Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Appl Lab Med ; 2(1): 4-16, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-33636955

ABSTRACT

BACKGROUND: Average telomere length in whole blood has become a biomarker of aging, disease, and mortality risk across a broad range of clinical conditions. The most common method of telomere length measurement for large patient sample sets is based on quantitative PCR (qPCR). For laboratory-developed tests to be performed on clinical samples, they must undergo a rigorous analytical validation, currently regulated under CLIA. METHODS: Whole blood samples from 40 donors were used in the analytical validation of methods for relative average telomere length (rATL) measurement. Three technical replicate DNA samples were extracted from each whole blood sample and placed in three independent wells on a sample plate. Each of these sample plates was assayed 12 times during the validation process. The study was conducted over a 20-day period, once in the morning and once in the evening, using 3 different operators. RESULTS: Our process of rATL measurement beginning with DNA extraction followed by qPCR-based assay resulted in repeatability and reproducibility CV of <5% and amplification efficiencies near 100%. The validated assay was used to establish a reference interval derived from 2 cohorts of individuals: (a) San Francisco Bay area (n = 504) and (b) a US cross-sectional, demographic population (n = 357). CONCLUSIONS: We present advances in the establishment of a highly reproducible analytically validated process for determining rATLs in a CLIA laboratory environment.

2.
Rejuvenation Res ; 19(6): 478-484, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26950204

ABSTRACT

TA-65 is a dietary supplement based on an improved formulation of a small molecule telomerase activator that was discovered in a systematic screening of natural product extracts from traditional Chinese medicines. This study summarizes the findings on telomere length (TL) changes from a randomized, double blind, placebo controlled study of TA-65 over a 1 year period. The study was conducted on 117 relatively healthy cytomegalovirus-positive subjects aged 53-87 years old. Subjects taking the low dose of TA-65 (250 U) significantly increased TL over the 12 months period (530 ± 180 bp; p = 0.005), whereas subjects in the placebo group significantly lost TL (290 ± 100 bp; p = 0.01). The high dose of TA-65 (1000 U) showed a trend of improvements in TL compared with that of the placebo group; however, the improvements did not reach statistical significance. TL changes in the low-dose group were similar for both median and 20th percentile TLs. The findings suggest that TA-65 can lengthen telomeres in a statistically and possibly clinically significant manner.


Subject(s)
Biological Products/pharmacology , Medicine, Chinese Traditional , Telomerase/physiology , Telomere/drug effects , Aged , Aged, 80 and over , Cross-Sectional Studies , Dose-Response Relationship, Drug , Double-Blind Method , Enzyme Activation/drug effects , Humans , Middle Aged
3.
Clin Ophthalmol ; 10: 243-9, 2016.
Article in English | MEDLINE | ID: mdl-26869760

ABSTRACT

PURPOSE: Telomere attrition and corresponding cellular senescence of the retinal pigment epithelium contribute to the changes of age-related macular degeneration. Activation of the enzyme telomerase can add telomeric DNA to retinal pigment epithelium chromosomal ends and has been proposed as a treatment for age-related macular degeneration. We report the use of a small molecule, oral telomerase activator (TA)-65 in early macular degeneration. This study, focusing on early macular degeneration, provides a model for the use of TAs in age-related disease. METHOD: Thirty-eight (38) patients were randomly assigned to a 1-year, double-blinded, placebo-controlled interventional study with arms for oral TA-65 or placebo. Macular functions via micro-perimetry were the primary measured outcomes. RESULTS: The macular function in the arm receiving the TA-65 showed significant improvement relative to the placebo control. The improvement was manifest at 6 months and was maintained at 1 year: macular threshold sensitivity (measured as average dB [logarithmic decibel scale of light attenuation]) improved 0.97 dB compared to placebo (P-value 0.02) and percent reduced thresholds lessened 8.2% compared to the placebo arm (P-value 0.04). CONCLUSION: The oral TA significantly improved the macular function of treatment subjects compared to controls. Although this study was a pilot and a larger study is being planned, it is noteworthy in that it is, to our knowledge, the first randomized placebo-controlled study of a TA supplement.

4.
Neurosignals ; 22(1): 52-63, 2014.
Article in English | MEDLINE | ID: mdl-25095809

ABSTRACT

Cycloastragenol (CAG) is an aglycone of astragaloside IV. It was first identified when screening Astragalus membranaceus extracts for active ingredients with antiaging properties. The present study demonstrates that CAG stimulates telomerase activity and cell proliferation in human neonatal keratinocytes. In particular, CAG promotes scratch wound closure of human neonatal keratinocyte monolayers in vitro. The distinct telomerase-activating property of CAG prompted evaluation of its potential application in the treatment of neurological disorders. Accordingly, CAG induced telomerase activity and cAMP response element binding (CREB) activation in PC12 cells and primary neurons. Blockade of CREB expression in neuronal cells by RNA interference reduced basal telomerase activity, and CAG was no longer efficacious in increasing telomerase activity. CAG treatment not only induced the expression of bcl2, a CREB-regulated gene, but also the expression of telomerase reverse transcriptase in primary cortical neurons. Interestingly, oral administration of CAG for 7 days attenuated depression-like behavior in experimental mice. In conclusion, CAG stimulates telomerase activity in human neonatal keratinocytes and rat neuronal cells, and induces CREB activation followed by tert and bcl2 expression. Furthermore, CAG may have a novel therapeutic role in depression.


Subject(s)
Depression/drug therapy , Neurons/drug effects , Neurons/metabolism , Sapogenins/administration & dosage , Telomerase/metabolism , Animals , Antidepressive Agents/administration & dosage , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Nerve Growth Factor/metabolism , PC12 Cells , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Sapogenins/chemical synthesis
5.
Rejuvenation Res ; 16(5): 386-95, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23808324

ABSTRACT

A short average telomere length is associated with low telomerase activity and certain degenerative diseases. Studies in animals and with human cells confirm a causal mechanism for cell or tissue dysfunction triggered by critically short telomeres, suggesting that telomerase activation may be an approach to health maintenance. Previously, we reported on positive immune remodeling in humans taking a commercial health maintenance program, PattonProtocol-1, composed of TA-65® (a natural product-derived telomerase activator) and other dietary supplements. In over a 5-year period and an estimated 7000 person-years of use, no adverse events or effects have been attributed to TA-65 by physicians licensed to sell the product. Here we report on changes in metabolic markers measured at baseline (n=97-107 subjects) and every 3-6 months (n=27-59 subjects) during the first 12 months of study. Rates of change per year from baseline determined by a multi-level model were -3.72 mg/dL for fasting glucose (p=0.02), -1.32 mIU/mL for insulin (p=0.01), -13.2 and -11.8 mg/dL for total cholesterol and low-density lipoprotein cholesterol (LDL-C) (p=0.002, p=0.002, respectively), -17.3 and -4.2 mmHg for systolic and diastolic blood pressure (p=0.007 and 0.001, respectively), and -3.6 µmole/L homocysteine (p=0.001). In a subset of individuals with bone mineral density (BMD) measured at baseline and 12 months, density increased 2.0% in the spine (p=0.003). We conclude that in addition to apparent positive immune remodeling, PattonProtocol-1 may improve markers of metabolic, bone, and cardiovascular health.


Subject(s)
Biological Products/pharmacology , Cardiovascular System/drug effects , Cardiovascular System/metabolism , Enzyme Activators/pharmacology , Health , Telomerase/metabolism , Aging/blood , Aging/drug effects , Aging/metabolism , Biomarkers/blood , Blood Glucose/metabolism , Blood Pressure/drug effects , Bone Density/drug effects , Cholesterol/blood , Female , Humans , Inflammation/blood , Male , Middle Aged , Multilevel Analysis , Vitamins/blood
6.
PLoS One ; 8(3): e58423, 2013.
Article in English | MEDLINE | ID: mdl-23516479

ABSTRACT

The emergence of diseases associated with telomere dysfunction, including AIDS, aplastic anemia and pulmonary fibrosis, has bolstered interest in telomerase activators. We report identification of a new small molecule activator, GRN510, with activity ex vivo and in vivo. Using a novel mouse model, we tested the potential of GRN510 to limit fibrosis induced by bleomycin in mTERT heterozygous mice. Treatment with GRN510 at 10 mg/kg/day activated telomerase 2-4 fold both in hematopoietic progenitors ex vivo and in bone marrow and lung tissue in vivo, respectively. Telomerase activation was countered by co-treatment with Imetelstat (GRN163L), a potent telomerase inhibitor. In this model of bleomycin-induced fibrosis, treatment with GRN510 suppressed the development of fibrosis and accumulation of senescent cells in the lung via a mechanism dependent upon telomerase activation. Treatment of small airway epithelial cells (SAEC) or lung fibroblasts ex vivo with GRN510 revealed telomerase activating and replicative lifespan promoting effects only in the SAEC, suggesting that the mechanism accounting for the protective effects of GRN510 against induced lung fibrosis involves specific types of lung cells. Together, these results support the use of small molecule activators of telomerase in therapies to treat idiopathic pulmonary fibrosis.


Subject(s)
Enzyme Activators/pharmacology , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Lung/drug effects , Lung/pathology , Sapogenins/pharmacology , Telomerase/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Bleomycin/adverse effects , Cellular Senescence/drug effects , Collagen/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activators/administration & dosage , Fibroblasts/drug effects , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Lung/metabolism , Mice , Respiratory Mucosa/drug effects , Respiratory Mucosa/enzymology , Sapogenins/administration & dosage
7.
Planta Med ; 78(2): 115-21, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22083896

ABSTRACT

Two Chinese herb-derived small molecule telomerase activators, astragaloside IV (AG-IV) and cycloastragenol (CAG), have recently been shown to improve the proliferative response of CD8+ T lymphocytes from HIV-infected patients by upregulating telomerase activity. Here, we examined the signaling mechanism of AG-IV and CAG. Telomerase activity in human embryonic kidney HEK293 fibroblasts was increased upon treatment with increasing concentrations of AG-IV or CAG. Both compounds induced the phosphorylation of extracellular signal-regulated protein kinase (ERK) in a time- and dose-dependent manner in HEK293 cells and HEK-neo keratinocytes. AG-IV and CAG also stimulated ERK phosphorylation in other cell lines of lung, brain, mammary, endothelial, and hematopoietic origins. Use of selective inhibitors and dominant negative mutants revealed the involvement of c-Src, MEK (ERK kinase), and epidermal growth factor receptor in CAG-induced ERK phosphorylation. Our data indicate that AG-IV and CAG may exert their cellular effects through the activation of the Src/MEK/ERK pathway.


Subject(s)
Astragalus Plant/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Plant Extracts/pharmacology , Sapogenins/pharmacology , Saponins/pharmacology , Telomerase/metabolism , Triterpenes/pharmacology , Brain/drug effects , Brain/metabolism , Breast/drug effects , Breast/metabolism , Cell Line , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , ErbB Receptors/metabolism , Humans , Lung/drug effects , Lung/metabolism , Phosphorylation , src-Family Kinases/metabolism
8.
Aging Cell ; 10(4): 604-21, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21426483

ABSTRACT

Here, we show that a small-molecule activator of telomerase (TA-65) purified from the root of Astragalus membranaceus is capable of increasing average telomere length and decreasing the percentage of critically short telomeres and of DNA damage in haploinsufficient mouse embryonic fibroblasts (MEFs) that harbor critically short telomeres and a single copy of the telomerase RNA Terc gene (G3 Terc(+/-) MEFs). Importantly, TA-65 does not cause telomere elongation or rescue DNA damage in similarly treated telomerase-deficient G3 Terc(-/-) littermate MEFs. These results indicate that TA-65 treatment results in telomerase-dependent elongation of short telomeres and rescue of associated DNA damage, thus demonstrating that TA-65 mechanism of action is through the telomerase pathway. In addition, we demonstrate that TA-65 is capable of increasing mouse telomerase reverse transcriptase levels in some mouse tissues and elongating critically short telomeres when supplemented as part of a standard diet in mice. Finally, TA-65 dietary supplementation in female mice leads to an improvement of certain health-span indicators including glucose tolerance, osteoporosis and skin fitness, without significantly increasing global cancer incidence.


Subject(s)
Anticarcinogenic Agents/pharmacology , Neoplasms/prevention & control , Telomerase/metabolism , Telomere/drug effects , Animals , Astragalus propinquus/chemistry , DNA Damage , Diet , Embryo, Mammalian/cytology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/metabolism , RNA/genetics , RNA/metabolism , Telomerase/genetics , Telomere/metabolism , Telomere/ultrastructure
9.
Clin Cancer Res ; 17(1): 111-21, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21208905

ABSTRACT

PURPOSE: Cancer recurrence is one of the major setbacks in oncology. Maintaining telomeres is essential for sustaining the limitless replicative potential of such cancers. Because telomerase is thought to be active in all tumor cells and normal stem cells, telomerase inhibition may be nonspecific and have detrimental effects on tissue maintenance and development by affecting normal stem cell self-renewal. METHODS: We examined telomerase activity, telomere maintenance, and stem cell maturation in tumor subpopulations from freshly resected gliomas, long-term, primary, neural tumor-initiating cells (TIC) and corresponding normal stem cell lines. We then tested the efficacy of the telomerase inhibitor Imetelstat on propagation and self-renewal capacity of TIC and normal stem cells in vitro and in vivo. RESULTS: Telomerase was undetectable in the majority of tumor cells and specific to the TIC subpopulation that possessed critically short telomeres. In contrast, normal tissue stem cells had longer telomeres and undetectable telomerase activity and were insensitive to telomerase inhibition, which results in proliferation arrest, cell maturation, and DNA damage in neural TIC. Significant survival benefit and late tumor growth arrest of neuroblastoma TIC were observed in a xenograft model (P = 0.02). Furthermore, neural TIC exhibited irreversible loss of self-renewal and stem cell capabilities even after cessation of treatment in vitro and in vivo. CONCLUSIONS: TIC exhaustion with telomerase inhibition and lack of telomerase dependency in normal stem cells add new dimensions to the telomere hypothesis and suggest that targeting TIC with telomerase inhibitors may represent a specific and safe therapeutic approach for tumors of neural origin.


Subject(s)
Glioma/therapy , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Neuroblastoma/therapy , Telomerase/antagonists & inhibitors , Telomere/metabolism , Animals , Cell Line , Cell Proliferation , Glioma/enzymology , Glioma/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neural Crest , Neuroblastoma/enzymology , Neuroblastoma/metabolism , Telomerase/metabolism
10.
Rejuvenation Res ; 14(1): 45-56, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20822369

ABSTRACT

Most human cells lack sufficient telomerase to maintain telomeres, hence these genetic elements shorten with time and stress, contributing to aging and disease. In January, 2007, a commercial health maintenance program, PattonProtocol-1, was launched that included a natural product-derived telomerase activator (TA-65®, 10-50 mg daily), a comprehensive dietary supplement pack, and physician counseling/laboratory tests at baseline and every 3-6 months thereafter. We report here analysis of the first year of data focusing on the immune system. Low nanomolar levels of TA-65® moderately activated telomerase in human keratinocytes, fibroblasts, and immune cells in culture; similar plasma levels of TA-65® were achieved in pilot human pharmacokinetic studies with single 10- to 50-mg doses. The most striking in vivo effects were declines in the percent senescent cytotoxic (CD8(+)/CD28(-)) T cells (1.5, 4.4, 8.6, and 7.5% at 3, 6, 9, and 12 months, respectively; p = not significant [N.S.], 0.018, 0.0024, 0.0062) and natural killer cells at 6 and 12 months (p = 0.028 and 0.00013, respectively). Most of these decreases were seen in cytomegalovirus (CMV) seropositive subjects. In a subset of subjects, the distribution of telomere lengths in leukocytes at baseline and 12 months was measured. Although mean telomere length did not increase, there was a significant reduction in the percent short (<4 kbp) telomeres (p = 0.037). No adverse events were attributed to PattonProtocol-1. We conclude that the protocol lengthens critically short telomeres and remodels the relative proportions of circulating leukocytes of CMV(+) subjects toward the more "youthful" profile of CMV(-) subjects. Controlled randomized trials are planned to assess TA-65®-specific effects in humans.


Subject(s)
Biological Products/pharmacology , Enzyme Activators/pharmacology , Health , Telomerase/metabolism , Cell Count , Cells, Cultured , Cytomegalovirus/drug effects , Female , Fetus/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Humans , Immune System/drug effects , Infant, Newborn , Keratinocytes/drug effects , Keratinocytes/enzymology , Male , Middle Aged , Telomere/metabolism , Time Factors
11.
Nat Protoc ; 5(9): 1596-607, 2010 Sep.
Article in English | MEDLINE | ID: mdl-21085125

ABSTRACT

In this protocol we describe a method to obtain telomere length parameters using Southern blots of terminal restriction fragments (TRFs). We use this approach primarily for epidemiological studies that examine leukocyte telomere length. However, the method can be adapted for telomere length measurements in other cells whose telomere lengths are within its detection boundaries. After extraction, DNA is inspected for integrity, digested, resolved by gel electrophoresis, transferred to a membrane, hybridized with labeled probes and exposed to X-ray film using chemiluminescence. Although precise and highly accurate, the method requires a considerable amount of DNA (3 µg per sample) and it measures both the canonical and noncanonical components of telomeres. The method also provides parameters of telomere length distribution in each DNA sample, which are useful in answering questions beyond those focusing on the mean length of telomeres in a given sample. A skilled technician can measure TRF length in ∼130 samples per week.


Subject(s)
Blotting, Southern , Genetic Techniques , Telomere/genetics , Epidemiologic Studies , Humans , Restriction Mapping
12.
Cancer Res ; 70(22): 9494-504, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21062983

ABSTRACT

Cancer stem cells (CSC) are rare drug-resistant cancer cell subsets proposed to be responsible for the maintenance and recurrence of cancer and metastasis. Telomerase is constitutively active in both bulk tumor cell and CSC populations but has only limited expression in normal tissues. Thus, inhibition of telomerase has been shown to be a viable approach in controlling cancer growth in nonclinical studies and is currently in phase II clinical trials. In this study, we investigated the effects of imetelstat (GRN163L), a potent telomerase inhibitor, on both the bulk cancer cells and putative CSCs. When breast and pancreatic cancer cell lines were treated with imetelstat in vitro, telomerase activity in the bulk tumor cells and CSC subpopulations were inhibited. Additionally, imetelstat treatment reduced the CSC fractions present in the breast and pancreatic cell lines. In vitro treatment with imetelstat, but not control oligonucleotides, also reduced the proliferation and self-renewal potential of MCF7 mammospheres and resulted in cell death after <4 weeks of treatment. In vitro treatment of PANC1 cells showed reduced tumor engraftment in nude mice, concomitant with a reduction in the CSC levels. Differences between telomerase activity expression levels or telomere length of CSCs and bulk tumor cells in these cell lines did not correlate with the increased sensitivity of CSCs to imetelstat, suggesting a mechanism of action independent of telomere shortening for the effects of imetelstat on the CSC subpopulations. Our results suggest that imetelstat-mediated depletion of CSCs may offer an alternative mechanism by which telomerase inhibition may be exploited for cancer therapy.


Subject(s)
Indoles/pharmacology , Neoplastic Stem Cells/drug effects , Niacinamide/analogs & derivatives , Telomerase/antagonists & inhibitors , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Flow Cytometry , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/enzymology , Niacinamide/pharmacology , Oligonucleotides , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Telomerase/metabolism , Xenograft Model Antitumor Assays
13.
PLoS One ; 5(9)2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20824134

ABSTRACT

BACKGROUND: Plasma cells constitute the majority of tumor cells in multiple myeloma (MM) but lack the potential for sustained clonogenic growth. In contrast, clonotypic B cells can engraft and recapitulate disease in immunodeficient mice suggesting they serve as the MM cancer stem cell (CSC). These tumor initiating B cells also share functional features with normal stem cells such as drug resistance and self-renewal potential. Therefore, the cellular processes that regulate normal stem cells may serve as therapeutic targets in MM. Telomerase activity is required for the maintenance of normal adult stem cells, and we examined the activity of the telomerase inhibitor imetelstat against MM CSC. Moreover, we carried out both long and short-term inhibition studies to examine telomere length-dependent and independent activities. METHODOLOGY/PRINCIPAL FINDINGS: Human MM CSC were isolated from cell lines and primary clinical specimens and treated with imetelstat, a specific inhibitor of the reverse transcriptase activity of telomerase. Two weeks of exposure to imetelstat resulted in a significant reduction in telomere length and the inhibition of clonogenic MM growth both in vitro and in vivo. In addition to these relatively long-term effects, 72 hours of imetelstat treatment inhibited clonogenic growth that was associated with MM CSC differentiation based on expression of the plasma cell antigen CD138 and the stem cell marker aldehyde dehydrogenase. Short-term treatment of MM CSC also decreased the expression of genes typically expressed by stem cells (OCT3/4, SOX2, NANOG, and BMI1) as revealed by quantitative real-time PCR. CONCLUSIONS: Telomerase activity regulates the clonogenic growth of MM CSC. Moreover, reductions in MM growth following both long and short-term telomerase inhibition suggest that it impacts CSC through telomere length-dependent and independent mechanisms.


Subject(s)
Cell Proliferation , Multiple Myeloma/enzymology , Multiple Myeloma/physiopathology , Telomerase/metabolism , Telomere/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Clone Cells , Down-Regulation , Humans , Mice , Mice, SCID , Multiple Myeloma/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/metabolism , Tumor Cells, Cultured
14.
Drug Metab Pharmacokinet ; 25(5): 477-86, 2010.
Article in English | MEDLINE | ID: mdl-20877137

ABSTRACT

Cycloastragenol (CAG) is the aglycone derivative of astragaloside IV which has recently been demonstrated to activate telomerase and represents a potential drug candidate for the treatment of degenerative diseases. In the present study, intestinal absorption and metabolism of CAG were examined using the Caco-2 model and liver microsomes, respectively. The results showed that CAG rapidly passes through the Caco-2 cell monolayer by passive diffusion. Four different glucuronide conjugates and two oxidized CAG metabolites were found in the apical and basolateral sides of Caco-2 monolayer, suggesting that first-pass intestinal metabolism of CAG might occur upon passage through the intestinal epithelium. CAG underwent extensive metabolism in rat and human liver microsomes with only 17.4% and 8.2%, respectively, of the starting amount of CAG remaining after 30 min of incubation. Monohydroxylation of the parent and oxidization of the hydroxylated CAG were found in the liver samples. The present study indicates that CAG is efficiently absorbed through intestinal epithelium. However, extensive first-pass hepatic metabolism would limit the oral bioavailability of this compound.


Subject(s)
Enzyme Activators/metabolism , Intestinal Absorption/physiology , Intestinal Mucosa/metabolism , Liver/metabolism , Sapogenins/metabolism , Telomerase , Animals , Caco-2 Cells , Carrier Proteins/antagonists & inhibitors , Diffusion/drug effects , Egtazic Acid/pharmacology , Enterocytes/drug effects , Enterocytes/metabolism , Glucuronides/metabolism , Humans , Hydroxylation , Intestinal Absorption/drug effects , Kinetics , Male , Metabolic Detoxication, Phase I/physiology , Metabolic Detoxication, Phase II/physiology , Microsomes, Liver/metabolism , Oxidation-Reduction , Permeability/drug effects , Rats , Species Specificity , Spectrometry, Mass, Electrospray Ionization , Transcytosis/drug effects , Transcytosis/physiology
15.
Recent Results Cancer Res ; 184: 221-34, 2010.
Article in English | MEDLINE | ID: mdl-20072842

ABSTRACT

Telomeres and telomerase play essential roles in the regulation of the lifespan of human cells. While normal human somatic cells do not or only transiently express telomerase and therefore shorten their telomeres with each cell division, most human cancer cells typically express high levels of telomerase and show unlimited cell proliferation. High telomerase expression allows cells to proliferate and expand long-term and therefore supports tumor growth. Owing to the high expression and its role, telomerase has become an attractive diagnostic and therapeutic cancer target. Imetelstat (GRN163L) is a potent and specific telomerase inhibitor and so far the only drug of its class in clinical trials. Here, we report on the structure and the mechanism of action of imetelstat as well as about the preclinical and clinical data and future prospects using imetelstat in cancer therapy.


Subject(s)
Neoplasms/drug therapy , Oligopeptides/therapeutic use , Telomerase/antagonists & inhibitors , Animals , Clinical Trials as Topic , Humans , Oligonucleotides , Telomerase/chemistry , Telomerase/physiology
16.
J Immunol ; 181(10): 7400-6, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981163

ABSTRACT

Telomerase reverse transcribes telomere DNA onto the ends of linear chromosomes and retards cellular aging. In contrast to most normal somatic cells, which show little or no telomerase activity, immune cells up-regulate telomerase in concert with activation. Nevertheless, during aging and chronic HIV-1 infection, there are high proportions of dysfunctional CD8(+) CTL with short telomeres, suggesting that telomerase is limiting. The present study shows that exposure of CD8(+) T lymphocytes from HIV-infected human donors to a small molecule telomerase activator (TAT2) modestly retards telomere shortening, increases proliferative potential, and, importantly, enhances cytokine/chemokine production and antiviral activity. The enhanced antiviral effects were abrogated in the presence of a potent and specific telomerase inhibitor, suggesting that TAT2 acts primarily through telomerase activation. Our study is the first to use a pharmacological telomerase-based approach to enhance immune function, thus directly addressing the telomere loss immunopathologic facet of chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , HIV Infections/metabolism , Sapogenins/pharmacology , Telomerase/drug effects , CD8-Positive T-Lymphocytes/immunology , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Oligonucleotides , Oligopeptides/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/metabolism
17.
Nat Rev Cancer ; 8(3): 167-79, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18256617

ABSTRACT

Telomerase is an attractive cancer target as it appears to be required in essentially all tumours for immortalization of a subset of cells, including cancer stem cells. Moreover, differences in telomerase expression, telomere length and cell kinetics between normal and tumour tissues suggest that targeting telomerase would be relatively safe. Clinical trials are ongoing with a potent and specific telomerase inhibitor, GRN163L, and with several versions of telomerase therapeutic vaccines. The prospect of adding telomerase-based therapies to the growing list of new anticancer products is promising, but what are the advantages and limitations of different approaches, and which patients are the most likely to respond?


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/therapy , Telomerase/physiology , Antineoplastic Agents/pharmacology , Cancer Vaccines , Clinical Trials as Topic , Enzyme Inhibitors/pharmacology , Humans , Immunotherapy/methods , Kinetics , Medical Oncology/methods , Models, Biological , Oligonucleotides , Oligopeptides/pharmacology , Telomerase/antagonists & inhibitors , Telomerase/metabolism
18.
Nucleosides Nucleotides Nucleic Acids ; 26(10-12): 1577-9, 2007.
Article in English | MEDLINE | ID: mdl-18066830

ABSTRACT

Telomerase is one of the key enzymes responsible for the proliferative immortality of the majority of cancer cells. We recently introduced a new telomerase inhibitor, a 13-mer oligonucleotide N3' --> P5'-thio-phosphoramidate lipid conjugate, designated as GRN163L. This compound inhibits telomerase activity in various tumor cell lines with IC(50) values of 3-300 nM without any cellular uptake enhancers. GRN163L demonstrated potent and sequence specific anti-cancer activity in vivo in multiple animal models. This compound was able to significantly affect not only the growth of primary tumors, but also the spread and proliferation of metastases. GRN163L is currently in Phase I and Phase I/II clinical studies in patients with solid tumors and CLL, respectively.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Oligopeptides/pharmacology , Telomerase/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/therapeutic use , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mice , Oligonucleotides , Oligopeptides/therapeutic use , Xenograft Model Antitumor Assays
19.
Hum Gene Ther ; 17(6): 651-60, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16776573

ABSTRACT

Chronic ischemic wounds are major clinical problems, and are especially prevalent in elderly patients. Management of these wounds costs billions of dollars annually in the United States. Because of the severe impairment in tissue repair, ischemic wounds among the aged are major challenges for physicians. For example, transforming growth factor-beta1 stimulates healing of young patients' ischemic wounds, but it is totally ineffective in treating the ischemic wounds of aged patients. Therefore, our goal is to develop a better therapeutic strategy for elderly patient ischemic wounds. Because human telomerase reverse transcriptase (hTERT) has emerged as having a role in promoting cell proliferation, we hypothesized that hTERT overexpression may improve ischemic wound healing in the elderly. We successfully tested this hypothesis by demonstrating for the first time that gene delivery of hTERT by adenovirus (Ad-hTERT) dramatically improved ischemic wound healing in an aged rabbit model. Importantly, our histological data indicate that no deleterious immune response was induced in the aged rabbits. This finding has broad implications for the field of gene therapy because the foremost obstacle in the use of adenoviral vectors for gene therapy is that they provoke strong innate and adaptive immune responses in the host. Moreover, Ad-hTERT significantly improved survival of primary rabbit dermal fibroblasts that were treated with hypoxia and hydrogen peroxide (oxidative stress). This model is clinically relevant because it simulates the ischemia cycle of an ischemia-reperfusion injury, which can lead to stroke, myocardial infarction, and other tissue injuries. We conclude that Ad-hTERT is an effective and novel approach to treating the ischemic wounds of elderly patients.


Subject(s)
Adenoviridae/genetics , DNA-Binding Proteins/genetics , Gene Transfer Techniques , Genetic Therapy , Ischemia/therapy , Telomerase/genetics , Wound Healing , Animals , Cells, Cultured , DNA-Binding Proteins/immunology , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/physiology , Fibroblasts/virology , Humans , Immunohistochemistry , Ischemia/pathology , Oxidants/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Skin/injuries , Skin/pathology , Skin Ulcer/pathology , Skin Ulcer/therapy , Telomerase/immunology , Wound Healing/genetics
20.
Hepatology ; 42(5): 1127-36, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16114043

ABSTRACT

Most cancer cells have an immortal growth capacity as a consequence of telomerase reactivation. Inhibition of this enzyme leads to increased telomere dysfunction, which limits the proliferative capacity of tumor cells; thus, telomerase inhibition represents a potentially safe and universal target for cancer treatment. We evaluated the potential of two thio-phosphoramidate oligonucleotide inhibitors of telomerase, GRN163 and GRN163L, as drug candidates for the treatment of human hepatoma. GRN163 and GRN163L were tested in preclinical studies using systemic administration to treat flank xenografts of different human hepatoma cell lines (Hep3B and Huh7) in nude mice. The studies showed that both GRN163 and GRN163L inhibited telomerase activity and tumor cell growth in a dose-dependent manner in vitro and in vivo. The potency and efficacy of the lipid-conjugated antagonist, GRN163L, was superior to the nonlipidated parent compound, GRN163. Impaired tumor growth in vivo was associated with critical telomere shortening, induction of telomere dysfunction, reduced rate of cell proliferation, and increased apoptosis in the treatment groups. In vitro, GRN163L administration led to higher prevalence of chromosomal telomere-free ends and DNA damage foci in both hepatoma cell lines. In addition, in vitro chemosensitivity assay showed that pretreatment with GRN163L increased doxorubicin sensitivity of Hep3B. In conclusion, our data support the development of GRN163L, a novel lipidated conjugate of the telomerase inhibitor GRN163, for systemic treatment of human hepatoma. In addition to limiting the proliferative capacity of hepatoma, GRN163L might also increase the sensitivity of this tumor type to conventional chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Oligonucleotides/pharmacology , Oligopeptides/pharmacology , Telomerase/antagonists & inhibitors , Anaphase , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Doxorubicin/pharmacology , Humans , Liver Neoplasms/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Telomere/genetics , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...