Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Kidney Int ; 105(5): 980-996, 2024 May.
Article in English | MEDLINE | ID: mdl-38423182

ABSTRACT

Collapsing focal segmental glomerulosclerosis (FSGS), also known as collapsing glomerulopathy (CG), is the most aggressive variant of FSGS and is characterized by a rapid progression to kidney failure. Understanding CG pathogenesis represents a key step for the development of targeted therapies. Previous work implicated the telomerase protein component TERT in CG pathogenesis, as transgenic TERT expression in adult mice resulted in a CG resembling that seen in human primary CG and HIV-associated nephropathy (HIVAN). Here, we used the telomerase-induced mouse model of CG (i-TERTci mice) to identify mechanisms to inhibit CG pathogenesis. Inactivation of WIP1 phosphatase, a p53 target acting in a negative feedback loop, blocked disease initiation in i-TERTci mice. Repression of disease initiation upon WIP1 deficiency was associated with senescence enhancement and required transforming growth factor-ß functions. The efficacy of a pharmacologic treatment to reduce disease severity in both i-TERTci mice and in a mouse model of HIVAN (Tg26 mice) was then assessed. Pharmacologic inhibition of WIP1 enzymatic activity in either the telomerase mice with CG or in the Tg26 mice promoted partial remission of proteinuria and ameliorated kidney histopathologic features. Histological as well as high-throughput sequencing methods further showed that selective inhibition of WIP1 does not promote kidney fibrosis or inflammation. Thus, our findings suggest that targeting WIP1 may be an effective therapeutic strategy for patients with CG.


Subject(s)
AIDS-Associated Nephropathy , Glomerulosclerosis, Focal Segmental , Renal Insufficiency , Telomerase , Adult , Humans , Mice , Animals , Glomerulosclerosis, Focal Segmental/pathology , Telomerase/therapeutic use , AIDS-Associated Nephropathy/pathology , Proteinuria , Renal Insufficiency/complications , Disease Models, Animal
2.
Int Immunopharmacol ; 126: 111225, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37988911

ABSTRACT

Therapeutic cancer vaccines are novel immuno-therapeutics, aiming to improve clinical outcomes with other immunotherapies. However, obstacles to their successful clinical development remain, which model-informed drug development approaches may address. UV1 is a telomerase based therapeutic cancer vaccine candidate being investigated in phase I clinical trials for multiple indications. We developed a mechanism-based model structure, using a nonlinear mixed-effects modeling techniques, based on longitudinal tumor sizes (sum of the longest diameters, SLD), UV1-specific immunological assessment (stimulation index, SI) and overall survival (OS) data obtained from a UV1 phase I trial including non-small cell lung cancer (NSCLC) patients and a phase I/IIa trial including malignant melanoma (MM) patients. The final structure comprised a mechanistic tumor growth dynamics (TGD) model, a model describing the probability of observing a UV1-specific immune response (SI ≥ 3) and a time-to-event model for OS. The mechanistic TGD model accounted for the interplay between the vaccine peptides, immune system and tumor. The model-predicted UV1-specific effector CD4+ T cells induced tumor shrinkage with half-lives of 103 and 154 days in NSCLC and MM patients, respectively. The probability of observing a UV1-specific immune response was mainly driven by the model-predicted UV1-specific effector and memory CD4+ T cells. A high baseline SLD and a high relative increase from nadir were identified as main predictors for a reduced OS in NSCLC and MM patients, respectively. Our model predictions highlighted that additional maintenance doses, i.e. UV1 administration for longer periods, may result in more sustained tumor size shrinkage.


Subject(s)
Cancer Vaccines , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Melanoma , Telomerase , Humans , Cancer Vaccines/therapeutic use , Telomerase/therapeutic use , Lung Neoplasms/pathology , Peptides/therapeutic use
3.
Chin J Physiol ; 66(3): 119-128, 2023.
Article in English | MEDLINE | ID: mdl-37322622

ABSTRACT

Glioblastoma (GB) is one of the most aggressive and malignant tumors of the central nervous system. Conventional treatment for GB requires surgical resection followed by radiotherapy combined with temozolomide chemotherapy; however, the median survival time is only 12-15 months. Angelica sinensis Radix (AS) is commonly used as a traditional medicinal herb or a food/dietary supplement in Asia, Europe, and North America. This study aimed to investigate the effect of AS-acetone extract (AS-A) on the progression of GB and the potential mechanisms underlying its effects. The results indicated that AS-A used in this study showed potency in growth inhibition of GB cells and reduction of telomerase activity. In addition, AS-A blocked the cell cycle at the G0/G1 phase by regulating the expression of p53 and p16. Furthermore, apoptotic morphology, such as chromatin condensation, DNA fragmentation, and apoptotic bodies, was observed in AS-A-treated cells, induced by the activation of the mitochondria-mediated pathway. In an animal study, AS-A reduced tumor volume and prolonged lifespans of mice, with no significant changes in body weight or obvious organ toxicity. This study confirmed the anticancer effects of AS-A by inhibiting cell proliferation, reducing telomerase activity, altering cell cycle progression, and inducing apoptosis. These findings suggest that AS-A has great potential for development as a novel agent or dietary supplement against GB.


Subject(s)
Glioblastoma , Telomerase , Humans , Mice , Animals , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Telomerase/metabolism , Telomerase/pharmacology , Telomerase/therapeutic use , Apoptosis , Cell Cycle Checkpoints , Cell Cycle , Cell Proliferation , Telomere/metabolism , Telomere/pathology , Mitochondria , Cell Line, Tumor
4.
Arch Immunol Ther Exp (Warsz) ; 71(1): 6, 2023 Feb 20.
Article in English | MEDLINE | ID: mdl-36807774

ABSTRACT

Colon cancer etiology involves a wide spectrum of genetic and epigenetic alterations, finding it challenging to find effective therapeutic strategies. Quercetin exhibits potent anti-proliferative/apoptotic properties. In the present study, we aimed to elucidate the anti-cancer and anti-aging effect of quercetin in colon cancer cell lines. The anti-proliferative effect of quercetin was assessed in vitro by CCK-8 in normal and colon cancer cell lines. To check the anti-aging potential of quercetin, collagenase, elastase, and hyaluronidase inhibitory activity assays were performed. The epigenetic and DNA damage assays were performed using the human NAD-dependent deacetylase Sirtuin-6, proteasome 20S, Klotho, Cytochrome-C, and telomerase ELISA kits. Furthermore, the aging-associated miRNA expression profiling was performed on colon cancer cells. The treatment with quercetin inhibited cell proliferation of colon cancer cells in a dose-dependent manner. Quercetin arrested colon cancer cell growth by modulating expression of aging proteins including Sirtuin-6 and Klotho and also by inhibiting telomerase activity to restrict the telomere length which is evident from qPCR analysis. Quercetin also exhibited DNA damage protection by reducing proteasome 20S levels. The miRNA expression profiling results displayed differential expression of miRNA in colon cancer cell, and in addition, the highly upregulated miRNA was involved in the regulation of cell cycle, proliferation, and transcription. Our data suggest that quercetin treatment inhibited cell proliferation in colon cancer cells through regulating the anti-aging protein expression and provides better understanding for quercetin's potential use in colon cancer treatment.


Subject(s)
Colonic Neoplasms , MicroRNAs , Sirtuins , Telomerase , Humans , Apoptosis , Cell Proliferation , Epigenesis, Genetic , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/pharmacology , Proteasome Endopeptidase Complex/therapeutic use , Quercetin/pharmacology , Quercetin/therapeutic use , Sirtuins/metabolism , Sirtuins/pharmacology , Sirtuins/therapeutic use , Telomerase/metabolism , Telomerase/pharmacology , Telomerase/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
5.
Biol Trace Elem Res ; 201(7): 3300-3310, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36192614

ABSTRACT

Tumour illness and its resistance against existing anticancer therapies pose a serious health concern globally despite the progressive advancement of therapeutic options. The prevailing treatment of HCC using numerous antitumor agents has inflated long-lived complete remissions, but a percentage of individuals still die due to disease recurrence, indicating a need for further exploration of possible anti-tumour regimes. We aim to boost the effectiveness of the HCC treatment by conducting current investigations evaluating the effect of arsenic trioxide (ATO) with different herbal compounds like quercetin and aloe-emodin against liver tumour via inhibition of telomerase, a pro-cancer enzyme. The anticancer activity of ATO with herbal compounds was investigated in human control liver cell line (Wrl-68) and cancer liver cell line (HepG2) at different time intervals. Viability and cytotoxicity in response to combinatorial drugs were assessed in vitro by trypan blue dye exclusion assay and MTT and WST assay. Apoptosis was analysed by annexin V/PI assay, and the expression of telomerase and apoptosis-regulating proteins was evaluated by immunoblotting and qRT-PCR. Arsenic trioxide in combination with quercetin and aloe-emodin reduced cell viability in cancerous cells compared to normal cells by inducing apoptosis, downregulating telomerase and Bcl-2 (anti-apoptotic protein) and upregulating the expression of Bax (pro-apoptotic protein). ATO exhibited significant anticancer effects due to the synergistic effects of quercetin and aloe-emodin in liver tumour cells. The current study data collectively suggest that a successful inhibition of cancer growth by the combination of ATO and tested herbal medicines against liver tumour growth is via the inhibition of telomerase activity.


Subject(s)
Antineoplastic Agents , Arsenic , Arsenicals , Carcinoma, Hepatocellular , Emodin , Liver Neoplasms , Telomerase , Humans , Arsenic Trioxide/pharmacology , Arsenic/metabolism , Liver Neoplasms/drug therapy , Telomerase/metabolism , Telomerase/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Arsenicals/pharmacology , Oxides/pharmacology , Oxides/metabolism , Emodin/pharmacology , Emodin/therapeutic use , Quercetin/pharmacology , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Apoptosis , Cell Proliferation
6.
Curr Hematol Malig Rep ; 17(5): 140-154, 2022 10.
Article in English | MEDLINE | ID: mdl-35984598

ABSTRACT

PURPOSE OF REVIEW: To discuss the current treatment paradigm, review novel targets, and summarize completed and ongoing clinical trials that may lead to a paradigm shifts in the management of myelofibrosis (MF). RECENT FINDINGS: In addition to the recent approval and ongoing late-stage development of multiple novel JAK inhibitors, recent clinical studies demonstrate therapeutic potential of targeting multiple alternate proteins and pathways including BET, MDM2, telomerase, BCL2, LSD1, PI3K, SMAC, and PTX2 in patients with MF. MF is a myeloproliferative neoplasm characterized by clonal proliferation of myeloid cells and bone marrow fibrosis often causing cytopenias, extramedullary hematopoiesis resulting in hepatosplenomegaly, and increased pro-inflammatory cytokine production driving systemic symptoms. A significant proportion of morbidity and mortality is related to the propensity to transform to acute leukemia. Allogeneic hematopoietic stem cell transplantation is the only curative therapy; however, due to the high associated mortality, this treatment is not an option for the majority of patients with MF. Currently, there are three targeted Food and Drug Administration (FDA)-approved therapies for MF which include ruxolitinib, fedratinib, and pacritinib, all part of the JAK inhibitor class. Many patients are unable to tolerate, do not respond, or develop resistance to existing therapies, leaving a large unmet medical need. In this review, we discuss the current treatment paradigm and novel therapies in development for the treatment of MF. We review the scientific rationale of each targeted pathway. We summarize updated clinical data and ongoing trials that may lead to FDA approval of these agents.


Subject(s)
Janus Kinase Inhibitors , Primary Myelofibrosis , Telomerase , Cytokines , Histone Demethylases , Humans , Janus Kinase Inhibitors/therapeutic use , Phosphatidylinositol 3-Kinases , Primary Myelofibrosis/diagnosis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-bcl-2 , Telomerase/therapeutic use
7.
Curr Hematol Malig Rep ; 17(5): 155-169, 2022 10.
Article in English | MEDLINE | ID: mdl-35932395

ABSTRACT

PURPOSE OF REVIEW: Estimating and modifying thrombotic risk is currently the mainstay of care for patients with polycythemia vera (PV) and essential thrombocythemia (ET). In recent years, however, increased attention has shifted towards quality of life and disease modification. In this review, we discuss recent advances in risk stratification, present updated results for ruxolitinib and interferon randomized clinical trials, discuss new approaches in antiplatelet and anticoagulant treatment, and summarize early phase trials of novel agents and emerging therapeutic concepts for the treatment of PV and ET. RECENT FINDINGS: International collaborations and novel technologies, i.e., next-generation sequencing and machine learning techniques, have demonstrated excellent abilities to improve thrombotic risk stratification in PV and ET. Updated results from ruxolitinib and interferon randomized clinical trials have confirmed excellent efficacy and safety of these agents, both as first- and second-line treatments. Early trials of novel agents (histone deacetylase inhibitors, telomerase inhibitors, lysine-specific demethylase-1 inhibitors, human double-minute 2 inhibitors, and hepcidin mimetics) have shown encouraging efficacy and safety in blood count control, reduction of splenomegaly, and alleviation of disease-related symptoms. Finally, accumulating evidence suggested that direct oral anticoagulants may be a valid therapeutic alternative to warfarin for prolonged thromboprophylaxis. International collaborations ("big data") with the help of new technologies represent an exciting new approach to analyze rare outcomes in rare diseases, especially for identifying novel prognostic biomarkers in PV and ET. Randomized clinical trials are also needed to fully elucidate whether novel agents may establish new standards of care.


Subject(s)
Polycythemia Vera , Telomerase , Thrombocythemia, Essential , Thrombosis , Venous Thromboembolism , Anticoagulants/adverse effects , Biomarkers , Hepcidins/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Humans , Interferons/therapeutic use , Lysine/therapeutic use , Nitriles , Polycythemia Vera/diagnosis , Polycythemia Vera/drug therapy , Pyrazoles , Pyrimidines , Quality of Life , Risk Assessment , Telomerase/therapeutic use , Thrombocythemia, Essential/diagnosis , Thrombocythemia, Essential/drug therapy , Warfarin/therapeutic use
8.
Lung Cancer ; 167: 98-106, 2022 05.
Article in English | MEDLINE | ID: mdl-35183375

ABSTRACT

INTRODUCTION: Among the different mechanisms of acquired resistance to EGFR tyrosine kinase inhibitors (TKIs) reported in EGFR-mutated lung adenocarcinoma (EGFR-LUAD) patients, histological transformation into small cell carcinoma (SCLC) occurs in 3-14% of resistant cases, regardless of the generation of EGFR-TKI. In recent studies, bi-allelic inactivation of TP53 and RB1 has been identified in a vast majority of transformed SCLCs. However, the molecular mechanisms driving this histologic transformation remain largely unknown, mainly due to the rarity of samples. PATIENTS AND METHODS: Out of an initial cohort of 64 patients, tumor tissues of adequate quality and quantity for whole exome sequencing (WES) analysis were available for nine tumors for six patients: paired pre- and post-SCLC transformation samples for three Patients and post-SCLC transformation samples for three other patients. RESULTS: Mutational analyses showed concurrent TP53 mutations and Rb pathway alterations in five of the six patients analyzed, confirming their suggested role as predisposing genetic alterations to SCLC transformation. In addition, TERT amplification was detected in four of the six patients and found to be an event acquired during SCLC transformation. Clonal history evolution analyses from the paired LUAD/SCLC samples showed different evolution patterns. In two patients, a large proportion of mutations were present in the most recent common ancestor cell of the initial LUAD and the transformed SCLC clones, whereas in the third patient, few clonal mutations were common between the LUAD and SCLC samples and the ancestor clone that lead to SCLC was present at low frequency in the initial LUAD. CONCLUSION: Despite varied clinical presentations and clonal history evolution patterns, in addition to p53 and Rb pathways alterations, TERT amplification emerged as another common genetic mechanism of EGFR-LUAD to SCLC transformation in our cohort, and could represent a candidate therapeutic target in this subset of SCLC tumors.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Small Cell , Lung Neoplasms , Small Cell Lung Carcinoma , Telomerase , Adenocarcinoma of Lung/pathology , Carcinoma, Small Cell/drug therapy , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Humans , Lung Neoplasms/pathology , Mutation , Protein Kinase Inhibitors/therapeutic use , Retinoblastoma Protein/metabolism , Small Cell Lung Carcinoma/drug therapy , Telomerase/genetics , Telomerase/therapeutic use , Tumor Suppressor Protein p53/genetics
9.
Front Immunol ; 11: 652, 2020.
Article in English | MEDLINE | ID: mdl-32508804

ABSTRACT

Previously, a telomerase-derived 16-mer peptide, GV1001, developed as an anticancer vaccine, was reported to exert antiviral effects on human immunodeficiency virus or hepatitis C virus in a heat shock protein-dependent manner. Here we investigated whether GV1001 exerts antiviral effects on hepatitis B virus (HBV) and elucidated its underlying mechanisms. GV1001 inhibited HBV replication and hepatitis B surface antigen (HBsAg) secretion in a dose-dependent manner, showing synergistic antiviral effects with nucleos(t)ide analogs (NAs) including entecavir and lamivudine. This peptide also inhibited viral cccDNA and pgRNA. The intravenous GV1001 treatment of transgenic mice had anti-HBV effects. Our mechanistic studies revealed that GV1001 suppresses HBV replication by inhibiting capsid formation via type I interferon-mediated induction of heme oxygenase-1 (HO-1). GV1001 promoted the mitochondrial DNA stress-mediated release of oxidized DNA into the cytosol, resulting in IFN-I-dependent anti-HBV effects via the STING-IRF3 axis. We found that the anti-HBV effect of GV1001 was due to its ability to penetrate into the cytosol via extracellular heat shock protein, leading to phagosomal escape-mediated mtDNA stress. We demonstrated that the cell-penetrating and cytosolic localization capacity of GV1001 results in antiviral effects on HBV infections via mtDNA stress-mediated IFN-I production. Thus, GV1001, a peptide proven to be safe for human use, may be an anti-HBV drug that can be synergistically used with nucleot(s)ide analog.


Subject(s)
Antiviral Agents/metabolism , DNA Damage/immunology , DNA, Mitochondrial/genetics , Hepatitis B virus/physiology , Hepatitis B/immunology , Interferon Type I/metabolism , Peptide Fragments/pharmacology , Telomerase/pharmacology , Animals , Drug Synergism , Guanine/analogs & derivatives , Guanine/pharmacology , Heme Oxygenase-1/metabolism , Hep G2 Cells , Hepatitis B/drug therapy , Hepatitis B Surface Antigens/metabolism , Humans , Interferon Regulatory Factor-3/metabolism , Lamivudine/pharmacology , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Peptide Fragments/therapeutic use , Phagosomes/metabolism , Signal Transduction , Telomerase/therapeutic use , Virus Replication
10.
Curr Aging Sci ; 13(1): 11-21, 2020.
Article in English | MEDLINE | ID: mdl-31544708

ABSTRACT

BACKGROUND: Although telomerase has potential for age-related disease intervention, the overexpression of telomerase in about 90% of cancers, and in HIV virus reservoirs, cautions against se in anti-aging telomerase therapeutics. While multiple reviews document the canonical function of telomerase for maintenance of telomeres, as well as an increasing numbers of reviews that reveal new non-canonical functions of telomerase, there was no systematic review that focuses on the array of associates of the subunit of Telomerase Reverse transcriptase protein (TERT) as pieces of the puzzle to assemble a picture of the how specific TERT complexes uniquely impact aging and age-related diseases and more can be expected. METHODS: A structured search of bibliographic data on TERT complexes was undertaken using databases from the National Center for Biotechnology Information Pubmed with extensive access to biomedical and genomic information in order to obtain a unique documented and cited overview of TERT complexes that may uniquely impact aging and age-related diseases. RESULTS: The TERT associations include proper folding, intracellular TERT transport, metabolism, mitochondrial ROS (Reactive Oxygen Species) regulation, inflammation, cell division, cell death, and gene expression, in addition to the well-known telomere maintenance. While increase of cell cycle inhibitors promote aging, in cancer, the cell cycle check-point regulators are ambushed in favor of cell proliferation, while cytoplasmic TERT protects a cell cycle inhibitor in oxidative stress. The oncogene cMyc regulates gene expression for overexpression of TERT, and reduction of cell cycle inhibitors-the perfect storm for cancer promotion. TERT binds with the oncogene RMRP RNA, and TERT-RMRP function can regulate levels of that oncogene RNA, and TERT in a TBN complex can regulate heterochromatin. Telomerase benefit and novel function in neurology and cardiology studies open new anti- aging hope. GV1001, a 16 amino acid peptide of TERT that associates with Heat Shock Proteins (HSP's), bypasses the cell membrane with remarkable anti disease potential. CONCLUSIONS: TERT "associates" are anti-cancer targets for downregulation, but upregulation in antiaging therapy. The overview revealed that unique TERT associations that impact all seven pillars of aging identified by the Trans-NIH Geroscience Initiative that influence aging and urge research for appropriate targeted telomerase supplements/ stimulation, and inclusion in National Institute on Aging Intervention Testing Program. The preference for use of available "smart drugs", targeted to only cancer, not off-target anti- aging telomerase is implied by the multiplicity of TERT associates functions.


Subject(s)
Aging/metabolism , Neoplasms/enzymology , Telomerase/metabolism , Telomere Homeostasis , Telomere Shortening , Telomere/metabolism , Age Factors , Aging/genetics , Aging/pathology , Animals , Cancer Vaccines/therapeutic use , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Peptide Fragments/therapeutic use , Telomerase/genetics , Telomerase/therapeutic use , Telomere/genetics
11.
Endocr Relat Cancer ; 26(2): 147-162, 2019 02.
Article in English | MEDLINE | ID: mdl-30400054

ABSTRACT

GV1001, a 16-amino acid fragment of the human telomerase reverse transcriptase catalytic subunit (hTERT), has been developed as an injectable formulation of cancer vaccine. Here, we revealed for the first time that GV1001 is a novel ligand for gonadotropin-releasing hormone receptor (GnRHR). The docking prediction for GV1001 against GnRHR showed high binding affinity. Binding of GV1001 to GnRHR stimulated the Gαs-coupled cAMP signaling pathway and antagonized Gαq-coupled Ca2+ release by leuprolide acetate (LA), a GnRHR agonist. Repeated injection of GV1001 attenuated both serum testosterone level and seminal vesicle weight via desensitization of hypothalamic-pituitary-gonadal (HPG) axis. We then tested whether GV1001 has an inhibitory effect on tumor growth of LNCaP cells, androgen receptor-positive human prostate cancer (PCa) cells. GV1001 significantly inhibited tumor growth and induced apoptosis in LNCaP-implanted xenografts. Interestingly, mRNA expressions of matrix metalloproteinase 2 and matrix metalloproteinase 9 were suppressed by GV1001, but not by LA. Moreover, GV1001 significantly inhibited the proliferation and migration of PCa cells and induced apoptosis in a concentration-dependent manner. Our findings suggest that GV1001 functions as a biased GnRHR ligand to selectively stimulate the Gαs/cAMP pathway, with anti-proliferative and anti-migratory effects on human PCa.


Subject(s)
Cancer Vaccines/therapeutic use , Peptide Fragments/therapeutic use , Prostatic Neoplasms/drug therapy , Receptors, LHRH/genetics , Telomerase/therapeutic use , Animals , Cancer Vaccines/pharmacology , Humans , Ligands , Male , Mice , Peptide Fragments/pharmacology , Prostatic Neoplasms/pathology , Signal Transduction , Telomerase/pharmacology
12.
Otol Neurotol ; 39(8): e731-e737, 2018 09.
Article in English | MEDLINE | ID: mdl-30015752

ABSTRACT

HYPOTHESIS: We tested whether GV1001 has any ototoxic side effects at different doses and whether it protects hearing in an aminoglycoside-induced ototoxicity mouse model. BACKGROUND: GV1001, a novel peptide vaccine currently being examined in a Phase 3 clinical trial to treat pancreatic cancer, also has anti-inflammatory and antioxidant effects. METHODS: In the first experiment, C57/BL6 mice were injected with GV1001 preparations at concentrations of 0.1 to 100 mg/kg for 7 days to evaluate the toxicity of GV1001 on the inner ear and kidneys. In the second experiment, the protective effect of GV1001 was tested in an ototoxicity mouse model that was generated by injecting 800 mg/kg kanamycin (KM) for 2 weeks. The hearing threshold and hair cell loss were compared between the KM + GV1001 group (treated with 10 mg/kg GV1001 for 2 wk) and the KM + saline group. The hearing threshold was measured before, and 7, 14, and 21 days after the initial treatment. The blood urea nitrogen level was measured. RESULTS: No ototoxicity or renal toxicity was found following treatment with different doses of GV1001 (0.1-100 mg/kg). The KM + saline group showed impaired auditory function and markedly disoriented and missing cochlear hair cells, while the KM + GV1001 group showed significant hearing and hair cell preservation in comparison (p < 0.05). CONCLUSION: GV1001 itself did not have any detrimental effects on the inner ear or kidney. In the KM induced ototoxicity model, concomitant administration of GV1001 protected against cochlear hair cell damage and preserve hearing.


Subject(s)
Anti-Bacterial Agents/adverse effects , Hearing Loss/prevention & control , Kanamycin/adverse effects , Peptide Fragments/therapeutic use , Telomerase/therapeutic use , Animals , Disease Models, Animal , Ear, Inner/drug effects , Hair Cells, Auditory/drug effects , Hearing/drug effects , Hearing Loss/chemically induced , Male , Mice , Vaccines, Subunit
13.
Mol Cancer Ther ; 17(7): 1504-1514, 2018 07.
Article in English | MEDLINE | ID: mdl-29654065

ABSTRACT

Brain tumors remain the leading cause of cancer-related deaths in children and often are associated with long-term sequelae among survivors of current therapies. Hence, there is an urgent need to identify actionable targets and to develop more effective therapies. Telomerase and telomeres play important roles in cancer, representing attractive therapeutic targets to treat children with poor-prognosis brain tumors such as diffuse intrinsic pontine glioma (DIPG), high-grade glioma (HGG), and high-risk medulloblastoma. We have previously shown that DIPG, HGG, and medulloblastoma frequently express telomerase activity. Here, we show that the telomerase-dependent incorporation of 6-thio-2'deoxyguanosine (6-thio-dG), a telomerase substrate precursor analogue, into telomeres leads to telomere dysfunction-induced foci (TIF) along with extensive genomic DNA damage, cell growth inhibition, and cell death of primary stem-like cells derived from patients with DIPG, HGG, and medulloblastoma. Importantly, the effect of 6-thio-dG is persistent even after drug withdrawal. Treatment with 6-thio-dG elicits a sequential activation of ATR and ATM pathways and induces G2-M arrest. In vivo treatment of mice bearing medulloblastoma xenografts with 6-thio-dG delays tumor growth and increases in-tumor TIFs and apoptosis. Furthermore, 6-thio-dG crosses the blood-brain barrier and specifically targets tumor cells in an orthotopic mouse model of DIPG. Together, our findings suggest that 6-thio-dG is a promising novel approach to treat therapy-resistant telomerase-positive pediatric brain tumors. Mol Cancer Ther; 17(7); 1504-14. ©2018 AACR.


Subject(s)
Brain Neoplasms/therapy , Brain Stem Neoplasms/therapy , Glioma/therapy , Telomerase/genetics , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/pathology , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Medulloblastoma/therapy , Mice , Neoplastic Stem Cells/drug effects , Prognosis , Telomerase/therapeutic use , Telomere/drug effects , Telomere/genetics , Thionucleosides/pharmacology , Xenograft Model Antitumor Assays
14.
J Biomed Sci ; 25(1): 22, 2018 Mar 12.
Article in English | MEDLINE | ID: mdl-29526163

ABSTRACT

BACKGROUND: Limitless self-renewal is one of the hallmarks of cancer and is attained by telomere maintenance, essentially through telomerase (hTERT) activation. Transcriptional regulation of hTERT is believed to play a major role in telomerase activation in human cancers. MAIN BODY: The dominant interest in telomerase results from its role in cancer. The role of telomeres and telomere maintenance mechanisms is well established as a major driving force in generating chromosomal and genomic instability. Cancer cells have acquired the ability to overcome their fate of senescence via telomere length maintenance mechanisms, mainly by telomerase activation. hTERT expression is up-regulated in tumors via multiple genetic and epigenetic mechanisms including hTERT amplifications, hTERT structural variants, hTERT promoter mutations and epigenetic modifications through hTERT promoter methylation. Genetic (hTERT promoter mutations) and epigenetic (hTERT promoter methylation and miRNAs) events were shown to have clinical implications in cancers that depend on hTERT activation. Knowing that telomeres are crucial for cellular self-renewal, the mechanisms responsible for telomere maintenance have a crucial role in cancer diseases and might be important oncological biomarkers. Thus, rather than quantifying TERT expression and its correlation with telomerase activation, the discovery and the assessment of the mechanisms responsible for TERT upregulation offers important information that may be used for diagnosis, prognosis, and treatment monitoring in oncology. Furthermore, a better understanding of these mechanisms may promote their translation into effective targeted cancer therapies. CONCLUSION: Herein, we reviewed the underlying mechanisms of hTERT regulation, their role in oncogenesis, and the potential clinical applications in telomerase-dependent cancers.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasms/genetics , Telomerase/genetics , Telomerase/therapeutic use , Humans
15.
Nefrología (Madr.) ; 37(6): 592-597, nov.-dic. 2017. graf, tab
Article in Spanish | IBECS | ID: ibc-168664

ABSTRACT

Antecedentes: Los mecanismos moleculares responsables del aumento de la mortalidad cardiovascular en la enfermedad renal crónica (ERC) asociada a la edad biológica no se conocen bien. Los estudios recientes apoyan la hipótesis de que los factores comunes responsables de este fenómeno son el envejecimiento celular y la disfunción telomérica. Objetivos: El objetivo de este estudio fue investigar la relación entre la actividad de la enzima telomerasa y los estadios de ERC. Métodos: El estudio incluyó a 120 pacientes que fueron seguidos para la ERC en los estadios 2-5D; cada estadio incluyó a 30 pacientes y a 30 voluntarios sanos sin ninguna enfermedad conocida que fueron admitidos en nuestro hospital para los controles periódicos. La actividad de la telomerasa en células mononucleares de sangre periférica (CMSP) se midió usando el método de TRAP. Resultados: Se observó una diferencia significativa en la actividad telomerasa en las CMSP entre los diferentes grupos. Los niveles más bajos fueron los del grupo de controles sanos (0,15±0,02) y los más altos los del grupo de pacientes con ERC en el estadio 5D (0,23±0,04). En los pacientes con ERC, la actividad telomerasa en las CMSP se correlacionó positivamente con el estadio de ERC y los niveles plasmáticos de potasio, hormona paratiroidea y creatinina, y se correlacionó negativamente con la tasa de filtración glomerular estimada (eTFG), el índice de masa corporal (IMC), el recuento de plaquetas y el calcio en suero. Los predictores independientes para la actividad telomerasa alta en pacientes con ERC fueron la eTFG y el IMC, de acuerdo con el análisis de regresión lineal. Conclusión: La actividad telomerasa en CMSP aumenta con el avance en el estadio de ERC. El aumento de la actividad telomerasa en CMSP se asocia con la eTFG y el IMC (AU)


Background: Molecular mechanisms of increased cardiovascular mortality in chronic kidney disease (CKD) associated with biological age are not well understood. Recent studies support the hypothesis that common factors responsible for this phenomenon are cellular aging and telomere dysfunction. Objectives: The purpose of this study was to investigate the relation between telomerase activity and CKD stages. Methods: The study included 120 patients who were followed-up for CKD stage 2-5D, composed of 30 patients of each stage and 30 healthy volunteers without any known disease who were admitted to our hospital for routine check-ups. Telomerase activity in peripheral blood mononuclear cells (PBMC) was measured using the TRAP assay. Results: A significant difference was observed for telomerase activity in PBMC between groups. The detected levels were lowest in the healthy control group (0.15±0.02), and highest in CKD stage 5D patients (0.23±0.04). In CKD patients, telomerase activity in PBMC was positively correlated with the CKD stage, serum creatinine, potassium and parathormone levels, and negatively correlated with estimated glomerular filtration rate (eGFR), body mass index (BMI), platelet count and serum calcium levels. According to the linear regression analysis, independent predictors for high telomerase activity in CKD patients were eGFR and BMI. Conclusion: Telomerase activity in PBMC increases with advancing CKD stage in CKD patients. Increased telomerase activity in PBMC is associated with eGFR and BMI(AU)


Subject(s)
Humans , Male , Adult , Middle Aged , Renal Insufficiency, Chronic/classification , Renal Insufficiency, Chronic/diagnosis , Telomerase/therapeutic use , Telomerase/metabolism , Renal Insufficiency, Chronic/enzymology , Linear Models , 28599
16.
Mol Med Rep ; 16(5): 7315-7320, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28944828

ABSTRACT

The potential cardioprotective effects of the novel vaccine peptide GV1001 were evaluated in myocardial ischemia­reperfusion injury induced rat models. GV1001 is a human telomerase reverse transcriptase derived peptide, which has been reported to possess both anti­tumor and anti­inflammatory effects. The normal saline (control group) and various concentrations (0.001­10 mg/kg) of GV1001 were administered directly to the right ventricle anterior wall before induction of ischemia. The was induced by Tightening the snare around the left anterior descending coronary artery for 40 min, before releasing the snare for 10 min induced the myocardial ischemia­reperfusion injury and was conducted in Sprague­Dawley rats. The area at risk, histology, apoptotic cells, neutrophils and inflammatory cytokines were analyzed from the excised heart tissue following myocardial ischemia­reperfusion injury. The area at risk was protected by concentrations of GV1001 equal to or higher than 0.01 mg/kg. At 0.1 mg/kg and higher concentrations of GV1001, the hemorrhage in the heart was attenuated, while severe congestion was reported in the control group. Apoptotic cells, myeloperoxidase activity and inflammatory cytokines [tumor necrosis factor (TNF)­α and interleukin (IL)­6] revealed decreased levels in a dose­dependent manner with respect to GV1001 concentration. The group treated with 10 mg/kg GV1001 demonstrated 59.73% apoptotic cells (P<0.001), 48.14% neutrophil contents (P<0.001), 55.63% TNF­α (P<0.01) and 42.35% IL­6 (P<0.01) levels, compared with the control group. The novel vaccine peptide GV1001 provided protective effects on myocardial ischemia­reperfusion injury and, therefore, it should be considered as an alternative potential anti­inflammatory agent for myocardial ischemia­reperfusion injury.


Subject(s)
Myocardial Reperfusion Injury/prevention & control , Peptide Fragments/therapeutic use , Protective Agents/therapeutic use , Telomerase/therapeutic use , Animals , Apoptosis/drug effects , Down-Regulation/drug effects , Enzyme-Linked Immunosorbent Assay , Hyperemia/etiology , Interleukin-6/analysis , Interleukin-6/metabolism , Male , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Neutrophils/cytology , Peptide Fragments/adverse effects , Peptide Fragments/pharmacology , Peroxidase/metabolism , Protective Agents/adverse effects , Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Telomerase/adverse effects , Telomerase/pharmacology , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/metabolism
17.
Int J Oncol ; 51(2): 478-488, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28627695

ABSTRACT

Loss of caudal type homeobox 2 (CDX2) is associated with the development of human colorectal cancer, while human telomerase reverse transcriptase (hTERT) frequently occurs in variety of human cancers. We investigated the effects of restoration of CDX2 expression using a hypoxia-inducible hTERT promoter-driven vector (pLVX-5HRE-hTERTp-CDX2-3FLAG) on colon cancer cell viability, cell cycle distribution, apoptosis, colony formation, invasion ability and xenograft tumor growth in nude mice. CDX2 overexpression significantly inhibited viability, colony formation, and the invasion and migration ability of LoVo cells, and induced cell cycle arrest and apoptosis in vitro, especially under hypoxic culture conditions. Overexpression of CDX2 under normoxic conditions significantly suppressed the expression of TGF-ß, cyclin D1, uPA, MMP-9, MMP-2, and Bcl-2, and stimulated the expression of collagen IV, laminin-1, and Bax. Overexpression of CDX2 reduced colon cancer xenograft tumor formation in nude mice which was associated with downregulation of Ki-67. In conclusion, overexpression of CDX2 using a hypoxia-inducible hTERT promoter-driven vector suppressed malignant progression of colon cancer cells in vitro and in vivo. These results suggest that pLVX-5HRE-hTERTp-CDX2-3FLAG gene therapy may be a promising novel approach to treat colon cancer.


Subject(s)
CDX2 Transcription Factor/genetics , Cell Hypoxia/genetics , Colonic Neoplasms/genetics , Genetic Therapy , Telomerase/genetics , Animals , CDX2 Transcription Factor/therapeutic use , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Genetic Vectors , Humans , Mice , Neoplasm Invasiveness/genetics , Promoter Regions, Genetic/genetics , Telomerase/therapeutic use , Xenograft Model Antitumor Assays
18.
Tumour Biol ; 39(4): 1010428317698341, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28443470

ABSTRACT

The abilities to escape apoptosis induced by anticancer drugs are an essential factor of carcinogenesis and a hallmark of resistance to cancer therapy. In this study, we identified hTERTR-FAM96A (human telomerase reverse transcriptase-family with sequence similarity 96 member A) as a new efficient agent for apoptosome-activating and anti-tumor protein and investigated the potential tumor suppressor function in hepatocellular carcinoma. The hTERTR-FAM96A fusion protein was constructed by genetic engineering and its anticancer function of hTERTR-FAM96A was explored in vitro and in vivo by investigating the possible preclinical outcomes. Effects of hTERTR-FAM96A on improvement of apoptotic sensitivity and inhibition of migration and invasion were examined in cancer cells and tumors. Our results showed that the therapeutic effects of hTERTR-FAM96A were highly effective for inhibiting tumor growth and inducing apoptosis of hepatocellular carcinoma cells in H22-bearing nude mice. The hTERTR-FAM96A fusion protein could specifically bind with Apaf-1 and hTERT, which further induced apoptosis of hepatocellular carcinoma cells and improved apoptosis sensitivity. Our results indicated that hTERTR-FAM96A treatment enhanced cytotoxic effects by upregulation of cytotoxic T lymphocyte responses, interferon-γ release, and T lymphocyte infiltration. In addition, hTERTR-FAM96A led to tumor-specific immunologic cytotoxicity through increasing apoptotic body on hepatocellular tumors. Furthermore, hTERTR-FAM96A dramatically inhibited tumor growth, reduced death rate, and prolonged mice survival in hepatocellular carcinoma mice derived from three independent hepatocellular carcinoma mice cohorts compared to control groups. In summary, our data suggest that hTERTR-FAM96A may serve as an efficient anti-tumor agent for the treatment of hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/therapy , Carrier Proteins/genetics , Genetic Therapy , Liver Neoplasms/therapy , Telomerase/genetics , Animals , Apoptosis/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carrier Proteins/therapeutic use , Cell Line, Tumor , Cell Proliferation/genetics , Genetic Vectors , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Metalloproteins , Mice , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/therapeutic use , Telomerase/therapeutic use , Xenograft Model Antitumor Assays
19.
Int J Oncol ; 50(5): 1612-1622, 2017 May.
Article in English | MEDLINE | ID: mdl-28393254

ABSTRACT

The Escherichia coli purine nucleoside phosphorylase/Fludarabine phosphate (ePNP/Fludara) suicide system has several drawbacks, such as side-effects and the low efficiency of ePNP expression. In this study, we evaluated the antitumor effects of the dual-specific 8HSEs-hTERTp-ePNP/Fludara suicide system under hyperthermia in vitro and in vivo. Luciferase activities from the 8HSEs­hTERT and CMV promoters were compared using the dual luciferase assay in SW480 (high hTERT expression) and MKN74 cells (hTERT-negative) in the presence and absence of hyperthermia. Then, we investigated the effects of overexpressing the suicide gene ePNP using 8HSEs­hTERT-driven lentiviral vectors with Fludara on in vitro cell viability, side-effects, apoptosis, cycle distribution, colony formation and in vivo xenograft tumor growth. At 43˚C, luciferase activity from the 8HSEs­hTERT promoter was significantly increased in SW480 cells, but not in MKN74 cells. Importantly, luciferase activities from the 8HSEs­hTERT promoter were much higher than from the CMV promoter in hTERT-expressing SW480 cells under heated conditions. The in vitro quantitative analysis showed a 4-fold higher ePNP protein expression from the 8HSEs­hTERT promoter at 43˚C than at 37˚C in SW480 cells and the ePNP mRNA expression in SW480 cells at 43˚C was also higher than at 37˚C. Conversely, ePNP mRNA and protein expression were low, almost absent, in hTERT-negative MKN74 cells with or without hyperthermia. After Fludara addition, cell cytotoxicity assays showed that the significant inhibitory effect of the 8HSEs­hTERTp-ePNP on SW480 cells was dose- and time-dependent with hyperthermia. The 8HSEs­hTERTp-ePNP/Fludara suicide system significantly inhibited SW480 cell viability, colony formation, cell cycle progression and induced apoptosis in vitro, but also induced significant bystander effects, especially under the heated conditions. At the protein level, the suicide system significantly promoted Bax, caspase-3 and p53 expression and suppressed Bcl-2 expression. In sections from mouse xenografts, TUNEL assays showed that the suicide system reduced xenograft growth and induced SW480 apoptosis. These results indicated that the combinatorial cancer- and heat-specific promoter system has great potential for improving the efficacy of cancer treatment with hyperthermia. The 8HSEs­hTERTp-ePNP/Fludara system may serve as a powerful strategy for cancer gene therapy combined with hyperthermia.


Subject(s)
Genetic Therapy , Genetic Vectors/therapeutic use , Neoplasms/genetics , Neoplasms/therapy , Purine-Nucleoside Phosphorylase/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Escherichia coli/genetics , Gene Expression Regulation, Enzymologic , Genes, Transgenic, Suicide/genetics , Genetic Vectors/genetics , Green Fluorescent Proteins , Humans , Lentivirus/genetics , Mice , Neoplasms/pathology , Purine-Nucleoside Phosphorylase/biosynthesis , Purine-Nucleoside Phosphorylase/therapeutic use , Telomerase/genetics , Telomerase/therapeutic use , Xenograft Model Antitumor Assays
20.
Cancer Immunol Immunother ; 66(7): 891-901, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28391357

ABSTRACT

In newly diagnosed metastatic hormone-naive prostate cancer (mPC), telomerase-based immunotherapy with the novel hTERT peptide vaccine UV1 can induce immune responses with potential clinical benefit. This phase I dose escalation study of UV1 evaluated safety, immune response, effects on prostate-specific antigen (PSA) levels, and preliminary clinical outcome. Twenty-two patients with newly diagnosed metastatic hormone-naïve PC (mPC) were enrolled; all had started androgen deprivation therapy and had no visceral metastases. Bone metastases were present in 17 (77%) patients and 16 (73%) patients had affected lymph nodes. Three dose levels of UV1 were given as intradermal injections combined with GM-CSF (Leukine®). Twenty-one patients in the intention-to-treat population (95%) received conformal radiotherapy. Adverse events reported were predominantly grade 1, most frequently injection site pruritus (86.4%). Serious adverse events considered possibly related to UV1 and/or GM-CSF included anaphylactic reaction in two patients and thrombocytopenia in one patient. Immune responses against UV1 peptides were confirmed in 18/21 evaluable patients (85.7%), PSA declined to <0.5 ng/mL in 14 (64%) patients and in ten patients (45%) no evidence of persisting tumour was seen on MRI in the prostatic gland. At the end of the nine-month reporting period for the study, 17 patients had clinically stable disease. Treatment with UV1 and GM-CSF gave few adverse events and induced specific immune responses in a large proportion of patients unselected for HLA type. The intermediate dose of 0.3 mg UV1 resulted in the highest proportion of, and most rapid UV1-specific immune responses with an acceptable safety profile. These results warrant further clinical studies in mPC.


Subject(s)
Adenocarcinoma/therapy , Cancer Vaccines/therapeutic use , Immunotherapy/methods , Peptide Fragments/therapeutic use , Prostate-Specific Antigen/blood , Prostatic Neoplasms/therapy , Telomerase/therapeutic use , Adenocarcinoma/blood , Adenocarcinoma/immunology , Adenocarcinoma/secondary , Aged , Bone Neoplasms/secondary , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Cohort Studies , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Immunity, Active/immunology , Lymphatic Metastasis , Magnetic Resonance Imaging , Male , Middle Aged , Peptide Fragments/adverse effects , Peptide Fragments/immunology , Prostate-Specific Antigen/immunology , Prostatic Neoplasms/blood , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Telomerase/adverse effects , Telomerase/immunology , Vaccines, Subunit/adverse effects , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...