Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Health Phys ; 121(4): 304-330, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34546214

ABSTRACT

ABSTRACT: Medical countermeasure development under the US Food and Drug Administration animal rule requires validated animal models of acute radiation effects. The key large animal model is the non-human primate, rhesus macaque. To date, only the rhesus macaque has been used for both critical supportive data and pivotal efficacy trials seeking US Food and Drug Administration approval. The potential for use of the rhesus for other high priority studies such as vaccine development underscores the need to identify another non-human primate model to account for the current lack of rhesus for medical countermeasure development. The cynomolgus macaque, Macaca fascicularis, has an existing database of medical countermeasure development against the hematopoietic acute radiation syndrome, as well as the use of radiation exposure protocols that mimic the likely nonuniform and heterogenous exposure consequent to a nuclear terrorist event. The review herein describes published studies of adult male cynomolgus macaques that used two exposure protocols-unilateral, nonuniform total-body irradiation and partial-body irradiation with bone marrow sparing-with the administration of subject-based medical management to assess mitigation against the hematopoietic acute radiation syndrome. These studies assessed the efficacy of cytokine combinations and cell-based therapy to mitigate acute radiation-induced myelosuppression. Both therapeutics were shown to mitigate the myelosuppression of the hematopoietic acute radiation syndrome. Additional studies being presented herein further defined the dose-dependent hematopoietic acute radiation syndrome of cynomolgus and rhesus macaques and a differential dose-dependent effect with young male and female cynomolgus macaques. The database supports the investigation of the cynomolgus macaque as a comparable non-human primate for efficacy testing under the US Food and Drug Administration animal rule. Critical gaps in knowledge required to validate the models and exposure protocols are also identified.


Subject(s)
Acute Radiation Syndrome , Medical Countermeasures , Radiation Exposure , Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/prevention & control , Animals , Female , Macaca fascicularis , Macaca mulatta , Male , Radiation Exposure/adverse effects
2.
Metabolites ; 11(8)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34436481

ABSTRACT

The acute radiation syndrome is defined in large part by radiation injury in the hematopoietic and gastrointestinal (GI) systems. To identify new pathways involved in radiation-induced GI injury, this study assessed dose- and time-dependent changes in plasma metabolites in a nonhuman primate model of whole abdominal irradiation. Male and female adult Rhesus monkeys were exposed to 6 MV photons to the abdomen at doses ranging between 8 and 14 Gy. At time points from 1 to 60 days after irradiation, plasma samples were collected and subjected to untargeted metabolomics. With the limited sample size of females, different discovery times after irradiation between males and females were observed in metabolomics pattern. Detailed analyses are restricted to only males for the discovery power. Radiation caused an increase in fatty acid oxidation and circulating levels of corticosteroids which may be an indication of physiological stress, and amino acids, indicative of a cellular repair response. The largest changes were observed at days 9 and 10 post-irradiation, with most returning to baseline at day 30. In addition, dysregulated metabolites involved in amino acid pathways, which might indicate changes in the microbiome, were detected. In conclusion, abdominal irradiation in a nonhuman primate model caused a plasma metabolome profile indicative of GI injury. These results point to pathways that may be targeted for intervention or used as early indicators of GI radiation injury. Moreover, our results suggest that effects are sex-specific and that interventions may need to be tailored accordingly.

3.
Int J Radiat Biol ; 97(sup1): S63-S72, 2021.
Article in English | MEDLINE | ID: mdl-32924712

ABSTRACT

PURPOSE: Total body irradiation of the Gottingen minipig results in a characteristic hematopoietic response, including anemia, neutropenia, lymphocytopenia, and thrombocytopenia. Currently, there are no well-characterized large or small animal models for radiation-induced thrombocytopenia. The study described here using the Gottingen minipig was focused on understanding which aspects of the coagulation cascade leads to radiation-induced coagulopathy. In this study, multiple clinical pathology parameters were determined prior to and for 45-days following total body irradiation using a 6 MV photon linear accelerator. MATERIALS AND METHODS: Following irradiation, frequent analyses of conventional hematology and coagulation parameters provided time-course information on the onset and recovery of thrombocytopenia. In addition, thromboelastography (TEG) was utilized to monitor coagulation dysfunction, namely clotting time, clot formation time, clot strength, and fibrinolysis. Coagulation factor activity levels were measured for factors II, V, VII, VIII, IX, X, XI, XII, XIII, Protein C, fibrin monomers, antiplasmin and D-dimer using a Siemen's coagulation analyzer to provide time course information of changes in activity post irradiation exposure. RESULTS: These analyses revealed that in total body irradiated minipigs, TEG tracings demonstrate long R (time to initial clot formation) and K (time to achieve a certain clot strength) times, and low alpha-angle (rate of clot formation) and MA (overall stability of the clot) during onset of thrombocytopenia (typically post irradiation day 10-15). Low alpha-angle and MA directly correlated with decreased platelet counts. A long R time is suggestive of a deficiency in clotting factors and was compared to measured activity levels of individual coagulation factors. The data indicates that coagulation factors are significantly changed early after irradiation exposure prior to thrombocytopenia and factors VIII, XI, XII and XIII are markedly altered during the critical point of thrombocytopenia. CONCLUSION: These data support the continued use of multiple approaches to evaluate the coagulation cascade in order to provide the most meaningful interpretation of the hematopoietic changes that occur post irradiation.


Subject(s)
Blood Coagulation Disorders , Thrombocytopenia , Animals , Blood Coagulation Factors/metabolism , Swine , Swine, Miniature/metabolism , Thrombelastography/methods , Thrombocytopenia/etiology
4.
Int J Radiat Biol ; 97(sup1): S88-S99, 2021.
Article in English | MEDLINE | ID: mdl-32909856

ABSTRACT

PURPOSE: Well-characterized animal models that mimic the human response to potentially lethal doses of radiation are necessary in order to assess the efficacy of candidate medical countermeasures under the criteria of the U.S. Food and Drug Administration 'Animal Rule'. Development of a model requires the determination of the radiation dose response relationship and time course of mortality and morbidity under scenarios likely to be present in the human population during mass casualty situations. These scenarios include understanding the impact of medical management on survival of the hematopoietic acute radiation syndrome (H-ARS). Little information is available to compare the impact of medical management under identical study conditions. The work presented here provides a comparison of the impact of different levels of medical management (supportive care) on the survival outcome in two large animal models: the male Gottingen minipig and the male rhesus macaque (NHP). MATERIALS AND METHODS: In the context of this comparison, limited supportive care consisted of administration of analgesics only, standard supportive care consisted of prophylactic administration of analgesics, antibiotics and fluids (minipigs) or analgesics, antibiotics, antidiarrheals, nutritional and fluid support (NHP) on a set schedule regardless of indication, and full supportive care (NHP only) consisted of analgesics, antibiotics, antidiarrheals, nutritional and fluid support, antiemetics and blood transfusions on an individual animal, trigger-to-treat regimen. Regardless of level of supportive care, minipigs were exposed to total body irradiation using a Co60 source and NHPs were exposed to total body irradiation using 6 MV photon energy. RESULTS: Based on estimated LD50 values, the inclusion of antimicrobial or broad-spectrum antibiotics provided a dose modifying factor (DMF) of 1.09 in the minipig, and by 1.15 in the NHP (standard supportive care to limited supportive care ratio. For the NHP, the administration of supportive care based on symptomology rather than a set schedule, and inclusion of blood transfusions yielded a DMF of 1.05 (full supportive care to standard supportive care ratio). Conversely, comparison of the estimated LD50 values between full supportive care and limited supportive care in the NHP provided a DMF of 1.21. CONCLUSION: The study reported here provides a comparison of the impact of antibiotic administration on radiation-induced lethality.


Subject(s)
Acute Radiation Syndrome , Whole-Body Irradiation , Acute Radiation Syndrome/therapy , Animals , Anti-Bacterial Agents , Antidiarrheals , Disease Models, Animal , Macaca mulatta , Male , Models, Animal , Swine , Swine, Miniature , Whole-Body Irradiation/adverse effects
5.
Health Phys ; 116(3): 354-365, 2019 03.
Article in English | MEDLINE | ID: mdl-30688697

ABSTRACT

Radiation-induced lung injury is a characteristic, dose- and time-dependent sequela of potentially lethal, delayed effects of acute radiation exposure. Understanding of these delayed effects to include development of medical countermeasures requires well-characterized and validated animal models that mimic the human response to acute radiation and adhere to the criteria of the US Food and Drug Administration Animal Rule. The objective herein was to establish a nonhuman primate model of whole-thorax lung irradiation in female rhesus macaques. Definition of the dose-response relationship to include key signs of morbidity and mortality in the female macaque served to independently validate the recent model performed with male macaques and importantly, to establish the lack of sex and institutional bias across the dose-response relationship for radiation-induced lung injury. The study design was similar to that described previously, with the exception that female rhesus macaques were utilized. In brief, a computed tomography scan was conducted prior to irradiation and used for treatment planning. Animals in 5 cohorts (n = 8 per cohort) were exposed to a single 6-MV photon exposure focused on the lung as determined by the computed tomography scan and treatment planning at a dose of 9.5, 10, 10.5, 11, or 11.5 Gy. Subject-based supportive care, including administration of dexamethasone, was based on trigger-to-treat criteria. Clearly defined euthanasia criteria were used to determine a moribund condition over the 180-day study duration post-whole-thorax lung irradiation. Percent mortality per radiation dose was 12.5% at 9.5 Gy, 25% at 10 Gy, 62.5% at 10.5 Gy, 87.5% at 11 Gy, and 100% at 11.5 Gy. The resulting probit plot for the whole-thorax lung irradiation model estimated an LD50/180 of 10.28 Gy, which was not significantly different from the published estimate of 10.27 Gy for the male rhesus. The key parameters of morbidity and mortality support the conclusion that there is an absence of a sex influence on the radiation dose-response relationship for whole-thorax lung irradiation in the rhesus macaque. This work also provides a significant interlaboratory validation of the previously published model.


Subject(s)
Lung Injury/etiology , Radiation Injuries, Experimental/etiology , Animals , Dose-Response Relationship, Radiation , Female , Lung/diagnostic imaging , Lung/pathology , Lung/radiation effects , Lung Injury/diagnostic imaging , Lung Injury/mortality , Lung Injury/pathology , Macaca mulatta , Male , Radiation Injuries, Experimental/diagnostic imaging , Radiation Injuries, Experimental/mortality , Sex Factors , Tomography, X-Ray Computed
7.
Health Phys ; 109(5): 502-10, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26425909

ABSTRACT

The Medical Countermeasures against Radiological Threats (MCART) consortium has established a dose response relationship for the hematopoietic acute radiation syndrome (HARS) in the rhesus macaque conducted under an individualized supportive care protocol, including blood transfusions. Application of this animal model as a platform for demonstrating efficacy of candidate medical countermeasures is significantly strengthened when the model is independently validated at multiple institutions. The study reported here describes implementation of standard operating procedures at an institute outside the consortium in order to evaluate the ability to establish an equivalent radiation dose response relationship in a selected species. Validation of the animal model is a significant component for consideration of the model protocol as an FDA-recommended drug development tool in the context of the "Animal Rule." In the current study, 48 male rhesus macaques (4-8 kg) were exposed to total-body irradiation (TBI) using 6 MV photon energy at a dose rate of approximately 0.8 Gy min. Results show that onset and duration of the hematological response, including anemia, neutropenia, and thrombocytopenia, following TBI ranging from 6.25 to 8.75 Gy correlate well with previously reported findings. The lethality values at 60 d following TBI were estimated to be 6.88 Gy (LD30/60), 7.43 Gy (LD50/60), and 7.98 Gy (LD70/60). These values are equivalent to those published previously of 7.06 Gy (LD30/60), 7.52 Gy (LD50/60), and 7.99 Gy (LD70/60); the DRR slope (p = 0.68) and y-intercepts show agreement along the complete dose range for HARS. The ability to replicate the previously established institutional lethality profile (PROBIT) and model outcomes through careful implementation of defined procedures is a testament to the robustness of the model and highlights the need for consistency in procedures.


Subject(s)
Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/physiopathology , Disease Models, Animal , Dose-Response Relationship, Radiation , Whole-Body Irradiation/adverse effects , Whole-Body Irradiation/standards , Acute Radiation Syndrome/diagnosis , Animals , Laboratories , Macaca mulatta , Male , Reproducibility of Results , Sensitivity and Specificity , United States
8.
Health Phys ; 108(5): 538-46, 2015 May.
Article in English | MEDLINE | ID: mdl-25811151

ABSTRACT

There is widespread interest in the development of tools to estimate radiation exposures. Exhaled breath provides a novel matrix for assessing biomarkers that could be correlated with exposures. The use of exhaled breath for estimating radiation exposure is warranted, as studies have shown that external exposure to ionizing radiation causes oxidative stress that accelerates lipid peroxidation of polyunsaturated fatty acids, liberating alkanes and alkane metabolites that are excreted in the breath as volatile organic compounds (VOCs). As a proof of principle study, small groups (n = 4) of Göttingen minipigs were whole-body irradiated with gamma rays delivered by a 60Co source at absorbed doses of 0, 0.25, 0.5, 0.75, 1, 1.25, 2, and 4 Gy. Additional groups (n = 4) were treated with lipopolysaccharide (LPS) or granulocyte colony stimulating factor (G-CSF), with and without concurrent 60Co exposure, at an absorbed dose of 1 Gy. Breath and background air VOC samples were collected on days -3, -2, -1, 0 pre-irradiation, then at 0.25, 24, 48, 72, and 168 h post-irradiation. VOCs were analyzed by automated thermal desorption with two-dimensional gas chromatography and time-of-flight mass spectrometry (ATD GCxGC TOF MS). The results show significant changes in 58 breath VOCs post-irradiation, mainly consisting of methylated and other derivatives of alkanes, alkenes, and benzene. Using a multivariate combination of these VOCs, a radiation response function was constructed, which was significantly elevated at 15 min post irradiation and remained elevated throughout the study (to 168 h post irradiation). As a binary test of radiation absorbed doses ≥ 0.25 Gy, the radiation response function distinguished irradiated animals from shams (0 Gy) with 83-84% accuracy. A randomly derived radiation response function was robust: When half of the biomarkers were removed, accuracy was 75%. An optimally derived function with two biomarkers was 82% accurate. As a binary test of radiation absorbed doses ≥ 0.5 Gy, the radiation response function identified irradiated animals with an accuracy of 87% at 15 min post irradiation and 75.5% at 168 h post irradiation. Treatment with LPS and G-CSF did not affect the radiation response function. This proof-of-principle study supports the hypothesis that breath VOCs may be used for estimating radiation exposures. Further studies will be required to validate the sensitivity and specificity of these potential biomarkers.


Subject(s)
Breath Tests , Volatile Organic Compounds/analysis , Whole-Body Irradiation , Animals , Biomarkers/analysis , Gamma Rays , Male , Radiometry , Swine , Swine, Miniature
9.
Health Phys ; 105(3): 245-252, 2013 Sep.
Article in English | MEDLINE | ID: mdl-30522248

ABSTRACT

There is a great deal of interest in the establishment of a standardized animal model for the acute radiation syndrome to allow development of diagnostic approaches and countermeasure treatments following radiological terrorist events. Due to physiological, anatomical, and biochemical similarities to humans, the minipig is an attractive large animal model for evaluating countermeasure efficacy. This study was conducted in order to aid in the establishment of the minipig, and the Göttingen minipig in particular, as an animal model for the hematopoietic acute radiation syndrome. Animals were exposed whole-body to Co at doses of 0 (sham control), 0.25, 0.5, 0.75, 1.0, and 2.0 Gy, and hematological parameters followed in time from pre-irradiation to post-irradiation Day 7. Following irradiation, a dose-dependent decrease in total white blood cells was observed, which was determined to be statistically different as compared to control animals at all dose levels above 0.25 Gy at 24 h post-irradiation. Similarly, a dose-dependent reduction in both absolute lymphocyte count and absolute neutrophil count occurred by the earliest time point measured for all exposed animals. A significant decrease in platelets was observed at post-irradiation Day 7 in animals exposed only at the highest (2.0 Gy) level. The platelet-to-lymphocyte ratio generated for exposures ranging from 0.25-2.0 Gy was able to differentiate response between high and low exposure levels even at 7 d post exposure. In conclusion, the present study supports the development of the Göttingen minipig as a suitable large animal model to study radiation-induced hematopoietic syndrome.

10.
Health Phys ; 101(2): 118-27, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21709497

ABSTRACT

Cuprimine® and Syprine® are therapeutics approved by the USFDA to treat copper overload in Wilson Disease (a genetic defect in copper transport) by chelation and accelerated excretion of internally-deposited copper. These oral therapeutics are based on the respective active ingredients D-penicillamine (DPA) and N,N'-bis (2-aminoethyl) -1,2-ethanediamine dihydrochloride (Trien). Cuprimine is considered the primary treatment, although physicians are increasingly turning to Syprine as a first-line therapy. Both drugs exhibit oral systemic activity and low toxicity; their biological effects and safety are established. Previous in vivo studies using a rodent animal model established the decorporation potential of Cuprimine and Syprine for (60)Co and (210)Po. Currently these studies are being expanded to evaluate the in vivo decorporation efficacy of these drugs for several additional radionuclides. In this report, results of this investigation are discussed using the radionuclides (137)Cs, (60)Co, (192)Ir and (85)Sr. Short-term 48-h pilot studies were undertaken to evaluate DPA and Trien for their in vivo decorporation potential using male Wistar-Han rats. In these studies, a radionuclide solution was administered to the animals by intravenous (IV) injection, followed by a single IV dose of either DPA or Trien. Control animals received the radionuclide alone. Results show effective decorporation of (60)Co by DPA within the time frame evaluated. DPA and Trien were also modestly effective in decorporation of (137)Cs and (85)Sr, respectively. The study did not find DPA or Trien effective for decorporation of (192)Ir. Based on these encouraging findings, further studies to evaluate the dose-response profiles and timing of the chelator administration post exposure to radionuclides are warranted.


Subject(s)
Radiation Monitoring/methods , Radioisotopes/toxicity , Animals , Cesium/administration & dosage , Cesium/pharmacokinetics , Cesium/toxicity , Cobalt/administration & dosage , Cobalt/pharmacokinetics , Cobalt/toxicity , Injections, Intraventricular , Iridium/administration & dosage , Iridium/pharmacokinetics , Iridium/toxicity , Male , Pilot Projects , Radioisotopes/administration & dosage , Radioisotopes/pharmacokinetics , Rats , Rats, Wistar , Risk Assessment/methods , Strontium/administration & dosage , Strontium/pharmacokinetics , Strontium/toxicity , Tissue Distribution
11.
Health Phys ; 99(3): 394-400, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20699703

ABSTRACT

Although four stable isotopes of strontium occur naturally, Sr is produced by nuclear fission and is present in surface soil around the world as a result of fallout from atmospheric nuclear weapons tests. It can easily transfer to humans in the event of a nuclear/radiological emergency or through the plant-animal-human food chain causing long-term exposures. Strontium is chemically and biologically similar to calcium, and is incorporated primarily into bone following internal deposition. Alginic acid (alginate) obtained from seaweed (kelp) extract selectively binds ingested strontium in the gastrointestinal tract blocking its systemic uptake and reducing distribution to bone in rats, while other natural polysaccharides including chitosan and hyaluronic acid had little in vivo affinity for strontium. Alginate exhibits the unique ability to discriminate between strontium and calcium and has been previously shown to reduce intestinal absorption and skeletal retention of strontium without changing calcium metabolism. In our studies, the effect of commercially available alginate on intestinal absorption of strontium was examined. One problem associated with alginate treatment is its limited solubility and gel formation in water. The aqueous solubility of sodium alginate was improved in a sodium chloride/sodium bicarbonate electrolyte solution containing low molecular weight polyethylene glycol (PEG). Furthermore, oral administration of the combined alginate/electrolyte/PEG solution accelerated removal of internal strontium in rats when compared to treatment with individual sodium alginate/electrolyte or electrolyte/PEG solutions. Importantly, both alginate and PEG are nontoxic, readily available materials that can be easily administered orally in case of a national emergency when potentially large numbers of the population may require medical treatment for internal depositions. Our results suggest further studies to optimize in vivo decorporation performance of engineered alginate material via modification of its chemical and physicochemical properties are warranted.


Subject(s)
Biocompatible Materials/pharmacology , Calcium Isotopes/toxicity , Food Contamination, Radioactive , Intestinal Absorption/drug effects , Intestinal Absorption/radiation effects , Strontium Radioisotopes/toxicity , Administration, Oral , Alginates , Animals , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Calcium Isotopes/administration & dosage , Calcium Isotopes/metabolism , Chelating Agents/administration & dosage , Chelating Agents/chemistry , Chelating Agents/pharmacology , Chitosan/metabolism , Electrolytes , Glucuronic Acid , Hexuronic Acids , Hyaluronic Acid/metabolism , Injections, Intravenous , Intestinal Absorption/physiology , Molecular Weight , Muscle, Skeletal/metabolism , Polyethylene Glycols , Rats , Solubility , Strontium Radioisotopes/administration & dosage , Strontium Radioisotopes/metabolism , Time Factors
13.
Health Phys ; 98(3): 471-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20147789

ABSTRACT

The acknowledged risk of deliberate release of radionuclides into local environments by terrorist activities has prompted a drive to improve novel materials and methods for removing internally deposited radionuclides. These decorporation treatments will also benefit workers in the nuclear industry, should an exposure occur. Cuprimine and Syprine are oral therapeutics based on the active ingredients D-penicillamine and N,N'-bis-(2-aminoethyl)-1,2-ethanediamine dihydrochloride, respectively. These therapeutic drugs have been used for several decades to treat Wilson's disease, a genetic defect leading to copper overload, by chelation and accelerated excretion of internally deposited copper. Studies were undertaken to evaluate these FDA-approved drugs for the in vivo decorporation of radioactive cobalt (Co) and polonium (Po) using male Wistar-Han rats. In these studies, Co or Po was administered to animals by IV injection, followed by oral gavage doses of either Cuprimine or Syprine. Control animals received the radionuclide alone. For Co studies, animals received a single dose of Cuprimine or Syprine, while for Po studies animals were repeatedly dosed at 24-h intervals for a total of 5 doses. Results show that Syprine significantly increased urinary elimination and skeletal concentrations of Co compared to controls. While Cuprimine had little effect on total excretion of Co, the skeletal, kidney, liver, muscle, and stomach tissues had significantly lower radioactivity compared to control animals. The low overall excretion of Po made it difficult to reliably measure urinary or fecal radioactivity and draw a definitive conclusion on the effect of Cuprimine or Syprine treatment on excretion. However, Cuprimine treatment was effective at reducing spleen levels of Po compared to controls. Similarly, Syprine treatment produced statistically significant reductions of Po in the spleen and skeletal tissues compared to control animals. Based on these promising findings, further studies to evaluate the dose-response pharmacokinetic profiles for decorporation are warranted.


Subject(s)
Cobalt Radioisotopes/isolation & purification , Penicillamine/chemistry , Penicillamine/pharmacology , Polonium/chemistry , Polonium/isolation & purification , Trientine/chemistry , Trientine/pharmacology , Animals , Chelating Agents/administration & dosage , Chelating Agents/chemistry , Chelating Agents/pharmacology , Cobalt Radioisotopes/chemistry , Cobalt Radioisotopes/pharmacokinetics , Humans , Male , Penicillamine/administration & dosage , Polonium/pharmacokinetics , Rats , Rats, Wistar , Tissue Distribution , Trientine/administration & dosage
14.
Health Phys ; 98(1): 53-60, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19959951

ABSTRACT

This report provides a comparison of the oral decorporation efficacy of L-glutathione (GSH), L-cysteine (Cys), and a liposomal GSH formulation (ReadiSorb) toward systemic (60)Co to that observed following intravenous administration of GSH and Cys in F344 rats. Aminoacid L-histidine (His) containing no thiol functionality was tested intravenously to compare in vivo efficacy of the aminothiol (GSH, Cys) chelators with that of the aminoimidazole (His) chelator. In these studies, (60)Co was administered to animals by intravenous injection, followed by intravenous or oral gavage doses of a chelator repeated at 24-h intervals for a total of 5 doses. The results suggest that GSH and Cys are potent decorporation agents for (60)Co in the rat model, although the efficacy of treatment depends largely on the systemic availability of the chelator. The intravenous route of administration of GSH or Cys was most effective in reducing tissue (60)Co levels and in increasing excretion of radioactivity compared to control animals. Liposomal encapsulation was found to markedly enhance the oral bioavailability of GSH compared to non-formulated GSH. The oral administration of liposomal GSH reduced (60)Co levels in nearly all tissues by 12-43% compared to that observed for non-formulated GSH. Efficacy of oral Cys was only slightly reduced in comparison with intravenous Cys. Further studies to optimize the dosing regimen in order to maximize decorporation efficiency are warranted.


Subject(s)
Cobalt Radioisotopes/pharmacokinetics , Cobalt Radioisotopes/toxicity , Cysteine/administration & dosage , Glutathione/administration & dosage , Administration, Oral , Animals , Antidotes/administration & dosage , Chelating Agents/administration & dosage , Chelating Agents/metabolism , Cobalt Radioisotopes/administration & dosage , Cysteine/metabolism , Glutathione/metabolism , Histidine/metabolism , Injections, Intravenous , Liposomes , Male , Metabolic Clearance Rate/drug effects , Rats , Rats, Inbred F344 , Receptors, Cell Surface/metabolism , Terrorism , Tissue Distribution
15.
Health Phys ; 97(2): 115-24, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19590271

ABSTRACT

With the increased threat of terrorist release of radioactive materials, there is a need for non-toxic decorporation agents to treat internal contamination with radionuclides. In this study, low molecular weight chitosan was evaluated for decorporation of radioactive cobalt (60Co). The affinity of chitosan for Co(II) was tested in vitro using spectrophotometric and potentiometric titration techniques. For in vivo studies, the effect of chitosan on ingested 60Co was evaluated using F344 rats administered a single dose followed by oral chitosan. Chitosan was also evaluated for systemic decorporation of 60Co following intravenous injection with repeated chitosan administration over 5 d. Control animals received 60Co without chelation treatment. Excreta and tissues were collected for analysis using gamma-counting techniques. Results from in vitro experiments confirmed the binding of Co(II) to chitosan, with the postulated formation of a mixed cobalt-chitosan-hydroxide complex species; a stability constant was calculated for this complex. For in vivo studies, oral administration of chitosan significantly reduced systemic absorption of orally administered 60Co as evidenced by an increase in fecal elimination and decrease in urinary elimination. However, oral administration of chitosan lactate slightly decreased fecal excretion of 60Co. Further, oral administration of chitosan significantly reduced 60Co levels in kidney, liver, and skeleton compared to control animals receiving 60Co alone. By the i.v. route, chitosan slightly reduced levels of 60Co in tissues compared to controls, although statistically significant reductions were only observed for blood and kidney. Overall, this commercially available chitosan oligosaccharide exhibited promising potential; further studies are warranted to evaluate the optimal dosing regimen and chemical modifications to increase effectiveness.


Subject(s)
Chelating Agents/administration & dosage , Chitosan/administration & dosage , Cobalt Radioisotopes/pharmacokinetics , Administration, Oral , Animals , Bone and Bones/metabolism , Feces , Injections, Intravenous , Intestinal Absorption , Kidney/metabolism , Liver/metabolism , Male , Metabolic Clearance Rate , Rats , Rats, Inbred F344 , Tissue Distribution
16.
Inhal Toxicol ; 21(6): 524-30, 2009 May.
Article in English | MEDLINE | ID: mdl-19519152

ABSTRACT

Methyl iodide (MeI) is an intermediate in the manufacture of some pesticides and pharmaceuticals, and is under review for US registration as a non-ozone depleting alternative for methyl bromide for pre-plant soil fumigation. MeI is primarily metabolized via conjugation with glutathione (GSH), with further metabolism to S-methyl cysteine and methanethiol. To facilitate extrapolations of animal pharmacokinetic data to humans, rate constants for the GSH metabolism of MeI were determined in cytosols prepared from the liver and kidneys of rats, human donors, female rabbits, and rabbit fetuses, from rabbit olfactory and respiratory epithelium, and from rabbit and rat blood using a headspace vial equilibration technique and two-compartment mathematical model. MeI was metabolized in liver and kidney from adults of all three species, but metabolism was not detectable in fetal rabbit kidney. Maximal metabolic rates (V(max)) were similar in liver from rat and human donors (approximately 40 and 47 nmol/min/mg, respectively) whereas the V(max) rates in kidney cytosols varied approximately three-fold between the three species. No difference was observed in the loss of MeI from active and inactive whole blood from either rats or rabbits. The metabolism in olfactory and respiratory epithelial cytosol had Michaelis-Menten constant (K(m)) values that were several times higher than for any other tissue, suggesting essentially first-order metabolism in the nose. The metabolism of MeI in human liver cytosol prepared from five individual donors indicated two potential populations, one high affinity/low capacity and one with a lower affinity but higher capacity.


Subject(s)
Glutathione/blood , Glutathione/chemistry , Hydrocarbons, Iodinated/blood , Hydrocarbons, Iodinated/pharmacology , Adult , Aged , Animals , Female , Glutathione/metabolism , Humans , Hydrocarbons, Iodinated/metabolism , Male , Middle Aged , Pregnancy , Rabbits , Rats , Rats, Sprague-Dawley , Tissue Distribution/drug effects , Tissue Distribution/physiology
17.
Inhal Toxicol ; 21(6): 552-82, 2009 May.
Article in English | MEDLINE | ID: mdl-19519155

ABSTRACT

Methyl iodide (MeI) has been proposed as an alternative to methyl bromide as a pre-plant soil fumigant that does not deplete stratospheric ozone. In inhalation toxicity studies performed in animals as part of the registration process, three effects have been identified that warrant consideration in developing toxicity reference values for human risk assessment: nasal lesions (rat), acute neurotoxicity (rat), and fetal loss (rabbit). Uncertainties in the risk assessment can be reduced by using an internal measure of target tissue dose that is linked to the likely mode of action (MOA) for the toxicity of MeI, rather than the external exposure concentration. Physiologically based pharmacokinetic (PBPK) models have been developed for MeI and used to reduce uncertainties in the risk assessment extrapolations (e.g. interspecies, high to low dose, exposure scenario). PBPK model-derived human equivalent concentrations comparable to the animal study NOAELs (no observed adverse effect levels) for the endpoints of interest were developed for a 1-day, 24-hr exposure of bystanders or 8 hr/day exposure of workers. Variability analyses of the PBPK models support application of uncertainty factors (UF) of approximately 2 for intrahuman pharmacokinetic variability for the nasal effects and acute neurotoxicity.


Subject(s)
Hydrocarbons, Iodinated/pharmacokinetics , Models, Biological , Adipose Tissue/metabolism , Adult , Aged , Animals , Brain/metabolism , Child , Female , Humans , Infant , Kidney/metabolism , Male , Nasal Mucosa/metabolism , Pregnancy , Rabbits , Rats , Rats, Sprague-Dawley , Respiratory Physiological Phenomena
18.
Inhal Toxicol ; 21(6): 531-6, 2009 May.
Article in English | MEDLINE | ID: mdl-19519153

ABSTRACT

Nasal dosimetry models that combine computational fluid dynamics and physiologically based pharmacokinetic modeling incorporate information on species-specific anatomical differences, including nasal airflow, mucosal diffusion, clearance-extraction, and metabolism specific to different epithelial layers. As such, these hybrid models have the potential to improve interspecies dosimetric comparisons, and may ultimately reduce uncertainty associated with calculation of reference concentrations. Validation of these models, however, will require unique experimental data. To this end, a method for evaluating the uptake of a prototypical compound, methyl iodide (MeI), in the nasal cavity of the intact animal was developed. The procedure involved insertion of a small-diameter air-sampling probe in the depth of the nasal cavity to the nasopharynx region in anesthetized animals. The exterior portion of the probe was connected directly to a mass spectrometer to provide a continual real-time analysis of concentrations of MeI in the nasal cavity. A plethysmography system was used to monitor breathing parameters, including frequency and tidal volume for each animal. Animals were placed in a sealed glass chamber and exposed to MeI at initial chamber concentrations ranging from 1 to 50 ppm. Studies were conducted on n = 3 rabbits per exposure concentration for a total of nine animals and n = 6 rats at a single exposure concentration of 1 ppm. In the rabbit, the percent of MeI absorbed in the nasal cavity ranged from 57 to 92% (average 72 +/- 11) regardless of exposure concentration. Similarly, the percent of MeI absorbed in the nasal cavity of the rat ranged from 51 to 71% (average 63 +/- 8).


Subject(s)
Anesthesia , Computer Systems/standards , Hydrocarbons, Iodinated/metabolism , Nasal Mucosa/metabolism , Volatile Organic Compounds/metabolism , Absorption/drug effects , Absorption/physiology , Anesthesia/methods , Animals , Female , Hydrocarbons, Iodinated/administration & dosage , Inhalation Exposure , Male , Nasal Mucosa/drug effects , Rabbits , Rats , Rats, Sprague-Dawley , Volatile Organic Compounds/administration & dosage
19.
Inhal Toxicol ; 21(6): 519-23, 2009 May.
Article in English | MEDLINE | ID: mdl-19337875

ABSTRACT

Methyl iodide (MeI) is a water soluble monohalomethane that is metabolized in vivo to release iodide (I-). A physiologically based pharmacokinetic (PBPK) model exists for iodide in adult rats, pregnant rats and fetuses, and lactating rats and neonates, but not for pregnant rabbits and fetuses, which have been used extensively for toxicity testing with MeI. Thus, this study was conducted to determine the blood and tissue distribution kinetics of radioiodide in pregnant rabbits and fetuses. Timed-pregnant New Zealand White rabbits received a single intravenous injection of the sodium salt of iodine-131 (Na131I) at either a high (10 mg/kg body weight) or low (0.75 mg/kg body weight) dose on gestation day 25. At various intervals ranging from 0.5 to 24h post- injection, blood and tissues (thyroid, stomach contents, and skin) were collected from each doe, and blood, stomach contents, thyroid, trachea, and amniotic fluid were collected from a random sampling of three fetuses per doe per time point. Radioiodide accumulated as expected in the thyroid of maternal animals, where concentrations were the highest of any maternal tissues measured in both dose groups. Radioiodide also accumulated in fetal blood and tissues; levels were consistently higher than maternal levels and, unlike maternal tissues, showed no evidence of clearance over the 24-h sampling period. In contrast to observations in the maternal animals, fetal stomach contents showed the highest accumulation of radioiodide for both dose groups by 1-2h after dosing, followed by the trachea and thyroid tissues, with the lowest concentrations of radioiodide in the amniotic fluid and blood. There was no evidence for preferential accumulation of radioiodide in fetal thyroid tissues.


Subject(s)
Hydrocarbons, Iodinated/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Models, Biological , Sodium Iodide/pharmacokinetics , Animals , Female , Pregnancy , Rabbits , Tissue Distribution/drug effects , Tissue Distribution/physiology
20.
J Long Term Eff Med Implants ; 18(2): 133-44, 2008.
Article in English | MEDLINE | ID: mdl-19968622

ABSTRACT

A physiologically based pharmacokinetic model was developed to describe the silicone constituent octamethylcyclotetrasiloxane (D4) and its migration from intact or ruptured silicone gel-filled breast implants into surrounding tissues. D4 is a representative low-molecular weight constituent of silicone gel that is soluble enough in biological fluids to migrate from the implant and into surrounding tissues. The simulations were based on a representative young adult (premenopausal) woman and a mature (postmenopausal) woman using worst-case exposure conditions (i.e., complete rupture of the largest implant available, maximum levels of D4 in silicone, equal solubility of D4 in breast tissue and gel, and a range of breast tissue fat contents). The results indicate that D4 is cleared primarily by exhalation with highest concentrations achieved briefly in breast tissues of a representative postmenopausal woman. Maximum D4 levels in breast tissues for this scenario were estimated to be approximately 750 ppb with over 90% cleared in about 20 days. Thus, it is unlikely that D4 would be detected in any tissue within a few weeks of receiving an implant, even if immediately ruptured, under the assumptions used in this model.


Subject(s)
Adjuvants, Immunologic/pharmacokinetics , Breast Implants/adverse effects , Foreign-Body Migration/etiology , Models, Biological , Prosthesis Design/adverse effects , Prosthesis Failure , Siloxanes/pharmacokinetics , Female , Humans , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL
...