ArticlePDF AvailableLiterature Review

Commonalities Between COVID-19 and Radiation Injury

Authors:

Abstract and Figures

As the multi-systemic components of COVID-19 emerge, parallel etiologies can be drawn between SARS-CoV-2 infection and radiation injuries. While some SARS-CoV-2-infected individuals present as asymptomatic, others exhibit mild symptoms that may include fever, cough, chills, and unusual symptoms like loss of taste and smell and reddening in the extremities (e.g., ''COVID toes,'' suggestive of microvessel damage). Still others alarm healthcare providers with extreme and rapid onset of high-risk indicators of mortality that include acute respiratory distress syndrome (ARDS), multi-organ hypercoagulation, hypoxia and cardiovascular damage. Researchers are quickly refocusing their science to address this enigmatic virus that seems to unveil itself in new ways without discrimination. As investigators begin to identify early markers of disease, identification of common threads with other pathologies may provide some clues. Interestingly, years of research in the field of radiation biology documents the complex multiorgan nature of another disease state that occurs after exposure to high doses of radiation: the acute radiation syndrome (ARS). Inflammation is a key common player in COVID-19 and ARS, and drives the multi-system damage that dramatically alters biological homeostasis. Both conditions initiate a cytokine storm, with similar pro-inflammatory molecules increased and other anti-inflammatory molecules decreased. These changes manifest in a variety of ways, with a demonstrably higher health impact in patients having underlying medical conditions. The potentially dramatic human impact of ARS has guided the science that has identified many biomarkers of radiation exposure, established medical management strategies for ARS, and led to the development of medical countermeasures for use in the event of a radiation public health emergency. These efforts can now be leveraged to help elucidate mechanisms of action of COVID-19 injuries. Furthermore, this intersection between COVID-19 and ARS may point to approaches that could accelerate the discovery of treatments for both. © 2021 by Radiation Research Society. All rights of reproduction in any form reserved.
Content may be subject to copyright.
RADIATION RESEARCH 195, 1–24 (2021)
0033-7587/21 $15.00
Ó2021 by Radiation Research Society.
All rights of reproduction in any form reserved.
DOI: 10.1667/RADE-20-00188.1
REVIEW
Commonalities Between COVID-19 and Radiation Injury
Carmen I. Rios,
1
David R. Cassatt, Brynn A. Hollingsworth, Merriline M. Satyamitra, Yeabsera S. Tadesse,
Lanyn P. Taliaferro, Thomas A. Winters and Andrea L. DiCarlo
Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy
and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
Rios, C. I., Cassatt, D. R., Hollingsworth, B. A., Satyamitra,
M. M., Tadesse, Y. S., Taliaferro, L. P., Winters, T. A. and
DiCarlo, A. L. Commonalities Between COVID-19 and
Radiation Injury. Radiat. Res. 195, 1–24 (2021).
As the multi-systemic components of COVID-19 emerge,
parallel etiologies can be drawn between SARS-CoV-2
infection and radiation injuries. While some SARS-CoV-2-
infected individuals present as asymptomatic, others exhibit
mild symptoms that may include fever, cough, chills, and
unusual symptoms like loss of taste and smell and reddening
in the extremities (e.g., ‘‘COVID toes,’’ suggestive of
microvessel damage). Still others alarm healthcare provid-
ers with extreme and rapid onset of high-risk indicators of
mortality that include acute respiratory distress syndrome
(ARDS), multi-organ hypercoagulation, hypoxia and car-
diovascular damage. Researchers are quickly refocusing
their science to address this enigmatic virus that seems to
unveil itself in new ways without discrimination. As
investigators begin to identify early markers of disease,
identification of common threads with other pathologies
may provide some clues. Interestingly, years of research in
the field of radiation biology documents the complex multi-
organ nature of another disease state that occurs after
exposure to high doses of radiation: the acute radiation
syndrome (ARS). Inflammation is a key common player in
COVID-19 and ARS, and drives the multi-system damage
that dramatically alters biological homeostasis. Both condi-
tions initiate a cytokine storm, with similar pro-inflamma-
tory molecules increased and other anti-inflammatory
molecules decreased. These changes manifest in a variety
of ways, with a demonstrably higher health impact in
patients having underlying medical conditions. The poten-
tially dramatic human impact of ARS has guided the science
that has identified many biomarkers of radiation exposure,
established medical management strategies for ARS, and led
to the development of medical countermeasures for use in
the event of a radiation public health emergency. These
efforts can now be leveraged to help elucidate mechanisms
of action of COVID-19 injuries. Furthermore, this intersec-
tion between COVID-19 and ARS may point to approaches
that could accelerate the discovery of treatments for
both. Ó2021 by Radiation Research Society
INTRODUCTION
The world is currently in the grip of a global pandemic.
As of September 10, 2020, over 50 million cases of
COVID-19, the disease caused by severe acute respiratory
syndrome coronavirus 2 (SARS-CoV-2), have been report-
ed worldwide. At the forefront of directing research
activities focused on combating COVID-19 is the National
Institute of Allergy and Infectious Diseases (NIAID) within
the U.S. National Institutes of Health (NIH). In April 2020,
the NIAID published the NIAID Strategic Plan for COVID-
19 Research
2
describing NIAID’s efforts to better under-
stand SARS-CoV-2 and to accelerate the development of
safe biomedical tools. The plan is a well-defined document
that focuses on four research priorities: improving funda-
mental knowledge of SARS-CoV-2 and COVID-19;
supporting the development of diagnostics and assays;
characterizing and testing therapeutics; and developing safe
and effective vaccines against SARS-CoV-2. A detailed
research plan for each priority area is described in the
document.
The novelty of the 2019 novel coronavirus disease
(COVID-19) is underscored by the fact that there are no
FDA-approved or licensed therapeutics specific for corona-
viruses. The traditional drug development pathway for
therapeutics (and diagnostic tools) can be a years-long
process with milestones that require extensive resources to
achieve. However, the urgency of the COVID-19 pandemic
emphasizes the need for rapid development and testing of
promising therapeutic and diagnostic candidates. Approach-
es to accelerate the development process are being explored
in other areas of science where overlapping trends can be
1
Address for correspondence: RNCP, DAIT, NIAID, NIH, 5601
Fishers Lane, Room 7A69, Rockville, MD 20892-9828 (20852 for
express mail); email: carmen.rios@nih.gov.
2
NIAID strategic plan for COVID-19 research FY2020–FY2024,
April 22, 2020. (https://bit.ly/3mFnb2e)
1
investigated. Therapeutics developed for other clinical
indications could potentially be repurposed to treat
COVID-19; some of those possibilities are discussed in
this review. Likewise, pathological features and manner of
progression of other indications similar to COVID-19 could
elucidate how to effectively mitigate and treat this disease.
In this review, we describe observed parallels between
COVID-19 and the biological effects of radiation injury that
include immediate and long-term components: the acute
radiation syndrome (ARS) and the delayed effects of acute
radiation exposure (DEARE).
The Radiation and Nuclear Countermeasure Program
(RNCP) within the NIAID is one such program in which the
scientific work is relevant to the COVID-19 response. The
RNCP was initiated in 2004, with the mission of supporting
research to develop medical countermeasures to diagnose
(biodosimetry) and treat radiation injuries leading to ARS
and DEARE in the wake of a radiation public health
emergency. The marked similarities between COVID-19
and radiation injuries described in this review have a major
underpinning: the inflammatory response. Several products
currently in development within the RNCP to treat radiation
exposure operate by targeting inflammation that can lead to
tissue damage. In fact, approaches developed to treat
radiation-induced lung fibrosis and vascular damage merit
investigating and are discussed here in the context of
COVID-19.
TARGETS
Here we highlight some of the systems and immunolog-
ical areas affected by both COVID-19 and acute radiation
exposure, albeit to different levels. In both cases, the result
is a systemic insult that can cause damage to many parts of
the body, including the vascular system, lung, heart,
kidneys, liver, gut, eyes and brain. Regardless of the target
organ, the hyperactivation of the immunogenic pathways
are at the heart of the body’s response to overcome SARS-
CoV-2 and acute radiation exposure (1). Cytokines are
produced by a variety of immune cells (i.e., macrophages, B
lymphocytes, T lymphocytes and mast) and non-immune
cells (i.e., endothelial, fibroblasts and stromal). Under
normal circumstances, cytokines have a short half-life and
act as local mediators within a microenvironment; therefore,
circulating cytokines in the blood are below the limit of
detection of most commercially available assay kits (2).
This complex communication network provides a healthy
immune system with the proper signals to mount a
proportionate response against an infectious agent or
inflammatory stimuli. In other cases, the reaction is so
strong that circulating cytokine levels surge, resulting in a
‘‘cytokine storm’’ (also called hypercytokinemia
3
)oran
overaction of the immune system creating a generalized
inflammatory response that can lead to systemic tissue
damage. The cytokine storm is the nexus between SARS-
CoV-2 infection and radiation exposure; both result in
systemic inflammation that ravages the body (1, 3).
A wealth of early literature has described the cytokine
storm syndrome (CSS) in COVID-19 patients (4–15). For
example, in a study of 50 patients, expression levels of 14 of
48 cytokines studied were associated with disease severity
and progression, with interferon (IFN)-c-induced protein 10
and monocyte chemotactic protein-3 noted as excellent
predictors of disease progression (16). Increases in
granulocyte-macrophage colony-stimulating factor (GM-
CSF) and interleukin-6 (IL-6) due to T-cell overactivation
were also noted (17). Another retrospective study with
3,939 patients shows both mild and severe forms of
COVID-19 disease resulting in changes in circulating
leukocyte subsets and cytokine secretion (8). In particular,
Vaninov et al. noted that persistent high levels of three
cytokines (CXCL10, CCL7 and IL-1 receptor antagonist)
were associated with increased viral load, loss of lung
function, lung injury and a fatal outcome (12). Based on
these kinds of findings of elevated levels of specific
cytokines, in June 2020 the FDA issued an Emergency
Use Authorization (EUA) for an in vitro diagnostic test
based on measuring the circulating IL-6 levels in serum or
plasma for the management of patients with COVID-19
4
(discussed in more detail below). However, emerging
technologies measuring ‘‘cytokine signatures’’ demonstrate
variability across subjects and highlight the need for the
development of personalized treatments based on these data.
As observed with COVID-19, cytokines are also released
by many cells after radiation exposure, including endothe-
lial cells, fibroblasts, immune cells and parenchymal cells.
The interplay and early activation of inflammatory reactions
involving proteins in cytokine cascade, such as fibroblast
growth factor (FGF), transforming growth factor (TGF),
tumor necrosis factor (TNF-a), and interleukins (ILs) is
thought to be responsible for DEARE. Cytokines and
chemokines that attract immune cells and lead to inflam-
mation include IL-1aand IL-6. Inflammatory cells cause
numerous other changes to occur, such as cell death,
promotion of fibrosis and swelling of the tissue. These
cytokines are involved in both early and late reactions, like
the major cytokines in the response of skin cells to ionizing
radiation, and include IL-1, IL-6, tumor necrosis factor
(TNF)-a, transforming growth factor (TGF)-b, which can be
pro-or anti-inflammatory depending on the tissue and
context of release, and the chemokines IL-8 and eotaxin
(18). Cytokines can be broadly grouped as pro-inflamma-
tory cytokines such as TNF-a,IL-1aand b,IL-17;
angiogenic/vascular endothelial growth factor (VEGF),
3
NIH/National Cancer Institute. NCI Dictionaries. Cytokine
storm. (https://bit.ly/35RIp73)
4
U.S. Food and Drug Administration. Letter to Linda McCam-
mack, Senior Regulations Manager, Roche Diagnostics. Emergency
use authorization for Elecsys IL-6. June 2, 2020. (https://bit.ly/
32NzWzG)
2RIOS ET AL.
TNF-aand FGF; anti-inflammatory IL-4, IL-10 and TGF-b;
pro-fibrotic IL-6 and TGF-b; immune IL-2, IL-4 and IL-7;
and hematopoietic CSF1, GM-CSF, IL-3 and EPO (19).
Interestingly, persistence of inflammatory cytokines, che-
mokines and growth factors, such as TGF-b, IFN-c, ET-1,
IL-4, IL-13, lead to pulmonary fibrosis (20). Notably, IL-6,
which appears to be a key player in the response to SARS-
CoV-2, is altered after irradiation as well; however, its role
in radiation-induced lung injury is still unclear (21).
In addition to the cytokine storm, dysregulation of the
renin-angiotensin system (RAS) plays a critical role in the
pathogenesis of COVID-19 (22) and radiation exposure.
The RAS pathway, which regulates the body’s fluid
regulation system, is dependent on angiotensin converting
enzyme (ACE) and ACE2. ACE regulates blood pressure,
water, and sodium levels as well as inflammation,
proliferation and vasoconstriction. ACE2, the target recep-
tor of SARS-CoV-2, is involved in degrading AngII to
produce Ang 1-7 and then further to angiotensin 1-5, which
is the effector peptide (23). ACE2 can be found in a variety
of organs (e.g., heart, kidney, adipose tissue, vascular
smooth muscle cells, brain tissue, testis, gastrointestinal
(GI) tract, etc.). Together ACE/ACE2 promote vasodilation
to reduce blood pressure and maintain homeostasis of the
RAS. ACE2 is expressed throughout the body, so it is no
surprise that it plays many physiological roles, which extend
beyond its ability to reduce blood pressure. These include
cardiac and lung protection by ACE2. A mouse study by
Imai et al. shows that AngII, upregulated by ACE, drives
severe lung failure through the AT1a receptor while ACE2
and the AT2 receptors protect against lung injury. The
group showed that exogenous recombinant human ACE2
reduces acute lung failure in ACE2 knockout and wild-type
mice (24, 25), and impacts GI dysregulation and diabetic
complications (26). As SARS-CoV-2 (27) binds ACE2, it is
possible that ACE2 receptors are downregulated, causing an
imbalance in the RAS (28).
Both cytokine and RAS disequilibria may have implica-
tions in the development of lung and other organ injuries for
both COVID-19 (3, 22, 27) and ionizing radiation exposure
(29, 30), although the interplay is complex and not
completely understood. In general, SARS-CoV-2 infection
(31) and ionizing radiation exposure (31) trigger pro-
inflammatory cytokines (e.g., IFN-a, IFN-c, IL-1b, IL-6,
IL-12, IL-18, IL-33, TNF-a, TGF-b, etc.). In the case of
COVID-19, the lung is the most prevalent initial target and,
in some cases, the injury progresses from pneumonitis (32,
33) to acute respiratory distress syndrome (ARDS). This
heightened chronic inflammatory response creates a pro-
fibrotic environment that yields long-term fibrosis followed
by organ dysfunction. Similarly, exposure to radiation can
also result in lung fibrosis (34–36) as well as injury to other
organs (37–39).
The vasculature is also targeted by SARS-CoV-2, as
evidenced by the prevalence of systemic thrombotic events
(40), endotheliitis (endothelial inflammation) (41)and
‘‘COVID toes’’ (suggesting dysfunction of microvessels
in the extremities) (42, 43). The virus has been shown to
directly infect endothelial cells (41), and their subsequent
dysregulation likely explains the many vascular coagulop-
athies and thromboses that have been noted in COVID-19
patients. Recently, Zamboni (44) described COVID-19 as a
vascular disease, with the endothelial cell emerging as a
potent target for SARS-CoV-2, while the endothelium is
called ‘‘the lynchpin’’ of COVID-19 pathogenesis, orches-
trating the cytokine storm and damage to multiple organs
(45). In severe cases, this dysfunction has led to strokes (46)
and other cardiovascular involvement (47). Similarly, acute
radiation exposure leads to vascular injury (48) by way of
apoptosis and senescence of endothelial cells as well as via
an increase in cell adhesion molecules (49) and dysregu-
lation of coagulation homeostasis (50).
Another common target of SARS-CoV-2 and acute
radiation exposure is the pyroptosis pathway or caspase-1-
dependent programmed cell death of eukaryotic cells.
Pyroptosis is a mechanism by which the body’s innate
immune system clears pathogens and promotes an adaptive
immune response. Caspase-1 initiates inflammation that
results in the formation of plasma-membrane pores in
pathogen-infected cells, enabling water to enter, causing
swelling and osmotic lysis. In addition, caspase-1 promotes
cleavage of chromosomal DNA and nuclear condensation.
Caspase-1 is activated by Toll-like receptors (TLRs) and
NOD-like receptors (NLRs) present on immune cells, such
as macrophages or epithelial cells, which sense extracellular
and intracellular danger signals produced by pathogens or
tissue injury. This cellular communication network results
in an inflammatory cascade initiated by NF-jB, mitogen-
activated protein kinase (MAPK)- and interferon-regulatory
factor (IRF)- dependent pathways, activating IFN-a/b,
TNF-a/b, IL-12, IL-6, IL-8 and pro-IL-1b. The pyroptosis
pathway is also implicated in immune cell death (e.g., in the
spleen) after acute radiation exposure (51, 52). Furthermore,
the NF-jB/TNF-asignaling pathway is also activated in
response to ionizing radiation, resulting in a similar
cytokine cascade. In fact, a recently published study
showed that pyroptosis may play a role in radiation-induced
lung inflammation and fibrosis (53).
BIOMARKERS
Biomarkers in Pathophysiology of COVID-19 and Radiation
Injury
There is a remarkable similarity in the pattern of biomarker
response to SARS-CoV-2 infection and radiation exposure.
Significant changes in hematological, chemical, inflammato-
ry and immune biomarkers are seen in COVID-19 and in
irradiated patients. Although the insult (infection or acute
radiation exposure) is vastly different, the ensuing patholo-
gies converge to multiorgan dysfunction (MOD), resulting in
multiorgan failure (MOF) and mortality. Biomarkers unique
REVIEW 3
to each insult, such as smell dysfunction for COVID-19, or
cytogenetic, genomic or metabolomic approaches for radia-
tion biodosimetry are not discussed here.
Hematology
The classical approach of hematology has emerged as a
valuable tool in predicting outcome as well as stratifying
and management of COVID-19 patients (54–59). For
example, a retrospective analysis compared hemocyte
counts (neutrophils, lymphocytes and platelets; or ‘‘NLP
score’’) and found a strong correlation between NLP score
and COVID-19 disease progression, while another pub-
lished study noted lymphocyte decrease associated with
COVID-19 severity (60, 61). Furthermore, Chen et al.
conducted a retrospective study on 548 patients and noted
that the counts of lymphocytes, T-cell subsets, eosinophils
and platelets decreased markedly, especially in severe/
critical and fatal patients. Increased neutrophil count and
neutrophil-to-lymphocyte ratio were predominant in severe/
critical cases or non-survivors (61). From separate studies, it
was observed that an increase in the neutrophil-to-
lymphocyte ratio (NLR) is an early warning signal for
severe COVID-19 (59, 62, 63). These data are further
supported by findings from a published study of 75 patients
from Suzhuo, China with confirmed COVID-19 infection;
common COVID-19 abnormal hematological indexes on
admission included hyperfibrinogenemia, lymphopenia,
elevation of D-dimer, and leukopenia, which were signif-
icantly different between the mild/moderate and severe
COVID-19 groups. Furthermore, the dynamic change of
NLR and D-dimer level can distinguish severe COVID-19
cases from the mild/moderate (64).
Hematological data could be further refined to interrogate
a single cell population (such as lymphocytes or platelets)
that yielded significant correlation with COVID-19 pro-
gression and severity. A meta-analysis of 3,099 patients
from 24 different studies showed that patients who did not
fare well had low lymphocyte counts (mean difference of –
361.06/ll) compared to patients who had good outcomes
(65). Other researchers have also reported lymphopenia as a
key biomarker in COVID-19 patients (66–68); Terpos et al.
(68) referred to lymphopenia as ‘‘a cardinal laboratory
finding, with prognostic potential,’’ urging for the need for
longitudinal evaluation of parameters to follow the
dynamics of the disease progression. Thrombocytopenia is
also commonly noted among patients hospitalized with
COVID-19 and a low platelet count is associated with
higher mortality (57, 69–74). It is hypothesized that SARS-
CoV-2 infects both bone marrow cells and platelets via the
CD13 receptor, inducing growth inhibition and apoptosis.
This entry disrupts hematopoiesis, resulting in thrombocy-
topenia. Another cause of thrombocytopenia is attributed to
lung injury due to activation, aggregation, and retention of
platelets in the lung. The formation of thrombus at the site
of lung injury leads to decreased platelet production and
increased consumption (75, 76). Interestingly, in most cases
the platelet decreases did not reach a level where
spontaneous bleeding occurred.
As seen with COVID-19 patients, radiation exposure also
results in profound hematological perturbations in humans
as well as irradiated animal models, characterized by
granulocytopenia, lymphopenia and thrombocytopenia
(77–81). Unlike COVID-19, significant decreases in
neutrophils, in addition to other cytopenias, is a hallmark
of ARS (82). The SEARCH (System for Evaluation and
Archiving of Radiation accidents based on Case Histories)
(82) database contains 824 clinical cases from 81 radiation
accidents in 19 countries and allows detailed analysis of the
time course of ARS, with the intention to study all medical
aspects of ARS and to derive medical treatment protocols
for radiation accident victims (MEdical TREatment Proto-
cOLs; METREPOL) (83). The hematopoietic syndrome can
be characterized by granulocyte count kinetics (84). The
Radiation Emergency Medical Management (REMM)
5
website provides guidance on triaging radiation accident
victims based on lymphocyte depletion kinetics. The
kinetics of lymphocyte depletion have been shown to be
directly related to the absorbed radiation dose from 0.5 to 10
Gy (85–87). Furthermore, the ratio of neutrophil to
lymphocyte has been employed to determine the radiation
dose exposure (88–90). Interestingly, platelet depletion
alone has not been used to determine radiation dose,
although thrombocytopenia correlates directly with radia-
tion dose and platelet utilization at sites of active bleeding.
Serum Biomarkers
Patients with moderate and severe COVID-19 showed
significant increase in levels of serum amyloid A (SSA), C-
reactive protein (CRP) which positively correlated to
COVID-19 pneumonia (24), and serum albumin (ALB)
levels (P,0.05) (62). Radiation also induces increases in
the serum and urine proteome. CRP and serum amylase are
commonly elevated after radiation exposure (91–93). In the
criticality accident at Tokaimura, all three patients presented
with elevated serum amylase (94). CRP levels were reported
to correlate with clinical outcome in patients exposed to
radiation during the Chernobyl nuclear accident (95). From
a published study that identified 260 radiation-responsive
proteins (96), Partridge et al.(97) narrowed the panel to IL-
6, IL-1b, TNF-aand TGF-blevels as being strongly
correlated to irradiation, with IL-6 emerging as the best
marker for COVID-19 and acute radiation exposure.
Electrolytes
Electrolyte imbalance with reduced potassium, calcium,
chloride and sodium is observed in patients with COVID-19
5
U.S. Dept. of Health and Human Services. REMM/Radiation
Emergency Medical Management. Guidance on diagnosis and
treatment for healthcare providers. (https://www.remm.nlm.gov/)
4RIOS ET AL.
(72, 76, 98). Pooled analysis of data on serum electrolytes
confirms that hyponatremia, hypokalemia and hypocalce-
mia are associated with COVID-19 severity; however, the
authors cautioned that additional information such as
calcium concentrations, serum albumin levels and the
patients’ fluid status is necessary for accurate interpretation
of laboratory findings (99). Interestingly, the authors draw a
correlation between electrolyte imbalance and progression
of COVID-19 disease and MOF. SARS-CoV-2 binds to its
host receptor, ACE2, and reduces ACE2 expression, leading
to increased angiotensin II, which can cause increased
potassium excretion by the kidneys resulting in hypokale-
mia, while plasma angiotensin II is purported to be a
mediator of ARDS and ensuing MOF seen in a significant
number of COVID-19 patients (100). Another cause of
electrolyte loss in COVID-19 patients is attributed to GI
causes such as diarrhea and emesis (101). The U.S. Centers
for Disease Control (CDC) has described a similar
electrolyte imbalance, accompanied by diarrhea and emesis,
due to damage and disruption of the intestine after radiation
exposure (102, 103).
Immune Biomarkers
Lymphocyte counts provide a rapid snapshot of the
prognosis for patients with COVID-19. Delving into the
lymphocyte subsets provides a clearer understanding of the
patient’s immune status, with both diagnostic and prognos-
tic value (104). In a 103-patient study, a significant decrease
in T-cell populations were reported. In particular, the CD3
þ
,
CD4
þ
, CD8
þ
and NK cell counts dropped, with the CD4
þ
/
CD8
þ
ratio increased in COVID-19 patients compared to
healthy controls (105). In other published studies, a
correlation was noted between the severity of disease and
reduction in T-cell subunits (61, 66) as well as decreases in
B-cell numbers (106).
Persistent changes in the immune system after radiation
exposure are manifested as abnormalities in the lymphoid
populations and function (107). Radiation studies have
documented immediate changes in T-cell subsets (108) and
Bcells(109) linked to radiation dose. In particular,
depletion of CD8
þ
cells has been correlated with absorbed
radiation at low doses (110). In atomic bomb (A-bomb)
survivors, a decrease in CD4 helper T-cell populations,
attenuated T-cell function, as well as an increase in B-cell
populations, which could drive long-term inflammation.
Radiation dose-dependent reduction in CD4
þ
cell popula-
tion has also been reported (111, 112). While these metrics
are not used to monitor disease progression, they are
indications of the continued immune dysfunction observed
in patients after radiation exposure (113).
Vascular Dysfunction
Histological analysis of COVID-19 has shown that the
presence of SARS-CoV-2 within endothelial cells was
associated with clusters of inflammatory cells, suggesting
that infection initiates endotheliitis throughout the entire
human body, leading to systemic macro and microcircula-
tory dysfunction in vascular beds (44). VEGF-D was noted
as an indirect procoagulant biomarker of COVID-19
progression (114) and angiopoietin-2 (a marker for
endothelial activation), was associated with microvascular
dysfunction (115). Vascular abnormalities such as vascular
thickening, detected by thoracic computed tomography
(CT), was reported to be significantly associated with
COVID-19, when compared to non–COVID-19 pneumonia
(59% versus 22%, P,0.001) (116). Ultimately, endotheli-
opathy converges with COVID-19-associated coagulopa-
thies; a recently published study showed significant
elevation in markers of endothelial cell and platelet
activation with mortality strongly correlated to von
Willebrand factor (VWF) antigen and plasma thrombomo-
dulin (117).
Vascular dysfunction resulting from radiation exposure
has also been reported. Of the 28 people who died within 98
days of the Chernobyl criticality incident, deaths were
attributed to skin, GI and lung reactions, but most deaths
were characterized by circulatory problems, with a high
incidence of edema and focal hemorrhages (118). After the
Tokaimura Nuclear Plant accident in Tokai, Japan, Akashi
discussed the possible role of inflammation and hemorrhage
in radiation-induced MOF. In a review of 110 cases
histories of radiation accidents spanning 1945 through
2000, the authors analyzed MOF after total-body irradiation
(TBI) and stated that ‘‘. . .symptomatology of organ system
involvement could be traced not only to the pathophysiol-
ogy of the rapidly turning over cell renewal systems but – of
equal or more importance – to the vascular system and
specifically, to the endothelial components.’’ (119). The
primary target of radiation injury to the vasculature is the
endothelial cell. The acute phase of damage occurs within
hours to weeks postirradiation, and is characterized by
endothelial swelling, vascular permeability and edema,
lymphocyte adhesion and infiltration, and apoptosis (52).
Radiation-induced vascular biomarkers include inflamma-
tory signals, endothelial activation and adhesion markers,
and prothrombic markers (48, 120, 121), similar to those
reported for COVID-19.
As of September 10, 2020, the FDA has authorized EUAs
for more than 150 individual molecular diagnostic tests for
SARS-CoV-2. As a sampling, from February 4, 2020 to
June 18, 2020, a total of 85 tests received EUA
authorization, which included 37 tests for detection of
nucleic acids from SARS-CoV-2 and one antigen diagnostic
test, with required conditions for manufacturers and
authorized laboratories.
6
Similarly, as of mid-June 2020,
the FDA had authorized more than 20 serology biomarker
tests, with the caveat to ‘‘always refer to the complete
instructions for use to put these estimates into the proper
6
U.S. Food and Drug Administration. In vitro diagnostics EUAs.
(https://bit.ly/3mEFODt)
REVIEW 5
context and to understand how to use and interpret these
tests.’’ On June 2, 2020, the FDA authorized the only in
vitro diagnostic test for the management of patients with
COVID-19, which is based on measuring the circulating IL-
6 levels in these patients. In stark contrast, no radiation
biodosimetry test has been cleared/authorized by the FDA.
PATHOLOGY
As more information becomes available, it is increasingly
apparent that COVID-19 is not just a pulmonary affliction,
but a multi-organ disease. Curiously, many symptoms as
well as underlying pathogenesis in this multi-organ injury
caused by SARS-CoV-2 are similar to the multi-organ
injury caused by acute ionizing radiation exposure. In a
nuclear incident, a person’s entire body may be exposed to
large doses of damaging ionizing radiation, while SARS-
CoV-2 can only infect cells co-expressing angiotensin-
converting enzyme 2 and transmembrane serine protease 2
(TMPRSS2). The presence of these proteins on a wide
variety of cell types throughout the body such as airway
epithelial cells, alveolar epithelial cells, lung macrophages
and vascular endothelial cells (122, 123), absorptive
enterocytes of the ileum and colon (124), explains the
widespread damage caused by the infection (124). Indeed,
SARS-CoV-2 RNA has been detected in sputum, nasal
swabs, saliva, feces, blood, tears, urine and cerebrospinal
fluid (125–128). Despite the differences in the initial cause
of injury, systemic inflammation and coagulopathy, includ-
ing disseminated intravascular coagulation (DIC) are
hallmarks of both COVID-19 (123, 129–131) and acute
radiation injury, with pyroptosis (51, 53, 131, 132) and
neutrophil extracellular traps (133, 134) found in both. As
noted in COVID-19 patients, hematopoietic ARS patients
develop lymphopenia, thrombocytopenia and neutropenia
due to bone marrow damage (135), possibly further
contributing to multi-organ damage and failure. These
disease processes found in both COVID-19 and acute and
delayed radiation syndromes may directly cause or further
exacerbate injury and pathogenesis in multiple organ
systems (Fig. 1). In fact, multi-system inflammatory
syndrome in children with COVID-19 has recently been
described and affects a wide range of organs and systems
(136).
Pulmonary disease and symptoms are the most common
presentation of COVID-19 and respiratory failure is the
most common cause of death in those with COVID-19
disease (98, 137), whereas lung damage in irradiated
FIG. 1. Panel A: Schematic showing the human organ systems affected by radiation exposure and comprise
both acute and delayed radiation syndromes (Published and modified with permission; licensed from ClipArt
ETC: https://bit.ly/3mHcyvG). Panel B: A representative list of extrapulmonary symptoms observed in COVID-
19 (SARS-CoV-2 infection) (353). Coronavirus image published and modified, with permission, from the
University of Virginia School of Medicine (https://at.virginia.edu/32MutJv).
6RIOS ET AL.
patients is a later effect (compared to hematological and GI
manifestations). Nonetheless, these injuries can also be
severe and lead to death (138, 139). Pneumonitis and
subsequent drop in blood oxygen levels are seen in COVID-
19 patients (98, 123, 140), as well as after irradiation, which
often progresses to pulmonary fibrosis (36, 139). As with
lung damage seen in COVID-19 patients, fibrosis was also
seen in long-term follow-up of Middle East respiratory
syndrome (MERS) patients (141). An increase in local
neutrophils, cytokines and other immune factors is seen in
COVID-19 patients with lung damage (32, 33), and also in
patients and animal models of acute radiation exposure (34–
36). These factors may also contribute to pneumonitis and
provide further support to the hypothesis that lung injury
seen in COVID–19 patients may progress to lung fibrosis.
While pulmonary symptoms are the most common in
COVID-19 patients, GI symptoms are also common.
Nausea, vomiting and diarrhea are all common symptoms
in both COVID-19 and irradiated patients. In one published
study of 651 patients in China, it was found that 11.4% of
patients experienced at least one GI symptom (142) while in
another study, 5% and 3.8% of hospitalized COVID-19
patients experienced nausea/vomiting and diarrhea, respec-
tively (143). Additionally, SARS-CoV-2 RNA is often
found in stool samples of patients with and without GI
symptoms (144), which is not surprising given that ACE2 is
expressed throughout the intestines and co-expressed with
TMPRSS2 in enterocytes in the ileum and colon (124). In
addition, SARS-CoV-2 was recently found to infect enter-
ocytes in vitro (145). The intestinal damage and symptoms
seen in COVID-19 do not appear to be as extreme as those
observed in GI-ARS, where crypt stem cells die, leading to
loss of GI function and integrity, causing not only nausea,
vomiting and diarrhea, but also hemorrhage, endotoxemia,
bacterial infection and even death (34, 146). However, the
involvement of the gut microbiota should not be overlooked
in either disease process. While the effect of SARS-CoV-2
infection on the gut microbiome is not yet known, a healthy
gut microbiome may have contributed to a successful, but
not overly-inflammatory immune response and expedited
recovery with other respiratory diseases (147). Interestingly,
fecal microbiota transplants have been shown to increase
survival in a lethal irradiation mouse model (148).
Another organ of concern is the heart. Though the
coagulopathy seen in both COVID-19 and ARS may
contribute to cardiomyopathy and circulatory failure, direct
cardiac tissue remodeling is also seen in both disease
processes. Cardiac ischemia, inflammation, fibrosis and
wall thickening have been noted in COVID-19 patients
(149, 150) and after irradiation, though dependent upon
dose and time after irradiation (151, 152). SARS-CoV-2
infection and radiation both increase risk of myocardial
infarction, with one study from China reporting that 7% of
case fatalities had only myocardial damage and circulatory
failure without respiratory failure (137). Studies of A-bomb
survivors have shown that cardiovascular disease risk
increases 14% per Gy of exposure (153). The short- and
long-term effects of cardiac damage from both disease
processes is a concern.
Additionally, symptoms indicating damage to the central
nervous system have been observed in patients with
COVID-19 and those with ARS. Headache, disorientation,
cognitive dysfunction, ataxia, seizures, unconsciousness, as
well as other symptoms have been reported in patients who
received lethal high-dose radiation (135) and in adult and
juvenile COVID-19 patients (126, 154, 155). Radiation
causes vascular damage and inflammation leading to
hemorrhage and edema (156) and can increase risk of
stroke (157). Similarly, brain damage in COVID-19 could
be due to systemic inflammatory response and coagulopa-
thy, leading to stroke and other issues (158), or may be
directly due to infection of brain tissue, as SARS-CoV-2
RNA has been found in cerebrospinal fluid and in brain
tissue after autopsy (159). Brain damage due to SARS-
CoV-2 infection or irradiation can initiate or exacerbate
injury to other organs, including respiratory or circulatory
failure (159). Additionally, there is some evidence that
radiation exposure may cause long-term psychological
issues (160), and given the similarities between radiation-
induced central nervous system inflammation and coagu-
lopathy and that seen in COVID-19, long-term neurological
and psychological effects may be forthcoming.
Several published studies have outlined the cutaneous
manifestations of COVID-19. These symptoms appear at
different time points of the disease progression, either at
onset of disease or after hospitalization (161), and depend
on the severity (mild or severe) of the infection (162–164).
The most common symptoms identified in patients with
mild infections are chilblain-like eruptions (i.e., COVID
fingers or toes), and petechiae/purpuric rashes, while
patients with severe infections experience symptoms such
as acro-ischemia with finger and toe cyanosis, cutaneous
bullae, dry gangrene, chickenpox-like rash and maculopap-
ular lesions (162). Like COVID-19, cutaneous manifesta-
tions from ARS depend on the timepoint and severity of
exposure often assessed using a graded scale set forth by the
National Cancer Institute (165). The most common
symptoms are also the least severe, i.e., acute radiation
dermatitis and mild erythema, and are seen in patients
exposed to low-dose radiation. These complications usually
present within 90 days of radiation exposure (165).
Radiation-induced telangiectasias, keratoses, ulcers, hem-
angiomas, splinter hemorrhages in the distal nail bed,
lentiginous hyperpigmentation and severe subcutaneous
fibrosis may also occur. High-dose radiation exposure leads
to severe symptoms such as moist desquamation and
ulceration (165). Of note, these or similar injuries may
occur in the oral epithelium in both COVID-19 (166, 167),
and after radiation exposure (168). In comparing cutaneous
manifestations related to COVID-19 and ARS, the symp-
toms associated with ARS are more severe and long lasting.
Delayed effects can be seen months to years postirradiation,
REVIEW 7
while lesions due to COVID-19 infection appear to heal
more quickly, usually within a few days (161, 169). On the
other hand, vascular complications associated with COVID-
19 infection closely resemble mild radiation burns seen in
patients that have been exposed to mild (non-lethal) doses
of radiation. In both COVID-19 and mild radiation
cutaneous injury, vascular injury may be further contribut-
ing to the skin injury, and damage to the vasculature in the
upper layers of the skin may be involved (162, 163, 169).
Other symptomatic overlaps between COVID-19 and
radiation exposure include: acute kidney injury (72, 170–
172), whether coagulopathy-related or direct, as renal tubule
cells are a potential target for SARS-CoV-2 (173); liver
injury, though more severe and possibly fibrotic in radiation
hepatitis (174) compared to the usually mild elevation of
aspartate aminotransferase and alanine aminotransferase
levels seen in 14–53% of COVID-19 patients (175); and
conjunctivitis (127, 176, 177), an immediate effect of
radiation with possible long-term effects including macular
degeneration and cataracts (178, 179). Fertility issues have
been seen in irradiated individuals, and there is concern
regarding male fertility in COVID-19 patients as well, as
ACE2 receptors are also expressed in the testis, and some
male patients have reported scrotal discomfort. Nonetheless,
SARS-CoV-2 has yet to be found in semen, and this disease
may still be too new to identify fertility issues (180). High
rates of androgenetic alopecia in hospitalized COVID-19
patients have been documented, leading to the hypothesis
that the use of anti-androgen therapy (flutamide) may be a
possible treatment for COVID-19 patients (181).
Radiation-induced coagulopathies (RIC) are part of the
continuum of the irradiation sequalae (134, 182, 183), with
parallels to DIC. Hemorrhage, a hallmark of DIC, was
reported in 60% of the mortalities in the A-bomb-exposed
population, accompanied by petechial lesions and throm-
bocytopenia (184), and was also observed after (184), as
well as other radiation accidents (185, 186). Prevalence of
prolonged clot formation times, increased levels of
thrombin-antithrombin III (TAT) complex and increased
circulating nucleosome/histone (cNH) levels were noted in
blood from irradiated clinical samples (134). D-dimer has
not been reported in any of the radiation-related coagulop-
athy studies, but other metrics used to predict coagulopathy
for COVID-19 are similar to RIC. Though in many systems
radiation damage is much more severe and chronic, overall,
the similarities noted thus far between COVID-19 and ARS/
DEARE may provide insight into the late effects of
COVID-19, as well as shed light on possible targets for
diagnostics, prognostic markers and therapeutics.
MEDICAL COUNTERMEASURES
As noted, there are clear parallels between radiation
exposure, which is known to act systemically to cause
damage, and COVID-19, which has been implicated in
organ damage ranging from the lung and GI tract to the
heart, brain, kidney and vasculature. In fact, the character-
istics of radiation-induced pneumonitis are similar to SARS-
CoV-2 interstitial pneumonia (187). Therefore, it is not
surprising that there are a number of treatments for radiation
exposure under development that could prove to be
efficacious for COVID-19. Because it is not possible to
‘‘fight’’ radiation in a conventional sense, in the way that it
is possible to develop approaches directly targeting a
pathogen, researchers have relied instead on modifying the
host response to injury, to identify therapies to address
damage caused by exposure to radiation. In many cases,
these approaches have worked by harnessing the body’s
innate immunity, which is often dysregulated by radiation
exposure. These treatments fall into several general
categories, which include anti-oxidants, anti-inflammato-
ries, antibiotics, anti-fibrotics, growth factors, cellular
therapies, and products that target the vasculature or the
RAS. Especially important to emphasize is the ability to
repurpose these kinds of established drugs, some of which
are already in clinical use, to expedite their use in patients
with SARS-Cov-2 infection. These varied approaches will
be considered separately below, according to their mecha-
nisms of action.
Growth Factors
COVID-19 is characterized by damage to the lung and
vasculature, reducing blood oxygenation. In a review of the
effects of erythropoietin (EPO), Ehrenreich et al.(188)
noted that EPO acts on tissues beyond erythropoiesis; these
effects could be brought to bear in fighting SARS-CoV-2
pathology. EPO is produced in the body in response to low
oxygen levels, and in the short term, binds to receptors in
the brain stem to improve mechanical ventilation. EPO also
acts on airways and lung vasculature to reduce inflamma-
tion and promote vascularization and has been shown
clinically to be neuroprotective. Although at this writing, no
trials have been started, a randomized placebo-controlled
trial for proof-of-concept has been proposed. EPO has also
been shown to accelerate the expansion of erythroid
progenitors in mouse irradiation models (189, 190). Galal
et al.(191) described the effects of EPO beyond
erythropoiesis, through the reduction of oxidative stress
via upregulation of anti-inflammatory receptors. These
activities point to possible treatments to reduce inflamma-
tion contributing to radiation-induced GI, lung, or kidney
injury.
Leukinet(sargramostim or granulocyte-macrophage col-
ony-stimulating factor, GM-CSF; Partner Therapeutics Inc.,
Lexington, MA) is one of three leukocyte growth factors
approved by the U.S. FDA for treatment of ARS (192).
Lang et al.(193) noted that, just as immune system
stimulation can either help the body fight a viral infection or
produce a deleterious inflammatory response, administra-
tion or inhibition of GM-CSF may be useful therapies for
COVID-19. Lung macrophages depend on alveolar GM-
8RIOS ET AL.
CSF production for their maintenance, and GM-CSF
administration could provide protection against viral infection
in the early stages of ARDS and promote tissue repair. At this
writing, sargramostim is being proposed as a therapeutic
against COVID-19 in three trials
7
(NCT04400929,
NCT04326920, NCT04411680). As for deleterious effects,
GM-CSF can exacerbate the inflammatory response, driving
lung pathologies such as those resulting from COVID-19. In
this case, GM-CSF inhibition could reduce expression of the
pro-inflammatory cytokines IL-1, IL-6 and TNF, providing a
multi-pronged approach to dampen an overstimulated im-
mune system. Monoclonal antibody treatments targeting GM-
CSF or GM-CSF receptor that are ongoing include: otilimab
(NCT04376684), gimsilumab (NCT04351243), lenzilumab
(NCT04351152), TJM2 (NCT04341116) and mavrilimumab
(NCT04397497, NCT04399980, NCT04447469). Namilu-
mab is being used in the clinic in an expanded access
program. Patients receiving treatment in these experimental
protocols will need to be monitored carefully because of the
role that GM-CSF plays in immunological homeostasis. In
addition, it would appear that the timing of these kinds of
growth factor interventions is critical to their potential
efficacy.
Antioxidant Approaches
Radiation-induced damage is characterized by increases
in reactive oxygen species (ROS) and oxidative stress (194–
196). These increases, and the resulting inflammatory
response, can damage other sensitive tissues (197–200).
The antioxidant N-acetyl cysteine (NAC) has been shown to
mitigate radiation-induced damage to the GI tract and
improve 10- and 30-day survival in mice receiving total-
abdominal irradiation (201). NAC also decreased out-of-
field bone marrow damage and ROS levels, suggesting that
bone marrow damage contributes to some of the radiation-
induced GI injury. Because of similar patterns of tissue
damage, Corry et al. hypothesized that COVID-19-induced
damage to the lung could also be ameliorated by NAC
treatment (202). This hypothesis was further supported by
an earlier finding that NAC treatment of patients with acute
lung injury and ARDS resulted in reduced mortality (203).
Currently, there are several ongoing national clinical trials
to address the possible benefit of NAC treatment in
COVID-19 patients (NCT04374461, NCT04419025,
NCT04370288 and NCT04279197).
Another compound under investigation for COVID-19 is
the histamine H2-receptor antagonist famotidine. Typically
used to treat acid reflux and heartburn, the drug is also
known to have antioxidant activity (204). Although an
unlikely candidate to treat viral disease, famotidine first
came to the attention of researchers interested in repurpos-
ing already-licensed products (205). Generic and off-patent
drugs were of particular interest because of their safety and
affordability, supported by extensive data in humans (206).
In addition, in silico analysis suggested that the drug could
be useful as a therapeutic alternative in COVID-19 (207).
Clinicians have noted that hospitalized patients taking the
drug for other medical indications appeared to recuperate
from COVID-19 better than those who did not take the drug
(208). A review of over 6,000 patient records suggested that
famotidine use led to a death rate of ;14%, compared to
27% for those who had not taken famotidine. This finding
was further supported by a retrospective analysis of
COVID-19 patients who received the drug within 24 h of
hospital admission, which showed that its use reduced the
risk of intubation or death (209). One suggested mechanism
of action of famotidine is a direct action on the receptor,
leading to improved mast cell regulation (210). Additional,
prospective clinical trials to look at the efficacy of the drug
in COVID-19 patients are underway (NCT04370262,
NCT04389567). A recently published article has suggested
that famotidine is ineffective; however, the authors state that
‘‘We’re not challenging that famotidine might help. We’re
saying that the mechanism of action is not antiviral’’.
8
This
is consistent with the primary proposed antioxidant
mechanism of action.
Radiation researchers have also sought to understand the
protective effects of famotidine administration, with regards
to limitation of DNA damage and cellular protection. With
studies performed in vivo, in both pre-clinical irradiation
models as well as in patients undergoing radiotherapy, the
potential benefits of the drug have been demonstrated.
Famotidine was found to be radioprotective in mice that
were administered the drug prior to irradiation, as assessed
by micronuclei formation in cells of the bone marrow (211,
212). Pre-clinical work with mouse leukocytes collected
from irradiated animals documented consistent outcomes,
with reductions in DNA damage in animals treated with
famotidine prior to irradiation (213). Famotidine also
significantly reduced lymphocytopenia in prostate cancer
patients who received the drug a few hours prior to
undergoing radiotherapy (214). In another study, prostate
cancer patients given twice-daily oral doses of famotidine
during their radiotherapy led to a reduction in radiation-
induced injury to the normal rectal tissue (215). These
clinical findings in cancer patients were based on earlier
work using peripheral blood samples taken from normal
healthy volunteers (216). In those studies, blood was
irradiated ex vivo in the presence or absence of vitamin C
and famotidine. Comet assay results suggested that the
presence of famotidine was protective for radiation-induced
apoptosis, with an estimated dose reduction factor of 1.5.
The protective effects of the drug noted above suggest the
drug may have an antioxidant effect and ability to scavenge
7
NIH/U.S. National Library of Medicine. ClinicalTrials.gov.
(https://clinicaltrials.gov/)
8
Saey TH. A popular heartburn medicine doesn’t work as a
COVID-19 antiviral. ScienceNews 2020. (https://bit.ly/2ZSFtDi)
REVIEW 9
free radicals, a mechanism that would justify clinical use of
the drug for COVID-19.
Anti-inflammatory Approaches
Severe COVID-19 is characterized by a cytokine storm,
indicative of an overactive immune response to the infection
(217). Because elevated levels of pro-inflammatory cyto-
kines are associated with high morbidity and mortality,
various approaches to modulate the inflammatory response
have been proposed. Specifically, COVID-19 results in
elevated levels of serum IL-6 (217). The IL-6 receptor
antagonist, tocilizumab (Actemrat, Genentech, San Fran-
cisco, CA), indicated for rheumatoid arthritis, has also been
proposed to ameliorate radiation-induced tissue damage,
and has shown efficacy in diminishing the cytokine storm
resulting from cancer immunotherapy (218). In early
published clinical studies to assess the potential impact of
tocilizumab treatment in COVID-19, Somers et al.(219)
performed a single-site trial of the drug in 154 COVID-19
patients on mechanical ventilation. Treatments resulted in a
lower hazard of death, although the rate of superinfections
increased. Antinori et al.(220) noted the risk of a secondary
Candida infection from tocilizumab treatment, and therefore
suggested that the drug only be used in well-designed
clinical trials. Currently, a tocilizumab treatment arm has
been included as part of the University of Oxford’s
Randomised Evaluation of COVID-19 Therapy (RECOV-
ERY) trial,
9
the results of which have not been made
available at the time of this writing. In a study looking at the
effect of the drug in critically ill patients, treatment led to
improved oxygenation and blood counts (221). In addition,
another anti-IL-6 receptor antibody, sarilumab (Kevzarat;
Regeneron Pharmaceuticals Inc., Tarrytown, NY and
Sanofi, Paris, France) is also under clinical consideration
in more than 10 registered trials as a treatment for late-stage
COVID-19 patients.
Dexamethasone is a generic corticosteroid drug, which is
licensed for a broad range of indications including arthritis,
allergic reactions and immune system disorders. Dexameth-
asone has been shown to reduce multi-organ damage,
including lung injury, in rats that have been exposed to
localized radiation (222). In non-human primate (NHP)
models of radiation-induced lung injury, dexamethasone has
been used as a component of the medical management,
where it is given when there is an increase in the non-
sedated respiratory rate, which suggests respiratory distress
(138, 223, 224). This use of the drug is similar to its clinical
use for dyspnea (225). In these NHP studies, dexametha-
sone treatment reduced the elevated respiratory rate, lung
density, pleural effusion and pneumonitis, leading to
improved outcomes. Dexamethasone treatment has also
been included as an arm in the RECOVERY trial (226),
where its use has been shown to reduce COVID-19
mortality from 40.7% to 29% among patients who required
invasive mechanical ventilation, but did not appear to confer
a benefit to hospitalized patients who received only oxygen.
These data suggest that corticosteroid treatment may only be
effective if the immune system is overstimulated to the
extent that it is causing significant harm. The WHO Rapid
Evidence Appraisal for COVID-19 Therapies Working
Group performed a meta-analysis of seven randomized trials
and concluded that systemic corticosteroids reduced 28-day
all-cause mortality (227). Another treatment that has been
shown to improve survival in a mouse model of radiation-
induced lung injury is BIO 300 (Humanetics Corp., Edina,
MN), which is a nanosuspension of the soy isoflavonoid,
genistein. In mice, BIO 300 administration improved
survival and reduced other morbidities caused by lung
irradiation (228). Although genistein has antiviral activity
(229–231), it may also be useful to prevent radiation-
induced lung damage. Genistein is thought to act through
inactivation of NF-jB(232), and since NF-jB inhibition
has been shown to reduce inflammation in a mouse model
of COVID-19, it could be effective in treating lung
complicationscausedbySARS-CoV-2infection.Hu-
manetics has announced initiation of a clinical trial in
discharged COVID-19 patients, to determine if treatment
with the oral BIO 300 product can reduce late lung fibrosis
and improve quality of life in patients who are recovering
from the infection (NCT04482595).
Given that the autoimmune disease, rheumatoid arthritis
(RA) is caused by an overactive immune response that
targets normal joint tissue, it is not surprising that
approaches that have shown benefit in RA are being
considered for COVID-19. For example, anakinra (Kine-
rett; Swedish Orphan Biovitrum AB, Stockholm, Sweden),
a specific IL-1 receptor antagonist, has been proposed as a
possible treatment. Similarly, anakinra has been reported to
reduce vascular inflammation in a mouse model of radiation
exposure (233). In that study, administration of the drug for
two weeks postirradiation reduced the expression of
inflammatory mediators such as pro-caspase and caspase-
1. In an early case report from Italy, clinicians described a
critically ill patient who was successfully treated with
anakinra (234). Other studies have since followed; in fact,
King et al. have described ten ongoing clinical trials that
target hyper-inflammation. It is clear that many different
dosing regimens are being tested and that most of these
smaller studies (,400 patients) should be considered
preliminary, but data arising from these studies could lead
to larger-scale studies with more uniform treatments. The
outcomes of some of these anakinra studies have been
published. In the anakinra-COVID study performed in
France, 25% of patients who were treated with anakinra
required invasive mechanical ventilation or died, compared
to 44% of historical controls from the same hospital (235).
In another small retrospective cohort study from Italy, part
of the COVID-19 Biobank study, anakinra-treated patients
9
RECOVERY/Randomised Evaluation of COVID-19 Therapy.
Oxford, UK: University of Oxford; 2020. (https://www.recoverytrial.
net/)
10 RIOS ET AL.
showed improvement in respiratory parameters (reduced
need of supplemental oxygen, improved PaO
2
/FiO
2
ratio)
and reductions in the inflammatory marker, C-reactive
protein.
Anti-fibrotic Approaches
Lung inflammation caused by radiation can progress to
fibrosis in later stages of injury, causing shortness of breath
and reduced blood oxygen saturation. Drugs currently
approved to treat lung fibrosis include nintedanib (OFEVt;
Boehringer Ingelheim, Ingelheim am Rhein, Germany) and
pirfenidone (Esbriett; Genentech). Nintedanib is a tyrosine
kinase inhibitor approved for idiopathic pulmonary fibrosis.
Using a mouse model of localized irradiation, researchers
demonstrated that nintedanib protected against long-term
fibrosis, as detected microscopically at 39 weeks postirra-
diation (236). Similarly, studies showed protection in a
mouse model of thoracic irradiation by pirfenidone
treatment (237). In terms of COVID-19 and its progression,
it was noted that patients who experienced severe ARDS
often exhibit later lung fibrosis (238). Although anti-
inflammatory treatments could prevent late-stage disease,
it is not known if this will be the case for COVID-19. For
this reason, the authors propose that anti-fibrotics, such as
those described above, should be studied in clinical trials. In
one clinical study (NCT04338802), patients will be
randomized into a placebo-control or nintedanib treatment
group, and in another, the safety and efficacy of pirfenidone
will be studied in patients with SARS-CoV-2 infection
(NCT04282902). Similarly, imatinib (Gleevect; Novartis,
Basel, Switzerland), licensed for chronic myeloid leuke-
mia,
10
and previously shown to increase the survival time of
irradiated mice by delaying lung disease, has been
suggested as a COVID-19 treatment (239). Several clinical
trials are planned (NCT04357613) or recruiting
(NCT04394416) patients to study the drug as a possible
treatment for the disease.
Pentoxifylline is another drug that could potentially be
repurposed as a treatment for COVID-19. Originally
licensed to treat pain in individuals suffering from
intermittent claudication (peripheral arterial disease),
11
pentoxifylline improves blood flow, thereby increasing
tissue oxygenation. It has also been shown to inhibit
synthesis of pro-inflammatory cytokines, specifically TNF-
a(240). Pentoxifylline has been shown to reduce radiation-
induced fibrovascular injury in animal models (241) and in
the clinic (242). Because of these anti-fibrotic (and anti-
inflammatory) activities, pentoxifylline has been proposed
as a possible preventative of COVID-19 complications
(243) and will be tested in a clinical trial (NCT04433988).
Another driver of pulmonary fibrosis, TGF-b(244),
presents a potential target for the prevention of pulmonary
fibrosis in COVID-19 patients (245). For example, an anti-
sense mRNA product that targets TGF-b2 production, OT-
101 (Mateon Therapeutics, San Francisco, CA), is in phase
3 trials for several cancers, and has been proposed as a
COVID-19 treatment (246). Given the probable involve-
ment of TGF-bin the progression of COVID-19, a
preclinical, anti-TGFbreceptor 1 product, IPW5371
(Innovation Pathways, Palo Alto, CA) may also be a
promising candidate to treat COVID-19-induced lung
fibrosis, as it has previously been shown to reduce fibrosis
and improve survival in a mouse model of radiation
exposure (247).
RAS-Targeted Approaches
In the early stages of the pandemic, it became clear that
one method by which the SARS-CoV-2 virus gained access
to the internal cellular machinery was via the ACE2
receptor, the expression of which is most prevalent on lung
alveolar epithelial cells (248). This finding was similar to
SARS-CoV, which also used the ACE2 receptor to gain
entry into cells (249). Therefore, initial treatments consid-
ered for patients were angiotensin-converting enzyme
inhibitors (ACEIs) or angiotensin II type receptor blockers
(ARBs). Because many drugs, such as angiotensin-convert-
ing enzyme inhibitors (ACEIs), are generic and widely
available, they represent a valuable option in repurposed
drugs. Clear benefits include low cost, wealth of clinical
experience, established human data and minimal side
effects. During the initial stages of the COVID-19 response,
it was thought that individuals currently taking hypertensive
drugs could be at an increased risk of infection, and
therefore, their use should be discontinued in COVID-19
patients (250, 251). In one large retrospective study of over
12,000 patients, the relationship between prior use of ACEIs
and patient outcomes after infection was considered, with
the finding that there was no correlation between prior
hypertension medication use and COVID-19 risk (252).
In addition, it became evident that certain segments of the
population were more likely to have more severe forms of
disease and a propensity to develop ARDS. These
individuals included those with cardiovascular disease,
diabetes and hypertension, all of which have associations
with dysregulated aspects of RAS (253). Still other studies
suggested that these drugs should be considered as a
potential treatment due to their multi-prong effects (e.g.,
anti-inflammatory anti-oxidant and antifibrotic) (254).
Therefore, there remains a need to evaluate both angiotensin
agonists and antagonists for COVID-19 (255). In one study,
COVID-19 patients with hypertension were enrolled, to
explore if the use of ACEI or angiotensin receptor
antagonist treatments would impact the severity and
progression of the infection (256). As a biomarker of
10
Gleevec (imatinib mesylate) tablets for oral use. Prescribing
information. Stein, Switzerland: Novartis Pharma Stein AG; East
Hanover, NJ: Novartis Pharmaceuticals Corp.; 2008. (https://bit.ly/
3kyuqan)
11
Trentalt(pentoxifylline). Reference ID: 3873773. Parsippany,
NJ: Validus Pharmaceuticals LLC; 2016. (https://bit.ly/2ZYT6Rz)
REVIEW 11
efficacy, serum levels of IL-6 and circulating T-cell counts
were also evaluated. Both treatments were found to increase
T cells, decrease viral load and IL-6 levels, and reduce the
severity of the course of the disease. Therefore, the
recommendation was made to maintain ACEI and angio-
tensin receptor blocker treatments in patients with COVID-
19.
Similarly, the radiation community has established the
role that products targeting RAS can play on the progression
of radiation-induced organ injuries, primarily the lung and
kidneys. Many studies have demonstrated the ability of
ACEI products to address radiation-induced lung injuries.
Primarily conducted in rat models of injury, ACEIs were
found to increase survival and decrease lung, kidney and
vascular damage (257, 258). These findings were consistent
across different methods of radiation exposures, such as TBI
plus bone marrow transplant (BMT), whole-thorax lung
irradiation (WTLI), or partial-body irradiation (PBI) with a
percentage of the bone marrow spared using shielding
(259). Mitigation of lung and kidney injuries was
determined through assessment of circulating markers of
renal damage (e.g., blood urea nitrogen and creatinine) and
CT imaging of lungs in irradiated animals. In an irradiated
rat model, captopril and fosinopril both increased survival
after 11 Gy (TBI with BMT) and decreased lung injury
(257). Similarly, lisinopril was found to mitigate kidney
(260) and lung (259) damage after high-dose PBI in adult
rats and improve survival in juvenile and geriatric rats (38).
Enalapril mitigated injury and improved survival in a WTLI
rat model, even when initiated 35 days postirradiation (261).
Captopril, administered in a TBI model, improved survival,
although that benefit was diminished when coupled with
skin trauma (262). Finally, ramipril mitigated radiation-
induced damage to the spinal cord (263). In other preclinical
models of radiation injury, angiotensin (1–7) [A(1–7)], a
component of the RAS mentioned above, has also been
studied for its ability to improve survival in irradiated
rodents (264, 265). These peptides, which have been shown
to alter activity in many cell types, accelerated recovery of
the bone marrow in mice receiving TBI, and also improved
the platelet nadir in the animals (264). In later studies,
angiotensin peptides, even when administered days post-
lethal irradiation, improved mouse survival and reduced
bleeding time (265).
Approaches Targeting the Vasculature
The ability of the virus to directly infect and dysregulate
endothelial cells (41) is the driving force behind vascular
coagulopathies and thromboses observed in COVID-19
patients. In addition to vascular effects resulting from direct
viral infection, COVID-19 patients have been found to have
higher levels of VEGF as compared to healthy controls (266).
Elevated VEGF could further increase vessel permeability,
leading to some of the symptoms noted in patients.
Furthermore, studies have implicated VEGF as a target for
therapeutic intervention in ARDS (267). At the time of this
writing, several clinical trials of COVID-19 patients are being
planned or are recruiting to assess the potential efficacy of
bevacizumab, an anti-VEGF, long-lived, humanized mono-
clonal antibody, as a treatment for COVID-19-associated
ARDS (NCT04275414, NCT04344782, NCT04305106).
Also known as Avastint, the mechanism of action of the
drug is to bind to extracellular VEGF and prevent its
interaction with its receptor on endothelial cells (268).
Radiation exposure has long been known to lead to
vascular impairment, which is believed to explain the multi-
organ dysfunction that it causes (121). Many promising
clinical approaches that target the vasculature could have an
effect on radiation-induced damage. VEGF also represents a
molecule that is involved in radiation exposure, and thus, is
a target for reducing the negative effects. For example,
VEGF levels have been shown to be increased in mice after
irradiation (269, 270), and elevated levels of the growth
factor have been implicated in the development of radiation-
induced necrosis of normal tissue (271). Anti-VEGF
antibodies (bevacizumab) have been shown to mitigate
radiation necrosis in mouse brains (272), and in rats that
underwent gamma-knife radiosurgery (273), and bevacizu-
mab has been used in the clinic for radiation injury, where it
was found to reduce necrosis in nasopharyngeal carcinoma
patients who received radiotherapy (274).
As observations continue to be made in COVID-19
patients, it has become apparent that DIC may be
responsible for many of the complications that have been
seen (275–278). This condition, which is characterized by
the development and circulation of small blood clots, can
lead to the blockage of small vessels. As a follow-on effect,
the abnormal consumption of platelets can, in turn, lead to
thrombocytopenia and hemorrhage (279). In several animal
models of lethal radiation exposure, including ferrets (280)
and Yucatan minipigs (281), there has been evidence of
DIC, both in the early days after irradiation and at the time
of death. It is believed that DIC could be a contributor to
radiation-induced human mortality (50), as hemorrhage at
time of death has been seen clinically in irradiated patients.
A major finding in autopsies of humans who have died from
radiation exposure (282), widespread bleeding in the tissues
often occurs as a result of DIC. Coagulation abnormalities
have also clearly emerged as a key hallmark of COVID-19
infections (283). In addition to thromboses (284), throm-
bocytopenia has also been noted in some patients
experiencing COVID-19 infection. In a study from China
involving over 1,000 patients, 36.2% were thrombocytope-
nic, a finding that was greater in cases that were more
advanced (98). This association of low platelet count with
the infection is also supported by a meta-analysis in which
data from nine studies were examined, involving nearly
1,800 patients (71). Those researchers found that platelet
counts were much lower in patients with severe disease and
concluded that these lowered counts could indicate an
increased mortality risk. It is postulated that infection with
12 RIOS ET AL.
SARS-CoV-2 leads to this dysregulated platelet state via a
number of different causative pathways, including a
reduction in platelet production due to loss of progenitors
and growth inhibition, increased clearance due to evolution
of autoantibodies, and enhanced platelet consumption due
to lung injury, which leads to platelet activation and
formation of microthrombi (284).
Like COVID-19, radiation exposures, especially TBI,
are known to lead to a reduction in platelet levels. This
thrombocytopenia has been postulated to play a major role
in deaths from exposure (282). To address this manifes-
tation of radiation injury, drugs that promote platelet
production and are FDA-approved for other indications
[e.g., immune thrombocytopenic purpura (ITP)] have been
tested to see if they could mitigate damage and improve
survival. This has included preclinical and clinical studies
of drugs such as Nplatet(Amgen, Thousand Oaks, CA)
(6, 285), and Promactat(Novartis) (286, 287). Although
there is a case report documenting use of Promacta to treat
a COVID-19 patient who presented with ITP symptoms
while hospitalized (288) and responded well to the
treatment, it does not appear that these therapies have
been attempted on a broader scale thus far. This may be
due to the delicate balance between thrombocytopenia and
thrombosis in these patients; timing of interventions is
crucial to their efficacy or detriment. According to case
reports, heparin has been administered as a means of
countering hypercoagulation (289), and the International
Society of Thrombosis and Haemostasis is now recom-
mending use of heparin for all COVID-19 patients (290).
Clearly, clinical decisions concerning the use of treatments
that either enhance platelet counts or seek to reduce
clotting are complex, and the use of these kinds of
treatments represents an area of great interest. TP508, a 23
amino acid peptide that is a truncated form of human
prothrombin, has also been shown to mitigate radiation
normal tissue injury and increase survival in a mouse TBI
(LD100/15) model (291). Under study in a number of
other preclinical models [e.g., ischemia (292)and
musculoskeletal injuries (293), and in clinical trials to
address diabetic foot ulcers (294)], TP508 has been shown
to enhance tissue repair by targeting endothelial cells
(292). This product is in early preclinical testing as a
treatment for COVID-19
12
because it targets the vascula-
ture as its primary mechanism of action and has
generalized ability to mitigate tissue damage.
Statins as a Common Treatment for Vascular Injury
Statins represent another area of drug treatment overlap
between radiation and COVID-19 (295, 296). In addition to
an anti-inflammatory effect, statins may modify the entry of
viruses into cells, inducing autophagy of infected cells or
altering activation of the coagulation cascade (297). In silico
studies suggest that statins possess direct antiviral activity
through blocking infectivity (298) and have been shown to
enhance ACE2 levels (299) and protect against ARDS
(300).
In clinical trials of COVID-19 patients, observed benefits
were suggested to outweigh their potential risk (301). A
retrospective study of patients treated with statins for other
indications while hospitalized with COVID-19 showed a
lower risk of mortality (302). In animal models of radiation
injury, statins have demonstrated damage mitigation to
normal tissues. Simvastatin, a HMG-CoA reductase inhib-
itor with widespread clinical use, has been shown to
mitigate lung injury in a high-dose thoracic irradiation
mouse model (303), and protects the GI tract, bone marrow
(304) and salivary glands (305). Similarly, atorvastatin
limits radiation-induced heart damage in a rat model (306)
and kidney injury in mice (308).
Perhaps most significant, in terms of overlap with a
primary mechanism of action involved in COVID-19,
atorvastatin has been shown to induce a protective response
in irradiated human umbilical vein endothelial cells
(HUVECs) (308). Treatment of HUVECs with atorvastatin
decreased radiation-induced cell apoptosis, thought to be
driven by upregulation of thrombomodulin and protein C
activation. Finally, lovastatin, when administered after high-
dose irradiation in a WTLI mouse model, increased
survival, reduced levels of macrophages and lymphocytes
in the lung and decreased collagen (309). Activated protein
C (APC) has been tested for its ability to mitigate radiation
injuries (310) and has now been proposed as a therapy for
the vascular dysfunction and abnormal thrombosis (e.g.,
DIC) observed in COVID-19 patients (311). APC’s ability
to downregulate inflammation and generate thrombin
indicates its potential to reduce inflammation and limit
ischemic injury throughout the body.
Antibiotic Treatments for Radiation or SARS-CoV-2
Infection, beyond Antibacterial Properties
Given the need to rapidly assess potential treatments,
many clinicians have turned to antibiotics, which have the
advantage of safety data and clinical experience. Several
classes of antibiotics, which have activity against secondary
bacterial infections and have anti-inflammatory and antiviral
properties, have been suggested as potential treatments for
COVID-19. In silico modeling and other predictive studies
indicate efficacy of aminoglycoside compounds such as
streptomycin (312); tetracyclines, such as doxycycline (313,
314) and eravacycline (312); macrolide antibiotics, such as
azithromycin (315); streptogramins, such as quinupristin
(316); polyether ionophoric antibiotics, such as salinomycin
(317); and glycopeptides, such as teicoplanin. Teicoplanin,
a drug used to treat Staphylococcus infections, was found to
be effective against MERS and is predicted to also be
12
Chrysalis BioTherapeutics receives funding from the National
Institutes of Health for COVID-19 therapeutic development.
Galveston, TX: Chrysalis BioTherapeutics Inc.; 2020. (https://bit.
ly/2HkvCQm)
REVIEW 13
effective against SARS-CoV-2 (318, 319). There was hope
that azithromycin, in combination with hydroxychloroquine
(320), might be effective in reducing the severity of SARS-
CoV-2 infections (321). The drug, commonly used to treat
respiratory and other infections, is thought to strengthen
interferon-mediated antiviral responses (315). Unfortunate-
ly, clinical studies did not provide evidence of efficacy of
the drug (322). In contrast, some studies suggest limited
benefit from the use of antibiotics, either alone or in
combination (323) in both children and adults with COVID-
19 (324), and other research suggests that the use of some
antibiotics could actually worsen the progression of the
disease (325).
As with COVID-19, the efficacy of antibiotics, outside of
their normal antibacterial impact, have been observed in the
mitigation of radiation normal tissue injuries. In vitro
screening of a broad range of antimicrobial agents was
conducted to determine if any of these molecules could be
used a mitigators of radiation injury. In one screen that used
hematopoietic progenitor cells in a clonogenic survival
assay, tetracycline was identified as a significant mitigator
(326). In another in vitro screen of mouse lymphocytes, two
antibiotic classes, tetracyclines and fluoroquinolones (10
different molecules), were identified as potential radiation
mitigators, which the authors attributed to being separate
from their antibacterial properties. From these potential
mitigators, tetracycline showed efficacy in a TBI mouse
model of survival and further data mining confirmed these
earlier findings (327). The predicted impact of quinolones
on mitigation of radiation injuries was not surprising, given
earlier studies in mice that showed ciprofloxacin, sparflox-
acin and clinafloxacin could enhance colony-forming units
in the bone marrow and white blood cell counts in irradiated
mice (328). Similarly, a published study from 1961 showed
that the presence of chlortetracycline in rodent feed
decreased X-ray killing in mice (329). In more recent
studies, fluoroquinolones, such as ciprofloxacin and levo-
floxacin, as well as doxycycline, and neomycin, were all
found to increase the mean survival time in mice exposed to
lethal doses of radiation. Doxycycline and neomycin also
improved day-30 survival in the animals (330). In other
work, ciprofloxacin, an antibiotic notable for its efficacy in
treating bacterial pneumonia and other infections, was also
shown to enhance survival after radiation exposure in vivo
and in vitro. In studies using peripheral blood mononuclear
cells, ciprofloxacin protected against radiation exposure by
inhibiting p53 phosphorylation and increasing Bcl-2
production (331), and has also been shown to increase
survival in a mouse model of TBI (332).
Cellular Therapies
The development and use of cellular therapies, in both the
oncology clinic (333) and as a radiation treatment (334,
335), has long been an approach of interest. In particular,
mesenchymal stem (or stromal) cells (MSCs) have been
shown to mitigate the effects of radiation-induced lung
injury (336, 337). Stem cells, as well as extracellular
vesicles are also being considered as a means of treating
injuries caused by SARS-CoV-2 (338). Specifically, MSCs
derived from various sources, are under study to address
lung injuries in COVID-19 patients (339). These sources
include the bone marrow (340), umbilical cord (341),
adipose tissue (342), peripheral blood and placenta. Early
reported work indicates that the use of MSCs in patients is
safe, and their use is effective in improving lung functional
outcomes (343). There are more than 30 clinical trials
worldwide in which these cells are being used as COVID-19
treatments (clinicaltrials.gov). In fact, several cellular
therapies that are under active investigation for their
development as radiation normal tissue injury mitigators
are also showing promise as therapies in COVID-19 patient
trials. These trials include those for placental expanded
(PLX) cells
13
and multipotent adult progenitor cells (Multi-
Stemt; AthersystInc., Cleveland, OH).
14
CONCLUSIONS
The majority of SARS-CoV-2 infections are asymptom-
atic or symptomatically mild and do not require hospital-
ization. However, at the time of this writing, there are over
50 million confirmed infections worldwide, with more than
9 million confirmed cases in the U.S. alone, of which
hundreds of thousands have required hospitalization and
over 225,000 have died. While host factors such as
comorbidities, age and possibly genetics are strongly
associated with the severity of disease, it is clear that the
pathology of severe COVID-19 is characterized by a
dysregulated inflammatory response, the so-called ‘‘cyto-
kine storm,’’ along with a thrombotic response involving
elevated D-dimer levels and coagulopathies ranging from
small vessel thrombi to DIC. The cytokine storm is
manifested through high levels of pro-inflammatory cyto-
kines such as IL-1b, IL-6, IL-18, and TNFa.The
cumulative systemic effects of the hyperinflammatory
response and dysregulated thrombotic activity can lead to
multi-organ failure and death. In the current absence of any
effective prophylactic interventions, therapeutics capable of
beneficially altering these dysregulated processes and
supporting recovery are urgently needed.
A striking feature of the immune dysregulation, progres-
sion of disease and mechanisms of organ damage in COVID-
19 is its similarity to the biological responses to ionizing
radiation exposure at doses sufficiently large to cause ARS
(.2 Gy in humans). The similarity of inflammatory
responses and organ damages that are caused by COVID-
13
Pluristem provides 28-day follow up for ventilator-dependent
COVID-19 patients under compassionate use program in Israel and
U.S. Los Angeles, CA: GlobeNewswire, Inc.; 2020. (https://bit.ly/
3cjdzVX)
14
COVID-19 and other viral induced ARDS. Cleveland, OH:
Athersyst, Inc.: 2020. (https://bit.ly/33IsQMe)
14 RIOS ET AL.
19 and radiation offers an opportunity for possible COVID-
19 interventions. There is a wealth of data (detailed above)
and extensive experience from the field of radiation biology
for the development of radiation mitigators that target
radiation-induced dysregulation of inflammation, lung fibro-
sis and vascular damage that may lead to multi-organ failure
and death in a fashion similar to that seen in COVID-19.
Many of these agents are currently at preclinical levels of
development, but several are already licensed as radiation
mitigators and FDA approved for use in humans (Neupo-
gent, Neulastat,Leukinet), or have demonstrated safety in
human clinical trials for other indications, and are therefore
well-poised for possible translation to COVID-19 indications.
Finally, in perhaps the truest definition of overlap between
radiation and COVID-19, is the novel (and yet ‘‘old-school’’)
proposal, that low-dose radiation therapy (LDRT), involving
exposure of the thorax of the patient, may have efficacy in
countering lung infections, including those caused by SARS-
CoV-2 infection. This concept is based on studies on the use
of radiation exposures in the early 1900s, before the advent of
modern antibiotics, in which radiation was used to treat
pneumonias resulting from bacterial or viral infections (344).
This treatment proposal, however, is not without controversy.
Already, numerous editorials and comments have been
published (345–352) and discussion surrounding this treat-
ment modality will undoubtedly continue. Nonetheless,
LDRT is not considered to be a potential mitigator of high-
dose radiation damage to the lung, and therefore this
treatment generally falls outside the scope of the current
review.
While medical expertise in the fields of infectious disease,
pulmonology, immunology, rheumatology and hematology
are critical paths forward in the search for COVID-19
therapeutic interventions, the substantial overlap in patho-
biology between COVID-19 and ARS presents the
possibility of readily translatable, potentially high-impact
pharmacological interventions that were originally evaluat-
ed and/or developed to mitigate radiation injury in humans.
By the same token, it is possible that, given the broad range
of new treatment approaches that are being considered for
possible efficacy in COVID-19 infections, some of these
could one day be repurposed for use as radiation medical
countermeasures.
ACKNOWLEDGMENTS
In Memoriam
A friend and colleague of the NIAID, Col. (ret.) and Dr. David Barillo
sadly passed away due to complications from COVID 19 on August 11,
2020. David was an important mentor and voice in the community in the
area of cutaneous radiation injuries (as the Medical Director of Argentum),
due in part to his career devoted to the care of combat casualties, and as a
lead burn surgeon. His great personality, passion for science and
dedication to saving lives will be missed by all who were fortunate
enough to know him.
Received: August 3, 2020; accepted: September 14, 2020; published
online: October 16, 2020
REFERENCES
1. Wadman M, Couzin-Frankel J, Kaiser J, Matacic C. A rampage
through the body. Science 2020; 368:356–60.
2. Aziz N. Measurement of circulating cytokines and immune-
activation markers by multiplex technology in the clinical setting:
What are we really measuring? For Immunopathol Dis Therap
2015; 6:19–22.
3. Sun X, Wang T, Cai D, Hu Z, Chen J, Liao H, et al. Cytokine
storm intervention in the early stages of COVID-19 pneumonia.
Cytokine Growth Factor Rev 2020; 53:38–42.
4. Mahmudpour M, Roozbeh J, Keshavarz M, Farrokhi S, Nabipour
I. COVID-19 cytokine storm: The anger of inflammation.
Cytokine 2020; 133:155151.
5. Hu B, Huang S, Yin L. The cytokine storm and COVID-19. J
Med Virol 2020; Epub ahead of print.
6. Wong K, Chang PY, Fielden M, Downey AM, Bunin D, Bakke J,
et al. Pharmacodynamics of romiplostim alone and in combina-
tion with pegfilgrastim on acute radiation-induced thrombocyto-
penia and neutropenia in non-human primates. Int J Radiat Biol
2020; 96:155–66.
7. Ragab D, Salah Eldin H, Taeimah M, Khattab R, Salem R, The
COVID-19 cytokine storm; what we know so far. Front Immunol
2020; 11:1446.
8. Wang J, Jiang M, Chen X, Montaner LJ. Cytokine storm and
leukocyte changes in mild versus severe SARS-CoV-2 infection:
Review of 3939 COVID-19 patients in China and emerging
pathogenesis and therapy concepts. J Leukoc Biol 2020; 108:17–
41.
9. England JT, Abdulla A, Biggs CM, Lee AYY, Hay KA, Hoiland
RL, et al. Weathering the COVID-19 storm: Lessons from
hematologic cytokine syndromes. Blood Rev 2020; Epub ahead
of print.
10. Garg S, Garg M, Prabhakar N, Malhotra P, Agarwal R.
Unraveling the mystery of Covid-19 Cytokine storm: From skin
to organ systems. Dermatol Ther 2020; Epub ahead of print.
11. Sultan S, Sultan M. COVID-19 cytokine storm and novel truth.
Med Hypotheses 2020; 144:109875.
12. Vaninov N. In the eye of the COVID-19 cytokine storm. Nat Rev
Immunol 2020; 20:277.
13. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS,
Manson JJ. COVID-19: consider cytokine storm syndromes and
immunosuppression. Lancet 2020; 395:1033–34.
14. Ruscitti P, Berardicurti O, Iagnocco A, Giacomelli R. Cytokine
storm syndrome in severe COVID-19. Autoimmun Rev 2020;
19:102562.
15. Jose RJ, Manuel A. COVID-19 cytokine storm: the interplay
between inflammation and coagulation. Lancet Respir Med 2020;
8:e46–7.
16. Yang Y, Shen C, Li J, Yuan J, Wei J, Huang F, et al. Plasma IP–
10 and MCP-3 levels are highly associated with disease severity
and predict the progression of COVID-19. J Allergy Clin
Immunol 2020; 146:119–127.e.4.
17. Zhou Y, Fu B, Zheng X, Wang D, Zhao C, qi Y, et al. Aberrant
pathogenic GM–CSFþT cells and inflammatory CD14þCD16þ
monocytes in severe pulmonary syndrome patients of a new
coronavirus. bioRxiv 2020. (https://bit.ly/2ZWVFU7)
18. Muller K, Meineke V. Radiation-induced alterations in cytokine
production by skin cells. Exp Hematol 2007; 35:96–104.
19. Ray K, Hudak K, Citrin D, Stick M. Chapter 39 - Biomarkers of
radiation injury and response. In: Gupta RC, editor. Biomarkers
in toxicology. Cambridge, MA: Academic Press; 2014. p. 673–
87.
20. Huang Y, Zhang W, Yu F, Gao F. The cellular and molecular
mechanism of radiation-induced lung injury. Med Sci Monit
2017; 23:3446–50.
21. Lierova A, Jelicova M, Nemcova M, Proksova M, Pejchal J,
REVIEW 15
Zarybnicka L, et al. Cytokines and radiation-induced pulmonary
injuries. J Radiat Res 2018; 59:709–53.
22. Busse LW, Chow JH, McCurdy MT, Khanna AK. COVID-19
and the RAAS–a potential role for angiotensin II? Crit Care 2020;
24:136.
23. Santos RAS, Sampaio WO, Alzamora AC, Motta-Santos D,
Alenina N, Bader M, et al. The ACE2/angiotensin-(1-7)/MAS
axis of the renin-angiotensin system: focus on angiotensin-(1-7).
Physiol Rev 2018; 98:505–53.
24. Cheng H, Wang Y, Wang GQ. Organ-protective effect of
angiotensin-converting enzyme 2 and its effect on the prognosis
of COVID-19. J Med Virol 2020; 92:726–30.
25. Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, et al.
Angiotensin-converting enzyme 2 protects from severe acute lung
failure. Nature 2005; 436:112–6.
26. Gheblawi M, Wang K, Viveiros A, Nguyen Q, Zhong JC, Turner
AJ, et al. Angiotensin-converting enzyme 2: SARS-CoV-2
receptor and regulator of the renin-angiotensin system: celebrat-
ing the 20th anniversary of the discovery of ACE2. Circ Res
2020; 126:1456–74.
27. Zhang X, Li S, Niu S. ACE2 and COVID-19 and the resulting
ARDS. Postgrad Med J 2020; 96:403–07.
28. Wosten-van Asperen RM, Lutter R, Specht PA, Moll GN, van
Woensel JB, van der Loos CM, et al. Acute respiratory distress
syndrome leads to reduced ratio of ACE/ACE2 activities and is
prevented by angiotensin-(1-7) or an angiotensin II receptor
antagonist. J Pathol 2011; 225:618–27.
29. Korystova AF, Kublik LN, Levitman MK, Samokhvalova TV,
Shaposhnikova VV, Korystov YN. Ionizing radiation enhances
activity of angiotensin-converting enzyme in rat aorta. Bull Exp
Biol Med 2018; 165:216–9.
30. Robbins ME, Diz DI. Pathogenic role of the renin-angiotensin
system in modulating radiation-induced late effects. Int J Radiat
Oncol Biol Phys 2006; 64:6–12.
31. Coperchini F, Chiovato L, Croce L, Magri F, Rotondi M. The
cytokine storm in COVID-19: An overview of the involvement of
the chemokine/chemokine-receptor system. Cytokine Growth
Factor Rev 2020; 53:25–32.
32. Costela-Ruiz VJ, Illescas-Montes R, Puerta-Puerta JM, Ruiz C,
Melguizo-Rodriguez L. SARS-CoV-2 infection: The role of
cytokines in COVID-19 disease. Cytokine Growth Factor Rev
2020; 54:62–75.
33. Narasaraju T, Tang BM, Herrmann M, Muller S, Chow VTK,
Radic M. Neutrophilia and NETopathy as key pathologic drivers
of progressive lung impairment in patients with COVID-19. Front
Pharmacol 2020; 11:870.
34. Williams JP, McBride WH. After the bomb drops: a new look at
radiation-induced multiple organ dysfunction syndrome (MODS).
Int J Radiat Biol 2011; 87:851–68.
35. Hong JH, Chiang CS, Tsao CY, Lin PY, McBride WH, Wu CJ.
Rapid induction of cytokine gene expression in the lung after
single and fractionated doses of radiation. Int J Radiat Biol 1999;
75:1421–7.
36. Johnston CJ, Williams JP, Elder A, Hernady E, Finkelstein JN.
Inflammatory cell recruitment following thoracic irradiation. Exp
Lung Res 2004; 30:369–82.
37. MacVittie TJ, Bennett A, Booth C, Garofalo M, Tudor G, Ward
A, et al. The prolonged gastrointestinal syndrome in rhesus
macaques: the relationship between gastrointestinal, hematopoi-
etic, and delayed multi-organ sequelae following acute, poten-
tially lethal, partial-body irradiation. Health Phys 2012; 103:427–
53.
38. 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;
116:529–45.
39. Unthank JL, Miller SJ, Quickery AK, Ferguson EL, Wang M,
Sampson CH, et al. Delayed effects of acute radiation exposure in
a murine model of the H-ARS: Multiple-organ injury consequent
to ,10 Gy total body irradiation. Health Phys 2015; 109:511–21.
40. Cantador E, Nunez A, Sobrino P, Espejo V, Fabia L, Vela L, et
al. Incidence and consequences of systemic arterial thrombotic
events in COVID-19 patients. J Thromb Thrombolysis 2020;
50(3):543–547.
41. Varga Z, Flammer AJ, Steiger P, Haberecker M, Andermatt R,
Zinkernagel AS, et al. Endothelial cell infection and endotheliitis
in COVID-19. Lancet 2020; 395:1417–18.
42. Colmenero I, Santonja C, Alonso-Riano M, Noguera-Morel L,
Hernandez-Martin A, Andina D, et al. SARS-CoV-2 endothelial
infection causes COVID-19 chilblains: histopathological, immu-
nohistochemical and ultrastructural study of 7 paediatric cases. Br
J Dermatol 2020. (https://bit.ly/3iLnUMS)
43. Kanitakis J, Lesort C, Danset M, Jullien D. Chilblain-like acral
lesions during the Covid-19 pandemic (‘‘covid toes’’): histologic,
immunofluorescence and immunohistochemical study of 17
cases. J Am Acad Dermatol 2020; 83:870–5.
44. Zamboni P. COVID-19 as a vascular disease: Lesson learned
from imaging and blood biomarkers. Diagnostics (Basel) 2020;
10:440.
45. Gustafson D, Raju S, Wu R, Ching C, Veitch S, Rathnakumar K,
et al. Overcoming barriers: The endothelium as a linchpin of
coronavirus disease 2019 pathogenesis? Arterioscler Thromb
Vasc Biol 2020; 40:1818–29.
46. Trejo-Gabriel-Galan JM. Stroke as a complication and prognostic
factor of COVID-19. Neurologia 2020; 35:318–22.
47. Bansal M. Cardiovascular disease and COVID-19. Diabetes
Metab Syndr 2020; 14:247–50.
48. Venkatesulu BP, Mahadevan LS, Aliru ML, Yang X, Bodd MH,
Singh PK, et al. Radiation-induced endothelial vascular injury: a
review of possible mechanisms. JACC Basic Transl Sci 2018;
3:563–72.
49. Prabhakarpandian B, Goetz DJ, Swerlick RA, Chen X, Kiani MF.
Expression and functional significance of adhesion molecules on
cultured endothelial cells in response to ionizing radiation.
Microcirculation 2001; 8:355–64.
50. Krigsfeld GS, Kennedy AR. Is disseminated intravascular
coagulation the major cause of mortality from radiation at
relatively low whole body doses? Radiat Res 2013; 180:231–4.
51. Stoecklein VM, Osuka A, Ishikawa S, Lederer MR, Wanke-
Jellinek L, Lederer JA. Radiation exposure induces inflamma-
some pathway activation in immune cells. J Immunol 2015;
194:1178–89.
52. Paris F, Fuks Z, Kang A, Capodieci P, Juan G, Ehleiter D, et al.
Endothelial apoptosis as the primary lesion initiating intestinal
radiation damage in mice. Science 2001; 293:293–7.
53. Gao J, Peng S, Shan X, Deng G, Shen L, Sun J, et al. Inhibition of
AIM2 inflammasome-mediated pyroptosis by Andrographolide
contributes to amelioration of radiation-induced lung inflamma-
tion and fibrosis. Cell Death Dis 2019; 10:957.
54. Kermali M, Khalsa RK, Pillai K, Ismail Z, Harky A. The role of
biomarkers in diagnosis of COVID-19 - A systematic review.
Life Sci 2020; 254:117788.
55. Ponti G, Maccaferri M, Ruini C, Tomasi A, Ozben T. Biomarkers
associated with COVID-19 disease progression. Crit Rev Clin
Lab Sci 2020; 57:389–99.
56. Frater JL, Zini G, d’Onofrio G, Rogers HJ. COVID-19 and the
clinical hematology laboratory. Int J Lab Hematol 2020; 42:S11–
18.
57. Henry BM, de Oliveira MHS, Benoit S, Plebani M, Lippi G.
Hematologic, biochemical and immune biomarker abnormalities
associated with severe illness and mortality in coronavirus disease
2019 (COVID-19): a meta-analysis. Clin Chem Lab Med 2020;
58:1021–8.
58. Ferrari D, Motta A, Strollo M, Banfi G, Locatelli M. Routine
16 RIOS ET AL.
blood tests as a potential diagnostic tool for COVID-19. Clin
Chem Lab Med 2020; 58:1095–9.
59. Fei M, Tong F, Tao X, Wang J. Value of neutrophil-to-
lymphocyte ratio in the classification diagnosis of coronavirus
disease 2019. (Article in Chinese) Zhonghua Wei Zhong Bing Ji
Jiu Yi Xue 2020; 32:554–58.
60. Zheng Y, Zhang Y, Chi H, Chen S, Peng M, Luo L, et al. The
hemocyte counts as a potential biomarker for predicting disease
progression in COVID-19: a retrospective study. Clin Chem Lab
Med 2020; 58:1106–15.
61. Wu H, Zhu H, Yuan C, Yao C, Luo W, Shen X, et al. Clinical
and immune features of hospitalized pediatric patients with
coronavirus disease 2019 (COVID-19) in Wuhan, China. JAMA
Netw Open 2020; 3:e2010895.
62. Xia X, Wen M, Zhan S, He J, Chen W. An increased neutrophil/
lymphocyte ratio is an early warning signal of severe COVID-19.
(Article in Chinese). Nan Fang Yi Ke Da Xue Xue Bao 2020;
40:333–6.
63. Yang AP, Liu JP, Tao WQ, Li HM. The diagnostic and predictive
role of NLR, d–NLR and PLR in COVID-19 patients. Int
Immunopharmacol 2020; 84:106504.
64. Fu J, Kong J, Wang W, Wu M, Yao L, Wang Z, et al. The clinical
implication of dynamic neutrophil to lymphocyte ratio and D-
dimer in COVID-19: A retrospective study in Suzhou China.
Thromb Res 2020; 192:3–8.
65. Huang I, Pranata R, Lymphopenia in severe coronavirus disease-
2019 (COVID-19): systematic review and meta-analysis. J
Intensive Care 2020; 8:36.
66. Liu J, Li S, Liu J, Liang B, Wang X, Wang H, et al. Longitudinal
characteristics of lymphocyte responses and cytokine profiles in
the peripheral blood of SARS-CoV-2 infected patients. EBioMe-
dicine 2020; 55:102763.
67. Hou H, Zhang B, Huang H, Luo Y, Wu S, Tang G, et al. Using
IL-2R/lymphocytes for predicting the clinical progression of
patients with COVID-19. Clin Exp Immunol 2020; 201:76–84.
68. Terpos E, Ntanasis-Stathopoulos I, Elalamy I, Kastritis E,
Sergentanis TN, Politou M, et al. Hematological findings and
complications of COVID-19. Am J Hematol 2020; 95:834–47.
69. Yang X, Yang Q, Wang Y, Wu Y, Xu J, Yu Y, et al.
Thrombocytopenia and its association with mortality in patients
with COVID-19. J Thromb Haemost 2020; 18:1469–72.
70. Liu Y, Sun W, Guo Y, Chen L, Zhang L, Zhao S, et al.
Association between platelet parameters and mortality in
coronavirus disease 2019: Retrospective cohort study. Platelets
2020; 31:490–6.
71. Lippi G, Plebani M, Henry BM. Thrombocytopenia is associated
with severe coronavirus disease 2019 (COVID-19) infections: A
meta-analysis. Clin Chim Acta 2020; 506:145–8.
72. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical
features of patients infected with 2019 novel coronavirus in
Wuhan, China. Lancet 2020; 395:497–506.
73. Ahmed MZ, Khakwani M, Venkatadasari I, Horgan C, Giles H,
Jobanputra S, et al. Thrombocytopenia as an initial manifestation
of COVID-19; case series and literature review. Br J Haematol
2020; 189:1057–8.
74. Amgalan A, Othman M. Hemostatic laboratory derangements in
COVID-19 with a focus on platelet count. Platelets 2020:1–6.
75. Pilaczynska-Cemel M, Gołda R, Da
˛
browska A, Przybylski G.
Analysis of the level of selected parameters of inflammation,
circulating immune complexes, and related indicators (neutrophil/
lymphocyte, platelet/lymphocyte, CRP/CIC) in patients with
obstructive diseases. Cent Eur J Immunol 2019; 44:292–8.
76. Ciaccio M, Agnello L. Biochemical biomarkers alterations in
coronavirus disease 2019 (COVID-19). Diagnosis (Berl) 2020;
Epub ahead of print.
77. MacVittie TJ, Farese AM, Jackson W 3rd. The hematopoietic
syndrome of the acute radiation syndrome in rhesus macaques: A
systematic review of the lethal dose response relationship. Health
Phys 2015; 109:342–66.
78. Bilko NM, Dyagil IS, Russu IZ, Bilko DI. Circulating
hematopoietic progenitor cells in patients affected by Chernobyl
accident. Exp Oncol 2016; 38:242–4.
79. Snell FM, Neel JV, Ishibashi K. Hematologic studies in
Hiroshima and a control city two years after the atomic bombing.
Arch Intern Med 1949; 84:569–604.
80. LeRoy GV. The medical sequelae of the atomic bomb explosion.
JAMA 1947; 134:1143–8.
81. Fliedner TM. Nuclear terrorism: the role of hematology in coping
with its health consequences. Curr Opin Hematol 2006; 13:436–
44.
82. Fliedner TM, Graessle D, Meineke V, Dorr H. Pathophysiolog-
ical principles underlying the blood cell concentration responses
used to assess the severity of effect after accidental whole-body
radiation exposure: an essential basis for an evidence-based
clinical triage. Exp Hematol 2007; 35:8–16.
83. Friesecke I, Beyrer K, Wedel R, Reimers K, Fliedner TM.
SEARCH: a system for evaluation and archiving of radiation
accidents based on case histories. Radiat Environ Biophys 2000;
39:213–7.
84. Graessle DH, Dorr H, Bennett A, Shapiro A, Farese AM,
MacVittie TJ, et al. Comparing the hematopoetic syndrome time
course in the NHP animal model to radiation accident cases from
the database search. Health Phys 2015; 109:493–501.
85. Goans RE, Holloway EC, Berger ME, Ricks RC, Early dose
assessment in criticality accidents. Health Phys 2001; 81:446–9.
86. Azizova TV, Osovets SV, Day RD, Druzhinina MB, Sumina
MV, Pesternikova VS, et al. Predictability of acute radiation
injury severity. Health Phys 2008; 94:255–63.
87. 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; 140:1037–51.
88. Graessle DH, Fliedner TM. Computer-assisted severity of effect
assessment of hematopoietic cell renewal after radiation exposure
based on mathematical models. Health Phys 2010; 98:282–9.
89. Blakely W, Ossetrova N, Manglapus G, Salter C, Levine I,
Jackson W, et al. Amylase and blood cell-count hematological
radiation-injury biomarkers in a rhesus monkey radiation model–
use of multiparameter and integrated biological dosimetry. Radiat
Meas 2007; 42:1164–70.
90. Sullivan JM, Prasanna PG, Grace MB, Wathen LK, Wallace RL,
Koerner JF, et al. Assessment of biodosimetry methods for a
mass-casualty radiological incident: medical response and
management considerations. Health Phys 2013; 105:540–54.
91. Sproull M, Kramp T, Tandle A, Shankavaram U, Camphausen K.
Serum amyloid A as a biomarker for radiation exposure. Radiat
Res 2015; 184:14–23.
92. Ossetrova N, Sandgren D, Blakely W. C-reactive protein and
serum amyloid A as early-phase and prognostic indicators of
acute radiation exposure in nonhuman primate total-body
irradiation model. Radiation Meas 011; 46:1019–24.
93. Prasanna PG, Blakely WF, Bertho J-M, Chute JP, Cohen EP,
Goans RE, et al. Synopsis of partial-body radiation diagnostic
biomarkers and medical management of radiation injury
workshop. Radiat Res 2010; 173:245–53.
94. Akashi M, Hirama T, Tanosaki S, Kuroiwa N, Nakagawa K,
Tsuji H, et al. Initial symptoms of acute radiation syndrome in the
JCO criticality accident in Tokai-mura. J Radiat Res 2001;
42:S157–66.
95. Mal’tsev V, Ivanov A, Mikhailov V, Mazurik V. The individual
prognosis of the gravity and of the outcome of acute radiation
disease based on immunological indexes. (Article in Russian).
Radiats Biol Radioecol 2006; 46:152–8.
96. Marchetti F, Coleman MA, Jones IM, Wyrobek AJ. Candidate
REVIEW 17
protein biodosimeters of human exposure to ionizing radiation.
Int J Radiat Biol 2006; 82:605–39.
97. Partridge MA, Chai Y, Zhou H, Hei TK. High-throughput
antibody-based assays to identify and quantify radiation-respon-
sive protein biomarkers. Int J Radiat Biol 2010; 86:321–8.
98. Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, et al.
Clinical characteristics of coronavirus disease 2019 in China. N
Engl J Med 2020; 382:1708–20.
99. Lippi G, South AM, Henry BM. Electrolyte imbalances in
patients with severe coronavirus disease 2019 (COVID-19). Ann
Clin Biochem 2020; 57:262–5.
100. Kuba K, Imai Y, Rao S, Gao H, Guo F, Guan B, et al. A crucial
role of angiotensin converting enzyme 2 (ACE2) in SARS
coronavirus–induced lung injury. Nat Med 2005; 11:875–79.
101. Pan L, Mu M, Yang P, Sun Y, Wang R, Yan J, et al. Clinical
characteristics of COVID-19 patients with digestive symptoms in
Hubei, China: A descriptive, cross-sectional, multicenter study.
Am J Gastroenterol 2020; 115:766–73.
102. Zhang K, Yin L, Zhang M, Parker MD, Binder HJ, Salzman P, et
al. Radiation decreases murine small intestinal HCO3- secretion.
Int J Radiat Biol 2011; 87:878–88.
103. Zseboek ZB, Petranyi G Jr. Effect of the protective action of Aet
on sodium-water metabolism disorders in the so-called gastroin-
testinal radiation syndrome. (Article in German) Strahlentherapie
1964; 125:449–55.
104. Ganji A, Farahani I, Khansarinejad B, Ghazavi A, Mosayebi G.
Increased expression of CD8 marker on T-cells in COVID-19
patients. Blood Cells Mol Dis 2020; 83:102437.
105. Jiang M, Guo Y, Luo Q, Huang Z, Zhao R, Liu S, et al. T-cell
subset counts in peripheral blood can be used as discriminatory
biomarkers for diagnosis and severity prediction of coronavirus
disease 2019. J Infect Dis 2020; 222:198–202.
106. Xu B, Fan CY, Wang AL, Zou YL, Yu YH, He C, et al.
Suppressed T cell-mediated immunity in patients with COVID-
19: A clinical retrospective study in Wuhan, China. J Infect 2020;
81:e51–60.
107. Kusunoki Y, Hayashi T. Long-lasting alterations of the immune
system by ionizing radiation exposure: Implications for disease
development among atomic bomb survivors. Int J Radiat Biol
2008; 84:1–14.
108. Farese AM, Hankey KG, Cohen MV, MacVittie TJ. Lymphoid
and myeloid recovery in rhesus macaques following total body x-
irradiation. Health Phys 2015; 109:414–26.
109. Chambers KA, Harrington NP, Ross WM, Filion LG. Relative
alterations in blood mononuclear cell populations reflect radiation
injury in mice. Cytometry 1998; 31:45–52.
110. Vokurkova D, Sinkora J, Vavrova J, Rezacova M, Knizek J,
Ostereicher J. CD8þnatural killer cells have a potential of a
sensitive and reliable biodosimetric marker in vitro. Physiol Res
2006; 55:689–98.
111. Kusunoki Y, Akiyama M, Kyoizumi S, Bloom ET, Makinodan T.
Age-related alteration in the composition of immunocompetent
blood cells in atomic bomb survivors. Int J Radiat Biol 1988;
53:189–98.
112. Kusunoki Y, Kyoizumi S, Yamaoka M, Kasagi F, Kodama K,
Seyama T. Decreased proportion of CD4 T cells in the blood of
atomic bomb survivors with myocardial infarction. Radiat Res
1999; 152:539–43.
113. Dainiak N. Hematologic consequences of exposure to ionizing
radiation. Exp Hematol 2002; 30:513–28.
114. Kong Y, Han J, Wu X, Zeng H, Liu J, Zhang H. VEGF-D: a
novel biomarker for detection of COVID-19 progression. Crit
Care 2020; 24:373.
115. Smadja DM, Guerin CL, Chocron R, Yatim N, Boussier J,
Gendron N, et al. Angiopoietin-2 as a marker of endothelial
activation is a good predictor factor for intensive care unit
admission of COVID-19 patients. Angiogenesis 2020:1–10.
116. Qanadli SD, Beigelman-Aubry C, Rotzinger DC. Vascular
changes detected with thoracic CT in coronavirus disease
(COVID-19) might be significant determinants for accurate
diagnosis and optimal patient management. Am J Roentgenol
2020; 215:W15.
117. Goshua G, Pine AB, Meizlish ML, Chang CH, Zhang H, Bahel P,
et al. Endotheliopathy in COVID-19-associated coagulopathy:
evidence from a single-centre, cross-sectional study. Lancet
Haematol 2020; 7:e575–82.
118. United Nations Scientific Committee on the Effects of Atomic
Radiation (UNSCEAR), Report to the General Assembly. Annex
G: Early effects in man of high doses of radiation. New York:
United Nations; 1988.
119. Fliedner TM, Dorr HD, Meineke V. Multi-organ involvement as
a pathogenetic principle of the radiation syndromes: a study
involving 110 case histories documented in SEARCH and
classified as the bases of haematopoietic indicators of effect.
Brit J Radiol 2005; S27:1–8.
120. Baselet B, Sonveaux P, Baatout S, Aerts A. Pathological effects
of ionizing radiation: endothelial activation and dysfunction. Cell
Mol Life Sci 2019; 76:699–728.
121. 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; 186:99–111.
122. Abobaker A, Raba AA, Alzwi A. Extrapulmonary and atypical
clinical presentations of COVID-19. J Med Virol 2020; Epub
ahead of print.
123. Tay MZ, Poh CM, Renia L, MacAry PA, Ng LFP. The trinity of
COVID-19: immunity, inflammation and intervention. Nat Rev
Immunol 2020; 20:363–74.
124. Zhang H, Kang Z, Gong H, Xu D, Wang J, Li Z, et al. Digestive
system is a potential route of COVID-19: an analysis of single-
cell coexpression pattern of key proteins in viral entry process.
Gut 2020; 69:1010–8.
125. Wang W, Xu Y, Gao R, Lu R, Han K, Wu G, et al. Detection of
SARS-CoV-2 in different types of clinical specimens. JAMA
2020; 323:1843–4.
126. Moriguchi T, Harii N, Goto J, Harada D, Sugawara H, Takamino
J, et al. A first case of meningitis/encephalitis associated with
SARS-Coronavirus-2. Int J Infect Dis 2020; 94:55–8.
127. Karimi S, Arabi A, Shahraki T, Safi S. Detection of severe acute
respiratory syndrome Coronavirus-2 in the tears of patients with
coronavirus disease 2019. Eye 2020; 34:1220–3.
128. Chan KH, Poon LL, Cheng VC, Guan Y, Hung IF, Kong J, et al.
Detection of SARS coronavirus in patients with suspected SARS.
Emerg Infect Dis 2004; 10:294–9.
129. Price LC, McCabe C, Garfield B, Wort SJ. Thrombosis and
COVID-19 pneumonia: the clot thickens! Eur Respir J 2020;
56:2001608.
130. Giannis D, Ziogas IA, Gianni P. Coagulation disorders in
coronavirus infected patients: COVID-19, SARS-CoV-1, MERS-
CoV and lessons from the past. J Clin Virol 2020; 127:104362.
131. Leonard-Lorant I, Delabranche X, Severac F, Helms J, Pauzet C,
Collange O, et al. Acute pulmonary embolism in COVID-19
patients on CT angiography and relationship to D-Dimer levels.
Radiol 2020:201561.
132. Cookson BT, Brennan MA. Pro-inflammatory programmed cell
death. Trends Microbiol 2001; 9:113–4.
133. Middleton EA, He XY, Denorme F, Campbell RA, Ng D,
Salvatore SP, et al. Neutrophil extracellular traps (NETs)
contribute to immunothrombosis in COVID-19 acute respiratory
distress syndrome. Blood 2020; 136:1169–79.
134. Kennedy AR, Maity A, Sanzari JK. A review of radiation-
induced coagulopathy and new findings to support potential
18 RIOS ET AL.
prevention strategies and treatments. Radiat Res 2016; 186:121–
40.
135. Christensen DM, Iddins CJ, Parrillo SJ, Glassman ES, Goans RE.
Management of ionizing radiation injuries and illnesses, part 4:
acute radiation syndrome. J Am Osteopath Assoc 2014; 114:702–
11.
136. Simpson JM, Newburger JW. Multi-system inflammatory
syndrome in children in association with COVID-19. Circulation
2020; Epub ahead of print.
137. Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical predictors of
mortality due to COVID-19 based on an analysis of data of 150
patients from Wuhan, China. Intensive Care Med 2020; 46:846–
8, Erratum 1294–7.
138. Garofalo M, Bennett A, Farese AM, Harper J, Ward A, Taylor-
Howell C, et al. The delayed pulmonary syndrome following
acute high-dose irradiation: a rhesus macaque model. Health Phys
2014; 106:56–72.
139. Hanania AN, Mainwaring W, Ghebre YT, Hanania NA, Ludwig
M. Radiation-induced lung injury: assessment and management.
Chest 2019; 156:150–62.
140. Bosmuller H, Traxler S, Bitzer M, Haberle H, Raiser W, Nann D,
et al. The evolution of pulmonary pathology in fatal COVID-19
disease: an autopsy study with clinical correlation. Virchows
Arch 2020; 477:349–57.
141. Spagnolo P, Balestro E, Aliberti S, Cocconcelli E, Biondini D,
Casa GD, et al. Pulmonary fibrosis secondary to COVID-19: a
call to arms? Lancet Respir Med 2020; 8:750–2.
142. Jin X, Lian JS, Hu JH, Gao J, Zheng L, Zhang YM, et al.
Epidemiological, clinical and virological characteristics of 74
cases of coronavirus-infected disease 2019 (COVID-19) with
gastrointestinal symptoms. Gut 2020; 69:1002–9.
143. Guan W-j, Ni Z-y, Hu Y, Liang W-h, Ou C-q, He J-x, et al.
Clinical characteristics of coronavirus disease 2019 in China. N
Engl J Med 2020; 382:1708–20.
144. Chen Y, Chen L, Deng Q, Zhang G, Wu K, Ni L, et al. The
presence of SARS-CoV-2 RNA in the feces of COVID-19
patients. J Med Virol 2020; 92:833–40.
145. Lamers MM, Beumer J, van der Vaart J, Knoops K, Puschhof J,
Breugem TI, et al. SARS-CoV-2 productively infects human gut
enterocytes. Science 2020; 369:50–4.
146. Somosy Z, Horvath G, Telbisz A, Rez G, Palfia Z. Morphological
aspects of ionizing radiation response of small intestine. Micron
2002; 33:167–78.
147. Dhar D, Mohanty A. Gut microbiota and Covid-19- possible link
and implications. Virus Res 2020; 285:198018.
148. Cui M, Xiao H, Li Y, Zhou L, Zhao S, Luo D, et al. Faecal
microbiota transplantation protects against radiation-induced
toxicity. EMBO Mol Med 2017; 9:448–61.
149. Babapoor-Farrokhran S, Gill D, Walker J, Rasekhi RT,
Bozorgnia B, Amanullah A. Myocardial injury and COVID-19:
Possible mechanisms. Life Sci 2020; 253:117723.
150. Guzik TJ, Mohiddin SA, Dimarco A, Patel V, Savvatis K,
Marelli-Berg FM, et al. COVID-19 and the cardiovascular
system: implications for risk assessment, diagnosis, and treatment
options. Cardiovasc Res 2020; 116:1666–87.
151. Leiper AD. Late effects of total body irradiation. Arch Dis Child
1995; 72:382–5.
152. Bazyka OD, Belyi DO. Cardiovascular diseases and systolic
function of left ventricle in clean up workers of Chernobyl
accident (based on 30 years follow up). Probl Radiac Med
Radiobiol 2017; 22:292–305.
153. Shimizu Y, Pierce DA, Preston DL, Mabuchi K. Studies of the
mortality of atomic bomb survivors. Report 12, part II. Noncancer
mortality: 1950–1990. Radiat Res 1999; 152:374–89.
154. Mao L, Jin H, Wang M, Hu Y, Chen S, He Q, et al. Neurologic
manifestations of hospitalized patients with coronavirus disease
2019 in Wuhan, China. JAMA Neurol 2020; 77:683–90.
155. Rogers JP, Chesney E, Oliver D, Pollak TA, McGuire P, Fusar-
Poli P, et al. Psychiatric and neuropsychiatric presentations
associated with severe coronavirus infections: a systematic
review and meta-analysis with comparison to the COVID-19
pandemic. Lancet Psychiatry 2020; 7:611–27.
156. Price RE, Langford LA, Jackson EF, Stephens LC, Tinkey PT,
Ang KK. Radiation-induced morphologic changes in the rhesus
monkey (Macaca mulatta) brain. J Med Primatol 2001; 30:81–7.
157. Chen H, Li X, Zhang X, Xu W, Mao F, Bao M, et al. Late
delayed radiation-induced cerebral Arteriopathy by high-resolu-
tion magnetic resonance imaging: a case report. BMC Neurol
2019; 19:232.
158. Helms J, Kremer S, Merdji H, Clere-Jehl R, Schenck M,
Kummerlen C, et al. Neurologic Features in Severe SARS-CoV-2
Infection. N Engl J Med 2020; 382:2268–70.
159. Baig AM, Sanders EC. Potential neuroinvasive pathways of
SARS-CoV-2: Deciphering the spectrum of neurological deficit
seen in coronavirus disease-2019 (COVID-19). J Med Virol
2020; Epub ahead of print.
160. Loganovsky KN, Loganovskaja TK. Schizophrenia spectrum
disorders in persons exposed to ionizing radiation as a result of
the Chernobyl accident. Schizophr Bull 2000; 26:751–73.
161. Lopez M, Bell K, Annaswamy T, Juengst S, Ifejika N. COVID-
19 Guide for the rehabilitation clinician: a review of non-
pulmonary manifestations and complications. Am J Phys Med
Rehabil 2020; 99:669–73.
162. Wollina U, Karadag AS, Rowland-Payne C, Chiriac A, Lotti T.
Cutaneous signs in COVID-19 patients: A review. Dermatol Ther
2020:e13549.
163. Recalcati S. Cutaneous manifestations in COVID-19: a first
perspective. J Eur Acad Dermatol Venereol 2020; 34:e212–3.
164. Marzano AV, Cassano N, Genovese G, Moltrasio C, Vena GA.
Cutaneous manifestations in patients with COVID-19: A
preliminary review of an emerging issue. Br J Dermatol 2020;
183:431–42.
165. Bray FN, Simmons BJ, Wolfson AH, Nouri K. Acute and chronic
cutaneous reactions to ionizing radiation therapy. Dermatol Ther
(Heidelb) 2016; 6:185–206.
166. Vieira AR. Oral manifestations in coronavirus disease 2019
(COVID-19). Oral Dis 2020; Epub ahead of print.
167. Martin Carreras-Presas C, Amaro Sanchez J, Lopez-Sanchez AF,
Jane-Salas E, Somacarrera Perez ML. Oral vesiculobullous
lesions associated with SARS-CoV-2 infection. Oral Dis 2020;
Epub ahead of print.
168. Maria OM, Eliopoulos N, Muanza T. Radiation-induced oral
mucositis. Front Oncol 2017; 7:89.
169. DiCarlo AL, Bandremer AC, Hollingsworth BA, Kasim S,
Laniyonu A, Todd NF, et al. Workshop report. Cutaneous
radiation injuries: models, assessment and treatments. Radiat Res
2020; 194(3):315–344.
170. Chu KH, Tsang WK, Tang CS, Lam MF, Lai FM, To KF, et al.
Acute renal impairment in coronavirus-associated severe acute
respiratory syndrome. Kidney Int 2005; 67:698–705.
171. Cogan MG, Arieff AI. Radiation nephritis and intravascular
coagulation. Clin Nephrol 1978; 10:74–8.
172. Peter RU, Braun-Falco O, Birioukov A, Hacker N, Kerscher M,
Peterseim U, et al. Chronic cutaneous damage after accidental
exposure to ionizing radiation: the Chernobyl experience. J Am
Acad Dermatol 1994; 30:719–23.
173. Xia S, Wu M, Chen S, Zhang T, Ye L, Liu J, et al. Long term
culture of human kidney proximal tubule epithelial cells
maintains lineage functions and serves as an ex vivo model for
coronavirus associated kidney injury. Virol Sin 2020:1–10.
174. Kim J, Jung Y. Radiation-induced liver disease: current
understanding and future perspectives. Exp Mol Med 2017;
49:e359.
175. Su TH, Kao JH. The clinical manifestations and management of
REVIEW 19
COVID-19-related liver injury. J Formos Med Assoc 2020;
119:1016–8.
176. Hempel M, Hinkelbein W. Eye sequelae following external
irradiation. Recent Results Cancer Res 1993; 130:231–6.
177. Pinard CL, Mutsaers AJ, Mayer MN, Woods JP. Retrospective
study and review of ocular radiation side effects following
external-beam Cobalt-60 radiation therapy in 37 dogs and 12 cats.
Can Vet J 2012; 53:1301–7.
178. Gottlober P, Steinert M, Weiss M, Bebeshko V, Belyi D,
Nadejina N, et al. The outcome of local radiation injuries: 14
years of follow-up after the Chernobyl accident. Radiat Res 2001;
155:409–16.
179. Fedirko P, Babenko T, Kolosynska O, Dorichevska R, Garkava
N, Grek L, et al. Morphometric parameters of retinal macular
zone in reconvalescents of acute radiation sickness (in remote
period). Probl Radiac Med Radiobiol 2018; 23:481–9.
180. Verma S, Saksena S, Sadri-Ardekani H. ACE2 receptor
expression in testes: implications in coronavirus disease 2019
pathogenesis. Biol Reprod 2020; 103:449–51.
181. Wambier CG, Vano-Galvan S, McCoy J, Gomez-Zubiaur A,
Herrera S, Hermosa-Gelbard A, et al. Androgenetic alopecia
present in the majority of patients hospitalized with COVID-19:
The ‘‘Gabrin sign’’. J Am Acad Dermatol 2020; 83:680–2.
182. Krantz S, Lober M. Review: hemorrhagic diathesis resulting from
acute exposure to ionizing radiation. (Article in German). Folia
Haematol Int Mag Klin Morphol Blutforsch 1977; 104:1–10.
183. Pospisil J. Postirradiation haemorrhagic syndrome. Acta Univ
Carol Med Monogr 1987; 99:1–187.
184. Liebow AA, Warren S, DeCoursey E. Pathology of atomic bomb
casualties. Am J Pathol 1949; 25:853.
185. Valverde N, Cordeiro J, Oliveira A, Brandao Mello C. The acute
radiation syndrome in the 137 Cs Brazilian accident, 1987. In:
Ricks RC, Fry SA, editors. The medical basis for radiation
accident preparedness. II: Clinical experience and follow up since
1979. New York: Elsevier North Holland Inc.; 1990. p. 89–107.
186. Andrews GA. Criticality accidents in Vinca, Yugoslavia, and Oak
Ridge, Tennessee: Comparison of radiation injuries and results of
therapy. JAMA 1962; 179:191–7.
187. Ippolito E, Fiore M, Greco C, D’Angelillo RM, Ramella S.
COVID-19 and radiation induced pneumonitis: Overlapping
clinical features of different diseases. Radiother Oncol 2020;
148:201–2.
188. Ehrenreich H, Weissenborn K, Begemann M, Busch M, Vieta E,
Miskowiak KW. Erythropoietin as candidate for supportive
treatment of severe COVID-19. Mol Med 2020; 26:58.
189. Peslak SA, Wenger J, Bemis JC, Kingsley PD, Koniski AD,
McGrath KE, et al. EPO-mediated expansion of late-stage
erythroid progenitors in the bone marrow initiates recovery from
sublethal radiation stress. Blood 2012; 120:2501–11.
190. Drouet M, Grenier N, Herodin F. Revisiting emergency anti-
apoptotic cytokinotherapy: erythropoietin synergizes with stem
cell factor, FLT-3 ligand, trombopoietin and interleukin-3 to
rescue lethally-irradiated mice. Eur Cytokine Netw 2012; 23:56–
63.
191. Galal SM, Abdel-Rafei MK, Hasan HF. Cholinergic and
cytoprotective signaling cascades mediate the mitigative effect
of erythropoietin on acute radiation syndrome. Can J Physiol
Pharmacol 2018; 96:442–58.
192. Singh VK, Seed TM. An update on sargramostim for treatment of
acute radiation syndrome. Drugs Today (Barc) 2018; 54:679–93.
193. Lang FM, Lee KM, Teijaro JR, Becher B, Hamilton JA. GM-
CSF-based treatments in COVID-19: reconciling opposing
therapeutic approaches. Nat Rev Immunol 2020:1–8.
194. Avti PK, Pathak CM, Kumar S, Kaushik G, Kaushik T, Farooque
A, et al. Low dose gamma-irradiation differentially modulates
antioxidant defense in liver and lungs of Balb/c mice. Int J Radiat
Biol 2005; 81:901–10.
195. Biaglow JE, Ayene IS, Koch CJ, Donahue J, Stamato TD, Mieyal
JJ, et al. Radiation response of cells during altered protein thiol
redox. Radiat Res 2003; 159:484–94.
196. Hernandez-Flores G, Go´mez-Contreras PC, Dominguez-Rodri-
guez JR, Lerma-Diaz JM, Ortiz-Lazareno PC, Cervantes-
Munguia R, et al. Gamma-irradiation induced apoptosis in
peritoneal macrophages by oxidative stress. Implications of
antioxidants in caspase mitochondrial pathway. Anticancer Res
2005; 25:4091–100.
197. Koturbash I, Rugo RE, Hendricks CA, Loree J, Thibault B,
Kutanzi K, et al. Irradiation induces DNA damage and modulates
epigenetic effectors in distant bystander tissue in vivo. Oncogene
2006; 25:4267–75.
198. Parsons WB, Jr., Watkins CH, Pease GL, Childs DS, Jr. Changes
in sternal marrow following roentgen-ray therapy to the spleen in
chronic granulocytic leukemia. Cancer 1954; 7:179–89.
199. Petrovic N, Perovic J, Karanovic D, Todorovic L, Petrovic V.
Abscopal effects of local fractionated X-irradiation of face and
jaw region. Strahlentherapie 1982; 158:40–2.
200. Robin HI, AuBuchon J, Varanasi VR, Weinstein AB. The
abscopal effect: demonstration in lymphomatous involvement of
kidneys. Med Pediatr Oncol 1981; 9:473–6.
201. Jia D, Koonce NA, Griffin RJ, Jackson C, Corry PM. Prevention
and mitigation of acute death of mice after abdominal irradiation
by the antioxidant N-acetyl-cysteine (NAC). Radiat Res 2010;
173:579–89.
202. Corry PM, Griffin RJ. A radiation mitigator as a potential
treatment for COVID-19. Radiat Res 2020; 193:505.
203. Moradi M, Mojtahedzadeh M, Mandegari A, Soltan-Sharifi MS,
Najafi A, Khajavi MR, et al. The role of glutathione-S-transferase
polymorphisms on clinical outcome of ALI/ARDS patient treated
with N-acetylcysteine. Respir Med 2009; 103:434–41.
204. Ahmadi A, Ebrahimzadeh MA, Ahmad-Ashrafi S, Karami M,
Mahdavi MR, Saravi SS. Hepatoprotective, antinociceptive and
antioxidant activities of cimetidine, ranitidine and famotidine as
histamine H2 receptor antagonists. Fundam Clin Pharmacol
2011; 25:72–9.
205. Kandeel M, Abdelrahman AHM, Oh-Hashi K, Ibrahim A,
Venugopala KN, Morsy MA, et al. Repurposing of FDA-
approved antivirals, antibiotics, anthelmintics, antioxidants, and
cell protectives against SARS-CoV-2 papain-like protease. J
Biomol Struct Dyn 2020:1–8.
206. Rogosnitzky M, Berkowitz E, Jadad AR. Delivering benefits at
speed through real-world repurposing of off-patent drugs: the
COVID-19 pandemic as a case in point. JMIR Public Health
Surveill 2020; 6:e19199.
207. Ortega JT, Serrano ML, Jastrzebska B. Class A G protein-
coupled receptor antagonist famotidine as a therapeutic alterna-
tive against SARS-CoV2: an in silico analysis. Biomolecules
2020; 10:954.
208. Borrell B. New York clinical trial quietly tests heartburn remedy
against coronavirus. Science 2020. (https://bit.ly/3cgcsXg)
209. Freedberg DE, Conigliaro J, Wang TC, Tracey KJ, Callahan MV,
Abrams JA, et al. Famotidine use is associated with improved
clinical outcomes in hospitalized COVID-19 patients: a propen-
sity score matched retrospective cohort study. Gastroenterology
2020; 159:1129–31.e3.
210. Malone RW, Tisdall P, Fremont-Smith P, Liu Y, Huang XP,
White KM, et al. COVID-19: famotidine, histamine, mast cells,
and mechanisms. Res Sq 2020; Preprint. (https://bit.ly/3iLpUEV)
211. Naeeji A, Mozdarani H, Shabestani Monfared A, Faeghi F,
Ahmadi AA, Gholami M, et al. Oral administration of vitamin C,
cimetidine and famotidine on micronuclei induced by low dose
radiation in mouse bone marrow cells. J Biomed Phys Eng 2017;
7:117–26.
212. Zangeneh M, Mozdarani H, Mahmoudzadeh A. Potent radiopro-
tective effects of combined regimens of famotidine and vitamin C
20 RIOS ET AL.
against radiation-induced micronuclei in mouse bone marrow
erythrocytes. Radiat Environ Biophys 2015; 54:175–81.
213. Mozdarani H, Nasirian B, Haeri SA. In vivo gamma-rays induced
initial DNA damage and the effect of famotidine in mouse
leukocytes as assayed by the alkaline comet assay. J Radiat Res
2007; 48:129–34.
214. Razzaghdoust A, Mozdarani H, Mofid B, Aghamiri SM, Heidari
AH. Reduction in radiation-induced lymphocytopenia by famo-
tidine in patients undergoing radiotherapy for prostate cancer.
Prostate 2014; 74:41–7.
215. Razzaghdoust A, Mozdarani H, Mofid B. Famotidine as a
radioprotector for rectal mucosa in prostate cancer patients treated
with radiotherapy: phase I/II randomized placebo-controlled trial.
Strahlenther Onkol 2014; 190:739–44.
216. Mozdarani H, Ghoraeian P. Modulation of gamma-ray-induced
apoptosis in human peripheral blood leukocytes by famotidine
and vitamin C. Mutat Res 2008; 649:71–8.
217. Ye Q, Wang B, Mao J. The pathogenesis and treatment of the
‘Cytokine Storm’ in COVID-19. J Infect 2020; 80:607–13.
218. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M,
et al. Current concepts in the diagnosis and management of
cytokine release syndrome. Blood 2014; 124:188–95.
219. Somers EC, Eschenauer GA, Troost JP, Golob JL, Gandhi TN,
Wang L, et al. Tocilizumab for treatment of mechanically
ventilated patients with COVID-19. Clin Infect Dis 2020:ciaa954.
220. Antinori S, Bonazzetti C, Gubertini G, Capetti A, Pagani C,
Morena V, et al. Tocilizumab for cytokine storm syndrome in
COVID-19 pneumonia: an increased risk for candidemia?
Autoimmun Rev 2020; 19:102564.
221. Carvalho V, Turon R, Goncalves B, Ceotto V, Kurtz P, Righy C.
Effects of tocilizumab in critically ill patients with COVID-19: a
quasi-experimental study. medRxiv 2020. (https://bit.ly/
33MtLLF)
222. Geraci JP, Mariano MS, Jackson KL, Taylor DA, Still ER.
Effects of dexamethasone on late radiation injury following
partial-body and local organ exposures. Radiat Res 1992;
129:61–70.
223. Thrall KD, Mahendra S, Jackson MK, Jackson W 3rd, Farese
AM, MacVittie TJ. A comparative dose-response relationship
between sexes for mortality and morbidity of radiation-induced
lung injury in the rhesus macaque. Health Phys 2019; 116:354–
65.
224. MacVittie TJ, Farese AM, Parker GA, Jackson W 3rd. The time
course of radiation-induced lung injury in a nonhuman primate
model of partial-body irradiation with minimal bone marrow
sparing: clinical and radiographic evidence and the effect of
neupogen administration. Health Phys 2019; 116:366–82.
225. Hui D, Kilgore K, Frisbee-Hume S, Park M, Tsao A, Delgado
Guay M, et al. Dexamethasone for dyspnea in cancer patients: a
pilot double-blind, randomized, controlled trial. J Pain Symptom
Manage 2016; 52:8–16.e1.
226. Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L,
et al. Dexamethasone in hospitalized patients with COVID-19 -
preliminary report. N Engl J Med 2020; Epub ahead of print.
227. WHO Rapid Evidence Appraisal for COVID-19 Therapies
Working Group, Sterne JAC, Murthy S, Diaz JV, Slutsky AS,
Villar J, et al. Association between administration of systemic
corticosteroids and mortality among critically ill patients with
COVID-19: a meta-analysis. JAMA 2020; 324(13):1330–1341.
228. 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;
174:4738–50.
229. Andres A, Donovan SM, Kuhlenschmidt MS. Soy isoflavones
and virus infections. J Nutr Biochem 2009; 20:563–9.
230. Kolokoltsov AA, Adhikary S, Garver J, Johnson L, Davey RA,
Vela EM. Inhibition of Lassa virus and Ebola virus infection in
host cells treated with the kinase inhibitors genistein and
tyrphostin. Arch Virol 2012; 157:121–7.
231. Vela EM, Knostman KA, Mott JM, Warren RL, Garver JN, Vela
LJ, et al. Genistein, a general kinase inhibitor, as a potential
antiviral for arenaviral hemorrhagic fever as described in the
Pirital virus-Syrian golden hamster model. Antiviral Res 2010;
87:318–28.
232. Gong L, Li Y, Nedeljkovic-Kurepa A, Sarkar FH. Inactivation of
NF-kappaB by genistein is mediated via Akt signaling pathway in
breast cancer cells. Oncogene 2003; 22:4702–9.
233. Christersdottir T, Pirault J, Gistera A, Bergman O, Gallina AL,
Baumgartner R, et al. Prevention of radiotherapy-induced arterial
inflammation by interleukin-1 blockade. Eur Heart J 2019;
40:2495–503.
234. Filocamo G, Mangioni D, Tagliabue P, Aliberti S, Costantino G,
Minoia F, et al. Use of anakinra in severe COVID-19: a case
report. Int J Infect Dis 2020; 96:607–9.
235. Huet T, Beaussier H, Voisin O, Jouveshomme S, Dauriat G,
Lazareth I, et al. Anakinra for severe forms of COVID-19: a
cohort study. Lancet Rheumatol 2020; 2:e393–400.
236. De Ruysscher D, Granton PV, Lieuwes NG, van Hoof S, Wollin
L, Weynand B, et al. Nintedanib reduces radiation-induced
microscopic lung fibrosis but this cannot be monitored by CT
imaging: A preclinical study with a high precision image-guided
irradiator. Radiother Oncol 2017; 124:482–7.
237. Qin W, Liu B, Yi M, Li L, Tang Y, Wu B, et al. Antifibrotic
agent pirfenidone protects against development of radiation-
induced pulmonary fibrosis in a murine model. Radiat Res 2018;
190:396–403.
238. George PM, Wells AU, Jenkins RG. Pulmonary fibrosis and
COVID-19: the potential role for antifibrotic therapy. Lancet
Respir Med 2020; 8:807–15.
239. Bernal-Bello D, Jaenes-Barrios B, Morales-Ortega A, Ruiz-
Giardin JM, Garcia-Bermudez V, Frutos-Perez B, et al. Imatinib
might constitute a treatment option for lung involvement in
COVID-19. Autoimmun Rev 2020; 19:102565.
240. Zabel P, Schade FU, Schlaak M. Inhibition of endogenous TNF
formation by pentoxifylline. Immunobiology 1993; 187:447–63.
241. Okunieff P, Augustine E, Hicks JE, Cornelison TL, Altemus RM,
Naydich BG, et al. Pentoxifylline in the treatment of radiation-
induced fibrosis. J Clin Oncol 2004; 22:2207–13.
242. Patel V, McGurk M. Use of pentoxifylline and tocopherol in
radiation-induced fibrosis and fibroatrophy. Br J Oral Maxillofac
Surg 2017; 55:235–41.
243. Assimakopoulos SF, Seintis F, Marangos M. Pentoxifylline and
complicated COVID-19: A pathophysiologically based treatment
proposal. Med Hypotheses 2020; 143:109926.
244. Yue X, Shan B, Lasky JA. TGF-beta: Titan of lung fibrogenesis.
Curr Enzym Inhib 2010; 6:10.2174/10067.
245. Chen W. A potential treatment of COVID-19 with TGF-beta
blockade. Int J Biol Sci 2020; 16:1954–5.
246. Uckun FM, Hwang L, Trieu V. Selectively targeting TGF-beta
with Trabedersen/OT-101 in treatment of evolving and mild
ARDS in COVID-19. Clin Invest (Lond) 2020; 10:167–76.
247. Rabender C, Mezzaroma E, Mauro AG, Mullangi R, Abbate A,
Anscher M, et al. IPW-5371 proves effective as a radiation
countermeasure by mitigating radiation-induced late effects.
Radiat Res 2016; 186:478–88.
248. Ziegler CGK, Allon SJ, Nyquist SK, Mbano IM, Miao VN,
Tzouanas CN, et al. SARS-CoV-2 receptor ACE2 is an
interferon-stimulated gene in human airway epithelial cells and
is detected in specific cell subsets across tissues. Cell 2020;
181:1016–35.e19.
249. Li W, Moore MJ, Vasilieva N, Sui J, Wong SK, Berne MA, et al.
Angiotensin-converting enzyme 2 is a functional receptor for the
SARS coronavirus. Nature 2003; 426:450–4.
REVIEW 21
250. Danser AHJ, Epstein M, Batlle D. Renin-angiotensin system
blockers and the COVID-19 pandemic: at present there is no
evidence to abandon renin-angiotensin system blockers. Hyper-
tension 2020; 75:1382–5.
251. Kuster GM, Pfister O, Burkard T, Zhou Q, Twerenbold R, Haaf
P, et al. SARS-CoV2: should inhibitors of the renin-angiotensin
system be withdrawn in patients with COVID-19? Eur Heart J
2020; 41:1801–3.
252. Reynolds HR, Adhikari S, Pulgarin C, Troxel AB, Iturrate E,
Johnson SB, et al. Renin-angiotensin-aldosterone system inhib-
itors and risk of Covid-19. N Engl J Med 2020; 382:2441–8.
253. Hanff TC, Harhay MO, Brown TS, Cohen JB, Mohareb AM. Is
there an association between COVID-19 mortality and the renin-
angiotensin system? A call for epidemiologic investigations. Clin
Infect Dis 2020; 71:870–4.
254. Dalan R, Bornstein SR, El-Armouche A, Rodionov RN, Markov
A, Wielockx B, et al. The ACE-2 in COVID-19: foe or friend?
Horm Metab Res 2020; 52:257–63.
255. Shete A. Urgent need for evaluating agonists of angiotensin-(1-
7)/Mas receptor axis for treating patients with COVID-19. Int J
Infect Dis 2020; 96:348–51.
256. Meng J, Xiao G, Zhang J, He X, Ou M, Bi J, et al. Renin-
angiotensin system inhibitors improve the clinical outcomes of
COVID-19 patients with hypertension. Emerg Microbes Infect
2020; 9:757–60.
257. Medhora M, Gao F, Wu Q, Molthen RC, Jacobs ER, Moulder JE,
et al. Model development and use of ACE inhibitors for
preclinical mitigation of radiation-induced injury to multiple
organs. Radiat Res 2014; 182:545–55.
258. Medhora M, Gao F, Jacobs ER, Moulder JE. Radiation damage to
the lung: mitigation by angiotensin-converting enzyme (ACE)
inhibitors. Respirology 2012; 17:66–71.
259. Fish BL, Gao F, Narayanan J, Bergom C, Jacobs ER, Cohen EP,
et al. Combined hydration and antibiotics with lisinopril to
mitigate acute and delayed high-dose radiation injuries to
multiple organs. Health Phys 2016; 111:410–9.
260. Mehrvar S, la Cour MF, Medhora M, Camara AKS, Ranji M.
Optical metabolic imaging for assessment of radiation-induced
injury to rat kidney and mitigation by lisinopril. Ann Biomed Eng
2019; 47:1564–74.
261. Gao F, Fish BL, Moulder JE, Jacobs ER, Medhora M. Enalapril
mitigates radiation-induced pneumonitis and pulmonary fibrosis
if started 35 days after whole-thorax irradiation. Radiat Res 2013;
180:546–52.
262. Islam A, Bolduc DL, Zhai M, Kiang JG, Swift JM. Captopril
increases survival after whole-body ionizing irradiation but
decreases survival when combined with skin-burn trauma in
mice. Radiat Res 2015; 184:273–9.
263. Clausi MG, Stessin AM, Tsirka SE, Ryu S. Mitigation of
radiation myelopathy and reduction of microglial infiltration by
Ramipril, ACE inhibitor. Spinal Cord 2018; 56:733–40.
264. Rodgers KE, Xiong S, diZerega GS. Accelerated recovery from
irradiation injury by angiotensin peptides. Cancer Chemother
Pharmacol 2002; 49:403–11.
265. Rodgers KE, Espinoza T, Roda N, Meeks CJ, Hill C, Louie SG,
et al. Accelerated hematopoietic recovery with angiotensin-(1-7)
after total body radiation. Int J Radiat Biol 2012; 88:466–76.
266. Chi Y, Ge Y, Wu B, Zhang W, Wu T, Wen T, et al. Serum
cytokine and chemokine profile in relation to the severity of
coronavirus disease 2019 (COVID-19) in China. J Infect Dis
2020; 222:746–54.
267. Medford AR, Millar AB. Vascular endothelial growth factor
(VEGF) in acute lung injury (ALI) and acute respiratory distress
syndrome (ARDS): paradox or paradigm? Thorax 2006; 61:621–6.
268. Mukherji SK. Bevacizumab (Avastin). AJNR Am J Neuroradiol
2010; 31:235–6.
269. Makinde AY, Luo-Owen X, Rizvi A, Crapo JD, Pearlstein RD,
Slater JM, et al. Effect of a metalloporphyrin antioxidant (MnTE-
2-PyP) on the response of a mouse prostate cancer model to
radiation. Anticancer Res 2009; 29:107–18.
270. Liu Y, Kudo K, Abe Y, Aoki M, Hu DL, Kijima H, et al.
Hypoxia expression in radiation-induced late rectal injury. J
Radiat Res 2008; 49:261–8.
271. Dashti SR, Spalding A, Kadner RJ, Yao T, Kumar A, Sun DA, et
al. Targeted intraarterial anti-VEGF therapy for medically
refractory radiation necrosis in the brain. J Neurosurg Pediatr
2015; 15:20–5.
272. Jiang X, Engelbach JA, Yuan L, Cates J, Gao F, Drzymala RE, et
al. Anti-VEGF antibodies mitigate the development of radiation
necrosis in mouse brain. Clin Cancer Res 2014; 20:2695–702.
273. Aslan A, Kaya ZB, Bulduk EB, Ocal O, Ucar M, Erpolat OP, et
al. Prophylactic bevacizumab may mitigate radiation injury: an
experimental study. World Neurosurg 2018; 116:e791–800.
274. Xu Y, Rong X, Hu W, Huang X, Li Y, Zheng D, et al.
Bevacizumab monotherapy reduces radiation-induced brain
necrosis in nasopharyngeal carcinoma patients: a randomized
controlled trial. Int J Radiat Oncol Biol Phys 2018; 101:1087–95.
275. Lillicrap D. Disseminated intravascular coagulation in patients with
2019-nCoV pneumonia. J Thromb Haemost 2020; 18:786–7.
276. Kollias A, Kyriakoulis KG, Dimakakos E, Poulakou G, Stergiou
GS, Syrigos K. Thromboembolic risk and anticoagulant therapy
in COVID-19 patients: emerging evidence and call for action. Br
J Haematol 2020; 189:846–7.
277. Al-Samkari H, Karp Leaf RS, Dzik WH, Carlson JC, Fogerty AE,
Waheed A, et al. COVID and coagulation: bleeding and
thrombotic manifestations of SARS-CoV2 infection. Blood
2020; 136:489–500.
278. Harenberg J, Favaloro E. COVID-19: progression of disease and
intravascular coagulation - present status and future perspectives.
Clin Chem Lab Med 2020; 58:1029–36.
279. Gando S, Levi M, Toh CH. Disseminated intravascular
coagulation. Nat Rev Dis Primers 2016; 2:16037.
280. Krigsfeld GS, Savage AR, Billings PC, Lin L, Kennedy AR.
Evidence for radiation-induced disseminated intravascular coag-
ulation as a major cause of radiation-induced death in ferrets. Int J
Radiat Oncol Biol Phys 2014; 88:940–6.
281. Krigsfeld GS, Shah JB, Sanzari JK, Lin L, Kennedy AR.
Evidence of disseminated intravascular coagulation in a porcine
model following radiation exposure. Life Sci Space Res (Amst)
2014; 3:1–9.
282. DiCarlo AL, Kaminski JM, Hatchett RJ, Maidment BW. Role of
thrombocytopenia in radiation-induced mortality and review of
therapeutic approaches targeting platelet regeneration after
radiation exposure. J Radiat Oncol 2016; 5:19–32.
283. Han H, Yang L, Liu R, Liu F, Wu KL, Li J, et al. Prominent
changes in blood coagulation of patients with SARS-CoV-2
infection. Clin Chem Lab Med 2020; 58:1116–20.
284. Xu P, Zhou Q, Xu J. Mechanism of thrombocytopenia in
COVID-19 patients. Ann Hematol 2020; 99:1205–8.
285. Bunin DI, Bakke J, Green CE, Javitz HS, Fielden M, Chang PY.
Romiplostim (Nplate(R) as an effective radiation countermeasure
to improve survival and platelet recovery in mice. Int J Radiat
Biol 2020; 96:145–54.
286. Sun H, Tsai Y, Nowak I, Liesveld J, Chen Y. Eltrombopag, a
thrombopoietin receptor agonist, enhances human umbilical cord
blood hematopoietic stem/primitive progenitor cell expansion and
promotes multi-lineage hematopoiesis. Stem Cell Res 2012;
9:77–86.
287. Liesveld JL, Phillips GL, 2nd, Becker M, Constine LS, Friedberg
J, Andolina JR, et al. A phase 1 trial of eltrombopag in patients
undergoing stem cell transplantation after total body irradiation.
Biol Blood Marrow Transplant 2013; 19:1745–52.
288. Zulfiqar AA, Lorenzo-Villalba N, Hassler P, Andres E. Immune
22 RIOS ET AL.
thrombocytopenic purpura in a patient with COVID-19. N Engl J
Med 2020; 382:e43.
289. Riker RR, May TL, Fraser GL, Gagnon DJ, Bandara M, Zemrak
WR, et al. Heparin-induced thrombocytopenia with thrombosis in
COVID-19 adult respiratory distress syndrome. Res Pract
Thromb Haemost 2020; 4:936–41.
290. Thachil J TN, Gando S, Falanga A, Cattaneo M, Levi M. ISTH
interim guidance on recognition and management of coagulop-
athy in COVID-19. J Thromb Haemost 2020; 18:1023–6.
291. Kantara C, Moya SM, Houchen CW, Umar S, Ullrich RL, Singh
P, et al. Novel regenerative peptide TP508 mitigates radiation-
induced gastrointestinal damage by activating stem cells and
preserving crypt integrity. Lab Invest 2015; 95:1222–33.
292. Fossum TW, Olszewska-Pazdrak B, Mertens MM, Makarski LA,
Miller MW, Hein TW, et al. TP508 (Chrysalin) reverses
endothelial dysfunction and increases perfusion and myocardial
function in hearts with chronic ischemia. J Cardiovasc Pharmacol
Ther 2008; 13:214–25.
293. Ryaby JT, Sheller MR, Levine BP, Bramlet DG, Ladd AL,
Carney DH. Thrombin peptide TP508 stimulates cellular events
leading to angiogenesis, revascularization, and repair of dermal
and musculoskeletal tissues. J Bone Joint Surg Am 2006;
88:S132–9.
294. Fife C, Mader JT, Stone J, Brill L, Satterfield K, Norfleet A, et al.
Thrombin peptide Chrysalin stimulates healing of diabetic foot
ulcers in a placebo-controlled phase I/II study. Wound Repair
Regen 2007; 15:23–34.
295. Dashti-Khavidaki S, Khalili H. Considerations for statin therapy
in patients with COVID-19. Pharmacotherapy 2020; 40:484–6.
296. Lee KCH, Sewa DW, Phua GC. Potential role of statins in
COVID-19. Int J Infect Dis 2020.
297. Rodrigues-Diez RR, Tejera-Munoz A, Marquez-Exposito L,
Rayego-Mateos S, Sanchez LS, Marchant V, et al. Statins: Could
an old friend help the fight against COVID-19? Br J Pharmacol
2020; 177(21):4873-4886.
298. Reiner Z, Hatamipour M, Banach M, Pirro M, Al-Rasadi K,
Jamialahmadi T, et al. Statins and the COVID-19 main protease:
in silico evidence on direct interaction. Arch Med Sci 2020;
16:490–6.
299. South AM, Diz DI, Chappell MC. COVID-19, ACE2, and the
cardiovascular consequences. Am J Physiol Heart Circ Physiol
2020; 318:H1084–90.
300. Fedson DS, Opal SM, Rordam OM. Hiding in plain sight: an
approach to treating patients with severe COVID-19 infection.
mBio 2020; 11:e00398–20.
301. Kow CS, Hasan SS. Use of statins in patients with COVID-19.
QJM 2020; 113:604–5.
302. Zhang XJ, Qin JJ, Cheng X, Shen L, Zhao YC, Yuan Y, et al. In-
hospital use of statins is associated with a reduced risk of
mortality among individuals with COVID-19. Cell Metab 2020;
32:176–87.e4.
303. Mathew B, Huang Y, Jacobson JR, Berdyshev E, Gerhold LM,
Wang T, et al. Simvastatin attenuates radiation-induced murine
lung injury and dysregulated lung gene expression. Am J Respir
Cell Mol Biol 2011; 44:415–22.
304. Zhao X, Yang H, Jiang G, Ni M, Deng Y, Cai J, et al. Simvastatin
attenuates radiation-induced tissue damage in mice. J Radiat Res
2014; 55:257–64.
305. Xu L, Yang X, Chen J, Ge X, Qin Q, Zhu H, et al. Simvastatin
attenuates radiation-induced salivary gland dysfunction in mice.
Drug Des Devel Ther 2016; 10:2271–8.
306. Zhang K, He X, Zhou Y, Gao L, Qi Z, Chen J, et al. Atorvastatin
ameliorates radiation-induced cardiac fibrosis in rats. Radiat Res
2015; 184:611–20.
307. Talebpour Amiri F, Hamzeh M, Naeimi RA, Ghasemi A,
Hosseinimehr SJ. Radioprotective effect of atorvastatin against
ionizing radiation-induced nephrotoxicity in mice. Int J Radiat
Biol 2018; 94:106–13.
308. Ran XZ, Ran X, Zong ZW, Liu DQ, Xiang GM, Su YP, et al.
Protective effect of atorvastatin on radiation-induced vascular
endothelial cell injury in vitro. J Radiat Res 2010; 51:527–33.
309. Williams JP, Hernady E, Johnston CJ, Reed CM, Fenton B,
Okunieff P, et al. Effect of administration of lovastatin on the
development of late pulmonary effects after whole-lung irradia-
tion in a murine model. Radiat Res 2004; 161:560–7.
310. Geiger H, Pawar SA, Kerschen EJ, Nattamai KJ, Hernandez I,
Liang HP, et al. Pharmacological targeting of the thrombomo-
dulin-activated protein C pathway mitigates radiation toxicity.
Nat Med 2012; 18:1123–9.
311. Griffin JH, Lyden P. COVID-19 hypothesis: activated protein C
for therapy of virus-induced pathologic thromboinflammation.
Res Pract Thromb Haemost 2020; 4:506–9.
312. Wang J. Fast identification of possible drug treatment of
coronavirus disease-19 (COVID-19) through computational drug
repurposing study. J Chem Inf Model 2020; 60:3277–86.
313. Toma E. Doxycycline and pentoxifylline for mild and mild-to-
moderate Covid-19. 2020; Preprints 2020; (https://bit.ly/
2ZYyby1)
314. Conforti C, Giuffrida R, Zalaudek I, Di Meo N. Doxycycline, a
widely used antibiotic in dermatology with a possible anti-
inflammatory action against IL-6 in COVID-19 outbreak.
Dermatol Ther 2020; Epub ahead of print.
315. Schogler A, Kopf BS, Edwards MR, Johnston SL, Casaulta C,
Kieninger E, et al. Novel antiviral properties of azithromycin in
cystic fibrosis airway epithelial cells. Eur Respir J 2015; 45:428–
39.
316. Pokhrel R, Chapagain P, Siltberg-Liberles J. Potential RNA-
dependent RNA polymerase inhibitors as prospective therapeutics
against SARS-CoV-2. J Med Microbiol 2020; 69:864–73.
317. Pindiprolu S, Kumar CSP, Kumar Golla VS, Likitha P, Chandrta
S, Bashae, et al. Pulmonary delivery of nanostructured lipid
carriers for effective repurposing of salinomycin as an antiviral
agent. Med Hypotheses 2020; 143:109858.
318. Jean SS, Hsueh PR. Old and re-purposed drugs for the treatment
of COVID-19. Expert Rev Anti Infect Ther 2020:1–5.
319. Ceccarelli G, Alessandri F, d’Ettorre G, Borrazzo C, Spagnolello
O, Oliva A, et al. Is teicoplanin a complementary treatment option
for COVID-19? The question remains. Int J Antimicrob Agents
2020:106029.
320. Sivapalan P, Ulrik CS, Bojesen RD, Lapperre TS, Eklof JV,
Hakansson KEJ, et al. Proactive prophylaxis with azithromycin
and hydroxychloroquine in hospitalised patients with COVID-19
(ProPAC-COVID): a structured summary of a study protocol for
a randomised controlled trial. Trials 2020; 21:513.
321. Damle B, Vourvahis M, Wang E, Leaney J, Corrigan B. Clinical
pharmacology perspectives on the antiviral activity of azithro-
mycin and use in COVID-19. Clin Pharmacol Ther 2020;
108:201–11.
322. Gbinigie K, Frie K. Should azithromycin be used to treat
COVID-19? A rapid review. BJGP Open 2020; 4:
bjgpopen20X101094.
323. Du Y, Tu L, Zhu P, Mu M, Wang R, Yang P, et al. Clinical
features of 85 fatal cases of COVID-19 from Wuhan: a
retrospective observational study. Am J Respir Crit Care Med
2020; 201:1372–9.
324. Naseer MS, Chand R, Coppola S, Abreo A, Sharma M, Singh N.
Post-transplant de-novo renal phospholipidosis in a kidney
transplant recipient: Fabry disease or something else? Clin
Nephrol Case Stud 2020; 8:46–8.
325. Asaduzzaman M, Zaman F, Rousham E. Antibiotic consumption
may be linked to exaggeration of COVID-19. Med Hypotheses
2020; 143:109913.
326. Epperly MW, Franicola D, Shields D, Rwigema JC, Stone B,
REVIEW 23
Zhang X, et al. Screening of antimicrobial agents for in vitro
radiation protection and mitigation capacity, including those used
in supportive care regimens for bone marrow transplant
recipients. In Vivo 2010; 24:9–19.
327. Kim K, Damoiseaux R, Norris AJ, Rivina L, Bradley K, Jung
ME, et al. High throughput screening of small molecule libraries
for modifiers of radiation responses. Int J Radiat Biol 2011;
87:839–45.
328. Shalit I, Kletter Y, Weiss K, Gruss T, Fabian I. Enhanced
hematopoiesis in sublethally irradiated mice treated with various
quinolones. Eur J Haematol 1997; 58:92–8.
329. Linkenheimer WH, Berger H. The effect of continuous antibiotic
feeding on x-irradiation mortality in mice. Proc Soc Exp Biol
Med 1961; 108:676–80.
330. 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; 103:343–55.
331. Kiang JG, Garrison BR, Smith JT, Fukumoto R. Ciprofloxacin as
a potential radio-sensitizer to tumor cells and a radio-protectant
for normal cells: differential effects on gamma-H2AX formation,
p53 phosphorylation, Bcl-2 production, and cell death. Mol Cell
Biochem 2014; 393:133–43.
332. Kiang JG, Fukumoto R. Ciprofloxacin increases survival after
ionizing irradiation combined injury: gamma-H2AX formation,
cytokine/chemokine, and red blood cells. Health Phys 2014;
106:720–6.
333. A phase-II trial of fractionated total body radiation in bone
marrow transplantation for acute leukemia. UCLA Bone Marrow
Transplant Team. Transplant Proc 1979; 11:205–7.
334. Fukumoto R. Mesenchymal stem cell therapy for acute radiation
syndrome. Mil Med Res 2016; 3:17.
335. DiCarlo AL, Tamarat R, Rios CI, Benderitter M, Czarniecki CW,
Allio TC, et al. Cellular therapies for treatment of radiation
injury: report from a NIH/NIAID and IRSN workshop. Radiat
Res 2017; 188:e54–75.
336. Xu T, Zhang Y, Chang P, Gong S, Shao L, Dong L.
Mesenchymal stem cell-based therapy for radiation-induced lung
injury. Stem Cell Res Ther 2018; 9:18.
337. Klein D, Steens J, Wiesemann A, Schulz F, Kaschani F, Rock K,
et al. Mesenchymal stem cell therapy protects lungs from
radiation-induced endothelial cell loss by restoring superoxide
dismutase 1 expression. Antioxid Redox Signal 2017; 26:563–82.
338. Chrzanowski W, Kim SY, McClements L. Can stem cells beat
COVID-19: advancing stem cells and extracellular vesicles
toward mainstream medicine for lung injuries associated with
SARS-CoV-2 infections. Front Bioeng Biotechnol 2020; 8:554.
339. Golchin A, Seyedjafari E, Ardeshirylajimi A. Mesenchymal stem
cell therapy for COVID-19: present or future. Stem Cell Rev Rep
2020; 16:427–33.
340. Sengupta V, Sengupta S, Lazo A, Woods P, Nolan A, Bremer N.
Exosomes derived from bone marrow mesenchymal stem cells as
treatment for severe COVID-19. Stem Cells Dev 2020; 29:747–
54.
341. Atluri S, Manchikanti L, Hirsch JA. Expanded umbilical cord
mesenchymal stem cells (UC-MSCs) as a therapeutic strategy in
managing critically ill COVID-19 patients: the case for
compassionate use. Pain Physician 2020; 23:E71–83.
342. Rogers CJ, Harman RJ, Bunnell BA, Schreiber MA, Xiang C,
Wang FS, et al. Rationale for the clinical use of adipose-derived
mesenchymal stem cells for COVID-19 patients. J Transl Med
2020; 18:203.
343. Leng Z, Zhu R, Hou W, Feng Y, Yang Y, Han Q, et al.
Transplantation of ACE2(–) mesenchymal stem cells improves
the outcome of patients with COVID-19 pneumonia. Aging Dis
2020; 11:216–28.
344. Calabrese EJ, Dhawan G. How radiotherapy was historically used
to treat pneumonia: could it be useful today? Yale J Biol Med
2013; 86:555–70.
345. Trott KR, Zschaeck S, Beck M. Radiation therapy for COVID-19
pneumopathy. Radiother Oncol 2020; 147:210–11.
346. Rodel F, Arenas M, Ott OJ, Fournier C, Georgakilas AG, Tapio
S, et al. Low-dose radiation therapy for COVID-19 pneumopathy:
what is the evidence? Strahlenther Onkol 2020; 196:679–82.
347. Montero A, Arenas M, Algara M. Low-dose radiation therapy:
could it be a game-changer for COVID-19? Clin Transl Oncol
2020; Epub ahead of print.
348. Kirsch DG, Diehn M, Cucinotta FA, Weichselbaum R. Response
letter: Radiation therapy for COVID-19 pneumopathy. Radiother
Oncol 2020; 149:238–9.
349. Kirkby C, Mackenzie M. Is low dose radiation therapy a potential
treatment for COVID-19 pneumonia? Radiother Oncol 2020;
147:221.
350. Kefayat A, Ghahremani F. Low dose radiation therapy for
COVID-19 pneumonia: A double-edged sword. Radiother Oncol
2020; 147:224–5.
351. Lazzari G, Silvano G. RILI model and the Covid-19 pneumonia:
The radiation oncologist point of view. Radiother Oncol 2020;
147:222–3.
352. Schaue D, McBride WH. Flying by the seat of our pants: is low
dose radiation therapy for COVID-19 an option? Int J Radiat Biol
2020; 96(10):1219–1223.
353. Gupta A, Madhavan MV, Sehgal K, Nair N, Mahajan S,
Sehrawat TS, et al. Extrapulmonary manifestations of COVID-
19. Nat Med 2020; 26:1017–32.
24 RIOS ET AL.
... The global pandemic caused by SARS-CoV-2 (COVID- 19) resulted in acute respiratory distress syndrome (ARDS) with clinical presentations (1)(2)(3)(4)(5)(6)(7)(8)(9)(10)(11)(12)(13)(14)(15)(16) that were like those of ionizing irradiation-induced pneumonitis and fibrosis (17,18). Similarities between COVID-19 and ionizing irradiationinduced changes in the lung have led to speculation that some biomolecular and biochemical responses may involve common pathways (19,20). ...
... The global pandemic caused by SARS-CoV-2 (COVID- 19) resulted in acute respiratory distress syndrome (ARDS) with clinical presentations (1)(2)(3)(4)(5)(6)(7)(8)(9)(10)(11)(12)(13)(14)(15)(16) that were like those of ionizing irradiation-induced pneumonitis and fibrosis (17,18). Similarities between COVID-19 and ionizing irradiationinduced changes in the lung have led to speculation that some biomolecular and biochemical responses may involve common pathways (19,20). Prior research demonstrated potential additive or synergistic deleterious effects of pulmonary virus infections with damage induced by preexisting lung diseases, and with ionizing irradiation-induced lung damage (21)(22)(23)(24)(25). ...
Article
Background/aim: There is concern that people who had COVID-19 will develop pulmonary fibrosis. Using mouse models, we compared pulmonary inflammation following injection of the spike protein of SARS-CoV-2 (COVID-19) to radiation-induced inflammation to demonstrate similarities between the two models. SARS-CoV-2 (COVID-19) induces inflammatory cytokines and stress responses, which are also common to ionizing irradiation-induced acute pulmonary damage. Cellular senescence, which is a late effect following exposure to SARS-CoV-2 as well as radiation, was investigated. Materials and methods: We evaluated the effect of SARS-CoV-2 spike protein compared to ionizing irradiation in K18-hACE2 mouse lung, human lung cell lines, and in freshly explanted human lung. We measured reactive oxygen species, DNA double-strand breaks, stimulation of transforming growth factor-beta pathways, and cellular senescence following exposure to SARS-CoV-2 spike protein, irradiation or SARS-COV-2 and irradiation. We also measured the effects of the antioxidant radiation mitigator MMS350 following irradiation or exposure to SARS-CoV-2. Results: SARS-CoV-2 spike protein induced reactive oxygen species, DNA double-strand breaks, transforming growth factor-β signaling pathways, and senescence, which were exacerbated by prior or subsequent ionizing irradiation. The water-soluble radiation countermeasure, MMS350, reduced spike protein-induced changes. Conclusion: In both the SARS-Co-2 and the irradiation mouse models, similar responses were seen indicating that irradiation or exposure to SARS-CoV-2 virus may lead to similar lung diseases such as pulmonary fibrosis. Combination of irradiation and SARS-CoV-2 may result in a more severe case of pulmonary fibrosis. Cellular senescence may explain some of the late effects of exposure to SARS-CoV-2 spike protein and to ionizing irradiation.
... IR causes severe damage to cells and tissues through direct mechanisms such as the induction of DNA double-strand breaks (DSBs), an increase in ROS, and indirect mechanisms such as the upregulation of inflammatory factors leading to a cytokine storm and subsequent hyperactivated immune responses [5,6]. Interestingly, existing studies have reported that the SARS-CoV-2 infection triggers inflammation and immune responses similar to those triggered by irradiation [7,8]. Both SARS-CoV-2 infection and irradiation exposure can cause damage to multiple organs in the body, including the immune system, hematopoietic system, lung, liver, and so on. ...
... Both SARS-CoV-2 infection and irradiation exposure can cause damage to multiple organs in the body, including the immune system, hematopoietic system, lung, liver, and so on. This can be attributed to the induction of a systemic cytokine storm, characterized by the activation of proinflammatory cytokines such as interferon alpha (IFNA), interferon beta (IFNB), interleukin 1 beta (IL1B), interleukin 6, interleukin 18 (IL18), as well as the disruption of lymphocyte population and function [7]. Consequently, hyperactivated inflammation and dysregulated immune responses emerge as shared underlying factors in the development of multi-tissue injuries caused by SARS-CoV-2 infection and irradiation exposure. ...
Article
Full-text available
Ionizing radiation (IR) induces severe hematopoietic injury by causing DNA and RNA damage as well as activating the immune responses, necessitating the development of effective therapeutic strategies. Ribonuclease L (RNase L) as an innate immune response pathway is triggered by exogenous and endogenous abnormal dsRNA under viral infection and dyshomeostasis, thereby activating the immune responses. Thus, we investigated the effect of RNase L on irradiation-induced bone marrow damage using RNase L knockout (RNase L−/−) mice. Phenotypic analysis revealed that RNase L knockout mitigates irradiation-induced injury in the bone marrow. Further investigation into the mechanism of RNase L by RNA-seq, qRT-PCR, and CBA analysis demonstrated that RNase L deficiency counteracts the upregulation of genes related to immune responses induced by irradiation, including cytokines and interferon-stimulated genes. Moreover, RNase L deficiency inhibits the increased levels of immunoglobulins in serum induced by irradiation. These findings indicate that RNase L plays a role in the immune response induced by irradiation in the bone marrow. This study further enhances our understanding of the biological functions of RNase L in the immune response induced by irradiation and offers a novel approach for managing irradiation-induced bone marrow injury through the regulation of RNase L activation.
... For example, parallel etiologies between coronavirus disease (COVID-19) and irradiation injury implicate specific organ (e.g., vasculature, lungs, heart, kidneys, gut, and brain) and immune system involvement, which should be further explored. 4 Endothelial damage is observed following both radiological and chemical injury, as are delayed effects persisting months and years in the body's response to a variety of CRN threats. Traditionally, USG agencies have focused on studying the biological effects of CRN threats separately, with the goal to treat the biological damage from specific threats. ...
... In addition to the shift in thinking discussed above, BARDA continues to engage with the FDA/CDRH to revisit the a https://www.cdc.gov/biomonitoring/chemical_threat_agents.html 4 CI Rios et al. ...
Article
Full-text available
Although chemical and radiological agents cause toxicity through different mechanisms, the multiorgan injuries caused by these threats share similarities that convene on the level of basic biological responses. This publication will discuss these areas of convergence and explore “multi-utility” approaches that could be leveraged to address common injury mechanisms underlying actions of chemical and radiological agents in a threat-agnostic manner. In addition, we will provide an overview of the current state of radiological and chemical threat research, discuss the US Government’s efforts toward medical preparedness, and identify potential areas for collaboration geared toward enhancing preparedness and response against radiological and chemical threats. We also will discuss previous regulatory experience to provide insight on how to navigate regulatory paths for US Food and Drug Administration (FDA) approval/licensure/clearance for products addressing chemical or radiological/nuclear threats. This publication follows a 2022 trans-agency meeting titled, “Overlapping Science in Radiation and Sulfur Mustard Exposures of Skin and Lung: Consideration of Models, Mechanisms, Organ Systems, and Medical Countermeasures,” sponsored by the National Institute of Allergy and Infectious Diseases (NIAID), a part of the National Institutes of Health (NIH). Discussions from this meeting explored the overlapping nature of radiation and chemical injury and spurred increased interest in how preparedness for one threat leads to preparedness for the other. Herein, subject matter experts from the NIAID and the Biomedical Advanced Research and Development Authority (BARDA), a part of the Administration for Strategic Preparedness and Response (ASPR), summarize the knowledge gained from recently funded biomedical research, as well as insights from the 2022 meeting. These topics include identification of common areas for collaboration, potential use of biomarkers of injury to identify injuries caused by both hazards, and common and widely available treatments that could treat damage caused by radiological or chemical threats.
... 66 Pro-inflammatory cytokines such as IL-1β, IL-6, and TNFα play major roles in the development of radiation-induced intestinal damage. 12,67 In the present study we undertook a comprehensive evaluation of several GI injuryrelated pro-and anti-inflammatory cytokines in the serum (Figure 4) and colon lysates ( Figure 5). The majority of cytokines, including IL-1 β, TNFα and IL-6, were induced both systemically and in the colon in response to radiation. ...
Article
Full-text available
High-dose radiation exposure results in gastrointestinal (GI) acute radiation syndrome identified by the destruction of mucosal layer, intestinal growth barrier dysfunction, and aberrant inflammatory responses. Further, radiation causes gut microbiome dysbiosis characterized by diminished microbial diversity, mostly commensal bacteria, and the spread of bacterial pathogens that trigger the recruitment of immune cells and the production of pro-inflammatory factors that lead to further GI tissue damage. Currently, there are no U.S. Food and Drug Administration (FDA) approved countermeasures that can treat radiation-induced GI injuries. To meet this critical need, Synedgen Inc. has developed a glycopolymer radiomitigator (MIIST305) that is specifically targeted to the GI tract, which acts by intercalating into the mucus layer and the glycocalyx of intestinal epithelial cells that could potentially ameliorate the deleterious effects of radiation. Male C57BL/6J adult mice were exposed to 13 Gy partial body X–irradiation with 5% bone marrow shielding and MIIST305 was administered on days 1, 3, and 5 post-irradiation. Approximately 85% of the animals survived the irradiation exposure and were apparently healthy until the end of the 30-day study period. In contrast, no control, Vehicle-treated animals survived past day 10 at this radiation dose. We show that MIIST305 improved intestinal epithelial barrier function and suppressed systemic inflammatory responses mediated by radiation-induced pro-inflammatory cytokines. Taxonomic profiling and community structure of the fecal and colonic mucosa microbiota demonstrated that MIIST305 treatment increased microbial diversity and restored abundance of beneficial commensal bacteria, including Lactobacillus and Bifidobacterium genera while suppressing potentially pathogenic bacteria Enterococcus and Staphylococcus compared with Vehicle-treated animals. In summary, MIIST305 is a novel GI-targeted therapeutic that greatly enhances survival in mice exposed to lethal radiation and protects the GI tract from injury by restoring a balanced gut microbiota and reducing pro-inflammatory responses. Further development of this drug as an FDA-approved medical countermeasure is of critical importance.
... The observed sex difference in lung-DEARE in our rodent model mirrors the established sex disparity in COVID-19 respiratory syndrome patients where globally men have significantly higher death rates than women (Gagliardi et al., 2020;Mukherjee and Pahan, 2021). Interestingly, there are other established commonalities between radiation pneumonitis and COVID-19 acute respiratory syndrome including immune infiltration, inflammatory cytokine release, and dysregulation of the renin-angiotensin system (RAS) (Barhoumi et al., 2021;Rios et al., 2021;Geng et al., 2022). It has been hypothesized that the COVID-19 sex disparity may be attributable to dysregulation of RAS as the angiotensin converting enzyme (ACE) family member ACE2 serves as the entry point for the SARS-CoV-2 virus (Miličić Stanić et al., 2021). ...
Article
Full-text available
Introduction: In experimental animal models, biological sex-differences in the manifestation and severity of normal tissue radiation injury have been well-documented. Previously we demonstrated male and female rats have differential and highly reproducible responses to high-dose partial body irradiation (PBI) with male rats having greater susceptibility to both gastrointestinal acute radiation syndrome (GI-ARS) and radiation pneumonitis than female rats. Methods: In the current study, we have investigated whether differential expression of the renin-angiotensin system (RAS) enzymes angiotensin converting enzyme (ACE) and ACE2 contribute to the observed sex-related differences in radiation response. Results: During the period of symptomatic pneumonitis, the relative ratio of ACE to ACE2 (ACE/ACE2) protein in the whole lung was significantly increased by radiation in male rats alone. Systemic treatment with small molecule ACE2 agonist diminazene aceturate (DIZE) increased lung ACE2 activity and reduced morbidity during radiation pneumonitis in both sexes. Notably DIZE treatment also abrogated morbidity in male rats during GI-ARS. We then evaluated the contribution of the irradiated bone marrow (BM) compartment on lung immune cell infiltration and ACE imbalance during pneumonitis. Transplantation of bone marrow from irradiated donors increased both ACE-expressing myeloid cell infiltration and immune ACE activity in the lung during pneumonitis compared to non-irradiated donors. Discussion: Together, these data demonstrate radiation induces a sex-dependent imbalance in the renin-angiotensin system enzymes ACE and ACE2. Additionally, these data suggest a role for ACE-expressing myeloid cells in the pathogenesis of radiation pneumonitis. Finally, the observed sex-differences underscore the need for consideration of sex as a biological variable in the development of medical countermeasures for radiation exposure.
Article
The key role of structural cells in immune modulation has been revealed with the advent of single-cell multiomics, but the underlying mechanism remains poorly understood. Here, we revealed that the transcriptional activation of interferon regulatory factor 1 (IRF1) in response to ionizing radiation, cytotoxic chemicals and SARS-CoV-2 viral infection determines the fate of structural cells and regulates communication between structural and immune cells. Radiation-induced leakage of mtDNA initiates the nuclear translocation of IRF1, enabling it to regulate the transcription of inflammation- and cell death-related genes. Novel posttranslational modification (PTM) sites in the nuclear localization sequence (NLS) of IRF1 were identified. Functional analysis revealed that mutation of the acetylation site and the phosphorylation sites in the NLS blocked the transcriptional activation of IRF1 and reduced cell death in response to ionizing radiation. Mechanistically, reciprocal regulation between the single-stranded DNA sensors SSBP1 and IRF1, which restrains radiation-induced and STING/p300-mediated PTMs of IRF1, was revealed. In addition, genetic deletion or pharmacological inhibition of IRF1 tempered radiation-induced inflammatory cell death, and radiation mitigators also suppressed SARS-CoV-2 NSP-10-mediated activation of IRF1. Thus, we revealed a novel cytoplasm-oriented mechanism of IRF1 activation in structural cells that promotes inflammation and highlighted the potential effectiveness of IRF1 inhibitors against immune disorders.
Article
As severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) variants continue to wreak havoc worldwide, the “Cytokine Storm” (CS, also known as the inflammatory storm) or Cytokine Release Syndrome has reemerged in the public consciousness. CS is a significant contributor to the deterioration of infected individuals. Therefore, CS control is of great significance for the treatment of critically ill patients and the reduction of mortality rates. With the occurrence of variants, concerns regarding the efficacy of vaccines and antiviral drugs with a broad spectrum have grown. We should make an effort to modernize treatment strategies to address the challenges posed by mutations. Thus, in addition to the requirement for additional clinical data to monitor the long‐term effects of vaccines and broad‐spectrum antiviral drugs, we can use CS as an entry point and therapeutic target to alleviate the severity of the disease in patients. To effectively combat the mutation, new technologies for neutralizing or controlling CS must be developed. In recent years, nanotechnology has been widely applied in the biomedical field, opening up a plethora of opportunities for CS. Here, we put forward the view of cytokine storm as a therapeutic target can be used to treat critically ill patients by expounding the relationship between coronavirus disease 2019 (COVID‐19) and CS and the mechanisms associated with CS. We pay special attention to the representative strategies of nanomaterials in current neutral and CS research, as well as their potential chemical design and principles. We hope that the nanostrategies described in this review provide attractive treatment options for severe and critical COVID‐19 caused by CS.
Article
Full-text available
SARS-CoV-2 infection is required for COVID-19, but many signs and symptoms of COVID-19 differ from common acute viral diseases. SARS-CoV-2 infection is necessary but not sufficient for development of clinical COVID-19 disease. Currently, there are no approved pre- or post-exposure prophylactic COVID-19 medical countermeasures. Clinical data suggest that famotidine may mitigate COVID-19 disease, but both mechanism of action and rationale for dose selection remain obscure. We have investigated several plausible hypotheses for famotidine activity including antiviral and host-mediated mechanisms of action. We propose that the principal mechanism of action of famotidine for relieving COVID-19 symptoms involves on-target histamine receptor H2 activity, and that development of clinical COVID-19 involves dysfunctional mast cell activation and histamine release. Based on these findings and associated hypothesis, new COVID-19 multi-drug treatment strategies based on repurposing well-characterized drugs are being developed and clinically tested, and many of these drugs are available worldwide in inexpensive generic oral forms suitable for both outpatient and inpatient treatment of COVID-19 disease.
Article
Full-text available
i>Background: Coronavirus disease 2019 (COVID-19) is associated with diffuse lung damage. Corticosteroids may modulate inflammation-mediated lung injury, reducing progression to respiratory failure and death. Methods: The Randomised Evaluation of COVID-19 therapy (RECOVERY) trial is a randomized, controlled, open-label, platform trial comparing a range of possible treatments with usual care in patients hospitalized with COVID-19. We report the preliminary results for the comparison of dexamethasone 6 mg given once daily for up to ten days vs. usual care alone. The primary outcome was 28-day mortality. Results: 2104 patients randomly allocated to receive dexamethasone were compared with 4321 patients concurrently allocated to usual care. Overall, 482 (22.9%) patients allocated dexamethasone and 1110 (25.7%) patients allocated usual care died within 28 days (age-adjusted rate ratio [RR] 0.83; 95% confidence interval [CI] 0.75 to 0.93; P<0.001). The absolute mortality rate reductions varied depending on level of respiratory support at randomization: Dexamethasone reduced deaths in patients receiving invasive mechanical ventilation (29.3% vs. 41.4%, RR 0.64 [95% CI 0.51 to 0.81]), in patients receiving oxygen without invasive mechanical ventilation (23.3% vs. 26.2%, RR 0.82 [95% CI 0.72 to 0.94]), but did not reduce mortality in patients not receiving respiratory support at randomization (17.8% vs. 14.0%, RR 1.19 [95% CI 0.91 to 1.55]). Conclusions: In patients hospitalized with COVID-19, dexamethasone reduced 28-day mortality among those receiving invasive mechanical ventilation or oxygen at randomization, but not among patients not receiving respiratory support. (Funded by the Medical Research Council and National Institute for Health Research; ClinicalTrials.gov NCT04381936 and ISRCTN 50189673)
Preprint
Full-text available
Critically ill patients with COVID-19 may suffer from a cytokine release syndrome (CRS) characterized by remarkably high levels of interleukin 6 (IL-6). We assessed the effects of tocilizumab, an IL-6 receptor antagonist, on intra-hospital mortality and development of positive cultures in patients with COVID-19 admitted to the ICU. In this study, patients with COVID 19 admitted in the ICU who were treated with tocilizumab plus standard care were enrolled and compared to controls. Main outcome: 1) intra-hospital mortality; Secondary Outcomes: 1) the need for renal replacement therapy, 2) use of antibiotics and positive culture, and 3) inflammatory and oxygenation markers. Results: There was no difference in mortality, need for renal replacement therapy, use of antibiotics or positive cultures between the two groups. The use of corticosteroids was more frequent in the treatment group. Levels of C-reactive protein (CRP) and WBC (white blood cells) counts declined significantly faster in the treatment group. Oxygenation markers rose significantly higher in patients in the tocilizumab group as compared to controls. Conclusion: tocilizumab was associated with rapid improvement in oxygenation and a faster decrease of CRP and WBC counts in patients with COVID-19 and should be evaluated as rescue therapy for patients with progressive disease
Article
Full-text available
Background: Severe COVID-19 can manifest in rapid decompensation and respiratory failure with elevated inflammatory markers, consistent with cytokine release syndrome for which IL-6 blockade is approved treatment. Methods: We assessed effectiveness and safety of IL-6 blockade with tocilizumab in a single-center cohort of patients with COVID-19 requiring mechanical ventilation. The primary endpoint was survival probability post-intubation; secondary analyses included an ordinal illness severity scale integrating superinfections. Outcomes in patients who received tocilizumab compared to tocilizumab-untreated controls were evaluated using multivariable Cox regression with propensity score inverse probability weighting (IPTW). Results: 154 patients were included, of whom 78 received tocilizumab and 76 did not. Median follow-up was 47 days (range 28-67). Baseline characteristics were similar between groups, although tocilizumab-treated patients were younger (mean 55 vs. 60 years), less likely to have chronic pulmonary disease (10% vs. 28%), and had lower D-dimer values at time of intubation (median 2.4 vs. 6.5 mg/dL). In IPTW-adjusted models, tocilizumab was associated with a 45% reduction in hazard of death [hazard ratio 0.55 (95% CI 0.33, 0.90)] and improved status on the ordinal outcome scale [odds ratio per 1-level increase: 0.58 (0.36, 0.94)]. Though tocilizumab was associated with an increased proportion of patients with superinfections (54% vs. 26%; p<0.001), there was no difference in 28-day case fatality rate among tocilizumab-treated patients with versus without superinfection [22% vs. 15%; p=0.42]. Staphylococcus aureus accounted for ~50% of bacterial pneumonia. Conclusions: In this cohort of mechanically ventilated COVID-19 patients, tocilizumab was associated with lower mortality despite higher superinfection occurrence.
Article
Full-text available
Although COVID-19 is most well known for causing substantial respiratory pathology, it can also result in several extrapulmonary manifestations. These conditions include thrombotic complications, myocardial dysfunction and arrhythmia, acute coronary syndromes, acute kidney injury, gastrointestinal symptoms, hepatocellular injury, hyperglycemia and ketosis, neurologic illnesses, ocular symptoms, and dermatologic complications. Given that ACE2, the entry receptor for the causative coronavirus SARS-CoV-2, is expressed in multiple extrapulmonary tissues, direct viral tissue damage is a plausible mechanism of injury. In addition, endothelial damage and thromboinflammation, dysregulation of immune responses, and maladaptation of ACE2-related pathways might all contribute to these extrapulmonary manifestations of COVID-19. Here we review the extrapulmonary organ-specific pathophysiology, presentations and management considerations for patients with COVID-19 to aid clinicians and scientists in recognizing and monitoring the spectrum of manifestations, and in developing research priorities and therapeutic strategies for all organ systems involved.
Article
Full-text available
The pandemia of coronavirus disease 2019 (COVID-19) has caused more than 355,000 confirmed deaths worldwide. However, publications on postmortem findings are scarce. We present the pulmonary findings in four cases of fatal COVID-19 with a spectrum of lung pathology reflecting disease course and duration, invasive therapies, and laboratory features. Early disease is characterized by neutrophilic, exudative capillaritis with microthrombosis and high levels of IL-1beta and IL-6. Later stages are associated with diffuse alveolar damage and ongoing intravascular thrombosis in small to medium-sized pulmonary vessels, occasionally with areas of infarction equivalents, accompanied by laboratory features of disseminated intravascular coagulation. In late stages, organizing pneumonia with extensive intra-alveolar proliferation of fibroblasts and marked metaplasia of alveolar epithelium can be observed. Viral RNA is encountered in the lung, with virus particles in endothelial cells and pneumocytes. In many patients, multi-organ failure with severe liver damage sets in finally, possibly as consequence of an early-onset pro-inflammatory cytokine storm and/or thrombotic microangiopathy.
Article
Many cases of human exposures to high-dose radiation have been documented, including individuals exposed during the detonation of atomic bombs in Hiroshima and Nagasaki, nuclear power plant disasters (e.g., Chernobyl), as well as industrial and medical accidents. For many of these exposures, injuries to the skin have been present and have played a significant role in the progression of the injuries and survivability from the radiation exposure. There are also instances of radiation-induced skin complications in routine clinical radiotherapy and radiation diagnostic imaging procedures. In response to the threat of a radiological or nuclear mass casualty incident, the U.S. Department of Health and Human Services tasked the National Institute of Allergy and Infectious Diseases (NIAID) with identifying and funding early- to mid-stage medical countermeasure (MCM) development to treat radiation-induced injuries, including those to the skin. To appropriately assess the severity of radiation-induced skin injuries and determine efficacy of different approaches to mitigate/treat them, it is necessary to develop animal models that appropriately simulate what is seen in humans who have been exposed. In addition, it is important to understand the techniques that are used in other clinical indications (e.g., thermal burns, diabetic ulcers, etc.) to accurately assess the extent of skin injury and progression of healing. For these reasons, the NIAID partnered with two other U.S. Government funding and regulatory agencies, the Biomedical Advanced Research and Development Authority (BARDA) and the Food and Drug Administration (FDA), to identify state-of-the-art methods in assessment of skin injuries, explore animal models to better understand radiation-induced cutaneous damage and investigate treatment approaches. A two-day workshop was convened in May 2019 highlighting talks from 28 subject matter experts across five scientific sessions. This report provides an overview of information that was presented and the subsequent guided discussions.
Article
Background An important feature of severe acute respiratory syndrome coronavirus 2 pathogenesis is COVID-19-associated coagulopathy, characterised by increased thrombotic and microvascular complications. Previous studies have suggested a role for endothelial cell injury in COVID-19-associated coagulopathy. To determine whether endotheliopathy is involved in COVID-19-associated coagulopathy pathogenesis, we assessed markers of endothelial cell and platelet activation in critically and non-critically ill patients admitted to the hospital with COVID-19. Methods In this single-centre cross-sectional study, hospitalised adult (≥18 years) patients with laboratory-confirmed COVID-19 were identified in the medical intensive care unit (ICU) or a specialised non-ICU COVID-19 floor in our hospital. Asymptomatic, non-hospitalised controls were recruited as a comparator group for biomarkers that did not have a reference range. We assessed markers of endothelial cell and platelet activation, including von Willebrand Factor (VWF) antigen, soluble thrombomodulin, soluble P-selectin, and soluble CD40 ligand, as well as coagulation factors, endogenous anticoagulants, and fibrinolytic enzymes. We compared the level of each marker in ICU patients, non-ICU patients, and controls, where applicable. We assessed correlations between these laboratory results with clinical outcomes, including hospital discharge and mortality. Kaplan–Meier analysis was used to further explore the association between biochemical markers and survival. Findings 68 patients with COVID-19 were included in the study from April 13 to April 24, 2020, including 48 ICU and 20 non-ICU patients, as well as 13 non-hospitalised, asymptomatic controls. Markers of endothelial cell and platelet activation were significantly elevated in ICU patients compared with non-ICU patients, including VWF antigen (mean 565% [SD 199] in ICU patients vs 278% [133] in non-ICU patients; p<0·0001) and soluble P-selectin (15·9 ng/mL [4·8] vs 11·2 ng/mL [3·1]; p=0·0014). VWF antigen concentrations were also elevated above the normal range in 16 (80%) of 20 non-ICU patients. We found mortality to be significantly correlated with VWF antigen (r = 0·38; p=0·0022) and soluble thrombomodulin (r = 0·38; p=0·0078) among all patients. In all patients, soluble thrombomodulin concentrations greater than 3·26 ng/mL were associated with lower rates of hospital discharge (22 [88%] of 25 patients with low concentrations vs 13 [52%] of 25 patients with high concentrations; p=0·0050) and lower likelihood of survival on Kaplan–Meier analysis (hazard ratio 5·9, 95% CI 1·9–18·4; p=0·0087). Interpretation Our findings show that endotheliopathy is present in COVID-19 and is likely to be associated with critical illness and death. Early identification of endotheliopathy and strategies to mitigate its progression might improve outcomes in COVID-19. Funding This work was supported by a gift donation from Jack Levin to the Benign Hematology programme at Yale, and the National Institutes of Health.