Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Rep ; 13(1): 18840, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37914767

ABSTRACT

Rapid pathogen identification is a critical first step in patient isolation, treatment, and controlling an outbreak. Real-time PCR is a highly sensitive and specific approach commonly used for infectious disease diagnostics. However, mismatches in the primer or probe sequence and the target organism can cause decreased sensitivity, assay failure, and false negative results. Limited genomic sequences for rare pathogens such as Ebola virus (EBOV) can negatively impact assay performance due to undiscovered genetic diversity. We previously developed and validated several EBOV assays prior to the 2013-2016 EBOV outbreak in West Africa, and sequencing EBOV Makona identified sequence variants that could impact assay performance. Here, we assessed the impact sequence mismatches have on EBOV assay performance, finding one or two primer or probe mismatches resulted in a range of impact from minimal to almost two log sensitivity reduction. Redesigning this assay improved detection of all EBOV variants tested. Comparing the performance of the new assay with the previous assays across a panel of human EBOV samples confirmed increased assay sensitivity as reflected in decreased Cq values with detection of three positive that tested negative with the original assay.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Humans , Ebolavirus/genetics , Africa, Western , Disease Outbreaks , Genomics
2.
Microorganisms ; 9(3)2021 Mar 23.
Article in English | MEDLINE | ID: mdl-33806942

ABSTRACT

Ebola virus is a continuing threat to human populations, causing a virulent hemorrhagic fever disease characterized by dysregulation of both the innate and adaptive host immune responses. Severe cases are distinguished by an early, elevated pro-inflammatory response followed by a pronounced lymphopenia with B and T cells unable to mount an effective anti-viral response. The precise mechanisms underlying the dysregulation of the host immune system are poorly understood. In recent years, focus on host-derived miRNAs showed these molecules to play an important role in the host gene regulation arsenal. Here, we describe an investigation of RNA biomarkers in the fatal Ebola virus disease (EVD) cynomolgus macaque model. We monitored both host mRNA and miRNA responses in whole blood longitudinally over the disease course in these non-human primates (NHPs). Analysis of the interactions between these classes of RNAs revealed several miRNA markers significantly correlated with downregulation of genes; specifically, the analysis revealed those involved in dysregulated immune pathways associated with EVD. In particular, we noted strong interactions between the miRNAs hsa-miR-122-5p and hsa-miR-125b-5p with immunological genes regulating both B and T-cell activation. This promising set of biomarkers will be useful in future studies of severe EVD pathogenesis in both NHPs and humans and may serve as potential prognostic targets.

3.
PLoS Negl Trop Dis ; 14(11): e0008817, 2020 11.
Article in English | MEDLINE | ID: mdl-33141837

ABSTRACT

The 2013-2016 Ebola virus (EBOV) outbreak in West Africa and the ongoing cases in the Democratic Republic of the Congo have spurred development of a number of medical countermeasures, including rapid Ebola diagnostic tests. The likelihood of transmission increases as the disease progresses due to increasing viral load and potential for contact with others. Early diagnosis of EBOV is essential for halting spread of the disease. Polymerase chain reaction assays are the gold standard for diagnosing Ebola virus disease (EVD), however, they rely on infrastructure and trained personnel that are not available in most resource-limited settings. Rapid diagnostic tests that are capable of detecting virus with reliable sensitivity need to be made available for use in austere environments where laboratory testing is not feasible. The goal of this study was to produce candidate lateral flow immunoassay (LFI) prototypes specific to the EBOV glycoprotein and viral matrix protein, both targets known to be present during EVD. The LFI platform utilizes antibody-based technology to capture and detect targets and is well suited to the needs of EVD diagnosis as it can be performed at the point-of-care, requires no cold chain, provides results in less than twenty minutes and is low cost. Monoclonal antibodies were isolated, characterized and evaluated in the LFI platform. Top performing LFI prototypes were selected, further optimized and confirmed for sensitivity with cultured live EBOV and clinical samples from infected non-human primates. Comparison with a commercially available EBOV rapid diagnostic test that received emergency use approval demonstrates that the glycoprotein-specific LFI developed as a part of this study has improved sensitivity. The outcome of this work presents a diagnostic prototype with the potential to enable earlier diagnosis of EVD in clinical settings and provide healthcare workers with a vital tool for reducing the spread of disease during an outbreak.


Subject(s)
Antigens, Viral/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/diagnosis , Immunoassay/methods , Viral Envelope Proteins/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Democratic Republic of the Congo/epidemiology , Diagnostic Tests, Routine , Disease Outbreaks , Enzyme-Linked Immunosorbent Assay , Female , Hemorrhagic Fever, Ebola/epidemiology , Humans , Immunologic Tests , Mice , Point-of-Care Systems , Point-of-Care Testing , Polymerase Chain Reaction
4.
Sci Transl Med ; 10(434)2018 03 28.
Article in English | MEDLINE | ID: mdl-29593102

ABSTRACT

Ebola virus disease (EVD), caused by Ebola virus (EBOV), is a severe illness characterized by case fatality rates of up to 90%. The sporadic nature of outbreaks in resource-limited areas has hindered the ability to characterize the pathogenesis of EVD at all stages of infection but particularly early host responses. Pathogenesis is often studied in nonhuman primate (NHP) models of disease that replicate major aspects of human EVD. Typically, NHP models use a large infectious dose, are carried out through intramuscular or aerosol exposure, and have a fairly uniform disease course. By contrast, we report our analysis of the host response to EBOV after intranasal exposure. Twelve cynomolgus macaques were infected with 100 plaque-forming units of EBOV/Makona through intranasal exposure and presented with varying times to onset of EVD. We used RNA sequencing and a newly developed NanoString CodeSet to monitor the host response via changes in RNA transcripts over time. When individual animal gene expression data were phased based on the onset of sustained fever, the first clinical sign of severe disease, mathematical models indicated that interferon-stimulated genes appeared as early as 4 days before fever onset. This demonstrates that lethal EVD has a uniform and predictable response to infection regardless of time to onset. Furthermore, expression of a subset of genes could predict disease development before other host-based indications of infection such as fever.


Subject(s)
Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/virology , Administration, Intranasal , Animals , Disease Models, Animal , Hemorrhagic Fever, Ebola/immunology , Macaca fascicularis/virology
5.
PLoS One ; 12(8): e0183899, 2017.
Article in English | MEDLINE | ID: mdl-28859120

ABSTRACT

Antibiotic resistant bacterial infections are a significant problem in the healthcare setting, in many cases requiring the rapid administration of appropriate and effective antibiotic therapy. Diagnostic assays capable of quickly and accurately determining the pathogen resistance profile are therefore crucial to initiate or modify care. Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry (MS) is a standard method for species identification in many clinical microbiology laboratories and is well positioned to be applied towards antimicrobial susceptibility testing. One recently reported approach utilizes semi-quantitative MALDI-TOF MS for growth rate analysis to provide a resistance profile independent of resistance mechanism. This method was previously successfully applied to Gram-negative pathogens and mycobacteria; here, we evaluated this method with the Gram-positive pathogen Staphylococcus aureus. Specifically, we used 35 strains of S. aureus and four antibiotics to optimize and test the assay, resulting in an overall accuracy rate of 95%. Application of the optimized assay also successfully determined susceptibility from mock blood cultures, allowing both species identification and resistance determination for all four antibiotics within 3 hours of blood culture positivity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Staphylococcus aureus/drug effects , Cefepime , Cephalosporins/pharmacology , Ciprofloxacin/pharmacology , Humans , Microbial Sensitivity Tests , Oxacillin/pharmacology , Sensitivity and Specificity , Staphylococcal Infections/microbiology , Staphylococcus aureus/growth & development , Staphylococcus aureus/isolation & purification , Vancomycin/pharmacology
6.
Radiat Res ; 187(3): 298-318, 2017 03.
Article in English | MEDLINE | ID: mdl-28208025

ABSTRACT

Pneumonitis and fibrosis are potentially lethal, delayed effects of acute radiation exposure. In this study, male rhesus macaques received whole-thorax lung irradiation (WTLI) with a target dose of 10.74 Gy prescribed to midplane at a dose rate of 0.80 ± 0.05 Gy/min using 6 MV linear accelerator-derived photons. The study design was comprised of four animal cohorts: one control and three treated with AEOL 10150 (n = 20 animals per cohort). AEOL 10150, a metalloporphyrin antioxidant, superoxide dismutase mimetic was administered by daily subcutaneous injection at 5 mg/kg in each of three schedules, beginning 24 ± 2 h postirradiation: from day 1 to day 28, day 1 to day 60 or a divided regimen from day 1 to day 28 plus day 60 to day 88. Control animals received 0.9% saline injections from day 1 to day 28. All animals received medical management and were followed for 180 days. Computed tomography (CT) scan and baseline hematology values were assessed prior to WTLI. Postirradiation monthly CT scans were collected, and images were analyzed for evidence of lung injury (pneumonitis, fibrosis, pleural and pericardial effusion) based on differences in radiodensity characteristics of the normal versus damaged lung. The primary end point was survival to 180 days based on all-cause mortality. The latency, incidence and severity of lung injury were assessed through clinical, radiographic and histological parameters. A clear survival relationship was observed with the AEOL 10150 treatment schedule and time after lethal WTLI. The day 1-60 administration schedule increased survival from 25 to 50%, mean survival time of decedents and the latency to nonsedated respiratory rate to >60 or >80 breaths/min and diminished quantitative radiographic lung injury as determined by CT scans. It did not affect incidence or severity of pneumonitis/fibrosis as determined by histological evaluation, pleural effusion or pericardial effusion as determined by CT scans. Analysis of the Kaplan-Meier survival curves suggested that treatment efficacy could be increased by extending the treatment schedule to 90 days or longer after WTLI. No survival improvement was noted in the AEOL 10150 cohorts treated from day 1-28 or using the divided schedule of day 1-28 plus day 60-88. These results suggest that AEOL 10150 may be an effective medical countermeasure against severe and lethal radiation-induced lung injury.


Subject(s)
Lung Injury/drug therapy , Lung Injury/mortality , Metalloporphyrins/administration & dosage , Metalloporphyrins/pharmacology , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/mortality , Animals , Dose-Response Relationship, Radiation , Drug Administration Schedule , Kaplan-Meier Estimate , Macaca mulatta , Male , Metalloporphyrins/therapeutic use , Morbidity , Time Factors
7.
Sci Rep ; 6: 23612, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27029502

ABSTRACT

Cachexia, or muscle wasting, is a serious health threat to victims of radiological accidents or patients receiving radiotherapy. Here, we propose a non-human primate (NHP) radiation-induced cachexia model based on clinical and molecular pathology findings. NHP exposed to potentially lethal partial-body irradiation developed symptoms of cachexia such as body weight loss in a time- and dose-dependent manner. Severe body weight loss as high as 20-25% was observed which was refractory to nutritional intervention. Radiographic imaging indicated that cachectic NHP lost as much as 50% of skeletal muscle. Histological analysis of muscle tissues showed abnormalities such as presence of central nuclei, inflammation, fatty replacement of skeletal muscle, and muscle fiber degeneration. Biochemical parameters such as hemoglobin and albumin levels decreased after radiation exposure. Levels of FBXO32 (Atrogin-1), ActRIIB and myostatin were significantly changed in the irradiated cachectic NHP compared to the non-irradiated NHP. Our data suggest NHP that have been exposed to high dose radiation manifest cachexia-like symptoms in a time- and dose-dependent manner. This model provides a unique opportunity to study the mechanism of radiation-induced cachexia and will aid in efficacy studies of mitigators of this disease.


Subject(s)
Cachexia/veterinary , Gamma Rays/adverse effects , Muscle, Skeletal/radiation effects , Muscular Atrophy/veterinary , Weight Loss/radiation effects , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Cachexia/etiology , Cachexia/genetics , Cachexia/pathology , Disease Models, Animal , Dose-Response Relationship, Radiation , Gene Expression Regulation , Hemoglobins/metabolism , Humans , Macaca mulatta , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Myostatin/genetics , Myostatin/metabolism , Retrospective Studies , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Serum Albumin/metabolism , Time Factors
8.
Health Phys ; 109(5): 374-90, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26425899

ABSTRACT

Exposure to sufficiently high doses of ionizing radiation is known to cause fibrosis in many different organs and tissues. Connective tissue growth factor (CTGF/CCN2), a member of the CCN family of matricellular proteins, plays an important role in the development of fibrosis in multiple organs. The aim of the present study was to quantify the gene and protein expression of CTGF in a variety of organs from non-human primates (NHP) that were previously exposed to potentially lethal doses of radiation. Tissues from non-irradiated NHP and NHP exposed to whole thoracic lung irradiation (WTLI) or partial-body irradiation with 5% bone marrow sparing (PBI/BM5) were examined by real-time quantitative reverse transcription PCR, western blot, and immunohistochemistry. Expression of CTGF was elevated in the lung tissues of NHP exposed to WTLI relative to the lung tissues of the non-irradiated NHP. Increased expression of CTGF was also observed in multiple organs from NHP exposed to PBI/BM5 compared to non-irradiated NHP; these included the lung, kidney, spleen, thymus, and liver. These irradiated organs also exhibited histological evidence of increased collagen deposition compared to the control tissues. There was significant correlation of CTGF expression with collagen deposition in the lung and spleen of NHP exposed to PBI/BM5. Significant correlations were observed between spleen and multiple organs on CTGF expression and collagen deposition, respectively, suggesting possible crosstalk between spleen and other organs. These data suggest that CTGF levels are increased in multiple organs after radiation exposure and that inflammatory cell infiltration may contribute to the elevated levels of CTGF in multiple organs.


Subject(s)
Connective Tissue Growth Factor/biosynthesis , Disease Models, Animal , Radiation Exposure/analysis , Radiometry/methods , Viscera/metabolism , Viscera/radiation effects , Animals , Dose-Response Relationship, Radiation , Humans , Lethal Dose 50 , Macaca mulatta , Male , Organ Specificity/physiology , Up-Regulation/radiation effects
9.
Health Phys ; 109(5): 427-39, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26425903

ABSTRACT

A nonhuman primate (NHP) model of acute high-dose, partial-body irradiation with 5% bone marrow (PBI/BM5) sparing was used to assess the effect of Neupogen® [granulocyte colony stimulating factor (G-CSF)] to mitigate the associated myelosuppression when administered at an increasing interval between exposure and initiation of treatment. A secondary objective was to assess the effect of Neupogen® on the mortality or morbidity of the hematopoietic (H)- acute radiation syndrome (ARS) and concurrent acute gastrointestinal radiation syndrome (GI-ARS). NHP were exposed to 10.0 or 11.0 Gy with 6 MV LINAC-derived photons at approximately 0.80 Gy min. All NHP received medical management. NHP were dosed daily with control article (5% dextrose in water) initiated on day 1 post-exposure or Neupogen® (10 µg kg) initiated on day 1, day 3, or day 5 until recovery [absolute neutrophil count (ANC) ≥ 1,000 cells µL for three consecutive days]. Mortality in both the 10.0 Gy and 11.0 Gy cohorts suggested that early administration of Neupogen® at day 1 post exposure may affect acute GI-ARS mortality, while Neupogen® appeared to mitigate mortality due to the H-ARS. However, the study was not powered to detect statistically significant differences in survival. The ability of Neupogen® to stimulate granulopoiesis was assessed by evaluating key parameters for ANC recovery: the depth of nadir, duration of neutropenia (ANC < 500 cells µL) and recovery time to ANC ≥ 1,000 cells µL. Following 10.0 Gy PBI/BM5, the mean duration of neutropenia was 11.6 d in the control cohort vs. 3.5 d and 4.6 d in the day 1 and day 3 Neupogen® cohorts, respectively. The respective ANC nadirs were 94 cells µL, 220 cells µL, and 243 cells µL for the control and day 1 and day 3 Neupogen® cohorts. Following 11.0 Gy PBI/BM5, the duration of neutropenia was 10.9 d in the control cohort vs. 2.8 d, 3.8 d, and 4.5 d in the day 1, day 3, and day 5 Neupogen® cohorts, respectively. The respective ANC nadirs for the control and day 1, day 3, and day 5 Neupogen® cohorts were 131 cells µL, 292 cells µL, 236 cells µL, and 217 cells µL, respectively. Therefore, the acceleration of granulopoiesis by Neupogen® in this model is independent of the time interval between radiation exposure and treatment initiation up to 5 d post-exposure. The PBI/BM5 model can be used to assess medical countermeasure efficacy in the context of the concurrent GI- and H-ARS.


Subject(s)
Acute Radiation Syndrome/prevention & control , Acute Radiation Syndrome/physiopathology , Bone Marrow/radiation effects , Disease Models, Animal , Filgrastim/administration & dosage , Acute Radiation Syndrome/diagnosis , Animals , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Administration Schedule , Humans , Macaca mulatta , Male , Organ Sparing Treatments/methods , Radiation Dosage , Radiation-Protective Agents/therapeutic use , Treatment Outcome
10.
Health Phys ; 109(5): 479-92, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26425907

ABSTRACT

Computed Tomography (CT) and Echocardiography (EC) are two imaging modalities that produce critical longitudinal data that can be analyzed for radiation-induced organ-specific injury to the lung and heart. The Medical Countermeasures Against Radiological Threats (MCART) consortium has a well established animal model research platform that includes nonhuman primate (NHP) models of the acute radiation syndrome and the delayed effects of acute radiation exposure. These models call for a definition of the latency, incidence, severity, duration, and resolution of different organ-specific radiation-induced subsyndromes. The pulmonary subsyndromes and cardiac effects are a pair of interdependent syndromes impacted by exposure to potentially lethal doses of radiation. Establishing a connection between these will reveal important information about their interaction and progression of injury and recovery. Herein, the authors demonstrate the use of CT and EC data in the rhesus macaque models to define delayed organ injury, thereby establishing: a) consistent and reliable methodology to assess radiation-induced damage to the lung and heart; b) an extensive database in normal age-matched NHP for key primary and secondary endpoints; c) identified problematic variables in imaging techniques and proposed solutions to maintain data integrity; and d) initiated longitudinal analysis of potentially lethal radiation-induced damage to the lung and heart.


Subject(s)
Echocardiography/standards , Heart Injuries/diagnosis , Lung Injury/diagnosis , Multimodal Imaging/standards , Radiation Injuries/diagnosis , Tomography, X-Ray Computed/standards , Algorithms , Animals , Guidelines as Topic , Longitudinal Studies , Macaca mulatta , Male , Radiation Dosage , Reproducibility of Results , Sensitivity and Specificity
11.
Health Phys ; 109(5): 466-78, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26425906

ABSTRACT

Radiation-induced lung injury is highly complex and characterized by multiple pathologies, which occur over time and sporadically throughout the lung. This complexity makes biomarker investigations and medical countermeasure screenings challenging. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) has the ability to resolve differences spatially in molecular profiles within the lung following radiation exposure and can aid in biomarker identification and pharmaceutical efficacy investigations. MALDI-MSI was applied to the investigation of a whole-thorax lung irradiation model in non-human primates (NHP) for lipidomic analysis and medical countermeasure distribution.


Subject(s)
Lipids/analysis , Lung Injury/metabolism , Lung Injury/prevention & control , Radiation Injuries/metabolism , Radiation Injuries/prevention & control , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Biomarkers/analysis , Dose-Response Relationship, Drug , Lung Injury/diagnosis , Macaca mulatta , Male , Metalloporphyrins/administration & dosage , Radiation Injuries/diagnosis , Radiation-Protective Agents/administration & dosage , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome
12.
Health Phys ; 106(1): 56-72, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24276550

ABSTRACT

Several radiation dose- and time-dependent tissue sequelae develop following acute high-dose radiation exposure. One of the recognized delayed effects of such exposures is lung injury, characterized by respiratory failure as a result of pneumonitis that may subsequently develop into lung fibrosis. Since this pulmonary subsyndrome may be associated with high morbidity and mortality, comprehensive treatment following high-dose irradiation will ideally include treatments that mitigate both the acute hematologic and gastrointestinal subsyndromes as well as the delayed pulmonary syndrome. Currently, there are no drugs approved by the Food and Drug Administration to counteract the effects of acute radiation exposure. Moreover, there are no relevant large animal models of radiation-induced lung injury that permit efficacy testing of new generation medical countermeasures in combination with medical management protocols under the FDA animal rule criteria. Herein is described a nonhuman primate model of delayed lung injury resulting from whole thorax lung irradiation. Rhesus macaques were exposed to 6 MV photon radiation over a dose range of 9.0-12.0 Gy and medical management administered according to a standardized treatment protocol. The primary endpoint was all-cause mortality at 180 d. A comparative multiparameter analysis is provided, focusing on the lethal dose response relationship characterized by a lethal dose50/180 of 10.27 Gy [9.88, 10.66] and slope of 1.112 probits per linear dose. Latency, incidence, and severity of lung injury were evaluated through clinical and radiographic parameters including respiratory rate, saturation of peripheral oxygen, corticosteroid requirements, and serial computed tomography. Gross anatomical and histological analyses were performed to assess radiation-induced injury. The model defines the dose response relationship and time course of the delayed pulmonary sequelae and consequent morbidity and mortality. Therefore, it may provide an effective platform for the efficacy testing of candidate medical countermeasures against the delayed pulmonary syndrome.


Subject(s)
Disease Models, Animal , Lung/radiation effects , Radiation Dosage , Radiation Injuries, Experimental , Radiation Pneumonitis , Animals , Dexamethasone/pharmacology , Dose-Response Relationship, Radiation , Fibrosis , Hematologic Tests , Lung/drug effects , Lung/pathology , Lung/physiopathology , Macaca mulatta , Male , Organ Size/drug effects , Organ Size/radiation effects , Oxygen/metabolism , Radiation Injuries, Experimental/diagnostic imaging , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Radiation Pneumonitis/diagnostic imaging , Radiation Pneumonitis/etiology , Radiation Pneumonitis/pathology , Radiation Pneumonitis/physiopathology , Respiration/drug effects , Respiration/radiation effects , Survival Rate , Thorax/radiation effects , Time Factors , Tomography, X-Ray Computed
13.
Health Phys ; 106(1): 73-83, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24276551

ABSTRACT

The objective of this pilot study was to explore whether administration of a catalytic antioxidant, AEOL 10150 (C48H56C15MnN12), could reduce radiation-induced lung injury and improve overall survival when administered after 11.5 Gy of whole thorax lung irradiation in a non-human primate model. Thirteen animals were irradiated with a single exposure of 11.5 Gy, prescribed to midplane, and delivered with 6 MV photons at a dose rate of 0.8 Gy min. Beginning at 24 h post irradiation, the AEOL 10150 cohort (n = 7) received daily subcutaneous injections of the catalytic antioxidant at a concentration of 5 mg kg for a total of 4 wk. All animals received medical management, including dexamethasone, based on clinical signs during the planned 180-d in-life phase of the study. All decedent study animals were euthanized for failure to maintain saturation of peripheral oxygen > 88% on room air. Exposure of the whole thorax to 11.5 Gy resulted in radiation-induced lung injury in all animals. AEOL 10150, as administered in this pilot study, demonstrated potential efficacy as a mitigator against fatal radiation-induced lung injury. Treatment with the drug resulted in 28.6% survival following exposure to a radiation dose that proved to be 100% fatal in the control cohort (n = 6). Computed tomography scans demonstrated less quantitative radiographic injury (pneumonitis, fibrosis, effusions) in the AEOL 10150-treated cohort at day 60 post-exposure, and AEOL 10150-treated animals required less dexamethasone support during the in-life phase of the study. Analysis of serial plasma samples suggested that AEOL 10150 treatment led to lower relative transforming growth factor-Beta-1 levels when compared with the control animals. The results of this pilot study demonstrate that treatment with AEOL 10150 results in reduced clinical, radiographic, anatomic, and molecular evidence of radiation-induced lung injury and merits further study as a medical countermeasure against radiation-induced pulmonary injury.


Subject(s)
Antioxidants/pharmacology , Lung/drug effects , Lung/radiation effects , Metalloporphyrins/pharmacology , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/pharmacology , Animals , Antioxidants/administration & dosage , Antioxidants/chemistry , Antioxidants/therapeutic use , Catalysis , Dexamethasone/pharmacology , Lung/pathology , Lung/physiopathology , Macaca mulatta , Male , Metalloporphyrins/administration & dosage , Metalloporphyrins/chemistry , Metalloporphyrins/therapeutic use , Molecular Weight , Oxygen/metabolism , Pilot Projects , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Radiation Pneumonitis/blood , Radiation Pneumonitis/drug therapy , Radiation Pneumonitis/pathology , Radiation Pneumonitis/physiopathology , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/chemistry , Radiation-Protective Agents/therapeutic use , Respiration/drug effects , Respiration/radiation effects , Survival Rate , Tomography, X-Ray Computed , Transforming Growth Factor beta1/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...