ArticlePDF Available

Safety of High-Dose Vitamin C in Non-Intensive Care Hospitalized Patients with COVID-19: An Open-Label Clinical Study

MDPI
Journal of Clinical Medicine
Authors:

Abstract and Figures

Background: Vitamin C has been used as an antioxidant and has been proven effective in boosting immunity in different diseases, including coronavirus disease (COVID-19). An increasing awareness was directed to the role of intravenous vitamin C in COVID-19. Methods: In this study, we aimed to assess the safety of high-dose intravenous vitamin C added to the conventional regimens for patients with different stages of COVID-19. An open-label clinical trial was conducted on patients with COVID-19. One hundred four patients underwent high-dose intravenous administration of vitamin C (in addition to conventional therapy), precisely 10 g in 250 cc of saline solution in slow infusion (60 drops/min) for three consecutive days. At the same time, 42 patients took the standard-of-care therapy. Results: This study showed the safety of high-dose intravenous administration of vitamin C. No adverse reactions were found. When we evaluated the renal function indices and estimated the glomerular filtration rate (eGRF, calculated with the CKD-EPI Creatinine Equation) as the main side effect and contraindication related to chronic renal failure, no statistically significant differences between the two groups were found. High-dose vitamin C treatment was not associated with a statistically significant reduction in mortality and admission to the intensive care unit, even if the result was bound to the statistical significance. On the contrary, age was independently associated with admission to the intensive care unit and in-hospital mortality as well as noninvasive ventilation (N.I.V.) and continuous positive airway pressure (CPAP) (OR 2.17, 95% CI 1.41–3.35; OR 7.50, 95% CI 1.97–28.54; OR 8.84, 95% CI 2.62–29.88, respectively). When considering the length of hospital stay, treatment with high-dose vitamin C predicts shorter hospitalization (OR −4.95 CI −0.21–−9.69). Conclusions: Our findings showed that an intravenous high dose of vitamin C is configured as a safe and promising therapy for patients with moderate to severe COVID-19.
This content is subject to copyright.
Citation: Corrao, S.; Raspanti, M.;
Agugliaro, F.; Gervasi, F.; Di Bernardo,
F.; Natoli, G.; Argano, C., on behalf of
Internal Medicine IGR COVID-19
Investigators. Safety of High-Dose
Vitamin C in Non-Intensive Care
Hospitalized Patients with COVID-19:
An Open-Label Clinical Study. J. Clin.
Med. 2024,13, 3987. https://
doi.org/10.3390/jcm13133987
Academic Editors: Kiriakos
Karkoulias and Dimosthenis
Lykouras
Received: 11 May 2024
Revised: 30 June 2024
Accepted: 2 July 2024
Published: 8 July 2024
Copyright: © 2024 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
Journal of
Clinical Medicine
Article
Safety of High-Dose Vitamin C in Non-Intensive Care
Hospitalized Patients with COVID-19: An Open-Label
Clinical Study
Salvatore Corrao 1, 2, * , Massimo Raspanti 3, Federica Agugliaro 1, Francesco Gervasi 4, Francesca Di Bernardo 5,
Giuseppe Natoli 1and Christiano Argano 1, on behalf of Internal Medicine IGR COVID-19 Investigators
1
Department of Internal Medicine, National Relevance and High Specialization Hospital Trust ARNAS Civico,
Di Cristina, Benfratelli, 90127 Palermo, Italy; federica.agugliaro@gmail.com (F.A.);
peppenatoli@gmail.com (G.N.); chargano@yahoo.it (C.A.)
2Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical
Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
3Cardiology and Intensive Care Unit, A. Aiello Hospital, 91026 Mazzara del Vallo, Italy;
massimoraspanti1991@gmail.com
4Specialized Laboratory of Oncology, National Relevance and High Specialization Hospital Trust ARNAS
Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy; francesco.gervasi@arnascivico.it
5Department of Microbiology and Virology, National Relevance and High Specialization Hospital Trust
ARNAS Civico, Di Cristina, Benfratelli, 90127 Palermo, Italy; francesca.dibernardo@arnascivico.it
*
Correspondence: salvatore.corrao@unipa.it or s.corrao@tiscali.it; Tel.: +39-091-655-2065; Fax: +39-091-666-3167
Investigators of the Internal Medicine IGR COVID-19 is provided in the Acknowledgments.
Abstract: Background: Vitamin C has been used as an antioxidant and has been proven effective in
boosting immunity in different diseases, including coronavirus disease (COVID-19). An increasing
awareness was directed to the role of intravenous vitamin C in COVID-19. Methods: In this study,
we aimed to assess the safety of high-dose intravenous vitamin C added to the conventional regimens
for patients with different stages of COVID-19. An open-label clinical trial was conducted on patients
with COVID-19. One hundred four patients underwent high-dose intravenous administration of
vitamin C (in addition to conventional therapy), precisely 10 g in 250 cc of saline solution in slow
infusion (60 drops/min) for three consecutive days. At the same time, 42 patients took the standard-
of-care therapy. Results: This study showed the safety of high-dose intravenous administration of
vitamin C. No adverse reactions were found. When we evaluated the renal function indices and
estimated the glomerular filtration rate (eGRF, calculated with the CKD-EPI Creatinine Equation) as
the main side effect and contraindication related to chronic renal failure, no statistically significant
differences between the two groups were found. High-dose vitamin C treatment was not associated
with a statistically significant reduction in mortality and admission to the intensive care unit, even
if the result was bound to the statistical significance. On the contrary, age was independently
associated with admission to the intensive care unit and in-hospital mortality as well as noninvasive
ventilation (N.I.V.) and continuous positive airway pressure (CPAP) (OR 2.17, 95% CI 1.41–3.35;
OR 7.50,
95% CI 1.97–28.54
; OR 8.84, 95% CI 2.62–29.88, respectively). When considering the length
of hospital stay, treatment with high-dose vitamin C predicts shorter hospitalization (OR
4.95
CI 0.21–9.69
). Conclusions: Our findings showed that an intravenous high dose of vitamin C is
configured as a safe and promising therapy for patients with moderate to severe COVID-19.
Keywords: vitamin C; safety; length of hospital stay; in-hospital mortality; admission to ICU
1. Introduction
It is four years since the World Health Organization declared COVID-19 a pandemic
on 11 March 2020 [
1
] after the first pneumonia cases caused by severe acute respiratory
syndrome coronavirus (SARS-CoV2) occurred in late December 2019 [
2
]. Globally, as of
J. Clin. Med. 2024,13, 3987. https://doi.org/10.3390/jcm13133987 https://www.mdpi.com/journal/jcm
J. Clin. Med. 2024,13, 3987 2 of 17
3 March 2024, there have been 774,833,487 confirmed cases, and 7,036,994 deaths have been
reported worldwide [
3
]. This pandemic notably challenged the healthcare system and med-
ical care, highlighting the lack of preparation and inadequacy to manage this public health
crisis and simultaneously provide general and specialized medical care [
4
]. Despite a more
in-depth understanding of the pathophysiologic mechanisms behind COVID-19 [
5
,
6
] and
the recent progress in managing and averting COVID-19 by utilizing vaccines and antiviral
drugs, effective interventions have remained a significant challenge [
7
,
8
], especially in older
people with comorbidities [
9
]. From the beginning of COVID-19, researchers have studied
and used molecules and drugs with anti-inflammatory and antioxidant properties [
9
14
].
In this sense, an increasing interest was developed in utilizing vitamin C [
15
17
]. Vitamin
C, also known as ascorbic acid, is a water-soluble vitamin that acts as a strong antioxidant
and plays a role as a cofactor in various biosynthetic pathways in the immune system. It is
an essential nutrient that the human body cannot produce on its own [
18
]. Its antioxidant
properties stem from its ability to donate electrons, which helps protect molecules from
oxidative damage.
For this reason, many studies have focused on the antioxidant properties of vitamins,
particularly their potential anticancer and antiatherosclerosis properties [
19
]. In addition,
vitamin C stimulates immunity through the raised activity of neutrophils by increasing
chemotaxis and phagocytosis and in T-cell maturation, as well as through the mechanism
of ascorbate-mediated enhancement of immune function [
20
22
]. Individuals with a
deficiency in vitamin C face an increased likelihood of developing severe respiratory
infections, such as pneumonia [
23
,
24
]. A meta-analysis has shown that oral vitamin
C supplements can reduce the risk of pneumonia, especially in those with low dietary
intakes [
25
]. On the contrary, a recent meta-analysis showed no reduction in mortality
among people with community-acquired pneumonia [
26
]. Vitamin C deficiency can also
elevate the risk of sepsis during pneumonia.
Moreover, among patients with a diagnosis of sepsis and clinically significant vitamin
C deficiency, a more rapid progression of multiorgan failure was observed [
27
]. In this
sense, contrasting data are available about the combination of thiamine, vitamin C, and
hydrocortisone for the treatment of septic shock; in fact, Marik and colleagues [
28
] dis-
covered that early intravenous administration of vitamin C, complemented with thiamine
and corticosteroids, is effective in preventing progressive organ dysfunction. Litwak et al.
showed that triple therapy did not improve hospital or ICU mortality in patients with
septic shock [
29
]. Various clinical studies evaluating high-dose intravenous vitamin C ad-
ministration in patients with severe sepsis, acute lung injury, and acute respiratory distress
syndrome admitted to intensive care units have shown mixed results regarding laboratory
and clinical outcomes [
30
32
]. A systematic review and meta-analysis of intravenous
vitamin C therapy in critically ill patients showed favorable results regarding decreased
mechanical ventilation support to reduce overall mortality [
33
]. A recent systematic review
and meta-analysis showed no significant effect on infection episodes, length of stay in the
hospital or ICU, or duration of mechanical ventilation of administration of intravenous vi-
tamin C treatment in critically ill patients. However, the study did indicate a mild tendency
towards reducing mortality [
34
]. However, a recent meta-analysis revealed a decrease in
the duration of mechanical ventilation [35].
In addition, vitamin C inhibits viral growth, stimulates interferon production, and
enhances lung epithelial cells’ antiviral activity [
36
,
37
]. The antiviral properties of vitamin
C are believed to be particularly beneficial in treating COVID-19. However, how vitamin
C is administered is crucial for both its effectiveness and safety. It is crucial to remember
that while injections and infusions provide the highest dose of vitamin C, they also pose
a greater risk of overdose and renal side effects. This is because they bypass the natural
limits of intestinal transporters. It is important to note that higher doses of vitamin C
administered via infusion may lead to more serious side effects, such as severe kidney
injury [38].
J. Clin. Med. 2024,13, 3987 3 of 17
Since the outbreak of the SARS-CoV-2 pandemic, the safety and effectiveness of high-
dose vitamin C have been the focus of scientific research. Numerous studies have tried to
assess the effectiveness of vitamin C in preventing COVID-19 and its capacity to prevent the
progression to more severe diseases. Additionally, the impact of high doses of vitamin C
on critically ill patients was evaluated [
15
,
39
,
40
]. Given this background, this study aimed
at evaluating the safety and, secondly, the effectiveness of the administration of a high dose
of vitamin C (10 g for three consecutive days) in addition to standard-of-care therapy in a
cohort of hospitalized patients with COVID-19 in the non-intensive care ward, as well as
the effect on mortality, length of hospital stays, and admission to the intensive care unit.
2. Materials and Methods
From 30 March 2020 to 1 April 2021, 146 patients with confirmed COVID-19 (RT-PCR
positive for SARS-CoV-2 and presence of typical radiological signs) who were admitted
to the COVID Unit of the Department of Internal Medicine of the National Relevance
and High Specialization Hospital Trust ARNAS Civico, Di Cristina, Benfratelli, Italy, were
consecutively enrolled in this longitudinal study. The patients admitted to the Department
of Internal Medicine all provided written informed consent upon admission. Eligible
patients were adults with proven COVID-19 admitted to the hospital. There are no exclusion
criteria other than the lack of informed consent signature. The Ethics Committee has
approved the conduct of the study at our institution. The approval number assigned to this
study is 3143. Of these patients, 104 underwent high-dose intravenous administration of
vitamin C (in addition to conventional therapy); precisely, 10 g in 250 cc of saline solution
in slow infusion (60 drops/min) for three consecutive days, according to the therapeutic
protocol “Use of Ascorbic Acid in patients with COVID-19” developed by our center and
registered in March 2020 on the platform Clinicaltrials.gov (accessed on 14 January 2024).
NCT04323514 [
41
]. The other 42 patients underwent the standard of care therapy. Variables
taken into consideration for all patients at the time of admission included age, sex, pre-
existing comorbidities, whether or not oxygen therapy was required, the method used
[nasal cannula (N.C.), Venturi Mask (MV), Continuous Positive Airway Pressure (CPAP),
or noninvasive ventilation (N.I.V.)], and levels of inflammatory markers, such as lactate
dehydrogenase (L.D.H.), procalcitonin (P.C.T.), C-reactive protein (C.R.P.), D-Dimer (D-D),
and interleukin-6 (IL-6).
Statistical Analysis
Quantitative variables are summarized as mean (95% confidence intervals), medians
(interquartile range: first–third), and categorical variables, reported as percentages. The
safety of vitamin C administration represented the primary outcome. The secondary
outcome included mortality and length of hospital stay. In order to establish the safety of
administering high doses of intravenous ascorbic acid, all adverse events were recorded
during and after administration. Furthermore, a patient’s control group was consecutively
recruited to use multivariable logistic regression to study a possible association with the
hospital mortality of treated subjects. The following clinically relevant variables were
considered to demonstrate safety or lack of safety: sex, age, use of noninvasive ventilation
methods, obesity, and diabetes. The quantitative variables were compared using the non-
parametric Mann–Whitney U test. A multivariate regression analysis investigated the
relationship between variables and in-hospital mortality, admission to the intensive care
unit, and length of hospital stays.
The Hosmer–Lemeshow methodology [
42
] was used to select variables for analy-
sis [
39
]. After univariate analysis, only variables with a p-value less than 0.20 were included
in the final model. The variables were excluded through a backward process until each
variable reached a significance level of p< 0.20. The Hosmer–Lemeshow test was used
to measure how well the model fits the data without the researcher having to choose any
variables for the multivariate model. Statistical significance was set at a two-tailed p-value
J. Clin. Med. 2024,13, 3987 4 of 17
of less than 0.05. Stata Statistical Software 2017 (StataCorp, College Station, TX, USA) was
used for database management and analyses.
3. Results
During the recruitment period, 146 hospitalized patients were examined. Among these,
59.6% were male, with a median age of 64.3 years. Table 1shows the clinical characteristics
of the study population.
Table 1. Clinical variables of the analyzed population.
N 146
Patients undergoing Intravenous High-Dose Vitamin C (IHDVC) 104
Patients not undergoing Intravenous High-Dose Vitamin C (IHDVC) 42
Age §64.3 (54.9–76.0)
Men (%) * 59.6 (51.3–67.2)
Pre-existing comorbidities * 80.4 (73.0–86.0)
Hypertension * 70.0 (61.8–76.9)
Obesity (BMI 30 Kg/m2) * 40.8 (33.0–49.2)
Diabetes mellitus * 35.6 (28.2–43.9)
Chronic ischemic heart disease * 29.3 (22.4–37.4)
Chronic cerebrovascular disease * 29.1 (22.1–36.9)
Chronic renal failure * 14.7 (9.7–21.5)
Neoplasm (active or previous) * 11.1 (6.9–17.5)
Atrial fibrillation * 9.0 (5.3–15.1)
Body temperature at admission * 37.7 (37.1–38.4)
Oxygen support at admission: None * 37 (29.5–45.2)
Oxygen support at admission: Nose cannulas * 10.2 (6.3–16.4)
Oxygen support at admission: Venturi mask * 25.3 (18.9–33.1)
Oxygen support at admission: CPAP * 15.8 (10.7–22.7)
Oxygen support at admission: NIV S/T * 11.0 (6.8–17.2)
Hospitalization days §19.0 (13.0–29.5)
Admission to intensive care/ICU (%) 8.3
Death (%) 10.2
* Data reported as means (95% confidence intervals); §Data reported as medians (Q1–Q3).
In total, 80.4% of all patients had at least one pre-existing condition at admission.
Disease distribution showed that arterial hypertension, obesity (B.M.I.
30 kg/m
2
), and
diabetes mellitus were the most represented comorbidities in patients with COVID-19 (70%,
40.8%, and 35.6%, respectively). Furthermore, almost a third of patients reported a previous
major ischemic event (myocardial infarction and stroke). For approximately two-thirds
of patients, oxygen supportive therapy was necessary on admission or during a hospital
stay, with over a quarter of subjects requiring ventilation with continuous positive airway
pressure (CPAP) or noninvasive ventilation with pressure support ventilation (NIV/ST).
Due to severe hypoxemia, 8.3% were admitted to intensive care. Death occurred in 10.2%
of patients. The average length of hospital stay was 19 days.
The clinical characteristics of patients with COVID-19 who underwent intravenous
vitamin C and conventional therapy are shown in Table 2. No significant differences in
the main variables were found between the two groups. A more significant percentage
of patients not undergoing IHDVC required venturi mask and CPAP in comparison with
people undergoing IHDVC (30.9% vs. 23.1% and 23.8% vs. 12.5%, respectively), while the
percentage of patients that require NIV S/T was higher in people who received IHDVC
(12.5% vs. 7.1%). Almost 10 percent of patients taking vitamin C were admitted to the
J. Clin. Med. 2024,13, 3987 5 of 17
intensive care unit (9.7% vs. 5.0%). It is worth outlining that a shorter hospital length of stay
and mortality were present in patients who underwent high doses of vitamin C compared
to those who took conventional therapy (18.0% vs. 24.0% and 8.6% vs. 14.3%, respectively).
Table 2. Clinical variables of the analyzed population according to patients undergoing IHDVC
categorization.
Variable Patients Undergoing IHDVC
(n = 104)
Patients Not Undergoing IHDVC
(n = 42) p
Age §64 (53–76) 64 (56–76) 0.8086
Men (%) 61.5 54.8 0.4500
Pre-existing comorbidities (%) 79.6 82.5 0.6960
Hypertension (%) 71.8 65.0 0.4230
Obesity (BMI 30 Kg/m2)(%) 39.2 45.0 0.5282
Diabetes mellitus (%) 35.9 35.0 0.9177
Chronic ischemic heart disease or
Chronic cerebrovascular disease (%) 29.1 30.0 0.9180
Chronic renal failure (%) 13.6 17.5 0.5535
Neoplasm (active or previous) (%) 10.7 12.5 0.7566
Oxygen support at admission: None (%) 39.4 30.9 0.3372
Oxygen support at admission: Nose cannulas (%) 11.5 7.1 0.4284
Oxygen support at admission: Venturi mask (%) 23.1 30.9 0.3220
Oxygen support at admission: CPAP (%) 12.5 23.8 0.0895
Oxygen support at admission: NIV S/T (%) 12.5 7.1 0.3482
Hospitalization days §18 (13–27) 24 (13–31) 0.1596
Admission to intensive care/ICU (%) 9.7 5.0 0.3620
Death (%) 8.6 14.3 0.3103
§Data reported as medians (Q1–Q3).
Table 3shows the laboratory characteristics of patients who underwent intravenous
vitamin C and conventional therapy. This table represents the median percentage change
(Q1–Q3) between the value recorded at discharge and the value recorded at admission for
patients who took intravenous high doses of vitamin C and those who did not.
A significant increase in the median percentage change of neutrophils was found in
patients who took an intravenous high dose of vitamin C compared to those who took
conventional therapy (p= 0.0126). Compared to the control group, no significant changes
were highlighted in other laboratory variables, such as C-reactive protein and procalcitonin.
During the observation, there were no differences in terms of adverse drug events between
the intervention group and the control group. We did not record any adverse events. Since
the main side effect and the main contraindication are related to chronic renal failure, we
evaluated the renal function indices and estimated the glomerular filtration rate (eGRF)
(calculated with the CKD-EPI Creatinine Equation). No statistically significant differences
were found between the two groups. Furthermore, no allergic reactions occurred.
As illustrated in Figure 1, the high-dose vitamin C treatment did not result in a sta-
tistically significant decrease in mortality and admission to the intensive care unit, even
if the result bordered on statistical significance. On the contrary, age was independently
associated with in-hospital mortality and admission to the intensive care unit, as well as
N.I.V. and CPAP (OR 2.17, 95% CI 1.41–3.35; OR 7.50, 95% CI 1.97–28.54; OR 8.84, 95% CI
2.62–29.88, respectively). When we consider the length of hospital stay (Table 4), treatment
with high-dose vitamin C represents a predictor of shorter length of hospitalization. Al-
though length of hospital stay was not statistically significantly different between the two
groups, a statistically significant difference was found when correcting the confounding
variables. This difference was not only statistically significant but also clinically relevant.
J. Clin. Med. 2024,13, 3987 6 of 17
The data mentioned above allowed us to demonstrate an excellent safety profile of vitamin
C in the short term.
Table 3. Laboratory variables, % variation of the analyzed population (signed rank).
Variable
Patients Undergoing
High-Dose Vitamin C Treatment
(n = 104)
Patients Not Undergoing High-Dose
Vitamin C Treatment
(n = 42)
p
Hemoglobin (gr/dL) 5.2 (10.9–1.8) 3.6 (14.9–4.4) 0.5064
White cells (cell/uL) 24.4 (6.8–75.9) 1.1 (23.7–52.5) 0.0656
Neutrophils (cell/uL) 16.2 (22.9–81.5) 18.6 (40.6–31.8) 0.0126
Linfocytes (cell/uL) 43.2 (2.8–99.6) 38.6 (7.4–125) 0.9161
Platelets (cell/µL) 12.1 (19.4–48.6) 0 (15.8–27.7) 0.1735
PCR (mg/dL) 83.9 (95.0–10.1) 82.5 (95.0–19.2) 0.7957
Procalcitonin (PCT) (of/L) 0 (51.4–0) 0 (63.1–0) 0.5827
eGFR (mL/min/m2)7.7 (0.4–24.2) 4.5 (7.7–31.0) 0.3599
D-Dimer (ng/mL) 2.6 (50.3–40.7) 14.1 (58.6–2.1) 0.2604
Hematocrit (%) 4.1 (9.6–2.4) 1.3 (10.1–4.9) 0.3407
Monocytes (cell/µL) 21.6 (16.0–71.9) 18.7 (15.0–103.7) 0.9335
Sodium (mmol/L) 0 (2.1–2.2) 1.4 (0.7–2.9) 0.1853
Glycemia (mg/dL) 11.2 (29.9–24.5) 10.2 (28.8–16.5) 0.9702
Data reported as medians (Q1–Q3).
J. Clin. Med. 2024, 13, x FOR PEER REVIEW 7 of 18
Although length of hospital stay was not statistically signicantly dierent between the
two groups, a statistically signicant dierence was found when correcting the
confounding variables. This dierence was not only statistically signicant but also
clinically relevant. The data mentioned above allowed us to demonstrate an excellent
safety prole of vitamin C in the short term.
Figure 1. Multivariable logistic regression according to composite outcome mortality and/or
admission to the intensive care unit (p < 0.0001; pseudo R2 31.3%). Only the nal model is shown
according to the Hosmer–Lemeshow methodology for selecting variables; see the statistical analysis
section.
Table 4. Multivariable regression according to length of hospital stay. Only the nal model is shown
according to the Hosmer–Lemeshow methodology; for the selection of variables, see the statistical
analysis section.
Outcome: Length of Hospital Stays
Variables Coefficient (I.C. 95%) p Value
Age 0.13 (0.02–0.28) 0.089
Male sex 1.26 (3.06–5.59) 0.565
High-dose Vitamin C treatment 4.95 (0.21–9.69) 0.041
4. Discussion
4.1. Vitamin C (Ascorbic Acid)
Vitamin C (ascorbic acid) is a water-soluble vitamin that potentially benets patients
with dierent disease severities. It is a “scavenger” molecule (a substance capable of
transforming oxygen radicals into non-radical compounds, lacking reactivity and
therefore toxicity) that has anti-inammatory properties, inuences vascular integrity and
cellular immunity, works as a cofactor in the endogenous generation of catecholamines,
and has been studied in many disease states (predominantly inammatory states),
including COVID-19 [31]. Vitamin C is crucial in forming and depositing type IV collagen
in the basement membrane. It also promotes endothelial proliferation, inhibits apoptosis,
and preserves endothelial cell-derived nitric oxide to help regulate blood ow [43].
Vitamin C plays a vital role in the functioning and regulation of the immune system.
Leukemic cells and neutrophils accumulate vitamin C intracellularly, which depends on
its availability in plasma. In neutrophils, vitamin C aects the phagocytosis of
microorganisms and chemotaxis. In addition, vitamin C protects neutrophils and
phagocytes from damage following an oxidative burst through its antioxidant and
Figure 1. Multivariable logistic regression according to composite outcome mortality and/or admis-
sion to the intensive care unit (p< 0.0001; pseudo R
2
31.3%). Only the final model is shown according
to the Hosmer–Lemeshow methodology for selecting variables; see the statistical analysis section.
J. Clin. Med. 2024,13, 3987 7 of 17
Table 4. Multivariable regression according to length of hospital stay. Only the final model is shown
according to the Hosmer–Lemeshow methodology; for the selection of variables, see the statistical
analysis section.
Outcome: Length of Hospital Stays
Variables Coefficient (I.C. 95%) pValue
Age 0.13 (0.02–0.28) 0.089
Male sex 1.26 (3.06–5.59) 0.565
High-dose Vitamin C treatment 4.95 (0.21–9.69) 0.041
4. Discussion
4.1. Vitamin C (Ascorbic Acid)
Vitamin C (ascorbic acid) is a water-soluble vitamin that potentially benefits patients
with different disease severities. It is a “scavenger” molecule (a substance capable of
transforming oxygen radicals into non-radical compounds, lacking reactivity and therefore
toxicity) that has anti-inflammatory properties, influences vascular integrity and cellu-
lar immunity, works as a cofactor in the endogenous generation of catecholamines, and
has been studied in many disease states (predominantly inflammatory states), including
COVID-19 [
31
]. Vitamin C is crucial in forming and depositing type IV collagen in the
basement membrane. It also promotes endothelial proliferation, inhibits apoptosis, and
preserves endothelial cell-derived nitric oxide to help regulate blood flow [43].
Vitamin C plays a vital role in the functioning and regulation of the immune system.
Leukemic cells and neutrophils accumulate vitamin C intracellularly, which depends on its
availability in plasma. In neutrophils, vitamin C affects the phagocytosis of microorgan-
isms and chemotaxis. In addition, vitamin C protects neutrophils and phagocytes from
damage following an oxidative burst through its antioxidant and scavenging capacity and
activates a caspase-dependent cascade that induces programmed apoptosis and inhibits
necrosis [44,45].
A similar protective effect against oxidative stress has also been observed in lym-
phocytes. Vitamin C’s impact on controlling inflammation includes modifying nuclear
transcription factor kappa B (NFkB) and reducing inflammatory cytokine production [
46
].
A recent systematic review and meta-analysis showed that the use of vitamin C reduces
hospital mortality in patients with COVID-19 [17].
4.2. Role of Intravenous Vitamin C in Hospitalized Patients with COVID-19
The World Health Organization has identified vitamin C as a potential adjunctive ther-
apy for patients with critical COVID-19 [
47
]. Several trials have indicated some potentially
beneficial effects of intravenous vitamin C in severe COVID-19 [
48
,
49
]. In a pilot study
in China, 56 adults in the ICU with COVID-19 were given either vitamin C or a placebo.
The study was stopped early due to decreased COVID-19 cases. The results showed no
differences in mortality, mechanical ventilation duration, or SOFA score change between the
groups. The study showed that the treatment arm had more significant respiratory function
improvements than the placebo arm. The improvement was evaluated by calculating the
ratio of partial blood pressure of oxygen to inspired oxygen fraction (PaO
2
/FiO
2
) from
day 1 to day 7. The change in the treatment arm was +20.0, while in the placebo arm, it
was
51.9. The difference between the two arms was statistically significant (p= 0.04) [
48
].
Khumari found that intravenous vitamin C can significantly improve the clinical symptoms
of patients affected by COVID-19. Although it may not impact mortality and the need
for mechanical ventilation, patients who received vitamin C became asymptomatic earlier
(7.1 days vs. 9.6 days; p< 0.0001) and had a shorter duration of hospitalization (8.1 days
versus 10.7 days; p< 0.0001) compared to those who received standard therapy alone. There
were no significant differences in mortality and the need for mechanical ventilation [
49
].
Li and colleagues, on the contrary, demonstrated that adjunctive IV vitamin C may not
J. Clin. Med. 2024,13, 3987 8 of 17
reduce mortality, vasopressor requirements, SOFA scores, or ventilator settings in critically
ill COVID-19 patients [50]
4.3. The Immune System
The immune system is the body’s foremost defense against infectious agents. Through-
out biological evolution, it has developed two central and different defense systems: non-
specific (or innate) immunity and specific (or adaptive) immunity [51].
Innate and adaptive immune responses interact to create immune defenses [
52
]. The
innate immune response occurs immediately after infection and is usually involved in
virus elimination but with reduced antiviral capacity. Adaptive immunity is essential in
complete virus elimination [
52
]; this immune pathway is activated 4–7 days after infection.
The innate immune response is enhanced if the adaptive antiviral response fails to suppress
the virus in time. However, it cannot effectively eliminate the virus, leading to a systemic
inflammatory response with uncontrolled release of inflammatory cytokines [
53
56
]. El-
derly patients and patients with chronic diseases require more time to develop an adaptive
or innate immune response due to progressive cellular aging. Such patients rely solely on
an enhanced antiviral innate immune response in the early stages of infection and are at
increased risk for cytokinin storms. Whether the enhanced immunity is due to ongoing
viral replication or immunomodulatory dysregulation remains unclear [
57
]. Finally, the
NLR family containing pyrin domain 3 (NLRP3) is the most recognizable inflammasome
pattern in COVID-19 and includes most of the above-mentioned immune–inflammatory
pathways [
58
]. NLRP3 is a critical component of innate immunity and promotes inflam-
mation by producing IL-1
β
/18 and inducing pyroptosis. Later, IL-1
β
and IL-18 play a
role in promoting the release of other NLRP3 cytokines, including IL-6 [
59
]. For example,
a study demonstrated that activation of the NLRP3 inflammasome of S. suis results in
the production of IL-1
β
, leading to a cytokine release syndrome [
60
]. Consequently, a
positive correlation has been observed between IL-18, caspase-1, and other inflammatory
markers like C-reactive protein, lactate dehydrogenase (L.D.H.), and IL-6, in association
with inflammasome activation in COVID-19 [
61
], suggesting an essential role of the NLRP3
inflammasome in forming cytokine storms and the pathogenesis of COVID-19. Thus,
controlling NLRP3 inflammasome activation is a potential therapeutic strategy for these
conditions. Ascorbic acid, or vitamin C, has been studied for its effects on the NLRP3
inflammasome, given its roles in immune function, antioxidant defense, and inflammation
modulation. The interaction between ascorbic acid and the NLRP3 inflammasome involves
several mechanisms. One of the known triggers for NLRP3 inflammasome activation is
oxidative stress, characterized by a derangement between the generation of reactive oxygen
species (R.O.S.) and the body’s ability to detoxify these reactive products or repair the
resulting damage. Ascorbic acid, with its potent antioxidant properties, can scavenge
R.O.S., thereby reducing oxidative stress and potentially preventing the inappropriate
activation of the NLRP3 inflammasome. In addition, ascorbic acid can modulate the pro-
duction and activity of various inflammatory molecules and cytokines, some of which are
involved in the activation pathway of the NLRP3 inflammasome. By influencing these
pathways, ascorbic acid might indirectly impact the activation and function of the NLRP3
inflammasome. Finally, emerging evidence suggests that ascorbic acid may directly inhibit
the NLRP3 inflammasome. This direct inhibition can occur through the modulation of
mitochondrial integrity, reduction of mitochondrial R.O.S., and preservation of cellular
homeostasis, all of which are critical factors in activating the NLRP3 inflammasome. By
stabilizing mitochondria and reducing mitochondrial R.O.S., ascorbic acid may prevent the
assembly and activation of the NLRP3 inflammasome complex. However, while preclinical
studies have shown promising results, clinical evidence supporting the efficacy of ascorbic
acid in modulating the NLRP3 inflammasome in humans is still limited. Further research,
including rigorous clinical trials, is imperative for a comprehensive understanding of the
potential of ascorbic acid in managing conditions associated with NLRP3 inflammasome
J. Clin. Med. 2024,13, 3987 9 of 17
dysregulation. Additionally, the optimal dosing, safety, and long-term effects of ascorbic
acid supplementation for these specific purposes need to be thoroughly evaluated.
4.4. Role of the Immune System in COVID-19
Patients with COVID-19 often exhibit mild neutrophilia and T-cell lymphopenia, leading to
an increased neutrophil-to-lymphocyte ratio (NLR), which serves as a valuable prognostic marker
for the severity of COVID-19 [
62
]. Other leukocyte subpopulations also undergo characteristic
fluctuations and modifications, although these are more heterogeneous.
During an infection, viral PAMPs are detected by Toll-like receptors (TLRs), which
then trigger intracellular signaling cascades. These cascades activate transcription factors,
such as nuclear factor-kappa B (NF-
κ
B) and interferon regulatory factors, leading to the pro-
duction of type I interferons (IFNs) and proinflammatory cytokines. A proper IFN response
usually leads to an antiviral immune state in infected cells, limiting viral replication and
inducing apoptosis to protect the host from viral spread. However, certain SARS-CoV-2
proteins (e.g., open reading frames 6 and 3b) have been found to suppress type I IFN
production and the antiviral signal [
63
]. The initial delay of the IFN response is followed by
uncontrolled viral replication and spread in the infected host, contributing to a final surge in
IFN that may worsen hyperinflammation in the progression to severe disease [
64
]. Another
important component of the innate immune response is the complement system, which
serves as a rapid immune surveillance system against invading pathogens, connecting
innate and adaptive immunity.
In COVID-19 infection, excessive complement activation results in inflammation,
endothelial cell dysfunction, and intravascular coagulation [
65
]. Data indicated that in
patients with moderate to severe COVID-19 infection, there is an accumulation of Natural
Killer (NK) cells in the lungs. In contrast, the number of the same cells decreases in the
peripheral blood. Current evidence on the immune function of NK cells in COVID-19 is con-
tradictory. While some studies have reported signs of hyporesponsiveness and depletion
of NK cells in the blood of patients with COVID-19, others recorded a marked activation of
these cells [
66
]. Finally, the innate immune system interacts with coagulation—a process
known as “immuno-thrombosis”—which is thought to be dysregulated in severe
COVID-19
infection, probably because of increased tissue factor expression (TF). This action, in turn,
triggers the extrinsic pathway of coagulation [
57
]. Neutrophils, when activated, release
neutrophil extracellular traps (NETs) which are composed of neutrophil-derived DNA and
acetylated histones. These NETs function to trap and kill pathogens, but they can also
trigger a strong procoagulant response. They can promote the activation of the intrinsic
coagulation pathway through the activation of factor XII. Furthermore, they can bind TF
to activate the extrinsic coagulation pathway or form aggregates with platelets, influenc-
ing the severity of the disease [
67
]. The adaptive immune system is essential in clearing
SARS-CoV-2, utilizing activated cytotoxic T lymphocytes that destroy infected cells and
B lymphocytes that produce neutralizing antibodies against virus-specific antigens. A
significant characteristic of COVID-19 is blood lymphopenia, leading to reduced numbers
of CD4+ T cells, CD8+ T cells, and B cells [
68
]. The decrease in lymphocyte levels in
COVID-19 patients can be attributed to various factors. One reason is the low levels of
IFN-I, which hinders the production of the viral material necessary for antigen presen-
tation and the activation of adaptive immunity. Additionally, lymphopenia may occur
due to direct infection of T cells by SARS-CoV-2, cytokine-induced lymphocyte apoptosis,
and pyroptosis, MAS-related hemophagocytosis, the sequestration of lymphocytes in the
lungs or other organs, the reduction of bone marrow hematopoiesis, and the damage to
lymphoid organs induced by the virus (pathological alterations such as atrophy of the
splenic white pulp and alteration of the structure of the lymph nodes), suggesting that the
direct cytotoxicity of SARS-CoV-2 in lymphatic organs may impair the adaptive immune
response in COVID-19 [
69
]. The failure of germinal center formation in the spleen and
lymph nodes may explain why some individuals have suboptimal humoral immunity,
potentially leading to the risk of reinfection. However, most COVID-19 patients with
J. Clin. Med. 2024,13, 3987 10 of 17
mild-to-moderate disease have a strong adaptive immune response. This response includes
T cells that target antigens from protein S and nucleoprotein/membrane protein, as well
as neutralizing antibodies against protein S-derived antigens. These immune responses
can persist for months after the initial infection. It is crucial to note that coordinated adap-
tive immune responses specific to SARS-CoV-2 play a key role in mitigating the severity
of the disease [
70
]. Variations in individuals’ defense mechanisms may account for the
differences in disease progression following infection. Inadequate and uncoordinated
adaptive immune responses, often linked to aging, can result in a failure to control the
disease. This insufficient response can be attributed to “immunosenescence”, a concept
involving the age-related decline in immune function, which includes impairments in
both innate and adaptive immune responses, such as compromised pathogen recognition.
Other age-related factors contributing to this include low-level systemic inflammation in
older adults (“inflammasome”), a higher prevalence of comorbidities in this demographic,
and varying degrees of frailty [
71
74
]. The immune responses to SARS-CoV-2 may vary
based on sex, which could contribute to men being more susceptible to the disease. A
study examining differences between sexes in immune characteristics found that male
patients have a stronger initial immune response, indicated by higher levels of plasma
cytokines. On the other hand, female patients show a more effective T-cell activation [
75
].
Subjects with multiple underlying diseases or a single serious disease are at greater risk
of developing a severe form of COVID-19 (cardiovascular disease, arterial hypertension,
diabetes, lung disease, neurodegenerative disorders, immunodeficiencies, kidney disease,
obesity, and liver damage) [
76
]. In this regard, ACE2 expression and activity variations
between individuals are believed to impact vulnerability to COVID-19 progression. Para-
doxically, increased membrane-bound ACE2 may allow SARS-CoV-2 to invade host cells.
At the same time, downregulation of ACE2 (due to SARS-CoV-2-induced endocytosis)
precipitates tissue damage, (1) decreasing the inactivation of bradykinins with consequent
risk of developing angioedema and (2) dysregulating the RAAS failing to convert Ang II
into angiotensin (1–7) [
77
]. Finally, several other host factors are believed to impact the
progression of COVID-19, including epigenetic mechanisms, nutritional status, and ABO
blood type [78].
4.5. Cytokine Storm
The term “cytokine storm” describes overactive immune responses triggered by vari-
ous factors, including autoimmune diseases, viral infections, and immunotherapy [
79
82
].
Cytokine storms destroy pathogens and cause histotoxicity, affecting various organs [
83
,
84
].
Cytokine release syndrome (CRS) is a systemic inflammatory syndrome caused by cy-
tokine storms and has previously been observed in individuals infected with SARS-CoV
and MERS-CoV viruses. When the body is infected with a virus, certain molecules ac-
tivate neighboring cells’ antiviral responses and attract cells of the innate and adaptive
immune systems, such as natural killer (NK) cells, macrophages, and gamma delta (gd T)
cells
[8588]
. Interferon production helps protect neighboring epithelial cells from being in-
fected, while the release of IL-1b and IL-6 from other immune cells leads to the mobilization
of neutrophils and T cells. T cell activation or the lysis of immune cells triggers the secretion
of IFN-g and TNF-a, which in turn activates immune cells and endothelial cells, creating
a positive feedback loop leading to further release of inflammatory cytokines [
89
]. These
inflammatory mediators may contribute to thrombus formation [
57
]. This process, called
immuno-thrombosis, can also enhance cytokine production and has been attributed to the
link between thrombin and inflammasome activation and IL-1 production [
90
]. Vascular
endothelial cells are cells that line the blood vessels. These cells are exposed to various
immune mediators and cytokines circulating in the blood. Dysfunction in these cells due to
cytokine storms can cause coagulation disorders such as capillary leak syndrome, thrombus
formation, and even disseminated intravascular coagulation (D.I.C.). This dysfunction is
due to a connection between the immune response and the blood clotting system [
57
,
91
].
Cytokine storms inhibit further viral replication and cause secondary tissue damage by
J. Clin. Med. 2024,13, 3987 11 of 17
secreting large amounts of active mediators and inflammatory factors [
57
,
92
95
]. Inhibition
of this self-reinforcing inflammatory cascade may impair viral clearance and inhibit tissue
damage. In the COVID-19 study, Huang et al. discovered that individuals admitted to
intensive care units (ICUs) had elevated levels of the inflammatory cytokines IL-2, IL-7,
IL-10, G-CSF (granulocyte colony-stimulating factor), IFN-
γ
, M.C.P., and TNF-
α
in their
plasma compared to non-ICU patients.
High levels of cytokines were noted [
96
]. These cytokines indicated the presence of
both the Th1 response and Th2 response in COVID-19. In addition, monocyte activation
may indicate that the cytokine storm in COVID-19 is closely related to the imbalance
between innate and adaptive immunity. A recent study found that patients with severe
COVID-19 had significantly higher levels of IL-6 compared to those with mild or moderate
cases. Additionally, patients with severe COVID-19 showed decreased levels of CD4+ T
cells, CD8+ T cells, and NK cells, indicating immunosuppression [93].
On the other hand, T lymphocyte cells may be overactivated during cytokine storms
in COVID-19 patients, resulting in severe immune dysfunction [
97
]. A recent system-
atic review, based on autopsy findings, found fibrin clots with increased CD61-positive
platelets and megakaryocytes in the anterior and posterior capillaries. This was observed
without complete lumen obstruction in lung and other organ samples from COVID-19
patients [
98
]. As a result, cytokine storms can directly damage the pulmonary capillary
mucosa, leading to alveolar edema and further diffusion of inflammatory cytokines, which
damages alveolar structures and impairs pulmonary ventilation [
98
,
99
]. Similarly, cytokine
storms are associated with the order and severity of organ dysfunction in multiple organ
dysfunction syndrome (MODS). Therefore, cytokine storms are considered a crucial factor
in determining the outcome of patients with COVID-19 multiorgan pathology.
This study aimed to evaluate the safety and effectiveness of administering high-dose
vitamin C (as an add-on to the standard of care) to a population of patients with COVID-19
at different stages of the disease. The current findings suggest that high-dose vitamin
C—10 g
in 250 cc of saline solution in slow infusion (60 drops/min)—for three consecu-
tive days was safe and associated with shorter hospitalization in patients suffering from
COVID-19.
The assumption is based on the observation that ascorbic acid can directly reduce
the production of reactive oxygen species (R.O.S.), maintain endothelial barrier function,
promote vasodilation, and downregulate the expression of various proinflammatory mark-
ers [
100
]. Some studies have highlighted that vitamin C diminishes the production of
chemokines and cytokines such as IL1, IL6, IL8, and TNF
α
, thus counteracting the inflam-
matory alterations underlying the lung damage caused by sepsis; this was associated with
significantly lower mortality in severely ill patients with pneumonia [
32
,
101
]. Vitamin
C accumulates in neutrophils, enhancing their chemotaxis, phagocytosis, and microbial
killing. It also plays a vital role in apoptosis and the clearance of spent neutrophils from
infection sites by macrophages, thereby reducing necrosis and potential tissue damage.
Furthermore, vitamin C boosts the development and growth of B- and T-cells, possibly
due to its gene-regulating properties [
46
]. Vitamin C has been widely used in sepsis and
ARDS [
28
,
102
]. Severe inflammation and cytokine storms contribute to severe ARDS and
subsequent mortality in COVID-19 [
103
]. Considering these effects of the cytokine storm
and the close correlation with the prognosis in COVID-19 patients, treatment with high-
dose vitamin C was also evaluated, associated with a reduction in C-reactive protein (C.R.P.)
levels, procalcitonin (P.C.T.), Interleukin 8 (IL8), and attenuating pulmonary and systemic
inflammation. A pilot study conducted in China by Jing Zhang et al. [
48
] with the use
of high-dose vitamin C (24 g/day) in COVID-19 patients with critical illness, although
it did not lead to any results in what was the primary end-point (ventilator-free days
invasive mechanics), has, however, shown benefits in terms of improvement of the partial
pressure ratio of O
2
/inspiratory fraction of O
2
(P/F) and the safety of using the drug. A
retrospective cohort study conducted by D. Gao et al. [
104
] suggested a benefit in terms of
reduced mortality and improved oxygenation status in COVID-19 patients with the use of
J. Clin. Med. 2024,13, 3987 12 of 17
high-dose vitamin C. Multiple studies have demonstrated the beneficial effects of vitamin
C in lowering mortality and reducing hospital stays for patients with non-COVID-related
sepsis and ARDS. The CITRIS-ALI study was a significant clinical trial that included 167 pa-
tients with ARDS. It was a randomized, double-blind, placebo-controlled trial. Patients
were randomly assigned to receive 50 mg/kg of vitamin C every 6 h for four days. This
treatment resulted in a statistically significant decrease in 28-day all-cause mortality com-
pared to the placebo [
32
]. In patients who received a total of 200 mg/kg/day of high-dose
intravenous vitamin C (HDIVC) for four days (administered at 50 mg/kg/dose, every 6 h),
scores for organ failure were significantly lower than those receiving the placebo and even
lower than patients who received lower doses of intravenous vitamin C (50 mg/kg/day
administered at 12.5 mg/kg/dose, every 6 h for four days). In another study by Jamali
Moghadam Siahkali et al. [
105
], a dosage of 1.5 g of vitamin C given intravenously every
6 h for five days was used. In addition, improvements in peripheral oxygen saturation
and body temperature were found in both groups during hospitalization. The study did
not, however, find a statistically significant difference in the reduction in-hospital mortal-
ity of patients with moderate and severe disease in the intervention group compared to
the placebo, and the main reason could lie in the dosage of intravenous vitamin C used
(lower than to other studies conducted). A recent systematic review and meta-analysis of
randomized controlled trials and trial sequential analysis found that intravenous vitamin
C monotherapy may have mortality benefits for critically ill patients, especially for those at
high risk of death. However, the certainty of the evidence available is low [106].
In our study, we administered 10 g of vitamin C every 24 h for three consecutive days,
according to our therapeutic protocol, Use of Ascorbic Acid in patients with COVID-19”,
developed by our center and registered in March 2020 on the platform Clinicaltrials.gov.
We aimed to evaluate the safety of administering vitamin C; the treatment was compared
to the standard of care alone. The aim of the study was fully achieved, and there was a
clear tendency towards the effectiveness of endovenous acid ascorbic treatment even if the
statistical significance was not reached. In our study, we did not record any adverse events
and we did not record an increase in mortality. Mortality, which we consider an essential
safety signal, correcting for all the confounding variables, suggested the effectiveness of
vitamin C even if statistical significance was not reached. In addition, a statistical reduction
in the length of stay in the hospital was reached. It is worth outlining that our study has
some limitations, such as the heterogeneity of the population (presence of patients with
different disease stages) or the tendency to use the treatment mainly on patients with more
severe forms of the disease. This selection bias could explain the absence of statistically
significant effectiveness. Another critical limitation lies in the fact that our sample size
is small. The sample size calculated at the beginning of the study was not reached, and
this fact could affect the negative results about vitamin C’s effectiveness because the small
sample size increases the probability of false negatives. However, according to our results,
we have a reasonable certainty about safety.
Finally, the total daily dose of vitamin C used in our study was moderate compared
to studies on the effectiveness of intravenous vitamin C in COVID-19 for four to seven
days of intravenous vitamin C. A more extended treatment could be necessary to reach
effectiveness in infective diseases.
5. Conclusions
According to our results, high-dose vitamin C administration showed an excellent
short-term safety profile in moderate and severe COVID-19 patients. The infusion of
vitamin C is statistically significantly associated with reduced hospital stay. Moreover, high-
dose intravenous vitamin C may reduce inflammatory reactions, improve oxygen support,
and decrease mortality in COVID-19 patients and other inflammatory diseases without
adverse events. It is therefore indicated as a promising therapy for patients with moderate
to severe COVID-19, but also for other pathologies in which the hyperinflammatory and
inflammasome state play a crucial role in the pathogenesis. More research is necessary to
J. Clin. Med. 2024,13, 3987 13 of 17
validate these encouraging findings and better understand the role of intravenous vitamin
C in treating COVID-19 and other inflammatory conditions. Furthermore, since it is an
inexpensive and widely available drug, its possible application and usefulness could
represent a change of direction in the treatment strategy for this kind of patient and a large
number of inpatients, even to blunt the inflammatory state.
Author Contributions: Conceptualization, S.C. and C.A.; methodology, S.C., G.N. and C.A.; software,
G.N.; validation, S.C. and C.A.; formal analysis, F.G., F.D.B., M.R. and F.A.; investigation, M.R. and
F.A.; resources, F.G. and F.D.B.; data curation, G.N. and M.R.; writing—original draft preparation,
C.A.; writing—review and editing, S.C.; visualization, F.G., F.D.B. and G.N.; supervision, S.C.; project
administration, C.A. All authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding. The study was spontaneous research supported
by the ARNAS Civico, Di Cristina, Benfratelli, Palermo, Italy.
Institutional Review Board Statement: The Ethics Committee has approved the conduct of the study
at our institution. The approval number assigned to this study is 3143-2020.
Informed Consent Statement: Informed consent was obtained from all subjects involved in the study.
Data Availability Statement: The data presented in this study are available on request from the
corresponding author. The data are not publicly available due to privacy.
Acknowledgments: We would like to thank the Internal Medicine IGR COVID-19 Investigators
(Salvatore Corrao, Federica Agugliaro, Federica Bellini, Egle Briulotta, Giuseppe Brunori, Adele
Busardò, Luigi Calvo, Ignazio Cangemi, Maria Grazia Cecala, Bruno Curiale, Vincenzo De Blasi, Irene
Favatella, AnnaRita Giardina, Walter Granà, Fabio Lanzarone, Santi Lauria, Umberto Lupo, Carlo
Mannina, Martina Martorana, Silvia Messina, Gianni Mingoia, Davide Morana, Salvatore Morello,
Salvo Mularo, Maurilio Palazzo, Karen Pinelli, Giovanni Pistone, Agostino Racalbuto, Massimo
Raspanti, Serena Scardina, Martina Vacca, Giorgia Virzì, and Christiano Argano).
Conflicts of Interest: The authors declare no conflicts of interest.
References
1.
WHO Director-General’s Opening Remarks at the Media Briefing on COVID-19—11 March 2020. Available online:
https://www.who.int/director-general/speeches/detail/who-director-general-s-opening-remarks-at-the-media-briefing-on-
covid-19---11-march-2020 (accessed on 14 October 2023).
2.
Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R.; et al. A Novel Coronavirus from
Patients with Pneumonia in China, 2019. N Engl. J. Med. 2020,382, 727–733. [CrossRef] [PubMed]
3. WHO Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int (accessed on 14 October 2023).
4.
Filip, R.; Gheorghita Puscaselu, R.; Anchidin-Norocel, L.; Dimian, M.; Savage, W.K. Global Challenges to Public Health Care
Systems during the COVID-19 Pandemic: A Review of Pandemic Measures and Problems. J. Pers. Med. 2022,12, 1295. [CrossRef]
[PubMed]
5.
Parasher, A. COVID-19: Current Understanding of Its Pathophysiology, Clinical Presentation and Treatment. Postgrad. Med. J.
2021,97, 312–320. [CrossRef]
6.
Waterer, G. Understanding COVID-19 Pathophysiology: What Defines Progress? Am. J. Respir. Cell Mol. Biol. 2022,66, 120–121.
[CrossRef]
7. Lui, G.; Guaraldi, G. Drug Treatment of COVID-19 Infection. Curr. Opin. Pulm. Med. 2023,29, 174–183. [CrossRef] [PubMed]
8.
Gavriatopoulou, M.; Ntanasis-Stathopoulos, I.; Korompoki, E.; Fotiou, D.; Migkou, M.; Tzanninis, I.-G.; Psaltopoulou, T.; Kastritis,
E.; Terpos, E.; Dimopoulos, M.A. Emerging Treatment Strategies for COVID-19 Infection. Clin. Exp. Med. 2021,21, 167–179.
[CrossRef] [PubMed]
9.
Szarpak, L.; Pruc, M.; Gasecka, A.; Jaguszewski, M.J.; Michalski, T.; Peacock, F.W.; Smereka, J.; Pytkowska, K.; Filipiak, K.J.
Should We Supplement Zinc in COVID-19 Patients? Evidence from a Meta-Analysis. Pol. Arch. Intern. Med. 2021,131, 802–807.
[CrossRef] [PubMed]
10.
Berti, A.D.; Kale-Pradhan, P.B.; Giuliano, C.A.; Aprilliano, B.N.; Miller, C.R.; Alyashae, B.T.; Bhargava, A.; Johnson, L.B. Clinical
Outcomes of Zinc Supplementation Among COVID-19 Patients. Curr. Drug Saf. 2022,17, 366–369. [CrossRef] [PubMed]
11.
Bae, M.; Kim, H. Mini-Review on the Roles of Vitamin C, Vitamin D, and Selenium in the Immune System against COVID-19.
Molecules 2020,25, 5346. [CrossRef]
12.
Corrao, S.; Mallaci Bocchio, R.; Lo Monaco, M.; Natoli, G.; Cavezzi, A.; Troiani, E.; Argano, C. Does Evidence Exist to Blunt
Inflammatory Response by Nutraceutical Supplementation during COVID-19 Pandemic? An Overview of Systematic Reviews of
Vitamin D, Vitamin C, Melatonin, and Zinc. Nutrients 2021,13, 1261. [CrossRef]
J. Clin. Med. 2024,13, 3987 14 of 17
13.
Argano, C.; Mallaci Bocchio, R.; Lo Monaco, M.; Scibetta, S.; Natoli, G.; Cavezzi, A.; Troiani, E.; Corrao, S. An Overview of
Systematic Reviews of the Role of Vitamin D on Inflammation in Patients with Diabetes and the Potentiality of Its Application on
Diabetic Patients with COVID-19. Int. J. Mol. Sci. 2022,23, 2873. [CrossRef]
14.
Argano, C.; Mallaci Bocchio, R.; Natoli, G.; Scibetta, S.; Lo Monaco, M.; Corrao, S. Protective Effect of Vitamin D Supplementation
on COVID-19-Related Intensive Care Hospitalization and Mortality: Definitive Evidence from Meta-Analysis and Trial Sequential
Analysis. Pharmaceuticals 2023,16, 130. [CrossRef] [PubMed]
15.
Milani, G.P.; Macchi, M.; Guz-Mark, A. Vitamin C in the Treatment of COVID-19. Nutrients 2021,13, 1172. [CrossRef] [PubMed]
16.
Rawat, D.; Roy, A.; Maitra, S.; Shankar, V.; Khanna, P.; Baidya, D.K. Vitamin D Supplementation and COVID-19 Treatment: A
Systematic Review and Meta-Analysis. Diabetes Metab. Syndr. 2021,15, 102189. [CrossRef]
17.
Olczak-Pruc, M.; Swieczkowski, D.; Ladny, J.R.; Pruc, M.; Juarez-Vela, R.; Rafique, Z.; Peacock, F.W.; Szarpak, L. Vitamin C
Supplementation for the Treatment of COVID-19: A Systematic Review and Meta-Analysis. Nutrients 2022,14, 4217. [CrossRef]
[PubMed]
18.
Linster, C.L.; Van Schaftingen, E. Vitamin C. Biosynthesis, Recycling and Degradation in Mammals. FEBS J. 2007,274, 1–22.
[CrossRef] [PubMed]
19.
Padayatty, S.J.; Katz, A.; Wang, Y.; Eck, P.; Kwon, O.; Lee, J.-H.; Chen, S.; Corpe, C.; Dutta, A.; Dutta, S.K.; et al. Vitamin C as an
Antioxidant: Evaluation of Its Role in Disease Prevention. J. Am. Coll. Nutr. 2003,22, 18–35. [CrossRef] [PubMed]
20.
Shilotri, P.; Bhat, K. Effect of Mega Doses of Vitamin C on Bactericidal Ativity of Leukocytes. Am. J. Clin. Nutr. 1977,30, 1077–1081.
[CrossRef]
21.
Liugan, M.; Carr, A.C. Vitamin C and Neutrophil Function: Findings from Randomized Controlled Trials. Nutrients 2019,11, 2102.
[CrossRef]
22.
Manning, J.; Mitchell, B.; Appadurai, D.A.; Shakya, A.; Pierce, L.J.; Wang, H.; Nganga, V.; Swanson, P.C.; May, J.M.; Tantin, D.;
et al. Vitamin C Promotes Maturation of T-Cells. Antioxid. Redox Signal. 2013,19, 2054–2067. [CrossRef]
23. Hemilä, H.; Louhiala, P. Vitamin C May Affect Lung Infections. J. R. Soc. Med. 2007,100, 495–498. [CrossRef] [PubMed]
24.
Myint, P.K.; Wilson, A.M.; Clark, A.B.; Luben, R.N.; Wareham, N.J.; Khaw, K.-T. Plasma Vitamin C Concentrations and Risk of
Incident Respiratory Diseases and Mortality in the European Prospective Investigation into Cancer-Norfolk Population-Based
Cohort Study. Eur. J. Clin. Nutr. 2019,73, 1492–1500. [CrossRef] [PubMed]
25.
Hemilä, H.; Louhiala, P. Vitamin C for Preventing and Treating Pneumonia. Cochrane Database Syst. Rev. 2013,8, CD005532.
[CrossRef] [PubMed]
26.
Sharma, Y.; Sumanadasa, S.; Shahi, R.; Woodman, R.; Mangoni, A.A.; Bihari, S.; Thompson, C. Efficacy and Safety of Vitamin C
Supplementation in the Treatment of Community-Acquired Pneumonia: A Systematic Review and Meta-Analysis with Trial
Sequential Analysis. Sci. Rep. 2024,14, 11846. [CrossRef] [PubMed]
27.
Brant, E.B.; Angus, D.C. Is High-Dose Vitamin C Beneficial for Patients with Sepsis? JAMA 2019,322, 1257–1258. [CrossRef]
[PubMed]
28.
Marik, P.E.; Khangoora, V.; Rivera, R.; Hooper, M.H.; Catravas, J. Hydrocortisone, Vitamin C, and Thiamine for the Treatment of
Severe Sepsis and Septic Shock: A Retrospective Before-After Study. Chest 2017,151, 1229–1238. [CrossRef] [PubMed]
29.
Litwak, J.J.; Cho, N.; Nguyen, H.B.; Moussavi, K.; Bushell, T. Vitamin C, Hydrocortisone, and Thiamine for the Treatment of
Severe Sepsis and Septic Shock: A Retrospective Analysis of Real-World Application. J. Clin. Med. 2019,8, 478. [CrossRef]
[PubMed]
30.
Fowler, A.A.; Syed, A.A.; Knowlson, S.; Sculthorpe, R.; Farthing, D.; DeWilde, C.; Farthing, C.A.; Larus, T.L.; Martin, E.; Brophy,
D.F.; et al. Phase I Safety Trial of Intravenous Ascorbic Acid in Patients with Severe Sepsis. J. Transl. Med. 2014,12, 32. [CrossRef]
31.
Wei, X.; Wang, Z.; Liao, X.; Guo, W.; Wen, J.-Y.; Qin, T.; Wang, S. Efficacy of Vitamin C in Patients with Sepsis: An Updated
Meta-Analysis. Eur. J. Pharmacol. 2020,868, 172889. [CrossRef]
32.
Fowler, A.A.; Truwit, J.D.; Hite, R.D.; Morris, P.E.; DeWilde, C.; Priday, A.; Fisher, B.; Thacker, L.R.; Natarajan, R.; Brophy, D.F.;
et al. Effect of Vitamin C Infusion on Organ Failure and Biomarkers of Inflammation and Vascular Injury in Patients With Sepsis
and Severe Acute Respiratory Failure: The CITRIS-ALI Randomized Clinical Trial. JAMA 2019,322, 1261–1270. [CrossRef]
33.
Zhang, M.; Jativa, D.F. Vitamin C Supplementation in the Critically Ill: A Systematic Review and Meta-Analysis. SAGE Open Med.
2018,6, 2050312118807615. [CrossRef] [PubMed]
34.
Langlois, P.L.; Manzanares, W.; Adhikari, N.K.J.; Lamontagne, F.; Stoppe, C.; Hill, A.; Heyland, D.K. Vitamin C Administration to
the Critically Ill: A Systematic Review and Meta-Analysis. J. Parenter. Enter. Nutr. 2019,43, 335–346. [CrossRef]
35.
Hemilä, H.; Chalker, E. Vitamin C May Reduce the Duration of Mechanical Ventilation in Critically Ill Patients: A Meta-Regression
Analysis. J. Intensive Care 2020,8, 15. [CrossRef] [PubMed]
36.
Kim, Y.; Kim, H.; Bae, S.; Choi, J.; Lim, S.Y.; Lee, N.; Kong, J.M.; Hwang, Y.; Kang, J.S.; Lee, W.J. Vitamin C Is an Essential Factor
on the Anti-Viral Immune Responses through the Production of Interferon-α/βat the Initial Stage of Influenza A Virus (H3N2)
Infection. Immune Netw. 2013,13, 70–74. [CrossRef]
37.
Teafatiller, T.; Agrawal, S.; De Robles, G.; Rahmatpanah, F.; Subramanian, V.S.; Agrawal, A. Vitamin C Enhances Anti-viral
Functions of Lung Epithelial Cells. Biomolecules 2021,11, 1148. [CrossRef] [PubMed]
38.
Padayatty, S.J.; Sun, H.; Wang, Y.; Riordan, H.D.; Hewitt, S.M.; Katz, A.; Wesley, R.A.; Levine, M. Vitamin C Pharmacokinetics:
Implications for Oral and Intravenous Use. Ann. Intern. Med. 2004,140, 533–537. [CrossRef]
J. Clin. Med. 2024,13, 3987 15 of 17
39.
Vollbracht, C.; Kraft, K. Feasibility of Vitamin C in the Treatment of Post Viral Fatigue with Focus on Long COVID, Based on a
Systematic Review of IV Vitamin C on Fatigue. Nutrients 2021,13, 1154. [CrossRef] [PubMed]
40.
Colunga Biancatelli, R.M.L.; Berrill, M.; Catravas, J.D.; Marik, P.E. Quercetin and Vitamin C: An Experimental, Synergistic
Therapy for the Prevention and Treatment of SARS-CoV-2 Related Disease (COVID-19). Front. Immunol. 2020,11, 1451. [CrossRef]
41.
Corrao, S. Use of Ascorbic Acid in Patients with COVID 19. 2020. Available online: https://clinicaltrials.gov/ (accessed on
14 January 2024).
42.
Hosmer, D.W., Jr.; Lemeshow, S. Applied Logistic Regression; John Wiley & Sons: Hoboken, NJ, USA, 2004; ISBN 978-0-471-65402-5.
43.
May, J.M.; Harrison, F.E. Role of Vitamin C in the Function of the Vascular Endothelium. Antioxid. Redox Signal 2013,19, 2068–2083.
[CrossRef]
44.
Cerullo, G.; Negro, M.; Parimbelli, M.; Pecoraro, M.; Perna, S.; Liguori, G.; Rondanelli, M.; Cena, H.; D’Antona, G. The Long
History of Vitamin C: From Prevention of the Common Cold to Potential Aid in the Treatment of COVID-19. Front. Immunol.
2020,11, 574029. [CrossRef]
45.
Vissers, M.C.M.; Wilkie, R.P. Ascorbate Deficiency Results in Impaired Neutrophil Apoptosis and Clearance and Is Associated
with Up-Regulation of Hypoxia-Inducible Factor 1α.J. Leukoc. Biol. 2007,81, 1236–1244. [CrossRef] [PubMed]
46. Carr, A.C.; Maggini, S. Vitamin C and Immune Function. Nutrients 2017,9, 1211. [CrossRef]
47.
A Coordinated Global Research Roadmap. Available online: https://www.who.int/publications/m/item/a- coordinated-global-
research-roadmap (accessed on 22 October 2023).
48.
Zhang, J.; Rao, X.; Li, Y.; Zhu, Y.; Liu, F.; Guo, G.; Luo, G.; Meng, Z.; De Backer, D.; Xiang, H.; et al. Pilot Trial of High-Dose
Vitamin C in Critically Ill COVID-19 Patients. Ann. Intensive Care 2021,11, 5. [CrossRef] [PubMed]
49.
Kumari, P.; Dembra, S.; Dembra, P.; Bhawna, F.; Gul, A.; Ali, B.; Sohail, H.; Kumar, B.; Memon, M.K.; Rizwan, A. The Role of
Vitamin C as Adjuvant Therapy in COVID-19. Cureus 2020,12, e11779. [CrossRef]
50.
Li, M.; Ching, T.H.; Hipple, C.; Lopez, R.; Sahibzada, A.; Rahman, H. Use of Intravenous Vitamin C in Critically Ill Patients With
COVID-19 Infection. J. Pharm. Pract. 2023,36, 60–66. [CrossRef] [PubMed]
51. Chaplin, D.D. Overview of the Immune Response. J. Allergy Clin. Immunol. 2010,125, S3–S23. [CrossRef]
52.
Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J.; Wang, B.; Xiang, H.; Cheng, Z.; Xiong, Y.; et al. Clinical Characteristics
of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA 2020,323, 1061–1069.
[CrossRef]
53.
Bergsbaken, T.; Fink, S.L.; Cookson, B.T. Pyroptosis: Host Cell Death and Inflammation. Nat. Rev. Microbiol. 2009,7, 99–109.
[CrossRef]
54.
Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qiu, Y.; Wang, J.; Liu, Y.; Wei, Y.; et al. Epidemiological and Clinical
Characteristics of 99 Cases of 2019 Novel Coronavirus Pneumonia in Wuhan, China: A Descriptive Study. Lancet 2020,395,
507–513. [CrossRef]
55.
Corrao, S.; Gervasi, F.; Di Bernardo, F.; Natoli, G.; Raspanti, M.; Catalano, N.; Argano, C. Immunological Characteristics of
Non-Intensive Care Hospitalized COVID-19 Patients: A Preliminary Report. J. Clin. Med. 2021,10, 849. [CrossRef]
56. Corrao, S.; Gervasi, F.; Di Bernardo, F.; Argano, C. Immune Response Failure in Paucisymptomatic Long-Standing SARS-CoV-2
Spreaders. Clin. Pract. 2021,11, 151–161. [CrossRef]
57.
Engelmann, B.; Massberg, S. Thrombosis as an Intravascular Effector of Innate Immunity. Nat. Rev. Immunol. 2013,13, 34–45.
[CrossRef] [PubMed]
58.
Zhao, N.; Di, B.; Xu, L. The NLRP3 Inflammasome and COVID-19: Activation, Pathogenesis and Therapeutic Strategies. Cytokine
Growth Factor. Rev. 2021,61, 2–15. [CrossRef] [PubMed]
59.
Barker, B.R.; Taxman, D.J.; Ting, J.P.-Y. Cross-Regulation between the IL-1
β
/IL-18 Processing Inflammasome and Other Inflamma-
tory Cytokines. Curr. Opin. Immunol. 2011,23, 591–597. [CrossRef] [PubMed]
60.
Lin, L.; Xu, L.; Lv, W.; Han, L.; Xiang, Y.; Fu, L.; Jin, M.; Zhou, R.; Chen, H.; Zhang, A. An NLRP3 Inflammasome-Triggered
Cytokine Storm Contributes to Streptococcal Toxic Shock-like Syndrome (STSLS). PLoS Pathog. 2019,15, e1007795. [CrossRef]
61.
Rodrigues, T.S.; de Sá, K.S.G.; Ishimoto, A.Y.; Becerra, A.; Oliveira, S.; Almeida, L.; Gonçalves, A.V.; Perucello, D.B.; Andrade,
W.A.; Castro, R.; et al. Inflammasomes Are Activated in Response to SARS-CoV-2 Infection and Are Associated with COVID-19
Severity in Patients. J. Exp. Med. 2021,218, e20201707. [CrossRef] [PubMed]
62.
Liu, J.; Liu, Y.; Xiang, P.; Pu, L.; Xiong, H.; Li, C.; Zhang, M.; Tan, J.; Xu, Y.; Song, R.; et al. Neutrophil-to-Lymphocyte Ratio
Predicts Critical Illness Patients with 2019 Coronavirus Disease in the Early Stage. J. Transl. Med. 2020,18, 206. [CrossRef]
[PubMed]
63.
Xia, H.; Cao, Z.; Xie, X.; Zhang, X.; Chen, J.Y.-C.; Wang, H.; Menachery, V.D.; Rajsbaum, R.; Shi, P.-Y. Evasion of Type I Interferon
by SARS-CoV-2. Cell Rep. 2020,33, 108234. [CrossRef] [PubMed]
64.
Tian, W.; Zhang, N.; Jin, R.; Feng, Y.; Wang, S.; Gao, S.; Gao, R.; Wu, G.; Tian, D.; Tan, W.; et al. Immune Suppression in the Early
Stage of COVID-19 Disease. Nat. Commun. 2020,11, 5859. [CrossRef]
65.
Noris, M.; Benigni, A.; Remuzzi, G. The Case of Complement Activation in COVID-19 Multiorgan Impact. Kidney Int. 2020,98,
314–322. [CrossRef]
66.
Osuchowski, M.F.; Winkler, M.S.; Skirecki, T.; Cajander, S.; Shankar-Hari, M.; Lachmann, G.; Monneret, G.; Venet, F.; Bauer, M.;
Brunkhorst, F.M.; et al. The COVID-19 Puzzle: Deciphering Pathophysiology and Phenotypes of a New Disease Entity. Lancet
Respir. Med. 2021,9, 622–642. [CrossRef] [PubMed]
J. Clin. Med. 2024,13, 3987 16 of 17
67.
Zuo, Y.; Yalavarthi, S.; Shi, H.; Gockman, K.; Zuo, M.; Madison, J.A.; Blair, C.N.; Weber, A.; Barnes, B.J.; Egeblad, M.; et al.
Neutrophil Extracellular Traps in COVID-19. J.C.I. Insight 2020,5, e138999. [CrossRef] [PubMed]
68.
Zhang, X.; Tan, Y.; Ling, Y.; Lu, G.; Liu, F.; Yi, Z.; Jia, X.; Wu, M.; Shi, B.; Xu, S.; et al. Viral and Host Factors Related to the Clinical
Outcome of COVID-19. Nature 2020,583, 437–440. [CrossRef]
69.
Van Eijk, L.E.; Binkhorst, M.; Bourgonje, A.R.; Offringa, A.K.; Mulder, D.J.; Bos, E.M.; Kolundzic, N.; Abdulle, A.E.; Van Der
Voort, P.H.; Olde Rikkert, M.G.; et al. COVID-19: Immunopathology, Pathophysiological Mechanisms, and Treatment Options. J.
Pathol. 2021,254, 307–331. [CrossRef] [PubMed]
70.
Rydyznski Moderbacher, C.; Ramirez, S.I.; Dan, J.M.; Grifoni, A.; Hastie, K.M.; Weiskopf, D.; Belanger, S.; Abbott, R.K.; Kim, C.;
Choi, J.; et al. Antigen-Specific Adaptive Immunity to SARS-CoV-2 in Acute COVID-19 and Associations with Age and Disease
Severity. Cell 2020,183, 996–1012. [CrossRef]
71.
Hewitt, J.; Carter, B.; Vilches-Moraga, A.; Quinn, T.J.; Braude, P.; Verduri, A.; Pearce, L.; Stechman, M.; Short, R.; Price, A.; et al.
The Effect of Frailty on Survival in Patients with COVID-19 (COPE): A Multicentre, European, Observational Cohort Study. Lancet
Public Health 2020,5, e444–e451. [CrossRef]
72.
Argano, C.; Scichilone, N.; Natoli, G.; Nobili, A.; Corazza, G.R.; Mannucci, P.M.; Perticone, F.; Corrao, S.; REPOSI Investigators.
Pattern of Comorbidities and 1-Year Mortality in Elderly Patients with COPD Hospitalized in Internal Medicine Wards: Data
from the RePoSI Registry. Intern. Emerg. Med. 2021,16, 389–400. [CrossRef] [PubMed]
73.
Argano, C.; Natoli, G.; Mularo, S.; Nobili, A.; Monaco, M.L.; Mannucci, P.M.; Perticone, F.; Pietrangelo, A.; Corrao, S. Impact of
Diabetes Mellitus and Its Comorbidities on Elderly Patients Hospitalized in Internal Medicine Wards: Data from the RePoSi
Registry. Healthcare 2022,10, 86. [CrossRef]
74.
Corrao, S.; Natoli, G.; Nobili, A.; Mannucci, P.M.; Pietrangelo, A.; Perticone, F.; Argano, C.; RePoSI Investigators. Comorbidity
Does Not Mean Clinical Complexity: Evidence from the RePoSI Register. Intern. Emerg. Med. 2020,15, 621–628. [CrossRef]
75.
Takahashi, T.; Ellingson, M.K.; Wong, P.; Israelow, B.; Lucas, C.; Klein, J.; Silva, J.; Mao, T.; Oh, J.E.; Tokuyama, M.; et al. Sex
Differences in Immune Responses That Underlie COVID-19 Disease Outcomes. Nature 2020,588, 315–320. [CrossRef]
76.
Williamson, E.J.; Walker, A.J.; Bhaskaran, K.; Bacon, S.; Bates, C.; Morton, C.E.; Curtis, H.J.; Mehrkar, A.; Evans, D.; Inglesby, P.;
et al. Factors Associated with COVID-19-Related Death Using OpenSAFELY. Nature 2020,584, 430–436. [CrossRef] [PubMed]
77.
Van De Veerdonk, F.L.; Netea, M.G.; Van Deuren, M.; Van Der Meer, J.W.; De Mast, Q.; Brüggemann, R.J.; Van Der Hoeven, H.
Kallikrein-Kinin Blockade in Patients with COVID-19 to Prevent Acute Respiratory Distress Syndrome. eLife 2020,9, e57555.
[CrossRef] [PubMed]
78.
Briguglio, M.; Pregliasco, F.E.; Lombardi, G.; Perazzo, P.; Banfi, G. The Malnutritional Status of the Host as a Virulence Factor for
New Coronavirus SARS-CoV-2. Front. Med. 2020,7, 146. [CrossRef] [PubMed]
79.
Shimabukuro-Vornhagen, A.; Gödel, P.; Subklewe, M.; Stemmler, H.J.; Schlößer, H.A.; Schlaak, M.; Kochanek, M.; Böll, B.; von
Bergwelt-Baildon, M.S. Cytokine Release Syndrome. J. Immunother. Cancer 2018,6, 56. [CrossRef] [PubMed]
80.
Lee, D.W.; Gardner, R.; Porter, D.L.; Louis, C.U.; Ahmed, N.; Jensen, M.; Grupp, S.A.; Mackall, C.L. Current Concepts in the
Diagnosis and Management of Cytokine Release Syndrome. Blood 2014,124, 188–195. [CrossRef] [PubMed]
81. Fajgenbaum, D.C.; June, C.H. Cytokine Storm. N. Engl. J. Med. 2020,383, 2255–2273. [CrossRef] [PubMed]
82.
Karki, R.; Kanneganti, T.-D. The “Cytokine Storm”: Molecular Mechanisms and Therapeutic Prospects. Trends Immunol. 2021,42,
681–705. [CrossRef]
83.
Zaim, S.; Chong, J.H.; Sankaranarayanan, V.; Harky, A. COVID-19 and Multiorgan Response. Curr. Probl. Cardiol. 2020,45, 100618.
[CrossRef]
84. Hu, B.; Huang, S.; Yin, L. The Cytokine Storm and COVID-19. J. Med. Virol. 2021,93, 250–256. [CrossRef]
85. Mangalmurti, N.; Hunter, C.A. Cytokine Storms: Understanding COVID-19. Immunity 2020,53, 19–25. [CrossRef]
86.
Harker, J.A.; Lewis, G.M.; Mack, L.; Zuniga, E.I. Late Interleukin-6 Escalates T Follicular Helper Cell Responses and Controls a
Chronic Viral Infection. Science 2011,334, 825–829. [CrossRef] [PubMed]
87.
Azkur, A.K.; Akdis, M.; Azkur, D.; Sokolowska, M.; van de Veen, W.; Brüggen, M.; O’Mahony, L.; Gao, Y.; Nadeau, K.; Akdis, C.A.
Immune Response to SARS-CoV-2 and Mechanisms of Immunopathological Changes in COVID-19. Allergy 2020,75, 1564–1581.
[CrossRef] [PubMed]
88.
Medzhitov, R. Recognition of Microorganisms and Activation of the Immune Response. Nature 2007,449, 819–826. [CrossRef]
[PubMed]
89.
Klinkhammer, J.; Schnepf, D.; Ye, L.; Schwaderlapp, M.; Gad, H.H.; Hartmann, R.; Garcin, D.; Mahlakõiv, T.; Staeheli, P. IFN-
λ
Prevents Influenza Virus Spread from the Upper Airways to the Lungs and Limits Virus Transmission. eLife 2018,7, e33354.
[CrossRef] [PubMed]
90.
Betakova, T.; Kostrabova, A.; Lachova, V.; Turianova, L. Cytokines Induced During Influenza Virus Infection. Curr. Pharm. Des.
2017,23, 2616–2622. [CrossRef] [PubMed]
91. Kawai, T.; Akira, S. Innate Immune Recognition of Viral Infection. Nat. Immunol. 2006,7, 131–137. [CrossRef]
92.
Li, X.; Geng, M.; Peng, Y.; Meng, L.; Lu, S. Molecular Immune Pathogenesis and Diagnosis of COVID-19. J. Pharm. Anal. 2020,10,
102–108. [CrossRef]
93.
Iwasaki, A.; Medzhitov, R. Regulation of Adaptive Immunity by the Innate Immune System. Science 2010,327, 291–295. [CrossRef]
[PubMed]
J. Clin. Med. 2024,13, 3987 17 of 17
94.
Zhou, Y.; Fu, B.; Zheng, X.; Wang, D.; Zhao, C.; Qi, Y.; Sun, R.; Tian, Z.; Xu, X.; Wei, H. Pathogenic T-Cells and Inflammatory
Monocytes Incite Inflammatory Storms in Severe COVID-19 Patients. Natl. Sci. Rev. 2020,7, 998–1002. [CrossRef]
95.
Wang, F.; Nie, J.; Wang, H.; Zhao, Q.; Xiong, Y.; Deng, L.; Song, S.; Ma, Z.; Mo, P.; Zhang, Y. Characteristics of Peripheral
Lymphocyte Subset Alteration in COVID-19 Pneumonia. J. Infect. Dis. 2020,221, jiaa150. [CrossRef]
96.
Xu, Z.; Shi, L.; Wang, Y.; Zhang, J.; Huang, L.; Zhang, C.; Liu, S.; Zhao, P.; Liu, H.; Zhu, L.; et al. Pathological Findings of
COVID-19 Associated with Acute Respiratory Distress Syndrome. Lancet Respir. Med. 2020,8, 420–422. [CrossRef] [PubMed]
97.
Braciale, T.J.; Sun, J.; Kim, T.S. Regulating the Adaptive Immune Response to Respiratory Virus Infection. Nat. Rev. Immunol.
2012,12, 295–305. [CrossRef]
98.
D’Errico, S.; Zanon, M.; Montanaro, M.; Radaelli, D.; Sessa, F.; Di Mizio, G.; Montana, A.; Corrao, S.; Salerno, M.; Pomara, C. More
than Pneumonia: Distinctive Features of SARS-CoV-2 Infection. From Autopsy Findings to Clinical Implications: A Systematic
Review. Microorganisms 2020,8, 1642. [CrossRef] [PubMed]
99. Newton, A.H.; Cardani, A.; Braciale, T.J. The Host Immune Response in Respiratory Virus Infection: Balancing Virus Clearance
and Immunopathology. Semin. Immunopathol. 2016,38, 471–482. [CrossRef] [PubMed]
100. G˛egotek, A.; Skrzydlewska, E. Ascorbic Acid as Antioxidant. Vitam. Horm. 2023,121, 247–270. [CrossRef] [PubMed]
101.
Grudlewska-Buda, K.; Wiktorczyk-Kapischke, N.; Budzy´nska, A.; Kwieci´nska-Piróg, J.; Przekwas, J.; Kijewska, A.; Sabiniarz, D.;
Gospodarek-Komkowska, E.; Skowron, K. The Variable Nature of Vitamin C—Does It Help When Dealing with Coronavirus?
Antioxidants 2022,11, 1247. [CrossRef] [PubMed]
102.
Kim, W.-Y.; Jo, E.-J.; Eom, J.S.; Mok, J.; Kim, M.-H.; Kim, K.U.; Park, H.-K.; Lee, M.K.; Lee, K. Combined Vitamin C, Hydrocortisone,
and Thiamine Therapy for Patients with Severe Pneumonia Who Were Admitted to the Intensive Care Unit: Propensity Score-
Based Analysis of a before-after Cohort Study. J. Crit. Care 2018,47, 211–218. [CrossRef]
103.
Ramasamy, S.; Subbian, S. Critical Determinants of Cytokine Storm and Type I Interferon Response in COVID-19 Pathogenesis.
Clin. Microbiol. Rev. 2021,34, e00299-20. [CrossRef] [PubMed]
104.
Gao, D.; Xu, M.; Wang, G.; Lv, J.; Ma, X.; Guo, Y.; Zhang, D.; Yang, H.; Jiang, W.; Deng, F.; et al. The Efficiency and Safety of
High-Dose Vitamin C in Patients with COVID-19: A Retrospective Cohort Study. Aging 2021,13, 7020–7034. [CrossRef]
105.
JamaliMoghadamSiahkali, S.; Zarezade, B.; Koolaji, S.; SeyedAlinaghi, S.; Zendehdel, A.; Tabarestani, M.; Sekhavati Moghadam,
E.; Abbasian, L.; Dehghan Manshadi, S.A.; Salehi, M.; et al. Safety and Effectiveness of High-Dose Vitamin C in Patients with
COVID-19: A Randomized Open-Label Clinical Trial. Eur. J. Med. Res. 2021,26, 20. [CrossRef]
106.
Lee, Z.-Y.; Ortiz-Reyes, L.; Lew, C.C.H.; Hasan, M.S.; Ke, L.; Patel, J.J.; Stoppe, C.; Heyland, D.K. Intravenous Vitamin C
Monotherapy in Critically Ill Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Trials with Trial
Sequential Analysis. Ann. Intensive Care 2023,13, 14. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... Clinical studies have explored these properties, with some suggesting improvements in patient outcomes such as reduced ventilator dependency and shortened intensive care unit stays when high doses of vitamin C are administered. These findings highlight the therapeutic potential of vitamin C in managing severe infections and improving patient prognosis [6]. ...
Article
Full-text available
Vitamin C, a key nutrient with potent antioxidant and immunomodulatory properties, has been explored for its therapeutic potential in treating severe infections, particularly sepsis. This systematic review aims to evaluate the effectiveness of parenteral vitamin C in improving clinical outcomes in patients with severe infections. A comprehensive search of several databases, including PubMed, EMBASE, and the Cochrane Library, was conducted for studies published between January 2000 and June 2024. Included were randomized controlled trials, observational studies, and case reports that examined the use of parenteral vitamin C in adult patients with severe infections. Data extracted included study design, sample size, intervention specifics, and clinical outcomes. Quality was assessed using tools appropriate to each study design, such as the Cochrane Risk of Bias Tool and the Newcastle-Ottawa Scale. The review included nine studies with diverse methodologies. While individual studies reported benefits such as improved immune function and reduced oxidative stress, larger systematic reviews and meta-analyses did not demonstrate a significant reduction in mortality. The results indicate that while parenteral vitamin C may improve certain biochemical and physiological parameters, these improvements do not consistently translate into enhanced survival or substantial clinical benefits. Parenteral vitamin C shows potential in modulating immune response and reducing oxidative damage in severe infections. However, its impact on key clinical outcomes like mortality and long-term recovery remains uncertain. This review highlights the need for further high-quality, randomized controlled trials to clarify vitamin C's role in managing severe infections and define optimal therapeutic protocols.
Article
Full-text available
Community-acquired pneumonia (CAP) poses a significant global health challenge, prompting exploration of innovative treatments. This systematic review and meta-analysis aimed to evaluate the efficacy and safety of vitamin C supplementation in adults undergoing treatment for CAP. A comprehensive search of the MEDLINE, Embase, CINAHL, the Cochrane Central Register of Controlled Trials, and Clinical Trials.gov databases from inception to 17 November 2023 identified six randomized-controlled-trials (RCTs) meeting inclusion criteria. The primary outcome analysis revealed a non-significant trend towards reduced overall mortality in the vitamin C group compared to controls (RR 0.51; 95% CI 0.24 to 1.09; p = 0.052; I² = 0; p = 0.65). Sensitivity analysis, excluding corona-virus-disease 2019 (COVID-19) studies and considering the route of vitamin C administration, confirmed this trend. Secondary outcomes, including hospital length-of-stay (LOS), intensive-care-unit (ICU) LOS, and mechanical ventilation, exhibited mixed results. Notably, heterogeneity and publication bias were observed in hospital LOS analysis, necessitating cautious interpretation. Adverse effects were minimal, with isolated incidents of nausea, vomiting, hypotension, and tachycardia reported. This meta-analysis suggests potential benefits of vitamin C supplementation in CAP treatment. However, inconclusive findings and methodological limitations warrants cautious interpretation, emphasising the urgency for high-quality trials to elucidate the true impact of vitamin C supplementation in CAP management.
Article
Full-text available
Background: A recent landmark randomized controlled trial (RCT) in septic patients demonstrated an increased risk of death and persistent organ dysfunction with intravenous Vitamin C (IVVC) monotherapy, which represents a disparate result from previous systematic reviews and meta-analyses (SRMA). We performed an updated SRMA of IVVC monotherapy to summarize and explore heterogeneity across current trials and conduct trial sequential analysis (TSA) to guard against type-I or type-II statistical errors. Methods: RCTs evaluating IVVC in adult critically ill patients were included. Four databases were searched from inception to 22 June 2022 without language restrictions. The primary outcome was overall mortality. Random effect meta-analysis was performed to estimate the pooled risk ratio. TSA for mortality was performed using the DerSimonian-Laird random effect model, alpha 5%, beta 10%, and relative risk reduction (RRR) of 30%, 25%, and 20%. Results: We included 16 RCTs (n = 2130). IVVC monotherapy is associated with significant reduction in overall mortality [risk ratio (RR) 0.73, 95% confidence interval (CI) 0.60-0.89; p = 0.002; I2 = 42%]. This finding is supported by TSA using RRR of 30% and 25%, and sensitivity analysis using fixed-effect meta-analysis. However, the certainty of our mortality finding was rated low using GRADE due to the serious risk of bias and inconsistency. In a priori subgroup analyses, we found no differences between single vs multicenter, higher (≥ 10,000 mg/day) vs lower dose and sepsis vs non-sepsis trials. Post-hoc, we found no differences in subgroup analysis of earlier (< 24 h) vs delayed treatment, longer (> 4 days) vs shorter treatment duration, and low vs other risk of bias studies. IVVC may have the greatest benefit in trials that enrolled patients above (i.e., > 37.5%; RR 0.65, 95% CI 0.54-0.79) vs below (i.e., ≤ 37.5%; RR 0.89, 95% CI 0.68-1.16) median control group mortality (test for subgroup differences: p = 0.06), and TSA supported this. Conclusions: IVVC monotherapy may be associated with mortality benefits in critically ill patients, particularly in patients with a high risk of dying. Given the low certainty of evidence, this potentially life-saving therapy warrants further studies to identify the optimal timing, dosage, treatment duration, and patient population that will benefit most from IVVC monotherapy. PROSPERO Registration ID: CRD42022323880. Registered 7th May 2022.
Article
Full-text available
Background: The COVID-19 pandemic represents one of the world's most important challenges for global public healthcare. Various studies have found an association between severe vitamin D deficiency and COVID-19-related outcomes. Vitamin D plays a crucial role in immune function and inflammation. Recent data have suggested a protective role of vitamin D in COVID-19-related health outcomes. The purpose of this meta-analysis and trial sequential analysis (TSA) was to better explain the strength of the association between the protective role of vitamin D supplementation and the risk of mortality and admission to intensive care units (ICUs) in patients with COVID-19. Methods: We searched four databases on 20 September 2022. Two reviewers screened the randomized clinical trials (RCTs) and assessed the risk of bias, independently and in duplicate. The pre-specified outcomes of interest were mortality and ICU admission. Results: We identified 78 bibliographic citations. After the reviewers' screening, only five RCTs were found to be suitable for our analysis. We performed meta-analyses and then TSAs. Vitamin D administration results in a decreased risk of death and ICU admission (standardized mean difference (95% CI): 0.49 (0.34-0.72) and 0.28 (0.20-0.39), respectively). The TSA of the protective role of vitamin D and ICU admission showed that, since the pooling of the studies reached a definite sample size, the positive association is conclusive. The TSA of the protective role of vitamin D in mortality risk showed that the z-curve was inside the alpha boundaries, indicating that the positive results need further studies. Discussion: The results of the meta-analyses and respective TSAs suggest a definitive association between the protective role of vitamin D and ICU hospitalization.
Article
Full-text available
Since the outbreak of the coronavirus disease 2019 (COVID-19) pandemic, caused by the severe respiratory syndrome coronavirus 2 (SARS-CoV-2), millions of people have died, and the medical system has faced significant difficulties. Our purpose was to perform a meta-analysis to estimate the effect of vitamin C on in-hospital mortality and the ICU or hospital length of stay for patients diagnosed with COVID-19. We conducted a systematic review with meta-analysis in the following databases: PubMed, Web of Science, Scopus and Cochrane Central Register of Controlled Trials. We included studies that evaluated the effect of vitamin C supplementation, compared with standard treatment in COVID-19 patients who are ≥18 y of age. Nineteen trials were included in the meta-analysis. In-hospital mortality with and without vitamin C supplementation was 24.1% vs. 33.9%(OR = 0.59; 95%CI: 0.37 to 0.95; p = 0.03), respectively. Sub-analysis showed that, in random- ized clinical trials, in-hospital mortality varied and amounted to 23.9% vs. 35.8% (OR = 0.44; 95%CI: 0.25 to 0.76; p = 0.003), respectively. In the non-randomized trials, in-hospital mortality was 24.2% vs. 33.5%(OR = 0.72; 95%CI: 0.38 to 1.39; p = 0.33), respectively. The ICU length of stay was longer in patients treated with vitamin C vs. standard therapy, 11.1 (7.3) vs. 8.3 (4.7) days (MD = 1.91; 95%CI: 0.89 to 2.93; p < 0.001), respectively. Acute kidney injury in patients treated with and without vitamin C varied and amounted to 27.8% vs. 45.0% (OR = 0.56; 95%CI: 0.40 to 0.78; p < 0.001), re- spectively. There were no differences in the frequency of other adverse events among patients’ treat- ment with and without vitamin C (all p > 0.05). The use of vitamin C reduces hospital mortality. The length of stay in the ICU is longer among patients treated with vitamin C. In terms of patient safety, vitamin C has an acceptable profile. Low doses of vitamin C are effective and safe. Despite some evidence of the usefulness of vitamin C in modifying the course of COVID-19, it is too early to modify guidelines and recommendations. Further studies, in particular randomized clinical trials, are necessary.
Article
Full-text available
Beginning in December 2019, the world faced a critical new public health stressor with the emergence of SARS-CoV-2. Its spread was extraordinarily rapid, and in a matter of weeks countries across the world were affected, notably in their ability to manage health care needs. While many sectors of public structures were impacted by the pandemic, it particularly highlighted shortcomings in medical care infrastructures around the world that underscored the need to reorganize medical systems, as they were vastly unprepared and ill-equipped to manage a pandemic and simultaneously provide general and specialized medical care. This paper presents modalities in approaches to the pandemic by various countries, and the triaged reorganization of medical sections not considered first-line in the pandemic that was in many cases transformed into wards for treating COVID-19 cases. As new viruses and structural variants emerge, it is important to find solutions to streamline medical care in hospitals, which includes the expansion of digital network medicine (i.e., telemedicine and mobile health apps) for patients to continue to receive appropriate care without risking exposure to contagions. Mobile health app development continues to evolve with specialized diagnostics capabilities via external attachments that can provide rapid information sharing between patients and care providers while eliminating the need for office visits. Telemedicine, still in the early stages of adoption, especially in the developing world, can ensure access to medical information and contact with care providers, with the potential to release emergency rooms from excessive cases, and offer multidisciplinary access for patients and care providers that can also be a means to avoid contact during a pandemic. As this pandemic illustrated, an overhaul to streamline health care is essential, and a move towards greater use of mobile health and telemedicine will greatly benefit public health to control the spread of new variants and future outbreaks.
Article
Full-text available
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still spreading worldwide. For this reason, new treatment methods are constantly being researched. Consequently, new and already-known preparations are being investigated to potentially reduce the severe course of coronavirus disease 2019 (COVID-19). SARS-CoV-2 infection induces the production of pro-inflammatory cytokines and acute serum biomarkers in the host organism. In addition to antiviral drugs, there are other substances being used in the treatment of COVID-19, e.g., those with antioxidant properties, such as vitamin C (VC). Exciting aspects of the use of VC in antiviral therapy are its antioxidant and pro-oxidative abilities. In this review, we summarized both the positive effects of using VC in treating infections caused by SARS-CoV-2 in the light of the available research. We have tried to answer the question as to whether the use of high doses of VC brings the expected benefits in the treatment of COVID-19 and whether such treatment is the correct therapeutic choice. Each case requires individual assessment to determine whether the positives outweigh the negatives, especially in the light of populational studies concerning the genetic differentiation of genes encoding the solute carriers responsible forVC adsorption. Few data are available on the influence of VC on the course of SARS-CoV-2 infection. Deducing from already-published data, high-dose intravenous vitamin C (HDIVC) does not significantly lower the mortality or length of hospitalization. However, some data prove, among other things, its impact on the serum levels of inflammatory markers. Finally, the non-positive effect of VC administration is mainly neutral, but the negative effect is that it can result in urinary stones or nephropathies.
Article
Full-text available
Almost two years have passed since the outbreak reported for the first time in Wuhan of coronavirus disease 2019 (COVID-19), due to severe acute respiratory syndrome (SARS)-CoV-2 coronavirus, rapidly evolved into a pandemic. This infectious disease has stressed global health care systems. The mortality rate is higher, particularly in elderly population and in patients with comorbidities such as hypertension, diabetes mellitus, cardiovascular disease, chronic lung disease, chronic renal disease, and malignancy. Among them, subjects with diabetes have a high risk of developing severe form of COVID-19 and show increased mortality. How diabetes contributes to COVID-19 severity remains unclear. It has been hypothesized that it may be correlated with the effects of hyperglycemia on systemic inflammatory responses and immune system dysfunction. Vitamin D (VD) is a modulator of immune-response. Data from literature showed that vitamin D deficiency in COVID-19 patients increases COVID-19 severity, likely because of its negative impact on immune and inflammatory responses. Therefore, the use of vitamin D might play a role in some aspects of the infection, particularly the inflammatory state and the immune system function of patients. Moreover, a piece of evidence highlighted a link among vitamin D deficiency, obesity and diabetes, all factors associated with COVID-19 severity. Given this background, we performed an overview of the systematic reviews to assess the association between vitamin D supplementation and inflammatory markers in patients with diabetes; furthermore, vitamin D’s possible role in COVID-19 patients was assessed as well. Three databases, namely MEDLINE, PubMed Central and the Cochrane Library of Systematic Reviews, were reviewed to retrieve the pertinent data. The aim of this review is to provide insight into the recent advances about the molecular basis of the relationship between vitamin D, immune response, inflammation, diabetes and COVID-19.
Article
Purpose of review: COVID-19 pandemic has caused more than 6.6 million deaths globally. Tremendous efforts have been committed for the development of new and repurposed drugs for the treatment of COVID-19. Although different international and national guidelines share consensus in the management of COVID-19 disease with different levels of severity, new challenges have emerged, steering the need for ongoing research in advancing the clinical management of COVID-19. Recent findings: This review focuses on recent data from randomized trials and postmarketing real-world evidence for the treatment of mild to moderate disease in the outpatient setting and patients hospitalized for COVID-19 with varying level of severity. Relevant data for treatment of the latest omicron sub-variants in people who received vaccination are presented. Challenges in special populations, including immunocompromised hosts, patients with renal failure and pregnant women, are also discussed. Summary: Treatment of COVID-19 should be personalized according to host characteristics, degree of severity and available treatment options.
Chapter
Ascorbic acid, as one of the basic exogenous vitamins, is known for its tremendous antioxidant properties. This review has been prepared to show not only the importance of ascorbic acid as a free radical scavenger, but also to summarize its antioxidant action based on other mechanisms, including activation of intracellular antioxidant systems. Ascorbic acid interacts with small molecule antioxidants, including tocopherol, glutathione and thioredoxin, but also can stimulate the biosynthesis and activation of antioxidant enzymes, such as superoxide dismutase, catalase or glutathione peroxidase. Moreover, ascorbic acid promotes the activity of several transcription factors (Nrf2, Ref-1, AP-1), which enables the expression of genes encoding antioxidant proteins. Additionally, it supports the action of other exogenous antioxidants, mainly polyphenols. In this connection, both DNA, protein and lipids are protected against oxidation. Although ascorbic acid has strong antioxidant properties, it can also have pro-oxidant effects in the presence of free transition metals. However, its role in prevention of DNA mutation and cellular apoptosis, especially in relation to cancer cells is controversial.
Article
Background: Zinc supplementation is frequently prescribed during the treatment of COVID-19. However, the evidence supporting the efficacy of this intervention is mixed. Objective: Establish the clinical utility of zinc supplementation to alter disease severity in COVID-19 illness. Methods: We performed a multicenter, retrospective, observational chart review of patients admitted to Ascension St. John Hospital or Detroit Medical Center from January 1st, 2020 to May 31st, 2020. All included patients received concomitant hydroxychloroquine due to its zinc ionophore activity. Our primary outcome was a change in Sequential Organ Failure Assessment (SOFA) score with secondary outcomes including all-cause mortality, need for intubation, and QTc prolongation as a safety outcome. Results: We identified 489 patients who received zinc and 587 patients who did not. The primary outcome showed a small difference in the change in SOFA score in patients receiving zinc in univariate analysis (1.08 vs. 1.43, p=0.02), but this difference was not significant after adjustment for confounding factors such as receipt of corticosteroids and ICU admission. Mortality was not different between those that received zinc compared to those that did not (32.7% vs 35.9%, p=0.268). Conclusion: Our retrospective study including 1064 patients hospitalized in Detroit demonstrated no differences in mortality or disease severity with zinc combination. Furthermore, prospective studies are needed to establish the utility of zinc in the treatment of COVID-19.