1
|
Kobata K, Matsubara K, Nishikawa R, Narita Y, Matsuo K, Waku T, Kobori A. Oligonucleotide-based telomerase inhibitors with a photoresponsive α-chloroaldehyde. Bioorg Med Chem Lett 2025; 120:130138. [PMID: 39986632 DOI: 10.1016/j.bmcl.2025.130138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/30/2025] [Accepted: 02/09/2025] [Indexed: 02/24/2025]
Abstract
Telomerase, a ribonucleoprotein, is a reverse transcriptase that uses telomerase RNA component (hTR) as a template to elongate telomeric DNA at the ends of chromosomes. This enzyme plays a critical role in oncogenic cellular transformation. Therefore, the telomerase is considered an attractive target for cancer therapy. In this study, we synthesized photo-cross-linking oligodeoxyribonucleotides (ODNs) with a photoresponsive α-chloroaldehyde (PCA) moiety at the 3'-end or the 5'-end of the ODNs and evaluated the inhibition efficiencies for telomerase activity. PCA-modified ODNs with a sequence complementary to the template RNA of telomerase inhibited telomerase activity upon UV irradiation, whereas those with scramble sequence showed no significant inhibition. The 50 % inhibitory concentration (IC50) of PCA-modified ODN was significantly reduced by UV irradiation. These results indicate that PCA-modified ODN inhibits telomerase activity in a sequence-selective manner and that the inhibitory activity is enhanced by the cross-linking reaction following UV irradiation.
Collapse
Affiliation(s)
- Kentaro Kobata
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Kazuki Matsubara
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Risa Nishikawa
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Yuki Narita
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Kazuya Matsuo
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Tomonori Waku
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Akio Kobori
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| |
Collapse
|
2
|
Gao J, Pickett HA. Targeting telomeres: advances in telomere maintenance mechanism-specific cancer therapies. Nat Rev Cancer 2022; 22:515-532. [PMID: 35790854 DOI: 10.1038/s41568-022-00490-1] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2022] [Indexed: 12/31/2022]
Abstract
Cancer cells establish replicative immortality by activating a telomere-maintenance mechanism (TMM), be it telomerase or the alternative lengthening of telomeres (ALT) pathway. Targeting telomere maintenance represents an intriguing opportunity to treat the vast majority of all cancer types. Whilst telomerase inhibitors have historically been heralded as promising anticancer agents, the reality has been more challenging, and there are currently no therapeutic options for cancer types that use ALT despite their aggressive nature and poor prognosis. In this Review, we discuss the mechanistic differences between telomere maintenance by telomerase and ALT, the current methods used to detect each mechanism, the utility of these tests for clinical diagnosis, and recent developments in the therapeutic strategies being employed to target both telomerase and ALT. We present notable developments in repurposing established therapeutic agents and new avenues that are emerging to target cancer types according to which TMM they employ. These opportunities extend beyond inhibition of telomere maintenance, by finding and exploiting inherent weaknesses in the telomeres themselves to trigger rapid cellular effects that lead to cell death.
Collapse
Affiliation(s)
- Jixuan Gao
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Hilda A Pickett
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia.
| |
Collapse
|
3
|
Bavelaar BM, Song L, Jackson MR, Able S, Tietz O, Skaripa-Koukelli I, Waghorn PA, Gill MR, Carlisle RC, Tarsounas M, Vallis KA. Oligonucleotide-Functionalized Gold Nanoparticles for Synchronous Telomerase Inhibition, Radiosensitization, and Delivery of Theranostic Radionuclides. Mol Pharm 2021; 18:3820-3831. [PMID: 34449222 PMCID: PMC8493550 DOI: 10.1021/acs.molpharmaceut.1c00442] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/07/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022]
Abstract
Telomerase represents an attractive target in oncology as it is expressed in cancer but not in normal tissues. The oligonucleotide inhibitors of telomerase represent a promising anticancer strategy, although poor cellular uptake can restrict their efficacy. In this study, gold nanoparticles (AuNPs) were used to enhance oligonucleotide uptake. "match" oligonucleotides complementary to the telomerase RNA template subunit (hTR) and "scramble" (control) oligonucleotides were conjugated to diethylenetriamine pentaacetate (DTPA) for 111In-labeling. AuNPs (15.5 nm) were decorated with a monofunctional layer of oligonucleotides (ON-AuNP) or a multifunctional layer of oligonucleotides, PEG(polethylene glycol)800-SH (to reduce AuNP aggregation) and the cell-penetrating peptide Tat (ON-AuNP-Tat). Match-AuNP enhanced the cellular uptake of radiolabeled oligonucleotides while retaining the ability to inhibit telomerase activity. The addition of Tat to AuNPs increased nuclear localization. 111In-Match-AuNP-Tat induced DNA double-strand breaks and caused a dose-dependent reduction in clonogenic survival of telomerase-positive cells but not telomerase-negative cells. hTR inhibition has been reported to sensitize cancer cells to ionizing radiation, and 111In-Match-AuNP-Tat therefore holds promise as a vector for delivery of radionuclides into cancer cells while simultaneously sensitizing them to the effects of the emitted radiation.
Collapse
Affiliation(s)
- Bas M. Bavelaar
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Lei Song
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Mark R. Jackson
- Institute
of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, University Avenue, Glasgow G12 8QQ, U.K.
| | - Sarah Able
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Ole Tietz
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Irini Skaripa-Koukelli
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Philip A. Waghorn
- Charles
River Laboratories, Elphinstone Research Centre, Elphinstone, Tranent EH33 2NE, U.K.
| | - Martin R. Gill
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Robert C. Carlisle
- Institute
of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, U.K.
| | - Madalena Tarsounas
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Katherine A. Vallis
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| |
Collapse
|
4
|
Role of Lipid-Based and Polymer-Based Non-Viral Vectors in Nucleic Acid Delivery for Next-Generation Gene Therapy. Molecules 2020; 25:molecules25122866. [PMID: 32580326 PMCID: PMC7356024 DOI: 10.3390/molecules25122866] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
The field of gene therapy has experienced an insurgence of attention for its widespread ability to regulate gene expression by targeting genomic DNA, messenger RNA, microRNA, and short-interfering RNA for treating malignant and non-malignant disorders. Numerous nucleic acid analogs have been developed to target coding or non-coding sequences of the human genome for gene regulation. However, broader clinical applications of nucleic acid analogs have been limited due to their poor cell or organ-specific delivery. To resolve these issues, non-viral vectors based on nanoparticles, liposomes, and polyplexes have been developed to date. This review is centered on non-viral vectors mainly comprising of cationic lipids and polymers for nucleic acid-based delivery for numerous gene therapy-based applications.
Collapse
|
5
|
Pontes JF, Grenha A. Multifunctional Nanocarriers for Lung Drug Delivery. NANOMATERIALS 2020; 10:nano10020183. [PMID: 31973051 PMCID: PMC7074870 DOI: 10.3390/nano10020183] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/14/2022]
Abstract
Nanocarriers have been increasingly proposed for lung drug delivery applications. The strategy of combining the intrinsic and more general advantages of the nanostructures with specificities that improve the therapeutic outcomes of particular clinical situations is frequent. These include the surface engineering of the carriers by means of altering the material structure (i.e., chemical modifications), the addition of specific ligands so that predefined targets are reached, or even the tuning of the carrier properties to respond to specific stimuli. The devised strategies are mainly directed at three distinct areas of lung drug delivery, encompassing the delivery of proteins and protein-based materials, either for local or systemic application, the delivery of antibiotics, and the delivery of anticancer drugs-the latter two comprising local delivery approaches. This review addresses the applications of nanocarriers aimed at lung drug delivery of active biological and pharmaceutical ingredients, focusing with particular interest on nanocarriers that exhibit multifunctional properties. A final section addresses the expectations regarding the future use of nanocarriers in the area.
Collapse
Affiliation(s)
- Jorge F. Pontes
- Centre for Marine Sciences (CCMAR), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal;
- Drug Delivery Laboratory, Centre for Biomedical Research (CBMR), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Ana Grenha
- Centre for Marine Sciences (CCMAR), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal;
- Drug Delivery Laboratory, Centre for Biomedical Research (CBMR), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- Correspondence: ; Tel.: +351-289-244-441; Fax: +351-289-800-066
| |
Collapse
|
6
|
Jackson MR, Bavelaar BM, Waghorn PA, Gill MR, El-Sagheer AH, Brown T, Tarsounas M, Vallis KA. Radiolabeled Oligonucleotides Targeting the RNA Subunit of Telomerase Inhibit Telomerase and Induce DNA Damage in Telomerase-Positive Cancer Cells. Cancer Res 2019; 79:4627-4637. [PMID: 31311806 PMCID: PMC7611324 DOI: 10.1158/0008-5472.can-18-3594] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 05/01/2019] [Accepted: 07/12/2019] [Indexed: 12/17/2022]
Abstract
Telomerase is expressed in the majority (>85%) of tumors, but has restricted expression in normal tissues. Long-term telomerase inhibition in malignant cells results in progressive telomere shortening and reduction in cell proliferation. Here we report the synthesis and characterization of radiolabeled oligonucleotides that target the RNA subunit of telomerase, hTR, simultaneously inhibiting enzymatic activity and delivering radiation intracellularly. Oligonucleotides complementary (Match) and noncomplementary (Scramble or Mismatch) to hTR were conjugated to diethylenetriaminepentaacetic dianhydride (DTPA), allowing radiolabeling with the Auger electron-emitting radionuclide indium-111 (111In). Match oligonucleotides inhibited telomerase activity with high potency, which was not observed with Scramble or Mismatch oligonucleotides. DTPA-conjugation and 111In-labeling did not change telomerase inhibition. In telomerase-positive cancer cells, unlabeled Match oligonucleotides had no effect on survival, however, 111In-labeled Match oligonucleotides significantly reduced clonogenic survival and upregulated the DNA damage marker γH2AX. Minimal radiotoxicity and DNA damage was observed in telomerase-negative cells exposed to 111In-Match oligonucleotides. Match oligonucleotides localized in close proximity to nuclear Cajal bodies in telomerase-positive cells. In comparison with Match oligonucleotides, 111In-Scramble or 111In-Mismatch oligonucleotides demonstrated reduced retention and negligible impact on cell survival. This study indicates the therapeutic activity of radiolabeled oligonucleotides that specifically target hTR through potent telomerase inhibition and DNA damage induction in telomerase-expressing cancer cells and paves the way for the development of novel oligonucleotide radiotherapeutics targeting telomerase-positive cancers. SIGNIFICANCE: These findings present a novel radiolabeled oligonucleotide for targeting telomerase-positive cancer cells that exhibits dual activity by simultaneously inhibiting telomerase and promoting radiation-induced genomic DNA damage.
Collapse
Affiliation(s)
- Mark R Jackson
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Bas M Bavelaar
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Philip A Waghorn
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Martin R Gill
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Afaf H El-Sagheer
- Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Tom Brown
- Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Madalena Tarsounas
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Katherine A Vallis
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
7
|
Qin H, Zhao C, Sun Y, Ren J, Qu X. Metallo-supramolecular Complexes Enantioselectively Eradicate Cancer Stem Cells in Vivo. J Am Chem Soc 2017; 139:16201-16209. [DOI: 10.1021/jacs.7b07490] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hongshuang Qin
- Laboratory
of Chemical Biology and State Key Laboratory of Rare Earth Resource
Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Chuanqi Zhao
- Laboratory
of Chemical Biology and State Key Laboratory of Rare Earth Resource
Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Yuhuan Sun
- Laboratory
of Chemical Biology and State Key Laboratory of Rare Earth Resource
Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Jinsong Ren
- Laboratory
of Chemical Biology and State Key Laboratory of Rare Earth Resource
Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Xiaogang Qu
- Laboratory
of Chemical Biology and State Key Laboratory of Rare Earth Resource
Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| |
Collapse
|
8
|
Jäger K, Walter M. Therapeutic Targeting of Telomerase. Genes (Basel) 2016; 7:genes7070039. [PMID: 27455328 PMCID: PMC4962009 DOI: 10.3390/genes7070039] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/16/2016] [Accepted: 06/24/2016] [Indexed: 12/20/2022] Open
Abstract
Telomere length and cell function can be preserved by the human reverse transcriptase telomerase (hTERT), which synthesizes the new telomeric DNA from a RNA template, but is normally restricted to cells needing a high proliferative capacity, such as stem cells. Consequently, telomerase-based therapies to elongate short telomeres are developed, some of which have successfully reached the stage I in clinical trials. Telomerase is also permissive for tumorigenesis and 90% of all malignant tumors use telomerase to obtain immortality. Thus, reversal of telomerase upregulation in tumor cells is a potential strategy to treat cancer. Natural and small-molecule telomerase inhibitors, immunotherapeutic approaches, oligonucleotide inhibitors, and telomerase-directed gene therapy are useful treatment strategies. Telomerase is more widely expressed than any other tumor marker. The low expression in normal tissues, together with the longer telomeres in normal stem cells versus cancer cells, provides some degree of specificity with low risk of toxicity. However, long term telomerase inhibition may elicit negative effects in highly-proliferative cells which need telomerase for survival, and it may interfere with telomere-independent physiological functions. Moreover, only a few hTERT molecules are required to overcome senescence in cancer cells, and telomerase inhibition requires proliferating cells over a sufficient number of population doublings to induce tumor suppressive senescence. These limitations may explain the moderate success rates in many clinical studies. Despite extensive studies, only one vaccine and one telomerase antagonist are routinely used in clinical work. For complete eradication of all subpopulations of cancer cells a simultaneous targeting of several mechanisms will likely be needed. Possible technical improvements have been proposed including the development of more specific inhibitors, methods to increase the efficacy of vaccination methods, and personalized approaches. Telomerase activation and cell rejuvenation is successfully used in regenerative medicine for tissue engineering and reconstructive surgery. However, there are also a number of pitfalls in the treatment with telomerase activating procedures for the whole organism and for longer periods of time. Extended cell lifespan may accumulate rare genetic and epigenetic aberrations that can contribute to malignant transformation. Therefore, novel vector systems have been developed for a 'mild' integration of telomerase into the host genome and loss of the vector in rapidly-proliferating cells. It is currently unclear if this technique can also be used in human beings to treat chronic diseases, such as atherosclerosis.
Collapse
Affiliation(s)
- Kathrin Jäger
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin 13353, Germany.
| | - Michael Walter
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin 13353, Germany.
- Labor Berlin-Charité Vivantes Services GmbH, Sylter Str. 2, Berlin 13353, Germany.
| |
Collapse
|
9
|
Yaswen P, MacKenzie KL, Keith WN, Hentosh P, Rodier F, Zhu J, Firestone GL, Matheu A, Carnero A, Bilsland A, Sundin T, Honoki K, Fujii H, Georgakilas AG, Amedei A, Amin A, Helferich B, Boosani CS, Guha G, Ciriolo MR, Chen S, Mohammed SI, Azmi AS, Bhakta D, Halicka D, Niccolai E, Aquilano K, Ashraf SS, Nowsheen S, Yang X. Therapeutic targeting of replicative immortality. Semin Cancer Biol 2015; 35 Suppl:S104-S128. [PMID: 25869441 PMCID: PMC4600408 DOI: 10.1016/j.semcancer.2015.03.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 03/06/2015] [Accepted: 03/13/2015] [Indexed: 12/15/2022]
Abstract
One of the hallmarks of malignant cell populations is the ability to undergo continuous proliferation. This property allows clonal lineages to acquire sequential aberrations that can fuel increasingly autonomous growth, invasiveness, and therapeutic resistance. Innate cellular mechanisms have evolved to regulate replicative potential as a hedge against malignant progression. When activated in the absence of normal terminal differentiation cues, these mechanisms can result in a state of persistent cytostasis. This state, termed "senescence," can be triggered by intrinsic cellular processes such as telomere dysfunction and oncogene expression, and by exogenous factors such as DNA damaging agents or oxidative environments. Despite differences in upstream signaling, senescence often involves convergent interdependent activation of tumor suppressors p53 and p16/pRB, but can be induced, albeit with reduced sensitivity, when these suppressors are compromised. Doses of conventional genotoxic drugs required to achieve cancer cell senescence are often much lower than doses required to achieve outright cell death. Additional therapies, such as those targeting cyclin dependent kinases or components of the PI3K signaling pathway, may induce senescence specifically in cancer cells by circumventing defects in tumor suppressor pathways or exploiting cancer cells' heightened requirements for telomerase. Such treatments sufficient to induce cancer cell senescence could provide increased patient survival with fewer and less severe side effects than conventional cytotoxic regimens. This positive aspect is countered by important caveats regarding senescence reversibility, genomic instability, and paracrine effects that may increase heterogeneity and adaptive resistance of surviving cancer cells. Nevertheless, agents that effectively disrupt replicative immortality will likely be valuable components of new combinatorial approaches to cancer therapy.
Collapse
Affiliation(s)
- Paul Yaswen
- Life Sciences Division, Lawrence Berkeley National Lab, Berkeley, CA, United States.
| | - Karen L MacKenzie
- Children's Cancer Institute Australia, Kensington, New South Wales, Australia.
| | | | | | | | - Jiyue Zhu
- Washington State University College of Pharmacy, Pullman, WA, United States.
| | | | | | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, HUVR, Consejo Superior de Investigaciones Cientificas, Universdad de Sevilla, Seville, Spain.
| | | | | | | | | | | | | | - Amr Amin
- United Arab Emirates University, Al Ain, United Arab Emirates; Cairo University, Cairo, Egypt
| | - Bill Helferich
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| | | | - Gunjan Guha
- SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust, Guildford, Surrey, United Kingdom
| | | | - Asfar S Azmi
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | | | | | | | | | - S Salman Ashraf
- United Arab Emirates University, Al Ain, United Arab Emirates; Cairo University, Cairo, Egypt
| | | | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| |
Collapse
|
10
|
Radan L, Hughes CS, Teichroeb JH, Vieira Zamora FM, Jewer M, Postovit LM, Betts DH. Microenvironmental regulation of telomerase isoforms in human embryonic stem cells. Stem Cells Dev 2014; 23:2046-66. [PMID: 24749509 DOI: 10.1089/scd.2013.0373] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Recent evidence points to extra-telomeric, noncanonical roles for telomerase in regulating stem cell function. In this study, human embryonic stem cells (hESCs) were cultured in 20% or 2% O2 microenvironments for up to 5 days and evaluated for telomerase reverse transcriptase (TERT) expression and telomerase activity. Results showed increased cell survival and maintenance of the undifferentiated state with elevated levels of nuclear TERT in 2% O2-cultured hESCs despite no significant difference in telomerase activity compared with their high-O2-cultured counterparts. Pharmacological inhibition of telomerase activity using a synthetic tea catechin resulted in spontaneous hESC differentiation, while telomerase inhibition with a phosphorothioate oligonucleotide telomere mimic did not. Reverse transcription polymerase chain reaction (RT-PCR) analysis revealed variations in transcript levels of full-length and alternate splice variants of TERT in hESCs cultured under varying O2 atmospheres. Steric-blocking of Δα and Δβ hTERT splicing using morpholino oligonucleotides altered the hTERT splicing pattern and rapidly induced spontaneous hESC differentiation that appeared biased toward endomesodermal and neuroectodermal cell fates, respectively. Together, these results suggest that post-transcriptional regulation of TERT under varying O2 microenvironments may help regulate hESC survival, self-renewal, and differentiation capabilities through expression of extra-telomeric telomerase isoforms.
Collapse
Affiliation(s)
- Lida Radan
- 1 Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario , London, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
11
|
Sekaran V, Soares J, Jarstfer MB. Telomere Maintenance as a Target for Drug Discovery. J Med Chem 2013; 57:521-38. [DOI: 10.1021/jm400528t] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Vijay Sekaran
- Division of Chemical Biology
and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Joana Soares
- Division of Chemical Biology
and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Michael B. Jarstfer
- Division of Chemical Biology
and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
12
|
Sassano MF, Schlesinger AP, Jarstfer MB. Identification of G-Quadruplex Inducers Usinga Simple, Inexpensiveand Rapid High Throughput Assay, and TheirInhibition of Human Telomerase. THE OPEN MEDICINAL CHEMISTRY JOURNAL 2012; 6:20-8. [PMID: 23173022 PMCID: PMC3502892 DOI: 10.2174/1874104501206010020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 07/17/2012] [Accepted: 07/18/2012] [Indexed: 11/22/2022]
Abstract
Telomeres are protein and DNA complexes located atchromosome ends. Telomeric DNA is composed of a double stranded region of repetitive DNA followed by single-stranded 3' extension of aG-rich sequence. Single-stranded G-rich sequencescan fold into G-quadruplex structures,and molecules that stabilize G-quadruplexes are known to inhibit the enzyme telomerase and disrupt telomere maintenance. Because telomere maintenance is required for proliferation of cancer cells, G-quadruplex stabilizers have become attractive prospects for anticancer drug discovery.However, telomere-targeting G-quadruplex ligands have yet to enter the clinic owing in part to poor pharmacokinetics and target selectivity. Increasing the pharmacophore diversity of G-quadruplex and specifically telomeric-DNA targeting agents should assist in overcoming these shortcomings. In this work, we report the identification and validation ofligands that bind telomeric DNA and induce G-quadruplex formationusing the NCI Diversity Set I, providing validation of anextremely simple, rapid and high-throughput screen using FRET technology. Hits from the screen were validated by examining telomerase inhibition and G-quadruplex inductionusing CD spectroscopy and DNA polymerase stop assays. We show that two known DNA binding molecules, ellipticine derivativeNSC 176327 (apyridocarbazole) and NSC 305831 (an antiparasitic hetero-cyclediamidine referred to as furamidine and DB75),are selective induceG-quadruplex formation in the human telomeric sequence and bind telomeric DNA quadruplexes in the absence of stabilizing monovalent cations with molar ratios(molecule: DNA)of 4:1and 1.5:1, respectively.
Collapse
Affiliation(s)
- Maria Florencia Sassano
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | | | | |
Collapse
|
13
|
Insights into the biomedical effects of carboxylated single-wall carbon nanotubes on telomerase and telomeres. Nat Commun 2012; 3:1074. [DOI: 10.1038/ncomms2091] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 08/24/2012] [Indexed: 02/06/2023] Open
|
14
|
Sharma A, Rajappa M, Saxena A, Sharma M. Telomerase Activity as a Tumor Marker in Indian Women with Cervical Intraepithelial Neoplasia and Cervical Cancer. Mol Diagn Ther 2012; 11:193-201. [PMID: 17570741 DOI: 10.1007/bf03256241] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND OBJECTIVES Cervical cancer is the most common cancer in Indian women and is a leading cause of death in women worldwide. Cervical cancer develops from pre-neoplastic cervical intraepithelial neoplasia (CIN). This study was conducted to evaluate telomerase activity as a tumor marker for the detection of cancer in patients with CIN and cervical cancer. The results were compared with human papillomavirus (HPV) status, clinical staging, and histopathologic studies. METHODS Telomerase activity was detected using the PCR-based telomeric repeat amplification protocol (TRAP) assay in cervical tissues collected by routine punch biopsy from the uterine cervix of patients with suspected cervical cancer. High-risk (HR) HPV-16 and -18 status was determined in all the study groups, including controls. A total of 125 patients (including 50 patients with CIN and 75 patients with cervical cancer [including nine patients with adeno-squamous disease]) and 22 control subjects were studied. The sensitivity and specificity for detecting CIN and cervical cancer were calculated. RESULTS Patients with grade I, II, and III CIN showed 17%, 33%, and 57% positivity for telomerase, respectively. In patients with cervical cancer, those at early clinical stages (Ia-IIb) showed 68% positivity and those at later clinical stages showed 92% positivity for telomerase activity. In the present study, telomerase positivity correlated significantly with the detection of HR HPV-16 and -18 (p < 0.001). As a diagnostic test, none of the described analyses combined a sensitivity of > or =90% with a specificity of > or =90%, except in patients with advanced cancer when telomerase activity was used as a diagnostic test. CONCLUSION Our findings suggest that telomerase activation is a relatively early event in cervical carcinogenesis and correlates with the grade of cervical lesion, HR-HPV status (16 and 18 subtypes), and clinical staging. Hence, these associations suggest it as a possible target for detection of cervical cancer.
Collapse
Affiliation(s)
- Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | | | | |
Collapse
|
15
|
Dong M, Mürdter TE, Philippi C, Loretz B, Schaefer UF, Lehr CM, Schwab M, Ammon-Treiber S. Pulmonary delivery and tissue distribution of aerosolized antisense 2'-O-Methyl RNA containing nanoplexes in the isolated perfused and ventilated rat lung. Eur J Pharm Biopharm 2012; 81:478-85. [PMID: 22565122 DOI: 10.1016/j.ejpb.2012.04.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 04/25/2012] [Accepted: 04/27/2012] [Indexed: 12/31/2022]
Abstract
Pulmonary delivery of drugs, particularly in the treatment of lung cancer, is an attractive strategy for future targeted therapy. In this context, inhalation of nanoplexes might offer a new mode for drug delivery in gene therapy. However, limited data are currently available demonstrating pulmonary delivery, cellular uptake as well as local tolerability in lung tissue. The aim of this study was to elucidate the pulmonary delivery, tissue distribution and local tolerability of aerosolized chitosan-coated poly(lactide-co-glycolide) based nanoplexes containing antisense 2'-O-Methyl RNA (OMR). Therefore, an aerosol of OMR-nanoplexes or OMR alone was administered intra-tracheally using the model of the isolated perfused and ventilated rat lung. Localization of OMR in rat lung tissue was examined by immunohistochemistry. Administration of the OMR-nanoplex formulation resulted in significantly higher cellular OMR uptake of the respiratory epithelium in contrast to the administration of OMR alone, indicating that drug administration via aerosolized nanoplexes is able to target lung tissue. No prominent changes in lung physiology parameters were observed following inhalation, suggesting good local tolerability of OMR-nanoplex formulation.
Collapse
Affiliation(s)
- M Dong
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Gude L, Berkovitch SS, Santos WL, Kutchukian PS, Pawloski AR, Kuimelis R, McGall G, Verdine GL. Mapping targetable sites on human telomerase RNA pseudoknot/template domain using 2'-OMe RNA-interacting polynucleotide (RIPtide) microarrays. J Biol Chem 2012; 287:18843-53. [PMID: 22451672 DOI: 10.1074/jbc.m111.316596] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Most cellular RNAs engage in intrastrand base-pairing that gives rise to complex three-dimensional folds. This self-pairing presents an impediment toward binding of the RNA by nucleic acid-based ligands. An important step in the discovery of RNA-targeting ligands is therefore to identify those regions in a folded RNA that are accessible toward the nucleic acid-based ligand. Because the folding of RNA targets can involve interactions between nonadjacent regions and employ both Watson-Crick and non-Watson-Crick base-pairing, screening of candidate binder ensembles is typically necessary. Microarray-based screening approaches have shown great promise in this regard and have suggested that achieving complete sequence coverage would be a valuable attribute of a next generation system. Here, we report a custom microarray displaying a library of RNA-interacting polynucleotides comprising all possible 2'-OMe RNA sequences from 4- to 8-nucleotides in length. We demonstrate the utility of this array in identifying RNA-interacting polynucleotides that bind tightly and specifically to the highly conserved, functionally essential template/pseudoknot domain of human telomerase RNA and that inhibit telomerase function in vitro.
Collapse
Affiliation(s)
- Lourdes Gude
- Departmens of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Nafee N, Schneider M, Friebel K, Dong M, Schaefer UF, Mürdter TE, Lehr CM. Treatment of lung cancer via telomerase inhibition: self-assembled nanoplexes versus polymeric nanoparticles as vectors for 2'-O-Methyl-RNA. Eur J Pharm Biopharm 2011; 80:478-89. [PMID: 22198416 DOI: 10.1016/j.ejpb.2011.11.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Revised: 11/22/2011] [Accepted: 11/28/2011] [Indexed: 10/14/2022]
Abstract
Antisense oligonucleotide, 2'-O-Methyl-RNA (OMR), is known as potent telomerase inhibitor for the treatment of lung cancer but limited by poor intracellular uptake. Chitosan-coated polymeric nanoparticles were compared to chitosan solution as non-viral vectors for OMR. The study investigated the role of chitosan properties and concentration in improving the efficiency of the nanocarriers in terms of loading, viability, cellular uptake, and telomerase inhibition in human lung cancer cell lines. Certain concentration of chitosan on nanoparticle surface is necessary to significantly increase the cellular uptake. However, excessive chitosan negatively affected the transfection efficiency. Self-assembled nanoplexes with chitosan polymer are preferentially adsorbed to the cell membrane rather than being internalized. Thus, polymeric nanoparticles proved to be superior to cationic polymers as carrier for antisense oligonucleotides. Charge cannot be considered the principle factor behind improved transfection. Uptake studies carried out on air-interface cell cultures to mimic in vivo conditions supported the results on normal cultures showing enhanced uptake of nanoplexes over naked oligonucleotides. OMR nanoplexes reduced telomerase activity by ∼50% in A549 cells concluding the potential of the system as a safe, non-invasive, and efficient treatment for lung carcinoma. These data are prerequisites for the ongoing studies on lung perfusion model and in vivo experiments.
Collapse
Affiliation(s)
- N Nafee
- Biopharmaceutics and Pharmaceutical Technology, Saarland University, Saarbrücken, Germany
| | | | | | | | | | | | | |
Collapse
|
18
|
Jain ML, Bruice PY, Szabó IE, Bruice TC. Incorporation of positively charged linkages into DNA and RNA backbones: a novel strategy for antigene and antisense agents. Chem Rev 2011; 112:1284-309. [PMID: 22074477 DOI: 10.1021/cr1004265] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Moti L Jain
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, USA
| | | | | | | |
Collapse
|
19
|
Svinareva LV, Glukhov AI, Moskaleva EY, Shvets VI. Effect of modified DNA and RNA oligonucleotides on telomerase activity and tumor cell survival in vitro. APPL BIOCHEM MICRO+ 2011. [DOI: 10.1134/s0003683811080102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Tissue slice model of human lung cancer to investigate telomerase inhibition by nanoparticle delivery of antisense 2′-O-methyl-RNA. Int J Pharm 2011; 419:33-42. [DOI: 10.1016/j.ijpharm.2011.07.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 07/05/2011] [Accepted: 07/06/2011] [Indexed: 01/08/2023]
|
21
|
Guittat L, Alberti P, Gomez D, De Cian A, Pennarun G, Lemarteleur T, Belmokhtar C, Paterski R, Morjani H, Trentesaux C, Mandine E, Boussin F, Mailliet P, Lacroix L, Riou JF, Mergny JL. Targeting human telomerase for cancer therapeutics. Cytotechnology 2011; 45:75-90. [PMID: 19003245 DOI: 10.1007/s10616-004-5127-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2004] [Accepted: 09/21/2004] [Indexed: 01/28/2023] Open
Abstract
The enzyme telomerase is involved in the replication of telomeres, specialized structures that cap and protect the ends of chromosomes. Its activity is required for maintenance of telomeres and for unlimited lifespan, a hallmark of cancer cells. Telomerase is overexpressed in the vast majority of human cancer cells and therefore represents an attractive target for therapy. Several approaches have been developed to inhibit this enzyme through the targeting of its RNA or catalytic components as well as its DNA substrate, the single-stranded 3'-telomeric overhang. Telomerase inhibitors are chemically diverse and include modified oligonucleotides as well as small diffusable molecules, both natural and synthetic. This review presents an update of recent investigations pertaining to these agents and discusses their biological properties in the context of the initial paradigm that the exposure of cancer cells to these agents should lead to progressive telomere shortening followed by a delayed growth arrest response.
Collapse
Affiliation(s)
- Lionel Guittat
- Laboratoire de Biophysique, Muséum National d'Histoire Naturelle USM503, INSERM U 565, CNRS UMR 5153, 43, rue Cuvier, 75231, Paris cedex 05, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Okabe K, Harada Y, Zhang J, Tadakuma H, Tani T, Funatsu T. Real time monitoring of endogenous cytoplasmic mRNA using linear antisense 2'-O-methyl RNA probes in living cells. Nucleic Acids Res 2011; 39:e20. [PMID: 21106497 PMCID: PMC3045578 DOI: 10.1093/nar/gkq1196] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 10/27/2010] [Accepted: 11/05/2010] [Indexed: 12/21/2022] Open
Abstract
Visualization and monitoring of endogenous mRNA in the cytoplasm of living cells promises a significant comprehension of refined post-transcriptional regulation. Fluorescently labeled linear antisense oligonucleotides can bind to natural mRNA in a sequence-specific way and, therefore, provide a powerful tool in probing endogenous mRNA. Here, we investigated the feasibility of using linear antisense probes to monitor the variable and dynamic expression of endogenous cytoplasmic mRNAs. Two linear antisense 2'-O-methyl RNA probes, which have different interactive fluorophores at the 5'-end of one probe and at the 3'-end of the other, were used to allow fluorescence resonance energy transfer (FRET) upon hybridization to the target mRNA. By characterizing the formation of the probe-mRNA hybrids in living cells, we found that the probe composition and concentration are crucial parameters in the visualization of endogenous mRNA with high specificity. Furthermore, rapid hybridization (within 1 min) of the linear antisense probe enabled us to visualize dynamic processes of endogenous c-fos mRNA, such as fast elevation of levels after gene induction and the localization of c-fos mRNA in stress granules in response to cellular stress. Thus, our approach provides a basis for real time monitoring of endogenous cytoplasmic mRNA in living cells.
Collapse
Affiliation(s)
- Kohki Okabe
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, The Tokyo Metropolitan Institute of Medical Science, 1-6-2 Kamikitazawa Setagaya-ku, Tokyo 156-8506, The Institute for Integrated Cell-Material Sciences, Kyoto University, Yoshida-Honmachi Sakyo-ku, Kyoto 606-8501, Graduate School of Frontier Sciences, the University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, 2-39-1 Kurokami Kumamoto, Kumamoto 860-8555 and Center for NanoBio Integration, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yoshie Harada
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, The Tokyo Metropolitan Institute of Medical Science, 1-6-2 Kamikitazawa Setagaya-ku, Tokyo 156-8506, The Institute for Integrated Cell-Material Sciences, Kyoto University, Yoshida-Honmachi Sakyo-ku, Kyoto 606-8501, Graduate School of Frontier Sciences, the University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, 2-39-1 Kurokami Kumamoto, Kumamoto 860-8555 and Center for NanoBio Integration, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8656, Japan
| | - Junwei Zhang
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, The Tokyo Metropolitan Institute of Medical Science, 1-6-2 Kamikitazawa Setagaya-ku, Tokyo 156-8506, The Institute for Integrated Cell-Material Sciences, Kyoto University, Yoshida-Honmachi Sakyo-ku, Kyoto 606-8501, Graduate School of Frontier Sciences, the University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, 2-39-1 Kurokami Kumamoto, Kumamoto 860-8555 and Center for NanoBio Integration, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8656, Japan
| | - Hisashi Tadakuma
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, The Tokyo Metropolitan Institute of Medical Science, 1-6-2 Kamikitazawa Setagaya-ku, Tokyo 156-8506, The Institute for Integrated Cell-Material Sciences, Kyoto University, Yoshida-Honmachi Sakyo-ku, Kyoto 606-8501, Graduate School of Frontier Sciences, the University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, 2-39-1 Kurokami Kumamoto, Kumamoto 860-8555 and Center for NanoBio Integration, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tokio Tani
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, The Tokyo Metropolitan Institute of Medical Science, 1-6-2 Kamikitazawa Setagaya-ku, Tokyo 156-8506, The Institute for Integrated Cell-Material Sciences, Kyoto University, Yoshida-Honmachi Sakyo-ku, Kyoto 606-8501, Graduate School of Frontier Sciences, the University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, 2-39-1 Kurokami Kumamoto, Kumamoto 860-8555 and Center for NanoBio Integration, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8656, Japan
| | - Takashi Funatsu
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-0033, The Tokyo Metropolitan Institute of Medical Science, 1-6-2 Kamikitazawa Setagaya-ku, Tokyo 156-8506, The Institute for Integrated Cell-Material Sciences, Kyoto University, Yoshida-Honmachi Sakyo-ku, Kyoto 606-8501, Graduate School of Frontier Sciences, the University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa-shi, Chiba 277-8562, Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, 2-39-1 Kurokami Kumamoto, Kumamoto 860-8555 and Center for NanoBio Integration, the University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
23
|
Xu Y. Chemistry in human telomere biology: structure, function and targeting of telomere DNA/RNA. Chem Soc Rev 2011; 40:2719-40. [DOI: 10.1039/c0cs00134a] [Citation(s) in RCA: 249] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Maritz MF, Napier CE, Wen VW, MacKenzie KL. Targeting telomerase in hematologic malignancy. Future Oncol 2010; 6:769-89. [PMID: 20465390 DOI: 10.2217/fon.10.42] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Over the past two decades, it has become increasingly apparent that telomerase-mediated telomere maintenance plays a crucial role in hematopoiesis. Supporting evidence is underscored by recent findings of mutations in genes involved in telomerase-mediated telomere maintenance that contribute to the pathogenesis of bone marrow failure syndromes. More recently described telomere-independent functions of telomerase are also likely to contribute to both normal hematopoiesis and hematologic diseases. The high levels of telomerase detected in aggressive leukemias have fueled fervent investigation into diverse approaches to targeting telomerase in hematologic malignancies. Successful preclinical investigations that employed genetic strategies, oligonucleotides, small-molecule inhibitors and immunotherapy have resulted in a rapid translation to clinical trials. Further investigation of telomere-independent functions of telomerase and detailed preclinical studies of telomerase inhibition in both normal and malignant hematopoiesis will be invaluable for refining treatments to effectively and safely exploit telomerase as a therapeutic target in hematologic malignancies.
Collapse
Affiliation(s)
- Michelle F Maritz
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, New South Wales, Australia
| | | | | | | |
Collapse
|
25
|
Moses JE, Ritson DJ, Zhang F, Lombardo CM, Haider S, Oldham N, Neidle S. A click chemistry approach to C3 symmetric, G-quadruplex stabilising ligands. Org Biomol Chem 2010; 8:2926-30. [DOI: 10.1039/c005055e] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
26
|
Sreenivasulu K, Vijayalakshmi M, Sambasivarao KRS. Regulation studies of telomerase gene in cancer cells by lentinan. Avicenna J Med Biotechnol 2010; 2:181-5. [PMID: 23408759 PMCID: PMC3558165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Accepted: 12/09/2010] [Indexed: 11/24/2022] Open
Abstract
Lentinan a polysaccharide from medicinal mushroom i.e Lentinus, has been known to have anticancer properties. Telomerase activity is not observed in normal healthy cells, whereas in cancerous cells telomerase expression is high. Telomerase represents a promising cancer therapeutic target. We investigated the inhibitory effect of lentinan on telomerase reverse transcriptase gene (hTERT) which is essential for telomerase activity. To assess the transcriptional effect, DLD -1 cancer cells were cultured in the presence of various concentrations of lentinan. TRAP assay, RT-PCR analysis were performed to find telomerase activity and hTERT gene expression respectively. Since C-myc is known to regulate hTERT, expression of C-myc was also determined. Culturing cells with lentinan resulted in down regulation of hTERT and C-myc expression. These results indicate that lentinan inhibits telomerase activity by down regulating hTERT expression via suppression of C-myc in cancer cells.
Collapse
Affiliation(s)
- Kamma Sreenivasulu
- Department of Biotechnology, KL University, India,Corresponding author: Kamma Sreenivasulu, Ph.D., Department of Biotechnology, KL University, Vaddeswaram, India. E-mail:
| | | | | |
Collapse
|
27
|
Beisner J, Dong M, Taetz S, Nafee N, Griese EU, Schaefer U, Lehr CM, Klotz U, Mürdter TE. Nanoparticle mediated delivery of 2'-O-methyl-RNA leads to efficient telomerase inhibition and telomere shortening in human lung cancer cells. Lung Cancer 2009; 68:346-54. [PMID: 19695733 DOI: 10.1016/j.lungcan.2009.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 07/10/2009] [Accepted: 07/19/2009] [Indexed: 02/09/2023]
Abstract
A promising approach for treatment of non-small cell lung cancer (NSCLC) is based on the inhibition of telomerase in cancer cells. The antisense oligonucleotide 2'-O-methyl-RNA binding to the RNA component of telomerase acts as a selective telomerase inhibitor. We developed chitosan-coated polylactide-coglycolide (PLGA) nanoparticles to mediate efficient delivery of 2'-O-methyl-RNA into human lung cancer cells. Cellular uptake of the inhibitor mediated by chitosan-coated PLGA nanoparticles was greatly enhanced compared to the uptake of antisense oligonucleotide alone as shown by flow cytometry analysis. Confocal laser scanning microscopy clearly demonstrated internalization of 2'-O-methyl-RNA. 2'-O-methyl-RNA-nanoparticle complexes exhibited nearly no acute cytotoxicity in human lung cancer cells and did not influence the viability of primary tumor lung fibroblasts. Human NSCLC A549 cells treated with 2'-O-methyl-RNA-nanoparticle complexes showed 87% viability compared to untreated control cells. 2'-O-methyl-RNA delivered by nanoparticle complexes inhibited telomerase activity in a sequence-specific manner. During long-term treatment (15 weeks) telomerase activity was continuously reduced by approximately 80%. Furthermore, nanoparticle mediated delivery of 2'-O-methyl-RNA resulted in significant telomere shortening from 5.9kb to 4kb (p=0.008) in A549 cells. In summary, our data demonstrate that nanoparticle mediated delivery of 2'-O-methyl-RNA induces effective telomerase inhibition and telomere shortening in human lung cancer cells and therefore represents a novel and promising strategy for the treatment of lung cancer.
Collapse
Affiliation(s)
- Julia Beisner
- Dr. Margarete Fischer - Bosch-Institute of Clinical Pharmacology, University of Tuebingen, Stuttgart, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Patutina OA, Mironova NL, Vlassov VV, Zenkova MA. New approaches for cancer treatment: antitumor drugs based on gene-targeted nucleic acids. Acta Naturae 2009; 1:44-60. [PMID: 22649602 PMCID: PMC3347510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Currently, the main way to fight cancer is still chemotherapy. This method of treatment is at the height of its capacity, so, setting aside the need for further improvements in traditional treatments for neoplasia, it is vital to develop now approaches toward treating malignant tumors. This paper reviews innovational experimental approaches to treating malignant malformations based on the use of gene-targeted drugs, such as antisense oligonucleotides (asON), small interfering RNA (siRNA), ribozymes, and DNAzymes, which can all inhibit oncogene expression. The target genes for these drugs are thoroughly characterized, and the main results from pre-clinical and first-step clinical trials of these drugs are presented. It is shown that the gene-targeted oligonucleotides show considerable variations in their effect on tumor tissue, depending on the target gene in question. The effects range from slowing and stopping the proliferation of tumor cells to suppressing their invasive capabilities. Despite their similarity, not all the antisense drugs targeting the same region of the mRNA of the target-gene were equally effective. The result is determined by the combination of the drug type used and the region of the target-gene mRNA that it complements.
Collapse
Affiliation(s)
- O A Patutina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences
| | | | | | | |
Collapse
|
29
|
Beisner J, Dong M, Taetz S, Piotrowska K, Kleideiter E, Friedel G, Schaefer U, Lehr CM, Klotz U, Mürdter TE. Efficient telomerase inhibition in human non-small cell lung cancer cells by liposomal delivery of 2'-O-methyl-RNA. J Pharm Sci 2009; 98:1765-74. [PMID: 18803262 DOI: 10.1002/jps.21553] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The antisense oligonucleotide 2'-O-methyl-RNA is a selective telomerase inhibitor targeting the telomerase RNA component and represents a potential candidate for anticancer therapy. The poor cellular uptake of 2'-O-methyl-RNA is a limiting factor that may contribute to the lack of functional efficacy. To improve delivery of 2'-O-methyl-RNA and consequently antitumoral efficiency in human lung cancer cells, we have investigated several transfection reagents. The transfection reagents DOTAP, MegaFectin 60, SuperFect, FuGENE 6 and MATra-A were tested for intracellular delivery. A FAM-labeled 2'-O-methyl-RNA was used to assess the intracellular distribution by confocal laser scanning microscopy in A549 human non-small cell lung cancer cells. Telomerase activity was measured using the telomeric repeat amplification protocol. Cell viability after transfection was quantified by the MTT assay. All transfection reagents enhanced 2'-O-methyl-RNA uptake in A549 cells but the cationic lipid reagents DOTAP and MegaFectin 60 were most efficient in the delivery of 2'-O-methyl-RNA resulting in telomerase inhibition. Among both DOTAP exhibited the lowest cytotoxicity. Our experiments show that DOTAP is the most suitable transfection reagent for the delivery of 2'-O-methyl-RNA in human lung cancer cells according to its relatively low cytotoxicity and its ability to promote efficient uptake leading to the inhibition of telomerase.
Collapse
Affiliation(s)
- Julia Beisner
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology and University of Tübingen, Stuttgart, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Cerqueira L, Azevedo NF, Almeida C, Jardim T, Keevil CW, Vieira MJ. DNA mimics for the rapid identification of microorganisms by fluorescence in situ hybridization (FISH). Int J Mol Sci 2008; 9:1944-60. [PMID: 19325728 PMCID: PMC2635612 DOI: 10.3390/ijms9101944] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Revised: 09/24/2008] [Accepted: 10/06/2008] [Indexed: 12/23/2022] Open
Abstract
Fluorescence in situ hybridization (FISH) is a well-established technique that is used for a variety of purposes, ranging from pathogen detection in clinical diagnostics to the determination of chromosomal stability in stem cell research. The key step of FISH involves the detection of a nucleic acid region and as such, DNA molecules have typically been used to probe for the sequences of interest. However, since the turn of the century, an increasing number of laboratories have started to move on to the more robust DNA mimics methods, most notably peptide and locked nucleic acids (PNA and LNA). In this review, we will cover the state-of-the-art of the different DNA mimics in regard to their application as efficient markers for the presence of individual microbial cells, and consider their potential advantages and pitfalls. Available PNA probes are then reassessed in terms of sensitivity and specificity using rRNA databases. In addition, we also attempt to predict the applicability of DNA mimics in well-known techniques attempting to detect in situ low number of copies of specific nucleic acid sequences such as catalyzed reporter deposition (CARD) and recognition of individual genes (RING) FISH.
Collapse
Affiliation(s)
- Laura Cerqueira
- IBB - Institute for Biotechnology and Bioengineering, Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar 4710-057, Braga, Portugal. E-Mails:
(L. C.);
(C. A.);
(T. J.);
(M. V.)
| | - Nuno F. Azevedo
- IBB - Institute for Biotechnology and Bioengineering, Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar 4710-057, Braga, Portugal. E-Mails:
(L. C.);
(C. A.);
(T. J.);
(M. V.)
- Environmental Healthcare Unit, School of Biological Sciences, University of Southampton, Bassett Crescent East, Southampton SO16 7PX, UK. E-Mail:
(N. A.)
- Author to whom correspondence should be addressed; E-Mail:
; Tel. +351-253605413; Fax: +351-253678986
| | - Carina Almeida
- IBB - Institute for Biotechnology and Bioengineering, Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar 4710-057, Braga, Portugal. E-Mails:
(L. C.);
(C. A.);
(T. J.);
(M. V.)
- Environmental Healthcare Unit, School of Biological Sciences, University of Southampton, Bassett Crescent East, Southampton SO16 7PX, UK. E-Mail:
(N. A.)
| | - Tatiana Jardim
- IBB - Institute for Biotechnology and Bioengineering, Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar 4710-057, Braga, Portugal. E-Mails:
(L. C.);
(C. A.);
(T. J.);
(M. V.)
| | - Charles William Keevil
- Environmental Healthcare Unit, School of Biological Sciences, University of Southampton, Bassett Crescent East, Southampton SO16 7PX, UK. E-Mail:
(N. A.)
| | - Maria J. Vieira
- IBB - Institute for Biotechnology and Bioengineering, Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar 4710-057, Braga, Portugal. E-Mails:
(L. C.);
(C. A.);
(T. J.);
(M. V.)
| |
Collapse
|
31
|
Kieltyka R, Fakhoury J, Moitessier N, Sleiman H. Platinum Phenanthroimidazole Complexes as G-Quadruplex DNA Selective Binders. Chemistry 2008; 14:1145-54. [DOI: 10.1002/chem.200700783] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
32
|
Tárkányi I, Aradi J. Pharmacological intervention strategies for affecting telomerase activity: future prospects to treat cancer and degenerative disease. Biochimie 2007; 90:156-72. [PMID: 17945408 DOI: 10.1016/j.biochi.2007.09.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Accepted: 09/04/2007] [Indexed: 12/20/2022]
Abstract
Telomerase enzyme is a ribonucleoprotein maintaining the length of the telomeres by adding G-rich repeats to the end of the eukaryotic chromosomes. Normal human somatic cells, cultured in vitro, have a strictly limited proliferative potential undergoing senescence after about 50-70 population doublings. In contrast, most of the tumor cells have unlimited replicative potential. Although the mechanisms of immortalization are not understood completely at a genetic level, the key role of the telomere/telomerase system in the process is clear. The DNA replication machinery is not able to replicate fully the DNA at the very end of the chromosomes; therefore, about 50-200 nucleotides are lost during each of the replication cycles resulting in a gradual decrease of telomere length. Critically short telomere induces senescence, subsequent crisis and cell death. In tumor cells, however, the telomerase enzyme prevents the formation of critically short telomeres, adding GGTTAG repeats to the 3' end of the chromosomes immortalizing the cells. Immortality is one of the hallmarks of cancer. Besides the catalytic activity dependent telomere maintenance, catalytic activity-independent effects of telomerase may also be involved in the regulation of cell cycle. The telomere/telomerase system offers two possibilities to intervene the proliferative activity of the cell: (1) inhibition the telomere maintenance by inhibiting the telomerase activity; (2) activating the residual telomerase enzyme or inducing telomerase expression. Whilst the former approach could abolish the limitless replicative potential of malignant cells, the activation of telomerase might be utilized for treating degenerative diseases. Here, we review the current status of telomerase therapeutics, summarizing the activities of those pharmacological agents which either inhibit or activate the enzyme. We also discuss the future opportunities and challenges of research on pharmacological intervention of telomerase activity.
Collapse
Affiliation(s)
- I Tárkányi
- 3rd Department of Internal Medicine, University of Debrecen, 22 Moricz Zsigmond Krt., Debrecen 4004, Hungary
| | | |
Collapse
|
33
|
Nafee N, Taetz S, Schneider M, Schaefer UF, Lehr CM. Chitosan-coated PLGA nanoparticles for DNA/RNA delivery: effect of the formulation parameters on complexation and transfection of antisense oligonucleotides. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2007; 3:173-83. [PMID: 17692575 DOI: 10.1016/j.nano.2007.03.006] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Revised: 03/01/2007] [Accepted: 03/15/2007] [Indexed: 11/22/2022]
Abstract
Cationically modified poly(D,L-lactide-co-glycolide) (PLGA) nanoparticles have recently been introduced as novel carriers for DNA/RNA delivery. The colloidal characteristics of the nanoparticles--particle size and surface charge--are considered the most significant determinants in the cellular uptake and trafficking of the nanoparticles. Therefore, our aim was to introduce chitosan-coated PLGA nanoparticles, whose size and charge are tunable to adapt for a specific task. The results showed that biodegradable nanoparticles as small as 130 nm and adjustable surface charge can be tailored controlling the process parameters. As a proof of concept, the overall potential of these particulate carriers to bind the antisense oligonucleotides, 2'-O-methyl-RNA, and improve their cellular uptake was demonstrated. The study proved the efficacy of chitosan-coated PLGA nanoparticles as a flexible and efficient delivery system for antisense oligonucleotides to lung cancer cells.
Collapse
Affiliation(s)
- Noha Nafee
- Biopharmaceutics and Pharmaceutical Technology, Saarland University, Saarbrücken, Germany
| | | | | | | | | |
Collapse
|
34
|
De Cian A, Lacroix L, Douarre C, Temime-Smaali N, Trentesaux C, Riou JF, Mergny JL. Targeting telomeres and telomerase. Biochimie 2007; 90:131-55. [PMID: 17822826 DOI: 10.1016/j.biochi.2007.07.011] [Citation(s) in RCA: 484] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Accepted: 07/16/2007] [Indexed: 01/06/2023]
Abstract
Telomeres and telomerase represent, at least in theory, an extremely attractive target for cancer therapy. The objective of this review is to present the latest view on the mechanism(s) of action of telomerase inhibitors, with an emphasis on a specific class of telomere ligands called G-quadruplex ligands, and to discuss their potential use in oncology.
Collapse
Affiliation(s)
- Anne De Cian
- INSERM, U565, Acides nucléiques: dynamique, ciblage et fonctions biologiques, 43 rue Cuvier, CP26, Paris Cedex 05, F-75231, France
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Shortening of telomeres prevents cells from uncontrolled proliferation. Progressive telomere shortening occurs at each cell division until a critical telomeric length is reached. Telomerase expression is switched off after embryonic differentiation in most normal cells, but it is expressed in a very high percentage of tumors of different origin. Thus, telomerase is regarded as the best tumor marker and a promising novel molecular target for cancer treatment. Therefore, different strategies to inhibit telomerase have been developed. However, systematic screening of telomerase inhibitors has not been performed to compare their therapeutic potential. We propose a suitable strategy for estimation of the therapeutic potential of telomerase inhibitors, which is based on a systematic screening of different inhibitors in the same cell system. From the long list of compounds discussed in the literature, we have selected four telomerase inhibitors of different structure and mode of action: BRACO19 (G-quadruplex-interactive compound), BIBR1532 (non-nucleosidic reverse transcriptase inhibitor), 2'-O-methyl RNA, and peptide nucleic acids (PNAs; hTR antisense oligonucleotides). To determine minimal effective concentrations for telomerase inhibition, telomerase activity was measured using the cell-free telomerase repeat amplification protocol (TRAP) assay. We also tested inhibitors in long-term cell-culture experiments by exposing A-549 cells to non-cytotoxic concentrations of inhibitors for a period of 99 days. Subsequently, telomerase activity of A-549 cells was investigated using the TRAP assay, and telomere length of samples was assessed by telomere restriction fragment (TRF) Southern blot analysis.
Collapse
Affiliation(s)
- Elke Kleideiter
- Dr Margarete Fiscer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
| | | | | |
Collapse
|
36
|
Plant Breeding: Antisense ODN Inhibition in in vitro spike cultures as a powerful Diagnostic Tool in Studies on Cereal Grain Development. ACTA ACUST UNITED AC 2007. [DOI: 10.1007/978-3-540-36832-8_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
37
|
Paranjape JM, Xu D, Kushner DM, Okicki J, Lindner DJ, Cramer H, Silverman RH, Leaman DW. Human telomerase RNA degradation by 2'-5'-linked oligoadenylate antisense chimeras in a cell-free system, cultured tumor cells, and murine xenograft models. Oligonucleotides 2006; 16:225-38. [PMID: 16978086 DOI: 10.1089/oli.2006.16.225] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ribonuclease L (RNase L) is a latent single-stranded RNA-directed endoribonuclease that is activated on binding to short 2'-5'-linked oligoadenylates (2-5A), a feature that has led to its use in antisense therapeutic strategies. By attaching a 2-5A moiety to the 5' terminus of standard antisense oligonucleotides, it is possible to activate RNase L and guide it to specific RNAs for degradation. These 2-5A antisense chimeras have been used successfully to target a variety of cellular and viral RNAs. Telomerase is a nuclear ribonucleoprotein complex that elongates telomeric DNA and contributes to cellular immortalization. Telomerase is composed of a protein catalytic subunit and an RNA (hTR or TERC) component, both of which are critical for holoenzyme activity. We describe the characterization of 2-5A antisense chimeras targeting the hTR component of telomerase (2-5A antihTR). Newly designed 2-5A anti-hTR molecules were assayed for their abilities to selectively degrade hTR in a cell-free system. Of the five chimeras tested, one (RBI011) degraded hTR by 97%, and two others (RBI013 and RBI009) were also found to be highly active (73-76% degradation). The ability of transfected RBI011, and its homolog RBI254, to degrade hTR in cultured tumor cells was assessed by real-time RT-PCR. In these studies, RBI011 and RBI254 effectively degraded hTR in a variety of hTR-positive tumor cell lines. The hTR degradation studies were extended to growth assays to determine whether hTR ablation affected tumor cell viability or proliferation. RBI254 treatment resulted in reduced tumor cell viability over the course of 4-day growth assays, effects that were augmented by cotreatment with interferon-beta. To extend these results to an in vivo system, nude mice were implanted subcutaneously or orthotopically with hTR-positive prostate tumors and treated with RBI254. RBI254-treated mice exhibited enhanced tumor cell apoptosis and reduced tumor volume as compared with controls. These findings demonstrated the effectiveness of highly active forms of 2-5A antisense against hTR, and also highlight the usefulness of the cell-free system in predicting chimera efficacy before to inception of cell-based and in vivo studies.
Collapse
Affiliation(s)
- Jayashree M Paranjape
- Department of Cancer Biology and Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
García-Cuadrado D, Cuadro AM, Barchín BM, Nuñez A, Cañeque T, Alvarez-Builla J, Vaquero JJ. Palladium-Mediated Functionalization of Heteroaromatic Cations: Comparative Study on Quinolizinium Cations. J Org Chem 2006; 71:7989-95. [PMID: 17025285 DOI: 10.1021/jo060634+] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An efficient palladium-catalyzed cross-coupling reaction on heteroaromatic cations is described. A comparative study of the Stille and Suzuki reactions shows that only the Stille reaction is able to produce an efficient C-C bond formation between any of the four isomeric bromoquinolizinium bromides and a variety of stannanes. In the presence of the catalysts Pd(PPh3)4 or Pd2(dba)3P(o-Tol)3, vinyl, ethynyl, aryl, and heteroaryl groups are successfully incorporated into the quinolizinium system in satisfactory yields under mild reaction conditions. This procedure represents a marked improvement on the functionalization of this class of heteroaromatic cation.
Collapse
Affiliation(s)
- Domingo García-Cuadrado
- Departamento de Química Organica, Universidad de Alcala, 28871-Alcala de Henares, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
39
|
Keppler BR, Jarstfer MB. A high-throughput assay for a human telomerase protein-human telomerase RNA interaction. Anal Biochem 2006; 353:75-82. [PMID: 16620757 DOI: 10.1016/j.ab.2006.03.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Revised: 03/08/2006] [Accepted: 03/15/2006] [Indexed: 02/06/2023]
Abstract
The rapid rate at which cancer cells divide necessitates a mechanism for telomere maintenance, and in approximately 90% of all cancer types the enzyme telomerase is used to maintain the length of telomeric DNA. Telomerase is a multi-subunit enzyme that minimally contains a catalytic protein subunit, hTERT, and an RNA subunit, hTR. Proper assembly of telomerase is critical for its enzymatic activity and therefore is a requirement for the proliferation of most cancer cells. We have developed the first high-throughput screen capable of identifying small molecules that specifically perturb human telomerase assemblage. The screen uses a scintillation proximity assay to identify compounds that prevent a specific and required interaction between hTR and hTERT. Rather than attempting to disrupt all of the individual hTR-hTERT interactions, we focused the screen on the interaction of the CR4-CR5 domain of hTR with hTERT. The screen employs a biotin-labeled derivative of the CR4-CR5 domain of hTR that independently binds [(35)S]hTERT in a functionally relevant manner. The complex between hTERT and biotin-labeled RNA can be captured on streptavidin-coated scintillation proximity beads. Use of 96-well filter plates and a vacuum manifold enables rapid purification of the beads. After optimization, statistical evaluation of the screen generated a Z' factor of 0.6, demonstrating the high precision of the assay.
Collapse
Affiliation(s)
- Brian R Keppler
- Division of Medicinal Chemistry and Natural Products, School of Pharmacy, University of North Carolina, Chapel Hill, 27599, USA
| | | |
Collapse
|
40
|
Matthes E, Lehmann C, Stulich M, Wu Y, Dimitrova L, Uhlmann E, Janta-Lipinski MV. Potent inhibitory activity of chimeric oligonucleotides targeting two different sites of human telomerase. Oligonucleotides 2006; 15:255-68. [PMID: 16396620 DOI: 10.1089/oli.2005.15.255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Suppression of telomerase activity in tumor cells has been considered as a new anticancer strategy. Here, we present chimeric oligonucleotides (chimeric ODNs) as a new type of telomerase inhibitor that contains differently modified oligomers to address two different sites of telomerase: the RNA template and a suggested protein motif. We have shown previously that phosphorothioate-modified oligonucleotides (PS ODNs) interact in a length-dependent rather than in a sequence-dependent manner, presumably with the protein part of the primer-binding site of telomerase, causing strong inhibition of telomerase. In the present study, we demonstrate that extensions of these PS ODNs at their 3'-ends with an antisense oligomer partial sequence covering 11 bases of the RNA template cause significantly increased inhibitory activity, with IC(50) values between 0.60 and 0.95 nM in a Telomeric Repeat Amplification Protocol (TRAP) assay based on U-87 cell lysates. The enhanced inhibitory activity is observed regardless of whether the antisense part is modified (phosphodiester, PO; 2'-O-methylribosyl, 2'-OMe/PO; phosphoramidate, PAM). However, inside intact U-87 cells, these modifications of the antisense part proved to be essential for efficient telomerase inhibition 20 hours after transfection. In particular, the chimeric ODNs containing PAM or 2'-OMe/PO modifications, when complexed with lipofectin, were most efficient telomerase inhibitors (ID(50) = 0.04 and 0.06 microM, respectively). In conclusion, ODNs of this new type emerged as powerful inhibitors of human telomerase and are, therefore, promising candidates for further investigations of the anticancer strategy of telomerase inhibition.
Collapse
Affiliation(s)
- Eckart Matthes
- Max-Delbrück-Centrum für Molekulare Medizin, D-13125 Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
41
|
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: 3.3] [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
|
42
|
Liu T, Hu B, Chung MJ, Ullenbruch M, Jin H, Phan SH. Telomerase regulation of myofibroblast differentiation. Am J Respir Cell Mol Biol 2006; 34:625-33. [PMID: 16424384 PMCID: PMC2644224 DOI: 10.1165/rcmb.2005-0252oc] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Telomerase activity, which has wide expression in cancerous cells, is induced in lung proliferating fibroblasts. It is preferentially expressed in fibroblasts versus myofibroblasts. It is unknown whether regulation of telomerase expression is related to the process of fibroblast differentiation into myofibroblasts. The objective of this study was to clarify such a potential link between telomerase expression and myofibroblast differentiation. Telomerase inhibitor, 3'-azido-2',3'-dideoxythymidine, or antisense oligonucleotide to the telomerase RNA component was used to inhibit the induced fibroblast telomerase activity. The results showed that inhibition of induced telomerase increased alpha-smooth muscle actin expression, an indicator of myofibroblast differentiation. In contrast, induction of telomerase by basic fibroblast growth factor inhibited alpha-smooth muscle actin expression. These findings suggest that the loss of telomerase activity is closely associated with myofibroblast differentiation and possibly functions as a trigger for myofibroblast differentiation. Conversely, expression of telomerase suppresses myofibroblast differentiation.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
43
|
Cunningham AP, Love WK, Zhang RW, Andrews LG, Tollefsbol TO. Telomerase inhibition in cancer therapeutics: molecular-based approaches. Curr Med Chem 2006; 13:2875-88. [PMID: 17073634 PMCID: PMC2423208 DOI: 10.2174/092986706778521887] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Current standard cancer therapies (chemotherapy and radiation) often cause serious adverse off-target effects. Drug design strategies are therefore being developed that will more precisely target cancer cells for destruction while leaving surrounding normal cells relatively unaffected. Telomerase, widely expressed in most human cancers but almost undetectable in normal somatic cells, provides an exciting drug target. This review focuses on recent pharmacogenomic approaches to telomerase inhibition. Antisense oligonucleotides, RNA interference, ribozymes, mutant expression, and the exploitation of differential telomerase expression as a strategy for targeted oncolysis are discussed here in the context of cancer therapeutics. Reports of synergism between telomerase inhibitors and traditional cancer therapeutic agents are also analyzed.
Collapse
MESH Headings
- Drug Design
- Enzyme Inhibitors/pharmacology
- Enzyme Inhibitors/therapeutic use
- Humans
- Neoplasms/drug therapy
- Neoplasms/enzymology
- Neoplasms/pathology
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/pharmacology
- Oligonucleotides, Antisense/therapeutic use
- RNA, Antisense/genetics
- RNA, Antisense/pharmacology
- RNA, Antisense/therapeutic use
- RNA, Catalytic/genetics
- RNA, Catalytic/metabolism
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- Telomerase/antagonists & inhibitors
- Telomerase/genetics
- Telomerase/metabolism
Collapse
Affiliation(s)
- A P Cunningham
- Department of Biology, University of Alabama at Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
44
|
Eitsuka T, Nakagawa K, Suzuki T, Miyazawa T. Polyunsaturated fatty acids inhibit telomerase activity in DLD-1 human colorectal adenocarcinoma cells: A dual mechanism approach. Biochim Biophys Acta Mol Cell Biol Lipids 2005; 1737:1-10. [PMID: 16216547 DOI: 10.1016/j.bbalip.2005.08.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2004] [Revised: 08/07/2005] [Accepted: 08/24/2005] [Indexed: 11/29/2022]
Abstract
As high telomerase activity is detected in most cancer cells, telomerase represents a promising cancer therapeutic target. We investigated the inhibitory effect of various fatty acids on telomerase, with particular emphasis on those with antitumor properties, such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). To evaluate the direct effect of fatty acids on telomerase, cell lysates of DLD-1 human colorectal adenocarcinoma cells were mixed with sample fatty acids, and the telomerase activity was determined. Saturated fatty acids and trans-fatty acids showed very weak or no inhibition of telomerase. In contrast, cis-unsaturated fatty acids significantly inhibited the enzyme, and the inhibitory potency was elevated with an increase in the number of double bonds. Accordingly, polyunsaturated fatty acids (PUFAs), like EPA and DHA, appeared to be powerful telomerase inhibitors. To assess the transcriptional effect, DLD-1 cells were cultured in the presence of sample fatty acids, and telomerase activity and gene expression were subsequently evaluated. Culturing DLD-1 cells with either EPA or DHA resulted in a remarkable decrease in telomerase activity. EPA and DHA inhibited telomerase by down-regulating human telomerase reverse transcriptase (hTERT) and c-myc expression via protein kinase C inhibition. These results indicate that PUFAs can directly inhibit the enzymatic activity of telomerase as well as modulate the telomerase at the transcriptional level.
Collapse
Affiliation(s)
- Takahiro Eitsuka
- Food and Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, 1-1 Tsutsumidori Amamiyamachi, Sendai 981-8555, Japan
| | | | | | | |
Collapse
|
45
|
Natarajan S, Chen Z, Wancewicz EV, Monia BP, Corey DR. Telomerase reverse transcriptase (hTERT) mRNA and telomerase RNA (hTR) as targets for downregulation of telomerase activity. Oligonucleotides 2005; 14:263-73. [PMID: 15665594 DOI: 10.1089/oli.2004.14.263] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Telomerase is expressed in cancer cells but not in most normal cells, leading to the hypothesis that telomerase inhibitors may be a powerful approach to cancer therapy. It is possible that telomerase plays roles in the cell other than telomere elongation and that blocking telomerase expression may have consequences that differ from simply blocking the active site through competitive inhibition. Here, we test this hypothesis by comparing the effects of antisense oligonucleotides and small interfering RNAs (siRNAs) that target the telomerase reverse transcriptase (hTERT) mRNA with the effects of oligonucleotides that target the telomerase RNA component (hTR). We find that the use of anti-hTR oligomers is more effective in blocking telomerase expression than strategies that target hTERT mRNA. Anti-hTR compounds are active on addition to cells in the absence of lipid, whereas antisense oligonucleotides are not. The modest inhibition of hTERT expression caused by antisense oligonucleotides or siRNAs does not persist, suggesting development of resistance. These data suggest that strategies for telomerase inhibition that require downregulation of hTERT mRNA may be less straightforward than those that target hTR. In addition, we have not seen evidence for a role for hTERT other than in telomere maintenance.
Collapse
Affiliation(s)
- Shobhana Natarajan
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9041, USA
| | | | | | | | | |
Collapse
|
46
|
Shi F, Hoekstra D. Effective intracellular delivery of oligonucleotides in order to make sense of antisense. J Control Release 2005; 97:189-209. [PMID: 15196747 DOI: 10.1016/j.jconrel.2004.03.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2004] [Accepted: 03/12/2004] [Indexed: 01/28/2023]
Abstract
For more than two decades, antisense oligonucleotides (ODNs) have been used to modulate gene expression for the purpose of applications in cell biology and for development of novel sophisticated medical therapeutics. Conceptually, the antisense approach represents an elegant strategy, involving the targeting to and association of an ODN sequence with a specific mRNA via base-pairing, resulting in an impairment of functional and/or harmful protein expression in normal and diseased cells/tissue, respectively. Apart from ODN stability, its efficiency very much depends on intracellular delivery and release/access to the target side, issues that are still relatively poorly understood. Since free ODNs enter cells relatively poorly, appropriate carriers, often composed of polymers and cationic lipids, have been developed. Such carriers allow efficient delivery of ODNs into cells in vitro, and the mechanisms of delivery, both in terms of biophysical requirements for the carrier and cell biological features of uptake, are gradually becoming apparent. To become effective, ODNs require delivery into the nucleus, which necessitates release of internalized ODNs from endosomal compartments, an event that seems to depend on the nature of the delivery vehicle and distinct structural shape changes. Interestingly, evidence is accumulating which suggests that by modulating the surface properties of the carrier, the kinetics of such changes can be controlled, thus providing possibilities for programmable release of the carrier contents. Here, consideration will also be given to antisense design and chemistry, and the challenge of extra- and intracellular barriers to be overcome in the delivery process.
Collapse
Affiliation(s)
- Fuxin Shi
- Department of Membrane Cell Biology, Faculty of Medical Sciences, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | |
Collapse
|
47
|
Therapeutic uses of peptide nucleic acids (PNA) in oncology. Int J Pept Res Ther 2005. [DOI: 10.1007/s10989-005-4910-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
48
|
Yeo M, Rha SY, Jeung HC, Hu SX, Yang SH, Kim YS, An SW, Chung HC. Attenuation of telomerase activity by hammerhead ribozyme targeting human telomerase RNA induces growth retardation and apoptosis in human breast tumor cells. Int J Cancer 2005; 114:484-9. [PMID: 15551309 DOI: 10.1002/ijc.20720] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Ribozyme possesses specific endoribonuclease activity and catalyzes the hydrolysis of specific phosphodiester bonds, which results in the cleavage of target RNA sequences. Here, we evaluated the ability of hammerhead ribozymes targeting human telomerase RNA (hTR) to inhibit the catalytic activity of telomerase and the proliferation of cancer cells. Hammerhead ribozymes were designed against 7 NUX sequences located in open loops of the hTR secondary structure. We verified the ribozyme specificity by in vitro cleavage assay by using a synthetic RNA substrate. Subsequently, we introduced ribozyme expression vector into human breast tumor MCF-7 cells and assessed the biologic effects of ribozyme. Hammerhead ribozyme R1 targeting the template region of hTR efficiently cleaved hTR in vitro, and stable transfectants of this ribozyme induced the degradation of target hTR RNA and attenuated telomerase activity in MCF-7 cells. Moreover, the ribozyme R1 transfectant displayed a significant telomere shortening and a lower proliferation rate than parental cells. Clones with reduced proliferation capacity showed enlarged senescence-like shapes or highly differentiated dendritic morphologies of apoptosis. In conclusion, the inhibition of telomerase activity by hammerhead ribozyme targeting the template region of the hTR presents a promising strategy for inhibiting the growth of human breast cancer cells.
Collapse
Affiliation(s)
- Marie Yeo
- Cancer Metastasis Research Center, Yonsei Cancer Center, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Leonetti C, Amodei S, D'Angelo C, Rizzo A, Benassi B, Antonelli A, Elli R, Stevens MFG, D'Incalci M, Zupi G, Biroccio A. Biological activity of the G-quadruplex ligand RHPS4 (3,11-difluoro-6,8,13-trimethyl-8H-quino[4,3,2-kl]acridinium methosulfate) is associated with telomere capping alteration. Mol Pharmacol 2004; 66:1138-46. [PMID: 15304549 DOI: 10.1124/mol.104.001537] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study had two goals: 1) to evaluate the biological effect of the novel pentacyclic acridine 3,11-difluoro-6,8,13-trimethyl-8H-quino[4,3,2-kl]acridinium methosulfate (RHPS4) on human melanoma lines possessing long telomeres, and 2) to elucidate the relationship between G-quadruplex-based telomerase inhibitor-induced cellular effects and telomere length/dysfunction. The cellular pharmacological effects of RHPS4 have been evaluated by treating melanoma lines with increasing concentrations of RHPS4. A dose-dependent inhibition of cell proliferation was observed in all the lines during short-term treatment. Flow cytometric analysis demonstrated that RHPS4 induced a dose-dependent accumulation of cells in the S-G(2)/M phase of cell cycle. The RHPS4-induced cell cycle alteration was irreversible even at low doses, and the cells died from apoptosis. At high RHPS4 concentration, apoptosis was accompanied by the induction of a senescence phenotype: large cell size, vacuolated cytoplasm, and beta-galactosidase activity. The short-term biological activity of RHPS4 was not caused by telomere shortening, but it was associated with telomere dysfunction, in terms of presence of telomeric fusions, polynucleated cells, and typical images of telophase bridge. In conclusion, our results demonstrate that the G-quadruplex ligand RHPS4 can function in a telomere length-independent manner through its ability to cause telomere-capping alteration.
Collapse
Affiliation(s)
- Carlo Leonetti
- Experimental Chemotherapy Laboratory, Centro di Ricerca Sperimentale, Regina Elena Cancer Institute, University La Sapienza, Roma, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Núñez A, Cuadro AM, Alvarez-Builla J, Vaquero JJ. A Unified Approach to Quinolizinium Cations and Related Systems by Ring-Closing Metathesis. Org Lett 2004; 6:4125-7. [PMID: 15496115 DOI: 10.1021/ol048177b] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
[reaction: see text] The first example of an olefin ring-closing metathesis reaction on cationic heteroaromatic systems is described. Dihydroquinolizinium cations and a variety of related cationic systems are synthesized in an efficient approach from N-alkenyl alpha-vinyl azinium salts using Grubbs' catalysts.
Collapse
Affiliation(s)
- Ana Núñez
- Departamento de Química Orgánica, Universidad de Alcalá, 28871-Alcalá de Henares, Madrid, Spain
| | | | | | | |
Collapse
|