Abstract
The Centers for Medical Countermeasures against Radiation Consortium (CMCRC) has provided a strong research foundation for the radiobiology science that will follow. After 20 years, however, the CMCRC will continue to conduct research on preparedness in niche areas of radiobiology and advanced product development, many of which were initiated by the CMCRC. This manuscript offers a review of past and current strategies and advancements in medical countermeasures to address radiation injuries, carried out by the CMCRC and funded by the National Institute of Allergy and Infectious Diseases Radiation and Nuclear Countermeasures Program. It also explores the mechanisms of radiation-induced injuries, discusses existing medical countermeasures, and highlights emerging technologies and potential future directions for radiobiology researchers. This review aims to enhance our understanding of current medical countermeasures against radiation and contribute to the future development of more efficient and innovative approaches to mitigate and treat radiation-induced damage.
INTRODUCTION AND PROGRAMMATIC OVERVIEW
On September 11, 2001, the threat of terrorism underscored the significance of national security in relation to weapons of mass destruction, especially those involving ionizing radiation. These events revived concerns about U.S. vulnerability to radioactive contamination incidents, radiological dispersal devices, and potential attacks on or accidents at nuclear power plants—concerns that had waned after the Cold War concluded in 1991 (1). In response, Congress allocated funds in 2004 to direct the National Institute of Allergy and Infectious Diseases (NIAID) to develop the Radiation and Nuclear Countermeasures Program (RNCP) (2). In 2005, NIAID created the NIH Strategic Plan and Research Agenda for Medical Countermeasures Against Radiological and Nuclear Threats with input from a Blue Ribbon Panel of scientific experts (3). The strategic plan was subsequently updated, and a progress report was issued in 2012 (4).
Radiation exposure can lead to both acute radiation syndrome (ARS) and delayed effects of radiation exposure (DEARE). Figure 1 illustrates some major organ systems affected by radiation-related comorbidities studied by the Centers for Medical Countermeasures against Radiation Consortium (CMCRC), including gastrointestinal (GI-ARS), hematopoietic (H-ARS), bone marrow, cutaneous, and vascular injuries as part of ARS. In contrast, DEARE encompasses lung, kidney, liver, cardiovascular, endocrine, and neurological injuries.
FIG. 1.

Major organ systems studied through the CMCR program and possible radiation-associated comorbidities (created with BioRender).
For the past 20 years (2005–2025), the CMCRC, a collaborative network of national research centers, has built the basic research foundation to prepare the government for medical management of radiological or nuclear public health emergencies. At its peak, the CMCRC included eight centers, 36 scientific projects, and 42 cores, significantly advancing the field of radiobiology in line with NIAID’s Strategic Plan. Currently (2020–2025), the CMCRC includes three Centers, each with multiple research projects focused on multi-disciplinary, product-oriented development, an administrative core, and optional scientific cores. Additionally, four consortium-wide cores (Coordinating Center, Opportunities Fund Management, Radiation Physics and Nonhuman Primate Radiation Survivors) were historically managed by selected institutions. The organization is overseen by a steering committee consisting of the principal investigators from each center, the leads of the consortium-wide cores, the NIAID program officer, and an external advisory committee appointed by NIAID. The administrative cores manage coordination and compliance, while scientific cores provide essential support. Although there are currently only three CMCR, Fig. 2 shows that this consortium has published over 115 peer-reviewed publications in 48 journals with over 667 authors representing 30 countries since receipt of notice of grant award (2020) through 2024 (also see Supplementary Table S1;2 https://doi.org/10.1667/RADE-24-00275.1.S1).
FIG. 2.
The country collaboration map in the CMCRC consortium 2020–2024. Depicted here are connections from 115 peer-reviewed publications in 48 journals with over 667 authors representing 30 countries. The Collaboration Network was generated using the R package Bibliometrix default parameters.3
The CMCRC’s primary objectives include: 1. Developing innovative biodosimetry techniques and devices for measuring radiation exposure in the human body; 2. Identifying biomarkers that indicate tissue damage and recovery; 3. Advancing novel medical countermeasures (MCMs) to minimize tissue damage, expedite recovery, restore normal functions, and improve survival rates. To date, this program has explored numerous radiation mitigators (Table 1) and enhanced diagnostic tools to assess radiation exposure at various maturity stages (Table 2).
TABLE 1.
CMCRC MCMs Development 2005–2025
| Hematopoietica | Gastrointestinala | Skina | Lungb |
|---|---|---|---|
| Cord Blood | Clusterin | Bmil1 | BCL-xL |
| Cordyceps Sinensis | Duke 9 | Celecoxib | BIO300 |
| Dinoprostone | DJ001 | Curcumin Derivates | Lisinopril/Captopril/Fosinopril/Enalapril |
| dmPGE2 | Extracellular Vesicles | TH-curcumin | EUK-189, 207, 423,451 |
| Endothelial cells | EUK-207,451 | EsA & h-EsA | FGF-PT |
| EUK-451 | FGF-P | EUK-207, 423, 451 | Genistein |
| Flt-3L | FGF-PT | FGrIL-12 | Homspera |
| G-CSF | FSL-1 | FGF-PT | Lovastatin |
| Human growth hormone | Hepoxilln A3 (HXA3) | JP4–039 | MnTnHex-2-PyP5+ |
| JP4–039 | IL-22 | MnTE-2-PyP5+ | |
| KGF | JP4–039 | SecinH3 | |
| Liposomes | Kineret | ||
| Renalb |
Central Nervous Systemb |
||
| Mesenchymal stem cells | KGF | ||
| Meloxicam | Li2CO3 | Atorvastatin | Atorvastatin |
| MnSOD-plasmid | Necrostatin | Captopril | EUK-189, 207,423,451 |
| Nocloprost | N-methyl-D-aspartate | EUK-207 | Minocycline |
| Rivenprost | PD0332991 | Enalapril | Ramipril |
| Stem cell transplant | RP-1 | Losartan | SecinH3 |
| Sulprostone | OrbeShield | Lisinopril | |
| Pleiotrophin | SOM-230 | ||
| PUMA | STO-609 | ||
| XJB-5–131 | XJB-5–131 | ||
| YeL1 & YeL2 |
Note. Medical countermeasures investigated by the CMCRC from 2005 to 2025 for acute
and delayed
sub-syndromes of irradiation.
TABLE 2.
CMCRC BioDosimetry Tools 2006–2025
| Blood/lymphocytes | Serum/plasma |
|---|---|
| γ-H2AX | Metabolomics |
| Microculture | Lipidomics |
| Automated | Targeted LCMS kits and assays |
| ImageStream | Targeted methods ± FPSE |
| CBMN | Proteomics |
| Microculture | Endothelial microparticles* |
| Automated | Exosomes* |
| Accelerated | miRNA* |
| Centrifuge-free | Cell-free DNA |
| ImageStream | Saliva |
| DCA | Metabolomics |
| Microculture | Targeted methods ± FPSE |
| Automated | qRT-PCR* |
| PCC | Skin |
| Microculture* | Micronuclei |
| Proteomics | Metabolites in sebum* |
| Peptide arrays* | γ-H2AX* |
| ATM phosphorylation* | Feces |
| Mass spectrometry | Metabolomics* |
| ELISA | Metagenomics* |
| ImageStream | Urine |
| Vertical flow POC* | Metabolomics |
| Aptamers (POC)* | Targeted methods ± FPSE |
| Transcriptomics | Exosomes* |
| Microarray | miRNA* |
| RNA-Seq | Proteome |
| qRT-PCR | Nails |
| CMOS (POC)* | EPR |
| Aptamers (POC)* | Hair |
| nCounter* | γ-H2AX |
| Vertical flow (POC)* | Teeth |
| Cheek Swab | EPR |
| miRNA mutations* | |
| Blood / Lymphocytes | Serum/Plasma |
| γ-H2AX | Metabolomics |
| Microculture | Lipidomics |
| Automated | Targeted LCMS kits and assays |
| ImageStream | Targeted methods ± FPSE |
| CBMN | Proteomics |
| Microculture | Endothelial microparticles* |
| Automated | Exosomes* |
| Accelerated | miRNA* |
| Centrifuge-free | Cell-free DNA |
| ImageStream | Saliva |
| DCA | Metabolomics |
| Microculture | Targeted methods ± FPSE |
| Automated | qRT-PCR* |
| PCC | Skin |
| Microculture* | Micronuclei |
| Proteomics | Metabolites in sebum* |
| Peptide arrays* | γ-H2AX* |
| ATM phosphorylation* | Feces |
| Mass spectrometry | Metabolomics* |
| ELISA | Metagenomics* |
| ImageStream | Urine |
| Vertical flow POC* | Metabolomics |
| Aptamers (POC)* | Targeted methods ± FPSE |
| Transcriptomics | Exosomes* |
| Microarray | miRNA* |
| RNA-Seq | Proteome |
| qRT-PCR | Nails |
| CMOS (POC)* | EPR |
| Aptamers (POC)* | Hair |
| nCounter* | γ-H2AX |
| Vertical flow (POC)* | Teeth |
| Cheek Swab | EPR |
| miRNA mutations* |
Abbreviations: γH2AX, phospho(S139) H2A.X variant histone; CBMN, cytokinesis block micronucleus assay; DCA, dicentric chromosome assay; PCC, premature chromatin condensation; PNA, peptide nucleic acid; ATM, ataxia telangiectasia mutated; ELISA, enzyme-linked immunosorbent assay; POC, point of care; RNA-Seq, RNA sequencing; qRT-PCR, quantitative real-time PCR; CMOS, complementary metal-oxide-semiconductor; LCMS, liquid chromatography mass spectrometry; FPSE, fabric phase sorptive extraction; miRNA, microRNA; EPR, electron para-magnetic resonance.
Denotes pilot project.
Under normal conditions, organ systems and tissues work together to maintain physiological homeostasis. However, exposure to elevated levels of radiation, such as may occur during a radiological or nuclear incident, can severely disrupt these mechanisms, leading to multi-organ failure and sometimes mortality due to ARS. Survivors of ARS face a risk of developing DEARE, characterized by systemic inflammation, immune aging, lung fibrosis, and cardiovascular and metabolic diseases. Understanding DEARE is crucial for informing public health officials about future medical needs following a radiation emergency.
The CMCRC has significantly contributed to radiation preparedness research, establishing a strong foundation for independent studies on how radiation impacts various systems, organs, tissues, and cells, including the hematological, gastrointestinal, lung, cutaneous, and cardiovascular systems. They have also investigated more complex scenarios such as combined injuries (i.e., radiation plus another trauma), radiation-induced immune dysfunction, and the role of the microbiome in reacting to radiation injury. Additionally, the CMCRC has played a pivotal role in studying biodosimetry, which involves developing methods to estimate the radiation dose a person has received and, to a lesser extent, radionuclide decorporation, which aids in eliminating radioactive substances from the body.
The CMCRC established small and large animal models and in vitro models to evaluate radiation MCMs and biodosimetry approaches aimed at achieving these goals. Additionally, understanding the cellular and molecular mechanisms of radiation injuries, such as DNA damage, oxidative stress, inflammation, and tissue impairment, is crucial for developing effective treatments. Furthermore, stem cell-based therapies may aid in tissue regeneration and repair. The knowledge gained from CMCRC research has advanced the development of MCMs and biodosimetry tools, bringing them closer to potential U.S. Food and Drug Administration (FDA) approval and inclusion in the Strategic National Stockpile for emergency use. After two decades of pioneering research, the CMCRC will conclude its operations. Nevertheless, the NIAID is committed to continuing funding for specific and advanced areas of radiobiological research, many of which originated under the CMCRC’s auspices.
ANIMAL MODELS
Due to ethical reasons and the infeasibility of human studies to determine the effectiveness of MCMs against ARS and/or DEARE, the FDA employs the Animal Rule to regulate, approve, and license MCMs. This non-traditional regulatory pathway relies on demonstrating therapeutic efficacy in “adequate and well-controlled” studies conducted in one or more well-characterized animal models as a surrogate for human clinical trials. Relevant animal models that mimic the human response to acute, high-dose, and potentially lethal radiation exposure have been established and validated to test potential MCMs. The rationale for defining the requirements for these models was based on the current understanding of radiation effects on human survivors from the Hiroshima and Nagasaki atomic bombings, as well as Belarus and Ukraine survivors of the Chernobyl Nuclear Power Plant accident.
In a radiological or nuclear incident, exposed individuals may receive non-homogeneous radiation doses that affect multiple organs. The degree and severity of radiation-induced injuries depend not only on the type and dose of radiation but also on the affected tissue. Animal models were designed to evaluate and understand key organ-specific and multi-organ sequelae for survivable nuclear radiation exposures. Additionally, animal models are needed to define the radiation dose, incidence, severity, and time-dependent relationship for ARS to progress into DEARE and to identify longitudinal biomarkers to guide MCM development.
Early approaches to developing potential mitigators of radiation injury focused on specific organ toxicities (e.g., hematopoietic, gastrointestinal, cutaneous, neurologic, etc.) or “syndromes.” This reductive method facilitated investigation into the effects of various potential mitigators on specific syndromes. It also tracked the clinical manifestations of these syndromes, noting that lower doses of radiation resulted in damage to the bone marrow and the development of H-ARS, while higher doses caused damage to the GI and central nervous system, accompanied by associated symptomology. Importantly, concentrating on the single syndrome of H-ARS led to four FDA-approved products as radiation MCMs, as well as several biosimilars.
While informative, organ-based radiation exposures like whole thoracic lung irradiation (WTLI) do not accurately simulate exposures likely to occur during mass casualty incidents, where multiple tissues are exposed to high radiation doses. In such an incident, total-body irradiation (TBI) doses significantly above conventional clinical practice would likely occur, with some shielding of the bone marrow, resulting in the development of H-ARS and GI-ARS, along with survivors subsequently experiencing late multi-organ injuries. Consequently, multi-organ injury models are being developed to compare critical radiation endpoints across sexes, strains, and acute versus delayed radiation-related morbidity and mortality. This strategy also aligns with the FDA’s preference for a multi-organ injury model for MCM development.
Several animal models have been used to study radiation-induced injury and for the development of MCMs (6). Various species, including small rodents and larger animals such as swine and non-human primates (NHPs), have greatly contributed to the current understanding of ARS; however, none of these models can perfectly reproduce every aspect of radiation-induced injuries in humans. It is understood that although a single animal model cannot completely replicate all radiation-induced injuries, using more than one model to cover different aspects of the syndrome often provides sufficient information to grant FDA approval under the Animal Rule. Here, we summarize the advantages and disadvantages of the animal models commonly used at the CMCRC institutions.
Mouse
Small animals such as rodents (mice and rats) are useful for investigating the effects of H-ARS, GI-ARS, and DEARE and screening efficacy of potential MCMs. Radiation mouse models have been utilized for decades in radiation oncology, including fractionated, focal, electron, and proton FLASH radiation exposures. A multi-organ injury mouse model was developed using partial-body irradiation (PBI) with 2.5% bone marrow shielding (PBI/BM2.5) (7). The TBI/BM2.5 model recapitulates the breathing dysfunction and microscopic abnormalities consistent with radiation pneumonitis and fibrosis.
Early studies on radiation-induced lung pathology and MCM development focused on focal irradiation of the upper thorax using the WTLI model. Several MCMs have been shown to enhance pathology and reduce mortality in the WTLI model (8, 9). However, the MCMs that showed efficacy in WTLI models did not always demonstrate the same effectiveness in the PBI/BM2.5 model, likely due to the multi-organ damage induced. Nonetheless, this model offers a more relevant radiation exposure, consistent with a mass casualty scenario, which is needed for the continued development of MCMs that target mediators of DEARE, such as monocyte/macrophage polarization, fibrosis, and senescence.
Advantages.
Several mouse strains are available; they are easy to handle, and they have a short gestation period and lifespan. Housing can conveniently be accommodated at most institutions, and the mice are economical. Reagents for murine genomic manipulation are readily available, and easily accessible databases already exist.
Disadvantages.
Murine physiology and organ structures differ from those of humans. Small body size confounds the variable of radiation dose distribution and makes it difficult to follow the effects of radiation exposure longitudinally. Two additional challenges arise with the use of mice in radiation studies. First, radiosensitivity varies widely between strains and should be considered when attempting to reproduce a particular type of radiation-induced injury in humans (10). For example, mouse strains differ in their development of DEARE, with some strains being more prone to pneumonitis or fibrosis. The C57L/J mouse strain is believed to best mimic the dose-response relationship (DRR) and lung injury pathology observed in NHP (11). The C57BL/6 strain is more radioresistant than the C57L/J in the development of DEARE, with an extended timeline for the development of lung injury that does not fully recapitulate the injuries anticipated in humans. Given the differences in radiosensitivity, it is recommended that MCMs be tested in more than one mouse strain (e.g., one resistant and one sensitive). Second, chronoradiosensitivity has been recognized since the 1960s as an important consideration in rodent models used for radiation studies. Studies by the CMCRC have shown that rodents are more radiosensitive during the morning hours (12). Investigators must consider this effect when designing experiments involving the irradiation of models.
Rabbits
Several ethical issues and a dwindling supply of NHPs for MCM studies have raised awareness of the need to develop larger non-clinical models that recapitulate aspects of human sequelae following acute radiation exposure. Rabbits have shown usefulness in understanding the physiological and pathological processes following radiation-induced injuries. These animals are easily managed, readily available, and compatible with imaging technologies such as computed tomography (CT) and magnetic resonance imaging (MRI).
Rabbits are considered the largest small animal model by the FDA, providing sufficient blood volume to allow repetitive sampling or monitor disease onset, dynamics, and progression during a longitudinal study. In addition, the coagulation profile in New Zealand White (NZW) rabbits, unlike rodents, is similar to that of humans and, therefore, more suitable for MCM development. In a model of TBI-induced ARS, the clinical manifestations of NZW rabbits exposed to different radiation doses recapitulated several human symptoms (13).
With moderate levels of supportive care, severe hemorrhage and bone marrow failure in rabbits were reported to be similar to those in other species, including humans. For each radiation dose tested (6.5–9.5 Gy), the damage to the small intestine was greater than that to the colon, as previously reported in NHPs (14). In addition, thrombocytopenia always preceded the onset of severe anemia, and the fall in hematocrit and red blood cell counts indicated an impending mortality (13). Gross findings of vital organs (stomach, small and large intestine, kidneys) at necropsy indicated architectural damage, inflammation, and hemorrhage consistent with a multiorgan injury model. The NZW rabbit model thus recapitulates several pathological symptoms associated with ARS in humans and may provide an alternative to the mouse model for MCM efficacy screening, dose finding, and schedule optimization studies.
Advantages.
Rabbits are docile, efficiently handled animals that are small enough to easily accommodate while having sufficient blood volume to allow multiple blood samplings for longitudinal studies. They are readily available, cost-effective, have a longer lifespan than rodents, and are immunologically more similar to humans than rodents. Further, their size facilitates more reliable imaging assessments (e.g., CT, MRI).
Disadvantages.
Although rabbits have previously been used to support radiobiology research and radiation MCM development, the model has yet to be developed to the same extent as other species, such as rodents and NHPs. In addition, substantial physiological differences from humans, including heart size, heart rate, and body weight, may complicate interpretation and extrapolation of findings to human response to ARS and MCMs. Finally, compared to reagents for mice and NHPs, few rabbit-specific reagents are available. This reality places limitations on analyses of pharmacodynamics, biomarkers, and mechanisms of action.
Swine
Swine have been used in medical research for quite some time. Similarities in size, anatomy, pathophysiology, metabolism, and pathology to humans make swine suitable models in multiple areas of medicine. In addition, the pig genome has 66% sequence identity to the human genome, and epigenetics are highly conserved (15–18). In the context of radiation-induced injuries, swine have been the preferred models for the evaluation of cutaneous injuries due to the comparability of swine skin to human skin (6). They are also useful models for studying the natural history of ARS progression as well as potential MCM evaluation (19–21).
Minipig radiation models have gained traction as suitable animal models for radiation exposure because their size is more appropriate than that of larger swine for laboratory housing, husbandry, and radiation studies. Minipigs serve as models for studying GI-ARS since they exhibit physiological signs, such as vomiting, which are not present in rodent and rabbit models. Currently, Sinclair and Göttingen minipig models are under development in the current CMCRC. Both strains exhibit similar hematological profiles to those observed in other species, including humans, with neutrophils, lymphocytes, platelets, and hematocrit decreasing after acute radiation exposure. In addition, gross observation at necropsy shows hemorrhage in the lungs, skin, heart, and GI in both strains, as could be expected in multiorgan injury models. Moreover, radiation absorption patterns in minipigs are expected to be comparable to those of young adult humans, given their similar body thickness (22). The minipig thus provides an additional large animal model to screen potential radiation mitigators and study H-ARS and GI-ARS.
Advantages.
The pathophysiology of H-ARS in minipigs is similar to the human response. Minipigs are also relevant models for studying GI-ARS with endpoints of vomiting and diarrhea. Minipig skin and body thicknesses are comparable to humans, thus facilitating the extrapolation of radiation absorption data to human exposure. Minipigs have a longer lifespan compared to mice and rabbits, and their larger size supports their use as an additional large non-clinical model to evaluate MCM against acute radiation exposure.
Disadvantages.
Minipigs exhibit increased radiosensitivity compared to humans and other large animal species and may need supportive care to extend survival long enough to assess crypt-epithelial cell transit time and countermeasure efficacy. Radiosensitivity also varies widely between minipig strains. For example, after 2.3 Gy TBI, Göttingen minipigs had 25% survival at day 45 postirradiation, compared to 100% survival in Sinclair minipigs (17). The difference in radiation dose tolerance is unclear but may be related to the inbred nature of the Göttingen compared to the outbred nature of the Sinclair minipig. Furthermore, while a PBI model has been developed in Göttingen minipigs, only the abdomen and lower extremities were exposed to radiation, minimizing the contribution of multi-organ injury in the development of GI-ARS and DEARE (20).
Non-human Primates
The NHP model is the closest model related to humans, with 95% DNA sequence identity and a high degree of physiological receptor and pathway similarity. The NHP model most closely reproduces the clinical, histopathological, and pathophysiological aspects of radiation injury in humans; therefore, it is considered the gold standard by most investigators. Based on convenience, cost, and availability, the genus Macaca (rhesus and cynomolgus) has historically been the most commonly used NHP for biomedical research. Rhesus macaques are known as either Chinese or Indian, depending on their country of origin.
The rhesus macaque has anatomic, tissue composition, and tissue complexity similar to humans. Their response to radiation and treatments is analogous to the human radiation response, which facilitates the application of derived information to clinical situations. The model also allows for dose-effect correlation with humans due to similar requirements for supportive care, body size/thickness, and a longer lifespan than other animal models (6). The LD50/60 of humans that received TBI is proposed to be approximately 4.5 Gy without supportive care and 6.0 Gy with supportive care, in line with the 7.2 Gy LD50/60 reported for rhesus macaques with supportive care (23).
The pharmacokinetic/pharmacodynamic parameters for MCMs in NHPs approximate the human response. These criteria adhere to the FDA Animal Rule guidance for MCM approval (5). For these reasons, the rhesus macaques and, more specifically those of Chinese origin, had historically been the model of choice in biomedical research for the development of radiation MCMs. However, recent restrictions on the export of these animals have led to the increased use of other species, including rhesus macaques of Indian origin, cynomolgus (Macaca fasicularis) macaques, and squirrel monkeys. Although all these animals significantly contribute to advanced biomedical research, comparison between historical and current studies will be limited to animals of similar species and genetic backgrounds.
A particular advantage of NHP models is the opportunity to study DEARE in surviving irradiated NHPs. A multi-morbidity pattern has emerged, which in some ways resembles aging but also has a distinctive pattern of fibrosis, vascular pathology, metabolic derangement, gonadal injury, cataractogenesis, and neuroinflammation reflecting mechanisms characteristic of radiation injury (Fig. 3) (24). The Wake Forest Radiation Late Effects Cohort (RLEC) of rhesus macaque (Macaca mulatta) NHPs are a unique colony of long-term radiation survivors, consisting of animals exposed TBI, PBI, and WTLI. The cohort is composed of 290 NHPs (215 males/75 females), with 239 irradiated NHPs (168 males/71 females) and 51 non-irradiated controls (47 males/4 females). There are 217 animals under current observation, and 153 are deceased. Each irradiated animal received 1.14–8.5 Gy TBI (mean exposure was 6.13 Gy). The NHPs were irradiated at 2.3–15.5 years of age (mean age was = 4.6 years). This cohort has been under observation for nearly 18 years. All NHPs undergo clinical screening on an annual or semi-annual basis, including physical exams, MRI, CT, body composition analysis, echocardiograms, abdominal ultrasound, complete blood counts, and blood chemistries. Clinical diagnoses, including type II diabetes, heart disease, hypertension, renal disease, neoplasia, underweight, MRI brain lesions, and cataracts, are made using rigorously defined criteria (Fig. 3). Euthanized or dying animals receive a comprehensive postmortem and histopathologic exam performed by veterinary pathologists, and over 80 unique tissue samples are archived.
FIG. 3.

Chord diagram of the co-occurrence of major morbidities by organ or disease process in (N = 270) irradiated rhesus macaques. The category “no other” represents the low proportion of animals with only one morbidity.
NHPs receiving any dose of radiation had a mean accumulation of 4.3 morbidities (SD = 2.6), whereas the mean morbidity accumulation for unirradiated NHPs was 2.7 (SD = 1.9; P < 0.0003). Additionally, we have observed that certain morbidities, such as heart disease, tend to manifest earlier, with a mean age of onset of 9.3 years (SD = 4.4) after irradiation, than others, like type II diabetes which has a mean age of onset of 11.3 years (SD = 3.0 years; P < 0.05). This finding suggests unique organ responses to systemic injury, such as inflammation, vascular injury, and resultant fibrosis, routinely observed in the cohort. The ability to intensively monitor these NHPs over decades represents an unparalleled resource for modeling DEARE in a well-established large animal model.
Advantages.
The NHP model is as close as possible to humans, sharing more than 95% of identical DNA sequences, resulting in similar organ structure, metabolism, and a long life span. The model also allows easy sequential sampling and is suitable for monitoring GI symptoms, including vomiting and diarrhea. A comparable body size, thickness, and supportive care treatment facilitate the correlation of dose-effect relationships to human symptoms and treatment response.
Disadvantages.
The social and cognitive similarity between humans and NHPs raises ethical considerations unique to NHPs. A longer breeding period increases housing, feeding, and handling costs. Acquisition due to recent export restriction seriously limits their availability. Finally, cutaneous studies are complicated by the hirsute nature of NHP skin.
MEDICAL COUNTERMEAURES
The Strategic National Stockpile Radiation Working Group noted that animal models are key to MCM development and crucial for U.S. preparedness for a radiological or nuclear event (25). Table 1 shows the numerous MCMs studied by the CMCRC, which span many organ systems, given the expected multi-organ insults after radiation exposure. Early MCMs repurposed for radiation exposure include leukocyte growth factors such as granulocyte colony-stimulating factor (G-CSF), studied by the CMCRC, and granulocyte-macrophage colony-stimulating factor (GM-CSF). In 2015, collaborations between NIAID, Amgen, and the FDA led to the approval of Filgrastim (Neupogen®) (26) and Pegfilgrastim (Neulasta®) (27) for H-ARS based on studies conducted under the FDA Animal Rule (28). Preclinical studies demonstrated Neupogen’s efficacy in mice (29) and NHPs (29) when administered 24 h postirradiation; Neulasta showed similar efficacy, leading to its approval. The Biomedical Advanced Research and Development Authority (BARDA), in partnership with Partner Therapeutics and the FDA, spearheaded the approval of Leukine® (Sargramostim) in 2018, showing that delayed treatment up to 48 h postirradiation significantly reduced mortality and enhanced recovery of hemopoietic cell counts (30). In 2021, Nplate® (Romiplostim) (31), an anti-thrombocytopenic that stimulates platelet recovery, was approved through collaborations with NIAID, Amgen, and the FDA. These public-private partnerships also enabled the approval of several growth factor biosimilars, including Nypozi™ (filgrastim-txid), Udenyca (pegfilgrastim-cbqv), Stimufend (pegfilgrastim-FPGA), Zarxio (filgrastim-sndz), and Ziextenzo (pegfilgrastim-bmez). The development and approval of these MCMs underscore the importance of collaborative efforts in enhancing U.S. radiation medical preparedness. These advancements ensure that effective treatments are available to mitigate the adverse effects of radiation exposure on hematopoiesis.
Other MCMs tested under the CMCRC continue to advance through academic and industry collaborations. For example, a genetically engineered probiotic Limosilactobacillus reuteri that releases IL-22 is being developed for GI-ARS under an NIAID contract with ChromoLogic, LLC, and the University of Pittsburgh (32). Lisinopril is also being developed for lung DEARE under an NIAID contract with the Medical College of Wisconsin (33). BIO300 was being developed under the CMCRC and a NIAID contract as a mitigator (7, 8). Humanetics Corporation was awarded funding to pursue a Phase 2 clinical trial to study BIO300 as a therapeutic to mitigate lung injury in discharged COVID-19 patients.4 The Department of Defense has also funded BIO 300 studies for prophylactic radioprotection (34).
BIODOSIMETRY
Biodosimetry tools will be needed to triage a large population and guide medical management after a mass casualty radiation incident. Predicting the severity and specific manifestations of radiation injuries is complex due to the variability in symptoms, which can take days to years to evolve, and the influence of individual factors such as age, immune status, and genetic background. The CMCRC has been at the forefront of biodosimetry diagnostic tools and biomarker discovery, aiming to predict early and delayed damage to specific organs and tissues. This research enables precise and timely medical interventions, reducing morbidity and mortality.
Cytogenetic approaches have historically been the cornerstone of biodosimetry, using lymphocyte DNA damage to assess radiation exposure. The dicentric chromosome assay (DCA) is considered the “gold standard” in the field, where lymphocyte division is induced but halted at the metaphase stage, allowing the identification of dicentric chromosomes specific to radiation exposure (35, 36). The formation of dicentric chromosomes requires at least two double-strand breaks formed on adjacent chromosomes and is highly specific for radiation. The average number of dicentric chromosomes per metaphase has a linear-quadratic relationship with dose and can be used for dose reconstruction.
Another useful assay is the cytokinesis blocked micronucleus (CBMN) assay, where lymphocyte division is blocked in cytokinesis, forming a binucleate cell. Irradiated lymphocytes may contain micronuclei, representing chromosome fragments not incorporated into the daughter nuclei during division (36, 37). Micronuclei are small, generally round objects in the cytoplasm of the cells outside the main nucleus. They represent chromosome fragments not incorporated into the daughter nuclei during division. The average number of micronuclei per binucleate cell also has a linear-quadratic relationship with dose and can be used for dose reconstruction. The main disadvantage of these assays is that 2–3 days are required for culture, but this has been accelerated and automated within the CMCRC for faster and more reliable results (38–40).
Radiation exposure causes protein modifications, expression, and localization, as well as rapid transcriptional and metabolic cellular reprogramming. These changes can be detected in easily sampled biofluids such as blood, serum, and urine. From cell samples, the γ-H2AX assay can be used to measure DNA damage by immunolabeling phosphorylated histone H2AX, which localizes to double-strand breaks (41). This assay can provide a dose estimate within a few hours and has the potential for automation (42, 43). Scoring is done by counting the number of foci per cell or the total fluorescence within the nucleus. While focus counting is more precise at low doses, it requires higher resolution (ideally confocal) imaging and saturates at low doses. Total fluorescence is easier but precision is not as good at low doses (44, 45). The major limitation of γ-H2AX analysis is its low persistence. The peak signal is achieved at 2–4 h, followed by a decrease in yields until 24 h postirradiation, when levels are close to the background.
The CMCRC has been instrumental in developing effective clinical tools for detecting radiation exposure using multiple approaches to biodosimetry (Table 2). Although γ-H2AX (46) and cytogenetic markers such as DCA (47), CBMN (37), and premature chromosome condensation (PCC) (48) have been used for biodosimetry, the assays are not high throughput, so CMCRC efforts focused on increasing throughput. Assays were developed using microculture in 96-well plates (38, 39, 42, 49) on robotic platforms with different configurations [e.g., custom robotics (50), high-throughput screening platforms (38, 39, 51–53), and imaging flow cytometry (45, 54)].
Historically, individual gene and protein markers have been explored as tools for biodosimetry. The CMCRC enabled extensive efforts in discovery using platforms like microarrays, RNA-Seq, antibody screening, and mass spectrometry (MS) (55–58), followed by signature development (59, 60) and point of care (POC) assays that would identify irradiated individuals in 30 min or less (61–63). The CMCRC also pioneered analysis of small molecule metabolites obtainable from other easily accessible biofluids (e.g., serum/plasma, urine, saliva) (64, 65), mainly using non-targeted MS approaches in discovery and later stages to increase speed and throughput (66, 67). In the past, other CMCRC studies addressed different approaches, such as exploration of the fecal microbiome and metabolome (68), gene expression signatures in saliva (69), cell-free DNA in serum (70), and γH2AX in skin (71) and hair (72). Physical dosimetry approaches using biological samples (e.g., tooth- and fingernail-based electron paramagnetic resonance) were also refined (73, 74). These studies have relied heavily on in vivo mouse and ex vivo human samples for their development. Studies have been conducted in NHPs both in vivo and ex vivo (75–79), genetically engineered mouse models representing alterations in DNA repair and immune pathways (80, 81), and in mice reconstituted with a human immune system (82–84).
Several biodosimetry tools have progressed from basic research under the CMCRC to advanced development funded by NIAID and BARDA. For example, the Columbia University Center launched technologies such as ASELL CytoRADx™ (85) and a MRIGlobal ARad Biodosimetry assay in partnership with Thermo Fisher Scientific and Arizona State University (86). The Duke University Center contributed to the discovery phase of DxTerity Diagnostics’ demonstration of the utility of gene signatures in animal models to predict radiation exposure in humans (87). Other biodosimetry technologies studied under the CMCRC, such as tooth EPR and micro DNA, also transitioned to BARDA but were not commercialized.
Rapid individual radiation dose estimates are essential for optimal medical management of mass radiation casualties and for determining the appropriate administration of counter-measures. In a mass casualty incident, quick identification of those not exposed to radiation can prevent medical facilities from being overwhelmed, allowing resources to focus on those with radiation exposures. After a large-scale radiological or nuclear incident, rapid radiation dose reconstruction can estimate the affected organ systems and identify individuals most likely to benefit from specific MCMs (88). Significant progress has been made in this area since the launch of the CMCRC, with a recent focus on developing biomarkers for radiological injury to particular organs or organ systems (e.g., lung, GI, etc.) and monitoring treatment responses. International interlaboratory intercomparison exercises have also been conducted to standardize methods and compare results from multiple biodosimetry approaches (89–92).
Methods based on signatures of responsive biomolecules are also under development. The earliest to emerge were signatures of genes measured in peripheral blood cells (93–96) and proteins in serum (97, 98). More recently, non-coding RNAs in blood or serum (99) and metabolomic profiles in serum or urine (65, 100, 101) have also shown great promise. These methods all rely on highly dynamic changes over time after irradiation, but most have shown markers that persist for at least a week. The use of multiple biomolecules in signatures is thought to militate against confounding by other exposures, disease states, or individual variation, but these factors are not yet completely understood. Measuring biomolecules in fluids such as urine is particularly attractive for large-scale incidents because it is a non-invasive approach for biodosimetry, avoiding the major potential bottleneck presented by the need to draw blood for most assays. These assays also have a great potential for both automation and integration into POC devices for rapid in-field testing (61, 102–105), a growing area of biodosimetry research.
EMERGING DIRECTIONS
Advancements in science and technology offer promising avenues for the development of novel MCMs against radiation injury and biodosimetry development. With TBI and PBI, there is a systemic effect rather than a local organ effect, providing a better understanding of tissue injury and actions of countermeasures. There have been several fruitful areas of research.
Endothelial/Vascular Injury
One scientific premise is that endothelial cell (EC) injury is the sine qua nonincident for radiation injury, initiating both downstream events and the perpetuation of injury (106). Processes that regulate whether ECs repair/regenerate or die are fundamental to the outcomes of patients. Endothelial cellS elaborate cytokines to modulate other cells in an autocrine, paracrine, and endocrine fashion. After irradiation, ECs will elaborate cytokines that incite tissue damage or facilitate cellular repair (107). In addition, ECs have a role in innate and adaptive immune responses. For example, CCL5 is secreted by ECs and other cells, facilitating tissue injury repair, and also acts as a potent chemoattractant for T cells (108). Thus, ECs not only participate in T cell recruitment to areas of injury but also direct T cells to produce inflammatory cytokines (e.g., IFNg) and modulate T cell function. Endothelial cells also mediate innate immune responses through enhanced neutrophil trafficking, activation of toll-like receptors (109), and increased secretion of other inflammatory cytokines like IL-1 beta, IL-6, and IL-8 in response to pathogens (110). Targeting inflammatory proteins could reset the inflammatory pathways and ameliorate the bone marrow microenvironment.
Immunity
The immune response, discussed at a high level above, is a key system that encompasses the most important mechanisms for healing. For example, monocytes, which constitute 5–10% of circulating white blood cells in humans and NHPs, respond to tissue injury and may transform into tissue-resident macrophages upon exiting the vasculature (111, 112). Classical monocytes represent the majority of monocytes (~85%), with intermediate and non-classical monocytes comprising the remainder (~15% total). These monocyte subsets have disparate phenotypes (113), where classical monocytes are associated with inflammatory responses and anaerobic metabolism, intermediate monocytes are associated with antigen presentation and processing, and non-classical monocytes are associated with oxidative phosphorylation and protein metabolism. Depending on the signals received from the local and systemic environment, monocytes may adopt a spectrum of phenotypes, developing into pro-inflammatory (M1-like) or anti-inflammatory-tissue remodeling/profibrotic (M2-like) macrophages (114). These definitions are too simplistic, and the role monocytes and macrophages play as master regulators of inflammation and fibrosis needs to be explored in more detail.
Microbiome
As epithelial organs, the gut, lungs, and skin are targets of radiation injury and are also hosts to extensive communities of microorganisms. Hundreds of studies have demonstrated associations between the microbiome and human health and disease, including associations between the gut microbiota and radiation-induced damage (115, 116). The outcome from radiation may be affected by the microbiome, microorganisms that colonize epithelial niches and contribute to health and disease. For example, gut bacteria may attenuate radiation responses by interacting with host cells to promote or inhibit apoptosis. Thus, targeting the microbiome can help mitigate radiation injury to these organs and improve survival. Previously published murine studies have shown that while most C57BL/6 mice receiving 9.2 Gy TBI will die from radiation injury, a small population of “elite survivors” can live relatively normal life spans (117). These mice harbored a distinct gut microbiota with protection against radiation-induced damage and death.
Moreover, the microbiota transfer from elite survivors to specific pathogen-free mice conferred these protective benefits, improving survival after irradiation. These microbiomes were characterized by elevated levels of Lachnospiraceae and Enterococcaceae, which were also found to be more abundant in humans who had fewer radiation-related toxicities after receiving TBI as part of hematopoietic stem cell transplantation. In addition, elite survivor mice had increased levels of microbially derived propionate and other short-chain fatty acids, and administration of these specific lipids protected against radiation injury, including mitigating H-ARS and reducing inflammatory responses.
Fibrosis (Long-term Effect)
A major DEARE manifestation is radiation-induced fibrosis (RIF), a debilitating condition occurring 4–12 months postirradiation in humans that has been referred to as a “wound that fails to heal.” RIF has been documented in many tissues, including the heart, lungs, liver, and skin (118–120). The initiating signals and cells that lead to progression from quiescence to RIF are largely unknown but likely involve an aberrant healing response mediated by monocytes, macrophages, fibroblasts, myofibroblasts, endothelial cells, and platelets (121). Ultimately, this response results in excessive extracellular matrix deposition, which reduces tissue compliance, impairs functionality, and increases morbidity. Unfortunately, there are currently no therapies that can halt or reverse RIF.
Nonetheless, monocytes and macrophages are known to be master regulators of inflammation and fibrosis, playing key roles in initiating, producing, and resolving fibrosis (122). Increased intermediate monocyte polarization and subsequent tissue infiltration have been implicated in the pro-fibrotic activity in pathologic fibrotic disorders, including systemic sclerosis, renal fibrosis, idiopathic pulmonary fibrosis, hepatic fibrosis, cardiac fibrosis, and skeletal muscle fibrosis (123–125). Early monocyte and macrophage depletion by clodronate in a bleomycin-induced pulmonary fibrosis model reduces fibrosis (126). Depletion of circulating monocytes during the early stages after infarction impaired healing and increased necrotic debris accumulation, whereas depletion during the later stages decreased collagen deposition and endothelial cells (127). These findings suggest monocyte-macrophages may be a target for intervention to inhibit radiation-induced fibrosis.
CONCLUSIONS
The NIAID CMCRC program has contributed significantly over the past 20 years to basic radiobiology, novel MCMs development, dose reconstruction, and development of biodosimetry tools for triage and medical management of patients. The goal is to continue to develop and mature these technologies for use after a radiation mass casualty scenario. Many products were matured under NIAID funding and some were transferred to BARDA for advanced development.
A robust early product development pipeline was established by funding high-risk, high-reward projects through the CMCRC and associated pilot projects program. Pilot projects were initially selected and administered by the individual centers, and for the last ten years through the Opportunities Funds Management Core (OFMC). The scientists and projects brought into the CMCRC portfolio have been extremely productive. In the nearly ten years of the OFMC, pilot awardees have published at least 83 related peer-reviewed publications. They have reported that data generated in their pilot projects contributed to the submission of 52 grant proposals (11 of which were reported as funded), nine patent filings, one invention report, and one patent award.
The CMCRC has successfully met its original objectives, resulting in a comprehensive radiation medical preparedness research portfolio. As the CMCRC concludes its operations, the NIAID/RNCP views this as a transition to a stronger and more advanced research preparedness portfolio, thanks to the consortium’s pioneering efforts. Radiation MCMs play a crucial role in mitigating the adverse effects of radiation exposure, and the biodosimetry tools developed will help in the allocation of essential medical supplies, protective equipment, and MCMs. This review provides an overview of current strategies, challenges, and future directions in this field, underscoring the CMCRC’s lasting impact and the continued evolution of research and medical preparedness in radiobiology.
Supplementary Material
SUPPLEMENTARY MATERIALS
Supplementary Table S1. Detailed information on 115 publications during the period of notice of awards for the three centers for medical countermeasures against radiation, starting in 2020.
ACKNOWLEDGMENTS
The authors would like to express their deep appreciation to all past members of the CMCRC. We want to thank the project leads who have contributed to the success of this 4th generation of the CMCRC: Benny Chen, Phuong Doan, Al Fornace, Joga Gobburu, Isabel Lauren Jackson, David Kirsch, Evagelia Laiakis, Paul Okuneiff, Christie Orschell, Louis Pelus, Brian Ponnaiya, Luigi Racioppi, Igor Shuryak, Michael Terrin, Gabor Tigyi, Jenny Ting, and Zeljko Vujaskovic, Daohong Zhou. Also, a big thank you to all the co-investigators and technical staff of past and present CMCRCs, as well as previous NIAID CMCRC Program Officers (Narayani Ramakrishnan, Francesca Macchiarini, and Andrea DiCarlo) for their guidance. This review was supported by NIAID Cooperative Agreements: 5U19AI150574, 5U19AI067773, and 5U19AI067798.
Footnotes
Editor’s note. The online version of this article (DOI: https://doi.org/10.1667/RADE-24–0075.1) contains supplementary information that is available to all authorized users.
Aria M, Cuccurullo C, Bibliometrix: An R-tool for comprehensive science mapping analysis, An R-tool for comprehensive science mapping analysis. J Informetr 2017; 11:959–75.
REFERENCES
- 1.DiCarlo AL, Homer MJ, Coleman CN. United States medical preparedness for nuclear and radiological emergencies. J Radiol Prot 2021. Dec 6; 41(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Rios CI, Cassatt DR, Dicarlo AL, Macchiarini F, Ramakrishnan N, Norman MK, et al. Building the strategic national stockpile through the NIAID Radiation Nuclear Countermeasures Program. Drug Dev Res 2014. Feb; 75(1):23–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.NIH Strategic Plan and Research Agenda for medical counter-measures against radiological and nuclear threats Washington, DC: Department of Health and Human Services; NIH; 2005; Available from: https://www.niaid.nih.gov/sites/default/files/radnucstrategicplan.pdf. [Google Scholar]
- 4.Health NIo. National Institutes of Health Strategic Plan And Research Agenda for Medical Countermeasures Against Radiological and Nuclear Threats Progress Report: 2005–2011 and Future Research Directions: 2012–2016. 2012; Available from: https://www.niaid.nih.gov/sites/default/files/radnucprogressreport.pdf. [Google Scholar]
- 5.U.S. Food and Drug Administration. Product Development Under the Animal Rule. Center for Biologics Evaluation and Research, Center for Drug Evaluation and Research; 2015. [Google Scholar]
- 6.Singh VK, Newman VL, Berg AN, MacVittie TJ. Animal models for acute radiation syndrome drug discovery. Expert Opin Drug Discov 2015. May; 10(5):497–517. [DOI] [PubMed] [Google Scholar]
- 7.Gibbs A, Gupta P, Mali B, Poirier Y, Gopalakrishnan M, Newman D, et al. A C57L/J mouse model of the delayed effects of acute radiation exposure in the context of evolving multi-organ dysfunction and failure after total-body irradiation with 2.5% bone marrow sparing. Radiat Res 2023. Apr 1; 199(4):319–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Jackson IL, Zodda A, Gurung G, Pavlovic R, Kaytor MD, Kuskowski MA, et al. BIO 300, a nanosuspension of genistein, mitigates pneumonitis/fibrosis following high-dose radiation exposure in the C57L/J murine model. Br J Pharmacol 2017. Dec; 174(24):4738–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Murigi FN, Mohindra P, Hung C, Salimi S, Goetz W, Pavlovic R, et al. Dose optimization study of AEOL 10150 as a mitigator of radiation-induced lung injury in CBA/J mice. Radiat Res 2015. Oct; 184(4):422–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Williams JP, Brown SL, Georges GE, Hauer-Jensen M, Hill RP, Huser AK, et al. Animal models for medical countermeasures to radiation exposure. Radiat Res 2010. Apr; 173(4):557–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Jackson IL, Vujaskovic Z, Down JD. Revisiting strain-related differences in radiation sensitivity of the mouse lung: recognizing and avoiding the confounding effects of pleural effusions. Radiat Res 2010. Jan; 173(1):10–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Plett PA, Sampson CH, Chua HL, Joshi M, Booth C, Gough A, et al. Establishing a murine model of the hematopoietic syndrome of the acute radiation syndrome. Health Phys 2012. Oct; 103(4):343–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Jackson IL, Gurung G, Poirier Y, Gopalakrishnan M, Cohen EP, Donohue TS, et al. A New Zealand white rabbit model of thrombocytopenia and coagulopathy following total body irradiation across the dose range to induce the hematopoietic-subsyndrome of acute radiation syndrome. Int J Radiat Biol 2021; 97(sup1):S19–S31. [DOI] [PubMed] [Google Scholar]
- 14.MacVittie TJ, Farese AM, Bennett A, Gelfond D, Shea-Donohue T, Tudor G, et al. The acute gastrointestinal subsyndrome of the acute radiation syndrome: a rhesus macaque model. Health Phys 2012. Oct; 103(4):411–26. [DOI] [PubMed] [Google Scholar]
- 15.McAnulty PA. The minipig in biomedical research. Boca Raton: CRC Press; 2012. [Google Scholar]
- 16.Schachtschneider KM, Madsen O, Park C, Rund LA, Groenen MA, Schook LB. Adult porcine genome-wide DNA methylation patterns support pigs as a biomedical model. BMC Genomics 2015. Oct 5; 16:743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Walters EM, Wolf E, Whyte JJ, Mao J, Renner S, Nagashima H, et al. Completion of the swine genome will simplify the production of swine as a large animal biomedical model. BMC Med Genomics 2012. Nov 15; 5:55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Humphray SJ, Scott CE, Clark R, Marron B, Bender C, Camm N, et al. A high utility integrated map of the pig genome. Genome Biol 2007; 8(7):R139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Moroni M, Coolbaugh TV, Lombardini E, Mitchell JM, Moccia KD, Shelton LJ, et al. Hematopoietic radiation syndrome in the Gottingen minipig. Radiat Res 2011. Jul; 176(1):89–101. [DOI] [PubMed] [Google Scholar]
- 20.Kaur A, Ten Have GAM, Hritzo B, Deutz NEP, Olsen C, Moroni M. Morphological and functional impairment in the gut in a partial body irradiation minipig model of GI-ARS. Int J Radiat Biol 2020. Jan; 96(1):112–28. [DOI] [PubMed] [Google Scholar]
- 21.Singh VK, Thrall KD, Hauer-Jensen M. Minipigs as models in drug discovery. Expert Opin Drug Discov 2016. Dec; 11(12):1131–4. [DOI] [PubMed] [Google Scholar]
- 22.Zoetelief J, Hennen LA, Broerse JJ (1984). Some practical aspects of dosimetry and dose specification for whole body irradiation. In: Response of Different Species to Total Body Irradiation. Series in Radiology, vol 10. Springer, Dordrecht. 10.1007/978-94-009-6048-0_2 [DOI] [Google Scholar]
- 23.Farese AM, Cohen MV, Katz BP, Smith CP, Jackson W 3rd, Cohen DM, et al. A nonhuman primate model of the hematopoietic acute radiation syndrome plus medical management. Health Phys 2012. Oct; 103(4):367–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Schaaf GW, Justice JN, Quillen EE, Cline JM. Resilience, aging, and response to radiation exposure (RARRE) in nonhuman primates: a resource review. Geroscience 2023. Dec; 45(6):3371–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Waselenko JK, MacVittie TJ, Blakely WF, Pesik N, Wiley AL, Dickerson WE, et al. Medical management of the acute radiation syndrome: recommendations of the Strategic National Stockpile Radiation Working Group. Ann Intern Med 2004. Jun 15; 140(12):1037–51. [DOI] [PubMed] [Google Scholar]
- 26.FDA. NEUPOGEN® (filgrastim). 2021. [cited 2024 October 24]; Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/103353s5157lbl.pdf. [Google Scholar]
- 27.FDA. NEULASTA® (pegfilgrastim) injection, for subcutaneous use. 2019. [cited 2024 October 24]; Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/125031s198lbl.pdf. [Google Scholar]
- 28.Ishigure N Implementation of the NCRP wound model for interpretation of bioassay data for intake of radionuclides through contaminated wounds. J Radiat Res 2009. May; 50(3):267–76. [DOI] [PubMed] [Google Scholar]
- 29.Farese AM, Cohen MV, Katz BP, Smith CP, Gibbs A, Cohen DM, et al. Filgrastim improves survival in lethally irradiated nonhuman primates. Radiat Res 2013. Jan; 179(1):89–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. .Clayton NP, Khan-Malek RC, Dangler CA, Zhang D, Ascah A, Gains M, et al. Sargramostim (rhu GM-CSF) improves survival of non-human primates with severe bone marrow suppression after acute, high-dose, whole-body irradiation. Radiat Res 2021. Feb 1; 195(2):191–9. [DOI] [PubMed] [Google Scholar]
- 31.FDA. NPLATE® (romiplostim) for injection, for subcutaneous use Food and Drug Administration; 2022. [cited 2024 October 24]; Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/125268s172lbl.pdf. [Google Scholar]
- 32.Hamade DF, Epperly MW, Fisher R, Hou W, Shields D, van Pijkeren JP, et al. Genetically engineered probiotic Limosilactobacillus reuteri releasing IL-22 (LR-IL-22) modifies the tumor microenvironment, enabling irradiation in ovarian cancer. Cancers (Basel) 2024. Jan 23; 16(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Medhora M, Gao F, Gasperetti T, Narayanan J, Khan AH, Jacobs ER, et al. Delayed effects of acute radiation exposure (DEARE) in juvenile and old rats: Mitigation by Lisinopril. Health Phys 2019. Apr; 116(4):529–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Singh VK, Serebrenik AA, Fatanmi OO, Wise SY, Carpenter AD, Janocha BL, et al. The radioprotectant, BIO 300, protects the lungs from total-body irradiation injury in C57L/J mice. Radiat Res 2023. Mar 01; 199(3):294–300. [DOI] [PubMed] [Google Scholar]
- 35.Blumenthal DJ, Sugarman SL, Christensen DM, Wiley AL, Livingston GK, Glassman ES, et al. Role of dicentric analysis in an overarching biodosimetry strategy for use following a nuclear detonation in an urban environment. Health Phys 2014. Apr; 106(4):516–22. [DOI] [PubMed] [Google Scholar]
- 36.International Atomic Energy Agency. Cytogenetic analysis for radiation dose assessment: A manual. Vienna: IAEA; 2001. 127 p. [Google Scholar]
- 37.Fenech M Cytokinesis-block micronucleus cytome assay. Nat Protoc 2007; 2(5):1084–104. [DOI] [PubMed] [Google Scholar]
- 38.Royba E, Repin M, Pampou S, Karan C, Brenner DJ, Garty G. RABiT-II-DCA: A fully-automated dicentric chromosome assay in multiwell plates. Radiat Res 2019; 192(3):311–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. .Repin M, Pampou S, Karan C, Brenner DJ, Garty G. RABiT-II: Implementation of a high-throughput micronucleus biodosimetry assay on commercial biotech robotic systems. Radiat Res 2017; 187(4):502–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Garty G, Chen Y, Turner H, Zhang J, Lyulko OV, Bertucci A, et al. The RABiT: A Rapid Automated BIodosimetry Tool for radiological triage II. Technological developments. Int J Radiat Biol 2011; 87(8):776–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem 1998; 273(10):5858–68. [DOI] [PubMed] [Google Scholar]
- 42.Turner HC, Brenner DJ, Chen Y, Bertucci A, Zhang J, Wang H, et al. Adapting the γ-H2AX assay for automated processing in human lymphocytes. 1. Technological aspects. Radiat Res 2011; 175(3):282–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Turner HC, Sharma P, Perrier JR, Bertucci A, Smilenov L, Johnson G, et al. The RABiT: high-throughput technology for assessing global DSB repair. Radiat Environ Biophys 2014; 53(2):265–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lee Y, Wang Q, Seong KM, Turner HC. High-throughput γ-H2AX assay using imaging flow cytometry. Methods Mol Biol 2023; 2635:123–34. [DOI] [PubMed] [Google Scholar]
- 45.Lee Y, Wang Q, Shuryak I, Brenner DJ, Turner HC. Development of a high-throughput γ-H2AX assay based on imaging flow cytometry. Radiat Oncol 2019; 14(1):150-●●●. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Olive PL. Detection of DNA damage in individual cells by analysis of histone H2AX phosphorylation. Methods Cell Biol 2004; 75:355–73. [DOI] [PubMed] [Google Scholar]
- 47.Wilkins RC, Romm H, Kao T-C, Awa AA, Yoshida MA, Livingston GK, et al. Interlaboratory comparison of the dicentric chromosome assay for radiation biodosimetry in mass casualty events. Radiat Res 2008; 169(5):551–60. [DOI] [PubMed] [Google Scholar]
- 48.Prasanna PG, Escalada ND, Blakely WF. Induction of premature chromosome condensation by a phosphatase inhibitor and a protein kinase in unstimulated human peripheral blood lymphocytes: a simple and rapid technique to study chromosome aberrations using specific whole-chromosome DNA hybridization probes for biological dosimetry. Mutat Res 2000. Mar 23; 466(2):131–41. [DOI] [PubMed] [Google Scholar]
- 49.Royba E, Shuryak I, Ponnaiya B, Repin M, Pampou S, Karan C, et al. Multiwell-based G0-PCC assay for radiation biodosimetry. Sci Rep 2024; 14(1):19789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Garty G, Turner HC, Salerno A, Bertucci A, Zhang J, Chen Y, et al. The decade of the RABiT (2005–15). Radiat Prot Dosimetry 2016; 172(1–3):201–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Repin M, Pampou S, Garty G, Brenner DJ. RABiT-II: A fully-automated micronucleus assay system with shortened time to result. Radiat Res 2019. Mar; 191(3):232–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Repin M, pampou S, Brenner DJ, Garty G. The use of a centrifuge-free RABiT-II system for high-throughput micronucleus analysis. J Radiat Res 2020; 61(1):68–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Repin M, Garty G, Garippa RJ, Brenner DJ. RABiT-III: an automated micronucleus assay at a non-specialized biodosimetry facility. Radiat Res 2024. Mar 22; 201(6):567–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wang Q, Rodrigues MA, Repin M, Pampou S, Beaton-Green LA, Perrier J, et al. Automated triage radiation biodosimetry: Integrating imaging flow cytometry with high-throughput robotics to perform the vytokinesis-block micronucleus assay. Radiat Res 2019. Apr; 191(4):342–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Meadows SK, Dressman HK, Muramoto GG, Himburg H, Salter A, Wei Z, et al. Gene expression signatures of radiation response are specific, durable and accurate in mice and humans. PLoS One 2008. Apr 2; 3(4):e1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Amundson SA. Transcriptomics for radiation biodosimetry: progress and challenges. Int J Radiat Biol 2023; 99(6):925–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ivey RG, Subramanian O, Lorentzen TD, Paulovich AG. Antibody-based screen for ionizing radiation-dependent changes in the Mammalian proteome for use in biodosimetry. Radiat Res 2009. May; 171(5):549–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lee Y, Pujol Canadell M, Shuryak I, Perrier JR, Taveras M, Patel P, et al. Candidate protein markers for radiation biodosimetry in the hematopoietically humanized mouse model. Sci Rep 2018. Sep 10; 8(1):13557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lucas J, Dressman H, Suchindran S, Nakamura M, Chao N, Himburg H, et al. A translatable predictor of human radiation exposure. PLoS One 2014; 9(9):e107897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ghandhi SA, Shuryak I, Ponnaiya B, Wu X, Garty G, Morton SR, et al. Cross-platform validation of a mouse blood gene signature for quantitative reconstruction of radiation dose. Sci Rep 2022. Aug 19; 12(1):14124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Huang TC, Paul S, Gong P, Levicky R, Kymissis J, Amundson SA, et al. Gene expression analysis with an integrated CMOS microarray by time-resolved fluorescence detection. Biosens Bioelectron 2011. Jan 15; 26(5):2660–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sun H, Olsen T, Zhu J, Tao J, Ponnaiya B, Amundson SA, et al. A microfluidic approach to parallelized transcriptional profiling of single cells. Microfluid Nanofluidics 2015; 19(6):1429–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Meng X, Wen K, Zhao J, Han Y, Ghandhi SA, Kaur SP, et al. Microfluidic measurement of intracellular mRNA with a molecular beacon probe towards point-of-care radiation triage. Sens Diagn 2024. Aug 8; 3(8):1344–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Pannkuk E, Laiakis E, Authier S, Wong K, Fornace A. Gas chromatography/mass spectrometry metabolomics of urine and serum from nonhuman primates exposed to ionizing radiation: Impacts on the tricarboxylic acid cycle and protein metabolism. J Proteome Res 2017; 16(5):2091–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Pannkuk EL, Fornace AJ Jr., Laiakis EC. Metabolomic applications in radiation biodosimetry: exploring radiation effects through small molecules. Int J Radiat Biol 2017. Oct; 93(10):1151–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Pannkuk E, Laiakis E, Authier S, Wong K, Fornace A. Targeted metabolomics of nonhuman primate serum after exposure to ionizing radiation: Potential tools for high-throughput biodosimetry. RSC Adv 2016; 6(56):51192–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Laiakis EC, Pannkuk EL, Chauthe SK, Wang YW, Lian M, Mak TD, et al. A serum small molecule biosignature of radiation exposure from total body irradiated patients. J Proteome Res 2017. Oct 6; 16(10):3805–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Goudarzi M, Mak TD, Jacobs JP, Moon BH, Strawn SJ, Braun J, et al. An integrated multi-omic approach to assess radiation injury on the host-microbiome axis. Radiat Res 2016. Sep; 186(3):219–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Ostheim P, Tichy A, Badie C, Davidkova M, Kultova G, Stastna MM, et al. Applicability of gene expression in saliva as an alternative to blood for biodosimetry and prediction of radiation-induced health effects. Radiat Res 2024. May 1; 201(5):523–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Okunieff P, Swarts SG, Fenton B, Zhang SB, Zhang Z, Rice L, et al. Radiation biological toximetry using circulating cell-free DNA (cfDNA) for rapid radiation/nuclear triage. Radiat Res 2024. Jul 1; 202(1):70–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Redon CE, Dickey JS, Bonner WM, Sedelnikova OA. γ-H2AX as a biomarker of DNA damage induced by ionizing radiation in human peripheral blood lymphocytes and artificial skin. Adv Space Res 2009; 43(8):1171–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Redon CE, Nakamura AJ, Gouliaeva K, Rahman A, Blakely WF, Bonner WM. The use of gamma-H2AX as a biodosimeter for total-body radiation exposure in non-human primates. PLoS One 2010. Nov 23; 5(11):e15544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Williams BB, Dong R, Flood AB, Grinberg O, Kmiec M, Lesniewski PN, et al. A deployable in vivo EPR tooth dosimeter for triage after a radiation event involving large populations. Radiat Meas 2011. Sep 1; 46(9):772–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Flood AB, Sidabras JW, Swarts SG, Buehler PW, Schreiber W, Grinberg O, et al. Benefits and challenges of in vivo EPR nail biodosimetry in a second tier of medical triage in response to a large radiation event. Radiat Prot Dosimetry 2023. Sep 18; 199(14):1539–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Pannkuk E, Laiakis E, Mak T, Astarita G, Authier S, Wong K, et al. A lipidomic and metabolomic serum signature from nonhuman primates exposed to ionizing radiation. Metabolomics 2016. May; 12(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Park JG, Paul S, Briones N, Zeng J, Gillis K, Wallstrom G, et al. Developing human radiation biodosimetry models: Testing cross-species conversion approaches using an ex vivo model system. Radiat Res 2017; 187(6):708–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ghandhi SA, Turner HC, Shuryak I, Dugan GO, Bourland JD, Olson JD, et al. Whole thorax irradiation of non-human primates induces persistent nuclear damage and gene expression changes in peripheral blood cells. PLoS One 2018; 13(1):e0191402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ghandhi SA, Morton SR, Shuryak I, Lee Y, Soni RK, Perrier JR, et al. Longitudinal multi-omic changes in the transcriptome and proteome of peripheral blood cells after a 4 Gy total body radiation dose to Rhesus macaques. BMC Genomics 2023. Mar 21; 24(1):139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Royba E, Repin M, Balajee AS, Shuryak I, Pampou S, Karan C, et al. The RABiT-II DCA in the rhesus macaque model. Radiat Res 2020; 196(5):501–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Rudqvist N, Laiakis EC, Ghandhi SA, Kumar S, Knotts JD, Chowdhury M, et al. Global gene expression response in mouse models of DNA repair deficiency after gamma irradiation. Radiat Res 2018. Apr; 189(4):337–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Mukherjee S, Laiakis EC, Fornace AJ Jr., Amundson SA. Impact of inflammatory signaling on radiation biodosimetry: mouse model of inflammatory bowel disease. BMC Genomics 2019. May 2; 20(1):329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ghandhi SA, Smilenov L, Shuryak I, Pujol-Canadell M, Amundson SA. Discordant gene responses to radiation in humans and mice and the role of hematopoietically humanized mice in the search for radiation biomarkers. Sci Rep 2019. Dec 19; 9(1):19434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wang Q, Lee Y, Shuryak I, Pujol Canadell M, Taveras M, Perrier JR, et al. Development of the FAST-DOSE assay system for high-throughput biodosimetry and radiation triage. Sci Rep 2020. Jul 29; 10(1):12716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wang Q, Lee Y, Pujol-Canadell M, Perrier JR, Smilenov L, Harken A, et al. Cytogenetic damage of human lymphocytes in humanized mice exposed to neutrons and X rays 24 h after exposure. Cytogenet Genome Res 2021; 161(6–7):352–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Capaccio C, Perrier JR, Cunha L, Mahnke RC, Lörch T, Porter M, et al. CytoRADx: A high-throughput, standardized biodosimetry diagnostic system based on the cytokinesis-block micronucleus assay. Radiat Res 2021. Nov 1; 196(5):523–34. [DOI] [PubMed] [Google Scholar]
- 86.MRIGlobal. MRIGlobal, Thermo Fisher Scientific, Arizona State University Lead Testing of Diagnostic Medical Countermeasures Program for U.S Department of Health and Human Services’ Office of the Assistant Secretary for Preparedness Response (ASPR). 2024. [cited 2024 10/11/2024]; Available from: https://www.mriglobal.org/mriglobal-in-partnership-with-thermo-fisher-scientific-and-arizona-state-university-to-lead-testing-of-diagnostic-medical-countermeasures-program-for-u-s-department-of-health-and-human-services/. [Google Scholar]
- 87.Iversen ES, McCarthy JM, Burdett KB, Lipton G, Phillips G, Dressman H, et al. Bridging the gaps: using an NHP model to predict single dose radiation absorption in humans. Int J Radiat Biol 2020. Jan; 96(1):47–56. [DOI] [PubMed] [Google Scholar]
- 88.Satyamitra MM, Cassatt DR, Molinar-Inglis O, Rios CI, Taliaferro LP, Winters TA, et al. The NIAID/RNCP biodosimetry program: An overview. Cytogenet Genome Res 2023. Sep 22; 163(3–4):89–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ainsbury E, Badie C, Barnard S, Manning G, Moquet J, Abend M, et al. Integration of new biological and physical retrospective dosimetry methods into EU emergency response plans - joint RENEB and EURADOS inter-laboratory comparisons. Int J Radiat Biol 2017. Jan; 93(1):99–109. [DOI] [PubMed] [Google Scholar]
- 90.Abend M, Amundson SA, Badie C, Brzoska K, Hargitai R, Kriehuber R, et al. Inter-laboratory comparison of gene expression biodosimetry for protracted radiation exposures as part of the RENEB and EURADOS WG10 2019 exercise. Sci Rep 2021. May 7; 11(1):9756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Port M, Barquinero J-F, Endesfelder D, Moquet J, Oestreicher U, Terzoudi G, et al. RENEB inter-laboratory comparison 2021: Inter-assay comparison of eight dosimetry assays. Radiat Res 2023; 199(6):535–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Abend M, Amundson SA, Badie C, Brzoska K, Kriehuber R, Lacombe J, et al. RENEB inter-laboratory comparison 2021: The gene expression assay. Radiat Res Jun 1; 199(6):598–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Amundson SA, Do KT, Shahab S, Bittner M, Meltzer P, Trent J, et al. Identification of potential mRNA biomarkers in peripheral blood lymphocytes for human exposure to ionizing radiation. Radiat Res 2000. Sep; 154(3):342–6. [DOI] [PubMed] [Google Scholar]
- 94.Dressman H, Muramoto G, Chao N, Meadows S, Marshall D, Ginsburg G, et al. Gene expression signatures that predict radiation exposure in mice and humans. PLoS Med 2007. Apr; 4(4):e106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Paul S, Amundson SA. Development of gene expression signatures for practical radiation biodosimetry. Int J Radiat Oncol Biol Phys 2008. Jul; 71(4):1236–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Ghandhi SA, Shuryak I, Morton SR, Amundson SA, Brenner DJ. New Approaches for quantitative reconstruction of radiation dose in human blood cells. Sci Rep 2019. Dec 5; 9(1):18441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Marchetti F, Coleman MA, Jones IM, Wyrobek AJ. Candidate protein biodosimeters of human exposure to ionizing radiation. Int J Radiat Biol 2006. Sep; 82(9):605–39. [DOI] [PubMed] [Google Scholar]
- 98.Ossetrova NI, Blakely WF. Multiple blood-proteins approach for early-response exposure assessment using an in vivo murine radiation model. Int J Radiat Biol 2009; 85(10):837–50. [PubMed] [Google Scholar]
- 99.Acharya SS, Fendler W, Watson J, Hamilton A, Pan Y, Gaudiano E, et al. Serum microRNAs are early indicators of survival after radiation-induced hematopoietic injury. Sci Transl Med 2015. May 13; 7(287):287ra69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Laiakis E, Strassburg K, Bogumil R, Lai S, Vreeken R, Hankemeier T, et al. Metabolic phenotyping reveals a lipid mediator response to ionizing radiation. J Proteome Res 2014. Sep 05; 13(9):4143–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Goudarzi M, Weber W, Mak TD, Chung J, Doyle-Eisele M, Melo D, et al. Development of urinary biomarkers for internal exposure by cesium-137 using a metabolomics approach in mice. Radiat Res 2014; 181(1):54–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Brengues M, Smith S, Estes M, Zenhausern F. Integrated micro-fluidic system for gene expression based biodosimetry. In: Christensen D, Sugarman S, O F, editors. The Medical Basis for Radiation Accident Preparedness V: Advances in Diagnosis & Management of Ionizing Radiation Injuries & Illnesses: Oak Ridge Associated Unoiversities; 2013. p. 363–6. [Google Scholar]
- 103.Lacombe J, Phillips SL, Zenhausern F. Microfluidics as a new tool in radiation biology. Cancer Lett 2016. Feb 28; 371(2):292–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Taraboletti A, Goudarzi M, Kabir A, Moon BH, Laiakis EC, Lacombe J, et al. Fabric phase sorptive extraction - A metabolomic pre-processing approach for ionizing radiation exposure assessment. J Proteome Res 2019. May 15; 18(8):3020–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Akkad AR, Gu J, Duane B, Norquist A, Brenner DJ, Ramakumar A, et al. Automatic reagent handling and assay processing of human biospecimens inside a transportation container for a medical disaster response against radiation. PLoS One 2022; 17(5):e0268508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Satyamitra MM, DiCarlo AL, Taliaferro L. Understanding the pathophysiology and challenges of development of medical countermeasures for radiation-induced vascular/endothelial cell injuries: Report of a NIAID workshop, August 20, 2015. Radiat Res 2016. Aug; 186(2):99–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Wijerathne H, Langston JC, Yang Q, Sun S, Miyamoto C, Kilpatrick LE, et al. Mechanisms of radiation-induced endothelium damage: Emerging models and technologies. Radiother Oncol 2021. May; 158:21–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Griffith JW, Sokol CL, Luster AD. Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu Rev Immunol 2014; 32:659–702. [DOI] [PubMed] [Google Scholar]
- 109.Khakpour S, Wilhelmsen K, Hellman J. Vascular endothelial cell Toll-like receptor pathways in sepsis. Innate Immun 2015. Nov; 21(8):827–46. [DOI] [PubMed] [Google Scholar]
- 110.Kany S, Vollrath JT, Relja B. Cytokines in inflammatory disease. Int J Mol Sci 2019. Nov 28; 20(23). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol 2005. Dec; 5(12):953–64. [DOI] [PubMed] [Google Scholar]
- 112.Janssen WJ, Barthel L, Muldrow A, Oberley-Deegan RE, Kearns MT, Jakubzick C, et al. Fas determines differential fates of resident and recruited macrophages during resolution of acute lung injury. Am J Respir Crit Care Med 2011. Sep 1; 184(5):547–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Kratofil RM, Kubes P, Deniset JF. Monocyte Conversion During Inflammation and Injury. Arterioscler Thromb Vasc Biol 2017. Jan; 37(1):35–42. [DOI] [PubMed] [Google Scholar]
- 114.Dal-Secco D, Wang J, Zeng Z, Kolaczkowska E, Wong CH, Petri B, et al. A dynamic spectrum of monocytes arising from the in situ reprogramming of CCR2+ monocytes at a site of sterile injury. J Exp Med 2015. Apr 6; 212(4):447–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Hollingsworth BA, Cassatt DR, DiCarlo AL, Rios CI, Satyamitra MM, Winters TA, et al. Acute radiation syndrome and the microbiome: Impact and review. Front Pharmacol 2021; 12:643283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Shakyawar SK, Mishra NK, Vellichirammal NN, Cary L, Helikar T, Powers R, et al. A review of radiation-induced alterations of multi-omic profiles, radiation injury biomarkers, and countermeasures. Radiat Res 2023. Jan 1; 199(1):89–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Guo H, Chou WC, Lai Y, Liang K, Tam JW, Brickey WJ, et al. Multi-omics analyses of radiation survivors identify radioprotective microbes and metabolites. Science 2020. Oct 30; 370(6516). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Rosiello RA, Merrill WW. Radiation-induced lung injury. Clin Chest Med 1990. Mar; 11(1):65–71. [PubMed] [Google Scholar]
- 119.Virmani R, Farb A, Carter AJ, Jones RM. Pathology of radiation-induced coronary artery disease in human and pig. Cardiovasc Radiat Med 1999. Jan-Mar; 1(1):98–101. [DOI] [PubMed] [Google Scholar]
- 120.Yarnold J, Brotons MC. Pathogenetic mechanisms in radiation fibrosis. Radiother Oncol 2010. Oct; 97(1):149–61. [DOI] [PubMed] [Google Scholar]
- 121.Zheng M, Liu Z, He Y. Radiation-induced fibrosis: Mechanisms and therapeutic strategies from an immune microenvironment perspective. Immunology 2024. Aug; 172(4):533–46. [DOI] [PubMed] [Google Scholar]
- 122.Wynn TA, Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity 2016. Mar 15; 44(3):450–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Rosenbloom J, Castro SV, Jimenez SA. Narrative review: fibrotic diseases: cellular and molecular mechanisms and novel therapies. Ann Intern Med 2010. Feb 2; 152(3):159–66. [DOI] [PubMed] [Google Scholar]
- 124.Rockey DC, Bell PD, Hill JA. Fibrosis–a common pathway to organ injury and failure. N Engl J Med 2015. Mar 19; 372(12):1138–49. [DOI] [PubMed] [Google Scholar]
- 125.Piera-Velazquez S, Mendoza FA, Jimenez SA. Endothelial to mesenchymal transition (EndoMT) in the pathogenesis of human fibrotic diseases. J Clin Med 2016. Apr 11; 5(4):45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Gibbons MA, MacKinnon AC, Ramachandran P, Dhaliwal K, Duffin R, Phythian-Adams AT, et al. Ly6Chi monocytes direct alternatively activated profibrotic macrophage regulation of lung fibrosis. Am J Respir Crit Care Med 2011. Sep 1; 184(5):569–81. [DOI] [PubMed] [Google Scholar]
- 127.van Amerongen MJ, Harmsen MC, van Rooijen N, Petersen AH, van Luyn MJ. Macrophage depletion impairs wound healing and increases left ventricular remodeling after myocardial injury in mice. Am J Pathol 2007. Mar; 170(3):818–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.

