ArticlePDF Available

Low level laser therapy as a modality to attenuate cytokine storm at multiple levels, enhance recovery, and reduce the use of ventilators in COVID-19

Authors:

Abstract and Figures

S Mokmeli MD, M Vetrici MD. Low level laser therapy as a modality to attenuate cytokine storm at multiple levels, enhance recovery, and reduce the use of ventilators in COVID-19. Can J Respir Ther 2020;56:1-7. doi: 10.29390/cjrt-2019-015. The global pandemic COVID-19 is a contagious disease and its mortality rates ranging from 1% to 5% are likely due to acute respiratory distress syndrome (ARDS), and cytokine storm. A significant proportion of patients who require intubation succumb to the disease, despite the availability of ventilators and the best treatment practices. Researchers worldwide are in search of anti-inflammatory medicines in the hope of finding a cure for COVID-19. Low-level laser therapy (LLLT) has strong, anti-inflammatory effects confirmed by meta-analyses, and it may be therapeutic to ARDS. LLLT has been used for pain management, wound healing, and other health conditions by physicians, physiotherapists, and nurses worldwide for decades. In addition, it has been used in veterinary medicine for respiratory tract disease such as pneumonia. Laser light with low-power intensity is applied to the surface of the skin to produce local and systemic effects. Based on the clinical experience, peer-reviewed studies, and solid laboratory data in experimental animal models, LLLT attenuates cytokine storm at multiple levels and reduces the major inflammatory metabolites. LLLT is a safe, effective, low-cost modality without any side-effects that may be combined with conventional treatment of ARDS. We summarize the effects of LLLT on pulmonary inflammation and we provide a protocol for augmenting medical treatment in COVID-19 patients. LLLT combined with conventional medical therapy has the potential to prevent the progression of COVID-19, minimize the length of time needed on a ventilator, enhance the healing process, and shorten recovery time.
Content may be subject to copyright.
Can J Respir Ther Vol 56 1
This open-access article is distributed under the terms of the Creative Commons Attribution Non-Commercial License (CC BY-NC) (http://
creativecommons.org/licenses/by-nc/4.0/), which permits reuse, distribution and reproduction of the article, provided that the original work is
properly cited and the reuse is restricted to noncommercial purposes. For commercial reuse, contact editor@csrt.com
Published online at https://www.cjrt.ca on 21 July 2020
RESEARCH ARTICLE
Low level laser therapy as a modality to attenuate cytokine
storm at multiple levels, enhance recovery, and reduce
the use of ventilators in COVID-19
Soheila Mokmeli MD Anesthesiologist1, Mariana Vetrici MD, PhD2
S Mokmeli MD, M Vetrici. Low level laser therapy as a modality to attenuate cytokine storm at multiple levels, enhance recovery, and reduce
the use of ventilators in COVID-19. Can J Respir Ther 2020;56:1–7. doi: 10.29390/cjrt-2019-015.
The global pandemic COVID-19 is a contagious disease and its mortality rates ranging from 1% to 5% are likely due to acute respiratory distress syndrome
(ARDS), and cytokine storm. A significant proportion of patients who require intubation succumb to the disease, despite the availability of ventilators and
the best treatment practices. Researchers worldwide are in search of anti-inflammatory medicines in the hope of finding a cure for COVID-19. Low-level
laser therapy (LLLT) has strong, anti-inflammatory effects confirmed by meta-analyses, and it may be therapeutic to ARDS. LLLT has been used for pain
management, wound healing, and other health conditions by physicians, physiotherapists, and nurses worldwide for decades. In addition, it has been used
in veterinary medicine for respiratory tract disease such as pneumonia. Laser light with low-power intensity is applied to the surface of the skin to produce
local and systemic effects. Based on the clinical experience, peer-reviewed studies, and solid laboratory data in experimental animal models, LLLT attenu-
ates cytokine storm at multiple levels and reduces the major inflammatory metabolites. LLLT is a safe, effective, low-cost modality without any side-effects
that may be combined with conventional treatment of ARDS. We summarize the effects of LLLT on pulmonary inflammation and we provide a protocol
for augmenting medical treatment in COVID-19 patients. LLLT combined with conventional medical therapy has the potential to prevent the progression
of COVID-19, minimize the length of time needed on a ventilator, enhance the healing process, and shorten recovery time.
Key Words: COVID-19; ARDS; cytokine storm; low level laser therapy; anti-inflammator y; ventilator; photobiomodulation
INTRODUCTION
What is low level laser therapy?
Low level laser therapy (LLLT) is also known as cold laser therapy or photo-
biomodulation therapy. LLLT utilizes visible light and infrared laser beams
in the range of 450–1000 nm. Single wavelength or monochromatic light is
emitted from a low-intensity laser diode (<500 mW). The light source is
placed in contact with the skin, allowing the photon energy to penetrate
tissue, where it interacts with various intracellular biomolecules to restore
normal cell function and enhance the body’s healing processes [1]. This
contrasts with the thermal effects produced by the high-power lasers that are
used in cosmetic and surgical procedures to destroy tissue [1], as mentioned
in the PubMed Medical Subject Heading (MeSH) subheading for LLLT.
LLLT effects are not due to heat but rather to a photochemical reaction
that occurs when a photoacceptor molecule within the cell absorbs a pho-
ton of light, becomes activated, and changes the cell’s membrane permea-
bility and metabolism. Presently, cytochrome c oxidase, opsins and their
associated calcium channels, and water molecules have been identified as
the main mediators of the photochemical mechanisms [2]. This leads to
increased mRNA synthesis and cell proliferation. LLLT produces reactive
oxygen species (ROS) in normal cells, but ROS levels are lowered when it
is used in oxidatively stressed cells, like in animal models of disease. LLLT
up-regulates antioxidant defenses and decreases oxidative stress [2].
Low-level lasers are a safe, noninvasive technology approved by both
the US Food and Drug Administration and Health Canada for several
chronic and degenerative conditions, temporary pain relief, cellulite
treatment, body contouring, lymphedema reduction, hair growth, and
chronic musculoskeletal injuries. LLLT increases microcirculation, lym-
phatic drainage, and cellular metabolism, thereby relieving many acute
and chronic conditions.
The MeSH database in PubMed contains more than 7000 articles on
LLLT. The effects of LLLT have been confirmed through several meta-
analysis studies and include anti-inflammatory [3] and analgesic effects [4],
tissue healing [5], treating tendinopathy [6], and improving lymphedema
[7]. Recent lab and animal studies suggest LLLT is ready for clinical trials
over myocardial infarction [5]. In 2010, a meta-analysis concluded that there
was strong evidence of an anti-inflammatory effect of LLLT [3].
To date, published reports indicate that LLLT up-regulates antioxi-
dant defenses and decreases ROS in oxidatively stressed cells and animal
models of disease. The anti-inflammatory effect of LLLT directly
addresses the main pathology of disorders such as musculoskeletal,
lungs, wounds, brain, trauma, etc. LLLT reduces NF-kB, a protein com-
plex that controls transcription of DNA, in pathological conditions.
Reports have shown reductions in reactive nitrogen species and prosta-
glandins in various animal models [2].
LLLT has diverse effects [8]:
reduces pain related to inflammation via dose-dependent reduc-
tion of prostaglandin E2, prostaglandin-endoperoxide synthase-2,
IL-1, IL-6, TNFa, as well as the cellular influx of neutrophils, oxi-
dative stress, edema, and bleeding;
1Canadian Optic and Laser Center (Training Institute), Victoria, BC, Canada
2Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
Correspondence: Soheila Mokmeli, Canadian Optic and Laser Center (Training Institute), 744A Lindsay Street, Victoria, BC V8Z 3E1, Canada.
Tel.: +1 (250) 480-7868, E-mail: dr.mokmeli@yahoo.com Mariana A. Vetrici, Department of Biological Sciences, University of Lethbridge, 4401 University
Drive, Lethbridge, AB T1K 3M4, Canada, Tel.: +1 (865) 888-3095, E-mail: marianavetrici@gmail.com
Soheila Mokmeli and Mariana Vetrici
2 Can J Respir Ther Vol 56
decreases edema and swelling by increasing lymphatic drainage;
increases collagen and protein production, and cell proliferation;
accelerates wound healing and scar formation;
improves quality and tensile strength of tissue;
stimulates nerve function and regeneration;
accelerates bone regeneration and remineralization;
reduces the pain threshold and enhances endorphins;
washes inflammatory debris away from the injured site; and
augments blood flow.
LLLT has been used in respiratory tract diseases since 1978. Empirical
practice on over 1000 patients produced data pertaining to chronic pneu-
monia, acute pneumonia, asthma, and chronic bronchitis in children,
adults, and elderly. Common findings include reduced chest pain and
heaviness; normalization of respiratory function; improved blood, immu-
nological, and radiological parameters; and shortened recovery times. In
community-acquired pneumonia, intravenous LLLT of blood added to
conventional treatment significantly promoted the bactericidal activity of
neutrophils. In asthma, the addition of LLLT was more effective than
medical treatment alone and it shortened the duration of treatment and
recovered bronchial sensitivity to sympathomimetics [9–11]. In newborns
with pneumonia, LLLT combined with conventional medical regimens
optimized the treatment infectious and inflammatory diseases, reduced
the incidence of complications, and shortened recovery periods [12].
LLLT is a well-known treatment modality in veterinary medicine.
Upper and lower respiratory conditions in dogs and cats are common, and
viral and bacterial infections are often highly contagious. Regardless of
etiology, inflammation is the major pathology of these conditions. The
addition of LLLT to conventional treatment alleviates symptoms and stim-
ulates the healing process in tissues. General guidelines for the use of laser
therapy in animals and protocols for specific conditions are published [13].
The pathogenesis of COVID-19 in respiratory tract
Coronaviruses are a large group of viruses that affect animals. In humans,
they produce diseases such as the common cold, severe acute respiratory
syndrome (SARS) and Middle East respiratory syndrome. The disease
caused by the novel coronavirus, SARS-CoV-2, has been named COVID-
19 and the clinical manifestations range from asymptomatic to severe
acute respiratory distress syndrome (ARDS) to death [14].
Respiratory viruses infect either the upper or lower airways. Typical
upper-respiratory infections are milder, more contagious, and spread eas-
ily, whereas lower-respiratory infections spread much less frequently but
are more severe and dangerous. SARS-CoV-2 appears to infect both
upper and lower airways. It spreads while still limited to the upper air-
ways, before traveling into the deeper respiratory tract and leading to
severe symptoms [15].
SARS-CoV-2 attaches to a protein called angiotensin converting
enzyme (ACE2), on the surface of cells in the respiratory tract. As SARS-
CoV-2 attacks the cells, dead cells flow down and block the airways with
debris while the virus moves deeper into the lungs. Breathing becomes
difficult because the lungs become clogged with dead cells and fluid.
The immune system attacks the virus causing inflammation and fever. In
severe cases, the immune system goes wild, causing more damage to the
lungs than the actual virus. Blood vessels dilate to increase blood flow
and become more permeable to maximize transport of chemical and cel-
lular mediators the infection site. Inevitably, the lungs get filled with
fluid. This exaggerated immune response is called cytokine storm and it
leads to ARDS, fever, multiorgan failure, and death [15, 16].
During cytokine storm, the immune system attacks indiscriminately
without clearing the specific targets. Cytokine storm also affects other
organs, especially if people already have chronic diseases [15]. The sever-
ity of cytokine storm determines who is hospitalized and who will be
treated in the intensive care unit (ICU). The classification of COVID-19
is summarized in Table 1 [17].
The morbidity and mortality of COVID-19 are due to excessive
inflammatory cytokine production and immune hyperactivity. Alveolar
macrophage activation and cytokine storm are the main pathogenesis of
severe COVID-19. The pathological features include exudation and
hemorrhage, epithelial injuries, infiltration of macrophages into the
lungs, and fibrosis of lung tissue. The mucous plug with fibrinous exu-
date in the alveoli and the activation of alveolar macrophage are charac-
teristic abnormalities [18, 19]. Chemical and genetic studies have shown
that the pulmonary endothelium is a key component of the cytokine
storm. Therefore, modulation of the involved cellular signaling pathways
may have therapeutic effects [20, 21].
COVID-19 begins when SARS-CoV-2 uses ACE2 as the entry recep-
tor for infection [22]. This induces ACE2 downregulation and shedding.
Loss of ACE2 from the endothelium causes dysfunction of the renin-an-
giotensin system, and it enhances inflammation and vascular permeabil-
ity. Shedding of ACE2 from the endothelium releases enzymatically
active soluble ACE2 (sACE2), which is tightly linked to tumor necrosis
factor alpha (TNF-α) production in cell culture [23].
Multiple signaling pathways are activated during an immune
response and cytokine storm. The P2X purinoceptor 7 (P2X7r) is major
TABLE 1
The staging and classication of COVID-19 [17]
Class Symptoms Imaging Respiratory criteria
Mild infection Mild Negative signs of pneumonia Normal
Moderate infection Fever and upper respiratory
tract symptoms
Positive signs of pneumonia Normal
Severe infection Fever, upper and lower
respiratory tract symptoms
>50% lesion progression
within 24–48 hours
Respiratory rate ≥ 30 /min
O2 saturation ≤ 93% at rest
Arterial partial pressure of O2 (PaO2)/oxygen concentration (FiO2)
≤ 300 mm Hg
Critical infection Respiratory failure requiring
mechanical ventilator
and (or)
presence of shock
and (or)
other organ failure that requires
monitoring and (or) treatment in
the ICU
> 50% lesion progression
within 24–48 hours
Early stage:
• Oxygenation index 100.1–149.9 mmHg.
• Respiratory system compliance (RSC) ≥ 30 ml/cmH2O.
• No organ failure other than the lungs.
Middle stage:
• 60 mmHg < O2 index ≤ l00 mmHg.
• 30 mL/cmH2O > RSC ≥ 15 mL/cmH2O.
• Maybe complicated by mild or moderate dysfunction of other organs.
Late stage:
• O2 index ≤ 60 mmHg.
• RSC < 15 mL/cmH2O.
Diffuse consolidation of both lungs that requires the use of extracorpo-
real membrane oxygenation or failure of other vital organs.
Note: A conrmed case is based on the epidemiological history (including cluster transmission), clinical symptoms (fever and respiratory symptoms), lung imaging,
and results of SARS-CoV-2 nucleic acid detection and serum-specic antibodies [17].
Low level laser therapy for COVID-19
Can J Respir Ther Vol 56 3
factor involved in activation of the cytokine storm and lung pathology in
response to viruses [24, 25], infection, inflammation, hypoxia, or trauma
[26]. P2X7r is an adenosine triphosphate (ATP) gated, nonselective cat-
ion channel, allowing Ca2+ and Na+ influx and K+ efflux. Extracellular
ATP plays a central role in apoptotic cell death [27], the induction of
inflammation [28], and mitochondrial failure in monocytes [29]. P2X7r
mediates ATP-induced cell death in different cells and it promotes
assembly and release of proinflammatory interleukins (IL -1β and IL-18)
from immune cells after exposure to lipopolysaccharide and ATP [27].
P2X7r is constitutively expressed in many cells, including respiratory epi-
thelial cells and most immune cells like neutrophils, monocytes, macro-
phages, dendritic, natural killer, B and T lymphocytes [27].
Studies stratified COVID-19 patients as: (i) severe symptoms and
ICU admission and (ii) mild and moderate symptoms requiring hospital-
ization but not ICU [17, 19]. The severe patients have significantly
higher levels of plasma pro-inflammatory factors (IL-2, IL-7, IL -10,
GSCF, IP-10, MCP-1, MIP1A, TNF-α) [19] and (IL-2, IL-6, IL-10, TNF-α)
[18] than non-ICU patients, and they were likely in cytokine storm [17,
19]. These findings justify the use of IL-6 receptor antagonists [18, 19];
however, a therapy to reduce inflammation at multiple levels, such as
LLLT, could be more successful in controlling the unbalanced immune
response (Figure 1).
The effects of LLLT on pulmonary inflammation
LLLT is effective against cytokine storm and ARDS while promoting
healing and tissue regeneration. Experimental and animal models of pul-
monary disease and infection have revealed multiple cellular and molec-
ular effects, which are both local and systemic. LLLT reduces
inflammation without impairing lung function in acute lung injuries
and is a promising therapeutic approach for lung inflammatory diseases
such as Chronic obstructive pulmonary disease [26].
In murine models of acute inflammation of the airways and lungs,
transcutaneous LLLT delivered over the trachea decreases pulmonary
microvascular leakage [30, 31], IL-1b levels [26, 30], IL -6 [26, 32],
MIP-2 mRNA expression [30], and intracellular ROS production [24].
LLLT produces anti-inflammator y effects on tracheal hyperactivity, and
reduces neutrophil influx [26, 30, 32–34] by inhibiting COX-2-derived
metabolites [33]. In ARDS, LLLT elevates cyclic adenosine monophos-
phate [32, 34], a signaling molecule that stimulates IL-10 and G-CSF expres-
sion and blocks TNF-a and MIP-1. LLLT also reduces TNF-a levels in
bronchoalveolar lavage fluid and alveolar macrophages [26, 31–34]. In hem-
orrhagic lesions of the lungs, LLLT significantly reduces the hemorrhagic
index and myeloperoxidase activity, to levels comparable to Celecoxib [35].
LLLT contributes to the resolution of inflammation by upregulating
IL-10 and downregulating P2X7r. LLLT changes the profile of inflamma-
tory cytokines and elevates IL-10 [26, 31, 36], known as human cytokine
synthesis inhibitory factor, in the lung and abolishes lung inflammation
via a reduction of inflammatory cytokines and mast cell degranulation
[31]. LLLT decreases collagen deposition as well as the expression of the
P2X7r [26].
LLLT contributes to healing by promoting apoptosis of inflamma-
tory cells while suppressing apoptotic pathways in lung tissue. In a model
of acute lung injury, LLLT reduced DNA fragmentation and apoptotic
pathways via increased B-cell-lymphoma-2 (Bcl-2), the key regulator of the
intrinsic or mitochondrial pathway for apoptosis, in alveolar epithelial
cells while promoting DNA fragmentation in inflammatory cells [37]. In
pulmonary idiopathic fibrosis, LLLT inhibits pro-inflammatory cyto-
kines and increases expression of proliferating cell nuclear antigen [38],
attenuates airway remodeling by balancing pro- and anti-inflammatory
cytokines in lung tissue, and inhibiting fibroblast secretion of the
pro-fibrotic cytokines [36].
LLLT provides synergy in combination with medical treatment. It has
a synergic anti-inflammatory action over alveolar macrophages pretreated
with N-acetyl cysteine, an effective oral medicine for coughs and some lung
conditions [39]. The synergic effects of LLLT combined with conventional
treatments were reported on over 1000 patients in Russian studies [9–11].
FIGURE 1
The effects of SARS-CoV-2 on alveolar cell and cytokine storm.
Soheila Mokmeli and Mariana Vetrici
4 Can J Respir Ther Vol 56
Extended time on ventilators causes lung injury but LLLT minimizes
this side effect. In experimental models of ventilator-induced lung injury
(VILI), LLLT following VILI resulted in lower injury scores, decreased
total cell count and neutrophil count in bronchoalveolar lavage, and
reduced alveolar neutrophil infiltration. LLLT in an experimental model
of VILI in rats demonstrated the anti-inflammatory effect via decreased
lung injury scores and lower counts of neutrophils in alveolar, intersti-
tial, and bronchial lavage [39] (Figure 2).
Evidence from the literature supports the use of LLLT for the treat-
ment of COVID-19.
It has significant anti-inf lammatory effects confirmed by meta-
analyses. Eleven cell studies, 27 animal studies, and another six
animal studies for drug comparisons and LLLT interactions veri-
fied that there is strong evidence of an anti-inflammatory effect of
LLLT. The scale of the anti-inf lammatory effect is not significantly
different than non-steroidal anti-inflammatory drugs, but it is
slightly less than glucocorticoid steroids [3].
It has diverse applications and effects confirmed through several
meta-analysis studies include analgesia [4], tissue healing [5], treat-
ing tendinopathy [6], and improved lymphedema [7].
LLLT is approved by the US FDA and Health Canada for several
chronic and degenerative conditions, temporary pain relief, cellu-
lite treatment, body contouring, lymphedema reduction, and hair
growth. It has been used in veterinary medicine for upper and
lower respiratory conditions in dogs and cats [13].
It has been used for human respiratory tract disease. Empirical use
on over 1000 patients produced data pertaining to chronic pneu-
monia, acute pneumonia, asthma, and chronic bronchitis in chil-
dren, adults, and the elderly [9–12]. Light therapy and LLLT has
been mentioned as a potential treatment for pandemic coronavirus
infections [40].
The anti-inf lammatory effect of LLLT in lung inflammation is con-
firmed in at least 14 experimental animal studies. LLLT attenuates
cytokine storm at multiple levels and reduces the major inflamma-
tory metabolites such as IL-6 and TNF-α. IL-6 antagonists are
being investigated for treating COVID-19 but LLLT reduces the
production of IL-6, as well as other chemokines and metabolites
[26–39, 41].
There are US FDA and Health Canada approved laser machines
for pain management, lymphedema after breast cancer surgery,
and cellulite treatments that can be used and set to treat lung
inflammation.
LLLT is an affordable modality compared with other treatments
and medicines like IL-6 antagonists. LLLT is a safe, effective, low-
cost modality without any reported side-effects compared with
other approaches. A laser machine costs Can$35,000.00–
200,000.00, and each machine can fully treat 20,000 patients for
COVID-19. In comparison, an IL-6 antagonist costs US$1000.00
per injection, and each patient would need 3–6 injections for com-
plete COVID-19 treatment. Treating 20,000 patients would cost
US$ 60,000,000.00–US$ 120,000,000.00.
Based on this information, LLLT will accelerate recovery from COVID-
19 and will get patients off ventilator support and out of the ICU more
rapidly. This could significantly decompress our severely overburdened
health care systems.
Therapeutic technique and dosage of LLLT
Laser dose is the amount of energy delivered per second per cm2. The effect
of laser therapy is related to the amount of laser energy per cm2. The Arndt-
Schultz Law is considered the standard to describe the dose dependent
effects of LLLT [42]. The minimum therapeutic dose for a bio-stimulatory
effect for red and infrared laser is 0.01 J/cm2 while for ultraviolet, blue,
green laser it is 0.001 J/cm2. LLLT has a noticeable biphasic dose response.
The effective stimulation dose is 1 J/cm2 on the target tissue. Doses greater
than 10 J/cm2 produces inhibitory effects. The inhibitory effects are used
in conditions requiring inhibition and suppression [2].
FIGURE 2
The effects of SARS-CoV-2 versus LLLT on cytokine storm and lung tissue.
Low level laser therapy for COVID-19
Can J Respir Ther Vol 56 5
Therapeutic protocol: early phase of COVID-19: (Figure 3, Table 2)
Laser parameters:
Laser type: infrared laser (780–900 nm), or red laser (630–660 nm)
Average power: 50–100 mW
Dose: 4–6 J/cm2
Area: 10 cm2
Time: 1–2 minutes/cm2
Sessions: 3–8 once-daily sessions
Laser probe positions:
Intranasal: 2 minutes, noncontact technique
Over right and left tonsils: transcutaneous (place laser over the
skin)
Over the trachea: transcutaneous
Over the veins in the cubital areas: transcutaneous blood laser
therapy, 10–15 minutes
Therapeutic protocol: medium–severe phase of COVID-19:
(Figure 3, Table 3)
Laser parameters:
Laser type: infrared laser (780–900 nm) or red laser (630–660 nm)
Average power: 50–100 mW
Dose: 6–10 J/cm2
Area: 10 cm2
Time: 2–3 minutes/cm2
Sessions: 3–10 once-daily sessions
Laser Probe Positions:
Over the lungs: bilaterally over apical, middle, and lower lobes and
front and back of thorax, transcutaneous over the intercostal spaces
Over the trachea: transcutaneous
Over the bronchus: upper mediastinal area, transcutaneous
Over right and left tonsils: transcutaneous
Over the veins in the cubital areas: transcutaneous blood laser
therapy; 10–15 minutes
Contraindications and side effects of LLLT [42]
Although LLLT is safe and noninvasive and there are no reports of muta-
genicity, genotoxicity, or carcinogenicity of LLLT after 60 years of its use.
However, there are some contraindications:
work over the site of tumors and cancer;
benign tumors with possibility of converting to malignant tumors;
FIGURE 3
LLLT for COVID-19.
TABLE 2
Therapeutic protocol: Early phase of COVID-19
Laser system parameters
Wavelengths Infrared laser (780–900 nm), or red
laser (630–660 nm)
Average power 50–100 mW
Dose 4–6 J/cm2
Area 10 cm2
Sessions 3–8 once-daily sessions
Laser probe positions
Intranasal:
1 minute/cm2 (100 mW)
2 minutes/cm2 (50 mW)
Noncontact technique
Over right and left tonsils
1 minute/cm2 (100 mW)
2 minutes/cm2 (50 mW)
Transcutaneous
(place laser over the skin)
Over the trachea
1 minute/cm2 (100 mW)
2 minutes/cm2 (50 mW)
Transcutaneous
Over the veins in the cubital areas
8 minute/cm2 (100 mW)
15 minutes/cm2 (50 mW)
Transcutaneous blood laser therapy
Soheila Mokmeli and Mariana Vetrici
6 Can J Respir Ther Vol 56
the first 3 months of pregnancy (in the second and third trimes-
ters, avoid work on abdominal and spine area); and
light sensitivity conditions.
Precautions [42]
epiphyseal line in children;
glands: avoid ovaries, testes;
in patients with severe end organ damage: heart, kidney, liver, and
lung;
epilepsy: the possibility of ner ve discharge is increased in LLLT,
especially with low-frequency protocols, 5–10 HZ.
Side effects of LLLT
Optical side effects
Because of the high intensity of lasers and the absorption of its wave-
lengths by different parts of ocular system, there is a possibility of dam-
age to the eyes. It is important to use protective glasses that can absorb
the specific wavelength. Protective glasses for each wavelength are differ-
ent; therefore, choose the protective goggles specified for each wave-
length. Both therapists and clients should wear protective goggles [42].
Early sense of healing
The analgesic effect of laser manifests earlier than its healing effect, and
the patients feel better because of this, but the actual tissue damage has
not yet healed. Patients feel relaxed and more energetic because the pain
is gone. However, they must allow enough time for recovery [42].
Fatigue and tiredness
Fatigue is the most common symptom following LLLT. This is due to
hormonal and metabolite changes after laser therapy that increase
expression natural pain killers like endorphins and enkephalins. These
metabolites induce relaxation and sleepiness [42].
Low blood pressure and dizziness
Very rarely, when the treated area is close to large blood vessels, a patient
may experience a temporary drop in the blood pressure and orthostasis.
This is due to vasodilatation and increased circulation to the limbs. To
avoid dizziness, it is recommended that patients drink fluids before
LLLT, and then wait for a few minutes before getting up from the supine
position [42].
CONCLUSION
COVID-19 is potentially lethal because of cytokine storm and ARDS.
Although most patients who contract COVID-19 may recover at home,
a significant proportion require hospitalization and (or) ICU treat-
ment. Many of the patients that are placed on ventilators succumb to
the disease despite the best treatment practices. Often, patients are
maintained on ventilators for longer than expected, and this may con-
tribute to ventilator induced lung injury while depleting the patient’s
convalescent resources. Modulation of inflammatory factors and a
boost to healing are necessary to help patients come off the ventilators.
LLLT is a safe and noninvasive modality that has been used for decades
in pain management, wound healing, and health conditions including
diseases of the respiratory tract. LLLT was combined successfully with
standard medical care to optimize response to treatments, reduce
inflammation, promote healing, and accelerate recovery times.
Scientific evidence shows that LLLT attenuates the inflammator y cyto-
kines and chemokines in cytokine storm at multiple levels. In addition,
LLLT promotes apoptosis of inflammatory cells and protects alveolar
cells from damage. These findings suggest that LLLT is a feasible
modality in the treatment of ARDS. LLLT can be added to the conven-
tional treatment in COVID-19 at different stages of the disease.
Because of its anti-inflammatory effect, and ability to shorten recovery
times, LLLT can reduce the need of ventilators in the healing process.
Clinical trials are necessary to objectively evaluate the effect of LLLT
on COVID-19 treatment and recovery.
Contributors
Soheila Mokmeli and Mariana Vetrici contributed to the conception
and design of the work.
Competing interests
All authors have completed the ICMJE uniform disclosure form at www.
icmje.org/coi_disclosure.pdf and declare: no financial relationships
with any organizations that might have an interest in the submitted work
in the previous 3 years; no other relationships or activities that could
appear to have influenced the submitted work.
Ethical approval
Informed consent was obtained from all participants.
REFERENCES
1. Cotler HB, Chow RT, Hamblin MR, Carroll J. The use of Low Level
Laser Therapy (LLLT) for musculoskeletal pain. MOJ Orthop Rheumatol
2015;2(5):00068. doi: 10.15406/mojor.2015.02.00068.
2. Hamblin MR. Mechanisms and applications of the anti-inflammatory
effects of photobiomodulation. AIMS Biophys 2017;4(3):337–61. doi:
10.3934/biophy.2017.3.337.
3. Bjordal JM, Lopes-Martins RAB, Iversen VV. The anti-inflammatory
mechanism of low level laser therapy and its relevance for clinical use in
physiotherapy. Medicine 2010. 15: 286–293 Corpus ID: 27687471. doi:
10.1179/ 1743288X10Y.0000000001.
4. Jang H, Lee H. Meta-analysis of pain relief effects by laser irradiation on
joint areas. Photomed Laser Surg 2012 Aug;30(8):405–17. doi: 10.1089/
pho.2012.3240.
5. Woodruff LD, Bounkeo JM, Brannon WM, et al. The efficacy of laser
therapy in wound repair: a meta-analysis of the literature. Photomed
Laser Surg 2004;22(3):241. Available at: https://www.ncbi.nlm.nih.gov/
pubmed/15315732 [Accessed 1 April 2020.]
6. Tumilty S, Munn J, McDonough S, Hurley DA, Basford JR, Baxter GD.
Low level laser treatment of tendinopathy: a systematic review with
meta-analysis. Photomed Laser Surg 2010;28(1):3–16. Available at:
https://search.pedro.org.au/search-results/record-detail/25413
[Accessed 1 April 2020.]
7. Smoot B, Chiavola-Larson L, Lee J, Manibusan H, Allen DD. Effect of
low-level laser therapy on pain and swelling in women with breast can-
cer-related lymphedema: a systematic review and meta-analysis. J Cancer
Survivorship 2014;9(2). doi: 10.1007/s11764-014-0411-1.
TABLE 3
Therapeutic protocol: medium–severe phase of COVID-19
Laser system parameters
Wavelengths Infrared laser (780–900 nm), or red laser
(630–660 nm)
Average power 50–100 mW
Dose 6–10 J/cm2
Area 10 cm2
Sessions 3–10 once-daily sessions
Laser probes positions
Intranasal:
1 minute/cm2 (100 mW)
2 minutes/cm2 (50 mW)
Noncontact technique
Over right and left tonsils
1 minute/cm2 (100 mW)
2 minutes/cm2 (50 mW)
Transcutaneous
(place laser over the skin)
Over the trachea
1 minute/cm2 (100 mW)
2 minutes/cm2 (50 mW)
Transcutaneous
Over the veins in the cubital
areas
8 minute/cm2 (100 mW)
15 minutes/cm2 (50 mW)
Transcutaneous blood laser therapy
Over the lungs
1:30–2 minute/cm2 (100 mW)
2–3 minutes/cm2 (50 mW)
Bilaterally over apical, middle, and lower
lobes, front and back of thorax; transcuta-
neous over the intercostal spaces
Over the bronchus
1:30–2 minute/cm2 (100 mW)
2–3 minutes/cm2 (50 mW)
Upper mediastinal area: transcutaneous
Low level laser therapy for COVID-19
Can J Respir Ther Vol 56 7
8. Simunovic Z. (chapter XIV); Gerard Danhof (chapter V); Laser in medi-
cine and dentistry. European Medical Laser Association, CIP, University
Library of Rijekam Croatia. ISBN: 953-6059-30-4; 2000
9. Amirov NB. Parameters of membrane permeability, microcirculation,
external respiration, and trace element levels in the drug-laser treatment
of pneumonia [Article in Russian]. Ter Arkh 2002;74(3):40–3.
10. Derbenev VA, Mikhailov VA, Denisov IN. Use of low-level laser therapy
(LLLT) in the treatment of some pulmonary diseases: ten-year experi-
ence. Proc SPIE 2000;4166:323–5. doi: 10.1117/12.389506.
11. Ostronosova NS. Outpatient use of laser therapy in bronchial asthma.
TerArkh 2006;78(3):414.
12. Erkinovna TB, Tulkunovna MH. Efficacy of laser therapy in infants with
infectious-inflammatory respiratory diseases. АКР ОНИМ: European
Science Review; 2006. ISSN: Печатный: 2310-5577. Available at: https://
cyberleninka.ru/article/n/efficacy-of-laser -therapy-in -infants-with-infec-
tious-inflammatory-respiratory-diseases [Accessed 1 April 2020.]
13. Arza RA. The chapter of upper and lower respiratory conditions: photo-
biomodulation. Laser therapy in veterinary medicine. 2017. doi:
10.1002/9781119220190.ch14.
14. Wu Z, McGoogan JM. Characteristics of and important lessons from the
coronavirus disease 2019 (COVID-19) outbreak in China: summary of a
report of 72 314 cases from the Chinese Center for Disease Control and
Prevention. JAMA 2020. doi: 10.1001/jama.2020.2648.
15. Yong E. Why the coronavirus has been so successful? Science 2020.
Available at: https://www.theatlantic.com/science/archive/2020/03/
biography-new-coronavirus/608338/ [Accessed 1 April 2020.]
16. Mehta P, McAuley DF, Brown M, et al. COVID-19: consider cytokine
storm syndromes and immunosuppression. Lancet 2020;395(10229):
P1033–4. doi: 10.1016/S0140-6736(20)30628-0.
17. Liang T. According to clinical experience; Handbook of COVID-19 pre-
vention and treatment. Zhejiang University School of Medicine,
Hangzhou; 2020.
18. Wang C, Xie J, Zhao L, et al. Alveolar macrophage activation and cyto-
kine storm in the pathogenesis of severe COVID-19. Preprint from
Research Square; 2020. doi: 10.21203/rs.3.rs-19346/v1.
19. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with
2019 novel coronavirus in Wuhan. Lancet China 2020. doi: 10.1016/
S0140-6736(20)30183-5.
20. Damjanovic D, Small CL, Jeyanathan M, et al. Immunopathology in
influenza virus infection: uncoupling the friend from foe. 2012 Clin
Immunol 144:5769. doi: 10.1016/j.clim.2012.05.005.
21. Teijaro JR, Walsh KB, Cahalan S, et al. Endothelial cells as regulators of
cytokine storms during influenza infection. Cell 2011;146:980e91.
Available at: https://thorax.bmj.com/content/thoraxjnl/early/2011/
12/14/thoraxjnl-2011-201437.full.pdf [Accessed 29 March 2020.]
22. Zhou P, Yang XL, Wang XG, et al. A pneumonia outbreak associated
with a new coronavirus of probable bat origin. Nature 2020. doi:
10.1038/s41586-020-2012-7.
23. Fu Y, Cheng Y, Wu Y. Understanding SARS-CoV-2-mediated inflamma-
tory responses: from mechanisms to potential therapeutic tools. Virol
Sin 2020. doi: 10.1007/s12250-020-00207-4.
24. Leyva-Grado VH, Ermler ME, Schotsaert M, et al. Contribution of the puri-
nergic receptor P2X7 to development of lung immunopathology during
influenza virus infection. mBio 2017. doi: 10.1128/mBio.00229-17.
25. Rosli S, Kirby FJ, Lawlor KE, et al. Repurposing drugs targeting the P2X7
receptor to limit hyperinflammation and disease during influenza virus
infection. Br J Pharmacol 2019. doi: 10.1111/bph.14787.
26. da Cunha Moraes G, Vitoretti LB, de Brito AA, et al. Low-level laser
therapy reduces lung inflammation in an experimental model of chronic
obstructive pulmonary disease involving P2X7 receptor. Oxid Med Cell
Longev 2018 Mar 4;2018:6798238. doi: 10.1155/2018/6798238.
27. Kawano A, Tsukimoto M, Noguchi T, et al. Involvement of P2X4 recep-
tor in P2X7 receptor-dependent cell death of mouse macrophages.
Biochem Biophys Res Commun 2012;419(2):374–80. doi: 10.1016/j.
bbrc.2012.01.156.
28. He Y, Taylor N, Fourgeaud L, Bhattacharya A. The role of microglial
P2X7: modulation of cell death and cytokine release. J Neuroinflammation
2017. doi: 10.1186/s12974-017-0904-8.
29. Martínez-García JJ, Martínez-Banaclocha H, Angosto-Bazarra D, et al.
P2X7 receptor induces mitochondrial failure in monocytes and compro-
mises NLRP3 inflammasome activation during sepsis. Nat Commun
2019;10:2711. doi: 10.1038/s41467-019-10626-x.
30. Aimbire F, Ligeiro de Oliveira AP, Albertini R et al. Low level laser ther-
apy (LLLT) decreases pulmonary microvascular leakage, neutrophil
influx and IL-1beta levels in airway and lung from rat subjected to LPS-
induced inflammation. Inflammation 2008;31(3):189–97. doi: 10.1007/
s10753-008-9064-4.
31. Miranda da Silva C, Peres Leal M, Brochetti RA, et al. Low level laser
therapy reduces the development of lung inflammation induced by form-
aldehyde exposure. PLoS One 2015 Nov 16;10(11):e0142816. doi:
10.1371/journal.pone.0142816.
32. Oliveira MC Jr, Greiffo FR, Rigonato-Oliveira NC, et al. Low level laser
therapy reduces acute lung inflammation in a model of pulmonary and
extrapulmonary LPS-induced ARDS. J Photochem Photobiol B
2014;134:57–63. doi: 10.1016/j.jphotobiol.2014.03.021.
33. Aimbire F, Albertine R, de Magalhães RG, et al. Effect of LLLT
Ga-Al-As (685 nm) on LPS-induced inflammation of the airway and
lung in the rat. Lasers Med Sci 2005;20(1):11–20. doi: 10.1007/
s10103-005-0339-9.
34. de Lima FM, Moreira LM, Villaverde AB, Castro-Faria-Neto HC,
Aimbire F. Low-level laser therapy (LLLT) acts as cAMP-elevating agent in
acute respiratory distress syndrome. Lasers Med Sci 2011 May
24;26(3):389–400. doi: 10.1007/s10103-010-0874-x.
35. Aimbire F, Lopes-Martins RA, Albertini R, et al. Effect of low-level laser
therapy on hemorrhagic lesions induced by immune complex in rat
lungs. Photomed Laser Surg 2007 Apr;25(2):112–7. doi: 10.1089/
pho.2006.1041.
36. de Brito AA, da Silveira EC, Rigonato-Oliveira NC, et al. Low-level laser
therapy attenuates lung inflammation and airway remodeling in a
murine model of idiopathic pulmonary fibrosis: relevance to cytokines
secretion from lung structural cells. J Photochem Photobiol B 2020
Jan;203:111731. doi: 10.1016/j.jphotobiol.2019.111731.
37. Sergio LPDS, Thomé AMC, Trajano LADSN, et al. Photobiomodulation
prevents DNA fragmentation of alveolar epithelial cells and alters the
mRNA levels of caspase 3 and Bcl-2 genes in acute lung injury.
Photochem Photobiol Sci 2018 Jul 11;17(7):975–83. doi: 10.1039/
c8pp00109j.
38. Rigonato-Oliveira NC, Brito AA, Moraes G, et al. Effect of low-level laser
therapy in a model of pulmonary idiopathic fibrosis. Eur Respir J
2015;46:PA945. doi: 10.1183/13993003.congress-2015.PA945; Available
at: https://erj.ersjournals.com/content/46/suppl_59/PA945 [Accessed
21 March 2020.]
39. Mafra de Lima F, Balbin Villaverde A, Albertini R, Ligeiro de Oliveira
AP, Castro Faria Neto HC, Aimbire F. Low-level laser therapy associated
to N-acetylcysteine lowers macrophage inflammatory protein-2 (MIP-2)
mRNA expression and generation of intracellular reactive oxygen species
in alveolar macrophages. Photomed Laser Surg 2010;28(6):763–71. doi:
10.1089/pho.2009.2638.
40. Enwemekaa CS, Bumahab VV, Masson-Meyersc DS. Light as a potential
treatment for pandemic coronavirus infections: a perspective. J
Photochem Photobiol B 2020;111891. doi: 10.1164/ajrccm-confer-
ence.2019.199.1_MeetingAbstracts.A6152.
41. Lucinda LMF, Fazza TF, Fonseca LMC, et al. Effect of low-level laser
therapy on experimental model of ventilator-induced lung injury.
American Thoracic Society; International Conference Abstracts; B31.
Acute lung injury & ARDS: transitional and mechanistic studies; 2019
May 22.
42. Tuner J, Hode L. Laser therapy. Clinical practice and scientic back-
ground. BookGra¨ngesberg, Sweden: Prima Books AB; 2002. ISBN:
91-631-1344-9
... In the last decades, light has been increasingly applied in research, survey, treatment, and therapy of biological objects (considering here the term light for near-ultraviolet (UV), visible, and nearinfrared (NIR) irradiation); it is named as low-level laser therapy (LLLT) and more generally as photobiomodulation (PBM) or photobiomodulation therapy (PBMT) (1,2). There are few known independent light-sensitive cellular pathways as the activation of ion channels on cell membrane and endoplasmic reticulum (3,4), though, light principally interacts with mitochondria, which is the main source of cellular reactive oxygen species (ROS) (5)(6)(7). ...
... To date, the most discussed photoacceptor is cytochrome c oxidase (CCO) (6,13). CCO absorbs red/NIR light within wavelengths ranging between 600-1000 nm, resulting in the activation of metabolic process as well as migration and proliferation of various cell types: neurons (650-660/808 nm at 3-90 J/cm 2 (3,4,14), fibroblasts (660 nm at 5 J/cm 2 (15), 660 nm at 3-8 J/cm 2 (16), 809 nm at 1.96-7.84 J/cm 2 (17)), endothelial cells (635 nm at 24 J/cm 2 (18), 650 nm at 30 J/cm 2 (19), 665 and 675 nm at 10 J/cm 2 (20), 808 nm at 60 J/cm 2 (21)), stem cells (660 nm (22,23) and 808 nm at 3 J/cm 2 (24)), keratinocytes (780 nm at 0.45-0.95 ...
... To date, the most discussed photoacceptor is cytochrome c oxidase (CCO) (6,13). CCO absorbs red/NIR light within wavelengths ranging between 600-1000 nm, resulting in the activation of metabolic process as well as migration and proliferation of various cell types: neurons (650-660/808 nm at 3-90 J/cm 2 (3,4,14), fibroblasts (660 nm at 5 J/cm 2 (15), 660 nm at 3-8 J/cm 2 (16), 809 nm at 1.96-7.84 J/cm 2 (17)), endothelial cells (635 nm at 24 J/cm 2 (18), 650 nm at 30 J/cm 2 (19), 665 and 675 nm at 10 J/cm 2 (20), 808 nm at 60 J/cm 2 (21)), stem cells (660 nm (22,23) and 808 nm at 3 J/cm 2 (24)), keratinocytes (780 nm at 0.45-0.95 ...
Article
Full-text available
The effect of UV/visible/NIR light (380/450/530/650/808/1064 nm) on ROS generation, mitochondrial activity, and viability is experimentally compared in human neuroblastoma cancer cells. The absorption of photons by mitochondrial photoacceptors in Complexes I, III, IV is in‐detail investigated by sequential blocking with selective pharmaceutical blockers. Complex I absorbs UV/blue light by heme P450, resulting in a very high rate (14 times) of ROS generation leading to cell death. Complex III absorbs green light, by cytochromes b, c1 and c, and possesses less ability for ROS production (7 times), so that only irradiation lower than 10 mW/cm2 cause an increase in cell viability. Complex IV is well‐known as the primary photoacceptor for red/NIR light. Light of 650/808 nm at 10–100 mW/cm2 generates a physiological ROS level about 20% of a basal concentration, which enhance mitochondrial activity and cell survival. While, 1064 nm light does not show any distinguished effects. Further, ROS generation induced by low‐intensity red/NIR light is compared in neurons, immune and cancer cells. Red light seems to more rapidly stimulate ROS production, mitochondrial activity, and cell survival than 808 nm. At the same time, different cell lines demonstrate slightly various rates of ROS generation, peculiar for their cellular physiology.
... It remains unclear COVID-19 infection pathway and its consequences, which may be related to receptors widely present in gustatory and olfactory cells, leading to partial or total perception loss for a prolonged period. It is not well understood why these cells have such prolonged malfunction and, in some cases, patients persist with prolonged taste or olfactory disturbances after COVID-19 infection [9,10]. Symptoms can vary, according to different studies, ranging from complete ageusia to parageusia or even dysgeusia. ...
... Symptoms can vary, according to different studies, ranging from complete ageusia to parageusia or even dysgeusia. In addition to that, symptoms appearance rate is also not well known, varying according to the methodology and the analyzed population [10]. The presence of concomitant neurological symptoms with taste impairment is well described and was observed in this patient [11], and the presence of neurological alterations might be speculated as the cause of taste impairment. ...
Article
Full-text available
Objective: The aim of this case report is to describe Photobiomodulation Therapy (PBMT) as an alternative for the management of ageusia following infection by COVID-19. Method and Materials: PBMT was performed using a 660 nm and 808 nm diode laser (1 J/point, 100 mW, 10 sec, 40 points on tongue) and taste perception recovery was assessed by a quantitative test with hyper-concentrated substances (acid, bitter, sweet and salt), using a 0-10 score. Taste tests were carried out before treatment, after 8 and 16 PBMT sessions. Results: Positive evolution of taste perception recovery was shown throughout PBMT sessions, at 8 and 16 sessions, demonstrating improvement in taste perception. Conclusions: PBMT using the parameters set for the present clinical case was shown to be effective, noninvasive and an option of therapy for the management of prolonged dysgeusia COVID-19 related.
... The minimal therapeutic dose for a bio-stimulant impact is 0.01 J/cm 2 for red and infrared lasers and 0.001 J/cm 2 for blue, ultraviolet, and green lasers. When employed in settings that need suppression, such as lowering the inflammatory response to avoid or minimize the flood of pro-inflammatory cytokines, dosages greater than 10 J/cm2 have been found to elicit inhibitory effects [15]. ...
... A recent study found that two weeks of light-emitting diode PBM therapy is effective in reducing non-specific low back pain and fatigue in working nurses [13], and phototherapy has been shown to improve pain, quality of life, and sleep disorders in people with fibromyalgia ACCEPTED MANUSCRIPT [14]. A review of 27 articles published between 2008 and 2017 showed that low-level laser therapy could be used to improve performance and make people feel less tired after hard exercise [15]. Applying PBM six hours before and immediately before an isokinetic exercise procedure on eighteen physically active men resulted in the same total effort being produced but with lower fatigue, which may enable the performance of further exercise sets [16]. ...
... LLLT is ready for clinical trials over myocardial infarction [9]. Recently, it is recommended to use as a modality to attenuate cytokine storm at multiple levels, enhance recovery, and reduce the use of ventilators in COVID-19 [1], [14] - [16]. ...
Article
Trong những năm gần đây, việc ứng dụng phương pháp điều trị Laser công suất thấp đa bước sóng đối với một số bệnh cấp tính, mãn tính và nâng cao chất lượng điều trị đã mang lại nhiều thành tựu tích cực trong y học. Nó ngày càng phổ biến trong cả thử nghiệm thực nghiệm và thử nghiệm lâm sàng. Các thiết bị laser quang trị liệu cho các ứng dụng cụ thể đang có nhu cầu rất lớn. Trong nghiên cứu này, một thiết bị quang trị liệu đa bước sóng phát ở vùng đỏ đến vùng hồng ngoại gần, vùng cửa sổ quang trị liệu, được phát triển. Thiết bị sử dụng các mô-đun laser, cho phép dễ dàng dẫn chùm tia laser vào vùng cần được trị liệu. Để thuận tiện cho người sử dụng, thiết bị tích hợp hệ thống điều khiển kỹ thuật số (Digital control), màn hình cảm ứng dễ dàng lựa chọn các thông số như công suất, bước sóng, chế độ hoạt tối ưu với mục đích điều trị cụ thể. Bài báo này trình bày thiết kế và chế tạo hoàn chỉnh thiết bị cũng như kỹ thuật chế tạo mô-đun laser. Các thông số chính của thiết bị trị liệu laser được đưa ra, đáp ứng các thông số kỹ thuật của thiết bị quang trị liệu, phù hợp cho điều trị các vết thương hở và mãn tính dựa trên cơ chế điều biến quang sinh.
... Magnesium, iron, and calcium, important micronutrients from millets and sorghum, have significantly reduced COVID-19 patients' oxygen requirements, and this is critical in reducing oxygen demand during intensive care support (Kluge et al., 2021). Marco-and micronutrients from small grains have generally been reported to lower probability of COVID-19 contagion and improve COVID-19 recovery (Mokmeli & Vetrici, 2020). ...
Chapter
Full-text available
In this chapter, we use the concept of everyday practice to highlight the plight of urban residents and what it means/takes to survive the ongoing COVID-19 pandemic in a water-insecure city. We use data from four Wards on differentiated locations relative to storage tanks supplying water and different water rationing zones. The data was collected from 2020 to 2021 (2 years). A stratified random sampling technique was used to select a study sample of 303 respondents. Of these, 200 household heads were interviewed at their place of residency, while the remaining 103 respondents gave interviews while waiting to draw water from boreholes dotted around the four residential areas. Our results suggest that the policies for managing the pandemic paid less attention to everyday practices of getting around the more than two-decade-old water challenges in the urban areas. The water challenges in the urban areas further exposed the residents to COVID-19 infection, and the pandemic widened the gendered and spatial inequalities to access to water. We conclude that the search for and concerted efforts to access water to manage and prevent COVID-19 infection were equally associated with high chances of being infected and/or spreading COVID-19. The ongoing COVID-19 pandemic is not the last water-demanding crisis we will experience. This calls for a paradigm shift in urban water and sanitation access planning to include alternative water sources – groundwater – at the initial stages of residential planning.
... Bệnh COVID-19 là 1 bệnh truyền nhiễm nguy hiểm, hiện đang là đại dịch trên thế giới và ở Việt Nam. Theo tin tổng hợp ngày 31 (11/2/2019). Bệnh lây truyền chủ yếu thông qua giọt bắn hô hấp, cũng có thể qua đường tiếp xúc. ...
Article
Bệnh COVID-19 là 1 bệnh truyền nhiễm nguy hiểm do virus SARS-CoV-2 gây ra và hiện đang là đại dịch trên thế giới và ở Việt Nam. Trong chiến lược khống chế bệnh dịch này vai trò của điều trị khi bị mắc bệnh là rất quan trọng. Liệu pháp quang điều hòa sinh học và quang động học kháng virus là hướng điều trị mới trong điều trị bệnh nhân COVID-19. Đây là hướng điều trị kết hợp quang điều hòa sinh học và quang động học kháng virus với dùng thuốc thông thường cho thấy tiềm năng cao cũng như hứa hẹn mang lại lợi ích to lớn của phương pháp mới trong việc điều trị bệnh nhân COVID-19.
... Therapeutic protocol: COVID-19 moderatesevere forms(23) Clinical trials have shown that the laser used in COVID-19 pneumonia has reduced respiratory symptoms by normalizing respiratory function; the recovery time has been significantly shortened and all blood, immunological and radiological parameters have improved(24). We mention the post-treatment results for the patients in the first randomized pilot study that involved patients with confirmed COVID-19, conducted in August 2020 by Dr. Sigman: PCR normalized; from 15.1 mg/dL to 1.23 mg/dL 7. Ferritin decreased from 359 ng/mL to 175 ng/mL 8. Clinical recovery was a total of 3 weeks, while the mean time is usually 6 to 8 weeks. ...
Article
Full-text available
Introduction. An unprecedented public health crisis has been triggered worldwide by SARS-CoV-2’s high contagiosity and it’s mortality rates of 1-5%. Although the majority of COVID-19 cases have a good outcome, there is a small percentage that develop severe pneumonia and citokine storm and may be in the need of mechanical ventilation. Methods. Identifying the exact drivers of the excessive inflammation and the biomarkers that can predict a hyperinflammatory response to SARS-CoV-2 would be extremly helpful in finding efficient anti-inflammatory interventions that may stop the progression to acute respiratory distress syndrome (ARDS). Results. In the search for such interventions we have identified the promising effect of low level LASER therapy (LLLT) on lung inflammation from COVID-19 pneumonia. Due to its well known anti-inflammatory effect and modulatory activity on immune cells, laser therapy may be able to decrease lung and systemic inflammation without affecting lung function in acute lung lesions, relieve respiratory symptoms, normalize respiratory function and stimulate the healing process of lung tissue. The recovery time may also be significantly shortened and all blood, immunological and radiological parameters may improve. Conclusions. This findings need further confirmation from clinical trials but we are hopeful for their contribution on the global battle against COVID-19 pandemic.
Article
Full-text available
The pandemic outbreak of Coronavirus disease 2019 (COVID-19) which is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2), is a new viral infection in all countries around the world. An increase in inflammatory cytokines, fever, dry cough, and pneumonia are the main symptoms of COVID-19. A shared of growing clinical evidence confirmed that cytokine storm correlates with COVID-19 severity which is also a crucial cause of death from COVID-19. The success of anti-inflammatory therapies in the recovery process of COVID-19 patients has been well established. Over the years, phototherapy (PhT) has been identified as a promising non-invasive treatment approach for inflammatory conditions. New evidence suggests that PhT as an anti-inflammatory therapy may be effective in treating acute respiratory distress syndrome (ARDS) and COVID-19. This review aims to a comprehensive overview of the direct and indirect effects of anti-inflammatory mechanisms of PhT in ARDS and COVID-19 patients.
Article
Since March 2020, scientists across the globe are looking for ways to combat COVID-19, a viral disease of recent times which was declared Pandemic by WHO. Mounting an incompetent immune reverberation and the surge in pro-inflammatory cytokines are the main cause for complications. Hence, it is hypothesized that photobiomodulation therapy (PBMT) using low-level light therapy (LLLT) might be a successful therapeutic treatment for COVID-19 victims since it has been shown to be successful in enhancing immunity, aiding in tissue healing, and reducing pro-inflammatory cytokines. The goal of this scoping review is to investigate the potential use of PBMT as a promising neo-strategy for an alternative therapy for the treatment of patients with COVID-19 in light of the significance of underlying pathways in the mitigation, therapeutic interventions, and recovery of chronic diseases like COVID-19. The findings of the review revealed that PBMT using a low-level laser has been proven to be effective in treating COVID-19’s initial stages and prevent the progression to severe form of the disease. This approach needs to be confirmed by clinical trials and larger studies that include a wider population. Because of its proven effectiveness in reducing inflammation, thrombosis and accelerate wound healing, PBMT may be a promising approach to reduce the severity of COVID-19, especially in the lungs. Based on the findings of the review, we propose a novel hypothesis of utilizing PBMT to enhance the biodiversity of oral microbiome which will help in extenuating the imperilment of SARS CoV-2 infection.
Chapter
Integrative veterinary medicine (IVM) has the potential to advance the health and recovery capacity of patients with acute and chronic diseases through tested, science‐based treatment strategies. This chapter discusses the types of IVM care and legal, regulatory, and ethical concerns associated with provision of IVM. IVM presents a gamut of opportunities and difficulties when it comes to ethical practice. As scientific support for IVM grows, its legitimacy and acceptance are similarly expanding. For all IVM therapies, the first step in treating animals safely and effectively involves an accurate assessment and delineation of the patient's problems. Herbal medicine refers to the practice of prescribing plant products for the treatment of disease. Evidential support concerning use of plant products in veterinary patients remains scarce. Some critics have suggested that IVM practitioners may be opportunistic when offering clients treatments that support comfort, quality of life, and mobility near the end of life.
Article
Full-text available
The recent outbreak of COVID-19, which continues to ravage communities with high death tolls and untold psychosocial and catastrophic economic consequences, is a vivid reminder of nature's capacity to defy contemporary healthcare. The pandemic calls for rapid mobilization of every potential clinical tool, including phototherapy—one of the most effective treatments used to reduce the impact of the 1918 “Spanish influenza” pandemic. This paper cites several studies showing that phototherapy has immense potential to reduce the impact of coronavirus diseases, and offers suggested ways that the healthcare industry can integrate modern light technologies in the fight against COVID-19 and other infections. The evidence shows that violet/blue (400–470 nm) light is antimicrobial against numerous bacteria, and that it accounts for Niels Ryberg Finsen's Nobel-winning treatment of tuberculosis. Further evidence shows that blue light inactivates several viruses, including the common flu coronavirus, and that in experimental animals, red and near infrared light reduce respiratory disorders, similar to those complications associated with coronavirus infection. Moreover, in patients, red light has been shown to alleviate chronic obstructive lung disease and bronchial asthma. These findings call for urgent efforts to further explore the clinical value of light, and not wait for another pandemic to serve as a reminder. The ubiquity of inexpensive light emitting lasers and light emitting diodes (LEDs), makes it relatively easy to develop safe low-cost light-based devices with the potential to reduce infections, sanitize equipment, hospital facilities, emergency care vehicles, homes, and the general environment as pilot studies have shown.
Preprint
Full-text available
The coronavirus disease-19 (COVID-19) caused by SARS-CoV-2 infection can lead to a series of clinical settings from non-symptomatic viral carriers/spreaders to severe illness characterized by acute respiratory distress syndrome (ARDS)1,2. A sizable part of patients with COVID-19 have mild clinical symptoms at the early stage of infection, but the disease progression may become quite rapid in the later stage with ARDS as the common manifestation and followed by critical multiple organ failure, causing a high mortality rate of 7-10% in the elderly population with underlying chronic disease1-3. The pathological investigation in the lungs and other organs of fatal cases is fundamental for the mechanistic understanding of severe COVID-19 and the development of specific therapy in these cases. Gross anatomy and molecular markers allowed us to identify, in two fatal patients subject to necropsy, the main pathological features such as exudation and hemorrhage, epithelium injuries, infiltration of macrophages and fibrosis in the lungs. The mucous plug with fibrinous exudate in the alveoli and the activation of alveolar macrophages were characteristic abnormalities. These findings shed new insights into the pathogenesis of COVID-19 and justify the use of interleukin 6 (IL6) receptor antagonists and convalescent plasma with neutralizing antibodies against SARS-CoV-2 for severe patients. Authors Chaofu Wang, Jing Xie, Lei Zhao, Xiaochun Fei, Heng Zhang, and Yun Tan contributed equally to this work. Authors Chaofu Wang, Jun Cai, Rong Chen, Zhengli Shi, and Xiuwu Bian jointly supervised this work.
Article
Full-text available
Currently there is no effective antiviral therapy for SARS-CoV-2 infection, which frequently leads to fatal inflammatory responses and acute lung injury. Here, we discuss the various mechanisms of SARS-CoV-mediated inflammation. We also assume that SARS-CoV-2 likely shares similar inflammatory responses. Potential therapeutic tools to reduce SARS-CoV-2-induced inflammatory responses include various methods to block FcR activation. In the absence of a proven clinical FcR blocker, the use of intravenous immunoglobulin to block FcR activation may be a viable option for the urgent treatment of pulmonary inflammation to prevent severe lung injury. Such treatment may also be combined with systemic anti-inflammatory drugs or corticosteroids. However, these strategies, as proposed here, remain to be clinically tested for effectiveness.
Article
Full-text available
Since the SARS outbreak 18 years ago, a large number of severe acute respiratory syndrome-related coronaviruses (SARSr-CoV) have been discovered in their natural reservoir host, bats1–4. Previous studies indicated that some of those bat SARSr-CoVs have the potential to infect humans5–7. Here we report the identification and characterization of a novel coronavirus (2019-nCoV) which caused an epidemic of acute respiratory syndrome in humans in Wuhan, China. The epidemic, which started from 12 December 2019, has caused 2,050 laboratory-confirmed infections with 56 fatal cases by 26 January 2020. Full-length genome sequences were obtained from five patients at the early stage of the outbreak. They are almost identical to each other and share 79.5% sequence identify to SARS-CoV. Furthermore, it was found that 2019-nCoV is 96% identical at the whole-genome level to a bat coronavirus. The pairwise protein sequence analysis of seven conserved non-structural proteins show that this virus belongs to the species of SARSr-CoV. The 2019-nCoV virus was then isolated from the bronchoalveolar lavage fluid of a critically ill patient, which can be neutralized by sera from several patients. Importantly, we have confirmed that this novel CoV uses the same cell entry receptor, ACE2, as SARS-CoV.
Article
Full-text available
Background: A recent cluster of pneumonia cases in Wuhan, China, was caused by a novel betacoronavirus, the 2019 novel coronavirus (2019-nCoV). We report the epidemiological, clinical, laboratory, and radiological characteristics and treatment and clinical outcomes of these patients. Methods: All patients with suspected 2019-nCoV were admitted to a designated hospital in Wuhan. We prospectively collected and analysed data on patients with laboratory-confirmed 2019-nCoV infection by real-time RT-PCR and next-generation sequencing. Data were obtained with standardised data collection forms shared by the International Severe Acute Respiratory and Emerging Infection Consortium from electronic medical records. Researchers also directly communicated with patients or their families to ascertain epidemiological and symptom data. Outcomes were also compared between patients who had been admitted to the intensive care unit (ICU) and those who had not. Findings: By Jan 2, 2020, 41 admitted hospital patients had been identified as having laboratory-confirmed 2019-nCoV infection. Most of the infected patients were men (30 [73%] of 41); less than half had underlying diseases (13 [32%]), including diabetes (eight [20%]), hypertension (six [15%]), and cardiovascular disease (six [15%]). Median age was 49·0 years (IQR 41·0-58·0). 27 (66%) of 41 patients had been exposed to Huanan seafood market. One family cluster was found. Common symptoms at onset of illness were fever (40 [98%] of 41 patients), cough (31 [76%]), and myalgia or fatigue (18 [44%]); less common symptoms were sputum production (11 [28%] of 39), headache (three [8%] of 38), haemoptysis (two [5%] of 39), and diarrhoea (one [3%] of 38). Dyspnoea developed in 22 (55%) of 40 patients (median time from illness onset to dyspnoea 8·0 days [IQR 5·0-13·0]). 26 (63%) of 41 patients had lymphopenia. All 41 patients had pneumonia with abnormal findings on chest CT. Complications included acute respiratory distress syndrome (12 [29%]), RNAaemia (six [15%]), acute cardiac injury (five [12%]) and secondary infection (four [10%]). 13 (32%) patients were admitted to an ICU and six (15%) died. Compared with non-ICU patients, ICU patients had higher plasma levels of IL2, IL7, IL10, GSCF, IP10, MCP1, MIP1A, and TNFα. Interpretation: The 2019-nCoV infection caused clusters of severe respiratory illness similar to severe acute respiratory syndrome coronavirus and was associated with ICU admission and high mortality. Major gaps in our knowledge of the origin, epidemiology, duration of human transmission, and clinical spectrum of disease need fulfilment by future studies. Funding: Ministry of Science and Technology, Chinese Academy of Medical Sciences, National Natural Science Foundation of China, and Beijing Municipal Science and Technology Commission.
Article
Full-text available
Background and Purpose Severe influenza A virus (IAV) infections are associated with damaging hyperinflammation that can be fatal. There is an urgent need to identify new therapeutic agents to treat severe and pathogenic IAV infections. Repurposing of drugs with an existing and studied pharmacokinetic and safety profile is a highly attractive potential strategy. We have previously demonstrated that the NLRP3 inflammasome plays time‐dependent roles during severe IAV infection with early protective responses and later dysregulation leading to excessive inflammation, contributing to disease severity. Experimental Approach We tested two existing drugs, probenecid and AZ11645373, to target P2X7 receptor signalling and dampen NLRP3 inflammasome responses during severe IAV infection. In vitro, the drugs were assessed for their ability to limit NLRP3 inflammasome‐dependent IL‐1β secretion in macrophage cultures. In vivo, their effects were assessed on hyperinflammation and disease during severe IAV infection in C57BL/6 mice. Key Results Treatment of macrophages with probenecid or AZ11645373 in vitro diminished NLRP3 inflammasome‐dependent IL‐1β secretion. Intranasal therapeutic treatment of mice displaying severe influenza disease with probenecid or AZ11645373 reduced pro‐inflammatory cytokine production, cellular infiltrates in the lung, and provided protection against disease. Importantly, these drugs could be administered at either early or late stage of disease and provide therapeutic efficacy. Conclusions and Implications Our study demonstrates that the anti‐inflammatory drugs probenecid and AZ11645373, which have documented pharmacokinetics and safety profiles in humans, are effective at dampening hyperinflammation and severe influenza disease providing potentially new therapeutic strategies for treating severe or pathogenic IAV infections.
Article
Purpose: To evaluate the effect of low-level laser therapy (LLLT) on an experimental model of ventilator-induced lung injury (VILI). Methods: 24 adult Wistar rats were randomized in four groups: protective mechanical ventilation (PMV), PMV+laser, VILI and VILI+laser. The animals of the PMV and VILI groups were ventilated with tidal volume of 6 and 35 ml/Kg, respectively for 90 minutes. After the first 60 minutes of ventilation, the animals of the laser groups were irradiated (808 nm, 100 mW power density, 20 J/cm² energy density, continuous emission mode, and exposure time 5s) and after 30 minutes of irradiation, the animals were euthanized. Lung samples were removed to morphological analysis, bronchoalveolar lavage (BAL) and RT-qPCR technique. Results: The VILI group showed greater acute lung injury (ALI) score with an increase in neutrophil infiltration, higher neutrophil count in the BAL fluid and greater cytokine mRNA expression compared to the PMV groups (p<0.05). The VILI+laser group when compared to the VILI group showed lower ALI score (0.35 ± 0.08 vs. 0.54 ± 0.13, p<0.05), alveolar neutrophil infiltration (7.00 ± 5.73 vs. 21.50 ± 9.52, p<0.05), total cell count (1.90 ± 0.71 vs. 4.09 ± 0.96 x105, p<0.05) and neutrophil count in the BAL fluid (0.60 ± 0.37 vs. 2.28 ± 0.48 x105, p<0.05). Moreover, LLLT induced a decrease in pro-inflammatory and an increase of anti-inflammatory mRNA levels compared to the VILI group (p<0.05). Conclusion: LLLT reduced the inflammatory response in an experimental model of VILI.
Article
Idiopathic pulmonary fibrosis (IPF) is a progressive and chronic inflammatory disease with a poor prognosis and very few available treatment options. Low-level laser therapy (LLLT) has been gaining prominence as a new and effective anti-inflammatory and immunomodulatory agent. Can lung inflammation and the airway remodeling be regulated by LLLT in an experimental model of IPF in C57Bl/6 mice? The present study investigated if laser attenuates cellular migration to the lungs, the airway remodeling as well as pro-fibrotic cytokines secretion from type II pneumocytes and fibroblasts. Mice were irradiated (780 nm and 30 mW) and then euthanized fifteen days after bleomycin-induced lung fibrosis. Lung inflammation and airway remodeling were evaluated through leukocyte counting in bronchoalveolar lavage fluid (BALF) and analysis of collagen in lung, respectively. Inflammatory cells in blood were also measured. For in vitro assays, bleomycin-activated fibroblasts and type II pneumocytes were irradiated with laser. The pro- and anti-inflammatory cytokines level in BALF as well as cells supernatant were measured by ELISA, and the TGFβ in lung was evaluated by flow cytometry. Lung histology was used to analyze collagen fibers around the airways. LLLT reduced both migration of inflammatory cells and deposition of collagen fibers in the lungs. In addition, LLLT downregulated pro-inflammatory cytokines and upregulated the IL-10 secretion from fibroblasts and pneumocytes. Laser therapy greatly reduced total lung TGFβ. Systemically, LLLT also reduced the inflammatory cells counted in blood. There is no statistical difference in inflammatory parameters studied between mice of the basal group and the laser-treated mice. Data obtained indicate that laser effectively attenuates the lung inflammation, and the airway remodeling in experimental pulmonary fibrosis is driven to restore the balance between the pro- and anti-inflammatory cytokines in lung and inhibit the pro-fibrotic cytokines secretion from fibroblasts.