1
|
Gringmuth M, Walther J, Greiser S, Toussaint M, Schwalm B, Kool M, Kortmann RD, Glasow A, Patties I. Enhanced Survival of High-Risk Medulloblastoma-Bearing Mice after Multimodal Treatment with Radiotherapy, Decitabine, and Abacavir. Int J Mol Sci 2022; 23:ijms23073815. [PMID: 35409174 PMCID: PMC8998934 DOI: 10.3390/ijms23073815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/22/2022] [Accepted: 03/26/2022] [Indexed: 02/04/2023] Open
Abstract
Children with high-risk SHH/TP53-mut and Group 3 medulloblastoma (MB) have a 5-year overall survival of only 40%. Innovative approaches to enhance survival while preventing adverse effects are urgently needed. We investigated an innovative therapy approach combining irradiation (RT), decitabine (DEC), and abacavir (ABC) in a patient-derived orthotopic SHH/TP53-mut and Group 3 MB mouse model. MB-bearing mice were treated with DEC, ABC and RT. Mouse survival, tumor growth (BLI, MRT) tumor histology (H/E), proliferation (Ki-67), and endothelial (CD31) staining were analyzed. Gene expression was examined by microarray and RT-PCR (Ki-67, VEGF, CD31, CD15, CD133, nestin, CD68, IBA). The RT/DEC/ABC therapy inhibited tumor growth and enhanced mouse survival. Ki-67 decreased in SHH/TP53-mut MBs after RT, DEC, RT/ABC, and RT/DEC/ABC therapy. CD31 was higher in SHH/TP53-mut compared to Group 3 MBs and decreased after RT/DEC/ABC. Microarray analyses showed a therapy-induced downregulation of cell cycle genes. By RT-PCR, no therapy-induced effect on stem cell fraction or immune cell invasion/activation could be shown. We showed for the first time that RT/DEC/ABC therapy improves survival of orthotopic SHH/TP53-mut and Group 3 MB-bearing mice without inducing adverse effects suggesting the potential for an adjuvant application of this multimodal therapy approach in the human clinic.
Collapse
Affiliation(s)
- Marieke Gringmuth
- Department of Radiation Oncology, University of Leipzig, Stephanstraße 9a, 04103 Leipzig, Germany; (M.G.); (R.-D.K.); (A.G.)
| | - Jenny Walther
- Fraunhofer Center for Microelectronic and Optical Systems for Biomedicine, Herman-Hollerith-Straße 3, 99099 Erfurt, Germany; (J.W.); (S.G.)
- Fraunhofer Institute for Cell Therapy and Immunology, Perlickstraße 1, 04103 Leipzig, Germany
| | - Sebastian Greiser
- Fraunhofer Center for Microelectronic and Optical Systems for Biomedicine, Herman-Hollerith-Straße 3, 99099 Erfurt, Germany; (J.W.); (S.G.)
- Fraunhofer Institute for Cell Therapy and Immunology, Perlickstraße 1, 04103 Leipzig, Germany
| | - Magali Toussaint
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Research Site Leipzig, Permoserstraße 15, 04318 Leipzig, Germany;
| | - Benjamin Schwalm
- Hopp Children’s Cancer Center (KiTZ), Im Neuenheimer Feld 430, 69120 Heidelberg, Germany; (B.S.); (M.K.)
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Research Consortium (DKTK), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Marcel Kool
- Hopp Children’s Cancer Center (KiTZ), Im Neuenheimer Feld 430, 69120 Heidelberg, Germany; (B.S.); (M.K.)
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Research Consortium (DKTK), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Rolf-Dieter Kortmann
- Department of Radiation Oncology, University of Leipzig, Stephanstraße 9a, 04103 Leipzig, Germany; (M.G.); (R.-D.K.); (A.G.)
| | - Annegret Glasow
- Department of Radiation Oncology, University of Leipzig, Stephanstraße 9a, 04103 Leipzig, Germany; (M.G.); (R.-D.K.); (A.G.)
| | - Ina Patties
- Department of Radiation Oncology, University of Leipzig, Stephanstraße 9a, 04103 Leipzig, Germany; (M.G.); (R.-D.K.); (A.G.)
- Correspondence:
| |
Collapse
|
2
|
Biophysical evidence for differential gallated green tea catechins binding to membrane type-1 matrix metalloproteinase and its interactors. Biophys Chem 2018; 234:34-41. [PMID: 29407769 DOI: 10.1016/j.bpc.2018.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/16/2022]
Abstract
Membrane type-1 matrix metalloproteinase (MT1-MMP) is a transmembrane MMP which triggers intracellular signaling and regulates extracellular matrix proteolysis, two functions that are critical for tumor-associated angiogenesis and inflammation. While green tea catechins, particularly epigallocatechin gallate (EGCG), are considered very effective in preventing MT1-MMP-mediated functions, lack of structure-function studies and evidence regarding their direct interaction with MT1-MMP-mediated biological activities remain. Here, we assessed the impact in both cellular and biophysical assays of four ungallated catechins along with their gallated counterparts on MT1-MMP-mediated functions and molecular binding partners. Concanavalin-A (ConA) was used to trigger MT1-MMP-mediated proMMP-2 activation, expression of MT1-MMP and of endoplasmic reticulum stress biomarker GRP78 in U87 glioblastoma cells. We found that ConA-mediated MT1-MMP induction was inhibited by EGCG and catechin gallate (CG), that GRP78 induction was inhibited by EGCG, CG, and gallocatechin gallate (GCG), whereas proMMP-2 activation was inhibited by EGCG and GCG. Surface plasmon resonance was used to assess direct interaction between catechins and MT1-MMP interactors. We found that gallated catechins interacted better than their ungallated analogs with MT1-MMP as well as with MT1-MMP binding partners MMP-2, TIMP-2, MTCBP-1 and LRP1-clusterIV. Overall, current structure-function evidence supports a role for the galloyl moiety in both direct and indirect interactions of green tea catechins with MT1-MMP-mediated oncogenic processes.
Collapse
|
3
|
Feng C, Ho Y, Sun C, Xia G, Ding Q, Gu B. Epigallocatechin gallate inhibits the growth and promotes the apoptosis of bladder cancer cells. Exp Ther Med 2017; 14:3513-3518. [PMID: 29042941 PMCID: PMC5639296 DOI: 10.3892/etm.2017.4981] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 06/15/2017] [Indexed: 11/20/2022] Open
Abstract
Epigallocatechin gallate (EGCG) has been revealed to inhibit the proliferation and induce the apoptosis of several types of tumor, in addition to inhibiting DNA methyltransferase activity, leading to CpG demethylation. Tissue factor pathway inhibitor 2 (TFPI-2) expression is downregulated in bladder cancer. The present study revealed that this downregulation was partly due to hypermethylation of the TFPI-2 gene promoter, which was decreased by EGCG treatment. In addition, the present study demonstrated that EGCG could inhibit the viability and invasion, and induce the apoptosis, of bladder cancer T24 cells. Furthermore, western blot analysis and reverse transcription-quantitative polymerase chain reaction analyses demonstrated that EGCG could upregulate the expression of TFPI-2. These results suggest that EGCG inhibits the growth and induces the apoptosis of bladder cancer cells through restoring TFPI-2 expression. Thus, EGCG is a potential therapeutic candidate for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Chenchen Feng
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Yatfaat Ho
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Chuanyu Sun
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Guowei Xia
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Qiang Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Bin Gu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
4
|
A molecular biology and phase II study of imetelstat (GRN163L) in children with recurrent or refractory central nervous system malignancies: a pediatric brain tumor consortium study. J Neurooncol 2016; 129:443-451. [PMID: 27350411 DOI: 10.1007/s11060-016-2189-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/21/2016] [Indexed: 12/30/2022]
Abstract
Telomerase activation is critical in many cancers including central nervous system (CNS) tumors. Imetelstat is an oligonucleotide that binds to the template region of the RNA component of telomerase, inhibiting its enzymatic activity. We conducted an investigator-sponsored molecular biology (MB) and phase II study to estimate inhibition of tumor telomerase activity and sustained responses by imetelstat in children with recurrent CNS malignancies. In the MB study, patients with recurrent medulloblastoma, high-grade glioma (HGG) or ependymoma undergoing resection received one dose of imetelstat as a 2-h intravenous infusion at 285 mg/m(2), 12-24 h before surgery. Telomerase activity was evaluated in fresh tumor from surgery. Post-surgery and in the phase II study, patients received imetelstat IV (days 1 and 8 q21-days) at 285 mg/m(2). Imetelstat pharmacokinetic and pharmacodynamic studies were performed. Of two evaluable patients on the MB trial, intratumoral telomerase activity was inhibited by 95 % compared to baseline archival tissue in one patient and was inevaluable in one patient. Forty-two patients (40 evaluable for toxicity) were enrolled: 9 medulloblastomas, 18 HGG, 4 ependymomas, 9 diffuse intrinsic pontine gliomas. Most common grade 3/4 toxicities included thrombocytopenia (32.5 %), lymphopenia (17.5 %), neutropenia (12.5 %), ALT (7.5 %) and AST (5 %) elevation. Two patients died of intratumoral hemorrhage secondary to thrombocytopenia leading to premature study closure. No objective responses were observed. Telomerase inhibition was observed in peripheral blood mononuclear cells (PBMCs) for at least 8 days. Imetelstat demonstrated intratumoral and PBMC target inhibition; the regimen proved too toxic in children with recurrent CNS tumors.
Collapse
|
5
|
Alternative lengthening of telomeres is enriched in, and impacts survival of TP53 mutant pediatric malignant brain tumors. Acta Neuropathol 2014; 128:853-62. [PMID: 25315281 DOI: 10.1007/s00401-014-1348-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/23/2014] [Accepted: 09/26/2014] [Indexed: 12/18/2022]
Abstract
Although telomeres are maintained in most cancers by telomerase activation, a subset of tumors utilize alternative lengthening of telomeres (ALT) to sustain self-renewal capacity. In order to study the prevalence and significance of ALT in childhood brain tumors we screened 517 pediatric brain tumors using the novel C-circle assay. We examined the association of ALT with alterations in genes found to segregate with specific histological phenotypes and with clinical outcome. ALT was detected almost exclusively in malignant tumors (p = 0.001). ALT was highly enriched in primitive neuroectodermal tumors (12 %), choroid plexus carcinomas (23 %) and high-grade gliomas (22 %). Furthermore, in contrast to adult gliomas, pediatric low grade gliomas which progressed to high-grade tumors did not exhibit the ALT phenotype. Somatic but not germline TP53 mutations were highly associated with ALT (p = 1.01 × 10(-8)). Of the other alterations examined, only ATRX point mutations and reduced expression were associated with the ALT phenotype (p = 0.0005). Interestingly, ALT attenuated the poor outcome conferred by TP53 mutations in specific pediatric brain tumors. Due to very poor prognosis, one year overall survival was quantified in malignant gliomas, while in children with choroid plexus carcinoma, five year overall survival was investigated. For children with TP53 mutant malignant gliomas, one year overall survival was 63 ± 12 and 23 ± 10 % for ALT positive and negative tumors, respectively (p = 0.03), while for children with TP53 mutant choroid plexus carcinomas, 5 years overall survival was 67 ± 19 and 27 ± 13 % for ALT positive and negative tumors, respectively (p = 0.07). These observations suggest that the presence of ALT is limited to a specific group of childhood brain cancers which harbor somatic TP53 mutations and may influence the outcome of these patients. Analysis of ALT may contribute to risk stratification and targeted therapies to improve outcome for these children.
Collapse
|
6
|
Manoranjan B, Wang X, Hallett RM, Venugopal C, Mack SC, McFarlane N, Nolte SM, Scheinemann K, Gunnarsson T, Hassell JA, Taylor MD, Lee C, Triscott J, Foster CM, Dunham C, Hawkins C, Dunn SE, Singh SK. FoxG1 interacts with Bmi1 to regulate self-renewal and tumorigenicity of medulloblastoma stem cells. Stem Cells 2014; 31:1266-77. [PMID: 23592496 DOI: 10.1002/stem.1401] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 01/27/2013] [Accepted: 02/20/2013] [Indexed: 01/31/2023]
Abstract
Brain tumors represent the leading cause of childhood cancer mortality, of which medulloblastoma (MB) is the most frequent malignant tumor. Recent studies have demonstrated the presence of several MB molecular subgroups, each distinct in terms of prognosis and predicted therapeutic response. Groups 1 and 2 are characterized by relatively good clinical outcomes and activation of the Wnt and Shh pathways, respectively. In contrast, groups 3 and 4 ("non-Shh/Wnt MBs") are distinguished by metastatic disease, poor patient outcome, and lack a molecular pathway phenotype. Current gene expression platforms have not detected brain tumor-initiating cell (BTIC) self-renewal genes in groups 3 and 4 MBs as BTICs typically comprise a minority of tumor cells and may therefore go undetected on bulk tumor analyses. Since increasing BTIC frequency has been associated with increasing tumor aggressiveness and poor patient outcome, we investigated the subgroup-specific gene expression profile of candidate stem cell genes within 251 primary human MBs from four nonoverlapping MB transcriptional databases (Amsterdam, Memphis, Toronto, Boston) and 74 NanoString-subgrouped MBs (Vancouver). We assessed the functional relevance of two genes, FoxG1 and Bmi1, which were significantly enriched in non-Shh/Wnt MBs and showed these genes to mediate MB stem cell self-renewal and tumor initiation in mice. We also identified their transcriptional regulation through reciprocal promoter occupancy in CD15+ MB stem cells. Our work demonstrates the application of stem cell data gathered from genomic platforms to guide functional BTIC assays, which may then be used to develop novel BTIC self-renewal mechanisms amenable to therapeutic targeting.
Collapse
|
7
|
Dorris K, Sobo M, Onar-Thomas A, Panditharatna E, Stevenson CB, Gardner SL, Dewire MD, Pierson CR, Olshefski R, Rempel SA, Goldman S, Miles L, Fouladi M, Drissi R. Prognostic significance of telomere maintenance mechanisms in pediatric high-grade gliomas. J Neurooncol 2014; 117:67-76. [PMID: 24477622 DOI: 10.1007/s11060-014-1374-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 01/19/2014] [Indexed: 02/06/2023]
Abstract
Children with high-grade glioma, including diffuse intrinsic pontine glioma (DIPG), have a poor prognosis despite multimodal therapy. Identifying novel therapeutic targets is critical to improve their outcome. We evaluated prognostic roles of telomere maintenance mechanisms in children with HGG, including DIPG. A multi-institutional retrospective study was conducted involving 50 flash-frozen HGG (35 non-brainstem; 15 DIPG) tumors from 45 children (30 non-brainstem; 15 DIPG). Telomerase activity, expression of hTERT mRNA (encoding telomerase catalytic component) and TERC (telomerase RNA template) and alternative lengthening of telomeres (ALT) mechanism were assayed. Cox Proportional Hazard regression analyses assessed association of clinical and pathological variables, TERC and hTERT levels, telomerase activity, and ALT use with progression-free or overall survival (OS). High TERC and hTERT expression was detected in 13/28 non-brainstem HGG samples as compared to non-neoplastic controls. High TERC and hTERT expression was identified in 13/15 and 11/15 DIPG samples, respectively, compared to controls. Evidence of ALT was noted in 3/11 DIPG and 10/19 non-brainstem HGG specimens. ALT and telomerase use were identified in 4/19 non-brainstem HGG and 2/11 DIPG specimens. In multivariable analyses, increased TERC and hTERT levels were associated with worse OS in patients with non-brainstem HGG, after controlling for tumor grade or resection extent. Children with HGG and DIPG, have increased hTERT and TERC expression. In children with non-brainstem HGG, increased TERC and hTERT expression levels are associated with a worse OS, making telomerase a promising potential therapeutic target in pediatric HGG.
Collapse
Affiliation(s)
- Kathleen Dorris
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, MLC 7013, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Shalaby T, Fiaschetti G, Nagasawa K, Shin-ya K, Baumgartner M, Grotzer M. G-quadruplexes as potential therapeutic targets for embryonal tumors. Molecules 2013; 18:12500-37. [PMID: 24152672 PMCID: PMC6269990 DOI: 10.3390/molecules181012500] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/18/2013] [Accepted: 09/25/2013] [Indexed: 12/27/2022] Open
Abstract
Embryonal tumors include a heterogeneous group of highly malignant neoplasms that primarily affect infants and children and are characterized by a high rate of mortality and treatment-related morbidity, hence improved therapies are clearly needed. G-quadruplexes are special secondary structures adopted in guanine (G)-rich DNA sequences that are often present in biologically important regions, e.g. at the end of telomeres and in the regulatory regions of oncogenes such as MYC. Owing to the significant roles that both telomeres and MYC play in cancer cell biology, G-quadruplexes have been viewed as emerging therapeutic targets in oncology and as tools for novel anticancer drug design. Several compounds that target these structures have shown promising anticancer activity in tumor xenograft models and some of them have entered Phase II clinical trials. In this review we examine approaches to DNA targeted cancer therapy, summarize the recent developments of G-quadruplex ligands as anticancer drugs and speculate on the future direction of such structures as a potential novel therapeutic strategy for embryonal tumors of the nervous system.
Collapse
Affiliation(s)
- Tarek Shalaby
- Division of Oncology, University Children's Hospital of Zurich, Zurich 8032, Switzerland.
| | | | | | | | | | | |
Collapse
|
9
|
Thompson PA, Drissi R, Muscal JA, Panditharatna E, Fouladi M, Ingle AM, Ahern CH, Reid JM, Lin T, Weigel BJ, Blaney SM. A phase I trial of imetelstat in children with refractory or recurrent solid tumors: a Children's Oncology Group Phase I Consortium Study (ADVL1112). Clin Cancer Res 2013; 19:6578-84. [PMID: 24097866 DOI: 10.1158/1078-0432.ccr-13-1117] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Imetelstat is a covalently-lipidated 13-mer thiophosphoramidate oligonucleotide that acts as a potent specific inhibitor of telomerase. It binds with high affinity to the template region of the RNA component of human telomerase (hTERC) and is a competitive inhibitor of telomerase enzymatic activity. The purpose of this study was to determine the recommended phase II dose of imetelstat in children with recurrent or refractory solid tumors. EXPERIMENTAL DESIGN Imetelstat was administered intravenously more than two hours on days 1 and 8, every 21 days. Dose levels of 225, 285, and 360 mg/m(2) were evaluated, using the rolling-six design. Imetelstat pharmacokinetic and correlative biology studies were also performed during the first cycle. RESULTS Twenty subjects were enrolled (median age, 14 years; range, 3-21). Seventeen were evaluable for toxicity. The most common toxicities were neutropenia, thrombocytopenia, and lymphopenia, with dose-limiting myelosuppression in 2 of 6 patients at 360 mg/m(2). Pharmacokinetics is dose dependent with a lower clearance at the highest dose level. Telomerase inhibition was observed in peripheral blood mononuclear cells at 285 and 360 mg/m(2). Two confirmed partial responses, osteosarcoma (n = 1) and Ewing sarcoma (n = 1), were observed. CONCLUSIONS The recommended phase II dose of imetelstat given on days 1 and 8 of a 21-day cycle is 285 mg/m(2).
Collapse
Affiliation(s)
- Patrick A Thompson
- Authors' Affiliations: Texas Children's Cancer Center and Department of Pediatrics; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas; Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Children's Oncology Group, Arcadia; Geron Corporation, Menlo Park, California; Department of Oncology, Mayo Clinic, Rochester; and Department of Pediatrics, Hematology-Oncology, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
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: 1.4] [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
|
11
|
Mandrioli L, Biserni R, Panarese S, Morini M, Gandini G, Bettini G. Immunohistochemical Profiling and Telomerase Activity of a Canine Medulloblastoma. Vet Pathol 2010; 48:814-6. [DOI: 10.1177/0300985810390016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A well-demarcated mass was found by computed tomography in the left cerebellar hemisphere of a 4-year-old male Boxer with acute onset of progressive central vestibular syndrome. At necropsy, the pink, gelatinous mass was in the flocculonodular lobe. Histologically, neoplastic tissue arose from the granular layer of the cerebellar cortex and consisted of sheets of oval to round hyperchromatic cells, consistent with the diagnosis of medulloblastoma. Synaptophysin and neuron-specific enolase immunoreactivity supported the neuronal origin of the neoplastic cells; furthermore, a weak to moderate c-kit expression was detected, as reported in pediatric medulloblastoma. Telomerase activity of tumor cells was demonstrated by immunohistochemistry and by the telomere repeat amplification protocol, suggesting involvement of this enzymatic pathway.
Collapse
Affiliation(s)
- L. Mandrioli
- Department of Veterinary Public Health and Animal Pathology, Alma Mater Studiorum–University of Bologna, Ozzano Emilia, Italy
| | - R. Biserni
- Veterinary Clinical Department, Alma Mater Studiorum–University of Bologna, Ozzano Emilia, Italy
| | - S. Panarese
- Department of Veterinary Public Health and Animal Pathology, Alma Mater Studiorum–University of Bologna, Ozzano Emilia, Italy
| | - M. Morini
- Department of Veterinary Public Health and Animal Pathology, Alma Mater Studiorum–University of Bologna, Ozzano Emilia, Italy
| | - G. Gandini
- Veterinary Clinical Department, Alma Mater Studiorum–University of Bologna, Ozzano Emilia, Italy
| | - G. Bettini
- Department of Veterinary Public Health and Animal Pathology, Alma Mater Studiorum–University of Bologna, Ozzano Emilia, Italy
| |
Collapse
|
12
|
Rahman R, Osteso-Ibanez T, Hirst RA, Levesley J, Kilday JP, Quinn S, Peet A, O'Callaghan C, Coyle B, Grundy RG. Histone deacetylase inhibition attenuates cell growth with associated telomerase inhibition in high-grade childhood brain tumor cells. Mol Cancer Ther 2010; 9:2568-81. [PMID: 20643785 DOI: 10.1158/1535-7163.mct-10-0272] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aberrant epigenetic regulation of gene expression contributes to tumor initiation and progression. Studies from a plethora of hematologic and solid tumors support the use of histone deacetylase inhibitors (HDACi) as potent anticancer agents. However, the mechanism of HDACi action with respect to the temporal order of induced cellular events is unclear. The present study investigates the anticancer effects of the HDACi trichostatin A in high-grade childhood brain tumor cells. Acute exposure to trichostatin A resulted in marked inhibition of cell proliferation, an increase in the proportion of G(2)-M cells, activation of H2A.X, and subsequent induction of apoptosis in the majority of cell lines. These phenotypic effects were associated with abrogation of telomerase activity and human telomerase reverse transcriptase downregulation in the majority of cell lines. In contrast, no cytotoxicity was observed in primary ependymal cells with respect to cilia function. Thus, inhibition of histone deacetylases leads to antiproliferative and proapoptotic effects in childhood brain tumor cells, likely to involve altered chromatin regulation at the human telomerase reverse transcriptase promoter.
Collapse
Affiliation(s)
- Ruman Rahman
- Children's Brain Tumor Research Centre, Medical School, University of Nottingham, Nottingham, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Shalaby T, von Bueren AO, Hürlimann ML, Fiaschetti G, Castelletti D, Masayuki T, Nagasawa K, Arcaro A, Jelesarov I, Shin-ya K, Grotzer M. Disabling c-Myc in childhood medulloblastoma and atypical teratoid/rhabdoid tumor cells by the potent G-quadruplex interactive agent S2T1-6OTD. Mol Cancer Ther 2010; 9:167-79. [PMID: 20053783 DOI: 10.1158/1535-7163.mct-09-0586] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We investigated here the effects of S2T1-6OTD, a novel telomestatin derivative that is synthesized to target G-quadruplex-forming DNA sequences, on a representative panel of human medulloblastoma (MB) and atypical teratoid/rhabdoid (AT/RT) childhood brain cancer cell lines. S2T1-6OTD proved to be a potent c-Myc inhibitor through its high-affinity physical interaction with the G-quadruplex structure in the c-Myc promoter. Treatment with S2T1-6OTD reduced the mRNA and protein expressions of c-Myc and hTERT, which is transcriptionally regulated by c-Myc, and decreased the activities of both genes. In remarkable contrast to control cells, short-term (72-hour) treatment with S2T1-6OTD resulted in a dose- and time-dependent antiproliferative effect in all MB and AT/RT brain tumor cell lines tested (IC(50), 0.25-0.39 micromol/L). Under conditions where inhibition of both proliferation and c-Myc activity was observed, S2T1-6OTD treatment decreased the protein expression of the cell cycle activator cyclin-dependent kinase 2 and induced cell cycle arrest. Long-term treatment (5 weeks) with nontoxic concentrations of S2T1-6OTD resulted in a time-dependent (mainly c-Myc-dependent) telomere shortening. This was accompanied by cell growth arrest starting on day 28 followed by cell senescence and induction of apoptosis on day 35 in all of the five cell lines investigated. On in vivo animal testing, S2T1-6OTD may well represent a novel therapeutic strategy for childhood brain tumors.
Collapse
Affiliation(s)
- Tarek Shalaby
- Department of Oncology, University Children's Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Kilday JP, Rahman R, Dyer S, Ridley L, Lowe J, Coyle B, Grundy R. Pediatric ependymoma: biological perspectives. Mol Cancer Res 2009; 7:765-86. [PMID: 19531565 DOI: 10.1158/1541-7786.mcr-08-0584] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pediatric ependymomas are enigmatic tumors that continue to present a clinical management challenge despite advances in neurosurgery, neuroimaging techniques, and radiation therapy. Difficulty in predicting tumor behavior from clinical and histological factors has shifted the focus to the molecular and cellular biology of ependymoma in order to identify new correlates of disease outcome and novel therapeutic targets. This article reviews our current understanding of pediatric ependymoma biology and includes a meta-analysis of all comparative genomic hybridization (CGH) studies done on primary ependymomas to date, examining more than 300 tumors. From this meta-analysis and a review of the literature, we show that ependymomas in children exhibit a different genomic profile to those in adults and reinforce the evidence that ependymomas from different locations within the central nervous system (CNS) are distinguishable at a genomic level. Potential biological markers of prognosis in pediatric ependymoma are assessed and the ependymoma cancer stem cell hypothesis is highlighted with respect to tumor resistance and recurrence. We also discuss the shifting paradigm for treatment modalities in ependymoma that target molecular alterations in tumor-initiating cell populations.
Collapse
Affiliation(s)
- John-Paul Kilday
- The Children's Brain Tumour Research Centre, University of Nottingham, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
15
|
Rossi A, Russo G, Puca A, La Montagna R, Caputo M, Mattioli E, Lopez M, Giordano A, Pentimalli F. The antiretroviral nucleoside analogue Abacavir reduces cell growth and promotes differentiation of human medulloblastoma cells. Int J Cancer 2009; 125:235-43. [PMID: 19358275 DOI: 10.1002/ijc.24331] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Abacavir is one of the most efficacious nucleoside analogues, with a well-characterized inhibitory activity on reverse transcriptase enzymes of retroviral origin, and has been clinically approved for the treatment of AIDS. Recently, Abacavir has been shown to inhibit also the human telomerase activity. Telomerase activity seems to be required in essentially all tumours for the immortalization of a subset of cells, including cancer stem cells. In fact, many cancer cells are dependent on telomerase for their continued replication and therefore telomerase is an attractive target for cancer therapy. Telomerase expression is upregulated in primary primitive neuroectodermal tumours and in the majority of medulloblastomas suggesting that its activation is associated with the development of these diseases. Therefore, we decided to test Abacavir activity on human medulloblastoma cell lines with high telomerase activity. We report that exposure to Abacavir induces a dose-dependent decrease in the proliferation rate of medulloblastoma cells. This is associated with a cell accumulation in the G(2)/M phase of the cell cycle in the Daoy cell line, and with increased cell death in the D283-MED cell line, and is likely to be dependent on the inhibition of telomerase activity. Interestingly, both cell lines showed features of senescence after Abacavir treatment. Moreover, after Abacavir exposure we detected, by immunofluorescence staining, increased protein expression of the glial marker glial fibrillary acidic protein and the neuronal marker synaptophysin in both medulloblastoma cell lines. In conclusion, our results suggest that Abacavir reduces proliferation and induces differentiation of human medulloblastoma cells through the downregulation of telomerase activity. Thus, using Abacavir, alone or in combination with current therapies, might be an effective therapeutic strategy for the treatment of medulloblastoma.
Collapse
Affiliation(s)
- Alessandra Rossi
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia PA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Rahman R, Heath R, Grundy R. Cellular immortality in brain tumours: an integration of the cancer stem cell paradigm. Biochim Biophys Acta Mol Basis Dis 2009; 1792:280-8. [PMID: 19419702 DOI: 10.1016/j.bbadis.2009.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Revised: 01/21/2009] [Accepted: 01/21/2009] [Indexed: 02/01/2023]
Abstract
Brain tumours are a diverse group of neoplasms that continue to present a formidable challenge in our attempt to achieve curable intervention. Our conceptual framework of human brain cancer has been redrawn in the current decade. There is a gathering acceptance that brain tumour formation is a phenotypic outcome of dysregulated neurogenesis, with tumours viewed as abnormally differentiated neural tissue. In relation, there is accumulating evidence that brain tumours, similar to leukaemia and many solid tumours, are organized as a developmental hierarchy which is maintained by a small fraction of cells endowed with many shared properties of tissue stem cells. Proof that neurogenesis persists throughout adult life, compliments this concept. Although the cancer cell of origin is unclear, the proliferative zones that harbour stem cells in the embryonic, post-natal and adult brain are attractive candidates within which tumour-initiation may ensue. Dysregulated, unlimited proliferation and an ability to bypass senescence are acquired capabilities of cancerous cells. These abilities in part require the establishment of a telomere maintenance mechanism for counteracting the shortening of chromosomal termini. A strategy based upon the synthesis of telomeric repeat sequences by the ribonucleoprotein telomerase, is prevalent in approximately 90% of human tumours studied, including the majority of brain tumours. This review will provide a developmental perspective with respect to normal (neurogenesis) and aberrant (tumourigenesis) cellular turnover, differentiation and function. Within this context our current knowledge of brain tumour telomere/telomerase biology will be discussed with respect to both its developmental and therapeutic relevance to the hierarchical model of brain tumourigenesis presented by the cancer stem cell paradigm.
Collapse
Affiliation(s)
- Ruman Rahman
- School of Clinical Sciences, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| | | | | |
Collapse
|
17
|
RNA interference-mediated c-MYC inhibition prevents cell growth and decreases sensitivity to radio- and chemotherapy in childhood medulloblastoma cells. BMC Cancer 2009; 9:10. [PMID: 19134217 PMCID: PMC2648994 DOI: 10.1186/1471-2407-9-10] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Accepted: 01/10/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND With current treatment strategies, nearly half of all medulloblastoma (MB) patients die from progressive tumors. Accordingly, the identification of novel therapeutic strategies remains a major goal. Deregulation of c-MYC is evident in numerous human cancers. In MB, over-expression of c-MYC has been shown to cause anaplasia and correlate with unfavorable prognosis. METHODS To study the role of c-MYC in MB biology, we down-regulated c-MYC expression by using small interfering RNA (siRNA) and investigated changes in cellular proliferation, cell cycle analysis, apoptosis, telomere maintenance, and response to ionizing radiation (IR) and chemotherapeutics in a representative panel of human MB cell lines expressing different levels of c-MYC (DAOY wild-type, DAOY transfected with the empty vector, DAOY transfected with c-MYC, D341, and D425). RESULTS siRNA-mediated c-MYC down-regulation resulted in an inhibition of cellular proliferation and clonogenic growth, inhibition of G1-S phase cell cycle progression, and a decrease in human telomerase reverse transcriptase (hTERT) expression and telomerase activity. On the other hand, down-regulation of c-MYC reduced apoptosis and decreased the sensitivity of human MB cells to IR, cisplatin, and etoposide. This effect was more pronounced in DAOY cells expressing high levels of c-MYC when compared with DAOY wild-type or DAOY cells transfected with the empty vector. CONCLUSION In human MB cells, in addition to its roles in growth and proliferation, c-MYC is also a potent inducer of apoptosis. Therefore, targeting c-MYC might be of therapeutic benefit when used sequentially with chemo- and radiotherapy rather than concomitantly.
Collapse
|
18
|
Ridley L, Rahman R, Brundler MA, Ellison D, Lowe J, Robson K, Prebble E, Luckett I, Gilbertson RJ, Parkes S, Rand V, Coyle B, Grundy RG. Multifactorial analysis of predictors of outcome in pediatric intracranial ependymoma. Neuro Oncol 2008; 10:675-89. [PMID: 18701711 PMCID: PMC2666244 DOI: 10.1215/15228517-2008-036] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Accepted: 02/25/2008] [Indexed: 01/26/2023] Open
Abstract
Pediatric ependymomas are enigmatic tumors, and their clinical management remains one of the more difficult in pediatric oncology. The identification of biological correlates of outcome and therapeutic targets remains a significant challenge in this disease. We therefore analyzed a panel of potential biological markers to determine optimal prognostic markers. We constructed a tissue microarray from 97 intracranial tumors from 74 patients (WHO grade II-III) and analyzed the candidate markers nucleolin, telomerase catalytic subunit (hTERT; antibody clone 44F12), survivin, Ki-67, and members of the receptor tyrosine kinase I (RTK-I) family by immunohistochemistry. Telomerase activity was determined using the in vitro-based telomere repeat amplification protocol assay, and telomere length was measured using the telomere restriction fragment assay. Primary tumors with low versus high nucleolin protein expression had a 5-year event-free survival of 74%+/-13% and 31%+/-7%, respectively. Multivariate analysis identified low nucleolin expression to be independently associated with a more favorable prognosis (hazard ratio=6.25; 95% confidence interval, 1.6-24.2; p=0.008). Ki-67 and survivin correlated with histological grade but not with outcome. Immunohistochemical detection of the RTK-I family did not correlate with grade or outcome. Telomerase activity was evident in 19 of 22 primary tumors, with telomere lengthening and/or maintenance occurring in five of seven recurrent cases. Low nucleolin expression was the single most important biological predictor of outcome in pediatric intracranial ependymoma. Furthermore, telomerase reactivation and maintenance of telomeric repeats appear necessary for childhood ependymoma progression. These findings require corroboration in a clinical trial setting.
Collapse
Affiliation(s)
- Lee Ridley
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - Ruman Rahman
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - Marie-Anne Brundler
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - David Ellison
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - James Lowe
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - Keith Robson
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - Emma Prebble
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - Inga Luckett
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - Richard J. Gilbertson
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - Sheila Parkes
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - Vikki Rand
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - Beth Coyle
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| | - Richard G. Grundy
- Children’s Brain Tumor Research Centre, University of Nottingham, Nottingham, UK (L.R., R.R., J.L., K.R., V.R., B.C., R.G.G.); Department of Pathology (M.-A.B.) and West Midlands Regional Children’s Tumor Registry (S.P.), Birmingham Children’s Hospital, Birmingham, UK; St. Jude Children’s Research Hospital, Memphis, TN, USA (D.E., I.L., R.J.G.); Regional Genetics Laboratory, Birmingham Women’s Hospital, Birmingham, UK (E.P.); Department of Neuropathology, Nottingham University Hospital, Queens Medical Centre, Nottingham, UK (J.L., K.R.)
| |
Collapse
|
19
|
Avigad S, Naumov I, Ohali A, Jeison M, Berco GH, Mardoukh J, Stark B, Ash S, Cohen IJ, Meller I, Kollender Y, Issakov J, Yaniv I. Short telomeres: a novel potential predictor of relapse in Ewing sarcoma. Clin Cancer Res 2007; 13:5777-83. [PMID: 17908968 DOI: 10.1158/1078-0432.ccr-07-0308] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Despite advances in therapy, >50% of patients with Ewing sarcoma will relapse. The current prognostic factors are not optimal for risk prediction. Studies have shown that telomere length could predict outcome in different malignancies. Our aim was to evaluate whether telomere length could be a better prognostic factor in Ewing sarcoma and correlate the results with clinical variables, outcome, and chromosomal instability. EXPERIMENTAL DESIGN Telomere length was determined in the primary tumor and peripheral blood of 32 patients with Ewing sarcoma. Chromosomal instability was evaluated by combining classical cytogenetics, comparative genomic hybridization and random aneuploidy. Telomere length was correlated to clinical variables, chromosomal instability, and outcome. RESULTS In 75% of the tumors, changes in telomere length, when compared with the corresponding peripheral blood lymphocytes, were noted. The majority of changes consisted of a reduction in telomere length. Patients harboring shorter telomeres had a significantly adverse outcome (P = 0.015). Chromosomal instability was identified in 65% of tumors, significantly correlating with short telomeres (P = 0.0094). Using multivariate analysis, telomere length remained the only significant prognostic variable (P = 0.034). Patients with short telomeres had a 5.3-fold risk of relapse as compared to those with unchanged or longer telomeres. CONCLUSION We have shown that tumors with telomere length reduction result in genomic instability. In addition, telomere length reduction was the only significant predictor of outcome. We suggest that reduction of telomere length in tumor cells at diagnosis could serve as a prognostic marker in Ewing sarcoma.
Collapse
Affiliation(s)
- Smadar Avigad
- Molecular Oncology, Felsenstein Medical Research Center, Petach Tikva, Israel.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
One of the hallmarks of cancer is limitless proliferative capacity, which is tightly associated with the ability to maintain telomeres. Over the last decade, the telomere biology of pediatric cancers has begun to be elucidated. Most pediatric leukemias and embryonal solid tumors activate the enzyme telomerase, a specialized reverse transcriptase that adds nucleotide repeats to telomeres. In general, high levels of tumor telomerase expression are associated with unfavorable outcome, although results vary according to tumor type. Some pediatric tumors, including osteosarcoma and glioblastoma multiforme, lack telomerase activity and maintain telomeres via a recombination-based mechanism called ALT (alternative lengthening of telomeres). Telomerase is a highly attractive therapeutic target for pediatric cancer because the enzyme plays a key role in conferring cellular immortality, is present in most tumors, and is relatively specific for cancer cells. Telomerase inhibitors have been evaluated in preclinical models of adult cancers, but few studies have been conducted on pediatric cancers. Further research is required to define how telomere biology can be used to clinical advantage in malignancies of childhood.
Collapse
Affiliation(s)
- Uri Tabori
- Department of Hematology/Oncology, Hospital for Sick Children, Toronto, Canada
| | | |
Collapse
|
21
|
Ortiz-Plata A, Tena Suck ML, López-Gómez M, Heras A, Sánchez García A. Study of the telomerase hTERT fraction, PCNA and CD34 expression on pituitary adenomas. Association with clinical and demographic characteristics. J Neurooncol 2007; 84:159-66. [PMID: 17361328 DOI: 10.1007/s11060-007-9365-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Accepted: 02/20/2007] [Indexed: 01/09/2023]
Abstract
To determine the frequency of human telomerase reverse transcriptase (hTERT) catalytic fraction expression and its association with clinical and demographic characteristics of the patient, as well as with the expression of CD34 and proliferating cell nuclear antigen (PCNA) indexes on adenohypophyseal hormone tissues. A transverse study was realized with 49 cases of hypophyseal adenoma with analysis type cases and controls. The different adenohypophyseal hormones [prolactin (PRL), growth hormone (GH), follicle stimulating hormone, luteinizing hormone, thyroid gland stimulant hormone, adrenocorticotropic hormone (ACTH)], the catalytic fraction of the telomerase hTERT, the PCNA index and the CD34 density were determined by means of immunohistochemical techniques. The clinical, demographic and histopathological characteristics of the patients with and without hTERT expression were compared by means of Pearson's Chi-squared, Fisher's exact test and Mann-Whitney's U. Twenty-eight point six percent of the adenomas had positive expression for hTERT. The variables significantly correlated with hTERT's expression were younger age of presentation, diagnostic of adenoma producer, higher PCNA index, higher CD34 density, increased GH on serum and the expression on PRL tissue, GH and ACTH. Tobacco history had a negative association with hTERT's expression. The telomerase could be a marker of cellular proliferation associated with angiogenesis and hormonal activity. Evaluation of these variables could provide information about their biological behavior.
Collapse
Affiliation(s)
- Alma Ortiz-Plata
- Departamento de Neuropatología, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Av. Insurgentes Sur 3877 Col La Fama, 14269, Mexico, D.F., Mexico.
| | | | | | | | | |
Collapse
|
22
|
Binz N, Shalaby T, Rivera P, Shin-ya K, Grotzer MA. Telomerase inhibition, telomere shortening, cell growth suppression and induction of apoptosis by telomestatin in childhood neuroblastoma cells. Eur J Cancer 2005; 41:2873-81. [PMID: 16253503 DOI: 10.1016/j.ejca.2005.08.025] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2005] [Revised: 08/05/2005] [Accepted: 08/16/2005] [Indexed: 11/23/2022]
Abstract
Neuroblastoma is a tumour derived from primitive cells of the sympathetic nervous system and is the most common extracranial solid tumour in childhood. Unfavourable tumours are characterised not only by structural changes, including 1p deletion and amplification of the MYCN proto-oncogene, but also by high telomerase activity. Telomeric G-rich single-stranded DNA can adopt in vitro an intramolecular quadruplex structure, which has been shown to inhibit telomerase activity. In this study, we examined telomestatin, a G-quadruplex interactive agent, for its ability to inhibit telomere maintenance of neuroblastoma cells. Telomere length was determined by the terminal restriction fragment method, telomerase activity was measured by a quantitative telomeric repeat amplification protocol, and the expression of human telomerase by quantitative real-time polymerase chain reaction (RT-PCR). Short-term treatment with telomestatin resulted in dose-dependent cytotoxicity and induction of apoptosis. Long-term treatment with telomestatin at non-cytotoxic, but still telomerase activity-inhibiting, concentrations resulted in telomere shortening, growth arrest and induction of apoptosis. These results suggest that the effect of telomestatin is dose-dependent and at least 2-fold. Prolonged low-dose treatment with telomestatin limits the cellular lifespan of NB cells through disruption of telomere maintenance.
Collapse
Affiliation(s)
- N Binz
- Department of Oncology, University Children's Hospital of Zurich, Steinwiesstrasse 75, Hospital, 8032 Zurich, Switzerland
| | | | | | | | | |
Collapse
|