Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Expert Opin Investig Drugs ; 32(1): 25-35, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36655861

ABSTRACT

INTRODUCTION: The possibility of exposure to high doses of total- or partial-body ionizing radiation at a high dose rate due to radiological/nuclear accidents or terrorist attacks is increasing. Despite research and development during the last six decades, there is a shortage of nontoxic, safe, and effective radiation medical countermeasures (MCMs) for radiological and nuclear emergencies. To date, the US Food and Drug Administration (US FDA) has approved only four agents for the mitigation of hematopoietic acute radiation syndrome (H-ARS). AREA COVERED: We present the current status of a promising radiation countermeasure, gamma-tocotrienol (GT3; a component of vitamin E) as a radiation MCM that has been investigated in murine and nonhuman primate models of H-ARS. There is significant work with this agent using various omic platforms during the last few years to identify its efficacy biomarkers. EXPERT OPINION: GT3 is a newer type of radioprotector having significant injury-countering potential and is currently under advanced development for H-ARS. As a pre-exposure drug, it requires only single doses, lacks significant toxicity, and has minimal, ambient temperature storage requirements; thus, GT3 appears to be an ideal MCM for military and first responders as well as for storage in the Strategic National Stockpile.


Subject(s)
Acute Radiation Syndrome , Medical Countermeasures , Radiation-Protective Agents , Humans , Mice , Animals , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/prevention & control , Radiation-Protective Agents/adverse effects , Vitamin E/adverse effects
2.
Radiat Prot Dosimetry ; 162(1-2): 83-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24993008

ABSTRACT

Tocols induce high levels of granulocyte-colony-stimulating factor (G-CSF). G-CSF mobilises progenitors that allow mice that have been severely immunocompromised by exposure to acute, high-dose ionising irradiation to recover and to survive. The neutralisation of G-CSF abrogates the radioprotective efficacy of tocols. This article reviews studies in which CD2F1 mice were irradiated with sufficiently high doses to cause acute radiation syndrome symptoms and then administered (iv) progenitor-enriched whole blood or peripheral blood mononuclear cells from tocol- and AMD3100-injected donor mice (AMD3100 is a chemokine receptor antagonist used to improve the yield of mobilised progenitors). In some experiments, G-CSF was neutralised completely. Irradiated recipient mice were observed for 30 d post-irradiation for survival, a primary endpoint used for determining therapeutic effectiveness. Additionally, potential tocol-induced biomarkers (cytokines, chemokines and growth factors) were quantified. The authors suggest that tocols are highly effective agents for mobilising progenitors with significant therapeutic potential.


Subject(s)
Gamma Rays/adverse effects , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cell Mobilization , Heterocyclic Compounds/pharmacology , Leukocytes, Mononuclear/transplantation , Radiation Injuries/prevention & control , Radiation-Protective Agents/pharmacology , Tocopherols/pharmacology , Animals , Anti-HIV Agents/pharmacology , Benzylamines , Cyclams , Male , Mice , Radiation Injuries/etiology
3.
Health Phys ; 106(6): 689-98, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24776901

ABSTRACT

The authors demonstrate the efficacy of a bridging therapy in a preclinical animal model that allows the lymphohematopoietic system of severely immunocompromised individuals exposed to acute, high-dose ionizing irradiation to recover and to survive. CD2F1 mice were irradiated acutely with high doses causing severe, potentially fatal hematopoietic or gastrointestinal injuries and then transfused intravenously with progenitor-enriched, whole blood, or peripheral blood mononuclear cells from mice injected with tocopherol succinate- and AMD3100- (a chemokine receptor anatogonist used to improve the yield of mobilized progenitors). Survival of these mice over a 30-d period was used as the primary measured endpoint of therapeutic effectiveness. The authors demonstrate that tocopherol succinate and AMD3100 mobilize progenitors into peripheral circulation and that the infusion of mobilized progenitor enriched blood or mononuclear cells acts as a bridging therapy for lymphohematopoietic system recovery in mice exposed to whole-body ionizing irradiation. The results demonstrate that infusion of whole blood or blood mononuclear cells from tocopherol succinate (TS)- and AMD3100-injected mice improved the survival of mice receiving high radiation doses significantly. The efficacy of TS-injected donor mice blood or mononuclear cells was comparable to that of blood or cells obtained from mice injected with granulocyte colony-stimulating factor. Donor origin-mobilized progenitors were found to localize in various tissues. The authors suggest that tocopherol succinate is an optimal agent for mobilizing progenitors with significant therapeutic potential. The extent of progenitor mobilization that tocopherol succinate elicits in experimental mice is comparable quantitatively to clinically used drugs such as granulocyte-colony stimulating factor and AMD3100. Therefore, it is proposed that tocopherol succinate be considered for further translational development and ultimately for use in humans.


Subject(s)
Emergency Responders , Mass Casualty Incidents , Occupational Exposure/prevention & control , Radiation-Protective Agents/pharmacology , Radioactive Hazard Release , alpha-Tocopherol/pharmacology , Animals , Dose-Response Relationship, Radiation , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/radiation effects , Male , Mice , Risk , Survival Analysis , Terrorism , Transplants
4.
Int J Radiat Biol ; 89(5): 334-45, 2013 May.
Article in English | MEDLINE | ID: mdl-23270472

ABSTRACT

PURPOSE: The objective of this study was to elucidate the action of α-tocopherol succinate (TS)- and AMD3100-mobilized progenitors in mitigating radiation-induced injuries. MATERIAL AND METHODS: CD2F1 mice were exposed to a high dose of radiation and then transfused intravenously with 5 million peripheral blood mononuclear cells (PBMC) from TS- and AMD3100-injected mice after irradiation. Intestinal and splenic tissues were harvested after irradiation and cells of those tissues were analyzed for markers of apoptosis and mitosis. Bacterial translocation from gut to heart, spleen, and liver in TS-treated and irradiated mice was evaluated by bacterial culture. RESULTS: We observed that the infusion of PBMC from TS- and AMD3100-injected mice significantly inhibited apoptosis, increased cell proliferation in the analyzed tissues of recipient mice, and inhibited bacterial translocation to various organs compared to mice receiving cells from vehicle-mobilized cells. This study further supports our contention that the infusion of TS-mobilized progenitor-containing PBMC acts as a bridging therapy by inhibiting radiation-induced apoptosis, enhancing cell proliferation, and inhibiting bacterial translocation in irradiated mice. CONCLUSIONS: We suggest that this novel bridging therapeutic approach that involves the infusion of TS-mobilized hematopoietic progenitors following acute radiation injury might be applicable to humans as well.


Subject(s)
Cell Movement/drug effects , Intestines/drug effects , Intestines/radiation effects , Radiation-Protective Agents/pharmacology , Stem Cells/cytology , Whole-Body Irradiation/adverse effects , alpha-Tocopherol/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Bacteria/drug effects , Bacteria/radiation effects , Cell Movement/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , DNA Breaks/drug effects , DNA Breaks/radiation effects , Endotoxins/blood , Gamma Rays/adverse effects , Intestines/cytology , Intestines/microbiology , Jejunum/cytology , Jejunum/drug effects , Jejunum/radiation effects , Male , Mice , Radiation Injuries/prevention & control , Spleen/cytology , Spleen/drug effects , Spleen/radiation effects , Stem Cells/drug effects , Stem Cells/radiation effects
5.
Exp Hematol ; 40(5): 407-17, 2012 May.
Article in English | MEDLINE | ID: mdl-22240608

ABSTRACT

The goal of this study was to elucidate the role of α-tocopherol succinate (TS)- and AMD3100-mobilized progenitors in mitigating the ionizing-radiation-induced gastrointestinal syndrome in mice. We demonstrate the efficacy of a bridging therapy that will allow the lymphohematopoietic system of severely immunocompromised victims exposed to ionizing radiation to recover from high doses of radiation. CD2F1 mice were irradiated with a high dose of radiation causing gastrointestinal syndrome (11 Gy, cobalt-60 γ-radiation) and then transfused intravenously (retro-orbital sinus) with whole blood or peripheral blood mononuclear cells (PBMC) from TS- and AMD3100-injected mice 2, 24, or 48 hours post irradiation and monitored for 30-day survival. Jejunum sections were analyzed for tissue area, surviving crypts, villi, mitotic figures, and basal lamina enterocytes. Our results demonstrate that infusion of whole blood or PBMC from TS- and AMD3100-injected mice significantly improved survival of mice receiving a high dose of radiation. Histopathology and immunostaining of jejunum from irradiated and TS- and AMD3100-mobilized PBMC-transfused mice reveal significant protection of gastrointestinal tissue from radiation injury. We demonstrate that TS and AMD3100 mobilize progenitors into peripheral circulation and that the infusion of mobilized progenitor-containing blood or PBMC acts as a bridging therapy for immune-system recovery in mice exposed to high, potentially fatal, doses of ionizing radiation.


Subject(s)
Antioxidants/therapeutic use , Blood Cells/transplantation , Gastrointestinal Diseases/etiology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Heterocyclic Compounds/therapeutic use , Leukocytes, Mononuclear/transplantation , Radiation Injuries/surgery , Radiation-Protective Agents/therapeutic use , alpha-Tocopherol/therapeutic use , Animals , Antioxidants/pharmacology , Benzylamines , Cyclams , Filgrastim , Gamma Rays/adverse effects , Gastrointestinal Diseases/surgery , Granulocyte Colony-Stimulating Factor/therapeutic use , Heterocyclic Compounds/pharmacology , Intestinal Mucosa/radiation effects , Intestinal Mucosa/ultrastructure , Jejunum/pathology , Jejunum/radiation effects , Male , Mice , Radiation Chimera , Radiation-Protective Agents/pharmacology , Recombinant Proteins/therapeutic use , alpha-Tocopherol/pharmacology
6.
Health Phys ; 89(5): 531-45, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16217197

ABSTRACT

In today's heightened nuclear/biological/chemical threat environment, there is an increased need to have safe and effective means to protect not only special high-risk service groups, but also the general population at large, from the health hazards of unintended ionizing radiation exposures. An unfulfilled dream has been to have a globally effective pharmacologic that could be easily taken orally without any undue side effects prior to a suspected or impending nuclear/radiological event; such an ideal radioprotective agent has yet to be identified, let alone fully developed and approved for human use. No one would argue against the fact that this is problematic and needs to be corrected, but where might the ultimate solution to this difficult problem be found? Without question, representative species of the aminothiol family [e.g., Amifostine (MedImmune, Gaithersburg, Maryland)] have proven to be potent cytoprotectants for normal tissues subjected to irradiation or to radiomimetic chemicals. Although Amifostine is currently used clinically, drug toxicity, limited times of protection, and unfavorable routes of administration, all serve to limit the drug's utility in nonclinical settings. A full range of research and development strategies is being employed currently in the hunt for new safe and effective radioprotectants. These include: (1) large scale screening of new chemical classes or natural products; (2) restructuring/reformulating older protectants with proven efficacies but unwanted toxicities; (3) using nutraceuticals that are only moderately protective but are essentially nontoxic; (4) using low dose combinations of potentially toxic but efficacious agents that protect through different routes to foster radioprotective synergy; and (5) accepting a lower level of drug efficacy in lieu of reduced toxicity, banking on the premise that the protection afforded can be leveraged by post-exposure therapies. Although it is difficult to predict which of these strategies will ultimately prove to be successful, it is certain that the probability of a useful protectant being fielded is increased significantly. This is due to the resurgence of interest in radiation protection, increased resources being expended by federal agencies, and by the Food and Drug Administration's willingness to innovate relative to new approval guidance.


Subject(s)
Radiation Protection , Radiation-Protective Agents/pharmacology , Combinatorial Chemistry Techniques , Dietary Supplements , Electromagnetic Fields , Gene Transfer, Horizontal , Genetic Engineering , Humans , Radiation Injuries/prevention & control , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/adverse effects
7.
Peptides ; 26(3): 413-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15652647

ABSTRACT

Interleukin-1beta (IL-1beta) is a cytokine involved in homeostatic processes of the immune system and specifically in inflammatory reactions. The nonapeptide of human IL-1beta (VQGEESNDK, position 163-171) has been shown to retain adjuvant and immunostimulatory activities of the native molecule without any inflammatory and pyrogenic properties. A lipophilic derivative of IL-1beta nonapeptide having a palmitoyl residue at the amino terminus was synthesized in order to determine the effects of such structural modification on its bioactivities. The structurally modified peptide derivative, palmitoylated peptide, significantly protected C3H/HeN mice against potentially lethal doses of ionizing radiation. The dose reduction factor was found to be 1.07. Hematological studies show improved recovery of red blood cells and platelets in irradiated and palmitoylated peptide treated mice as compared with the untreated and irradiated group. These results suggest the importance of the derivatization of small peptides of radioprotective, but toxic cytokines in order to enhance radioprotective activity while reducing unwanted toxic side effects.


Subject(s)
Interleukin-1/chemistry , Palmitic Acid/metabolism , Peptides/chemistry , Radiation-Protective Agents/pharmacology , Adjuvants, Immunologic , Animals , Blood Platelets/drug effects , Blood Platelets/radiation effects , Cytokines/chemistry , Dose-Response Relationship, Radiation , Erythrocytes/drug effects , Erythrocytes/radiation effects , Humans , Inflammation , Interleukin-1/pharmacology , Male , Mice , Mice, Inbred C3H , Palmitic Acid/chemistry , Protein Structure, Tertiary , Radiation, Ionizing , Radiation-Protective Agents/chemistry , Time Factors
8.
Mil Med ; 167(2 Suppl): 57-9, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11873518

ABSTRACT

Low-level radiation injury is dependent on the radiation dose and dose rate. The major military use of any potential radioprotectant is to prevent the short-term effects of lethality and the long-term effects of cancer and other pathologies from radiation exposure that may occur in a nuclear battlefield or in a nuclear material contaminated field of operation. Therefore, a radioprotectant should not affect the ability of military personnel to perform tasks. Because exposure to ionizing radiation induces free radical species, effective antioxidants, either alone or in combination with other agents, can be used as potential radioprotectors. To test this hypothesis, we studied vitamin E for its radioprotective efficacy. Using CD2F1 male mice as the model system, we observed that vitamin E at a dose of 400 IU/kg acts as a good radioprotectant against lethal doses of cobalt-60 radiation. Vitamin E was more efficacious when given subcutaneously than when given orally.


Subject(s)
Radiation-Protective Agents/therapeutic use , Vitamin E/therapeutic use , Animals , Cobalt Radioisotopes , Humans , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL