superior telomerase inhibitory activity.
Collapse
Affiliation(s)
- Cristina Martins
- CRUK Biomolecular Structure Group, The School of Pharmacy, University of London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
407
|
Nasir L. Telomeres and telomerase: Biological and clinical importance in dogs. Vet J 2007; 175:155-63. [PMID: 17398127 DOI: 10.1016/j.tvjl.2007.01.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 01/25/2007] [Accepted: 01/26/2007] [Indexed: 10/23/2022]
Abstract
In recent years in human oncology the enzyme telomerase has emerged as an ideal target for cancer therapy. This has led to the assessment of telomerase in cancers in companion animals, mainly dogs and these studies confirm that in dogs, like humans, telomere maintenance by telomerase is the primary mechanism by which cancer cells overcome their mortality and extend their lifespan. This review aims to provide an introduction to the biology of telomeres and telomerase and to discuss some of the telomere/telomerase directed therapeutic methodologies currently under development which may be of benefit to the canine cancer patient.
Collapse
Affiliation(s)
- Lubna Nasir
- Institute of Comparative Medicine, University of Glasgow, Faculty of Veterinary Medicine, Bearsden Road, Glasgow G61 1QH, UK.
| |
Collapse
|
408
|
Cian AD, Mergny JL. Quadruplex ligands may act as molecular chaperones for tetramolecular quadruplex formation. Nucleic Acids Res 2007; 35:2483-93. [PMID: 17395639 PMCID: PMC1885647 DOI: 10.1093/nar/gkm098] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 02/03/2007] [Accepted: 02/03/2007] [Indexed: 11/24/2022] Open
Abstract
G-quadruplexes are a family of four-stranded DNA structures, stabilized by G-quartets, that form in the presence of monovalent cations. Efforts are currently being made to identify ligands that selectively bind to G-quadruplex motifs as these compounds may interfere with the telomere structure, telomere elongation/replication and proliferation of cancer cells. The kinetics of quadruplex-ligands interactions are poorly understood: it is not clear whether quadruplex ligands lock into the preformed structure (i.e. increase the lifetime of the structure by lowering the dissociation constant, k(off)) or whether ligands actively promote the formation of the complex and act as quadruplex chaperones by increasing the association constant, k(on). We studied the effect of a selective quadruplex ligand, a bisquinolinium pyridine dicarboxamide compound called 360A, to distinguish these two possibilities. We demonstrated that, in addition to binding to and locking into preformed quadruplexes, this molecule acted as a chaperone for tetramolecular complexes by acting on k(on). This observation has implications for in vitro and in vivo applications of quadruplexes and should be taken into account when evaluating the cellular responses to these agents.
Collapse
Affiliation(s)
| | - Jean-Louis Mergny
- Laboratoire de Biophysique, Muséum National d′Histoire Naturelle USM 503, INSERM UR 565, CNRS UMR 5153, 43 rue Cuvier, 75231 Paris cedex 05, France
| |
Collapse
|
409
|
De Cian A, Delemos E, Mergny JL, Teulade-Fichou MP, Monchaud D. Highly efficient G-quadruplex recognition by bisquinolinium compounds. J Am Chem Soc 2007; 129:1856-7. [PMID: 17260991 DOI: 10.1021/ja067352b] [Citation(s) in RCA: 343] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Anne De Cian
- Laboratoire de Chimie des Interactions Moléculaires, Collège de France, CNRS UPR285, 75005 Paris, France
| | | | | | | | | |
Collapse
|
410
|
Burger AM. Highlights in experimental therapeutics. Cancer Lett 2007; 245:11-21. [PMID: 16647200 DOI: 10.1016/j.canlet.2006.03.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Revised: 03/08/2006] [Accepted: 03/09/2006] [Indexed: 02/02/2023]
Abstract
The past two decades have seen a dramatic change in cancer treatment paradigms. Anticancer agents are no longer being developed based on empiricism and serendipity, but are now being aimed to inhibit a validated target that is relatively specific for tumours rather than normal cells. The vast majority of cancers arise from multiple genetic lesions; thus, sophisticated drug cocktails, or single drugs acting on multiple downstream targets will be needed for successful cancer therapy. Three emerging concepts that are addressing these therapeutic needs and that are key to blocking steps in tumourigenesis will be highlighted in this review: (a) attacking cancer cell immortality by targeting the telomere/telomerase complex; (b) targeting oncogene activation by inhibiting the molecular chaperone Hsp90; and (c) stabilizing tumour suppressor proteins by modulating the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Angelika M Burger
- Department of Pharmacology and Experimental Therapeutics, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
411
|
Fleisig HB, Wong JMY. Telomerase as a clinical target: Current strategies and potential applications. Exp Gerontol 2007; 42:102-12. [PMID: 16814507 DOI: 10.1016/j.exger.2006.05.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2006] [Accepted: 05/05/2006] [Indexed: 01/19/2023]
Abstract
Chromosome ends are capped by telomeres, protective DNA-protein complexes that distinguish natural ends from random DNA breaks. Telomeres erode with each successive cell division, and such divisions cease once telomeres become critically short. This proliferation limit is important as a tumor suppressive mechanism, but also contributes to the degenerative conditions associated with cellular aging. In cell types that require continuous renewal, transient expression of telomerase delays proliferation arrest by the de novo synthesis of telomere repeats. Data from our work and others' has shown that deficient telomerase activity has a negative impact on normal human physiology. In the bone marrow failure syndrome dyskeratosis congenita, telomerase enzyme deficiency leads to the premature shortening of telomeres. Premature telomere shortening most grievously affects tissues that have a rapid turnover, such as the hematopoietic and epithelial compartments. In the most severe cases, compromised renewal of hematopoietic stem cells leads to bone marrow failure and premature death. Telomerase activation/replacement shows potential as a therapy for telomere maintenance deficiency syndromes, and in tissue engineering for the degenerative conditions that are associated with normal aging. Conversely, clinical researchers are developing telomerase inhibition therapies to treat tumors, which overcome the short-telomere barrier to unrestricted proliferation by over-expressing telomerase.
Collapse
Affiliation(s)
- Helen B Fleisig
- Division of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | | |
Collapse
|
412
|
Huang HS, Chen IB, Huang KF, Lu WC, Shieh FY, Huang YY, Huang FC, Lin JJ. Synthesis and Human Telomerase Inhibition of a Series of Regioisomeric Disubstituted Amidoanthraquinones. Chem Pharm Bull (Tokyo) 2007; 55:284-92. [PMID: 17268103 DOI: 10.1248/cpb.55.284] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Telomerase is the enzymatic activity that maintains the ends of eukaryotic chromosomes. Telomerase activity is detected in most tumor cells whereas it is low or undetectable in most normal somatic cells. Expression of the telomerase catalytic component, the human telomerase reverse transcriptase (hTERT), is believed to be controlled primarily at the level of transcription. Because of this selective expression property of telomerase, it has been touted as a specific target for antitumor chemotherapeutics. However, a concern for the applicability of telomerase inhibitors is that they require a long lag time for telomeres to be shortened to critical length before cancer cells stop proliferating. Here we investigate telomerase inhibitory, cytotoxicity and the hTERT repressing effects on a number of synthesized 2,6-diamidoanthraquinones and 1,5-diamidoanthraquinones as compared to their disubstituted homologues. We found that several of the 1,5-diamidoanthraquinones and 2,6-diamidoanthraquinones inhibited telomerase activity effectively with IC50 at the sub-micro to micro molar range and caused acute cytotoxicity to cancer cells with EC50 similar or better than that of mitoxantrone. Particularly, 2,6-diamidoanthraquinone with 2-ethylaminoacetamido side chains 33, even though not affecting cell proliferation, showed to be endowed with a strong telomerase effect, probably related to a marked stabilization of the G-quadruplex-binding structure. The results suggested that these compounds caused multiple effects to cancer cells. More significantly, they overcome the long lag period problem of classical telomerase inhibitors that they are also potent cytotoxic agents. These results greatly expand the potential of tricyclic anthraquinone pharmacophore in preventive and/or curative therapy.
Collapse
Affiliation(s)
- Hsu-Shan Huang
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
413
|
Oganesian L, Bryan TM. Physiological relevance of telomeric G-quadruplex formation: a potential drug target. Bioessays 2007; 29:155-65. [PMID: 17226803 DOI: 10.1002/bies.20523] [Citation(s) in RCA: 251] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The concept of a G-quartet, a unique structural arrangement intrinsic to guanine-rich DNA, was first introduced by Gellert and colleagues over 40 years ago. For decades, it has been uncertain whether the G-quartet and the structure that it gives rise to, the G-quadruplex, are purely in vitro phenomena. Nevertheless, the presence of signature G-rich motifs in the eukaryotic genome, and the plethora of proteins that bind to, modify or resolve this nucleic acid structure in vitro have provided circumstantial evidence for its physiological relevance. More recently, direct visualisation of G-quadruplex DNA at native telomeres was achieved, bolstering the evidence for its existence in the cell. Furthermore, G-quadruplex folded telomeric DNA has been found to perturb telomere function and to impede the action of telomerase, an enzyme overexpressed in >85% of human cancers, hence opening up a novel avenue for cancer therapy in the form of G-quadruplex stabilising agents.
Collapse
Affiliation(s)
- Liana Oganesian
- Children's Medical Research Institute, Westmead, Sydney Australia
| | | |
Collapse
|
414
|
Reed JE, Neidle S, Vilar R. Stabilisation of human telomeric quadruplex DNA and inhibition of telomerase by a platinum–phenanthroline complex. Chem Commun (Camb) 2007:4366-8. [DOI: 10.1039/b709898g] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
415
|
Sasaki S, Bando T, Minoshima M, Shimizu T, Shinohara KI, Takaoka T, Sugiyama H. Sequence-Specific Alkylation of Double-Strand Human Telomere Repeat Sequence by Pyrrole-Imidazole Polyamides with Indole Linkers. J Am Chem Soc 2006; 128:12162-8. [PMID: 16967966 DOI: 10.1021/ja0626584] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We designed and synthesized pyrrole (Py)-imidazole (Im) hairpin polyamide 1-(chloromethyl)-5-hydroxy-1,2-dihydro-3H-benz[e]indole (seco-CBI) conjugates 1 and 2, which target both strands of the double-stranded region of the human telomere repeat sequences, 5'-d(TTAGGG)(n)-3'/5'-d(CCCTAA)(n)-3'. High-resolution denaturing polyacrylamide gel electrophoresis demonstrated that conjugates 1 and 2 alkylated DNA at the 3' A of 5'-ACCCTA-3' and 5'-AGGGTTA-3', respectively. Cytotoxicities of conjugates 1 and 2 were evaluated using 39 human cancer cell lines; averages of log IC(50) values for conjugates 1 and 2 were -6.96 (110 nM) and -7.24 (57.5 nM), respectively. Conjugates 1 and 2 have potential as antitumor drugs capable of targeting telomere repeat sequence.
Collapse
Affiliation(s)
- Shunta Sasaki
- Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo, Kyoto, 606-8502, Japan
| | | | | | | | | | | | | |
Collapse
|
416
|
Li W, Zhang M, Zhang JL, Li HQ, Zhang XC, Sun Q, Qiu CM. Interactions of daidzin with intramolecular G-quadruplex. FEBS Lett 2006; 580:4905-10. [PMID: 16920104 DOI: 10.1016/j.febslet.2006.08.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 07/06/2006] [Accepted: 08/01/2006] [Indexed: 02/01/2023]
Abstract
The potential interaction of daidzin, an ingredient of soy isoflavones, with human telomeric antiparallel G-quadruplex dAG(3)(T(2)AG(3))(3) was studied using ESI-MS, PAGE, CD and molecular simulation. Experimental studies indicated that daidzin molecules interacted with dAG(3)(T(2)AG(3))(3) and formed DNA-daidzin complex with the stoichiometric ratio of 1:1 and 1:2. The transition temperature of the G-quadruplex increased at higher ratio of daidzin to DNA. Under molecular crowding conditions the interactions between daidzin and the G-quadruplex become much stronger. Combining computational simulation and experimental results, it is demonstrated that the dAG(3)(T(2)AG(3))(3)/daidzin complex with a stoichiometric ratio of 1:1 is stabilized through the pi-pi conjugacy interactions and hydrogen bondings between daidzin and the bases of G-quadruplex. This work provides guidance not only on exploring the molecular anti-cancer mechanism of dietary isoflavones, but also searching small natural products as promising anticancer candidates that can inhibit telomerase activity.
Collapse
Affiliation(s)
- Wei Li
- Department of Chemical Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | | | | | | | | | | | | |
Collapse
|
417
|
Eddy J, Maizels N. Gene function correlates with potential for G4 DNA formation in the human genome. Nucleic Acids Res 2006; 34:3887-96. [PMID: 16914419 PMCID: PMC1557811 DOI: 10.1093/nar/gkl529] [Citation(s) in RCA: 397] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
G-rich genomic regions can form G4 DNA upon transcription or replication. We have quantified the potential for G4 DNA formation (G4P) of the 16 654 genes in the human RefSeq database, and then correlated gene function with G4P. We have found that very low and very high G4P correlates with specific functional classes of genes. Notably, tumor suppressor genes have very low G4P and proto-oncogenes have very high G4P. G4P of these genes is evenly distributed between exons and introns, and it does not reflect enrichment for CpG islands or local chromosomal environment. These results show that genomic structure undergoes selection based on gene function. Selection based on G4P could promote genomic stability (or instability) of specific classes of genes; or reflect mechanisms for global regulation of gene expression.
Collapse
Affiliation(s)
- Johanna Eddy
- Molecular and Cellular Biology Graduate Program, University of Washington School of Medicine1959 NE Pacific Street, Seattle, WA 98195-7650, USA
| | - Nancy Maizels
- Molecular and Cellular Biology Graduate Program, University of Washington School of Medicine1959 NE Pacific Street, Seattle, WA 98195-7650, USA
- Department of Immunology, University of Washington School of Medicine1959 NE Pacific Street, Seattle, WA 98195-7650, USA
- Department of Biochemistry, University of Washington School of Medicine1959 NE Pacific Street, Seattle, WA 98195-7650, USA
- To whom correspondence should be addressed. Tel: +1 206 221 6876; Fax: +1 206 221 6781;
| |
Collapse
|
418
|
Cerone MA, Londoño-Vallejo JA, Autexier C. Telomerase inhibition enhances the response to anticancer drug treatment in human breast cancer cells. Mol Cancer Ther 2006; 5:1669-75. [PMID: 16891452 DOI: 10.1158/1535-7163.mct-06-0033] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Breast cancer is the most common malignancy among women. Current therapies for breast tumors are based on the use of chemotherapeutic drugs that are quite toxic for the patients and often result in resistance. Telomerase is up-regulated in 95% of breast carcinomas but not in adjacent normal tissues. Therefore, it represents a very promising target for anticancer therapies. Unfortunately, the antiproliferative effects of telomerase inhibition require extensive telomere shortening before they are fully present. Combining telomerase inhibition with common chemotherapeutic drugs can be used to reduce this lag phase and induce tumor cell death more effectively. Few studies have analyzed the effects of telomerase inhibition in combination with anticancer drugs in breast cancer cells. In this study, we inhibited telomerase activity in two breast cancer cell lines using a dominant-negative human telomerase reverse transcriptase and analyzed cell viability after treatment with different anticancer compounds. We found that dominant-negative human telomerase reverse transcriptase efficiently inhibits telomerase activity and causes telomere shortening over time. Moreover, cells in which telomerase was suppressed were more sensitive to anticancer agents independently of their mechanism of action and this sensitization was dependent on the presence of shorter telomeres. Altogether, our data show that blocking telomere length maintenance in combination with anticancer drugs can be used as an effective way to induce death of breast cancer cells.
Collapse
Affiliation(s)
- Maria Antonietta Cerone
- Bloomfield Centre for Research in Aging, Lady Davis Institute, Sir Mortimer B. Davis Jewish General Hospital, 3755 Cote Ste Catherine, Montreal, Quebec, Canada H3T 1E2
| | | | | |
Collapse
|
419
|
White EW, Tanious F, Ismail MA, Reszka AP, Neidle S, Boykin DW, Wilson WD. Structure-specific recognition of quadruplex DNA by organic cations: influence of shape, substituents and charge. Biophys Chem 2006; 126:140-53. [PMID: 16831507 DOI: 10.1016/j.bpc.2006.06.006] [Citation(s) in RCA: 158] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Revised: 06/13/2006] [Accepted: 06/13/2006] [Indexed: 01/17/2023]
Abstract
Combining structure-specific recognition of nucleic acids with limited sequence reading is a promising method to reduce the size of the recognition unit required to achieve the necessary selectivity and binding affinity to control function. It has been demonstrated recently that G-quadruplex DNA structures can be targeted by organic cations in a structure-specific manner. Structural targets of quadruplexes include the planar end surfaces of the G-tetrad stacked columns and four grooves. These provide different geometries and functional groups relative to duplex DNA. We have used surface plasmon resonance and isothermal titration calorimetry to show that binding affinity and selectivity of a series of quadruplex end-stacking molecules to human telomeric DNA are sensitive to compound shape as well as substituent type and position. ITC results indicate that binding is largely enthalpy driven. Circular dichroism was also used to identify a group of structurally related compounds that selectively target quadruplex grooves.
Collapse
Affiliation(s)
- Elizabeth W White
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | | | | | | | | | | | | |
Collapse
|
420
|
Phan AT, Kuryavyi V, Patel DJ. DNA architecture: from G to Z. Curr Opin Struct Biol 2006; 16:288-98. [PMID: 16714104 PMCID: PMC4689308 DOI: 10.1016/j.sbi.2006.05.011] [Citation(s) in RCA: 214] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 04/10/2006] [Accepted: 05/10/2006] [Indexed: 12/27/2022]
Abstract
G-quadruplexes and Z-DNA are two important non-B forms of DNA architecture. Results on novel structural elements, folding and unfolding kinetics, and interactions with small molecules and proteins have been reported recently for these forms. These results will enhance our understanding of the biology of these structures and provide a platform for drug design.
Collapse
Affiliation(s)
- Anh Tuân Phan
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| | | | | |
Collapse
|
421
|
Guzmán MR, Liquier J, Brahmachari SK, Taillandier E. Characterization of parallel and antiparallel G-tetraplex structures by vibrational spectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2006; 64:495-503. [PMID: 16384730 DOI: 10.1016/j.saa.2005.07.049] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Accepted: 07/25/2005] [Indexed: 05/05/2023]
Abstract
A series of G-rich oligonucleotides able to form tetraplexes has been studied by FTIR spectroscopy. Characteristic markers of the formation of guanine tetrads are given. Moreover, we propose a new marker discriminating between parallel and antiparallel tetraplexes: the position of the C6O6 guanine carbonyl stretching vibration. In intermolecular parallel tetrameric structures formed by four separate strands this absorption is observed at 1693 cm-1 while for antiparallel tetrameric structures, either intramolecular or formed by dimerization of hairpins, this vibrational mode is observed at 1682 cm-1. These shifts to higher wavenumbers, when compared to the position of a free guanine C6O6 carbonyl stretching vibration observed at 1666 cm-1(Deltanu=27 cm-1 for parallel tetraplexes and Deltanu=16 cm-1 for antiparallel tetraplexes) reflect different strand orientations in the structures. This marker has been used to evidence the possibility of an antiparallel-parallel tetraplex reorganization for Oxytricha nova d(G4T4G4) and d((G4T4)3G4) and human d(G3T2AG3) telomeric sequences induced by Na+/K+ or Na+/Ca2+ ion exchange. Formation of the guanine tetrads, characterization of the phosphate geometries and of the sugar conformations have also been obtained by FTIR for the different tetraplexes.
Collapse
Affiliation(s)
- M Romero Guzmán
- Laboratoire BioMoCeTi, UMR CNRS 7033, Université Paris 13, 74, rue Marcel Cachin, F93017 Bobigny Cedex, France
| | | | | | | |
Collapse
|
422
|
Taetz S, Baldes C, Mürdter TE, Kleideiter E, Piotrowska K, Bock U, Haltner-Ukomadu E, Mueller J, Huwer H, Schaefer UF, Klotz U, Lehr CM. Biopharmaceutical Characterization of the Telomerase Inhibitor BRACO19. Pharm Res 2006; 23:1031-7. [PMID: 16715394 DOI: 10.1007/s11095-006-0026-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2005] [Accepted: 01/11/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE To characterize the telomerase inhibitor and G-quadruplex stabilizing substance 9-[4-(N,N-dimethylamino)phenylamino]-3,6-bis (3-pyrrolodino-propionamido) acridine x 3HCl (BRACO19) in terms of biopharmaceutical properties such as solubility, protein binding, interaction with membrane lipids, cytotoxicity, and permeability across pulmonary epithelial cells. METHODS Protein binding and interaction with membrane lipids were investigated by two high-performance liquid chromatography methods with immobilized human serum albumin and immobilized phosphatidylcholine, respectively. Cytotoxicity (methyl-thiazolyl-tetrazolium assay) and transport studies were performed with the bronchial cell lines 16HBE14o- and Calu-3, primary human alveolar epithelial cells, and the intestinal cell line Caco-2. Transport experiments were also done in the presence of cyclosporin A (10 microM) and tetraethylammonium chloride (5 mM) and at low temperature (4 degrees C). RESULTS BRACO19 has good solubility of at least 2 mg/mL in water and in physiological buffers of pH 7.4 and below. Protein binding to human serum albumin was 38%. No interaction with membrane lipids could be found. Cytotoxicity in 16HBE14o-, Calu-3, and human alveolar epithelial cells was in the range of IC50 = 3.5 to 13.5 microM. Caco-2 cells were not affected at concentrations up to 50 microM. No transport of BRACO19 was detected across either cell monolayer in absorptive direction. In secretory direction, permeability was very low, with P (app) values in the range of 0.25 x 10(-7) to 0.98 x 10(-7) cm/s for all epithelial cell cultures tested. The transport was not influenced by cyclosporin A or tetraethylammonium chloride or at 4 degrees C, indicating that no efflux/influx systems or active transport are involved. CONCLUSIONS From these results, we conclude that the very poor permeability of BRACO19 is its main biopharmaceutical limitation. Further applications will require a suitable formulation to warrant adequate delivery across cellular barriers.
Collapse
Affiliation(s)
- S Taetz
- Biopharmaceutics and Pharmaceutical Technology, Saarland University, P.O. Box 15 11 50, 66041 Saarbrücken, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
423
|
Reed JE, Arnal AA, Neidle S, Vilar R. Stabilization of G-Quadruplex DNA and Inhibition of Telomerase Activity by Square-Planar Nickel(II) Complexes. J Am Chem Soc 2006; 128:5992-3. [PMID: 16669641 DOI: 10.1021/ja058509n] [Citation(s) in RCA: 265] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Two new alkylamine-substituted nickel(II)-salphen complexes have been prepared and their interactions with DNA investigated. FRET studies have shown that these complexes have a remarkable ability to stabilize G-quadruplex DNA. Furthermore, TRAP/Taq assays have shown that these complexes inhibit telomerase at low micromolar concentrations.
Collapse
Affiliation(s)
- Julie E Reed
- Department of Chemistry, Imperial College London, South Kensington, UK
| | | | | | | |
Collapse
|
424
|
Zhou JM, Zhu XF, Lu YJ, Deng R, Huang ZS, Mei YP, Wang Y, Huang WL, Liu ZC, Gu LQ, Zeng YX. Senescence and telomere shortening induced by novel potent G-quadruplex interactive agents, quindoline derivatives, in human cancer cell lines. Oncogene 2006; 25:503-11. [PMID: 16170347 DOI: 10.1038/sj.onc.1209067] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Agents stabilizing G-quadruplexes have the potential to interfere with telomere replication by blocking the elongation step catalysed by telomerase or telomerase-independent mechanism and could therefore act as antitumor agents. In this study, we found that quindoline derivatives interacted preferentially with intramolecular G-quadruplex structures and were novel potent telomerase inhibitors. Treatment with quindoline derivatives reproducibly inhibited telomerase activity in human leukemia K562 cells and colon cancer SW620 cells. N'-(10H-Indolo [3,2-b] quinolin-11-yl)-N, N-dimethyl-propane-1,3-diamine (SYUIQ-5), (one of quindoline derivatives), when added to K562 and SW620 cell culture at nonacute cytotoxic concentrations, increased time of population doublings of K562 and SW620 cells, induced a marked cessation in cell growth and cellular senescence phenotype after 35 and 18 days, respectively. Growth cessation was accompanied by a shortening of telomere length, and induction of p16, p21 and p27 protein expression. However, another compound SYUIQ-7 with greater IC(50) for telomerase had no obvious cellular effect in nonacute cytotoxic concentrations. These results indicate that quindoline derivatives as novel potent G-quadruplex interactive agents induce senescence and telomere shortening in cancer cells and therefore are promising agents for cancer treatment.
Collapse
Affiliation(s)
- J-M Zhou
- State Key Laboratory of Oncology in Southern China, Cancer Center, Sun Yat-sen University, Guangzhou
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
425
|
Olaussen KA, Dubrana K, Domont J, Spano JP, Sabatier L, Soria JC. Telomeres and telomerase as targets for anticancer drug development. Crit Rev Oncol Hematol 2006; 57:191-214. [PMID: 16469501 DOI: 10.1016/j.critrevonc.2005.08.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2005] [Revised: 08/10/2005] [Accepted: 08/11/2005] [Indexed: 12/15/2022] Open
Abstract
In most human cancers, the telomere erosion problem has been bypassed through the activation of a telomere maintenance system (usually activation of telomerase). Therefore, telomere and telomerase are attractive targets for anti-cancer therapeutic interventions. Here, we review a large panel of strategies that have been explored to date, from small inhibitors of the catalytic sub-unit of telomerase to anti-telomerase immunotherapy and gene therapy. The many positive results that are reported from anti-telomere/telomerase assays suggest a prudent optimism for a possible clinical application in a close future. However, we discuss some of the main limits for these approaches of antitumour drug development and why significant work remains before a clinically useful drug can be proposed to patients.
Collapse
Affiliation(s)
- Ken André Olaussen
- Laboratory of Radiobiology and Oncology, DSV/DRR/LRO, CEA, Fontenay aux Roses, France
| | | | | | | | | | | |
Collapse
|
426
|
Kaiser M, De Cian A, Sainlos M, Renner C, Mergny JL, Teulade-Fichou MP. Neomycin-capped aromatic platforms: quadruplex DNA recognition and telomerase inhibition. Org Biomol Chem 2006; 4:1049-57. [PMID: 16525549 DOI: 10.1039/b516378a] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A series of aminoglycoside-capped macrocyclic structures has been prepared using intramolecular bis-tethering of neomycin on three aromatic platforms (phenanthroline, acridine, quinacridine). Based on NMR and calculations studies, it was found that the cyclic compounds adopt a highly flexible structure without conformational restriction of the aminoglycoside moiety. FRET-melting stabilization measurements showed that the series displays moderate to high affinity for the G4-conformation of human telomeric repeats, this effect being correlated with the size of the aromatic moiety. In addition, a FRET competition assay evidenced the poor binding ability of all macrocycles for duplex DNA and a clear binding preference for loop-containing intramolecular G4 structures compared to tetramolecular parallel G4 DNA. Finally, TRAP experiments demonstrated that the best G4-binder (quinacridine ) is also a potent and selective telomerase inhibitor with an IC(50) in the submicromolar range (200 nM).
Collapse
Affiliation(s)
- Markus Kaiser
- Laboratoire de Chimie des Interactions Moléculaires, Collège de France, CNRS UPR 285, 11, place Marcelin Berthelot, 75005 Paris, France
| | | | | | | | | | | |
Collapse
|
427
|
Moore MJB, Cuenca F, Searcey M, Neidle S. Synthesis of distamycin A polyamides targeting G-quadruplex DNA. Org Biomol Chem 2006; 4:3479-88. [PMID: 17036143 DOI: 10.1039/b607707b] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A number of amide-linked oligopyrroles based on distamycin molecules have been synthesized by solid-state methods, and their interactions with a human intramolecular G-quadruplex have been measured by a melting procedure. Several of these molecules show an enhanced ratio of quadruplex vs. duplex DNA binding compared to distamycin itself, including one with a 2,5-disubstituted pyrrole group. Quadruplex affinity increases with the number of pyrrole groups, and it is suggested that this is consistent with a mixed groove/G-quartet stacking binding mode.
Collapse
Affiliation(s)
- Michael J B Moore
- Cancer Research UK Biomolecular Structure Group, The School of Pharmacy, University of London, 29-39 Brunswick Square, London, UK WC1N 1AX
| | | | | | | |
Collapse
|
428
|
Cookson JC, Dai F, Smith V, Heald RA, Laughton CA, Stevens MFG, Burger AM. Pharmacodynamics of the G-quadruplex-stabilizing telomerase inhibitor 3,11-difluoro-6,8,13-trimethyl-8H-quino[4,3,2-kl]acridinium methosulfate (RHPS4) in vitro: activity in human tumor cells correlates with telomere length and can be enhanced, or antagonized, with cytotoxic agents. Mol Pharmacol 2005; 68:1551-8. [PMID: 16150933 DOI: 10.1124/mol.105.013300] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Telomeric integrity is required to maintain the replicative ability of cancer cells and is a target for the G-quadruplex-stabilizing drug 3,11-difluoro-6,8,13-trimethyl-8H-quino[4,3,2-kl]acridinium methosulfate (RHPS4). We report a senescent-like growth arrest in MCF-7 breast cancer cells, within 14 to 17 days, and a reduction in telomere length (from 5.2 kilobases (kb) to 4.7 and 4.3 kb after 17 days of treatment at 0.5 and 1 microM, respectively). These effects occurred at noncytotoxic drug concentrations (doses < 1 microM over a 14-day exposure) compatible with long-term drug dosing. The telomere length of cancer cells influences their sensitivity to growth inhibition by RHPS4: mutant (mt) human telomerase reverse transcriptase (hTERT)-expressing MCF-7 cells [short telomere restriction fragment (TRF) length, 1.9 kb; IC50, 0.2 microM] were 10 times more sensitive to RHPS4 compared with wild-type (wt) hTERT-expressing, vector-transfected control cells (longer TRF-length 5.2 kb; IC50 2 microM) in the 5 day SRB assay. This relationship was corroborated in a panel of 36 human tumor xenografts grown in vitro showing a positive correlation between telomere length and growth inhibitory potency of RHPS4 (15-day clonogenic assay, r = 0.75). These observations are consistent with loss of the protective capping status of telomeres mediated by RHPS4 G-quadruplex-stabilization, thus leading to greater susceptibility of cells with shorter telomeres. In combination studies, paclitaxel (Taxol), doxorubicin (Adriamycin), and the experimental therapeutic agent 17-(allylamino)-17-demethoxygeldanamycin, which inhibits the 90-kDa heat shock protein, conferred enhanced sensitivity in RHPS4 treated MCF-7 cells, whereas the DNA-interactive temozolomide and cisplatin antagonized the action of RHPS4. Our results support the combined use of certain classes of cytotoxic anticancer agents with RHPS4 to enhance potential clinical benefit.
Collapse
Affiliation(s)
- Jennifer C Cookson
- Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
429
|
Granotier C, Pennarun G, Riou L, Hoffschir F, Gauthier LR, De Cian A, Gomez D, Mandine E, Riou JF, Mergny JL, Mailliet P, Dutrillaux B, Boussin FD. Preferential binding of a G-quadruplex ligand to human chromosome ends. Nucleic Acids Res 2005; 33:4182-90. [PMID: 16052031 PMCID: PMC1181860 DOI: 10.1093/nar/gki722] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Revised: 06/17/2005] [Accepted: 07/05/2005] [Indexed: 02/01/2023] Open
Abstract
The G-overhangs of telomeres are thought to adopt particular conformations, such as T-loops or G-quadruplexes. It has been suggested that G-quadruplex structures could be stabilized by specific ligands in a new approach to cancer treatment consisting in inhibition of telomerase, an enzyme involved in telomere maintenance and cell immortality. Although the formation of G-quadruplexes was demonstrated in vitro many years ago, it has not been definitively demonstrated in living human cells. We therefore investigated the chromosomal binding of a tritiated G-quadruplex ligand, 3H-360A (2,6-N,N'-methyl-quinolinio-3-yl)-pyridine dicarboxamide [methyl-3H]. We verified the in vitro selectivity of 3H-360A for G-quadruplex structures by equilibrium dialysis. We then showed by binding experiments with human genomic DNA that 3H-360A has a very potent selectivity toward G-quadruplex structures of the telomeric 3'-overhang. Finally, we performed autoradiography of metaphase spreads from cells cultured with 3H-360A. We found that 3H-360A was preferentially bound to chromosome terminal regions of both human normal (peripheral blood lymphocytes) and tumor cells (T98G and CEM1301). In conclusion, our results provide evidence that a specific G-quadruplex ligand interacts with the terminal ends of human chromosomes. They support the hypothesis that G-quadruplex ligands induce and/or stabilize G-quadruplex structures at telomeres of human cells.
Collapse
Affiliation(s)
| | - Gaëlle Pennarun
- LRP, DRR, CEA92265 Fontenay-aux-Roses, France
- Laboratoire de Biophysique, INSERM U565, CNRS UMR 5153, Muséum National d'Histoire Naturelle USM 50375005 Paris, France
| | - Lydia Riou
- LRP, DRR, CEA92265 Fontenay-aux-Roses, France
| | | | | | - Anne De Cian
- Laboratoire de Biophysique, INSERM U565, CNRS UMR 5153, Muséum National d'Histoire Naturelle USM 50375005 Paris, France
| | - Dennis Gomez
- Laboratoire d'Onco-Pharmacologie, JE 2428, Université de Reims Champagne-Ardenne51096, Reims, France
| | - Eliane Mandine
- Aventis Pharma SA, Centre de Recherche de Paris94403 Vitry-sur-Seine, France
| | - Jean-François Riou
- Laboratoire d'Onco-Pharmacologie, JE 2428, Université de Reims Champagne-Ardenne51096, Reims, France
| | - Jean-Louis Mergny
- Laboratoire de Biophysique, INSERM U565, CNRS UMR 5153, Muséum National d'Histoire Naturelle USM 50375005 Paris, France
| | - Patrick Mailliet
- Aventis Pharma SA, Centre de Recherche de Paris94403 Vitry-sur-Seine, France
| | | | | |
Collapse
|
430
|
Zaug AJ, Podell ER, Cech TR. Human POT1 disrupts telomeric G-quadruplexes allowing telomerase extension in vitro. Proc Natl Acad Sci U S A 2005; 102:10864-9. [PMID: 16043710 PMCID: PMC1180509 DOI: 10.1073/pnas.0504744102] [Citation(s) in RCA: 298] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The POT1 (protection of telomeres 1) protein binds the ssDNA overhangs at the ends of chromosomes in diverse eukaryotes. POT1 is essential for chromosome end-protection, as best demonstrated in fission yeast. In human cells, hPOT1 is also involved in telomere-length regulation. We now show that telomeric oligonucleotides, such as d[GGG(TTAGGG)(3)], which form intramolecular G-quadruplexes through Hoogsteen base-pairing, serve as only marginal primers for extension by recombinant human telomerase; telomerase stalls after every nucleotide addition. Addition of hPOT1 to the reaction restores the normal processive elongation pattern seen with primers that cannot form G-quadruplexes. hPOT1 does not act catalytically but, instead, forms a stoichiometric complex with the DNA, freeing its 3' tail. An antisense oligonucleotide, which base-pairs near the 5' end of the telomeric sequence, leaving a telomerase-extendable 3' tail, duplicates the effect of hPOT1 on activation of G-quadruplex primers. Thus, hPOT1 may function simply by trapping the unfolded forms of these telomeric primers in an equilibrium population. We propose an additional role for hPOT1 in telomere maintenance: disrupting G-quadruplex structures in telomeric DNA, thereby allowing proper elongation by telomerase.
Collapse
Affiliation(s)
- Arthur J Zaug
- Howard Hughes Medical Institute and Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309-0215, USA
| | | | | |
Collapse
|
431
|
Ward RJ, Autexier C. Pharmacological telomerase inhibition can sensitize drug-resistant and drug-sensitive cells to chemotherapeutic treatment. Mol Pharmacol 2005; 68:779-86. [PMID: 15939802 DOI: 10.1124/mol.105.011494] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Effective strategies to reverse or prevent chemotherapeutic resistance are required before cancer therapies can be curative. Telomerase is the ribonucleoprotein responsible for de novo synthesis and maintenance of telomeres, and its activity is predominantly observed in cancer cells. The telomerase enzyme has been successfully inhibited or inactivated to sensitize cells to cellular stresses; however, no studies have determined yet the effect of combining a pharmacological inhibitor of telomerase catalysis and traditional chemotherapeutics for the treatment of drug-sensitive or drug-resistant cancers. Here, we describe the effect of 2-[(E)-3-naphtalen-2-yl-but-2-enoylamino]-benzoic acid (BIBR1532), a small-molecule inhibitor of telomerase catalytic activity, on drug-resistant leukemia and breast cancer cells and their parental counterparts when treated in combination with chemotherapeutics. We observed that BIBR1532-treated cells show progressive telomere shortening, decreased proliferative capacity, and sensitization to chemotherapeutic treatment. These effects are telomere length-dependent, because cells insensitive to BIBR1532 or cells released from telomerase inhibition did not demonstrate changes in growth ability or drug sensitivity. Our novel observations suggest that pharmacological telomerase inhibition in combination therapy may be a valid strategy for the treatment of both drug-sensitive and drug-resistant cancers.
Collapse
Affiliation(s)
- Ryan J Ward
- Faculty of Medicine, Department of Anatomy and Cell Biology , McGill University, Montréal, Québec, Canada
| | | |
Collapse
|
432
|
|
433
|
Kelland LR. Overcoming the immortality of tumour cells by telomere and telomerase based cancer therapeutics – current status and future prospects. Eur J Cancer 2005; 41:971-9. [PMID: 15862745 DOI: 10.1016/j.ejca.2004.11.024] [Citation(s) in RCA: 166] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2004] [Accepted: 11/24/2004] [Indexed: 10/25/2022]
Abstract
A key property of malignant tumours is their immortality or limitless replicative potential. Cell replication is associated with the maintenance of telomeres and in the great majority of cases, through the reactivation of the reverse transcriptase telomerase. Targeting the telomere/telomerase machinery offers a novel and potentially broad-spectrum anticancer therapeutic strategy since telomerase is constitutively overexpressed in the vast majority of human cancers. Telomeres are also critically short in most tumours compared to normal tissues. Strategies that exploit these differences include the direct targeting of components of telomerase: the protein component hTERT or RNA component hTR. Examples of such agents include the small molecule hTERT inhibitor BIBR1532 and GRN163L, a thio-phosphoramidate oligonucleotide targeting the template region of hTR as a "template antagonist". Anti-tumour effects have been observed in both cell lines and, especially for GRN163L, in xenografted human tumours in mice. Effects, however, are largely dependent upon initial telomere length, which can result in a substantial lag before antitumour activity is observed in tumours possessing relatively long telomeres. An alternative approach is to target the telomere itself (Telomere Targeting Agents, TTAs). Several classes of small molecules have been described that induce the G-rich single-stranded overhang of telomeric DNA to fold into 4-stranded G-quadruplex structures. Such folding is incompatible with telomerase function and may induce rapid telomere uncapping. These molecules have shown potent telomerase inhibition in nanomolar concentrations in vitro and the rapid induction of senescence in cancer cells. The trisubstituted acridine based TTA, BRACO19, has demonstrated single agent activity against human tumour xenografts with anti-tumour effects apparent from only 7 days of treatment. In the near future, it is expected that lead examples from both the direct telomerase targeted agents (e.g., GRN163L) and from the distinct class of those targeting telomeres (e.g., AS1410 based on BRACO19) will enter Phase I clinical trial where clinical benefit from this class of novel drugs will be determined.
Collapse
Affiliation(s)
- Lloyd R Kelland
- Antisoma Research Laboratories, St. Georges Hospital Medical School, Cranmer Terrace, London SW17 OQS, UK.
| |
Collapse
|
434
|
Douarre C, Gomez D, Morjani H, Zahm JM, O'Donohue MF, Eddabra L, Mailliet P, Riou JF, Trentesaux C. Overexpression of Bcl-2 is associated with apoptotic resistance to the G-quadruplex ligand 12459 but is not sufficient to confer resistance to long-term senescence. Nucleic Acids Res 2005; 33:2192-203. [PMID: 15831792 PMCID: PMC1079972 DOI: 10.1093/nar/gki514] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The triazine derivative 12459 is a potent G-quadruplex interacting agent that inhibits telomerase activity. This agent induces time- and dose-dependent telomere shortening, senescence-like growth arrest and apoptosis in the human A549 tumour cell line. We show here that 12459 induces a delayed apoptosis that activates the mitochondrial pathway. A549 cell lines selected for resistance to 12459 and previously characterized for an altered hTERT expression also showed Bcl-2 overexpression. Transfection of Bcl-2 into A549 cells induced a resistance to the short-term apoptotic effect triggered by 12459, suggesting that Bcl-2 is an important determinant for the activity of 12459. In sharp contrast, the Bcl-2 overexpression was not sufficient to confer resistance to the senescence-like growth arrest induced by prolonged treatment with 12459. We also show that 12459 provokes a rapid degradation of the telomeric G-overhang in conditions that paralleled the apoptosis induction. In contrast, the G-overhang degradation was not observed when apoptosis was induced by camptothecin. Bcl-2 overexpression did not modify the G-overhang degradation, suggesting that this event is an early process uncoupled from the final apoptotic pathway.
Collapse
Affiliation(s)
- Céline Douarre
- Laboratoire d'Onco-Pharmacologie, JE 2428, UFR de Pharmacie, Université de Reims Champagne-Ardenne51 rue Cognacq-Jay, 51096 Reims, France
| | - Dennis Gomez
- Laboratoire d'Onco-Pharmacologie, JE 2428, UFR de Pharmacie, Université de Reims Champagne-Ardenne51 rue Cognacq-Jay, 51096 Reims, France
- Laboratoire de Biophysique, Muséum National d'Histoire Naturelle, USM503, INSERM U565, CNRS UMR 515343 rue Cuvier, 75231 Paris cedex 05, France
| | - Hamid Morjani
- Laboratoire d'Onco-Pharmacologie, JE 2428, UFR de Pharmacie, Université de Reims Champagne-Ardenne51 rue Cognacq-Jay, 51096 Reims, France
| | - Jean-Marie Zahm
- INSERM UMR 514, CHU Maison Blanche45 rue Cognacq-Jay, 51092 Reims, France
| | - Marie-Françoise O'Donohue
- CNRS UMR 6142, UFR de Pharmacie, Université de Reims Champagne-Ardenne51 rue Cognacq-Jay, 51096, Reims, France
| | - Lahcen Eddabra
- Laboratoire d'Onco-Pharmacologie, JE 2428, UFR de Pharmacie, Université de Reims Champagne-Ardenne51 rue Cognacq-Jay, 51096 Reims, France
| | - Patrick Mailliet
- Sanofi-Aventis SA, Département de Chimie, Centre de Recherche de Paris13 quai Jules Guesde, 94403 Vitry sur Seine, France
| | - Jean-François Riou
- Laboratoire d'Onco-Pharmacologie, JE 2428, UFR de Pharmacie, Université de Reims Champagne-Ardenne51 rue Cognacq-Jay, 51096 Reims, France
- To whom correspondence should be addressed. Tel: +33 3 26 91 80 13; Fax: +33 3 26 91 89 26;
| | - Chantal Trentesaux
- Laboratoire d'Onco-Pharmacologie, JE 2428, UFR de Pharmacie, Université de Reims Champagne-Ardenne51 rue Cognacq-Jay, 51096 Reims, France
| |
Collapse
|
435
|
Abstract
The chemical sciences are essential for the process of anticancer-drug discovery, and a range of chemical research techniques is needed to develop clinically effective drugs. Improved understanding of the cellular, molecular and genetic basis of cancer has increased the number of drug targets available. What chemical approaches are used to develop agents that target specific features of cancer cells and make these therapeutics more effective? We outline the roles that chemical synthesis and understanding of drug uptake have had in drug discovery over the past 100 years, as well as the chemical insights derived from knowledge of the three-dimensional structure of targets.
Collapse
Affiliation(s)
- Stephen Neidle
- Cancer Research UK, Biomolecular Structure Group, The School of Pharmacy, University of London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | | |
Collapse
|