1
|
Bora RE, Bilgicli HG, Üç EM, Alagöz MA, Zengin M, Gulcin İ. Synthesis, characterization, Evaluation of Metabolic Enzyme Inhibitors and in silico Studies of Thymol Based 2-Amino Thiol and Sulfonic Acid Compounds. Chem Biol Interact 2022; 366:110134. [DOI: 10.1016/j.cbi.2022.110134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 11/03/2022]
|
2
|
Vasin MV. B-190 (Indralin) in Light of the History of the Formation of Ideas about the Mechanism of Action of Radioprotectors. BIOL BULL+ 2022. [DOI: 10.1134/s1062359021110091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
3
|
Barreiro-Lage D, Nicolafrancesco C, Kočišek J, Luna A, Kopyra J, Alcamí M, Huber BA, Martín F, Domaracka A, Rousseau P, Díaz-Tendero S. Controlling the diversity of ion-induced fragmentation pathways by N-methylation of amino acids. Phys Chem Chem Phys 2022; 24:941-954. [PMID: 34913940 DOI: 10.1039/d1cp04097a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We present a combined experimental and theoretical study of the fragmentation of singly and doubly N-methylated glycine (sarcosine and N,N-dimethyl glycine, respectively) induced by low-energy (keV) O6+ ions. Multicoincidence mass spectrometry techniques and quantum chemistry simulations (ab initio molecular dynamics and density functional theory) allow us to characterise different fragmentation pathways as well as the associated mechanisms. We focus on the fragmentation of doubly ionised species, for which coincidence measurements provide unambiguous information on the origin of the various charged fragments. We have found that single N-methylation leads to a larger variety of fragmentation channels than in no methylation of glycine, while double N-methylation effectively closes many of these fragmentation channels, including some of those appearing in pristine glycine. Importantly, the closure of fragmentation channels in the latter case does not imply a protective effect by the methyl group.
Collapse
Affiliation(s)
- Darío Barreiro-Lage
- Departamento de Química, Módulo 13, Universidad Autónoma de Madrid, Madrid 28049, Spain.
| | - Chiara Nicolafrancesco
- Normandie Univ, ENSICAEN, UNICAEN, CEA, CNRS, CIMAP, Caen 14000, France. .,Synchrotron SOLEIL, LOrme des Merisiers, St Aubin, BP 48, Gif sur Yvette Cedex 91192, France
| | - Jaroslav Kočišek
- J. Heyrovsky Institute of Physical Chemistry v.v.i., The Czech Academy of Sciences, Dolejskova 3, Prague 18223, Czech Republic
| | - Alberto Luna
- Centro de Computación Científica, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Janina Kopyra
- Faculty of Exact and Natural Sciences, Siedlce University of Natural Sciences and Humanities, 3 Maja 54, Siedlce 08-110, Poland
| | - Manuel Alcamí
- Departamento de Química, Módulo 13, Universidad Autónoma de Madrid, Madrid 28049, Spain. .,Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA-Nano), Campus de Cantoblanco, Madrid 28049, Spain.,Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Bernd A Huber
- Normandie Univ, ENSICAEN, UNICAEN, CEA, CNRS, CIMAP, Caen 14000, France.
| | - Fernando Martín
- Departamento de Química, Módulo 13, Universidad Autónoma de Madrid, Madrid 28049, Spain. .,Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA-Nano), Campus de Cantoblanco, Madrid 28049, Spain.,Condensed Matter Physics Center (IFIMAC), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Alicja Domaracka
- Normandie Univ, ENSICAEN, UNICAEN, CEA, CNRS, CIMAP, Caen 14000, France.
| | - Patrick Rousseau
- Normandie Univ, ENSICAEN, UNICAEN, CEA, CNRS, CIMAP, Caen 14000, France.
| | - Sergio Díaz-Tendero
- Departamento de Química, Módulo 13, Universidad Autónoma de Madrid, Madrid 28049, Spain. .,Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid, Madrid 28049, Spain.,Condensed Matter Physics Center (IFIMAC), Universidad Autónoma de Madrid, Madrid 28049, Spain
| |
Collapse
|
4
|
Iqbal S, Shah MA, Rasul A, Saadullah M, Tabassum S, Ali S, Zafar M, Muhammad H, Uddin MS, Batiha GES, Vargas-De-La-Cruz C. Radioprotective Potential of Nutraceuticals and their Underlying Mechanism of Action. Anticancer Agents Med Chem 2021; 22:40-52. [PMID: 33622231 DOI: 10.2174/1871520621666210223101246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 11/22/2022]
Abstract
Radiations are an efficient treatment modality in cancer therapy. Besides the treatment effects of radiations, the ionizing radiations interact with biological systems and generate reactive oxygen species that interfere with the normal cellular process. Previous investigations of synthetic radioprotectors have shown less effectiveness, mainly owing to some limiting effects. The nutraceuticals act as efficient radioprotectors to protect the tissues from the deleterious effects of radiation. The main radioprotection mechanism of nutraceuticals is the scavenging of free radicals while other strategies are involved modulation of signaling transduction of pathways like MAPK (JNK, ERK1/2, ERK5, and P38), NF-kB, cytokines, and their protein regulatory genes expression. The current review is focused on the radioprotective effects of nutraceuticals including vitamin E, -C, organosulphur compounds, phenylpropanoids, and polysaccharides. These natural entities protect against radiation-induced DNA damage. The review mainly entails the antioxidant perspective and mechanism of action of their radioprotective activities on a molecular level, DNA repair pathway, anti-inflammation, immunomodulatory effects, the effect on cellular signaling pathways, and regeneration of hematopoietic cells.
Collapse
Affiliation(s)
- Shabnoor Iqbal
- Department of Zoology, Faculty of Life Sciences, Government College University Faisalabad. Pakistan
| | - Muhammad A Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad. Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University Faisalabad. Pakistan
| | - Malik Saadullah
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad. Pakistan
| | - Sobia Tabassum
- Department of Biological Sciences, International Islamic University, Islamabad. Pakistan
| | - Shujat Ali
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013. China
| | - Muhammad Zafar
- Department of Plant Sciences, Quaid-i-Azam University, Islamabad. Pakistan
| | - Haji Muhammad
- Department of Chemistry, Federal Urdu University of Arts, Science & Technology, Karachi. Pakistan
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka. Pakistan
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira. Egypt
| | - Celia Vargas-De-La-Cruz
- Faculty of Pharmacy and Biochemistry, Academic Department of Pharmacology, Bromatology and Toxicology, Centro Latinoamericano de Enseñanza e Investigación en Bacteriología Alimentaria (CLEIBA), Universidad Nacional Mayor de San Marcos, Lima15001. Peru
| |
Collapse
|
5
|
Wu J, Duan Y, Cui J, Dong Y, Li H, Wang M, Fan S, Li D, Li Y. Protective effects of zingerone derivate on ionizing radiation-induced intestinal injury. JOURNAL OF RADIATION RESEARCH 2019; 60:740-746. [PMID: 31665488 PMCID: PMC7357233 DOI: 10.1093/jrr/rrz065] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/28/2019] [Indexed: 05/06/2023]
Abstract
Intestinal injury is the primary toxicity of radiotherapy for pelvic and abdominal tumors, and it is also one of the common acute complications of radiotherapy. At present, there are no effective drugs to prevent intestinal injury in the clinic. Zingerone is a natural product with radioprotective effects. In this study, a novel compound (thiazolidine hydrochloride, TZC01) was synthesized by structural modification of zingerone. The effects of TZC01 on preventing intestinal injury from radiation were further investigated in this study. C57BL/6N mice were exposed to a lethal dose of abdominal irradiation (ABI) with and without TZC01 treatments. The morphological changes of the intestine and various makers of intestinal crypt cells were investigated. Treatment with TZC01 improved the survival rate of mice exposed to 12 Gy ABI. Moreover, TZC01 protected the intestinal morphology of mice, decreased the apoptotic rate of intestinal crypt cells, maintained cell regeneration and promoted crypt cell proliferation and differentiation. This study suggests that TZC01 has preventive and therapeutic effects on radiation enteritis by promoting the proliferation and differentiation of crypt cells to protect the small intestine from the toxic effects of ionizing radiation. Furthermore, the study of TCZ01 lays a strong foundation for developing novel radioprotectors with multiple properties.
Collapse
Affiliation(s)
- Jing Wu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Yuqing Duan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Jie Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
- School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yinping Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Hongyan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Meifang Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
- Corresponding author. Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, No 238, Baidi Road, Nankai district, Tianjin 300192, China, Tel: +86 022-85682340; Fax: +86 022-85683033.
| | - Yiliang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
6
|
Cheng Y, Dong Y, Hou Q, Wu J, Zhang W, Tian H, Li D. The protective effects of XH-105 against radiation-induced intestinal injury. J Cell Mol Med 2019; 23:2238-2247. [PMID: 30663222 PMCID: PMC6378229 DOI: 10.1111/jcmm.14159] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/17/2018] [Accepted: 12/24/2018] [Indexed: 12/26/2022] Open
Abstract
Radiation-induced intestinal injury is one of the major side effects in patients receiving radiation therapy. There is no specific treatment for radiation enteritis in the clinic. We designed and synthesized a new compound named XH-105, which is expected to cleave into polyphenol and aminothiol in vivo to mitigate radiation injury. In the following study, we describe the beneficial effects of XH-105 against radiation-induced intestinal injury. C57BL/6J mice were treated by gavage with XH-105 1 hour before total body irradiation (TBI), and the survival rate was monitored. Histological changes were examined, and survival of Lgr5+ intestinal stem cells Ki67+ cells, villi+ enterocytes and lysozymes was determined by immunohistochemistry. DNA damage and cellular apoptosis in intestinal tissue were also evaluated. Compared to vehicle-treated mice after TBI, XH-105 treatment significantly enhanced the survival rate, attenuated structural damage of the small intestine, decreased the apoptotic rate, reduced DNA damage, maintained cell regeneration and promoted crypt proliferation and differentiation. XH-105 also reduced the expression of Bax and p53 in the small intestine. These data suggest that XH-105 is beneficial for the protection of radiation-induced intestinal injury by inhibiting the p53-dependent apoptosis signalling pathway.
Collapse
Affiliation(s)
- Ying Cheng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.,Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Yinping Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Qinlian Hou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Jing Wu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Wei Zhang
- Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| |
Collapse
|
7
|
Fischer N, Seo EJ, Efferth T. Prevention from radiation damage by natural products. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 47:192-200. [PMID: 30166104 DOI: 10.1016/j.phymed.2017.11.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 10/20/2017] [Accepted: 11/12/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND Radiotherapy is a mainstay of cancer treatment since decades. Ionizing radiation (IR) is used for destruction of cancer cells and shrinkage of tumors. However, the increase of radioresistance in cancer cells and radiation toxicity to normal tissues are severe concerns. The exposure to radiation generates intracellular reactive oxygen species (ROS), which leads to DNA damage by lipid peroxidation, removal of thiol groups from cellular and membrane proteins, strand breaks and base alterations. HYPOTHESIS Plants have to deal with radiation-induced damage (UV-light of sun, other natural radiation sources). Therefore, it is worth speculating that radioprotective mechanisms have evolved during evolution of life. We hypothesize that natural products from plants may also protect from radiation damage caused as adverse side effects of cancer radiotherapy. METHODS The basis of this systematic review, we searched the relevant literature in the PubMed database. RESULTS Flavonoids, such as genistein, epigallocatechin-3-gallate, epicatechin, apigenin and silibinin mainly act as antioxidant, free radical scavenging and anti-inflammatory compounds, thus, providing cytoprotection in addition to downregulation of several pro-inflammatory cytokines. Comparable effects have been found in phenylpropanoids, especially caffeic acid phenylethylester, curcumin, thymol and zingerone. Besides, resveratrol and quercetin are the most important cytoprotective polyphenols. Their radioprotective effects are mediated by a wide range of mechanisms mainly leading to direct or indirect reduction of cellular stress. Ascorbic acid is broadly used as antioxidant, but it has also shown activity in reducing cellular damage after irradiation mainly due to its antioxidant capabilities. The metal ion chelator, gallic acid, represents another natural product attenuating cellular damage caused by radiation. CONCLUSIONS Some secondary metabolites from plants reveal radioprotective features against cellular damage caused by irradiation. These results warrant further analysis to develop phytochemicals as radioprotectors for clinical use.
Collapse
Affiliation(s)
- Nicolas Fischer
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Ean-Jeong Seo
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
8
|
Raghuraman M, Verma P, Kunwar A, Phadnis PP, Jain VK, Priyadarsini KI. Cellular evaluation of diselenonicotinamide (DSNA) as a radioprotector against cell death and DNA damage. Metallomics 2017; 9:715-725. [DOI: 10.1039/c7mt00034k] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
9
|
Dihydroxyselenolane (DHS) supplementation improves survival following whole-body irradiation (WBI) by suppressing tissue-specific inflammatory responses. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2016; 807:33-46. [DOI: 10.1016/j.mrgentox.2016.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/24/2016] [Accepted: 07/05/2016] [Indexed: 01/09/2023]
|
10
|
Lachumy SJ, Oon CE, Deivanai S, Saravanan D, Vijayarathna S, Choong YS, Yeng C, Latha LY, Sasidharan S. Herbal Remedies for Combating Irradiation: a Green Anti-irradiation Approach. Asian Pac J Cancer Prev 2013; 14:5553-65. [DOI: 10.7314/apjcp.2013.14.10.5553] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
11
|
Lim Y, Hedayati M, Merchant AA, Zhang Y, Yu HHM, Kastan MB, Matsui W, Deweese TL. Chloroquine improves survival and hematopoietic recovery after lethal low-dose-rate radiation. Int J Radiat Oncol Biol Phys 2012; 84:800-6. [PMID: 22445004 DOI: 10.1016/j.ijrobp.2012.01.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 12/17/2011] [Accepted: 01/06/2012] [Indexed: 01/09/2023]
Abstract
PURPOSE We have previously shown that the antimalarial agent chloroquine can abrogate the lethal cellular effects of low-dose-rate (LDR) radiation in vitro, most likely by activating the ataxia-telangiectasia mutated (ATM) protein. Here, we demonstrate that chloroquine treatment also protects against lethal doses of LDR radiation in vivo. METHODS AND MATERIALS C57BL/6 mice were irradiated with a total of 12.8 Gy delivered at 9.4 cGy/hour. ATM null mice from the same background were used to determine the influence of ATM. Chloroquine was administered by two intraperitoneal injections of 59.4 μg per 17 g of body weight, 24 hours and 4 hours before irradiation. Bone marrow cells isolated from tibia, fibula, and vertebral bones were transplanted into lethally irradiated CD45 congenic recipient mice by retroorbital injection. Chimerism was assessed by flow cytometry. In vitro methylcellulose colony-forming assay of whole bone marrow cells and fluorescence activated cell sorting analysis of lineage depleted cells were used to assess the effect of chloroquine on progenitor cells. RESULTS Mice pretreated with chloroquine before radiation exhibited a significantly higher survival rate than did mice treated with radiation alone (80% vs. 31%, p = 0.0026). Chloroquine administration before radiation did not affect the survival of ATM null mice (p = 0.86). Chloroquine also had a significant effect on the early engraftment of bone marrow cells from the irradiated donor mice 6 weeks after transplantation (4.2% vs. 0.4%, p = 0.015). CONCLUSION Chloroquine administration before radiation had a significant effect on the survival of normal but not ATM null mice, strongly suggesting that the in vivo effect, like the in vitro effect, is also ATM dependent. Chloroquine improved the early engraftment of bone marrow cells from LDR-irradiated mice, presumably by protecting the progenitor cells from radiation injury. Chloroquine thus could serve as a very useful drug for protection against the harmful effects of LDR radiation.
Collapse
Affiliation(s)
- Yiting Lim
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Chaudhary P, Shukla SK, Sharma RK. REC-2006-A Fractionated Extract of Podophyllum hexandrum Protects Cellular DNA from Radiation-Induced Damage by Reducing the Initial Damage and Enhancing Its Repair In Vivo. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2011; 2011:473953. [PMID: 20008078 PMCID: PMC3136531 DOI: 10.1093/ecam/nep212] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 11/09/2009] [Indexed: 11/13/2022]
Abstract
Podophyllum hexandrum, a perennial herb commonly known as the Himalayan May Apple, is well known in Indian and Chinese traditional systems of medicine. P. hexandrum has been widely used for the treatment of venereal warts, skin infections, bacterial and viral infections, and different cancers of the brain, lung and bladder. This study aimed at elucidating the effect of REC-2006, a bioactive fractionated extract from the rhizome of P. hexandrum, on the kinetics of induction and repair of radiation-induced DNA damage in murine thymocytes in vivo. We evaluated its effect on non-specific radiation-induced DNA damage by the alkaline halo assay in terms of relative nuclear spreading factor (RNSF) and gene-specific radiation-induced DNA damage via semi-quantitative polymerase chain reaction. Whole body exposure of animals with gamma rays (10 Gy) caused a significant amount of DNA damage in thymocytes (RNSF values 17.7 ± 0.47, 12.96 ± 1.64 and 3.3 ± 0.014) and a reduction in the amplification of β-globin gene to 0, 28 and 43% at 0, 15 and 60 min, respectively. Administrating REC-2006 at a radioprotective concentration (15 mg kg−1 body weight) 1 h before irradiation resulted in time-dependent reduction of DNA damage evident as a decrease in RNSF values 6.156 ± 0.576, 1.647 ± 0.534 and 0.496 ± 0.012, and an increase in β-globin gene amplification 36, 95 and 99%, at 0, 15 and 60 min, respectively. REC-2006 scavenged radiation-induced hydroxyl radicals in a dose-dependent manner stabilized DPPH free radicals and also inhibited superoxide anions. Various polyphenols and flavonoides present in REC-2006 might contribute to scavenging of radiation-induced free radicals, thereby preventing DNA damage and stimulating its repair.
Collapse
Affiliation(s)
- Pankaj Chaudhary
- Institute of Nuclear Medicine and Allied Sciences (INMAS), Delhi, India
| | | | | |
Collapse
|
13
|
Lion C, Boukou-Poba JP, Charvy C. Synthèse Dans La Série Des Phenanthridines. IV. Préparation De Nouveaux Radioprotecteurs A Motif Phenanthridine. ACTA ACUST UNITED AC 2010. [DOI: 10.1002/bscb.19911000210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
14
|
Rudneva TN, Sanina NA, Lyssenko KA, Aldoshin SM, Antipin MY, Ovanesyan NS. Synthesis and structure of a water-soluble nitrosyl iron complex with cysteamine ligand. MENDELEEV COMMUNICATIONS 2009. [DOI: 10.1016/j.mencom.2009.09.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Stankiewicz A, Skrzydlewska E. Amifostine—Antioxidant Drug in Anticancer Therapy. Toxicol Mech Methods 2008; 16:181-8. [DOI: 10.1080/15376520500195608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
16
|
Paunesku D, Paunesku T, Wahl A, Kataoka Y, Murley J, Grdina DJ, Woloschak GE. Incidence of tissue toxicities in gamma ray and fission neutron-exposed mice treated with Amifostine. Int J Radiat Biol 2008; 84:623-34. [PMID: 18661379 DOI: 10.1080/09553000802241762] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE To determine the effects of Amifostine or WR-151,327 on the incidence of lethal and non-lethal toxicities in a large cohort of mice exposed to gamma-ray or fission-spectrum neutron radiation. METHODS To analyze data from 4000 B6CF1 mice which received a single whole body irradiation (WBI) with 206 cGy or 417 cGy cobalt-60 gamma rays or 10 cGy or 40 cGy of fission-spectrum neutrons (average energy 0.85 MeV) produced by the Janus reactor at Argonne National Laboratory. In the neutron cohort, Amifostine, WR-151,327, saline or nothing was injected once, intraperitoneally, 30 minutes before irradiation. In the cobalt-60 cohort, WR-151327 was omitted from the same protocol. At the time of natural death, tissue toxicities found in these mice were recorded, and these records were analyzed. While all previous studies focused on the modulation of life shortening effects of WBI by Amifostine, in this study we calculated changes in the frequencies of 59 tissue toxicities and changes in the total number of toxicities per animal. RESULTS Amifostine protected against specific non-tumor pathological complications (67% of the non-tumor toxicities induced by gamma irradiation, 31% of the neutron induced specific toxicities), as well as specific tumors (56% of the tumor toxicities induced by gamma irradiation, 25% of the neutron induced tumors). Amifostine also reduced the total number of toxicities per animal for both genders in the gamma ray exposed mice and in males in the neutron exposed mice. CONCLUSIONS Amifostine was protective against many, but not all, tissue toxicities caused by WBI gamma and neutron irradiation.
Collapse
Affiliation(s)
- David Paunesku
- Department of Radiation Oncology, University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Shukla SK, Chaudhary P, Kumar IP, Samanta N, Afrin F, Gupta ML, Sharma UK, Sinha AK, Sharma YK, Sharma RK. Protection from radiation-induced mitochondrial and genomic DNA damage by an extract of Hippophae rhamnoides. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2006; 47:647-56. [PMID: 16948057 DOI: 10.1002/em.20251] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Hippophae rhamnoides or seabuckthorn is used extensively in Indian and Tibetan traditional medicine for the treatment of circulatory disorders, ischemic heart disease, hepatic injury, and neoplasia. In the present study, we have evaluated the radioprotective potential of REC-1001, a fraction isolated from the berries of H. rhamnoides. Chemical analysis of the extract indicated that REC-1001 was approximately 68% by weight polyphenols, and contained kaempferol, isorhamnetin, and quercetin. The effect of REC-1001 on modulating radiation-induced DNA damage was determined in murine thymocytes by measuring nonspecific nuclear DNA damage at the whole genome level using the alkaline halo assay and by measuring sequence/gene-specific DNA damage both in nuclear DNA (beta-globin gene) and in mitochondrial DNA using a quantitative polymerase chain reaction. Treatment with 10 Gy resulted in a significant amount of DNA damage in the halo assay and reductions in the amplification of both the beta-globin gene and mitochondrial DNA. REC-1001 dose-dependently reduced the amount of damage detected in each assay, with the maximum protective effects observed at the highest REC-1001 dose evaluated (250 micro g/ml). Studies measuring the nicking of naked plasmid DNA further established the radioprotective effect of REC-1001. To elucidate possible mechanisms of action, the antioxidant properties and the free-radical scavenging activities of REC-1001 were evaluated. REC-1001 dose-dependently scavenged radiation-induced hydroxyl radicals, chemically-generated superoxide anions, stabilized DPPH radicals, and reduced Fe(3+) to Fe(2+). The results of the study indicate that the REC-1001 extract of H. rhamnoides protects mitochondrial and genomic DNA from radiation-induced damage. The polyphenols/flavonoids present in the extract might be responsible for the free radical scavenging and DNA protection afforded by REC-1001.
Collapse
Affiliation(s)
- Sandeep Kumar Shukla
- Division of Radioimaging, Bioinformatics, and Radiation Biology, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Rharass T, Vigo J, Salmon JM, Ribou AC. Variation of 1-pyrenebutyric acid fluorescence lifetime in single living cells treated with molecules increasing or decreasing reactive oxygen species levels. Anal Biochem 2006; 357:1-8. [PMID: 16920058 DOI: 10.1016/j.ab.2006.07.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Revised: 07/04/2006] [Accepted: 07/07/2006] [Indexed: 10/24/2022]
Abstract
1-Pyrenebutyric acid (PBA) is a fluorescent probe whose fluorescence lifetime depends on local oxygen and free radical concentrations. We propose to use PBA fluorescence lifetime to quantify reactive oxygen species (ROS) in biological samples. Time-resolved microfluorimetry was used to record the fluorescence decay of single living cells loaded with this probe. We measured intracellular PBA fluorescence lifetimes and reduced nicotinamide adenine dinucleotide phosphate intensities under various oxygen concentrations. To confirm the feasibility of the new method, CCRF-CEM cells were treated with drugs that are known to increase or decrease ROS production. After treatment with adriamycin, we observed a decrease of PBA fluorescence lifetime. This corresponded to an increase of ROS concentration (80%). After treatment with cysteamine, we observed a reduction of the ROS concentration by 67%. Moreover, addition of exogenous H(2)O(2) solution resulted in a decrease of PBA fluorescence lifetime due to a raising of the intracellular ROS concentration. These results support our hypothesis linking a part of PBA fluorescence lifetime variations to intracellular fluctuation of ROS.
Collapse
Affiliation(s)
- Tareck Rharass
- BDSI Biophysic and Dynamic of Integrated Systems, University of Perpignan, 52, av. Paul Alduy, 66860 Perpignan, France
| | | | | | | |
Collapse
|
19
|
Kronrád L, Kozák I. Transformation of aminoethylisouronium salts. 1, S-aminoethylisothiouronium bromide hydrobromide-14C. ACTA ACUST UNITED AC 2006. [DOI: 10.1002/jlcr.2590090113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
20
|
La Scala GC, O'Donovan DA, Yeung I, Darko J, Addison PD, Neligan PC, Pang CY, Forrest CR. Radiation-induced craniofacial bone growth inhibition: efficacy of cytoprotection following a fractionated dose regimen. Plast Reconstr Surg 2006; 115:1973-85. [PMID: 15923846 DOI: 10.1097/01.prs.0000163322.22436.3b] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Severe craniofacial growth disturbances are noted in 66 to 100 percent of children with head and neck cancers who received radiotherapy during their growing years. The authors have previously demonstrated the prevention of radiation-induced craniofacial bone growth inhibition following single-dose orthovoltage radiation to the orbitozygomatic complex in an infant rabbit model through the administration of the cytoprotective agent amifostine (WR-2721) before radiation treatment. The purpose of this study was to investigate the efficacy of cytoprotection using a fractionated dose regimen that better approximates the clinical application of radiation therapy. METHODS Thirty 7-week-old male New Zealand rabbits were randomized into three groups (n = 10), each receiving six fractions of orthovoltage radiation to the right orbitozygomatic complex: group C, sham irradiation control; group F35, total dose of 35 Gy; and group F35A, total dose of 35 Gy with administration of amifostine 200 mg/kg intravenously 20 minutes before each fraction. Bone growth was evaluated up to skeletal maturity (age 21 weeks) with serial radiographs and computed tomography scans for cephalometric analysis, bone volume, and bone density measurements. RESULTS Fractionated radiation resulted in significant (p < 0.05) bone growth inhibition compared with sham radiation in 16 of 21 cephalometric parameters measured and significantly (p < 0.05) reduced bone volume of the rabbit orbitozygomatic complex. Pretreatment with amifostine before each radiation fraction prevented growth deformities in four cephalometric parameters and significantly (p < 0.05) attenuated these effects in another seven parameters compared with radiated animals. Bone volumes were also significantly (p < 0.05) improved in F35A animals compared with F35 animals. CONCLUSIONS This study establishes that fractionation of orthovoltage radiation does not prevent the development of growth disturbances of the rabbit craniofacial skeleton and also demonstrates that preirradiation administration of amifostine is highly effective in the prevention and attenuation of radiation-induced craniofacial bone growth inhibition.
Collapse
Affiliation(s)
- Giorgio C La Scala
- Division of Plastic Surgery, The Hospital for Sick Children Center for Craniofacial Care and Research, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Amifostine (Ethyol, WR-2721; MedImmune, Inc, Gaithersburg, MD) is a member of a sulfhydryl-containing class of compounds that protects normal tissue and organs against ionizing radiation damage by scavenging radiation-induced radicals. The goal of this study was to assess the preclinical and clinical data on the protective effect of amifostine in normal organs and tissue. The current literature was reviewed and assessed for progress in the pathogenesis of radiation-induced pulmonary injury. Preclinical and clinical data on the protective effect of amifostine in radiation-induced lung and esophageal injuries were also critically assessed. Significant progress has been made in understanding the pathogenesis of radiation pneumonitis. Preclinical studies have shown strong evidence of the protective effect of amifostine in radiation-induced toxicities in rodents and monkeys. However, available clinical data are not conclusive in showing the protective effect of amifostine in radiation pneumonitis and esophagitis. Amifostine has been well tolerated with a low incidence of toxicities, which included nausea and vomiting (3% to 5%) and transient hypotension during intravenous infusion (7%). Preclinical data are promising for amifostine in protecting thoracic organs from radiation-induced toxicities. Studies measuring the magnitude of gain in tumor control and survival as a result of the enhanced protective effect of amifostine on normal tissue over that of tumor tissue are lacking. Such data would help in designing new approaches to maximize outcome. Additional well-designed phase III studies are necessary to confirm the clinical benefit of amifostine in minimizing radiation- and chemoradiation-related toxicities in patients with lung cancer.
Collapse
Affiliation(s)
- Noah C Choi
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| |
Collapse
|
22
|
Athanassiou H, Antonadou D, Coliarakis N, Kouveli A, Synodinou M, Paraskevaidis M, Sarris G, Georgakopoulos GR, Panousaki K, Karageorgis P, Throuvalas N. Protective effect of amifostine during fractionated radiotherapy in patients with pelvic carcinomas: results of a randomized trial. Int J Radiat Oncol Biol Phys 2003; 56:1154-60. [PMID: 12829154 DOI: 10.1016/s0360-3016(03)00187-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
PURPOSE To evaluate whether pretreatment with amifostine can reduce treatment-induced toxicity in patients with pelvic malignancies undergoing radiotherapy (RT). METHODS AND MATERIALS A total of 205 patients with pelvic malignancies (rectal, 32; bladder, 47; prostate, 40; gynecologic, 86) were randomized to receive RT (Group 1, n = 95) or RT plus amifostine (Group 2, n = 110). The patient characteristics for both treatment groups were well balanced. Amifostine was administered at 340 mg/m(2) i.v., 15 min before RT, with standard antiemetics 30 min before. All patients received conventional RT, radical (65-70 Gy) or postoperative (50 Gy), with 45 Gy given to the whole pelvis at daily fractions of 1.8-2.0 Gy, 5 d/wk. Skin, bowel, bladder, and hematologic toxicities were evaluated according to the Radiation Therapy Oncology Group/European Organization Research and Treatment of Cancer scoring system. RESULTS A significant reduction occurred in acute Grade 2-3 bladder and lower GI tract toxicities in the amifostine group (p <0.05, Weeks 3-7). With a median follow-up of 12 months, few late Grade 2-3 effects were observed in either group. No statistically significant difference between the two groups was observed in terms of response 6 weeks after RT completion (complete response plus partial response, 96.8% in the control and 98.3% in the amifostine arm). Amifostine was well tolerated, with only moderate hypotension occurring in 2 patients and moderate nausea in 1 patient. CONCLUSIONS The results of this randomized trial support the role of amifostine in reducing acute radiation-related toxicity of the bladder and lower GI tract in patients with pelvic malignancies, without evidence of tumor protection.
Collapse
Affiliation(s)
- Helen Athanassiou
- Department of Radiation Oncology, "Agioi Anargyri" Hospital, Athens, Greece.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
The potential of antioxidants to reduce the cellular damage induced by ionizing radiation has been studied in animal models for more than 50 years. The application of antioxidant radioprotectors to various human exposure situations has not been extensive although it is generally accepted that endogenous antioxidants, such as cellular non-protein thiols and antioxidant enzymes, provide some degree of protection. This review focuses on the radioprotective efficacy of naturally occurring antioxidants, specifically antioxidant nutrients and phytochemicals, and how they might influence various endpoints of radiation damage. Results from animal experiments indicate that antioxidant nutrients, such as vitamin E and selenium compounds, are protective against lethality and other radiation effects but to a lesser degree than most synthetic protectors. Some antioxidant nutrients and phytochemicals have the advantage of low toxicity although they are generally protective when administered at pharmacological doses. Naturally occurring antioxidants also may provide an extended window of protection against low-dose, low-dose-rate irradiation, including therapeutic potential when administered after irradiation. A number of phytochemicals, including caffeine, genistein, and melatonin, have multiple physiological effects, as well as antioxidant activity, which result in radioprotection in vivo. Many antioxidant nutrients and phytochemicals have antimutagenic properties, and their modulation of long-term radiation effects, such as cancer, needs further examination. In addition, further studies are required to determine the potential value of specific antioxidant nutrients and phytochemicals during radiotherapy for cancer.
Collapse
Affiliation(s)
- Joseph F Weiss
- Office of Health Studies, US Department of Energy, EH-6/270 Corporate Square, 1000 Independence Avenue, SW, Washington, DC 20585-0270, USA.
| | | |
Collapse
|
24
|
Basavaraju SR, Easterly CE. Pathophysiological effects of radiation on atherosclerosis development and progression, and the incidence of cardiovascular complications. Med Phys 2002; 29:2391-403. [PMID: 12408314 DOI: 10.1118/1.1509442] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Radiation therapy while important in the management of several diseases, is implicated in the causation of atherosclerosis and other cardiovascular complications. Cancer and atherosclerosis go through the same stages of initiation, promotion, and complication, beginning with a mutation in a single cell. Clinical observations before the 1960s lead to the belief that the heart is relatively resistant to the doses of radiation used in radiotherapy. Subsequently, it was discovered that the heart is sensitive to radiation and many cardiac structures may be damaged by radiation exposure. A significantly higher risk of death due to ischemic heart disease has been reported for patients treated with radiation for Hodgkin's disease and breast cancer. Certain cytokines and growth factors, such as TGF-beta1 and IL-1 beta, may stimulate radiation-induced endothelial proliferation, fibroblast proliferation, collagen deposition, and fibrosis leading to advanced lesions of atherosclerosis. The treatment for radiation-induced ischemic heart disease includes conventional pharmacological therapy, balloon angioplasty, and bypass surgery. Endovascular irradiation has been shown to be effective in reducing restenosis-like response to balloon-catheter injury in animal models. Caution must be exercised when radiation therapy is combined with doxorubicin because there appears to be a synergistic toxic effect on the myocardium. Damage to endothelial cells is a central event in the pathogenesis of damage to the coronary arteries. Certain growth factors that interfere with the apoptotic pathway may provide new therapeutic strategies for reducing the risk of radiation-induced damage to the heart. Exposure to low level occupational or environmental radiation appears to pose no undue risk of atherosclerosis development or cardiovascular mortality. But, other radiation-induced processes such as the bystander effects, abscopal effects, hormesis, and individual variations in radiosensitivity may be important in certain circumstances.
Collapse
|
25
|
Syng-ai C, Basu Baul TS, Chatterjee A. Antiproliferative and cytotoxic effect of a novel organotin compound on mammalian cells both in vitro and in vivo. Mutat Res 2002; 513:49-59. [PMID: 11719089 DOI: 10.1016/s1383-5718(01)00289-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Organotin compounds are organometallic compounds showing various toxicological properties. Several organotin compounds also showed an antineoplastic effect. However, their relative mutagenic potential is not well established. In this study Et(2)SnCl(2).L [L=N-(2-pyridylmethylene)-4-toluidine] (OTC) has been subjected to investigation for its cytotoxic effect in mouse bone marrow cells (BMCs) and human peripheral blood lymphocyte cells (HPBLs). The Sn [bond] N bond in OTC is 2.46A which is greater than 2.39A and therefore, a better formation of tin-DNA complex can be expected. The present data indicate that OTC induced significant delay in cell kinetics and sister chromatid exchanges (SCEs) in both BMCs and HPBLs, whereas, induction of chromosome aberrations was found only in HPBLs. The presence of buthionine sulfoximine (BSO) modulated cellular sensitivity towards OTC in both cell systems. It may be inferred that the OTC could bind on DNA more easily owing to its structural advantage and this may explain the induction of DNA damage and the delay in cell proliferation. Since the cytotoxic effect of OTC is more in glutathione depleted cells, the concentration of OTC may be reduced to get an antitumour effect in GSH-depleted cells and thus minimizes its toxic side effect.
Collapse
Affiliation(s)
- Christine Syng-ai
- Genetics Laboratory, Department of Zoology, North-Eastern Hill University, Shillong 793022, India
| | | | | |
Collapse
|
26
|
Antonadou D, Coliarakis N, Synodinou M, Athanassiou H, Kouveli A, Verigos C, Georgakopoulos G, Panoussaki K, Karageorgis P, Throuvalas N. Randomized phase III trial of radiation treatment +/- amifostine in patients with advanced-stage lung cancer. Int J Radiat Oncol Biol Phys 2001; 51:915-22. [PMID: 11704311 DOI: 10.1016/s0360-3016(01)01713-8] [Citation(s) in RCA: 136] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE This multicenter trial investigated whether daily pretreatment with amifostine (A) could reduce the incidence of acute and late lung toxicity and esophagitis without affecting antitumor efficacy of radiation in advanced lung cancer. PATIENTS AND METHODS Radiotherapy (XRT) patients (n = 146) received a daily fraction of 2 Gy/5 days/week to a total of 55-60 Gy +/- amifostine 340 mg/m(2) administered daily 15 min before irradiation. Acute and late toxicities were graded from 0 to 4 according to the Radiation Therapy Oncology Group/European Organization for the Research and Treatment of Cancer system. RESULTS Ninety-seven patients were evaluated 2 months post-XRT for the incidence of pneumonitis; 43% (23/53) of patients in the XRT arm and 9% (4/44) in the A + XRT arm experienced > or = Grade 2 pneumonitis (p < 0.001) [corrected]. Forty-nine percent (26/53) of patients in the XRT arm and 16% (7/44) in the A+XRT arm demonstrated changes representative of > or = Grade 2 lung damage (p < 0.001). At 6 months, fibrosis was present in 53% (19/36) receiving XRT vs. 28% (9/32) receiving A+XRT (p < 0.05). Incidence of esophagitis > or = Grade 2 during Week 4 was 42% (31/73) in the XRT arm vs. 4% (3/73) in the A+XRT arm (p < 0.001). Among 97 patients evaluable for response 2 months after XRT, complete or partial response was present in 76% (40/53) of patients in the XRT arm and 75% (33/44) in the A+XRT arm (p = 1.0). CONCLUSION Amifostine reduces the incidence of pneumonitis, lung fibrosis, and esophagitis in radiotherapy patients with lung cancer without compromising antitumor efficacy.
Collapse
Affiliation(s)
- D Antonadou
- Radiation Oncology Department, Metaxas Cancer Hospital, Pireus, Greece.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Grdina DJ, Kataoka Y, Murley JS. Amifostine: mechanisms of action underlying cytoprotection and chemoprevention. DRUG METABOLISM AND DRUG INTERACTIONS 2001; 16:237-79. [PMID: 11201306 DOI: 10.1515/dmdi.2000.16.4.237] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Amifostine is an important drug in the new field of cytoprotection. It was developed by the Antiradiation Drug Development Program of the US Army Medical Research and Development Command as a radioprotective compound and was the first drug from that Program to be approved for clinical use in the protection of dose limiting normal tissues in patients against the damaging effects of radiation and chemotherapy. Its unique polyamine-like structure and attached sulfhydryl group give it the potential to participate in a range of cellular processes that make it an exciting candidate for use in both cytoprotection and chemoprevention. Amifostine protects against the DNA damaging effects of ionizing radiation and chemotherapy drug associated reactive species. It possesses anti-mutagenic and anti-carcinogenic properties. At the molecular level, it has been demonstrated to affect redox sensitive transcription factors, gene expression, chromatin stability, and enzymatic activity. At the cellular level it has important effects on growth and cell cycle progression. This review focuses on relating its unique chemical design to mechanisms of action that underlie its broad usefulness as both a cytoprotective and chemopreventive agent for use in cancer therapy.
Collapse
Affiliation(s)
- D J Grdina
- Department of Radiation and Cellular Oncology, University of Chicago, MC 1105, Rm ES ESB 11B, 5841 S. Maryland Avenue, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
28
|
Guest I, Uetrecht J. Bone marrow stem cell protection from chemotherapy by low--molecular-weight compounds. Exp Hematol 2001; 29:123-37. [PMID: 11166451 DOI: 10.1016/s0301-472x(00)00621-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The stem cells of the bone marrow have the capacity for both self-renewal and derivation of all the blood cell lineages. Consequently, toxicity to these cells can result in neutropenia, agranulocytosis, thrombocytopenia, pancytopenia, or aplastic anemia. Many anticancer drugs adversely affect the bone marrow, and neutropenia is a common limiting factor in dose escalation. In this review, we discuss agents that appear to have potential as bone marrow sparing agents. Computerized catalogs of the National Library of Medicine and Medline were searched for reports on low-molecular-weight compounds that detailed effects on the hematopoietic progenitor cells. The most promising agents are the endogenous peptides p-glutamic acid-glutamic acid-aspartic acid-cysteine-lysine and acetyl-serine-aspartic acid-lysine-proline, and the exogenous compounds amifostine and ammonium trichloro[dioxoethylene-O,O']tellurate, but several others are also discussed. These compounds preserve stem cell function in the presence of antineoplastic drugs of diverse pharmacological classes, and they do so by various mechanisms of action. Their present status in clinical practice is also detailed. More needs to be learned about their mechanisms of action and therapeutic potential, but the results are encouraging for some of these compounds and more clinical trials should be expected.
Collapse
Affiliation(s)
- I Guest
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
29
|
Koukourakis MI, Kyrias G, Kakolyris S, Kouroussis C, Frangiadaki C, Giatromanolaki A, Retalis G, Georgoulias V. Subcutaneous administration of amifostine during fractionated radiotherapy: a randomized phase II study. J Clin Oncol 2000; 18:2226-33. [PMID: 10829042 DOI: 10.1200/jco.2000.18.11.2226] [Citation(s) in RCA: 204] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Amifostine (WR-2721) is an important cytoprotective agent. Although intravenous administration is the standard route, pharmacokinetic studies have shown acceptable plasma levels of the active metabolite of amifostine (WR-1605) after subcutaneous administration. The subcutaneous route, due to its simplicity, presents multiple advantages over the intravenous route when amifostine is used during fractionated radiotherapy. PATIENTS AND METHODS Sixty patients with thoracic, 40 with head and neck, and 40 with pelvic tumors who were undergoing radical radiotherapy were enrolled onto a randomized phase II trial to assess the feasibility, tolerance, and cytoprotective efficacy of amifostine administered subcutaneously. A flat dose of amifostine 500 mg, diluted in 2.5 mL of normal saline, was injected subcutaneously 20 minutes before each radiotherapy fraction. RESULTS The subcutaneous amifostine regimen was well tolerated by 85% of patients. In approximately 5% of patients, amifostine therapy was interrupted due to cumulative asthenia, and in 10%, due to a fever/rash reaction. Hypotension was never noted, whereas nausea was frequent. A significant reduction of pharyngeal, esophageal, and rectal mucositis was noted in the amifostine arm (P <.04). The delays in radiotherapy because of grade 3 mucositis were significantly longer in the group of patients treated with radiotherapy alone (P <.04). Amifostine significantly reduced the incidence of acute perineal skin and bladder toxicity (P <.0006). CONCLUSION Subcutaneous administration of amifostine is well tolerated, effectively reduces radiotherapy's early toxicity, and prevents delays in radiotherapy. The subcutaneous route is much simpler and saves time compared with the intravenous route of administration and can be safely and effectively applied in the daily, busy radiotherapy practice.
Collapse
Affiliation(s)
- M I Koukourakis
- Department of Radiotherapy/Oncology and Medical Oncology, University Hospital of Iraklion, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Tamurian RM, Damron TA, Spadaro JA. Sparing radiation-induced damage to the physis by radioprotectant drugs: laboratory analysis in a rat model. J Orthop Res 1999; 17:286-92. [PMID: 10221847 DOI: 10.1002/jor.1100170219] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The radioprotectant compound amifostine (S-2[3-aminopropylamino]-ethylphosphorothioic acid), administered prior to radiotherapy, has been demonstrated to provide differential protection of normal cells from the damaging effects of ionizing radiation. The aim of this pilot was to determine if amifostine could preserve the integrity of, or minimize the damage to, the physis during exposure to radiation in an animal model. Thirty weanling Sprague-Dawley rats were randomized into five groups of six animals each. Groups 1 and 2 received a single exposure to radiation consisting of 12.5 and 17.5 Gy, respectively. Groups 3 and 4 received similar exposures of 12.5 and 17.5 Gy, respectively, but with prior administration of amifostine at 100 mg/kg. Group 5 (control) received neither radiation nor amifostine. At 6 weeks, femoral and tibial lengths were measured in treated and untreated hindlimbs and compared with the baseline lengths to calculate growth. Concordant with previous reports in the literature, the radiation doses of 12.5 and 17.5 Gy reduced net femoral growth in length by a mean of 23% (range = 12-33%, SD = 7.41) and 59% (range = 54-64%, SD = 4.45), respectively, in the irradiated limb. Amifostine reduced anticipated growth loss normally resulting from a single 12.5-Gy radiation dose by 48.9% in the femur, 13.1% in the tibia, and 27.6% overall in the total limb (p < or = 0.05). Similarly, anticipated growth loss from a single 17.5-Gy radiation dose was reduced by 30.8% in the femur, 20.3% in the tibia, and 25.7% overall in the total limb (p < or = 0.05). Amifostine administered prior to clinically relevant radiation exposures significantly reduced the amount of anticipated growth arrest in our animal model.
Collapse
Affiliation(s)
- R M Tamurian
- Department of Orthopedic Surgery, State University of New York Health and Science Center at Syracuse, 13202, USA
| | | | | |
Collapse
|
31
|
Baul SB, Baul TSB, Rivarola E, Dakternieks D, Tiekink ERT, Syng-ai C, Chatterjee A. Synthesis and characterization of diorganotin(IV) complexes ofN-(2-pyridylmethylene)arylamines and mutagenicity testingin vivo of Et2SnCl2�[L4=N-(2-pyridylmethylene)-4-toluidine]. Appl Organomet Chem 1998. [DOI: 10.1002/(sici)1099-0739(199807)12:7<503::aid-aoc746>3.0.co;2-p] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
32
|
Jeitner TM, Delikatny EJ, Bartier WA, Capper HR, Hunt NH. Inhibition of drug-naive and -resistant leukemia cell proliferation by low molecular weight thiols. Biochem Pharmacol 1998; 55:793-802. [PMID: 9586951 DOI: 10.1016/s0006-2952(97)00575-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of these studies was to investigate the ability of cysteamine and its congeners to arrest the proliferation of leukemic cells and to determine the physico-chemical properties responsible for this ability. Fifteen low molecular weight thiol-bearing compounds were shown to arrest the proliferation of CCRF-CEM cells and a methotrexate-resistant subline, with IC50 values between 10(-5) and 10(-4) M. Cysteamine arrested proliferation by slowing the passage of cells through S phase. These cells subsequently resumed cycling, although a proportion went on to die by apoptosis. The antiproliferative action of cysteamine was shown to depend, in part, on H2O2 production. This ability to generate peroxide is shared by many thiol compounds, and molecular modeling indicated that thiol groups were required for the antiproliferative actions of the congeners of cysteamine. Molecular modeling also revealed that the most efficacious antiproliferative agents were those that had their amino acid and thiol moieties separated by an intramolecular distance of 3.17 to 5.9 A, as exemplified by WR 1065 and the aminothiophenols. These findings indicate that thiol-bearing compounds may have some efficacy in the treatment of drug-naive and -resistant leukemia cells.
Collapse
Affiliation(s)
- T M Jeitner
- Department of Pathology, University of Sydney, New South Wales, Australia.
| | | | | | | | | |
Collapse
|
33
|
VARANDA EA, TAVARES DC. RADIOPROTECTION: MECHANISMS AND RADIOPROTECTIVE AGENTS INCLUDING HONEYBEE VENOM. ACTA ACUST UNITED AC 1998. [DOI: 10.1590/s0104-79301998000100002] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- E. A. VARANDA
- São Paulo State University; UNESP, São Paulo State University, Botucatu, SP, Brazil
| | | |
Collapse
|
34
|
Abstract
The induction of micronucleated polychromatic erythrocytes (MNPCEs) was assessed in the bone marrow of adult male Swiss mice treated with MEA (cysteamine HCl), AET (2-aminoethylisothiouronium Br.HBr), or WR-2721 (S-2-(3-aminopropylamino)ethyl phosphorothioic acid), at a dose of 200 mg/kg body weight, and/or exposed to 6 Gy X-rays. MEA, AET, or WR-2721 was given alone or 15 min prior to X-ray exposure, and the frequency of MNPCEs was determined 24 h after the aminothiol treatment and X-irradiation of mice. A genotoxic effect was shown for MEA, AET, WR-2721, and X-rays, as well as a protective effect of the aminothiols against X-ray-induced genotoxicity in the mouse erythropoietic system. The aminothiol drugs given alone, without subsequent X-irradiation, elevated the frequency of MNPCEs, and WR-2721 appeared to be less toxic than AET and MEA. After exposure of mice to X-rays, the number of MNPCEs was distinctly increased. MEA, AET, or WR-2721 administration prior to X-irradiation resulted in a reduction of the X-ray-induced elevation of the frequency of micronuclei, but a stronger radioprotective effect was obtained following WR-2721 and AET treatment than after MEA application. So, the genotoxic and radioprotective effect of the aminothiols was dependent on the compound applied.
Collapse
Affiliation(s)
- L Mazur
- Department of Animal Physiology, Jagiellonian University, Cracow, Poland
| |
Collapse
|
35
|
Chatterjee A, Chattopadhyay A, Lawlor CJ. Effect of glutathione on sister-chromatid exchanges in normal and buthionine sulfoximine-treated mice. Mutat Res 1995; 327:171-7. [PMID: 7870086 DOI: 10.1016/0027-5107(94)00185-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Based on their ability to induce sister-chromatid exchanges (SCEs) it is evident that thiol-containing radioprotectors can induce DNA damage. However, there were contradictory findings when reduced glutathione (GSH) was tested using two cell lines. The present study demonstrated that GSH can induce SCEs and also delay in cell proliferation in mouse bone marrow cells in vivo. The presence of catalase significantly reduced GSH-induced SCE frequency down to catalase alone levels. An attempt was made to evaluate the effect of GSH treatment in buthionine sulfoximine (BSO)-treated mice (GSH-depleted mice) and the data indicate that induction of SCEs takes place without inducing a delay in cell proliferation or the generation of hydrogen peroxide. Probably, some unknown route is involved by which GSH-degraded product(s) induce SCEs in BSO-treated mice. Therefore, the induction of SCEs by GSH in normal mice may be largely due to hydrogen peroxide generation; however, the involvement of the binding ability of GSH to chromatin and the probable (unknown) route by which GSH-degraded product(s) may cause smaller fraction of SCEs cannot be ruled out.
Collapse
Affiliation(s)
- A Chatterjee
- Department of Zoology, School of Life Sciences, North-Eastern Hill University, Shillong, India
| | | | | |
Collapse
|
36
|
Murray D, Simpson R, Rosenberg E, Carraway A, Britten R. Correlation between gamma-ray-induced DNA double-strand breakage and cell killing after biologically relevant doses: analysis by pulsed-field gel electrophoresis. Int J Radiat Biol 1994; 65:419-26. [PMID: 7908929 DOI: 10.1080/09553009414550491] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We examined the degree of correlation between gamma-ray-induced lethality and DNA double-strand breaks (dsbs) after biologically relevant doses of radiation. Radiation lethality was modified by treating 14C-labelled Chinese hamster ovary cells with either of two aminothiols (WR-1065 or WR-25591) and the associated effect on dsb induction was determined by pulsed-field gel electrophoresis (PFGE). The use of phosphorimaging to analyse the distribution of 14C-activity in the gel greatly improved the low-dose resolution of the PFGE assay. Both WR-1065 and WR-25591 protected against dsb induction and lethality to a similar extent after low doses of radiation, although this correlation broke down when supralethal doses were used to induce dsbs. Thus, the level of dsbs induced in these cells as measured by PFGE after survival-curve doses of gamma-radiation is consistently predictive of the degree of lethality obtained, implying a case-effect relationship between these two parameters and confirming previous results obtained using the neutral filter elution assay for dsbs.
Collapse
Affiliation(s)
- D Murray
- Department of Radiobiology, Cross Cancer Institute, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
37
|
Chatterjee A, Raman MJ. Protective effect of cysteine against X-ray- and bleomycin-induced chromosomal aberrations and cell cycle delay. Mutat Res 1993; 290:231-8. [PMID: 7694114 DOI: 10.1016/0027-5107(93)90163-a] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The protective effect of cysteine was studied in muntjac and human lymphocytes in vitro scoring chromosomal aberrations in harlequin stained first cycle metaphases, induced by X-irradiation at G0. Its protective efficiency was also studied against the radiomimetic clastogen, bleomycin, in muntjac cells. 30 micrograms and 1 mg/ml of cysteine were given prior to 2, 3, and 4 Gy, and 2 mg/ml prior to only 4 Gy. 30 micrograms cysteine protected only against deletions in 4 Gy-treated cells while 1 mg protected against deletions by all three doses of X-rays. However, rearrangements were not reduced significantly in any of these, probably due to their low frequency. But when cysteine was increased to 2 mg, both types of aberrations were reduced significantly. This shows that a sufficient number of aberrations and an optimum concentration of the protector are essential for eliciting the best protective effect. This conclusion is further supported by the results of 2 mg cysteine treatment in human lymphocytes which yielded higher frequencies of rearrangements with 2 and 3 Gy X-rays than 4 Gy in muntjac, but had a relatively lower frequency of deletions. Thus the most abundant categories of aberration, i.e., deletions in muntjac and exchanges in humans, were reduced significantly by 2 mg cysteine, associated with a prominent reduction in the frequency of aberrant metaphases. Therefore, the differential protection observed with a low concentration of the protector and an insufficient yield of aberrations induced only indicates protection provided to the most frequent type of aberration by a protector when present in lower concentration. Cysteine pretreatment yielded weak protection against the effects of bleomycin, but posttreatment caused a mild potentiation of the clastogenic effect of BLM without altering the cell cycle kinetics. In this context, an action of cysteine as a reducing agent on BLM is suggested. Although cysteine alone caused severe retardation of the cell cycle, when given prior to X-irradiation, not only its delaying effect was not observed, but also it reduced the X-ray-induced cell cycle delay. This might be due to the oxidation of cysteine by its radical scavenging action.
Collapse
Affiliation(s)
- A Chatterjee
- Cytogenetics Laboratory, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
38
|
Carnes BA, Grdina DJ. In vivo protection by the aminothiol WR-2721 against neutron-induced carcinogenesis. Int J Radiat Biol 1992; 61:567-76. [PMID: 1349621 DOI: 10.1080/09553009214551381] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The ability of the compound S-2-(aminopropylamino)ethylphosphorothioic acid, designated WR-2721, to protect against neutron-induced carcinogenesis was investigated. Both sexes of the B6CF1 mouse were injected i.p. with 400 mg/kg of WR-2721 30 min prior to being irradiated by 10 cGy of neutrons. Neoplastic mortality in the groups receiving thiol was either reduced or delayed relative to irradiated mice not given protector. However, the time at which the protective effect of WR-2721 was expressed depended on the sex of the animal. Thiol-related shifts in the time of neoplastic death in females occurred only in the first half of the lifespan. Once a female survived to the mean age at death, no difference in the pattern of mortality could be detected between control and WR-2721-treated mice exposed to neutrons. Irrespective of thiol treatment, the timing of tumour-related death was nearly identical during the first half of life for males exposed to neutrons. In the last half of the lifespan, survival of thiol-protected males was enhanced relative to saline-injected males and even exceeded that observed in the control population.
Collapse
Affiliation(s)
- B A Carnes
- Biological and Medical Research Division, Argonne National Laboratory, IL 60439-4833
| | | |
Collapse
|
39
|
Grdina DJ, Garnes BA, Nagy B. Protection by WR-2721 and WR-151327 against late effects of gamma rays and neutrons. ADVANCES IN SPACE RESEARCH : THE OFFICIAL JOURNAL OF THE COMMITTEE ON SPACE RESEARCH (COSPAR) 1992; 12:257-263. [PMID: 11537016 DOI: 10.1016/0273-1177(92)90115-e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Two thiophosphoroate compounds WR-2721 and WR-151327 were assessed for their ability to modify the deleterious effects (life shortening and carcinogenesis) of fission-spectrum neutrons (kerma-weighted mean energy of 0.85 MeV) or gamma rays on B6CF1 hybrid mice. Male and female mice, 200 of each sex per experimental group, were irradiated individually at 110 days of age. Radioprotectors (400 mg/kg of WR-2721 or 580 mg/kg of WR-151327) were administered intraperitoneally 30 min prior to irradiation. Neutron doses were 10 cGy or 40 cGy and gamma ray doses were 206 cGy or 417 cGy. Animals were housed five to a cage; cage locations in the holding rooms were randomized by computer. Animals were checked daily and all deceased animals were necropsied. WR-2721 afforded protection against both neutron- and gamma-ray-induced carcinogenesis and subsequent life shortening. Cumulative survival curves for unirradiated mice of either sex were unaffectecd by protectors. WR-2721 protected irradiated groups against life shortening by approximately 10 cGy of neutrons or 100 cGy of gamma rays. WR-151327 was as effective as WR-2721 against neutron irradiation.
Collapse
Affiliation(s)
- D J Grdina
- Biological and Medical Research Division, Argonne National Laboratory, Argonne, Illinois 60439, USA
| | | | | |
Collapse
|
40
|
Sigdestad CP, Bergquist BL, Grdina DJ. The effect of chemical radiation protectors on cell cycle progression after gamma or neutron irradiation. Cell Prolif 1991; 24:271-80. [PMID: 2039803 DOI: 10.1111/j.1365-2184.1991.tb01156.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The effects of two chemical radiation protectors, WR-1065 and WR-151326, were characterized in V79 Chinese hamster cells after either cobalt-60 (60Co) gamma or fission spectrum neutron irradiation. Each protector was administered at a concentration of 4 mM to exponentially growing cultures for 30 min prior to and during irradiation with either 60Co gamma or JANUS fission spectrum neutrons. After irradiation the cells were either plated immediately for survival or returned to the incubator and assayed for cell progression. Aliquots of cells were removed at selected times, counted, fixed and stained with 4'6-diamidino-2-phenylindole (DAPI). Analysis of DNA histograms indicate that the presence of the protector during irradiation reduced the division delay experienced at the G2-M interface. Implications of these effects are discussed.
Collapse
Affiliation(s)
- C P Sigdestad
- Biological & Medical Research Division, Argonne National Laboratory, IL 60439
| | | | | |
Collapse
|
41
|
Murray D, Prager A, Vanankeren SC, Altschuler EM, Kerr MS, Terry NH, Milas L. Comparative effect of the thiols dithiothreitol, cysteamine and WR-151326 on survival and on the induction of DNA damage in cultured Chinese hamster ovary cells exposed to gamma-radiation. Int J Radiat Biol 1990; 58:71-91. [PMID: 1973441 DOI: 10.1080/09553009014551441] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We compared the ability of three thiols--dithiothreitol (DTT), cysteamine and WR-151326--to protect aerated Chinese hamster ovary cells from the lethal and DNA-damaging effects of gamma-radiation. These results were compared with earlier measurements for WR-1065 and WR-255591. The time-course and the concentration dependence of protection against cell killing was determined after 10 Gy of gamma-rays. The aminothiols cysteamine and WR-151326 protected at much lower extracellular concentrations than the simple thiol DTT; however, there was no clear difference between the behaviour of cysteamine, WR-151326, WR-1065 and WR-255591 in this respect. Protection by DTT and cysteamine was complete within 1 min, whereas for WR-151326, WR-1065 and WR-255591 about 30 min was required before protection began to reach a plateau. Based on these data, complete radiation survival curves were generated for each thiol and protection factors calculated. Effects on the induction of DNA single-strand breaks (ssb) and double-strand breaks (dsb) by gamma-rays were measured using alkaline (pH 12.1) and neutral (pH 7.0 and 9.6) elution, respectively. All three thiols protected against ssb induction, although to a significantly lower extent than against cell killing measured under identical conditions. Each thiol also protected against dsb induction. After high radiation doses the protection factors for dsb induction were also less than the protection factors for cell survival; however, when dsb were assayed using the low-dose replicate plating neutral elution method, the relative effect of each thiol on cell survival and on dsb induction appeared to be equivalent. The hierarchy of protection against both ssb and dsb induction (based on the extracellular thiol concentration required to produce a given degree of protection) was similar to that for cell survival, i.e., WR-151326 congruent to cysteamine less than DTT.
Collapse
Affiliation(s)
- D Murray
- Department of Experimental Radiotherapy, University of Texas, M.D. Anderson Cancer Center, Houston 77030
| | | | | | | | | | | | | |
Collapse
|
42
|
Maurizis JC, Madelmont JC, Godeneche D, Moreau MF, Oiry J, Imbach JL, Veyre A, Meyniel G. Metabolism of a new radioprotector; S-acetyl-N-glycyl cysteamine. II. Main tissue metabolites in mice bearing EMT6 tumours. Xenobiotica 1989; 19:833-42. [PMID: 2815826 DOI: 10.3109/00498258909043144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
1. The major tissue metabolites of the radioprotector S-acetyl-N-glycl cysteamine (I) labelled with 14C on the cysteamine group, were quantified and identified in normal tissues and EMT6 tumours implanted in mice, by chromatographic comparison with authentic reference compounds. 2. In all tissues the radioprotector undergoes rapid deacetylation and hydrolysis leading to the formation of cysteamine, which is the main metabolite involved in radioprotection. A major part of this metabolite is reversibly inactivated by binding to endogenous SH. 3. The differential radioprotection of healthy tissues versus EMT6 tumour is explained both by a lower uptake of radioprotector, and a weaker deacetylation and hydrolysis rate, in the tumour cells.
Collapse
|
43
|
Affiliation(s)
- D J Grdina
- Biological and Environmental Research Division, Argonne National Laboratory, Illinois 60439
| | | |
Collapse
|
44
|
Grdina DJ, Sigdestad CP, Dale PJ, Perrin JM. The effect of 2-[(aminopropyl)amino] ethanethiol on fission-neutron-induced DNA damage and repair. Br J Cancer 1989; 59:17-21. [PMID: 2667608 PMCID: PMC2246967 DOI: 10.1038/bjc.1989.5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The effect(s) of the radioprotector 2-[(aminopropyl)amino] ethanethiol (WR 1065) on fission-neutron-induced DNA damage and repair in V79 Chinese hamster cells was determined by using a neutral filter elution procedure (pH 7.2). When required, WR1065, at a final working concentration of 4 mM, was added to the culture medium, either 30 min before and during irradiation with fission spectrum neutrons (beam energy of 0.85 MeV) from the JANUS research reactor, or for selected intervals of time following exposure. The frequency of neutron-induced DNA strand breaks as measured by neutral elution as a function of dose equalled that observed for 60Co gamma-ray-induced damage (relative biological effectiveness of one). In contrast to the protective effect exhibited by WR1065 in reducing 60Co-induced DNA damage, WR1065 was ineffective in reducing or protecting against induction of DNA strand breaks by JANUS neutrons. The kinetics of DNA double-strand rejoining were measured following neutron irradiation. In the absence of WR1065, considerable DNA degradation by cellular enzymes was observed. This process was inhibited when WR1065 was present. These results indicate that, under the conditions used, the quality (i.e. nature), rather than quantity, of DNA lesions (measured by neutral elution) formed by neutrons was significantly different from that formed by gamma-rays.
Collapse
Affiliation(s)
- D J Grdina
- Biological, Environmental and Medical Research Division, Argonne National Laboratory, IL 60439-4833
| | | | | | | |
Collapse
|
45
|
Murray D, Altschuler EM, Kerr MS, Vanankeren SC, Meyn RE, Milas L. Radioprotection of cultured Chinese hamster ovary cells by WR-255591. Int J Radiat Biol 1988; 54:269-83. [PMID: 2900283 DOI: 10.1080/09553008814551701] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We examined the radioprotective effect of the aminothiol WR-255591 and its phosphorothioate derivative WR-3689 on aerated cultured Chinese hamster ovary cells. At concentrations up to 10 mmol dm-3, WR-3689 afforded little protection from the lethal effects of gamma-radiation. The free thiol WR-255591, on the other hand, efficiently protected these cells, giving a protection factor (PF) for cell survival of 2.3 at a concentration of 6 mmol dm-3. The effects of WR-255591 on the induction and rejoining of gamma-ray-induced DNA single-strand breaks (ssb) and double-strand breaks (dsb) were measured using alkaline (pH 12.1) and neutral (pH 7.0 or 9.6) elution, respectively. PFs calculated from these data were compared with the PFs measured for cell survival. WR-255591 (6 mmol dm-3) protected against the induction of both DNA ssb and dsb; however, the magnitude of the modification of both ssb (PF of 1.23) and dsb (PF of 1.83 at pH 7.0 and 1.70 at pH 9.6) was less than that for cell survival (PF of 2.3) measured under identical conditions (irradiation on ice). Treatment of cells with WR-255591 prior to irradiation retarded the subsequent rate of ssb rejoining but had no effect on dsb rejoining. Postirradiation treatment with the drug slightly retarded ssb rejoining but had no effect on cell survival. The observation of lower PFs for DNA strand breaks than for cell survival suggests that radioprotection by WR-255591 probably does not result from a uniform decrease in the induction of all types of DNA lesions. Rather, the drug may differentially protect against the induction of subclasses of DNA damage--which could also explain the effects on the kinetics of ssb rejoining--and/or enhance cellular recovery processes.
Collapse
Affiliation(s)
- D Murray
- Department of Experimental Radiotherapy, University of Texas System Cancer Center, M.D. Anderson Hospital and Tumor Institute, Houston 77030
| | | | | | | | | | | |
Collapse
|
46
|
Milas L, Murray D, Brock WA, Meyn RE. Radioprotectors in tumor radiotherapy: factors and settings determining therapeutic ratio. Pharmacol Ther 1988; 39:179-87. [PMID: 2849122 DOI: 10.1016/0163-7258(88)90059-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
WR-2721 and DDC have been used most frequently in our studies on radioprotective agents. WR-2721 was a much more potent radioprotector of murine normal tissues, both against early and late injuries of several organs and tissues, than was DDC. Protection factors for WR-2721 usually ranged between 1.5 and 2.5. Both agents protected solid murine tumors only minimally. While WR-2721 increased therapeutic ratios commonly, DDC did so only rarely. Micrometastatic foci were amenable to radioprotection more than established solitary tumors. Additional factors that influenced the degree of therapeutic benefit included dose of WR-2721, dose of irradiation (single versus fractionated), and time of WR-2721 administration in relation to radiation delivery. The ability of WR-2721 to prevent radiation-induced immunosuppression, metastatic spread, and carcinogenesis are additional benefits in the therapeutic use of this agent. Our current research on the improvement of radioprotectors for therapeutic use is focused on (a) a search for new radioprotective agents that are equal to or better than WR-2721 but less toxic and/or more specific for normal tissue, (b) understanding the basic mechanisms of action of these radioprotective agents at the molecular level, both in cells and tissues, and thus understanding the mechanisms leading to selective or preferential radioprotection of normal tissues, and (c) in vitro testing of primary human tumor cultures for their (non)susceptibility to radioprotection.
Collapse
Affiliation(s)
- L Milas
- Department of Experimental Radiotherapy, University of Texas System Cancer Center, M. D. Anderson Hospital and Tumor Institute, Houston 77030
| | | | | | | |
Collapse
|
47
|
Livesey JC, Reed DJ. Chemical Protection against Ionizing Radiation. ADVANCES IN RADIATION BIOLOGY 1987. [DOI: 10.1016/b978-0-12-035413-9.50010-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
48
|
Chatterjee A, Jacob-Raman M. Modifying effect of reduced glutathione on X-ray-induced chromosome aberration and cell-cycle delay in muntjac lymphocytes in vitro. ACTA ACUST UNITED AC 1986. [DOI: 10.1016/0165-7992(86)90128-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
49
|
Abstract
We examined the SH compounds glutathione (GSH), cysteine (CYS), cysteamine (MEA) and 2-mercaptoethanol (MET) with regard to their capacity for inducing SCEs. Whereas cysteamine and 2-mercaptoethanol increased the SCE frequency, this did not happen with glutathione and cysteine. By a comparative analysis of oxidized forms and structurally analogous compounds, it was confirmed that the induction of SCEs has no connection with the reducing property of the SH compounds. These findings are discussed in terms of other cytogenetic effects produced by SH compounds and the possible causes responsible for the induction of SCEs by SH compounds.
Collapse
|
50
|
Speit G, Wick C, Wolf M. Induction of sister chromatid exchanges by hydroxylamine, hydrazine and isoniazid and their inhibition by cysteine. Hum Genet 1980; 54:155-8. [PMID: 7390490 DOI: 10.1007/bf00278964] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Experiments were performed in order to gain information about the primary process leading to the production of sister chromatid exchanges (SCEs). Radical-forming substances (hydroxylamine, hydrazine and the antituberculous drug isoniazid) were examined for their effectiveness in inducing SCEs. All three substances proved successful in the induction of SCEs in the V-79 cell line of the Chinese hamster. By simultaneous application of a sulfhydryl compound (cysteine), a reduction of the hydrazine- and isoniazid-induced SCEs was achieved. Isoniazid was additionally examined in the in vivo SCE-test. At concentrations of 2-100 mg/kg body weight, it does not increase the rate of SCEs in the bone marrow of the Chinese hamster.
Collapse
|