Skip to main content Accessibility help
×
Hostname: page-component-84b7d79bbc-x5cpj Total loading time: 0 Render date: 2024-07-30T05:41:07.777Z Has data issue: false hasContentIssue false

37 - Telomerase: target for cancer treatment

from Part 2.4 - Molecular pathways underlying carcinogenesis: DNA repair

Published online by Cambridge University Press:  05 February 2015

Jerry W. Shay
Affiliation:
University of Texas Southwestern Medical Center, Department of Cell Biology, Dallas, TX, USA
Woodring E. Wright
Affiliation:
University of Texas Southwestern Medical Center, Department of Cell Biology, Dallas, TX, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Telomerase, a cellular reverse transcriptase that adds DNA to the ends of chromosomes, is reactivated or up-regulated in the vast majority of human advanced malignancies, and is thus an almost universal target for human cancer. Most human tumors not only express telomerase, but also have very short telomeres, whereas telomerase activity is absent or at lower levels in normal tissues, which also have longer telomeres. This relationship between activation of telomerase activity and short telomeres in human malignancies makes the inhibition of telomerase a novel target for cancer therapeutics. Importantly, the mode of action of telomerase inhibitors predicts minimal side effects on normal stem cells that can express telomerase. Here we summarize the role of telomeres and telomerase in cancer and review the current status of ongoing telomerase clinical trials. This chapter will also discuss the relationship of telomerase to cancer stem cells. Central questions remaining include: What are the key safety concerns, such as the effect of telomerase inhibitors on normal stem cells that express some telomerase? Do cancer stem cells express telomerase activity? What effect will telomerase inhibitors have if cancer stem cells are more quiescent than the bulk of the more differentiated tumor cells? Will human cancers become resistant to telomerase inhibitors?

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 442 - 451
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

de Lange, T. Mammalian telomeres. In: deLange T, Lundblad V, and Blackburn E, editors. Telomeres, 2nd edn. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press;2006:387–431.Google Scholar
von Zglinicki, T. Oxidative stress shortens telomeres. Trends in Biochemical Sciences 2002;27:339–44.CrossRefGoogle ScholarPubMed
Shay, JW, Wright, WE. Telomerase therapeutics for cancer: challenges and new directions. Nature Reviews Drug Discovery 2006;5:577–84.CrossRefGoogle ScholarPubMed
Wu, X, Amos, CI, Zhu, Y, et al. Telomere dysfunction: a potential cancer predisposition factor. Journal of the National Cancer Institute 2003;95:1211–18.CrossRefGoogle ScholarPubMed
Slagboom, PE, Droog, S, Boomsma, DI. Genetic determination of telomere size in humans: a twin study of three age groups. American Journal of Human Genetics 1994;55:876–82.Google ScholarPubMed
Shay, JW, Roninson, IB. Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene 2004;23:2919–33.CrossRefGoogle ScholarPubMed
Campisi, J. Senescent cells, tumor suppression, and organismal aging: good citizen, bad neighbors. Cell 2005;120:513–22.CrossRefGoogle Scholar
Campisi, J, d’Adda di Fagagna, F.Cellular senescence: when bad things happen to good cells. Nature Reviews Molecular Cell Biology 2007;8:729–40.CrossRefGoogle ScholarPubMed
Brouilette, S, Singh, RK, Thompson, JR, Goodall, AH, Samani, NJ. White cell telomere length and risk of premature myocardial infarction. Arteriosclerosis, Thrombosis and Vascular Biology 2003;23:842–6.CrossRefGoogle ScholarPubMed
von Zglinicki, T, Serra, V, Lorenz, M et al. Short telomeres in patients with vascular dementia: an indicator of low antioxidative capacity and possible risk factors? Laboratory Investigations 2000; 80:1739–47.CrossRefGoogle Scholar
Benetos, A, Gardner, JP, Zureik, M, et al. Short telomeres are associated with increased carotid atherosclerosis in hypertensive subjects. Hypertension 2004;43:182–5.CrossRefGoogle ScholarPubMed
Okuda, K, Khan, MY, Skurnick, J, et al. Telomere attrition of the human abdominal aorta: relationships with age and atherosclerosis. Atherosclerosis 2000;152:391–8.CrossRefGoogle ScholarPubMed
Samani, NJ, Boultby, R, Butler, R, Thompson, JR, Goodall, AH. Telomere shortening in atherosclerosis. Lancet 2001;358:472–3.CrossRefGoogle ScholarPubMed
Panossian, LA, Porter, VR, Valenzuela, HF et al. Telomere shortening in T cells correlate with Alzheimer's disease status. Neurobiology of Aging 2003;24:77–84.CrossRefGoogle Scholar
Meeker, A. K., Hicks, JL, Platz, EA, et al. Telomere length abnormalities occur early in the initiation of epithelial carcinogenesis. Clinical Cancer Research 2004;10:317–26.CrossRefGoogle ScholarPubMed
Rudolph, KL, Millard, M, Bosenberg, MW, DePinho, RA. Telomere dysfunction and evolution of intestinal carcinoma in mice and humans. Nature Genetics 2001;28:155–9.CrossRefGoogle ScholarPubMed
O’Sullivan, JN, Bronner, MP, Brentnall, TA, et al. Chromosomal instability in ulcerative colitis is related to telomere shortening. Nature Genetics 2002;32:280–4.CrossRefGoogle ScholarPubMed
Wiemann, SU, Satyanarayana, A, Tsahuridu, M, et al. Hepatocyte telomere shortening and senescence are general markers of human liver cirrhosis. FASEB Journal 2002;16:935–42.CrossRefGoogle ScholarPubMed
Farazi, PA, Glickman, J, Jiang, S, et al. Differential impact of telomere dysfunction on initiation and progression of hepatocellular carcinoma. Cancer Research 2003;63:5021–7.Google ScholarPubMed
Cawthon, RM, Smith, KR, O’Brien, E, Sivatchenko, A, Kerber, RA. Association between telomere length in blood and mortality in people aged 60 years or older. Lancet 2003;361:393–5.CrossRefGoogle ScholarPubMed
Aviv, A.Telomeres and human aging: facts and fibs. Science of Aging Knowledge Environment 2004;51:43–5.Google Scholar
Aviv, A, Valdes, A, Gardner, JP, et al. Menopause modifies the association of leukocyte telomere length with insulin resistance and inflammation. Journal of Clinical Endocrinology and Metabolism 2006;91:635–40.CrossRefGoogle ScholarPubMed
Andrew, T, Aviv, A, Falchi, M, et al. Mapping genetic loci that determine leukocyte telomere length in a large sample of unselected, female sibling pairs. American Journal of Human Genetics 2006;78:480–6.CrossRefGoogle Scholar
Benetos, A, Okuda, K, Lajemi, M, et al. Telomere length as an indicator of biological aging: the gender effect and relation with pulse pressure and pulse wave velocity. Hypertension 2001;37:381–5.CrossRefGoogle ScholarPubMed
Valdes, AM, Andrew, T, Gardner, JP et al. Obesity, cigarette smoking, and telomere length in women. Lancet 2005;366:662–4.CrossRefGoogle ScholarPubMed
Gardner, JP, Li, S, Srinivasan, SR, Chen, W et al. Rise in insulin resistance is associated with escalated telomere attrition. Circulation 2005;111:2171–7.CrossRefGoogle ScholarPubMed
Nawrot, TS, Staessen, JA, Gardner, JP, Aviv, A. Telomere length and possible link to X chromosome. Lancet 2004;363:507–10.CrossRefGoogle ScholarPubMed
Martin-Ruiz, CM, Gussekloo, J, van Heemst, D, von Zglinicki, T, Westendrop, RG. Telomere length in white blood cells is not associated with morbidity or mortality in the oldest old: a population-based study. Aging Cell 2005;4:287–90.CrossRefGoogle ScholarPubMed
Bischoff, C, Petersen, HC, Graakjaer, J, et al. No association between telomere length and survival among the elderly and oldest old. Epidemiology 2006;17:190–4.CrossRefGoogle ScholarPubMed
Jeanclos, E, Schork, NJ, Kyvik, KO, et al. Telomere length inversely correlates with pulse pressure and is highly familial. Hypertension 2000;36:195–200.CrossRefGoogle ScholarPubMed
Unryn, BM, Cook, LS, Riabowol, KT. Paternal age is positively linked to telomere length of children. Aging Cell 2005;4:97–101.CrossRefGoogle ScholarPubMed
Epel, ES, Blackburn, EH, Lin, J, et al. Accelerated telomere shortening in response to life stress. Proceedings of the National Academy of Science USA 2004;101:17 312–15.CrossRefGoogle ScholarPubMed
Martin-Ruis, C, Dickinson, HO, Key, B, et al. Telomere length predicts poststroke mortality, dementia, and cognitive decline. Annals of Neurology 2006;60:174–80.CrossRefGoogle Scholar
Honig, LS, Schupf, N, Lee, JH, Tang, MX, Mayeux, R. Shorter telomeres are associated with mortality in those with APOE E4 and dementia. Annals of Neurology 2006;60:181–7.CrossRefGoogle ScholarPubMed
Rando, T.Prognostic value of telomere length: the long and short of it. Annals of Neurology 2006;60:155–7.CrossRefGoogle Scholar
Walne, A, Marrone, A, Dokal, I. Dyskeratosis congenita: a disorder of defective telomere maintenance? International Journal of Hematology 2005;82:184–9.CrossRefGoogle ScholarPubMed
Demissie, S, Levy, D, Benjamin, EJ, et al. Insulin resistance, oxidative stress, hypertension, and leukocyte telomere length in men from the Framingham Heart Study. Aging Cell 2006;5:325–30.CrossRefGoogle ScholarPubMed
Fitzpatrick, AL, Kronmal, RA, Gardner, JP, et al. Leukocyte telomere length and cardiovascular disease in the cardiovascular health study. American Journal of Epidemiology 2007;165:14–21.CrossRefGoogle ScholarPubMed
Wright, WE, Pereira-Smith, OM, Shay, JW. Reversible cellular senescence: A two-stage model for the immortalization of normal human diploid fibroblasts. Molecular and Cellular Biology 1989;9:3088–92.CrossRefGoogle ScholarPubMed
Shay, JW, Wright, WE. Senescence and immortalization: role of telomeres and telomerase. Carcinogenesis 2004;2,1–8.Google Scholar
Shay, JW, Wright, WE. Telomerase and human cancer. In: deLange T, Lundblad V, and Blackburn E, editors. Telomeres 2nd edn. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press;2005:81–108.Google Scholar
de Lange, T, Shiue, L, Meyers, RM, et al. Structure and variability of human chromosome ends. Molecular and Cellular Biology 1990:10:518–27.CrossRefGoogle ScholarPubMed
Blackburn, EH. Telomerases. Annual Review of Biochemistry 1992;61:113–29.CrossRefGoogle ScholarPubMed
Greider, CW, Blackburn, EH. Identification of a specific telomere terminal transferase activity in Tetrahymena extracts. Cell 1985;43:405–13.CrossRefGoogle ScholarPubMed
Cristofari, G, Lingner, J. The telomerase ribonucleoprotein particle. In: Telomeres, 2nd edn. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press;2006:21–48.Google Scholar
Nakamura, TM, Morin, GB, Chapman, KB, et al. Telomerase catalytic subunit homologs from fission yeast and humans. Science 1997;277:955–9.CrossRefGoogle Scholar
Feng, J, Funk, WD, Wang, S-S, et al. The RNA component of human telomerase. Science 1995;269:1236–41.CrossRefGoogle ScholarPubMed
Mason, PJ, Wilson, DB, Bessler, M. Dyskeratosis congenital: a disease of dysfunctional telomere maintenance. Current Molecular Medicine 2005;5:159–70.CrossRefGoogle ScholarPubMed
Yamaguchi, H, Calado, RT, Ly, H, et al. Mutations in TERT, the gene for telomerase reverse transcriptase, in aplastic anemia. New England Journal of Medicine 2005;352:1413–24.CrossRefGoogle ScholarPubMed
Mitchell, JR, Wood, E, Collins, KA. A telomerase component is defective in the human disease dyskeratosis congenita. Nature 1999;402:551–5.CrossRefGoogle ScholarPubMed
Shay, JW, Wright, WE. Telomeres in dyskeratosis congenita. Nature Genetics 1999;36:437–8.CrossRefGoogle Scholar
Shay, JW, Wright, WE. Mutant dyskerin ends relationship with telomerase. Science 2004;286:2284–5.CrossRefGoogle Scholar
Vulliamy, T, Marrone, A, Goldman, F, et al. The RNA component of telomerase is mutated in autosomal dominant dyskeratosis congenita. Nature 2001;413:432–5.CrossRefGoogle ScholarPubMed
Heiss, NS, Knight, SW, Vulliamy, TJ, et al. X-linked dyskeratosis congenita is caused by mutations in a highly conserved gene with putative nucleolar functions. Nature Genetics 1998;19:32–8.CrossRefGoogle Scholar
Fogarty, PF, Yamaguchi, H, Wiestner, A, et al. Late presentation of dyskeratosis congenital as apparently acquired aplastic anemia due to mutations in telomerase RNA. Lancet 2003;362:628–30.CrossRefGoogle ScholarPubMed
Holt, SE, Aisner, DL, Baur, J, et al. Functional requirement of p23 and hsp90 in telomerase complexes. Genes and Development 1999;13:817–26.CrossRefGoogle ScholarPubMed
Wright, WE, Piatyszek, MA, Rainey, WE, Byrd, W, Shay, JW. Telomerase activity in human germline and embryonic tissues and cells. Developmental Genetics 1996;18:173–9.3.0.CO;2-3>CrossRefGoogle ScholarPubMed
Harley, CB, Futcher, BA, Greider, CW. Telomeres shorten during aging of human fibroblasts. Nature 1990;345:458–60.CrossRefGoogle ScholarPubMed
Hastie, ND, Dempster, M, Dunlop, MG, et al. Telomere reduction in human colorectal carcinoma and with ageing. Nature 1990;346:866–8.CrossRefGoogle ScholarPubMed
Lindsey, J, McGill, NI, Lindsey, LA, Green, DK, Cooke, HJ. In vivo loss of telomeric repeats with age in humans. Mutation Research 1991;256:45–8.CrossRefGoogle ScholarPubMed
Bodnar, AG, Ouellete, M, Frolkis, M, et al. Extension of lifespan by introduction of telomerase in normal human cells. Science 1998;279:349–52.CrossRefGoogle Scholar
Vaziri, H, Benchimol, S. Reconstitution of telomerase activity in normal human cells leads to elongation of telomeres and extended replicative life span. Current Biology 1998;8:279–82.CrossRefGoogle ScholarPubMed
Garcia, CK, Wright, WE, Shay, JW.Human diseases of telomerase dysfunction: insights into tissue aging. Nucleic Acids Research 2007;35:7406–16.CrossRefGoogle ScholarPubMed
Tsakiri, KD, Cronkite, JT, Kuan, PJ, et al. Adult-onset pulmonary fibrosis caused by mutations in telomerase. Proceedings of the National Academy of Sciences USA 2007;104:7552–7.CrossRefGoogle ScholarPubMed
Armanios, MY, Chen, JJ, Cogan, JD, et al. Telomerase mutations in families with idiopathic pulmonary fibrosis. New England Journal of Medicine 2007;356:1370–2.CrossRefGoogle ScholarPubMed
Shay, JW, Bacchetti, S. A survey of telomerase in human cancer. European Journal of Cancer 1997;33:787–91.CrossRefGoogle ScholarPubMed
Jakupciak, JP, Wang, W, Barker, PE, Srivastava, S, Atha, DH. Analytical validation of telomerase activity for cancer early detection: TRAP/PCR-CE and hTERT mRNA quantification assay for high-throughput screening of tumor cells. Journal of Molecular Diagnostics 2004;6:157–65.CrossRefGoogle ScholarPubMed
Shay, JW, Werbin, H, Wright, WE. Telomerase assays in the diagnosis and prognosis of cancer. In: Telomeres and Telomerase (Ciba Foundation Symposium 211). Chichester, UK: John Wiley & Sons, Ltd;1997:148–60.Google Scholar
Herbert, B-S, Shay, JW, Wright, WE. Analysis of telomeres and telomerase. In Current Protocols in Cell Biology. New York, NY: John Wiley & Sons, Inc.;2003:8.6.6, 2003.Google Scholar
Harley, CB. Telomerase and cancer therapeutics. Nature Reviews Cancer 2008;8:167–79.CrossRefGoogle ScholarPubMed
Shay, JW. Telomerase in cancer: diagnostic, prognostic and therapeutic implications. Cancer Journal from Scientific American 1998;4:26–34.Google ScholarPubMed
White, L, Wright, WE, Shay, JW. Telomerase inhibitors. Trends in Biotechnology 2001;19:114–20.CrossRefGoogle ScholarPubMed
Shay, JW, Wright, WE. Telomerase: a target for cancer therapy. Cancer Cell 2002;2:257–65.CrossRefGoogle Scholar
Shay, JW. Telomerase therapeutics: telomeres recognized as a DNA damage signal. Clinical Cancer Research 2003;9:3521–5.Google ScholarPubMed
Gellert, GC, Jackson, SR, Dikmen, G, Wright, WE, Shay, JW. Telomerase as a therapeutic target in cancer. Drug Discovery Today 2005;2:159–64.CrossRefGoogle Scholar
Shay, JW, Wright, WE. Mechanism-based combination telomerase inhibition therapy. Cancer Cell 2005;7:1–2.CrossRefGoogle ScholarPubMed
Shay, JW, Keith, WN. Targeting telomerase for cancer therapeutics British Journal of Cancer 2008;98:677–83.
Keith, WN, Thomson, CM, Howcroft, J, Maitland, NJ, Shay, JW. Seeding drug discovery: integrating telomerase cancer biology and cellular senescence to uncover new therapeutic opportunities in targeting cancer stem cells. Drug Discovery Today 2007;12:611–21.CrossRefGoogle ScholarPubMed
Shay, JW, Wright, WE. Hallmarks of telomeres in ageing research. Journal of Pathology 2007;211:114–23.CrossRefGoogle ScholarPubMed
Vonderheide, RH, Schultze, JL, Anderson, KS, et al. Equivalent induction of telomerase-specific cytotoxic T lymphocytes from tumor-bearing patients and healthy individuals. Cancer Research 2001;61:8366–70.Google ScholarPubMed
Vonderheide, RH, Anderson, KS, Hahn, WC, et al. Characterization of HLA-A3-restricted cytotoxic T lymphocytes reactive against the widely expressed tumor antigen telomerase. Clinical Cancer Research 2001;7: 343–8.Google ScholarPubMed
Vonderheide, RH, Domchek, SM, Schultze, JL, et al. Vaccination of cancer patients against telomerase induces functional anti-tumor CD8+ T lymphocytes. Clinical Cancer Research 2004;10:828–39.CrossRefGoogle Scholar
Vonderheide, RH. Telomerase as a universal tumor-associated antigen for cancer immunotherapy. Oncogene 202;21:674–9.
Brunsvig, PF, Aamdal, S, Gjertsen, MK, et al. Telomerase peptide vaccination: a Phase I/II study in patients with non-small cell lung cancer. Cancer Immunology and Immunotherapy 2006;55:1553–64.CrossRefGoogle ScholarPubMed
Su, Z, Dannull, J, Yang, BK, et al. Telomerase mRNA-transfected dendritic cells stimulate antigen-specific CD8+ and CD4+ cell responses in patients with metastatic prostate cancer. Journal of Immunology 2005;174:3798–807.CrossRefGoogle ScholarPubMed
Carpenter, EL, Vonderheide, RH. Telomerase-based immunotherapy of cancer. Expert Opinion on Biological Therapy 2006;10:1031–9.CrossRefGoogle Scholar
Vonderheide, RH. Universal tumor antigens for cancer vaccination: targeting telomerase for immunoprevention. Discovery Medicine 2007;7:103–8.Google ScholarPubMed
Domchek, SM, Recio, A, Mick, R, et al. Telomerase-specific T-cell immunity in breast cancer: impact of vaccination on immunosurveillance. Cancer Research 2007;67:10 546–55.CrossRefGoogle Scholar
Danet-Desnoyers, GH, Luongo, JL, Bonnet, DA, Domchek, SM, Vonderheide, RH. Telomerase vaccination has no detectable effect on SCID-repopulating and colony-forming activities in the bone marrow of cancer patients. Experimental Hematology 2005;33:1275–80.CrossRefGoogle ScholarPubMed
Minev, B, Hipp, J, Firat, H, et al. Cytotoxic T cell immunity against telomerase reverse transcriptase in humans. Proceedings of the National Academy of Sciences USA 2000;97:4796–801.CrossRefGoogle ScholarPubMed
Nair, SK, Heiser, A, Boczkowski, D, et al. Induction of cytotoxic T lymphocyte responses and tumor immunity against unrelated tumors using telomerase reverse transcriptase RNA transfected dendritic cells. Nature Medicine 2000;6:1011–17.CrossRefGoogle ScholarPubMed
Nava-Parafa, P, Emems, LAGV10001, an injectable telomerase peptide vaccine for the treatment of solid cancers. Current Opinion in Molecular Therapy 2007;9:490–7.Google Scholar
Mavroudis, D, Bolonakis, I, Cornet, S. A Phase I study of the optimized cryptic peptide TERT(572y) in patients with advanced malignancies. Oncology 2006;70:306–14.CrossRefGoogle ScholarPubMed
Dikmen, ZG, Wright, WE, Shay, JW, Gryaznov, SM. Telomerase targeted oligonucleotide thio-phosphoramidates in T24-luc bladder cancer cells. Journal of Cellular Biochemistry 2007;104:444–52.CrossRefGoogle Scholar
Dikmen, ZG, Gellert, GC, Jackson, S, et al. In vivo inhibition of lung cancer by GRN163L: a novel human telomerase inhibitor. Cancer Research 2005;65:7866–73.CrossRefGoogle ScholarPubMed
Pongracz, K, Li, S, Herbert, B-S, et al. Novel short oligonucleotide conjugates as inhibitors of human telomerase. Nucleosides, Nucleotides, and Nucleic Acids 2003;22:1627–9.CrossRef
Gryaznov, SM, Jackson, SDikmen, G, et al. Oligonucleotide conjugate GRN163L targeting human telomerase as potential anticancer and antimetastatic agent. Nucleoside, Nucleotides, and Nucleic Acids 2007;26:1–3.CrossRefGoogle ScholarPubMed
Jackson, SR, Zhu, C-H, Paulson, V, et al. Anti-adhesive effects of GRN163L – an oligonucleotide, N3ʹ-P5ʹ thio-phosphoramidate targeting telomerase. Cancer Research 2007;67:1121–9.CrossRefGoogle Scholar
Gellert, GC, Dikmen, ZG, Wright, WE, Gryaznov, S, Shay, JW. Effects of a novel telomerase inhibitor, GRN163L, in human breast cancer. Breast Cancer Research and Treatment 2006;96:73–81.CrossRefGoogle ScholarPubMed
Herbert, B-S, Gellert, GC, Hochreiter, A, et al. Lipid modification of oligonucleotide N3ʹ→P5ʹ: thio-phosphoramidates enhances the potency of telomerase inhibitionOncogene 2005;24:5262–8.CrossRefGoogle ScholarPubMed
Hochreiter, AE, Xiao, H, Goldblatt, EM. Telomerase template antagonist GRN163L disrupts telomere maintenance, tumor growth, and metastasis of breast cancer. Clinical Cancer Research 2006;12:3184–92.CrossRefGoogle ScholarPubMed
Gomez-Millan, J, Goldblatt, EM, Gryaznov, SM, Mendonca, MS, Herbert, BS. Specific telomere dysfunction induced by GRN163L increases radiation sensitivity in breast cancer cells. International Journal of Radiation Oncology, Biology, Physics 2007;67:897–905.CrossRefGoogle ScholarPubMed
Djojosubroto, MW, Chin, AC, Go, N, et al. Telomerase antagonists GRN163 and GRN163L inhibit tumor growth and increase chemosensitivity of human hepatoma. Hepatology 2005;42:1127–36.CrossRefGoogle ScholarPubMed
Ozawa, T, Gryaznov, SM, Hu, LJ, et al. Antitumor effects of specific telomerase inhibitor GRN163 in human glioblastoma xenografts. Neuro-Oncology 2004;6:218–26.CrossRefGoogle ScholarPubMed
Wang, ES, Wu, K, Chin, AC, et al. Telomerase inhibition with an oligonucleotide telomerase template antagonist: in vitro and in vivo studies in multiple myeloma and lymphoma. Blood 2004;103:258–66CrossRefGoogle ScholarPubMed
Akiyama, M, Hideshima, T, Shammas, MA, et al. Effects of oligonucleotide N3ʹ→P5ʹ thio-phosphoramidate (GRN163) targeting telomerase RNA in human multiple myeloma cells. Cancer Research 2003;63:6187–94.Google ScholarPubMed
Seimiya, H, Muramatsu, Y, Ohishi, T, Tsuruo, T. Tankyrase 1 as a target for telomere-directed molecular cancer therapeutics. Cancer Cell 2005;7:25–37.CrossRefGoogle ScholarPubMed
Seimiya, H.The telomeric PARP, tankyrases, as targets for cancer therapy. British Journal of Cancer 2006;94:341–5.CrossRefGoogle ScholarPubMed
Clarke, MF, Dick, JE, Dirks, PB, et al. Cancer stem cells –perspectives on current status and future directions: AACR Workshop on Cancer Stem Cells. Cancer Research 2006;66:9339–44.CrossRefGoogle ScholarPubMed
Ju, Z, Rudolph, KL. Telomeres and telomerase in cancer stem cells. European Journal of Cancer 2006;42:1197–203.CrossRefGoogle ScholarPubMed
Armanios, M, Greider, CW. Telomerase and cancer stem cells. Cold Spring Harbor Symposia on Quantitative Biology 2005;70:205–8.CrossRefGoogle ScholarPubMed
Holt, SE, Aisner, DL, Shay, JW, Wright, WE. Lack of cell cycle regulation of telomerase activity in human cells. Proceedings of the National Academy of Sciences USA 1997;94:10 687–92.CrossRefGoogle ScholarPubMed
Ponti, D, Costa, A, Zaffaroni, N et al. Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Research 2005;65:5506–11.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×