1
|
Kageler L, Aquilanti E. Discovery of telomerase inhibitors: existing strategies and emerging innovations. Biochem Soc Trans 2024; 52:1957-1968. [PMID: 39194999 DOI: 10.1042/bst20230264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024]
Abstract
Telomerase, crucial for maintaining telomere length, is an attractive target for cancer therapy due to its role in cellular immortality. Despite three decades of research efforts, no small-molecule telomerase inhibitors have been clinically approved, highlighting the extensive challenges in developing effective telomerase-based therapeutics. This review examines conventional and emerging methods to measure telomerase activity and discusses existing inhibitors, including oligonucleotides and small molecules. Furthermore, this review highlights recent breakthroughs in structural studies of telomerase using cryo-electron microscopy, which can facilitate improved structure-based drug design. Altogether, advancements in structural methodologies and high-throughput screening offer promising prospects for telomerase-based cancer therapeutic development.
Collapse
Affiliation(s)
- Lauren Kageler
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, U.S.A
| | - Elisa Aquilanti
- Division of Neuro Oncology, Dana Farber Cancer Institute, Boston, MA, U.S.A
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, U.S.A
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, U.S.A
| |
Collapse
|
2
|
Sabokrouh A, Hajivand S, Atabi F. Comparison of anti-cancer effects of platinum ribavirin and ribavirin via telomerase and Bcl-2 gene expression. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3907-3915. [PMID: 37975929 DOI: 10.1007/s00210-023-02841-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
Among the common treatments for cancers, chemotherapy is widely used. One of the ways to evaluate the effectiveness of anti-cancer drugs is by checking the expression of tumor markers. Hence, this study aimed to evaluate the anti-cancer effects of the newly synthesized platinum ribavirin (Pt-Rb) compared to ribavirin (Rb) through biomarkers. In this study, cell lines were divided into four groups: groups A and B as healthy negative control group and untreated cancer group respectively. Group C and D were treated with, Rb and Pt-Rb, a novel anti-cancer drug, respectively. After evaluating LC50 for the drugs by MTT test, the expression of telomerase and Bcl-2 (B cell lymphoma-2) genes was evaluated using real-time PCR (RT-qPCR). The results showed a significant decrease in telomerase (0.020 ± 0.007) and Bcl-2(0.120 ± 0.005) gene expression in cancer cells treated with Pt-Rb (group D) compared to telomerase (0.040 ± 0.014) and Bcl-2(0.220 ± 0.014) treated with Rb (group C) and also between group D and telomerase (70.76 ± 0.330) and Bcl-2 (99.52 ± 0.670) in group B. The majority of the groups under investigation showed a significant difference (p < 0.05), suggesting that Pt-Rb had stronger anti-cancer effects than Rb and untreated cancer cells. Additionally, Pt-Rb treatment results demonstrated more increased apoptosis than Rb. Our results demonstrated that Pt-Rb is an effective medication in cancer treatment by lowering anti-apoptotic indicators. Therefore, this chemical has the potential to be an effective anti-cancer therapy, pending further research on animal models and then human volunteers.
Collapse
Affiliation(s)
- Abdolreza Sabokrouh
- Department of Biochemistry, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Soheyla Hajivand
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fereshteh Atabi
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
3
|
Palamarchuk AI, Kovalenko EI, Streltsova MA. Multiple Actions of Telomerase Reverse Transcriptase in Cell Death Regulation. Biomedicines 2023; 11:biomedicines11041091. [PMID: 37189709 DOI: 10.3390/biomedicines11041091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/25/2023] [Accepted: 04/02/2023] [Indexed: 04/07/2023] Open
Abstract
Telomerase reverse transcriptase (TERT), a core part of telomerase, has been known for a long time only for its telomere lengthening function by reverse transcription of RNA template. Currently, TERT is considered as an intriguing link between multiple signaling pathways. The diverse intracellular localization of TERT corresponds to a wide range of functional activities. In addition to the canonical function of protecting chromosome ends, TERT by itself or as a part of the telomerase complex participates in cell stress responses, gene regulation and mitochondria functioning. Upregulation of TERT expression and increased telomerase activity in cancer and somatic cells relate to improved survival and persistence of such cells. In this review, we summarize the data for a comprehensive understanding of the role of TERT in cell death regulation, with a focus on the interaction of TERT with signaling pathways involved in cell survival and stress response.
Collapse
Affiliation(s)
- Anastasia I. Palamarchuk
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Elena I. Kovalenko
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Maria A. Streltsova
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| |
Collapse
|
4
|
Tsai WH, Zeng YH, Lee CC, Tsai MC. Mortality factors in recurrent parathyroid cancer: a pooled analysis. J Bone Miner Metab 2022; 40:508-517. [PMID: 35184206 DOI: 10.1007/s00774-021-01305-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/19/2021] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Parathyroid cancer is a rare disease with high recurrence rate. The prognostic factors for recurrent parathyroid cancer are yet to be ascertained. We aimed to establish the association between recurrent parathyroid cancer and previously reported prognostic factors. MATERIALS AND METHODS We conducted a PubMed search using the keywords 'parathyroid cancer', 'parathyroid neoplasm', and 'hypercalcemia' during 1966-2019 and included 3272 articles. We focused on 73 patients with recurrent parathyroid cancer from 55 studies. We conducted a survival analysis using the Cox proportional hazards model with 95% confidence interval. RESULTS For the 73 patients included in the analysis, the mean age (± standard deviation) was 44 ± 13.2 years, wherein 36 patients were women (49.3%). During the 5236 person-months at risk (mean follow-up 71.7 months, range 3-264), 38 patients died. The incidence of local recurrence, lymph-node metastasis, lung metastasis, and bone metastasis were 60.3, 12.3, 56.2, and 24.7, respectively. Bone metastasis, disease-free interval < 1 year, and total surgeries < 3 were significant prognostic factors in univariate analysis (log-rank test P = 0.0063, P = 0.0006, and P = 0.0056, respectively). In the multivariate-adjusted analysis, the mortality risk was significantly increased in patients with bone metastasis with a hazard ratio (HR) of 4.83 (95% CI 1.16-20.2; P = 0.03), disease-free interval <=1 year of 5.92 (95% CI 1.85-18.99; P = 0.003), and total surgeries <3 of 11.29 (95% CI 2.82-45.22; P = 0.001), considering these as possible predictive prognostic factors. CONCLUSION Bone metastasis, duration of disease-free interval, and total number of surgeries predict survival in recurrent parathyroid cancer.
Collapse
Affiliation(s)
- Wen-Hsuan Tsai
- Division of Endocrinology and Metabolism, Department of Endocrinology, Department of Internal Medicine, Mackay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Zhongshan Dist, Taipei City, 104, Taiwan (ROC)
| | - Yi-Hong Zeng
- Division of Endocrinology and Metabolism, Department of Endocrinology, Department of Internal Medicine, Mackay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Zhongshan Dist, Taipei City, 104, Taiwan (ROC)
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (ROC)
| | - Chun-Chuan Lee
- Division of Endocrinology and Metabolism, Department of Endocrinology, Department of Internal Medicine, Mackay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Zhongshan Dist, Taipei City, 104, Taiwan (ROC)
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (ROC)
| | - Ming-Chieh Tsai
- Division of Endocrinology and Metabolism, Department of Endocrinology, Department of Internal Medicine, Mackay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Zhongshan Dist, Taipei City, 104, Taiwan (ROC).
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (ROC).
| |
Collapse
|
5
|
Repurposing of Antimicrobial Agents for Cancer Therapy: What Do We Know? Cancers (Basel) 2021; 13:cancers13133193. [PMID: 34206772 PMCID: PMC8269327 DOI: 10.3390/cancers13133193] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
The substantial costs of clinical trials, the lengthy timelines of new drug discovery and development, along the high attrition rates underscore the need for alternative strategies for finding quickly suitable therapeutics agents. Given that most approved drugs possess more than one target tightly linked to other diseases, it encourages promptly testing these drugs in patients. Over the past decades, this has led to considerable attention for drug repurposing, which relies on identifying new uses for approved or investigational drugs outside the scope of the original medical indication. The known safety of approved drugs minimizes the possibility of failure for adverse toxicology, making them attractive de-risked compounds for new applications with potentially lower overall development costs and shorter development timelines. This latter case is an exciting opportunity, specifically in oncology, due to increased resistance towards the current therapies. Indeed, a large body of evidence shows that a wealth of non-cancer drugs has beneficial effects against cancer. Interestingly, 335 drugs are currently being evaluated in different clinical trials for their potential activities against various cancers (Redo database). This review aims to provide an extensive discussion about the anti-cancer activities exerted by antimicrobial agents and presents information about their mechanism(s) of action and stage of development/evaluation.
Collapse
|
6
|
Anwar A, Khan NA, Siddiqui R. Repurposing of Drugs Is a Viable Approach to Develop Therapeutic Strategies against Central Nervous System Related Pathogenic Amoebae. ACS Chem Neurosci 2020; 11:2378-2384. [PMID: 32073257 DOI: 10.1021/acschemneuro.9b00613] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Brain-eating amoebae including Acanthamoeba spp., Naegleria fowleri, and Balamuthia mandrillaris cause rare infections of the central nervous system that almost always result in death. The high mortality rate, lack of interest for drug development from pharmaceutical industries, and no available effective drugs present an alarming challenge. The current drugs employed in the management and therapy of these devastating diseases are amphotericin B, miltefosine, chlorhexidine, pentamidine, and voriconazole which are generally used in combination. However, clinical evidence shows that these drugs have limited efficacy and high host cell cytotoxicity. Repurposing of drugs is a practical approach to utilize commercially available, U.S. Food and Drug Administration approved drugs for one disease against rare diseases caused by brain-eating amoebae. In this Perspective, we highlight some of the success stories of drugs repositioned against neglected parasitic diseases and identify future potential for effective and sustainable drug development against brain-eating amoebae infections.
Collapse
Affiliation(s)
- Ayaz Anwar
- Department of Biological Sciences, School of Science and Technology, Sunway University, Subang Jaya 47500, Selangor, Malaysia
| | - Naveed Ahmed Khan
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Ruqaiyyah Siddiqui
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah 26666, United Arab Emirates
| |
Collapse
|
7
|
Berrino E, Angeli A, Zhdanov DD, Kiryukhina AP, Milaneschi A, De Luca A, Bozdag M, Carradori S, Selleri S, Bartolucci G, Peat TS, Ferraroni M, Supuran CT, Carta F. Azidothymidine "Clicked" into 1,2,3-Triazoles: First Report on Carbonic Anhydrase-Telomerase Dual-Hybrid Inhibitors. J Med Chem 2020; 63:7392-7409. [PMID: 32463228 PMCID: PMC8154556 DOI: 10.1021/acs.jmedchem.0c00636] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Cancer cells rely on the enzyme telomerase
(EC 2.7.7.49) to promote
cellular immortality. Telomerase inhibitors (i.e., azidothymidine)
can represent promising antitumor agents, although showing high toxicity
when administered alone. Better outcomes were observed within a multipharmacological
approach instead. In this context, we exploited the validated antitumor
targets carbonic anhydrases (CAs; EC 4.2.1.1) IX and XII to attain
the first proof of concept on CA–telomerase dual-hybrid inhibitors.
Compounds 1b, 7b, 8b, and 11b showed good in vitro
inhibition potency against the CAs IX and XII, with KI values in the low nanomolar range, and strong antitelomerase
activity in PC-3 and HT-29 cells (IC50 values ranging from
5.2 to 9.1 μM). High-resolution X-ray crystallography on selected
derivatives in the adduct with hCA II as a model study allowed to
determine their binding modes and thus to set the structural determinants
necessary for further development of compounds selectively targeting
the tumoral cells.
Collapse
Affiliation(s)
- Emanuela Berrino
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Andrea Angeli
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Dmitry D Zhdanov
- Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia.,Peoples Friendship University of Russia (RUDN University), Miklukho-Maklaya st. 6, 117198 Moscow, Russia
| | - Anna P Kiryukhina
- Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia
| | - Andrea Milaneschi
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Alessandro De Luca
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Murat Bozdag
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Simone Carradori
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Silvia Selleri
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Gianluca Bartolucci
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Thomas S Peat
- CSIRO, 343 Royal Parade, Parkville, Victoria 3052, Australia
| | - Marta Ferraroni
- Dipartimento di Chimica "Ugo Schiff", Università di Firenze, Via della Lastruccia 3-13, 50019 Sesto Fiorentino (Florence), Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| | - Fabrizio Carta
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Sesto Fiorentino (Florence), Italy
| |
Collapse
|
8
|
Armando RG, Gómez DLM, Gomez DE. New drugs are not enough‑drug repositioning in oncology: An update. Int J Oncol 2020; 56:651-684. [PMID: 32124955 PMCID: PMC7010222 DOI: 10.3892/ijo.2020.4966] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/16/2019] [Indexed: 11/24/2022] Open
Abstract
Drug repositioning refers to the concept of discovering novel clinical benefits of drugs that are already known for use treating other diseases. The advantages of this are that several important drug characteristics are already established (including efficacy, pharmacokinetics, pharmacodynamics and toxicity), making the process of research for a putative drug quicker and less costly. Drug repositioning in oncology has received extensive focus. The present review summarizes the most prominent examples of drug repositioning for the treatment of cancer, taking into consideration their primary use, proposed anticancer mechanisms and current development status.
Collapse
Affiliation(s)
- Romina Gabriela Armando
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Diego Luis Mengual Gómez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Daniel Eduardo Gomez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| |
Collapse
|
9
|
Joshy KS, Augustine R, Mayeen A, Alex SM, Hasan A, Thomas S, Chi H. NiFe2O4/poly(ethylene glycol)/lipid–polymer hybrid nanoparticles for anti-cancer drug delivery. NEW J CHEM 2020. [DOI: 10.1039/d0nj01163k] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The present study reports the fabrication of hybrid nanoparticles consisting of nickel ferrite (NFO) for anti cancer drug delivery.
Collapse
Affiliation(s)
- K. S. Joshy
- Shandong Provincial Key Laboratory of Molecular Engineering
- School of Chemistry and Chemical Engineering
- Qilu University of Technology (Shandong Academy of Sciences)
- Jinan
- China
| | - Robin Augustine
- Department of Mechanical and Industrial Engineering
- College of Engineering
- Qatar University
- Doha
- Qatar
| | - Anshida Mayeen
- Department of Physics
- Thangal Kunju Musliar College of Engineering
- Kollam – 691005
- India
| | | | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering
- College of Engineering
- Qatar University
- Doha
- Qatar
| | - Sabu Thomas
- Mahatma Gandhi University
- Kottayam – 686 560
- India
| | - Hong Chi
- Shandong Provincial Key Laboratory of Molecular Engineering
- School of Chemistry and Chemical Engineering
- Qilu University of Technology (Shandong Academy of Sciences)
- Jinan
- China
| |
Collapse
|
10
|
Qiu S, Zhao F, Zenasni O, Li J, Shih WC. Catalytic assembly of DNA nanostructures on a nanoporous gold array as 3D architectures for label-free telomerase activity sensing. NANOSCALE HORIZONS 2017; 2:217-224. [PMID: 32260643 DOI: 10.1039/c7nh00042a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Telomerase, an enzyme known to catalyze telomere elongation by adding TTAGGG [thymine (T), adenine (A), and guanine (G)] repeats to the end of telomeres, is vital for cell proliferation. Overexpression of telomerase has been found in most tumor cells, resulting in telomere dysfunction and uncontrolled cellular proliferation. Thus, telomerase has been considered as a potential cancer biomarker, as well as a potential target in cancer therapy. In this study, telomerase-catalyzed growth of tandem G-quadruplex (G4) assembled on a nanoporous gold array (NPGA) resulted in the formation of three-dimensional hybrid nanoarchitectures. The generated nanostructure then captured malachite green (MG) (reporter molecule) without the need of a complicated labeling process. Upon laser irradiation, the captured MG molecules produced a surface-enhanced Raman scattering (SERS) signal that was generated by an abundant amount of plasmonic hot spots in the NPGA substrates. A limit of detection (LOD) of 10-10 IU along with a linear range, which was 3 orders of magnitude, was achieved, which was equivalent to the telomerase amount extracted from 20 HeLa cells. The LOD is 2 orders of magnitude better than that of the commercial enzyme-linked immunosorbent assay (ELISA), and it approaches that of the most sensitive technique, telomeric repeat amplification protocols (TRAP), which require a laborious and equipment-intensive polymerase chain reaction (PCR). In addition, X-ray photoelectron spectroscopy (XPS) was used to chemically identify and quantify the telomerase activity on the sensitized NPGA surface. Furthermore, the sensor was applied to screen the effectiveness of anti-telomerase drugs such as zidovudine, thus demonstrating the potential use of the sensor in telomerase-based diagnosis and drug development. Moreover, the framework represents a novel paradigm of collaborative plasmonic intensification and catalytic multiplication (c-PI/CM) for label-free biosensing.
Collapse
Affiliation(s)
- Suyan Qiu
- Department of Electrical and Computer Engineering, University of Houston, 4800 Calhoun Road, Houston, TX 77204, USA.
| | | | | | | | | |
Collapse
|
11
|
Wang H, Zhou J, He Q, Dong Y, Liu Y. Azidothymidine inhibits cell growth and telomerase activity and induces DNA damage in human esophageal cancer. Mol Med Rep 2017; 15:4055-4060. [PMID: 28487971 PMCID: PMC5436214 DOI: 10.3892/mmr.2017.6549] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 02/27/2017] [Indexed: 12/26/2022] Open
Abstract
Esophageal cancer is one of the most common type of malignancies. Telomerase activity, which is absent or weakly detected in the majority of human somatic cells, is elevated in esophageal cancer. Although azidothymidine (AZT), a reverse transcriptase inhibitor, has been utilized as a treatment for tumors, its role in treating esophageal cancer has not been confirmed. The aim of the present study was to determine the effect of AZT on telomerase activity and the proliferation of the human esophageal cancer cell line TE-11. A telomeric repeat amplification assay was utilized to detect telomerase activity following treatment of TE-11 cells with AZT. The effect of AZT on TE-11 cell cycle distribution was determined by flow cytometry. Cellular DNA damage was evaluated by a comet assay and an MTT assay demonstrated that AZT significantly inhibited the viability of TE-11 cells, in a time-and dose-dependent manner. In addition, TE-11 cells treated with various concentrations of AZT exhibited a significant reduction in telomerase activity and percentage of cells in the G1/G0 phase, and an increase in the percentage of cells in the S phase. High doses of AZT caused DNA damage, and enhanced the expression levels of γ-H2A histone family member X and phosphorylated checkpoint kinase 2 in TE-11 cells. These results demonstrated that AZT effectively inhibits proliferation of the TE-11 human esophageal cancer cell line in vitro. The growth inhibitory effects were associated with a reduction in telomerase activity, S and G2/M phase cell cycle arrest, and enhanced DNA damage, suggesting that AZT may be utilized in the clinic for the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Haoli Wang
- Department of Pathology, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong 510080, P.R. China
| | - Jianwen Zhou
- Department of Pathology, First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Qiong He
- Department of Pathology, First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yu Dong
- Department of Pathology, First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yanhui Liu
- Department of Pathology, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
12
|
Shahabadi N, Falsafi M, Feizi F, Khodarahmi R. Functionalization of γ-Fe2O3@SiO2 nanoparticles using the antiviral drug zidovudine: synthesis, characterization, in vitro cytotoxicity and DNA interaction studies. RSC Adv 2016. [DOI: 10.1039/c6ra16564h] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The aim of this study was to design and prepare γ-Fe2O3@SiO2-zidovudine magnetic nanoparticles (MNPs) for magnetic guided drug targeting and biological applications.
Collapse
Affiliation(s)
- Nahid Shahabadi
- Department of Inorganic Chemistry
- Faculty of Chemistry
- Razi University
- Kermanshah
- Iran
| | - Monireh Falsafi
- Department of Inorganic Chemistry
- Faculty of Chemistry
- Razi University
- Kermanshah
- Iran
| | - Foroozan Feizi
- Department of Analytical Chemistry
- Faculty of Chemistry
- Razi University
- Kermanshah
- Iran
| | - Reza Khodarahmi
- Medical Biology Research Center (MBRC)
- Kermanshah University of Medical Sciences
- Kermanshah
- Iran
| |
Collapse
|
13
|
Haglund F, Juhlin CC, Brown T, Ghaderi M, Liu T, Stenman A, Dinets A, Prasad M, Korah R, Xu D, Carling T, Larsson C. TERT promoter mutations are rare in parathyroid tumors. Endocr Relat Cancer 2015; 22:L9-L11. [PMID: 25876648 DOI: 10.1530/erc-15-0121] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/14/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Felix Haglund
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| | - Carl Christofer Juhlin
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| | - Taylor Brown
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| | - Mehran Ghaderi
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| | - Tiantian Liu
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| | - Adam Stenman
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| | - Andrii Dinets
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| | - Manju Prasad
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| | - Reju Korah
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| | - Dawei Xu
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| | - Tobias Carling
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| | - Catharina Larsson
- Department of Oncology-PathologyKarolinska Institutet, Cancer Center Karolinska (CCK), Karolinska University HospitalSE-171 76, StockholmSweden
| |
Collapse
|
14
|
de Souza D, Mariano DOC, Nedel F, Schultze E, Campos VF, Seixas F, da Silva RS, Munchen TS, Ilha V, Dornelles L, Braga AL, Rocha JBT, Collares T, Rodrigues OED. New Organochalcogen Multitarget Drug: Synthesis and Antioxidant and Antitumoral Activities of Chalcogenozidovudine Derivatives. J Med Chem 2015; 58:3329-39. [DOI: 10.1021/jm5015296] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Diego de Souza
- LabSelen-NanoBio
- Departamento de Química, Universidade Federal de Santa Maria, 97105-900 Santa Maria, Brazil
| | - Douglas O. C. Mariano
- LabSelen-NanoBio
- Departamento de Química, Universidade Federal de Santa Maria, 97105-900 Santa Maria, Brazil
| | - Fernanda Nedel
- Molecular
and Cellular Oncology Group, Graduate Program in Biotechnology, Universidade Federal de Pelotas, 96010-610 Pelotas, Brazil
| | - Eduarda Schultze
- Molecular
and Cellular Oncology Group, Graduate Program in Biotechnology, Universidade Federal de Pelotas, 96010-610 Pelotas, Brazil
| | - Vinícius F. Campos
- Molecular
and Cellular Oncology Group, Graduate Program in Biotechnology, Universidade Federal de Pelotas, 96010-610 Pelotas, Brazil
| | - Fabiana Seixas
- Molecular
and Cellular Oncology Group, Graduate Program in Biotechnology, Universidade Federal de Pelotas, 96010-610 Pelotas, Brazil
| | - Rafael S. da Silva
- LabSelen-NanoBio
- Departamento de Química, Universidade Federal de Santa Maria, 97105-900 Santa Maria, Brazil
| | - Taiana S. Munchen
- LabSelen-NanoBio
- Departamento de Química, Universidade Federal de Santa Maria, 97105-900 Santa Maria, Brazil
| | - Vinicius Ilha
- LabSelen-NanoBio
- Departamento de Química, Universidade Federal de Santa Maria, 97105-900 Santa Maria, Brazil
| | - Luciano Dornelles
- LabSelen-NanoBio
- Departamento de Química, Universidade Federal de Santa Maria, 97105-900 Santa Maria, Brazil
| | - Antonio L. Braga
- Departamento
de Química, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, Brazil
| | - João B. T. Rocha
- LabSelen-NanoBio
- Departamento de Química, Universidade Federal de Santa Maria, 97105-900 Santa Maria, Brazil
| | - Tiago Collares
- Molecular
and Cellular Oncology Group, Graduate Program in Biotechnology, Universidade Federal de Pelotas, 96010-610 Pelotas, Brazil
| | - Oscar E. D. Rodrigues
- LabSelen-NanoBio
- Departamento de Química, Universidade Federal de Santa Maria, 97105-900 Santa Maria, Brazil
| |
Collapse
|
15
|
Wang J, Wang YJ, Chen ZS, Kwon CH. Synthesis and evaluation of sulfonylethyl-containing phosphotriesters of 3′-azido-3′-deoxythymidine as anticancer prodrugs. Bioorg Med Chem 2014; 22:5747-56. [DOI: 10.1016/j.bmc.2014.09.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/13/2014] [Accepted: 09/22/2014] [Indexed: 10/24/2022]
|
16
|
Fang JL, Han T, Wu Q, Beland FA, Chang CW, Guo L, Fuscoe JC. Differential gene expression in human hepatocyte cell lines exposed to the antiretroviral agent zidovudine. Arch Toxicol 2014; 88:609-23. [PMID: 24292225 PMCID: PMC5901687 DOI: 10.1007/s00204-013-1169-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 11/13/2013] [Indexed: 01/27/2023]
Abstract
Zidovudine (3'-azido-3'-deoxythymidine; AZT) is the most widely used nucleoside reverse transcriptase inhibitor for the treatment of AIDS patients and prevention of mother-to-child transmission of HIV-1. Previously, we demonstrated that AZT had significantly greater growth inhibitory effects upon the human liver carcinoma cell line HepG2 as compared to the immortalized human liver cell line THLE2. We have now used gene expression profiling to determine the molecular pathways associated with toxicity in both cell lines. HepG2 cells were incubated with 0, 2, 20, or 100 μM AZT for 2 weeks; THLE2 cells were treated with 0, 50, 500, or 2,500 μM AZT, concentrations that were equi-toxic to those used in the HepG2 cells. After the treatment, total RNA was isolated and subjected to microarray analysis. Global analysis of gene expression, with a false discovery rate ≤0.01 and a fold change ≥1.5, indicated that 6- to 70-fold more genes were differentially expressed in a significant concentration-dependent manner in HepG2 cells when compared to THLE2 cells. Comparative analysis indicated that 7 % of these genes were common to both cell lines. Among the common differentially expressed genes, 70 % changed in the same direction, most of which were associated with cell death and survival, cell cycle, cell growth and proliferation, and DNA replication, recombination, and repair. As determined by the uptake of [methyl-(3)H]AZT, the intracellular levels of total AZT were approximately twofold higher in THLE2 cells than in HepG2 cells. The expression of thymidine kinase 1 (TK1) and UDP-glucuronosyltransferase 2B7 (UGT2B7) genes that regulate the metabolic activation and deactivation of AZT, respectively, was increased in HepG2 cells but decreased in THLE2 cells after treatment with AZT. This differential response in AZT metabolism was confirmed by real-time PCR, western blotting, and/or enzymatic assays. These data indicate that molecular pathways involved with cell death and survival, cell cycle, cell growth and proliferation, and DNA replication, recombination, and repair are involved in the toxicities associated with AZT in both human cell lines, and that the difference in expression of TK1 and UGT2B7 in response to AZT treatment in HepG2 cells and THLE2 cells might explain why HepG2 cells are more sensitive than THLE2 cells to the toxicity of AZT.
Collapse
Affiliation(s)
- Jia-Long Fang
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, AR, 72079, USA,
| | | | | | | | | | | | | |
Collapse
|
17
|
Shen T, Ma J, Zhang L, Yu X, Liu M, Hou Y, Wang Y, Ma C, Li S, Zhu D. Positive feedback-loop of telomerase reverse transcriptase and 15-lipoxygenase-2 promotes pulmonary hypertension. PLoS One 2013; 8:e83132. [PMID: 24376652 PMCID: PMC3871619 DOI: 10.1371/journal.pone.0083132] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 10/31/2013] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Pulmonary hypertension (PH) is characterized with pulmonary vasoconstriction and vascular remodeling mediated by 15-lipoxygenase (15-LO)/15-hydroxyeicosatetraenoic acid (15-HETE) according to our previous studies. Meanwhile, telomerase reverse transcriptase (TERT) activity is highly correlated with vascular injury and remodeling, suggesting that TERT may be an essential determinant in the development of PH. The aim of this study was to determine the contribution and molecular mechanisms of TERT in the pathogenesis of PH. APPROACH AND RESULTS We measured the right ventricular systolic pressure (RVSP) and ventricular weight, analyzed morphometric change of the pulmonary vessels in the hypoxia or monocrotaline treated rats. Bromodeoxyuridine incorporation, transwell assay and flow cytometry in pulmonary smooth muscle cells were performed to investigate the roles and relationship of TERT and 15-LO/15-HETE in PH. We revealed that the expression of TERT was increased in pulmonary vasculature of patients with PH and in the monocrotaline or hypoxia rat model of PH. The up-regulation of TERT was associated with experimental elevated RVSP and pulmonary vascular remodeling. Coimmunoprecipitation experiments identified TERT as a novel interacting partner of 15-LO-2. TERT and 15-LO-2 augmented protein expression of each other. In addition, the proliferation, migration and cell-cycle transition from G0/G1 phase to S phase induced by hypoxia were inhibited by TERT knockdown, which were rescued by 15-HETE addition. CONCLUSIONS These results demonstrate that TERT regulates pulmonary vascular remodeling. TERT and 15-LO-2 form a positive feedback loop and together promote proliferation and migration of pulmonary artery smooth muscle cells, creating a self-amplifying circuit which propels pulmonary hypertension.
Collapse
Affiliation(s)
- Tingting Shen
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, Heilongjiang Province, China
| | - Jun Ma
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, Heilongjiang Province, China
| | - Lei Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, Heilongjiang Province, China
| | - Xiufeng Yu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, Heilongjiang Province, China
| | - Mengmeng Liu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, Heilongjiang Province, China
| | - Yunlong Hou
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yanyan Wang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, Heilongjiang Province, China
| | - Cui Ma
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, Heilongjiang Province, China
| | - Shuzhen Li
- Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, Heilongjiang Province, China
- Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
18
|
Chen P, Liu Y, Sun Y, Chen C, Qi Y, Zhang Y. AZT and emodin exhibit synergistic growth-inhibitory effects on K562/ADM cells by inducing S phase cell cycle arrest and suppressing MDR1 mRNA/p-gp protein expression. PHARMACEUTICAL BIOLOGY 2013; 51:1586-1591. [PMID: 24004004 DOI: 10.3109/13880209.2013.803257] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
CONTEXT Previous studies have demonstrated that both 3'-azido-3'-deoxythymidine (AZT) and emodin, a traditional chemotherapy agent, can inhibit the growth of many types of cancer cells. OBJECTIVE This study aimed to evaluate the effect of AZT and emodin on adriamycin-resistant human chronic myelogenous leukemia (K562/ADM) cells, determine the expression of multidrug resistance 1 (MDR1) mRNA/p-glycoprotein (p-gp) protein, a protein known to induce resistance to anticancer agents, and to elucidate the underlying molecular mechanisms. MATERIALS AND METHODS K562/ADM cells were treated with AZT (10-160 μM) or emodin (5-80 μM) for 24, 48 and 72 h and cell viability was measured using the MTT assay. The effect of AZT (16.5, 33 and 66 μM) and emodin (6.1, 17.6 and 33.2 μM) on K562/ADM cell cycle distribution was determined by flow cytometry, and MDR1 mRNA/p-gp protein expression was determined by real time RT-PCR and western blotting. RESULTS The growth suppression of emodin was dramatically enhanced by AZT in K562/ADM cells. The IC50 of AZT and emodin was lower than that of emodin alone. All examined combinations of AZT and emodin yielded a synergetic effect (CI < 1). Furthermore, AZT and emodin altered the cell cycle distribution and led to an accumulation of cells in S phase. Meanwhile, the expression of MDR1 mRNA/p-gp protein was markedly decreased. DISCUSSION AND CONCLUSION These results show a synergistic growth-inhibitory effect of AZT and emodin in K562/ADM cells, which is achieved through S phase arrest. MDR1 might ultimately be responsible for these phenomena.
Collapse
Affiliation(s)
- Peng Chen
- School of Life Science, Lanzhou University , Lanzhou , China
| | | | | | | | | | | |
Collapse
|
19
|
Bowyer SE, White AM, Ransom DT, Davidson JA. Resistant hypercalcaemia in metastatic parathyroid carcinoma. Med J Aust 2013; 198:559-61. [DOI: 10.5694/mja12.11243] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 02/11/2013] [Indexed: 11/17/2022]
Affiliation(s)
| | - Alison M White
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Perth, WA
| | - David T Ransom
- Department of Medical Oncology, Royal Perth Hospital, Perth, WA
| | - John A Davidson
- Department of Medical Oncology, Royal Perth Hospital, Perth, WA
| |
Collapse
|
20
|
Bollmann FM. Telomerase inhibition may contribute to accelerated mitochondrial aging induced by anti-retroviral HIV treatment. Med Hypotheses 2013; 81:285-7. [PMID: 23679995 DOI: 10.1016/j.mehy.2013.04.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 04/08/2013] [Accepted: 04/16/2013] [Indexed: 01/27/2023]
Abstract
HIV-infected individuals undergoing long-term anti-retroviral treatment tend to show premature senescence. Accelerated mitochondrial aging induced by nucleoside reverse transcriptase inhibitors (NRTIs) has been implicated as a part of this phenomenon. Traditionally, this has been attributed to inhibition of mtDNA polymerase γ by these drugs, but alternative explanations have been proposed. It is known that NRTIs can not only inhibit viral reverse transcriptase, but also human telomerase. A number of extratelomeric roles of telomerase, including protection of mitochondrial DNA and function, have emerged recently. In this paper, I propose that inhibition of mitochondrial telomerase activity by NRTI drugs contributes to the mitochondrial toxicity and premature aging seen in treated HIV patients, and discuss objections and experimental testing of the hypothesis.
Collapse
Affiliation(s)
- F M Bollmann
- University Medical Center Tübingen, Wilhelmstr 27, 72016 Tübingen, Germany.
| |
Collapse
|
21
|
Azidothymidine hinders arsenic trioxide-induced apoptosis in acute promyelocytic leukemia cells by induction of p21 and attenuation of G2/M arrest. Ann Hematol 2013; 92:1207-20. [DOI: 10.1007/s00277-013-1763-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Accepted: 04/12/2013] [Indexed: 12/12/2022]
|
22
|
Souza Sobrinho CPD, Gragnani A, Santos IDAO, Oliveira AF, Lipay MVN, Ferreira LM. AZT on telomerase activity and cell proliferation in HS 839.T melanoma cells. Acta Cir Bras 2012. [PMID: 23207751 DOI: 10.1590/s0102-86502012001200005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
PURPOSE To evaluate telomerase activity and proliferation of HS839.T melanoma cells, subjected to the action of AZT. METHODS Cells were grown in triplicate, AZT at different concentrations: 50, 100 and 200 μM, was added and left for 24 and 48 hours, and its effects were compared with the control group. Telomerase activity was detected by PCR and cell proliferation was evaluated by MTT. RESULTS After 24 hours, there was no inhibition of cell proliferation or telomerase activity when compared to the control group. After 48 hours, there was a momentary decrease, suggesting that the cell lines used in this study are sensitive to AZT, but quickly recover both the enzyme activity and cell proliferation. CONCLUSION The action of AZT on the melanoma cells studied, at the concentrations and times tested, did not inhibit telomerase activity nor affect cell proliferation.
Collapse
|
23
|
Gomez DE, Armando RG, Alonso DF. AZT as a telomerase inhibitor. Front Oncol 2012; 2:113. [PMID: 22973556 PMCID: PMC3434370 DOI: 10.3389/fonc.2012.00113] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 08/17/2012] [Indexed: 01/23/2023] Open
Abstract
Telomerase is a highly specialized reverse transcriptase (RT) and the maintenance of telomeric length is determined by this specific enzyme. The human holoenzyme telomerase is a ribonucleoprotein composed by a catalytic subunit, hTERT, an RNA component, hTR, and a group of associated proteins. Telomerase is normally expressed in embryonic cells and is repressed during adulthood. The enzyme is reexpressed in around 85% of solid tumors. This observation makes it a potential target for developing drugs that could be developed for therapeutic purposes. The identification of the hTERT as a functional catalytic RT prompted studies of inhibiting telomerase with the HIV RT inhibitor azidothymidine (AZT). Previously, we have demonstrated that AZT binds preferentially to telomeres, inhibits telomerase and enhances tumor cell senescence, and apoptosis after AZT treatment in breast mammary adenocarcinoma cells. Since then, several studies have considered AZT for telomerase inhibition and have led to potential clinical strategies for anticancer therapy. This review covers present thinking of the inhibition of telomerase by AZT and future treatment protocols using the drug.
Collapse
Affiliation(s)
- Daniel E Gomez
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal Buenos Aires, Argentina
| | | | | |
Collapse
|
24
|
Falchetti A, Cavalli L, Cavalli T, Giusti F, Marcucci G, Marini F, Brandi ML. Molecular diagnosis of parathyroid carcinoma: a reality in the near future. ACTA ACUST UNITED AC 2011; 6:27-37. [PMID: 23480618 DOI: 10.1517/17530059.2012.634796] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Parathyroid carcinoma (PC) is a rare endocrine malignancy that still suffers from a problem of delayed clinical diagnosis. Consequently, it usually is not recognized preoperatively and often is not conclusively identified during the operation either. AREAS COVERED The role played by parafibromin in the development of PC, representing an important advance in understanding the pathogenesis of this malignancy, is discussed. Through a careful search of the international literature, using "parathyroid carcinoma", "molecular genetics of parathyroid carcinoma" and "parathyroid tumorigenesis" as key words, other less mentioned molecular mechanisms are reappraised as potential molecular markers of PC; we also discuss their potential role in 20 parathyroid outgrowths. Finally, both the major efforts and the limitations of reported molecular diagnostic techniques and diagnostic markers are considered. EXPERT OPINION Currently, several critical issues still need to be addressed, such as the lack of: i) common criteria for the histopathological diagnosis of parathyroid malignancy and ii) timely appropriated preoperative diagnosis of PC. The latter issue would be of fundamental importance to assist the surgeon in performing a complete resection of all carcinomatous tissue at the time of the initial surgery, allowing for the greatest likelihood of a cure.
Collapse
Affiliation(s)
- Alberto Falchetti
- University of Florence and Regional Centre for Hereditary Endocrine Tumors, University Hospital of Careggi , Department of Internal Medicine , Unit of Metabolic Bone Diseases, Viale Pieraccini, 6, 50139 Florence , Italy
| | | | | | | | | | | | | |
Collapse
|
25
|
Owen RP, Silver CE, Pellitteri PK, Shaha AR, Devaney KO, Werner JA, Rinaldo A, Ferlito A. Parathyroid carcinoma: a review. Head Neck 2011; 33:429-36. [PMID: 20310041 DOI: 10.1002/hed.21376] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Randall P Owen
- Department of Surgery, Division of Metabolic, Endocrine and Minimally Invasive Surgery, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
OBJECTIVE To review the current knowledge pertaining to the etiology, molecular pathogenesis, and management of parathyroid carcinoma, a rare presentation of primary hyperparathyroidism. METHODS The existing MEDLINE English-language literature was reviewed using the search terms "parathyroid" and "carcinoma" or "cancer." RESULTS Parathyroid cancer is a rare endocrine tumor accounting for a small proportion of cases of primary hyperparathyroidism. Recent database studies indicate increasing incidence rates. Its etiology is unknown, although numerous molecular alterations have been described, and the tumors also occur in association with germline mutations in the CDC73 gene. Most affected patients present with severe hypercalcemia; however, the diagnosis can be challenging. Complete surgical resection remains the mainstay of treatment and provides the best chance of cure, although data from small series suggest that external beam radiation may also reduce the high recurrence rates. No effective chemotherapy regimens are currently available. A significant number of patients develop recurrent disease and need additional procedures; however, long-term survival is possible with palliative surgery. Medical management of chronic and debilitating hypercalcemia with calcimimetics is often necessary and is an important adjunct in patients with recurrent and metastatic disease. CONCLUSIONS Further elucidation of the molecular pathogenesis of parathyroid carcinomas will enhance our understanding of etiology and behavior of this uncommon entity. Future research must be directed at identifying more effective therapies for this condition.
Collapse
Affiliation(s)
- Sandy H Fang
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
27
|
Manda KR, Banerjee A, Banks WA, Ercal N. Highly active antiretroviral therapy drug combination induces oxidative stress and mitochondrial dysfunction in immortalized human blood-brain barrier endothelial cells. Free Radic Biol Med 2011; 50:801-10. [PMID: 21193030 PMCID: PMC5997409 DOI: 10.1016/j.freeradbiomed.2010.12.029] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 12/10/2010] [Accepted: 12/17/2010] [Indexed: 10/18/2022]
Abstract
The era of highly active antiretroviral therapy (HAART) has controlled AIDS and its related disorders considerably; however, the prevalence of HIV-1-associated neurocognitive disorders has been on the rise in the post-HAART era. In view of these developments, we investigated whether a HAART drug combination of 3'-azido-2',3'-deoxythymidine (AZT) and indinavir (IDV) can alter the functionality of the blood-brain barrier (BBB) endothelial cells, thereby exacerbating this condition. The viability of hCMEC/D3 cells (in vitro model of BBB) that were exposed to these drugs was significantly reduced after 72h treatment, in a dose-dependent manner. Reactive oxygen species were highly elevated after the exposure, indicating that mechanisms that induce oxidative stress were involved. Measures of oxidative stress parameters, such as glutathione and malondialdehyde, were altered in the treated groups. Loss of mitochondrial membrane potential, as assessed by fluorescence microscopy and decreased levels of ATP, indicated that cytotoxicity was mediated through mitochondrial dysfunction. Furthermore, AZT+IDV treatment caused apoptosis in endothelial cells, as assessed by the expression of cytochrome c and procaspase-3 proteins. Pretreatment with the thiol antioxidant N-acetylcysteine amide reversed some of the pro-oxidant effects of AZT+IDV. Results from our in vitro studies indicate that the AZT+IDV combination may affect the BBB in HIV-infected individuals treated with HAART drugs.
Collapse
Affiliation(s)
- Kalyan Reddy Manda
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO, USA
| | - Atrayee Banerjee
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO, USA
| | - William A. Banks
- GRECC-VA, Puget Sound Health Care System and Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington, Seattle, WA, USA
| | - Nuran Ercal
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO, USA
- Corresponding Author Address: Department of Chemistry, Missouri University of Science and Technology, 400 West 11th Street, Rolla, MO 65409, Phone: 573-341-6950, Fax: 573-341-6033,
| |
Collapse
|
28
|
Abstract
Telomeres are nucleoprotein complexes located at the ends of chromosomes that have a critical role in the maintenance of chromosomal integrity. This involvement is based on complex secondary and tertiary structures that rely on DNA-DNA, DNA-protein and protein-protein interactions. De novo synthesis and maintenance of telomere repeats is controlled by telomerase, a specialized complex that consists of a telomerase RNA component and a protein component--telomerase reverse transcriptase. When telomerase is silent (its default state in differentiated somatic cells), chromosomes shorten with every cell division, thus limiting the lifespan of the cells (the process of senescence) and preventing unlimited cell proliferation, which might eventually lead to the development of cancer. During this process, occasionally, a cell can activate telomerase, which stabilizes short telomeres and enables immortalization-a process essential for malignant transformation. Thus, although telomere erosion is a barrier to malignant progression, paradoxically, in certain circumstances it might also trigger tumorigenesis. A number of studies have demonstrated unequivocally that reactivation of telomerase in the presence of short telomeres is one of the most common features of human cancers, including those of the endocrine system.
Collapse
Affiliation(s)
- Furio Pacini
- Department of Internal Medicine, Endocrinology & Metabolism and Biochemistry, University of Siena, Siena 53100, Italy.
| | | | | | | |
Collapse
|
29
|
Wu Q, Beland FA, Chang CW, Fang JL. XPC is essential for nucleotide excision repair of zidovudine-induced DNA damage in human hepatoma cells. Toxicol Appl Pharmacol 2010; 251:155-62. [PMID: 21192964 DOI: 10.1016/j.taap.2010.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 12/14/2010] [Accepted: 12/16/2010] [Indexed: 12/18/2022]
Abstract
Zidovudine (3'-azido-3'-dexoythymidine, AZT), a nucleoside reverse transcriptase inhibitor, can be incorporated into DNA and cause DNA damage. The mechanisms underlying the repair of AZT-induced DNA damage are unknown. To investigate the pathways involved in the recognition and repair of AZT-induced DNA damage, human hepatoma HepG2 cells were incubated with AZT for 2 weeks and the expression of DNA damage signaling pathways was determined using a pathway-based real-time PCR array. Compared to control cultures, damaged DNA binding and nucleotide excision repair (NER) pathways showed significantly increased gene expression. Further analysis indicated that AZT treatment increased the expression of genes associated with NER, including XPC, XPA, RPA1, GTF2H1, and ERCC1. Western blot analysis demonstrated that the protein levels of XPC and GTF2H1 were also significantly up-regulated. To explore further the function of XPC in the repair of AZT-induced DNA damage, XPC expression was stably knocked down by 71% using short hairpin RNA interference. In the XPC knocked-down cells, 100 μM AZT treatment significantly increased [³H]AZT incorporation into DNA, decreased the total number of viable cells, increased the release of lactate dehydrogenase, induced apoptosis, and caused a more extensive G2/M cell cycle arrest when compared to non-transfected HepG2 cells or HepG2 cells transfected with a scrambled short hairpin RNA sequence. Overall, these data indicate that XPC plays an essential role in the NER repair of AZT-induced DNA damage.
Collapse
Affiliation(s)
- Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR 72079, USA
| | | | | | | |
Collapse
|
30
|
Bagossi P, Bander P, Bozóki B, Tözsér J. Discovery and significance of new human T-lymphotropic viruses: HTLV-3 and HTLV-4. Expert Rev Anti Infect Ther 2010; 7:1235-49. [PMID: 19968515 DOI: 10.1586/eri.09.97] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Human T-lymphotropic virus type 1 (HTLV-1) and type 2 (HTLV-2) were discovered approximately 30 years ago and they are associated with various lymphoproliferative and neurological diseases. The estimated number of infected people is 10-20 million worldwide. In 2005, two new HTLV-1/HTLV-2-related viruses were detected, HTLV-3 and HTLV-4, from the same geographical area of Africa. In the last 4 years, their complete genomic sequences were determined and some of their characteristic features were studied in detail. These newly discovered retroviruses alongside their human (HTLV-1 and -2) and animal relatives (simian T-lymphotropic virus type 1-3) are reviewed. The potential risks associated with these viruses and the potential antiretroviral therapies are also discussed.
Collapse
Affiliation(s)
- Péter Bagossi
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary.
| | | | | | | |
Collapse
|
31
|
Fang JL, Beland FA. Long-term exposure to zidovudine delays cell cycle progression, induces apoptosis, and decreases telomerase activity in human hepatocytes. Toxicol Sci 2009; 111:120-30. [PMID: 19541796 DOI: 10.1093/toxsci/kfp136] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Zidovudine (3'-azido-3'-deoxythymidine; AZT), which is currently used in the treatment of acquired immunodeficiency syndrome, has been shown to have anticancer properties. In the present study, we examined the mechanisms contributing to increased sensitivity of cancer cells to the growth-inhibitory effects of AZT. This was accomplished by incubating a hepatoma cell line (HepG2) and a normal liver cell line (THLE2) with AZT in continuous culture for up to 4 weeks and evaluating the number of viable and necrotic cells, the induction of apoptosis, cell cycle alterations, and telomerase activity. In HepG2 cells, AZT (2-100 microM) caused significant dose-dependent decreases in the number of viable cells at exposures > 24 h. During a 1-week recover period, there was only a slight increase in the number of viable cells treated with AZT. The decrease in viable cells was associated with an induction of apoptosis, a decrease in telomerase activity, and S and G2/M phase arrest of the cell cycle. During the recovery period, the extent of apoptosis and telomerase activity returned to control levels, whereas the disruption of cell cycle progression persisted. Western blot analysis indicated that AZT caused a decrease in checkpoint kinase 1 (Chk1) and kinase 2 (Chk2) and an increase in phosphorylated Chk1 (Ser345) and Chk2 (Thr68). Similar effects, to lesser extent, were observed in THLE2 cells given much higher concentrations of AZT (50-2500 microM). These data show that HepG2 cells are much more sensitive than THLE2 cells to AZT. They also indicate that a combination of a delay of cell cycle progression, an induction of apoptosis, and a decrease in telomerase activity is contributing to the decrease in the number of viable cells from AZT treatment, and that checkpoint enzymes Chk1 and Chk2 may play an important role in the delay of cell cycle progression.
Collapse
Affiliation(s)
- Jia-Long Fang
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079, USA.
| | | |
Collapse
|
32
|
Kucherenko Y, Geiger C, Shumilina E, Föller M, Lang F. Inhibition of cation channels and suicidal death of human erythrocytes by zidovudine. Toxicology 2008; 253:62-9. [PMID: 18822339 DOI: 10.1016/j.tox.2008.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2008] [Revised: 08/20/2008] [Accepted: 08/21/2008] [Indexed: 12/13/2022]
Abstract
Zidovudine, a drug widely used in the treatment of AIDS, has been shown to influence cytosolic calcium activity in HIV-infected lymphocytes. Thus, zidovudine may modify the activity of Ca(2+)-permeable ion channels. In erythrocytes, activation of Ca(2+)-permeable cation channels stimulates eryptosis, the suicidal erythrocyte death. Eryptosis is characterized by cell shrinkage (apparent from a decrease of forward scatter) and phosphatidylserine (PS) exposure (apparent from annexin V-binding) at the erythrocyte surface. Triggers of eryptosis include isotonic cell shrinkage (Cl(-) replacement by gluconate), energy depletion (removal of glucose) or exposure to a variety of drugs including azathioprine. The present study explored, whether zidovudine influences the activity of erythrocytic Ca(2+)-permeable cation channels and eryptosis. Whole-cell patch-clamp recordings indeed revealed that zidovudine blocked the Ca(2+)-permeable cation channels activated by Cl(-) removal. In the presence of Cl(-) and glucose, the percentage of annexin V-binding cells was low and not significantly modified by the presence of zidovudine. Both, Cl(-) removal and glucose depletion increased annexin V-binding and decreased forward scatter, effects significantly blunted by zidovudine (2 microg/ml). According to Fluo3 fluorescence, zidovudine (2 microg/ml) did not significantly modify cytosolic Ca(2+) concentration under control conditions, but significantly blunted the increase in cytosolic Ca(2+) activity following glucose depletion. Furthermore, zidovudine significantly inhibited azathioprine-induced eryptosis. The present observations disclose a completely novel effect of zidovudine, i.e. its inhibitory influence on Ca(2+) entry and subsequent suicidal erythrocyte death during isotonic cell shrinkage or energy depletion.
Collapse
|
33
|
Scruggs ER, Dirks Naylor AJ. Mechanisms of Zidovudine-Induced Mitochondrial Toxicity and Myopathy. Pharmacology 2008; 82:83-8. [PMID: 18504416 DOI: 10.1159/000134943] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Accepted: 02/22/2008] [Indexed: 01/06/2023]
Affiliation(s)
- Erin R Scruggs
- Wingate University School of Pharmacy, Wingate, NC 28174, USA
| | | |
Collapse
|
34
|
Ishitsuka K, Tamura K. Treatment of adult T-cell leukemia/lymphoma: past, present, and future. Eur J Haematol 2007; 80:185-96. [PMID: 18081707 DOI: 10.1111/j.1600-0609.2007.01016.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Adult T-cell leukemia/lymphoma (ATLL) is a peripheral T-cell malignancy caused by human T-cell lymphotrophic virus type I. Clinical manifestations of ATLL range from smoldering to chronic, lymphoma and acute. Patients with acute and lymphoma type ATLL require therapeutic intervention. Conventional chemotherapeutic regimens used against other malignant lymphoma have been administered to ATLL patients, but the therapeutic outcomes of acute and lymphoma type ATLL remain very poor. Promising results of allogeneic stem cell transplantation (SCT) for ATLL patients have recently been reported and the treatment outcome might be improved for some ATLL patients. Besides conventional chemotherapy and SCT, interferon, zidovudine, arsenic trioxide, targeted therapy against surface molecule on ATLL cells, retinoid derivatives, and bortezomib have been administered to ATLL patients in pilot or phase I/II studies. Further studies are required to confirm the clinical benefits of these novel therapeutics. This article reviews the current status and future directions of ATLL treatment.
Collapse
Affiliation(s)
- Kenji Ishitsuka
- Internal Medicine, Division of Hematology and Oncology, Fukuoka University, Fukuoka, Japan.
| | | |
Collapse
|
35
|
De Cian A, Lacroix L, Douarre C, Temime-Smaali N, Trentesaux C, Riou JF, Mergny JL. Targeting telomeres and telomerase. Biochimie 2007; 90:131-55. [PMID: 17822826 DOI: 10.1016/j.biochi.2007.07.011] [Citation(s) in RCA: 477] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Accepted: 07/16/2007] [Indexed: 01/06/2023]
Abstract
Telomeres and telomerase represent, at least in theory, an extremely attractive target for cancer therapy. The objective of this review is to present the latest view on the mechanism(s) of action of telomerase inhibitors, with an emphasis on a specific class of telomere ligands called G-quadruplex ligands, and to discuss their potential use in oncology.
Collapse
Affiliation(s)
- Anne De Cian
- INSERM, U565, Acides nucléiques: dynamique, ciblage et fonctions biologiques, 43 rue Cuvier, CP26, Paris Cedex 05, F-75231, France
| | | | | | | | | | | | | |
Collapse
|
36
|
Wu D, Ji S, Wu Y, Ju Y, Zhao Y. Design, synthesis, and antitumor activity of bile acid–polyamine–nucleoside conjugates. Bioorg Med Chem Lett 2007; 17:2983-6. [PMID: 17416522 DOI: 10.1016/j.bmcl.2007.03.067] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Revised: 03/19/2007] [Accepted: 03/22/2007] [Indexed: 10/23/2022]
Abstract
A series of bile acid-polyamine amides conjugated with 3'-azido-3'-deoxythymidine (AZT) as potential antitumor prodrugs in the form of phosphoramidates were synthesized in good yields and their antitumor activities were assayed against two human cancer cells in vitro: cervix cancer HeLa cells and renal cancer 7860 cells. The improved antitumor activity probably derived from the enhanced delivery efficiency of AZT due to bile acid-polyamine conjugates.
Collapse
Affiliation(s)
- Dimao Wu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Ministry of Education, Department of Chemistry, Tsinghua University, Beijing 100084, PR China
| | | | | | | | | |
Collapse
|
37
|
Olivero OA. Mechanisms of genotoxicity of nucleoside reverse transcriptase inhibitors. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2007; 48:215-23. [PMID: 16395695 DOI: 10.1002/em.20195] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Nucleoside analogs were first approved by the U.S. Food and Drug Administration for use against HIV-AIDS in 1987. Since then, these agents, now commonly referred to as nucleoside reverse transcriptase inhibitors (NRTIs), have become essential components of the Highly Active Antiretroviral Therapy (HAART) drug combinations used for treatment of Human Immunodeficiency Virus-1 (HIV-1) infections. Their antiretroviral activity is likely two-fold: incorporation of the drug into viral DNA and inhibition of the viral reverse transcriptase. However, incorporation of the drug into host nuclear and mitochondrial DNA may be largely responsible for dose-limiting toxicities. Azidothymidine (AZT, 3'-azido-3'-deoxythymidine, zidovudine), the first NRTI approved for the therapy of HIV-1, is incorporated into DNA, causes mutations in the hypoxanthine-guanine phosphoribosyl-transferase (HPRT) and thymidine kinase (TK) genes, and induces micronuclei, chromosomal aberrations, sister chromatid exchange, shortened telomeres, and other genotoxic effects in cultured cells. Genomic instability would be predicted as a consequence of these events. Metabolic pathways that result in the phosphorylation of AZT play a crucial role in AZT-DNA incorporation, and may be altered after prolonged treatment. For example, thymidine kinase 1, the enzyme responsible for AZT mono-phosphorylation, is down-regulated during long-term exposure and appears to be associated with AZT-induced replication inhibition and the accumulation of cells in S-phase. Detailed information on the mechanisms underlying NRTI-associated antiretroviral efficacy, toxicity, and metabolic resistance were not available when AZT was first approved for use as an antiretroviral agent. Current insights, based on 15 years of research, may lead to intervention strategies to attenuate toxicity without altering drug efficacy.
Collapse
Affiliation(s)
- Ofelia A Olivero
- Carcinogen-DNA Interactions Section, Laboratory of Cellular Carcinogenesis and Tumor Promotion, National Cancer Institute, NIH, Bethesda, Maryland, USA.
| |
Collapse
|
38
|
Chang YJ, Mittal V, Remine S, Manyam H, Sabir M, Richardson T, Young S. Correlation between Clinical and Histological Findings in Parathyroid Tumors Suspicious for Carcinoma. Am Surg 2006. [DOI: 10.1177/000313480607200511] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Carcinoma of the parathyroid is a rare malignancy that can be cured surgically if the proper diagnosis and treatment is given initially. Arriving to the clinical suspicion of a malignancy preoperatively is by far the most important step for a good prognosis. Our goal is to review the correlation between clinical and final histopathological findings that can arouse the suspicion of such malignancy and their true predictive value in the diagnosis. All patients that underwent surgical removal of the parathyroid mass between March of 1992 and March of 2003 were reviewed retrospectively at Providence Hospital and Medical Centers. Among 168 patients who underwent parathyroid excision, 14 (8.3%) had hyperplasia of the parathyroid, 121 (72%) had benign adenoma, 25 (14.8%) had other benign lesions, and 8 (4.7%) patients had primary carcinoma of the parathyroid confirmed by pathology. Our mean serum calcium level was 11.57 mg/dL, which was lower than the mean level (12 mg/dL) for benign hyperparathyroidism. The mean tumor size was 2.18 cm, smaller than the proposed for malignant criteria, and none of the eight patients (0%) had any symptoms of hypercalcemia at the time of diagnosis. Seven of eight patients (87.5%) had frank signs of invasion together with other histological features, and two patients had associated papillary carcinoma of the thyroid. Five patients from our series did not meet clinical criteria for malignancy (tumor size > 3 cm, palpable mass, and serum calcium > 14 mg/dL), but had undisputable histological findings (high mitotic pattern, fibrous trabeculae, capsular invasion, vascular invasion, and nodular involvement). On the other hand, 17 patients with benign histology had tumors greater than 3 cm, and an additional 18 had palpable masses on physical examination. We believe that these patients need to be followed closely. The patients with diagnosis of parathyroid carcinoma, their kindred, and those with large adenomas may benefit from genetic screening for HRTP2 gene mutations in search of early detection of tumors suspicious for malignancy. This is based on the fact that we did not find correlation between the clinical presentation and the histological features in our patients with proven malignancy.
Collapse
Affiliation(s)
- Yeon-Jeen Chang
- From the Department of Surgery, Providence Hospital and Medical Centers, Southfield, Michigan
| | - Vijay Mittal
- From the Department of Surgery, Providence Hospital and Medical Centers, Southfield, Michigan
| | - Stephen Remine
- From the Department of Surgery, Providence Hospital and Medical Centers, Southfield, Michigan
| | - Harish Manyam
- From the Department of Surgery, Providence Hospital and Medical Centers, Southfield, Michigan
| | - Mubashir Sabir
- From the Department of Surgery, Providence Hospital and Medical Centers, Southfield, Michigan
| | - Todd Richardson
- From the Department of Surgery, Providence Hospital and Medical Centers, Southfield, Michigan
| | - Shun Young
- From the Department of Surgery, Providence Hospital and Medical Centers, Southfield, Michigan
| |
Collapse
|
39
|
Torres PU, Prié D, Beck L, Friedlander G. New Therapies for Uremic Secondary Hyperparathyroidism. J Ren Nutr 2006; 16:87-99. [PMID: 16567265 DOI: 10.1053/j.jrn.2006.01.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Indexed: 11/11/2022] Open
Abstract
Secondary hyperparathyroidism (SHPT) is a common and serious complication of chronic kidney disease (CKD). It affects more than 300,000 end-stage renal disease patients treated by dialysis and probably more than 3 million patients with CKD worldwide. For a long time, traditional therapies for SHPT had consisted of correcting the hypocalcemia using calcium salts and vitamin D derivatives, preventing the hyperphosphatemia by calcium- or aluminum-containing intestinal phosphate binders, and recently by using no metal-containing intestinal phosphate binders; however, these therapies are limited by the occurrence of hypercalcemia, hyperphosphatemia, and the lack of specificity and long-term efficacy. Moreover, surgical parathyroidectomy (PTX), which remains the gold standard therapy, is not exempt from risk. PTX exposes patients to anesthesia risks, presurgical and postsurgical complications, and in many cases a permanent state of hypoparathyroidism. Thus, the medical treatment of SHPT became an ideal target for the development of new therapies and strategies. The purpose of this article is to provide an overview of these new therapies, including vitamin D analogs, intestinal phosphate binders, calcimimetics, parathyroidectomies, tyrosine kinase inhibitors, azydothymidine, anticalcineurins, N-terminal truncated parathyroid hormone fragments, bisphosphonates, calcitonin, osteoprotegerin, and others. The use of these new therapies alone or in combination may help to optimize the future treatment of SHPT in CKD patients.
Collapse
Affiliation(s)
- Pablo Ureña Torres
- Service de Néphrologie et Dialyse, Clinique de l'Orangerie, Aubervilliers, France.
| | | | | | | |
Collapse
|