imetelstat and Disease-Models--Animal

imetelstat has been researched along with Disease-Models--Animal* in 5 studies

Other Studies

5 other study(ies) available for imetelstat and Disease-Models--Animal

ArticleYear
Role for telomerase in pulmonary hypertension.
    Circulation, 2015, Feb-24, Volume: 131, Issue:8

    Cells exhibiting dysregulated growth may express telomerase reverse transcriptase (TERT), the dual function of which consists of maintaining telomere length, in association with the RNA template molecule TERC, and controlling cell growth. Here, we investigated lung TERT in human and experimental pulmonary hypertension (PH) and its role in controlling pulmonary artery smooth muscle cell (PA-SMC) proliferation.. Marked TERT expression or activity was found in lungs from patients with idiopathic PH and from mice with PH induced by hypoxia or serotonin-transporter overexpression (SM22-5HTT(+) mice), chiefly within PA-SMCs. In cultured mouse PA-SMCs, TERT was expressed on growth stimulation by serum. The TERT inhibitor imetelstat and the TERT activator TA65 abrogated and stimulated PA-SMC growth, respectively. PA-SMCs from PH mice showed a heightened proliferative phenotype associated with increased TERT expression, which was suppressed by imetelstat treatment. TERC(-/-) mice at generation 2 and TERT(-/-) mice at generations 2, 3, and 4 developed less severe PH than did wild-type mice exposed to chronic hypoxia, with less distal pulmonary artery muscularization and fewer Ki67-stained proliferating PA-SMCs. Telomere length differed between TERC(-/-) and TERT(-/-) mice, whereas PH severity was similar in the 2 strains and across generations. Chronic imetelstat treatment reduced hypoxia-induced PH in wild-type mice or partially reversed established PH in SM22-5HTT(+) mice while simultaneously decreasing TERT expression. Opposite effects occurred in mice treated with TA65.. Telomerase exerts telomere-independent effects on PA-SMC growth in PH and may constitute a treatment target for PH.

    Topics: Adult; Animals; Cell Proliferation; Cells, Cultured; Disease Models, Animal; Female; Humans; Hypertension, Pulmonary; Hypoxia; Indoles; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Mice, Mutant Strains; Middle Aged; Muscle, Smooth, Vascular; Niacinamide; Oligonucleotides; Pulmonary Artery; Serotonin Plasma Membrane Transport Proteins; Telomerase

2015
The HSP90 inhibitor alvespimycin enhances the potency of telomerase inhibition by imetelstat in human osteosarcoma.
    Cancer biology & therapy, 2015, Volume: 16, Issue:6

    The unsatisfactory outcomes for osteosarcoma necessitate novel therapeutic strategies. This study evaluated the effect of the telomerase inhibitor imetelstat in pre-clinical models of human osteosarcoma. Because the chaperone molecule HSP90 facilitates the assembly of telomerase protein, the ability of the HSP90 inhibitor alvespimycin to potentiate the effect of the telomerase inhibitor was assessed. The effect of single or combined treatment with imetelstat and alvespimycin on long-term growth was assessed in osteosarcoma cell lines (143B, HOS and MG-63) and xenografts derived from 143B cells. Results indicated that imetelstat as a single agent inhibited telomerase activity, induced telomere shortening, and inhibited growth in all 3 osteosarcoma cell lines, though the bulk cell cultures did not undergo growth arrest. Combined treatment with imetelstat and alvespimycin resulted in diminished telomerase activity and shorter telomeres compared to either agent alone as well as higher levels of γH2AX and cleaved caspase-3, indicative of increased DNA damage and apoptosis. With dual telomerase and HSP90 inhibition, complete growth arrest of bulk cell cultures was achieved. In xenograft models, all 3 treatment groups significantly inhibited tumor growth compared with the placebo-treated control group, with the greatest effect seen in the combined treatment group (imetelstat, p = 0.045, alvespimycin, p = 0.034; combined treatment, p = 0.004). In conclusion, HSP90 inhibition enhanced the effect of telomerase inhibition in pre-clinical models of osteosarcoma. Dual targeting of telomerase and HSP90 warrants further investigation as a therapeutic strategy.

    Topics: Animals; Apoptosis; Ataxia Telangiectasia Mutated Proteins; Benzoquinones; Cell Line, Tumor; Cell Proliferation; Cell Survival; Disease Models, Animal; DNA Damage; Enzyme Activation; Enzyme Inhibitors; HSP90 Heat-Shock Proteins; Humans; Indoles; Lactams, Macrocyclic; Mice; Niacinamide; Oligonucleotides; Osteosarcoma; Signal Transduction; Telomerase; Telomere Shortening; Tumor Suppressor Protein p53; Xenograft Model Antitumor Assays

2015
Telomerase inhibition improves tumor response to radiotherapy in a murine orthotopic model of human glioblastoma.
    Molecular cancer, 2015, Jul-17, Volume: 14

    Glioblastoma (GBM) is the most frequent and aggressive type of adult brain tumor. Most GBMs express telomerase; a high level of intra-tumoral telomerase activity (TA) is predictive of poor prognosis. Thus, telomerase inhibitors are promising options to treat GBM. These inhibitors increase the response to radiotherapy (RT), in vitro as well as in vivo. Since typical treatments for GBM include RT, our objective was to evaluate the efficiency of Imetelstat (TA inhibitor) combined with RT.. We used a murine orthotopic model of human GBM (N = 8 to11 mice per group) and μMRI imaging to evaluate the efficacy of Imetelstat (delivered by intra-peritoneal injection) alone and combined with RT. Using a clinically established protocol, we demonstrated that Imetelstat significantly: (i) inhibited the TA in the very center of the tumor, (ii) reduced tumor volume as a proportion of TA inhibition, and (iii) increased the response to RT, in terms of tumor volume regression and survival increase.. Imetelstat is currently evaluated in refractory brain tumors in young patients (without RT). Our results support its clinical evaluation combined with RT to treat GBM.

    Topics: Animals; Antineoplastic Agents; Brain Neoplasms; Disease Models, Animal; Glioblastoma; Humans; Indoles; Mice; Niacinamide; Oligonucleotides; Radiation Tolerance; Telomerase; Xenograft Model Antitumor Assays

2015
Telomerase inhibition effectively targets mouse and human AML stem cells and delays relapse following chemotherapy.
    Cell stem cell, 2014, Dec-04, Volume: 15, Issue:6

    Acute myeloid leukemia (AML) is an aggressive and lethal blood cancer maintained by rare populations of leukemia stem cells (LSCs). Selective targeting of LSCs is a promising approach for treating AML and preventing relapse following chemotherapy, and developing such therapeutic modalities is a key priority. Here, we show that targeting telomerase activity eradicates AML LSCs. Genetic deletion of the telomerase subunit Terc in a retroviral mouse AML model induces cell-cycle arrest and apoptosis of LSCs, and depletion of telomerase-deficient LSCs is partially rescued by p53 knockdown. Murine Terc(-/-) LSCs express a specific gene expression signature that can be identified in human AML patient cohorts and is positively correlated with patient survival following chemotherapy. In xenografts of primary human AML, genetic or pharmacological inhibition of telomerase targets LSCs, impairs leukemia progression, and delays relapse following chemotherapy. Altogether, these results establish telomerase inhibition as an effective strategy for eliminating AML LSCs.

    Topics: Animals; Apoptosis; Cell Cycle Checkpoints; Cells, Cultured; Disease Models, Animal; Gene Expression Regulation, Neoplastic; Gene Knockout Techniques; Humans; Indoles; Leukemia, Myeloid, Acute; Mice; Mice, Inbred C57BL; Neoplastic Stem Cells; Niacinamide; Oligonucleotides; Recurrence; RNA, Small Interfering; Telomerase; Tumor Suppressor Protein p53; Xenograft Model Antitumor Assays

2014
Augmentation of radiation response by motesanib, a multikinase inhibitor that targets vascular endothelial growth factor receptors.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2010, Jul-15, Volume: 16, Issue:14

    Motesanib is a potent inhibitor of vascular endothelial growth factor receptors (VEGFR) 1, 2, and 3, platelet-derived growth factor receptor, and Kit receptors. In this report we examine the interaction between motesanib and radiation in vitro and in head and neck squamous cell carcinoma (HNSCC) xenograft models.. In vitro assays were done to assess the impact of motesanib on VEGFR2 signaling pathways in human umbilical vein endothelial cells (HUVEC). HNSCC lines grown as tumor xenografts in athymic nude mice were utilized to assess the in vivo activity of motesanib alone and in combination with radiation.. Motesanib inhibited VEGF-stimulated HUVEC proliferation in vitro, as well as VEGFR2 kinase activity. Additionally, motesanib and fractionated radiation showed additive inhibitory effects on HUVEC proliferation. In vivo combination therapy with motesanib and radiation showed increased response compared with drug or radiation alone in UM-SCC1 (P < 0.002) and SCC-1483 xenografts (P = 0.001); however, the combination was not significantly more efficacious than radiation alone in UM-SCC6 xenografts. Xenografts treated with motesanib showed a reduction of vessel penetration into tumor parenchyma, compared with control tumors. Furthermore, triple immunohistochemical staining for vasculature, proliferation, and hypoxia showed well-defined spatial relationships among these parameters in HNSCC xenografts. Motesanib significantly enhanced intratumoral hypoxia in the presence and absence of fractionated radiation.. These studies identify a favorable interaction when combining radiation and motesanib in HNSCC models. The data presented suggest that motesanib reduces blood vessel penetration into tumors and thereby increases intratumoral hypoxia. These findings suggest that clinical investigations examining combinations of radiation and motesanib are warranted in HNSCC.

    Topics: Animals; Antineoplastic Agents; Cell Proliferation; Disease Models, Animal; Drug Therapy, Combination; Endothelial Cells; Head and Neck Neoplasms; Humans; Indoles; Male; Mice; Mice, Nude; Neoplasm Transplantation; Niacinamide; Oligonucleotides; Structure-Activity Relationship; Vascular Endothelial Growth Factor Receptor-2; Xenograft Model Antitumor Assays

2010