ArticlePDF Available

Treatment with metformin glycinate reduces SARS-CoV-2 viral load: An in vitro model and randomized, double-blind, Phase IIb clinical trial

Authors:

Abstract

The health crisis caused by the new coronavirus SARS-CoV-2 highlights the need to identify new treatment strategies for this viral infection. During the past year, over 400 coronavirus disease (COVID-19) treatment patents have been registered; nevertheless, the presence of new virus variants has triggered more severe disease presentations and reduced treatment effectiveness, highlighting the need for new treatment options for the COVID-19. This study evaluates the Metformin Glycinate (MG) effect on the SARS-CoV-2 in vitro and in vivo viral load. The in vitro study was conducted in a model of Vero E6 cells, while the in vivo study was an adaptive, two-armed, randomized, prospective, longitudinal, double-blind, multicentric, and phase IIb clinical trial. Our in vitro results revealed that MG effectively inhibits viral replication after 48 h of exposure to the drug, with no cytotoxic effect in doses up to 100 µM. The effect of the MG was also tested against three variants of interest (alpha, delta, and epsilon), showing increased survival rates in cells treated with MG. These results are aligned with our clinical data, which indicates that MG treatment reduces SARS-CoV2-infected patients´ viral load in just 3.3 days and supplementary oxygen requirements compared with the control group. We expect our results can guide efforts to position MG as a therapeutic option for COVID-19 patients.
Biomedicine & Pharmacotherapy 152 (2022) 113223
Available online 2 June 2022
0753-3322/© 2022 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/).
Treatment with metformin glycinate reduces SARS-CoV-2 viral load: An in
vitro model and randomized, double-blind, Phase IIb clinical trial
Claudia Ventura-L´
opez
a
,
1
, Karla Cervantes-Luevano
a
,
1
, Janet S. Aguirre-S´
anchez
b
,
Juan C. Flores-Caballero
b
, Carolina Alvarez-Delgado
a
, Johanna Bernaldez-Sarabia
a
,
Noemí S´
anchez-Campos
a
, Laura A. Lugo-S´
anchez
c
, Ileana C. Rodríguez-V´
azquez
c
,
Jose G. Sander-Padilla
c
, Yulia Romero-Antonio
c
, María M. Arguedas-Nú˜
nez
c
, Jorge Gonz´
alez-
Canudas
c
, Alexei F. Licea-Navarro
a
,
*
a
Departamento de Innovaci´
on Biom´
edica, CICESE, Carretera Ensenada-Tijuana 3918, Zona Playitas, Ensenada, BC 22860, Mexico
b
The American British Cowdray Medical Center I.A.P. (Centro M´
edico ABC), Mexico
c
Laboratorio Silanes S.A. de C.V., CdMx, Mexico
ARTICLE INFO
Keywords:
SARS-CoV-2 viral load
COVID-19 treatment
Metformin glycinate
SARS-CoV-2 variants
ABSTRACT
The health crisis caused by the new coronavirus SARS-CoV-2 highlights the need to identify new treatment
strategies for this viral infection. During the past year, over 400 coronavirus disease (COVID-19) treatment
patents have been registered; nevertheless, the presence of new virus variants has triggered more severe disease
presentations and reduced treatment effectiveness, highlighting the need for new treatment options for the
COVID-19. This study evaluates the Metformin Glycinate (MG) effect on the SARS-CoV-2 in vitro and in vivo viral
load. The in vitro study was conducted in a model of Vero E6 cells, while the in vivo study was an adaptive, two-
armed, randomized, prospective, longitudinal, double-blind, multicentric, and phase IIb clinical trial. Our in vitro
results revealed that MG effectively inhibits viral replication after 48 h of exposure to the drug, with no cytotoxic
effect in doses up to 100 µM. The effect of the MG was also tested against three variants of interest (alpha, delta,
and epsilon), showing increased survival rates in cells treated with MG. These results are aligned with our clinical
data, which indicates that MG treatment reduces SARS-CoV2-infected patients´viral load in just 3.3 days and
supplementary oxygen requirements compared with the control group. We expect our results can guide efforts to
position MG as a therapeutic option for COVID-19 patients.
1. Introduction
A novel coronavirus was recently discovered and termed SARS-CoV-
2. Human infection can cause coronavirus disease 2019 (COVID-19), for
which, at this point, over 510 million cases have resulted in more than 6
million deaths in over 40 countries; in Mexico, there are over 5 million
cases with more than 300,000 deaths [1]. SARS-CoV-2 infects different
organs and causes a systemic disease; as clearance of the virus occurs,
the symptoms tend to worsen, implicating an aberrant immune response
as pathogenesis of infection [2]. COVID-19 has presented one of the
most signicant challenges to humanity concerning the development of
diagnostic systems, vaccines, and new treatments. A detailed search in
the Orbit database indicated that between January 2020 to August 2021,
439 relevant patents and applications were registered relating to the
treatment of the infection caused by the coronavirus SARS-CoV-2,
including compounds of traditional Chinese medicine, as well as a
wide variety of treatments comprising placenta-derived natural killer
cells, ILC3 cells derived from a population of hematopoietic stem or
* Corresponding author.
E-mail addresses: cventura@cicese.mx (C. Ventura-L´
opez), kcervates@cicese.mx (K. Cervantes-Luevano), janetaguirre@yahoo.com (J.S. Aguirre-S´
anchez),
juancarlos18@hotmail.com (J.C. Flores-Caballero), alvarezc@cicese.mx (C. Alvarez-Delgado), jbernald@cicese.mx (J. Bernaldez-Sarabia), lsanchez@cicese.mx
(N. S´
anchez-Campos), llugo@silanes.com.mx (L.A. Lugo-S´
anchez), icrodriguez@silanes.com.mx (I.C. Rodríguez-V´
azquez), jgsander@silanes.com.mx (J.G. Sander-
Padilla), yromero@silanes.com.mx (Y. Romero-Antonio), marguedas@silanes.com.mx (M.M. Arguedas-Nú˜
nez), jogonzalez@silanes.com.mx (J. Gonz´
alez-
Canudas), alicea@cicese.mx (A.F. Licea-Navarro).
1
These authors contribute equally to this work.
Contents lists available at ScienceDirect
Biomedicine & Pharmacotherapy
journal homepage: www.elsevier.com/locate/biopha
https://doi.org/10.1016/j.biopha.2022.113223
Received 20 March 2022; Received in revised form 25 May 2022; Accepted 30 May 2022
Biomedicine & Pharmacotherapy 152 (2022) 113223
2
progenitor cells, antibodies, an alpaca nanobody, modied viruses,
bacteria (Pseudomonas), recombinant plasmids, oligomannuronic acid
phosphate, chitosan, ketoamide-based compounds, extracellular poly-
saccharide metabolite of yeast, "halogen or halogenated" compounds for
inhibiting ion channel activity of SARS-CoV-2E protein, thio-
imidazolidinone to dysregulate Angiotensin-converting enzyme (ACE)
and TMPRSS2 (Spike) proteins, GP73 inhibitor to reduce the enrichment
of ACE2 in the cell membrane preventing the virus infection, xanthine
oxidase inhibitor and an active oxygen scavenger that modies disulde
bonds in viral receptors, altering the afnity between the viral receptor
and the spike protein (S), inhibiting the virus proliferation, membrane
fusion inhibitor polypeptide, viral replication inhibitors, pinocytosis
inhibitors of the virus in host cells, activity inhibitor of virus protease
3CL pro and viral RNA-dependent RNA polymerase (RdRp) inhibitor.
Treatments involving drug repositioning also stand out, such as
Remdesivir, Ivermectin, and their respective modications. However,
the presence of mutations or substitutions in amino acids of the S protein
give place to new virus variants with implications for public health such,
as an increase in transmissibility, more severe disease presentations, and
reduced effectiveness of treatments or vaccines; highlighting the need to
identify new treatment strategies for these viral infections.
Metformin (Met) is a biguanide widely used to treat type 2 diabetes
[3], a disease that increases the risk of mortality in patients with
COVID-19 infection. Recently, metformin glycinate (MG), a new drug
derived from Met, has been shown to have better bioavailability, ab-
sorption, and safety prole than Met, with comparable anti-
hyperglycemic effects [4,5]. Met and MG have different mechanisms of
action: Met has been shown to selectively inhibit mitochondrial complex
I (NADH dehydrogenase), leading to less oxygen consumption rate,
lower ATP levels, and the activation of AMP-activated protein kinase
(AMPK) [6].
On the other hand, four lines of work characterize the mechanism of
action of MG. They are a function of different proteins: Goodpasture
antigen-binding Protein (GPBP), Liver Kinase B1 (LKB1), AMPK, and Akt
substrate of 160 kDa (AS160), which are essential in regulating the ac-
tivity of the ceramide transporter (CERT), energy, and glucose meta-
bolism [6,7]. So far, based on the evidence [814] it has been
determined that: it inhibits kinase activity GPBP/CERT activity.
GPBP/CERT and LKB1 synergistically enhance their kinase activity,
AMPK increases GPBP/CERT activity, increases the activity of LKB1
(metformin does not), and inhibits the cross-activation of GPBP/CERT
and LKB1. Provides a different prole modulation of the immune
response, especially with the migration of M1 to M2 by inhibiting the
synthesis of Interleukin 10 (IL10), translocate the Glucose transporter
(GLUT4) more efciently, consequently acts via the VAPA-VAMP2
interaction, and participates in the regulation of AS160. Metformin
glycinate is the only commercially available inhibitor of kinase activity
CERT with safety and efcacy studies.
In this study, we established a human cell culture model for infection
of lung cells H1299 with SARS-CoV-2 clinically isolated. Employing this
system, we determined the SARS-CoV-2 viral load at different times after
infection. In the context of our drug repositioning hypothesis, we tested
the capacity of MG to inhibit infection by SARS-CoV-2 in an in vitro
model of Vero E6 cells. Furthermore, we studied the efcacy and safety
of MG for the treatment of hospitalized patients with acute severe res-
piratory syndrome secondary to SARS-CoV-2, in a randomized, double-
blind, phase IIb clinical trial. The fact that MG reduces the protein
secretion pathway led us to hypothesize it could also inhibit the secre-
tion of viral particles from infected cells and thus be a candidate for drug
repositioning against SARS-CoV-2.
2. Materials and methods
2.1. Viral isolation and cell culture
Cell line H1299 (carcinoma; non-small cell lung cancer) and Vero E6
cell line were obtained from ATCC. Cells were maintained in a Dulbec-
cos Modied Eagles Medium (DMEM) medium (Corning) containing 10
% of fetal bovine serum (FBS) (Biowest), and 1 % antibiotic/antimycotic
(Gibco, 10,000 units/mL of penicillin, 10,000 µg/mL of streptomycin,
and 25 µg/mL of Fungizone).
Nasopharyngeal swabs were obtained from patients and identied as
positive for SARS-CoV2 infection after RNA extraction with QIAamp
Viral RNA Mini Kit (Qiagen) and positive amplication of RNA-
dependent RNA polymerase (RdRP) gene by qPCR using qPCRBIO
probe 1 step Go No-ROX (PCR biosystems). For viral isolation, the Vero
E6 cell line was used at conuency in a T25 cm
2
ask in a modied
protocol [15]. Briey: complete media was removed, and the monolayer
was washed twice with phosphate buffer solution (PBS), trypsinized and
counted; for infection, a total of 3 ×10
6
cells were seeded in DMEM 2 %
FBS +1 % antibiotic/antimycotic (infection media) for each nasopha-
ryngeal swab, a control of mocked cell was seeded in parallel. After 24 h,
cells reached 80 % conuence and the monolayer was infected with 50
µL of the nasopharyngeal swab in 800 µL of infection media, ask were
incubated at 37 C and 5 % CO
2
and manually moved every 20 min for 2
h; after incubation supernatant was removed and 5 mL of new infection
media were added. Cultures were monitored every 24 h for cytopathic
effects (CPE). Isolated supernatant was used for sequencing and further
experiments; Tissue Culture Infectious Dose 50 % (TCID
50
) and Multi-
plicity of Infection (MOI) were calculated in Vero E6 cells [1618].
SARS-CoV2 variants were identied by whole-genome sequencing using
Illumina COVIDSeq Assay (Illumina), from viral amplications of
nasopharyngeal swabs with low Cq value; complete viral sequences
were assembled and characterized using the Illumina® DRAGEN COVID
Lineage App version 3.5.4 (Illumina) and submitted to GISAID.
2.2. In vitro effect of metformin glycinate
For viral load assays in H1299 cells, a total of 5 ×10
4
cells/well were
seeded 24 h before infection with SARS-CoV-2 MX/BC1/2020 at a MOI
of 100:1 particle per cell for 2 h in DMEM 2 %FBS +1 % antibiotic/
antimycotic (infection media). After incubation, the supernatant with
the inoculum was removed and 500 µL of new infection media was
added containing 0, 0.1, 1 or 10 µM of MG. At 24 and 48 h after the
drugs addition, the cell supernatant was collected and centrifuged for 5
min at 300 g to remove debris and stored at 80 C. Cell monolayers
were collected by scraping and resuspending in 500 µL of infection
media.
2.2.1. Dose effect of MG on SARS-CoV-2 viral load and cell survival
Both cell monolayers and supernatant were used to isolate RNA using
the QIAamp Viral RNA Mini Kit (Qiagen). The obtained RNA was used as
a template in the reverse transcription and amplication of the SARS-
CoV-2 E gene [19]. For this amplication, the qPCRBIO probe 1 step
Go No-ROX (PCR biosystems) kit was used in a nal volume of 20 µL
contained 1X reaction buffer, 0.4 µM of the forward
(5-ACAGGTACGTTAATAGTTAATAGCGT-3) and reverse
(5-ATATTGCAGCAGTACGCACACA-3) primers, 0.2 µM Probe
(5-FAM-ACACTAGCCATCCTTACTGCGCTTCG-BHQ1-3), 0.2 ×of
RTase Go, and 5 µL of RNA. Amplication conditions were,
Retro-transcription at 50 C for 15 min, denaturation at 95 C for 2 min,
followed by 45 cycles for 15 s at 95 C, 30 s at 60 C acquiring the
uorescence at this step. The RT-qPCR reactions were conducted in
duplicate for all samples using a 96-well and optical adhesive lm
(Bio-Rad, CA, USA) on a CFX96 Real-Time PCR Detection System
(Bio-Rad). The determination of the copy numbers was obtained by
extrapolation of Cq values in the linear regression curve. The standard
curve was obtained from 5 serial dilutions (dilution factor 1:10) of a
Synthetic RNA transcribed in vitro, ranging from 10
6
to 10
2
molecules
SARS-CoV-2 per µL. The synthesized RNA corresponds to a 1000 bp
sequence with the SARS-CoV-2 RdRP and E genes inserted. For the
statistical analysis, a factorial ANOVA was used to establish differences
C. Ventura-L´
opez et al.
Biomedicine & Pharmacotherapy 152 (2022) 113223
3
between drug concentration, and signicance was established at P <
0.05.
To evaluate cell death due to the active principle, toxicity controls
were set up in parallel with the infection experiments without infection.
In short, 0.1, 1.0,10 and 100 µM of MG were added to 5 ×10
4
H1299
cells/well in cell culture conditions using 10 % DMSO as a dead control,
and PBS as a negative control. The nal viability of cells after 48 h of
exposure was measured in a parallel experiment by a colorimetric
method [20,21] using the CellTiter 96® AQueous One Solution Cell
Proliferation Assay (Promega). Half-maximal inhibitory concentration
(IC50) values were tting using a dose-response curve in GraphPad
prism 9.
2.2.2. Inhibition of SARS-CoV-2 variants cytopathic effects by MG
A total of 1.5 ×10
4
Vero E6 cells/well were seeded in a 96 well plate
for 24 h in infection media as pre-conditioning. After this time, media
was removed and cells were pre-incubated with 300 µM of MG for 1 h
prior to virus infection with 0.01 pfu/cell of isolated viral strains B.1.387
(D614G); B.1.429 +B.1.427 (Epsilon); B.1.1.7 (alpha) or B.1.617.2
(delta). After 96 h incubation, cytopathic effect on cells was measured
by neutral red uptake assay and viral mediated cell death was compared
among variants. The nucleoside GS-441524 were used as a positive
control for inhibition of viral replication in the same conditions. Dif-
ferences between the media was evaluated by unpaired student t-test
with p <0.0001 with the software GraphPad prism 9.
2.3. Clinical study
An adaptive, two-armed, randomized, prospective, longitudinal,
double-blind, multicentric and phase IIb clinical trial was performed,
with the main objective of evaluating the efcacy and safety of MG
treatment vs placebo (Treatment A: Metformin Glycinate 620 mg,
administered orally, twice a day for 14 days, Treatment B: Placebo,
administered orally, twice a day for 14 days) in patients with severe
acute respiratory syndrome secondary to SARS-CoV-2 infection and
diagnosis of type 2 diabetes mellitus. The study was conducted from July
2020 to March 2021, with a follow-up period of 14 days. The evaluation
criteria were: a) Comparing the viral load between groups; b) analyzing
the viral load in the same group at days 0 (basal), 2, 5, 7, 9, and at the
end of the study; c) comparing the use of supplementary oxygen, arti-
cial mechanical ventilation, duration of hospital stay, normalization of
body temperature, oxygen saturation, and number of deaths, between
groups; d) evaluating the change in basal and nal levels of serum
creatinine, aspartate aminotransferase, and creatine kinase MB (CK-
MB); e) and prevalence of grades 3 and 4 adverse events at the beginning
and end of the study.
2.3.1. Study subjects
Twenty patients, over 18 years of age, both genders with coronavirus
infection, severe acute respiratory syndrome (SARS-CoV)-2 conrmed
by polymerase chain reaction (PCR) test 4 days before randomization,
hospitalized and with radiographic evidence of pulmonary inltrates,
were recruited. Patients with multiorgan failure, with mechanical
ventilation during the randomization and pregnant women were
excluded from the study. Patients were randomly and blindly distributed
into group A(treatment group that received 620 mg of MG, twice a
day) and group B(placebo), with 10 patients in each group. All pa-
tients signed their informed consent form. This study was conducted in
the American British Cowdray Medical Center and was accepted by the
Ethical Research Committee of the institution: with approval number
ABC-20-16. The study was registered in the Clinical Trials System (NC
T04625985) in October 2020, during the enrollment of participants,
as well as in the Clinical Trials National Registry (RNEC by its Spanish
acronym) with the number 203301410A0085 registered on June 30 of
2020.
2.3.2. Randomization and blinding
Randomization of the treatment group was performed by the prin-
cipal investigator using a randomization list generated by randomizer.
org. Allocation was done by simple random sampling, balanced by
treatment. During the enrollment and randomization, no participants
were excluded, all 20 participants were randomized and started the
study treatments. The blinding of the study was carried out by homol-
ogating the tablets and the packaging. The packages were identied by
kit number, both the patient and the principal investigator were un-
aware of the allocation of participants. Breaking of blinding of the study
was not required.
2.3.3. Statistical analysis
The efcacy of the treatment was statistically analyzed in all treated
patients(ATP), a group of all the randomized patients that received at
least one dose of MG, and that had a basal and subsequent reading.
Missing data were managed using the last observation carried forward
method. The comparison between the groups of viral load, oxygen
supplementation and days of hospital stay was carried out using Stu-
dents t-tests for independent samples. The comparison between the
groups of the laboratory variables was carried out using the non-
parametric Mann Whitney U test. Qualitative variables were analyzed
using Fishers exact-tests. All tests were performed with IBM SPSS Sta-
tistics for Windows, version 25 (IBM Corp., Armonk, N.Y., USA). Values
of p <0.05 were considered signicant.
The sample size was selected to evaluate the primary efcacy in 10
patients infected by SARS-CoV2 vs 10 patients with personalized treat-
ment for COVID-19 according to the criteria of the treating physician.
Using an adaptive model.
3. Results
3.1. In vitro effect of metformin glycinate
3.1.1. Dose effect and IC50
To test the antiviral effect, three doses of MG 0.1, 1, and 10 µM were
added to H1299 cells for 24 and 48 h. Quantication of the extracellular
viral load was performed in at least 6 independent replicates. At 24 h
after exposition to the drug, no signicant differences were observed
neither in the cell culture nor in the supernatant (data not shown). As
shown in Fig. 1, the major inhibition effect for the presence of virus in
the supernatant and cell-associated virus was observed at 48 h. MG
shows at 48 h, a 98 % decrease in the viral load in the cell-associated
virus (Fig. 1A); and a 86 % decrease of the viral load in the superna-
tant medium (Fig. 1B). The IC50 value to decrease the cell-associated
virus load was 189.8 µM of MG (Fig. 1C), and the IC50 value to
decrease the supernatant viral load was 355.4 µM of MG (Fig. 1D).
3.1.2. MG inhibits cytopathic effects mediated by SARS-CoV2 variants
After 48 h of exposure, the drug MG potently inhibits SARS-CoV-2
replication, and cytopathic effects in vitro with doses up to 100 µM
showing a survival percentage of 100 % (Fig. 2A). Furthermore, the
effect of MG was tested against three variants of concern: B.1.387
(D614G), B.1.429 +B.1.427 (Epsilon), B.1.1.7 (alpha) and B.1.617.2
(delta) showing a signicant cell survival after 96 h incubation
compared to infected cells with no MG administration (Fig. 2B).
3.2. In vivo effect of metformin glycinate
3.2.1. Demographic and basal characteristics from the clinical study
patients
The study included 20 patients (Fig. 3 shows the selection and
allocation of the participants) recruited from July 2020 to March 2021,
with a follow-up period of 14 days. The mean age was 47.48 (42.83 in
group A; 49.38 in group B), 85 % of the patients were male and the
average BMI was 28.54 kg/m
2
. Table 1 shows the basal parameters and
C. Ventura-L´
opez et al.
Biomedicine & Pharmacotherapy 152 (2022) 113223
4
Fig. 1. Metformin glycinate (MG) Inhibition effect on SARS-CoV-2 clinical isolated (MX/BC1/2020). Metformin Glycinate effect on the SARS-CoV-2
(MOI =100) viral load (RNA copies per mL) determined 48 h after infection in (A) supernatant and (B) whole cells (carcinoma; non-small cell lung cancer; Cell
line H1299). Half-maximal inhibitory concentration (IC50) of MG on cell viability in whole cells (C) and supernatant (D).
Fig. 2. Evaluation of cytotoxicity of MG in H1299 cells and inhibition of cytopathic effects in Vero E6. (A) Cell viability of H1299 cells after 48 h infection
with SARS-CoV-2 clinical isolated (MX/BC1/2020) exposed to different concentration (0.1, 1.0,10 and 100 µM) of MG. 10 % DMSO was used as a dead control, and
PBS a negative control (NT). (B) Cell viability of Vero E6 cells treated with MG (300 µM) and infected with 0.01pfu/cell of isolated viral strains B.1.387(D614G);
B.1.429 +B.1.427 (Epsilon); B.1.1.7 (alpha) or B.1.617.2 (delta).
C. Ventura-L´
opez et al.
Biomedicine & Pharmacotherapy 152 (2022) 113223
5
clinical characteristics of the patients. The comorbidities detected were:
diabetes 20.0 % (10 % per treatment group), hypertension 20.0 %
(group A, 5.0 %; group B, 15.0 %), dyslipidemia 15.0 % (group A, 5.0 %,
group B, 10.0 %). There were no important differences between groups,
except in the percentage of oxygen saturation, where group B had 93.0
and group A had 89 (the mean percentage was 92.0).
3.2.2. Comparison of the basal main biochemical and immunological
parameters between groups of patients from the clinical trial
Table 2 shows the biochemical data for both groups. Essentially,
there were no differences between the groups, except in the levels of
alkaline phosphatase, where group A had a basal level of 90.5 UI/L and
B group had 66.5 UI/L (mean from both groups was 70.5 UI/L), which
represents a statistically signicant difference (p =0.011).
3.2.3. Basal covid-related symptoms from each group of patients
Table 3 shows that both groups of patients entered the study with
similar covid-related symptoms. There were no signicant differences in
these variables, which included cough, fever, dyspnea, headache, my-
algias/arthralgias, diarrhea and a disturbance to the general condition
(loss of appetite, fatigue, and weight loss). Additionally, the frequency of
other symptoms, such as pharyngitis, tachycardia, abdominal pain,
nasal congestion, and abdominal distention, among others, were
studied. We did not nd signicant differences in the basal frequency or
percentage of these symptoms between groups.
3.2.4. Comparison of other basal biochemical and immunological
parameters between groups of treatment
In addition to analyzing the main biochemical and immunological
parameters shown in Table 2, we compared the levels of other variables
indicative of inammation, tissue damage, and severe disease. The re-
sults in Table 4 show that both groups of patients entered the study with
very similar basal levels of immunoglobulins (IgG and IgM), reactive C
protein (RCP), lactate dehydrogenase (DHL), and creatine phosphoki-
nase (CPK).
3.2.5. Comparison of biochemical and immunological parameters in the
same group at the beginning and end of the clinical study
There were statistically signicant changes in the levels of AST,
lymphocytes, neutrophils, D dimer, CRP, DHL, and IgG in the group
treated with MG in the beginning and end of the study. In the control
group, we observed signicant differences at the basal and nal state in
ALT, ferritin, CRP, and IgG (Table 5).
3.2.6. Evaluation of the need for external oxygen supplementation
Supplementary oxygen requirements were evaluated daily for each
Fig. 3. Flowchart of the selection and allocation of study participants. *No patients were excluded from the statistical analysis since the data obtained was
sufcient to be included in the nal analysis.
C. Ventura-L´
opez et al.
Biomedicine & Pharmacotherapy 152 (2022) 113223
6
patient during their hospitalization, using the system shown in Table 6.
At the end of the study, the points were added and the mean values were
compared between groups.
3.2.7. Evaluation of the main efcacy variables between groups
We analyzed three main variables indicative of treatment efcacy:
duration of hospitalization, oxygen requirement, and reduction of the
percentage of viral load. In the MG-treated group, we observed a sig-
nicant reduction in the viral load (93.2 %) from the beginning to the
end of the study. In the control group, there was only a 78.3 % decrease
in the viral load, importantly (Table 7).
Analyzing the number of days necessary to demonstrate an unde-
tectable viral load, it was found that, group A needed only 3.3 days while
group B 5.6 days (p =0.043) (Table 8), the average hospitalization was
8.8 days for group A and 9.8 days for group B, in relation to the need for
supplemental oxygen intake, it was found that group A required a lower
intake on days 2, 5 and 7 than group B (5.9 vs 10.6 points). The differ-
ences were statistically signicant for all the variables except for the
days of hospitalization (Table 7).
It was also determined that patients in group B are 6 times more
likely to have a detectable viral load after 4 days (OR =6.667)
compared to group A (Table 8).
Finally, in relation to safety, the number and type of adverse events
Table 1
Basal demographic and clinical characteristics in each group.
Variable Treatment A
n=10
Treatment B
n=10
Total n =20 p
Age (years) 42.83 (32.2;
65.0)
49.38 (45.7;
61.2)
47.48
(39.83;
63.0)
0.247
Sex
Male (n, %) 8 (40.0) 9 (45.0) 17 (85.0) 1.000
Female (n, %) 2 (10.0) 1 (5.0) 3 (15.0)
Weight (kg) 84.50 (71.5;
95.0)
85.50 (74.5;
95.0)
84.50
(73.25;
95.0)
0.684
Height (m) 1.72 (1.65;
1.79)
1.75
(1.711.77)
1.74 (1.68;
1.78)
0.579
BMI (kg/m
2
) 29.1 (22.75;
31.93)
27.72 (25.51;
30.81)
28.54
(24.83;
31.0)
0.684
Clinical
characteristics
SBP (mmHg) 113.0 (109.2;
125.0)
120.0 (118.0;
128.0)
120.0
(112.0;
125.0)
0.165
DBP (mmHg) 70 (65.0;
73.0)
70 (69.0; 78.0) 70.0 (68.0;
76.0)
0.579
CF (lpm) 79 (67.0;
84.0)
70 (66.0; 85.0) 70.0 (67.0;
84.0)
0.684
RF (rpm) 20 (18.0;
20.0)
19 (18.0; 20.0) 20.0 (18.0;
20.0)
0.393
O2 saturation (%) 89 (88.0;
95.0)
93 (90.0;
96.0)
92.0 (88.0;
96.0)
0.165
Temperature (C) 36.5 (36.0;
37.5)
36.2 (36.0;
36.7)
36.4 (36.0;
37.4)
0.529
Comorbidities
Diabetes (n, %) 2 (10) 2 (10) 4 (20) 1.000
Hypertension (n, %) 1 (5) 3 (15) 4 (20) 0.582
Dyslipidemias (n,
%)
1 (5) 2 (10) 3 (15) 1.000
Respiratory disease
(n, %)
2 (10) 0 (0) 2 (10) 0.474
Autoimmune
disease (n, %)
1 (5) 1 (5) 2 (10) 1.000
Tabaquism (n, %) 1 (5) 1 (5) 2 (10) 1.000
SBP: systolic blood pressure; DBP: diastolic blood pressure; CF: cardiac fre-
quency; RF: respiratory frequency; BMI: body mass index. Medians and inter-
quartile range were used for continuous variables, comparisons between the
groups were made through the Mann Whitney U test and for qualitative vari-
ables in frequencies and percentages the Fishers exact-tests were used.
Table 2
Basal main biochemical parameters between groups.
Variable Treatment A Treatment B Total p
Glucose (mg/dL) 151.5 (124.9;
184.3)
148.9 (135.0;
187.7)
148.9
(129.4;
177.3)
0.796
HBA1c (%) 5.95 (5.65;
6.50)
5.8 (4.40;
6.10)
5.8 (5.4; 6.1) 0.529
Total cholesterol
(mg/dL)
151.5 (143.0;
189.2)
156.5 (147.2;
173.5)
154.5
(147.0;
176.5)
0.796
HDL cholesterol
(mg/dL)
37.5 (26.7;
42.7)
37.5 (32.2;
43.0)
37.0 (29.7;
42.5)
0.529
LDL cholesterol
(mg/dL)
91.0 (75.2;
125.2)
102.5 (86.7;
105.2)
97.5 (86.7;
107.2)
0.684
Triglycerides (mg/
dL)
169.0 (138.5;
180.0)
137.0 (100.0;
207.5)
157.5
(104.7;
176.5)
0.529
ALT (U/L) 62.5 (30.0;
83.0)
44.0 (30.7;
63.0)
53.0 (30.7;
66.2)
0.436
AST (U/L) 33.8 (23.2;
92.0)
26.4 (21.8;
35.0)
27.6 (22.1;
50.6)
0.089
GGT (U/L) 89.5 (56.7;
112.7)
99.0 (36.0;
134.7)
94.5 (39.2;
129.5)
0.780
Serum creatinine
(mg/dl)
0.73 (0.61;
0.81)
0.81 (0.73;
0.93)
0.79 (0.67;
0.87)
0.156
Total bilirubin (mg/
dl)
0.29 (0.16;
0.48)
0.37 (0.28;
0.52)
0.31 (0.27;
0.50)
0.315
Albumin (g/L) 3.4 (3.8; 3.8) 3.3 (3.0; 3.4) 3.4 (3.0; 3.6) 0.089
Alkaline
phosphatase
(UI/L)
90.5 (71.2;
114.5)
66.5 (56.0;
73.2)
70.5 (64.2;
91.2)
0.011
Leukocytes (Cell/
uL)
12.6 (9.3;
15.0)
10.1 (6.6;
12.9)
10.8 (6.7;
13.6)
0.579
Erythrocytes (Cell/
uL)
4.8 (4.3; 5.2) 4.6 (4.2; 5.0) 4.8 (4.2; 5.1) 0.315
Hemoglobin (g/dl) 15.2 (13.5;
15.6)
14.3 (13.4;
15.1)
14.5 (13.5;
15.4)
0.315
Hematocrit (%) 43.4 (40.1;
44.6)
42.6 (39.9;
43.8)
42.9 (40.4;
44.2)
0.247
MCV (fL) 89.6 (84.4;
91.7)
91.6 (87.3;
92.9)
90.4 (87.3;
92.3)
0.247
MCH (pc) 31.2 (29.3;
31.7)
30.9 (29.7;
31.8)
31.0 (29.7;
31.8)
0.684
Platelets (10
9
/L) 257.0 (205.2;
339.7)
268.5 (214.5;
307.5)
266.0
(211.7;
307.5)
1.000
Lymphocytes (%) 8.1 (4.0; 11.8) 8.2 (6.7; 11.6) 8.2 (6.0;
11.6)
0.971
Neutrophiles (%) 85.2 (83.4;
89.3)
87.2 (82.5;
89.2)
86.1 (83.2;
89.2)
0.529
Monocytes (%) 4.0 (3.4; 5.7) 3.8 (2.7; 4.4) 3.9 (3.3; 5.0) 0.280
HBA1c: glycated hemoglobin; ALT: alanine aminotransferase; AST: aspartate
aminotransferase; GGT: gamma-glutamyl transferase; MCV: mean corpuscular
volume; MCH: mean corpuscular hemoglobin. Medians and interquartile range
were used for continuous variables, the Mann Whitney U test was for compari-
sons between treatment groups, qualitative variables frequencies and percent-
ages were used, comparisons were made using the Fishers exact-tests.
Table 3
Covid-related variables.
Variable Treatment
A
Treatment
B
Total p
n=10 n =10 n =20
Cough (n, %) 8 (40) 7 (35) 15 (75) 1.000
Fever (n, %) 9 (45) 6 (30) 15 (75) 0.303
Dyspnea (n, %) 4 (20) 7 (35) 11 (55) 0.370
Headache (n, %) 7 (35) 3 (15) 10 (50) 0.179
Myalgias/arthralgias (n, %) 6 (30) 6 (30) 12 (60) 1.000
Diarrhea (n, %) 4 (20) 2 (10) 6 (30) 0.628
General condition disturbances
(n, %)
5 (25) 7 (35) 12 (60) 0.650
%, percentage. Data presented as frequencies and percentages. Differences be-
tween groups were analyzed with the Fishers exact-tests.
C. Ventura-L´
opez et al.
Biomedicine & Pharmacotherapy 152 (2022) 113223
7
Table 4
Other biochemical and immunological parameters at the beginning of the trial
(basal).
Variable Treatment A Treatment B Total p
n=10 n =10 n =20
D dimer (ng/mL) 533.0 (233.5;
703.5)
480.0 (233.5;
703.5)
511.5 (322.5;
756.5)
0.739
Ferritin (ng/mL) 1335.0 (298.5;
2102.0)
1473.5 (991.2;
2003.0)
1473.5
(754.5;
1987.5)
0.853
Procalcitonin
(ng/mL)
0.04 (0.02;
0.13)
0.08 (0.04;
0.22)
0.06 (0.04;
0.17)
0.481
RCP (mg/L) 1.83 (0.47;
2.30)
4.16 (1.47;
11.13)
2.28 (0.74;
7.58)
0.063
DHL (UI/L) 326.5 (255.2;
464.0)
257.5 (237.7;
334.7)
305.5 (241.7;
366.0)
0.105
Interleukin 6
(pg/mL)
13.7 (2.20;
67.35)
8.9 (2.07;
30.62)
10.1 (2.17;
47.0)
0.631
Vitamin D (ng/
mL)
19.1 (15.2;
22.8)
23.2 (19.3;
28.0)
20.1 (18.4;
24.3)
0.043
IgG (mg/dl) 1012.0 (753.0;
1106.7)
969.0 (819.0;
1151.7)
1012.0
(819.0;
1116.5)
0.912
IgM (mg/dl) 91.7 (66.4;
119.0)
82.5 (69.0;
115.7)
91.1 (69.1;
115.1)
0.631
CPK (U/L) 41.0 (28.5;
72.7)
35.5 (25.7;
122.7)
39.0 (28.8;
80.5)
0.684
ng/mL, nanograms per milliliter; mg/L, milligrams per liter; IU/L, international
units per liter; CRP, C-reactive protein; IgG, immunoglobulin G; IgM, immuno-
globulin M; CPK, creatinine phostokinase. Medians and interquartile range were
used for continuous variables and comparisons between treatment groups were
made with the Mann Whitney U test.
Table 5
Biochemical and immunological parameters at the beginning and end of the trial.
Variable Treatment A Treatment B
Basal Final p +Basal Final p +p*
Glucose (mg/dL) 151.5 (124.9; 184.3) 110.3 (78.3;136.2) .173 148.9 (135.0; 187.7) 139.0 (126.8; 173.4) .345 .093
Total cholesterol (mg/dL) 151.5 (143.0; 189.2) 160.0 (132.2; 174.5) .400 156.5 (147.2; 173.5) 185.5 (164.0;203.0) .069 .142
HDL cholesterol (mg/dL) 37.5 (26.7; 42.7) 35.5 (21.0; 42.7) .917 37.5 (32.2; 43.0) 51.5 (36.0; 54.2) .050 .081
LDL cholesterol (mg/dL) 91.0 (75.2; 125.2) 92.5 (74.7; 114.7) .599 102.5 (86.7; 105.2) 122.5 (46.9; 138.7) .484 .414
Triglycerides (mg/dL) 169.0 (138.5; 180.0) 147.5 (113.2; 246.0) .599 137.0 (100.0; 207.5) 114.0 (99.2; 251.25) .944 .573
ALT (U/L) 62.5 (30.0; 83.0) 85.5 (35.0; 165.2) .075 44.0 (30.7; 63.0) 92.0 (78.0;110.0) .017 .662
AST (U/L) 33.8 (23.2; 92.0) 31.5 (22.1; 49.7) .028 26.4 (21.8; 35.0) 22.2 (17.9; 29.8) .889 .491
GGT (U/L) 89.5 (56.7; 112.7) 84 (33.7; 148.5) 1.000 99.0 (36.0; 134.7) 128.5 (102.7; 175.0) .262 .142
Serum creatinine (mg/dl) 0.73 (0.61; 0.81) 0.74 (0.60; 0.84) .600 0.81 (0.73; 0.93) 0.84 (0.72;0.92) .575 .142
Total bilirubin (mg/dl) 0.29 (0.16; 0.48) 0.35 (0.23; 0.58) .528 0.37 (0.28; 0.52) 0.47 (0.36; 0.73) .069 .228
Albumin (g/L) 3.4 (3.8; 3.8) 3.15 (3.07; 3.48) .345 3.3 (3.0; 3.4) 3.48 (2.95; 3.77) .440 .662
Alkaline phosphatase (UI/L) 90.5 (71.2; 114.5) 69.0 (56.7; 76.5) .116 66.5 (56.0; 73.2) 62.0 (59.5; 79.5) .726 .755
Leukocytes (Cell/uL) 12.6 (9.3; 15.0) 9.4 (6.9; 13.3) .345 10.1 (6.6; 12.9) 9.9 (6.7; 13.8) .674 .852
Erythrocytes (C´
el/uL) 4.8 (4.3; 5.2) 5.0 (4.17; 7.96) .345 4.6 (4.2; 5.0) 4.6 (4.4; 4.9) .441 .282
Hemoglobin (g/dl) 15.2 (13.5; 15.6) 14.8 (12.7; 15.3) .293 14.3 (13.4; 15.1) 14.5 (13.8; 15.6) 1.000 .852
Hematocrit (%) 43.4 (40.1; 44.6) 43.7 (38.7; 44.8) .674 42.6 (39.9; 43.8) 41.3 (38.7; 44.7) .208 .491
MCV (fL) 89.6 (84.4; 91.7) 88.5 (84.8; 91.5) .345 91.6 (87.3; 92.9) 89.4 (85.4; 92.1) .208 .573
MCH (pc) 31.2 (29.3; 31.7) 30.8 (27.2; 32.2) .917 30.9 (29.7; 31.8) 31.3 (29.8; 32.8) .671 .662
Platelets (10
9
/L) 257.0 (205.2; 339.7) 260 (190.7; 346.7) .916 268.5 (214.5; 307.5) 324.5 (228.5; 441.7) .208 .491
Lymphocytes (%) 8.1 (4.0; 11.8) 17.0 (5.7; 36.52) .028 8.2 (6.7; 11.6) 8.0 (5.7; 12.4) .263 .491
Neutrophiles (%) 85.2 (83.4; 89.3) 76.2 (42.3; 88.7) .046 87.2 (82.5; 89.2) 85.5 (80.9; 87.8) .293 .755
Monocytes (%) 4.0 (3.4; 5.7) 7.0 (4.7; 11.0) .173 3.8 (2.7; 4.4) 3.9 (3.5; 5.8) .207 .181
D Dimer (ng/mL) 480.0 (233.5; 703.5) 634.0 (171.7; 1315.0) .028 480.0 (233.5; 703.5) 356.0 (153.5; 1585.09 .237 .491
Ferritin (ng/mL) 1401.5 (360.2; 2523.0) 1289.5 (260.25; 3483.2) .600 1709.0 (1199.2; 2179.0) 1381.0 (1205.2; 1899.2) .036 .852
Procalcitonin (ng/mL) 0.08 (0.04; 0.22) 0.04 (0.02;0.21) .916 0.08 (0.04; 0.22) 0.04 (0.03;0.05) .018 .755
CRP (mg/L) 4.16 (1.47; 11.13) 0.10 (0.02;0.41) .046 4.16 (1.47; 11.13 0.09 (0.05; 1.10) .012 .662
DHL (UI/L) 257.5 (237.7; 334.7) 256.5 (223.7; 295.7) .028 257.5 (237.7; 334.7) 244.5 (211.0; 296.5) .208 .852
Interleukin 6 (pg/mL) 8.9 (2.07; 30.62) 2.10 (2.0; 26.2) .346 8.9 (2.07; 30.62) 2.4 (1.82; 23.8) .050 .662
Vitamin D (ng/mL) 23.2 (19.3; 28.0) 19.6 (18.5; 31.0) .249 23.2 (19.3; 28.0) 29.2 (22.1; 36.7) .484 .345
IgG (mg/dl) 969.0 (819.0; 1151.7) 781.0 (550.0; 873.2) .028 969.0 (819.0; 1151.7) 800.5 (643.7; 997.7) .017 .662
IgM (mg/dl) 82.5 (69.0; 115.7) 220.1 (81.0; 133.99 .116 82.5 (69.0; 115.7) 98.2 (80.9; 147.8) .327 .573
CPK (U/L) 35.5 (25.7; 122.7) 47.0 (20.7; 76.2) .833 35.5 (25.7; 122.7) 48.5 (25.7; 76.5) .674 .950
ng/mL, nanograms per milliliter; mg/L, milligrams per liter; IU/L, international units per liter; CRP, C-reactive protein; IgG, immunoglobulin G; IgM, immunoglobulin
M; CPK, creatinine phostokinase. Medians and interquartile range were used for continuous variables * Mann Whitney U test was use to observe differences between
treatment groups.
Wilcoxon test was used for comparisons of changes within groups 6 cases where no laboratories were performed were excluded from the count.
Table 6
Point system for evaluating the need for supplementary oxygen.
Need for supplementary oxygen Points
1 Not hospitalized 0
2 Hospitalized without oxygen supplementation 1
3 Hospitalized with the requirement of supplementary oxygen (not high
ux, non-invasive ventilation).
2
4 Hospitalized with the requirement of non-invasive ventilation and/or
high ux oxygen).
4
5 Hospitalized with the requirement of extracorporeal membrane
oxygenation and/or invasive mechanical ventilation.
8
6 Death 10
Table 7
Description of the main efcacy variables.
Variable Treatment A Treatment B P*
Days of hospitalization 8.8 ±6.1 9.8 ±5.4 0.352
Oxygen need (points) 5.9 ±4.6 10.6 ±6.2 0.030
LV% Reduction 93.2 ±15.4 78.3 ±62.7 0.013
Expression of results as mean and standard deviation. * Students t-tests for
independent samples were used to compare treatment groups. LV% Percentage
viral load.
C. Ventura-L´
opez et al.
Biomedicine & Pharmacotherapy 152 (2022) 113223
8
found were similar in both treatment groups, all of them of mild
severity. There were no patients with hypoglycemia.
4. Discussion
The potential role of metformin in COVID-19 disease has been
elucidated in diverse research highlighting its antioxidant, anti-
inammatory, immunomodulatory, and antiviral effects [2224]. Our
results demonstrate that MG has anti-viral action against the
SARS-CoV-2 clinical isolate in vitro, with a single dose able to control
viral replication within 48 h. SARSCoV-2 is a positive single-strain RNA
virus (+ssRNA) and depends on cellular membranes in all steps of the
viral life cycle and immunologically have similar characteristics con-
cerning host immune response with other +ssRNA viruses, which might
offer some insight into the treatment of COVID-19 [25,26].
Plus-stranded RNA viruses share the characteristic of remodeling
intracellular membranes in order to create membrane replication fac-
tories or replication organelles, which are vesicles where viral RNA
replication occurs [27,28]. These vesicles not only represent the site of
viral replication but also act as one of the strategies of viral immune
evasion mechanisms, shielding the viral RNA from cellular innate im-
mune sensors [2932]. We hypothesize that MG inhibits the replication
of SARS-CoV-2 by the inhibition of protein synthesis as has been
described for other RNA viruses. Sphingomyelin (SM) is required for the
replication of some RNA viruses as the hepatitis C virus (HCV); the
biosynthesis starts in the endoplasmic reticulum (ER) and gives rise to
ceramide, which is transported from the ER to the Golgi by the action of
ceramide transfer protein (CERT), where it can be converted to a SM. For
HCV, it has been demonstrated that inhibition of SM biosynthesis, either
by using small-molecule inhibitors or by knockout (KO) of CERT, sup-
pressed HCV replication in a genotype-independent manner. This
reduction in HCV replication was rescued by exogenous SM or ectopic
expression of the CERT protein, but not by ectopic expression of
nonfunctional CERT mutants. Observing low numbers of DMVs in stable
replicon cells treated with a SM biosynthesis inhibitor or in CERT-KO
cells transfected with either HCV replicon or with constructs that
drive HCV protein production in a replication-independent system
indicated the signicant importance of SM to DMVs. The degradation of
SM of the in vitro-isolated DMVs affected their morphology and
increased the vulnerability of HCV RNA and proteins to RNase and
protease treatment, respectively [33,34].
The SARS-CoV-2 can induce cell death after 4872 h of infection;
therefore, the cell viability is a surrogate measure of viral replication in
vitro. Pre-treatment of cells with a single-dose of 300 µM MG was able to
reduce cell death among different SARS-CoV2 variants, indicating that
MG cellular mechanism is kept despite differences in SARS-CoV2 spike.
The in vitro results are in line with our clinical study where we
demonstrated that MG reduces the viral load of Covid-19 patients to
undetectable levels in only 3.3 days. This reduction of infection is
associated with shorter hospitalization and less dependence on
supplementary oxygen. Except for a mildly elevated GGT in both groups,
in the basal and nal state, most of the biochemical parameters evalu-
ated at the beginning and end of the clinical study were normal for the
MG and control group. The only two-biochemical markers that changed
signicantly with MG treatment were AST and LDH levels. AST levels
were signicantly reduced at the end of the study in the group treated
with MG (33.831.5 U/L, basal vs nal, respectively), but both levels are
in the normal range (848 U/L). Similarly, LDH levels decreased in the
MG group from 257.5 to 256.5 IU/L, both of which are within the
normal range for this enzyme. D dimer was above the normal range in
both groups at the beginning of the study (480.0 ng/mL for both
groups), as can be expected for Covid-19 patients [35]. Interestingly, D
dimer increased signicantly only in the MG group at the end of the
treatment (480.0634.0 ng/mL), while a non-signicant decrease was
observed for the control group (480.0356.0 ng/mL).
Even though the majority of patients from both groups entered the
study with moderate disease, we did not observe the expected elevated C
reactive protein level for Covid patients (2050 mg/L) [36]. In fact, both
groups had normal CRP levels at the beginning (4.16 mg/L, both groups)
and these levels decreased signicantly in all patients at the end of the
study (0.10 and 0.09 mg/L, MG and control groups, respectively). This
could be explained by the fact that none of the patients that entered this
study presented with severe Covid symptoms. Basal fasting glucose
levels were elevated in both groups (151.5 and 148.9 mg/dL, respec-
tively). This is not surprising, as 50 % of hospitalized Covid patients
develop acute hyperglycemia, and 7 % of this population has diabetes
[37]. At the end of the study, glucose levels in the MG-treated group
were close to normal (110.3 mg/dL), as was expected from the
anti-hyperglycemic mode of action of the drug.
5. Conclusions
In the present study, we demonstrated the in vitro effect of metformin
glycinate on the viral load of SARS-CoV-2. The antiviral effect of MG was
supported by in vivo results according to biochemical and immunolog-
ical parameters; 620 mg of MG administered orally every 12 h signi-
cantly and safely decreased viral load in Covid 19 patients. Although
these results strongly suggest that MG could be repositioned for the
treatment of SARS-CoV2, further clinical studies need to be assessed to
establish the safety and efcacy of MG in large populations. The hos-
pitalization condition of the patients was one of the limitations of the
clinical study, which made the collection of complete data and follow-up
of subjects more complicated. A longer follow-up could elucidate if the
biochemical alterations found in this study (reduced AST and reduced
DHL) are clinically relevant or are the only biochemical modications.
Finally, the longer follow-up could determine if the ability to decrease
viral replication of MG would have an implication on subacute and/or
chronic complications of the COVID-19 patients.
CRediT authorship contribution statement
Claudia Ventura-Lopez: Formal analysis, Investigation, Methodol-
ogy Writing original draft. Karla Cervantes-Luevano: Formal anal-
ysis, Data curation, Methodology. Janet S. Aguirre-S´
anchez: Formal
analysis, Data curation, Methodology. Juan C. Flores-Caballero:
Formal analysis, Methodology. Carolina Alvarez-Delgado: Data cura-
tion, Writing original draft. Johanna Bernaldez-Sarabia: Supervi-
sion, Project administration. Noemí S´
anchez-Campos: Data curation.
Laura A. Lugo-S´
anchez: Methodology, Writing review & editing.
Ileana C. Rodríguez-V´
azquez: Methodology, Writing review &
editing. Jose G. Sander-Padilla: Methodology, Writing review &
editing. Yulia Romero-Antonio: Methodology, Writing review &
editing. María M. Arguedas-Nú˜
nez: Methodology, Writing review &
editing. Jorge Gonz´
alez-Canudas: Conceptualization, Resources, Su-
pervision, Writing review & editing. Alexei F. Licea-Navarro: Fund-
ing acquisition, Project administration, Supervision, Writing review &
Table 8
Comparison of negative viral load by treatment group.
Variable Treatment
A
Treatment
B
P OR (95 %IC)
n=10 n =10
Negative viral load
(days)
3.3 ±2.16 5.6 ±0.89 0.029
Negative viral load
<3.3 days (n, %)
4.0 (40.0) 0.0 (0.0) 0.043 6.667
(0.59674.490)
Negative viral load
>4.0 days (n, %)
6.0 (60.0) 10.0
(100.0)
Expression of results as mean and standard deviation. * For quantitative vari-
ables the Students t-tests for independent samples was used to compare treat-
ment groups. For qualitative variable the Fishers exact-tests was used for the
comparison between treatment groups.
C. Ventura-L´
opez et al.
Biomedicine & Pharmacotherapy 152 (2022) 113223
9
editing.
Funding
Funding for this work came partially from CICESE Grant 685-101.
Declaration of Competing Interest
The authors declare that they have no known competing nancial
interests or personal relationships that could have appeared to inuence
the work reported in this paper.
Data availability
Data will be made available on request.
Acknowledgments
We are grateful to Dr. Javier Perez-Robles, Dr. Jahaziel Gasperin and
M.A. Itandehui Betanzo Guti´
errez for support during samples
processing.
References
[1] World Health Organization, Coronavirus disease situation, 2022. https://covid19.
who.int/, (Accessed 27 April 2022).
[2] T.S. Fung, D.X. Liu, Human coronavirus: host-pathogen interaction, Annu. Rev.
Microbiol. 73 (2019) 529557, https://doi.org/10.1146/annurev-micro-020518-
115759.
[3] R.J. Ford, M.D. Fullerton, S.L. Pinkosky, E.A. Day, J.W. Scott, J.S. Oakhill, A.
L. Bujak, B.K. Smith, J.D. Crane, R.M. Blümer, K. Marcinko, B.E. Kemp, H.
C. Gerstein, G.R. Steinberg, Metformin and salicylate synergistically activate liver
AMPK, inhibit lipogenesis and improve insulin sensitivity, Biochem. J. 468 (2015)
125132, https://doi.org/10.1042/BJ20150125.
[4] National Library of Medicine (U.S.), Metformin Glycinate on Metabolic Control and
Inammatory Mediators in Type 2 Diabetes (COMET), Identier NCT01386671,
2018. Available from: https://clinicaltrials.gov/ct2/show/study/NCT01386671.
[5] J. Gonz´
alez-Canudas, Comet Group, 146-LB: efcacy and safety of metformin
glycinate vs. metformin hydrochloride in metabolic control and inammatory
mediators in Type 2 diabetes mellitus patients (T2DM), ADA (2019) 68, https://
doi.org/10.2337/db19-146-LB.
[6] P. Rada, A. Mosquera, J. Mutan´
e, F. Ferrandiz, L. Rodríguez-Ma˜
nas, F. de Pablo,
J. Gonz´
alez-Canudas, A.M. Malverde, Differential effects of metformin glycinate
and hydrochloride in glucose production, AMPK phosphorylation and insulin
sensitivity in hepatocytes from non-diabetic and diabetic mice, Food Chem.
Toxicol. 123 (2019) 470480, https://doi.org/10.1016/j.fct.2018.11.019.
[7] D.G. Hardie, F.A. Ross, S.A. Hawley, AMPK: a nutrient and energy sensor that
maintains energy homeostasis, Nat. Rev. Mol. Cell Biol. 13 (2012) 251262,
https://doi.org/10.1038/nrm3311.
[8] L.J. Foster, M.L. Weir, D.Y. Lim, Z. Liu, W.S. Trimble, A. Klip, A functional role for
VAP-33 in insulin-stimulated GLUT4 trafc, Trafc 1 (2000) 512521, https://doi.
org/10.1034/j.1600-0854.2000.010609.x.
[9] L. Florin, A. Pegel, E. Becker, A. Hausser, M.A. Olayioye, H. Kaufmann,
Heterologous expression of the lipid transfer protein CERT increases therapeutic
protein productivity of mammalian cells, J. Biotechnol. 141 (2009) 8490, https://
doi.org/10.1016/j.jbiotec.2009.02.014.
[10] A. Raya, F. Revert-Ros, P. Martinez-Martinez, S. Navarro, E. Rosello, B. Vieites,
F. Granero, J. Forteza, J. Saus, Goodpasture antigen-binding protein, the kinase
that phosphorylates the goodpasture antigen, is an alternatively spliced variant
implicated in autoimmune pathogenesis, J. Biol. Chem. 275 (2000) 4039240399,
https://doi.org/10.1074/jbc.M002769200.
[11] M. Kawano, K. Kumagai, M. Nishijima, K. Hanada, Efcient trafcking of ceramide
from the endoplasmic reticulum to the Golgi apparatus requires a VAMP-associated
protein-interacting FFAT motif of CERT, J. Biol. Chem. 281 (2006) 3027930288,
https://doi.org/10.1074/jbc.M605032200.
[12] S. Saito, H. Matsui, M. Kawano, K. Kumagai, N. Tomishige, K. Hanada, S. Echigo,
S. Tamura, T. Kobayashi, Protein phosphatase 2Cepsilon is an endoplasmic
reticulum integral membrane protein that dephosphorylates the ceramide
transport protein CERT to enhance its association with organelle membranes,
J. Biol. Chem. 283 (2008) 65846593, https://doi.org/10.1074/jbc.M707691200.
[13] D.C. Prosser, D. Tran, P.Y. Gougeon, C. Verly, J.K. Ngsee, FFAT rescues VAPA-
mediated inhibition of ER-to-Golgi transport and VAPB-mediated ER aggregation,
J. Cell Sci. 121 (2008) 30523061, https://doi.org/10.1242/jcs.028696.
[14] Y. Zhang, Y. Wang, C. Bao, Y. Xu, H. Shen, J. Chen, J. Yan, Y. Chen, Metformin
interacts with AMPK through binding to gamma subunit, Mol. Cell. Biochem. 368
(2012) 6976, https://doi.org/10.1007/s11010-012-1344-5.
[15] F.J. Díaz, W. Aguilar-Jim´
enez, L. Fl´
orez-´
Alvarez, G. Valencia, K. Laiton-Donato,
C. Franco-Mu˜
noz, D. ´
Alvarez-Díaz, M. Mercado-Reyes, M.T. Rugeles, Isolation and
characterization of an early SARS-CoV-2 isolate from the 2020 epidemic in
Medellín, Colombia. Aislamiento y caracterizaci´
on de una cepa temprana de SARS-
CoV-2 durante la epidemia de 2020 en Medellín, Colombia, Biomed.: Rev. Inst.
Nac. Salud 40 (2020) 148158, https://doi.org/10.7705/biomedica.5834.
[16] L.J. Reed, H. Muench, A simple method of estimating fty per cent endpoints, Am.
J. Epidemiol. 27 (1983) 493497, https://doi.org/10.1093/oxfordjournals.aje.
a118408.
[17] L.A. Sachs, D. Schnurr, S. Yagi, M.E. Lachowicz-Scroggins, J.H. Widdicombe,
Quantitative real-time PCR for rhinovirus, and its use in determining the
relationship between TCID50 and the number of viral particles, J. Virol. Methods
171 (2011) 212218, https://doi.org/10.1016/j.jviromet.2010.10.027.
[18] M.A. Ramakrishnan, Determination of 50 % endpoint titer using a simple formula,
World J. Virol. 5 (2016) 8586, https://doi.org/10.5501/wjv.v5.i2.85.
[19] V.M. Corman, O. Landt, M. Kaiser, R. Molenkamp, A. Meijer, D.K. Chu, T. Bleicker,
S. Brünink, J. Schneider, M.L. Schmidt, D.G. Mulders, B.L. Haagmans, B. Van der
Veer, S. Van den Brink, L. Wijsman, G. Goderski, J.L. Romette, J. Ellis, M. Zambon,
M. Peiris, H. Goossens, C. Reusken, M.P. Koopmans, C. Drosten, Detection of 2019
novel coronavirus (2019-nCoV) by real-time RT-PCR, Eur. Surveill. 25 (2020),
2000045, https://doi.org/10.2807/1560-7917.ES.2020.25.3.2000045 (Erratum
in: Euro Surveill. 2020 Apr;25(14): Erratum in: Euro Surveill. 2020 Jul;25(30):
Erratum in: Euro Surveill. 2021 Feb;26(5)).
[20] A.H. Cory, T.C. Owen, J.A. Barltrop, J.G. Cory, Use of an aqueous soluble
tetrazolium/formazan assay for cell growth assays in culture, Cancer Commun. 3
(1991) 207212, https://doi.org/10.3727/095535491820873191.
[21] T.L. Riss, R.A. Moravec, Comparison of MTT, XTT, and a novel tetrazolium
compound for MTS for in vitro proliferation and chemosensitivity assays, Mol. Biol.
Cell 3 (Suppl.) (1992) S184a.
[22] L. Ojeda-Fern´
andez, A. Foresta, G. Macaluso, P. Colacioppo, M. Tettamanti,
A. Zambon, S. Genovese, I. Fortino, O. Leoni, M.C. Roncaglioni, M. Baviera,
Metformin use is associated with a decrease in the risk of hospitalization and
mortality in COVID-19 patients with diabetes: a population-based study in
Lombardy, Diabetes Obes. Metab. 24 (5) (2022) 891898, https://doi.org/
10.1111/dom.14648.
[23] S.M. Samuel, E. Varghese, D. Büsselberg, Therapeutic potential of metformin in
COVID-19: reasoning for its protective role, Trends Microbiol. 29 (10) (2021)
894907, https://doi.org/10.1016/j.tim.2021.03.004.
[24] E. Varghese, S.M. Samuel, A. Liskova, P. Kubatka, D. Büsselberg, Diabetes and
coronavirus (SARS-CoV-2): molecular mechanism of metformin intervention and
the scientic basis of drug repurposing, PLoS Pathog. 17 (6) (2021), e1009634,
https://doi.org/10.1371/journal.ppat.1009634.
[25] N. Altan-Bonnet, Lipid tales of viral replication and transmission, Trends Cell Biol.
27 (2017) 201213, https://doi.org/10.1016/j.tcb.2016.09.011.
[26] H. Alothaid, M. Aldughaim, K. El Bakkouri, S. AlMashhadi, A.A. Al-Qahtani,
Similarities between the effect of SARS-CoV-2 and HCV on the cellular level, and
the possible role of ion channels in COVID19 progression: a review of potential
targets for diagnosis and treatment, Channels 14 (2020) 403412, https://doi.org/
10.1080/19336950.2020.1837439.
[27] D. Egger, B. Wolk, R. Gosert, L. Bianchi, H.E. Blum, D. Moradpour, K. Bienz,
Expression of hepatitis C virus proteins induces distinct membrane alterations
including a candidate viral replication complex, J. Virol. 76 (2002) 59745984,
https://doi.org/10.1128/jvi.76.12.5974-5984.2002.
[28] H. Wang, A.W. Tai, Mechanisms of cellular membrane reorganization to support
hepatitis C virus replication, Viruses 8 (2016) 142, https://doi.org/10.3390/
v8050142.
[29] M. Kikkert, Innate immune evasion by human respiratory RNA viruses, J. Innate
Immun. 12 (2020) 420, https://doi.org/10.1159/000503030.
[30] I. Romero-Brey, R. Bartenschlager, Membranous replication factories induced by
plus-strand RNA viruses, Viruses 6 (2014) 28262857, https://doi.org/10.3390/
v6072826.
[31] A. Garcia-Sastre, Ten strategies of interferon evasion by viruses, Cell Host Microbe
22 (2017) 176184, https://doi.org/10.1016/j.chom.2017.07.012.
[32] Y.T. Kao, M.M.C. Lai, C.Y. Yu, How dengue virus circumvents innate immunity,
Front. Immunol. 7 (2018) 2860, https://doi.org/10.3389/mmu.2018.02860.
[33] Y. Amako, G.H. Syed, A. Siddiqui, Protein kinase D negatively regulates hepatitis C
virus secretion through phosphorylation of oxysterol-binding protein and ceramide
transfer protein, J. Biol. Chem. 286 (2011) 1126511274, https://doi.org/
10.1074/jbc.M110.182097.
[34] H. Gewaid, H. Aoyagi, M. Arita, K. Watashi, R. Suzuki, S. Sakai, K. Kumagai,
T. Yamaji, M. Fukasawa, F. Kato, T. Hishiki, A. Mimata, Y. Sakamaki, S. Ichinose,
K. Hanada, M. Muramatsu, T. Wakita, H. Aizaki, Sphingomyelin is essential for the
structure and function of the double-membrane vesicles in hepatitis C virus RNA
replication factories, J. Virol. 94 (2020), https://doi.org/10.1128/JVI.01080-20
(e01080 20).
[35] Y. Yao, J. Cao, Q. Wang, Q. Shi, K. Liu, Z. Luo, X. Chen, S. Chen, K. Yu, Z. Hang, D-
dimer as a biomarker for disease severity and mortality in COVID-19 patients: a
case control study, J. Intensive Care 8 (2020) 49, https://doi.org/10.1186/s40560-
020-00466-z.
[36] N. Ali, Elevated level of C-reactive protein may be an early marker to predict risk
for severity of COVID-19, J. Med. Virol. 92 (2020) 24092411, https://doi.org/
10.1002/jmv.26097.
[37] A. Ceriello, Hyperglycemia and COVID-19: what was known and what is really
new? Diabetes Res. Clin. Pract. 167 (2020), 108383 https://doi.org/10.1016/j.
diabres.2020.108383.
C. Ventura-L´
opez et al.
... Some studies indicate metformin's potential in enhancing therapeutic outcomes, with its effectiveness demonstrated in bacterial and cancer cell activity [2,[10][11][12]. Metformin demonstrates pleiotropic benefits, contributing to the improvement of multiple conditions, including thyroid-related disorders and other diseases [13][14][15][16][17]. Metformin is associated with minimal side effects compared to other medications, with the most common being gastrointestinal disturbances, which are generally mild and transient [18][19][20][21][22]. 2 In the context of viral infections, metformin has shown the potential to interfere with viral replication and spread by creating an intracellular environment that is less conducive to viral survival [23,24] (Figure 1). One of the primary mechanisms underlying this antiviral effect is metformin's activation of AMPK, which shifts host cellular metabolism away from the high-energy states that many viruses exploit for their replication cycles [25,26]. ...
... Metformin's effects on the renin-angiotensin-aldosterone system (RAAS) have also been considered necessary, as SARS-CoV-2 utilizes the angioten-sin-converting enzyme 2 (ACE2] receptor for cell entry [120][121][122]. Although the exact relationship between metformin and ACE2 expression is not fully understood, it has been hypothesized that the drug might modulate ACE2 levels, potentially affecting viral entry or disease severity [24,120,123,124]. Moreover, metformin improves endothelial function, which may offer protection against vascular complications commonly observed in COVID-19, such as endothelial damage, elevated blood pressure and thrombosis [41,[125][126][127][128][129]. ...
Preprint
Full-text available
Metformin, a widely used antidiabetic medication, has emerged as a promising broad-spectrum antiviral agent due to its ability to modulate cellular pathways essential for viral replication. By activating AMPK, metformin depletes cellular energy reserves that viruses rely on, effectively limiting the replication of pathogens such as Influenza, HIV, SARS-CoV-2, HBV, and HCV. Its role in inhibiting the mTOR pathway, crucial for viral protein synthesis and reactivation, is particularly significant in managing infections caused by HIV, CMV, and EBV. Furthermore, metformin reduces oxidative stress and reactive oxygen species (ROS), which are critical for replicating arboviruses such as Zika and dengue. The drug also regulates immune responses, cellular differentiation, and inflammation, disrupting the life cycle of HPV and potentially other viruses. These diverse mechanisms suppress viral replication, enhance immune system functionality, and contribute to better clinical outcomes. This multifaceted approach highlights metformin's potential as an adjunctive therapy in treating a wide range of viral infections. Keywords: metformin; broad-spectrum antiviral; AMPK activation; mTOR inhibition; viral replication; inflammation
... One repurposed drug is metformin (N, N-dimethylbiguanide). Metformin, derived from galegine, was initially used as a first-line oral therapy for type 2 diabetes (T2D), mainly because of its ability to suppress hepatic gluconeogenesis 8 . Metformin has been reported to delay ageing and play critical roles in ageing-related diseases (such as Alzheimer's disease, cardiovascular diseases, and osteoarthritis) 9,10 , chronic immune-inflammatory diseases 11 , and even COVID-19 12 . Since it was reported in 2010 that T2D patients had a lower cancer incidence after treatment with metformin 13 , which indicates the anticancer potential of this drug, an increasing number of clinical trials of metformin have been conducted in nondiabetic patients with cancers, including breast cancer and non-small cell lung cancer (NSCLC), and have produced encouraging results 14,15 . ...
Article
Full-text available
Small cell lung cancer (SCLC) is a therapeutically challenging disease. Metformin, an effective agent for the treatment of type 2 diabetes, has been shown to have antitumour effects on many cancers, including non-small cell lung cancer (NSCLC) and breast cancer. Currently, the antitumour effects of metformin on SCLC and the underlying molecular mechanisms remain unclear. CCK-8, EdU, colony formation, flow cytometry, immunofluorescence, molecular docking, western blotting, nude mouse transplanted tumour model, and immunohistochemistry experiments were conducted to analyse gene functions and the underlying mechanism involved. In vitro experiments demonstrated that metformin inhibited the growth of SCLC cells (H446, H526, H446/DDP and H526/DDP), which was confirmed in xenograft mouse models in vivo. Additionally, metformin induced cell cycle arrest, apoptosis, and autophagy in these SCLC cells. The molecular docking results indicated that metformin has a certain binding affinity for EGFR. The western blotting results revealed that metformin decreased the expression of EGFR, p-EGFR, AKT, and p-AKT, which could be reversed by EGF and SC79. Moreover, metformin activated AMPK and inactivated mTOR, and compound C and SC79 increased the levels of p-mTOR. Metformin can not only enhance the antitumour effect of cisplatin but also alleviate the toxic effects of cisplatin on the organs of xenograft model animals. In summary, the current study revealed that metformin inhibits the growth of SCLC by inducing autophagy and apoptosis via suppression of the EGFR/AKT/AMPK/mTOR pathway. Metformin might be a promising candidate drug for combination therapy of SCLC.
... A clinical trial targeting patients with severe acute respiratory syndrome caused by SARS-CoV-2, who were also diagnosed with T2D and had elevated blood glucose levels, found that metformin glycinate (MG, 620 mg orally every 12 h) significantly and safely reduced the viral load in patients with COVID-19 while also lowering aspartate aminotransferase (AST) and lactate dehydrogenase (DHL) levels; at the end of the study, the glucose levels in the MG treatment group were close to normal values (110.3 mg/dL). 99 Another study also indicated that combined metabolic activators (CMAs), such as L -serine and N-acetyl-L-cysteine, can significantly improve these metabolic disorders and shorten the recovery time of patients with COVID-19. Phase 2 and 3 clinical trials have shown that CMA treatment significantly improves the levels of inflammation and antioxidant-related metabolic proteins and metabolites in plasma, suggesting that CMAs may accelerate patient recovery. ...
... Metformin did not prevent the hypoxemia, hospitalization or death associated with COVID-19 [73]. Contrary, treatment with metformin glycinate reduced the viral load in patients with COVID-19 [74]. Outpatient treatment with metformin reduced long COVID-19 incidence among patients with overweight or obesity [75]. ...
Article
Full-text available
Original Article, Pol J Public Health, Vol. 134 (2024): 47-51 Paulina Oleksa, Kacper Jasiński, Daria Żuraw, Mateusz Sobczyk, Monika Żybowska, Anna Rzewuska-Fijałkowska, Karolina Haczkur-Pawłowska, Piotr Więsyk Students’ Scientific Society at the Department of Epidemiology and Clinical Research Methodology, Medical University of Lublin, Poland Introduction. Metformin is an oral antidiabetic drug from the biguanide group, popularly referred as an aspirin of the 21st century. The therapeutic targets of metformin are expanding. It is characterized by antineoplastic, immunoregulatory, anti-aging and neuroprotective properties. We aimed to evaluate the pleiotropic effects of metformin, taking into account its different mechanisms, efficacy and safety in contemporary public health challenges. Material and methods. We conducted the literature review from 2014 to 2024 using the PubMed and Google Scholar. Results. Metformin, depending on the cancer and its stage, enhances the cancer treatment effects, prevents the drug resistance, lengthens overall time of survival, reduces the risk of recurrence. In the Parkinson’s disease, Alzheimer’s disease and depression metformin can even increase the risk of their occurrence, especially in high doses. Such doses predispose to the cobalamin deficiency, affecting the functioning of the nervous system. Metformin was effective in seizure control of epilepsy. It has positive impact on the course of some autoimmunological diseases. Among diabetics treatment, outcomes of COVID-19 and tuberculosis could be improved by metformin. Conclusions. Metformin is pluripotential drug. Possibilities of adjuvant metformin therapy are very promising, but it cannot be recommended as standard treatment. This issue requires further investigation, preferentially randomized controlled trials on the bigger research samples. Keywords: metformin and therapy, metformin and treatment, metformin and advances.
... There is growing evidence that metformin, a first line anti-hyperglycemic medication for the management of T2DM may be beneficial in the management of SARS-CoV-2 infection, likely due to both anti-viral and anti-inflammatory actions. [17][18][19][20][21] These properties along with reported anti-thrombotic properties may impact the development of severe COVID-19. [22][23][24] Given the shared etiology of COVID-19 and PASC and the hypothesized immunological and inflammatory pathway of PASC development, metformin may also be beneficial for PASC prevention. ...
Article
Full-text available
Background Observed activity of metformin in reducing the risk of severe COVID-19 suggests a potential use of the anti-hyperglycemic in the prevention of post-acute sequelae of SARS-CoV-2 infection (PASC). We assessed the 3-month and 6-month risk of PASC among patients with type 2 diabetes mellitus (T2DM) comparing metformin users to sulfonylureas (SU) or dipeptidyl peptidase-4 inhibitors (DPP4i) users. Methods We used de-identified patient level electronic health record data from the National Covid Cohort Collaborative (N3C) between October 2021 and April 2023. Participants were adults ≥ 18 years with T2DM who had at least one outpatient healthcare encounter in health institutions in the United States prior to COVID-19 diagnosis. The outcome of PASC was defined based on the presence of a diagnosis code for the illness or using a predicted probability based on a machine learning algorithm. We estimated the 3-month and 6-month risk of PASC and calculated crude and weighted risk ratios (RR), risk differences (RD), and differences in mean predicted probability. Results We identified 5596 (mean age: 61.1 years; SD: 12.6) and 1451 (mean age: 64.9 years; SD 12.5) eligible prevalent users of metformin and SU/DPP4i respectively. We did not find a significant difference in risk of PASC at 3 months (RR = 0.86 [0.56; 1.32], RD = −3.06 per 1000 [−12.14; 6.01]), or at 6 months (RR = 0.81 [0.55; 1.20], RD = −4.91 per 1000 [−14.75, 4.93]) comparing prevalent users of metformin to prevalent users of SU/ DPP4i. Similar observations were made for the outcome definition using the ML algorithm. Conclusion The observed estimates in our study are consistent with a reduced risk of PASC among prevalent users of metformin, however the uncertainty of our confidence intervals warrants cautious interpretations of the results. A standardized clinical definition of PASC is warranted for thorough evaluation of the effectiveness of therapies under assessment for the prevention of PASC.
Article
Full-text available
Metformin, a widely used antidiabetic medication, has emerged as a promising broad-spectrum antiviral agent due to its ability to modulate cellular pathways essential for viral replication. By activating AMPK, metformin depletes cellular energy reserves that viruses rely on, effectively limiting the replication of pathogens such as influenza, HIV, SARS-CoV-2, HBV, and HCV. Its role in inhibiting the mTOR pathway, crucial for viral protein synthesis and reactivation, is particularly significant in managing infections caused by HIV, CMV, and EBV. Furthermore, metformin reduces oxidative stress and reactive oxygen species (ROS), which are critical for replicating arboviruses such as Zika and dengue. The drug also regulates immune responses, cellular differentiation, and inflammation, disrupting the life cycle of HPV and potentially other viruses. These diverse mechanisms suppress viral replication, enhance immune system functionality, and contribute to better clinical outcomes. This multifaceted approach highlights metformin’s potential as an adjunctive therapy in treating a wide range of viral infections.
Article
OBJECTIVE Studies show metformin use before and during SARS-CoV-2 infection reduces severe COVID-19 and postacute sequelae of SARS-CoV-2 (PASC) in adults. Our objective was to describe the incidence of PASC and possible associations with prevalent metformin use in adults with type 2 diabetes mellitus (T2DM). RESEARCH DESIGN AND METHODS This is a retrospective cohort analysis using the National COVID Cohort Collaborative (N3C) and Patient-Centered Clinical Research Network (PCORnet) electronic health record (EHR) databases with an active comparator design that examined metformin-exposed individuals versus nonmetformin-exposed individuals who were taking other diabetes medications. T2DM was defined by HbA1C ≥6.5 or T2DM EHR diagnosis code. The outcome was death or PASC within 6 months, defined by EHR code or computable phenotype. RESULTS In the N3C, the hazard ratio (HR) for death or PASC with a U09.9 diagnosis code (PASC-U09.0) was 0.79 (95% CI 0.71–0.88; P < 0.001), and for death or N3C computable phenotype PASC (PASC-N3C) was 0.85 (95% CI 0.78–0.92; P < 0.001). In PCORnet, the HR for death or PASC-U09.9 was 0.87 (95% CI 0.66–1.14; P = 0.08), and for death or PCORnet computable phenotype PASC (PASC-PCORnet) was 1.04 (95% CI 0.97–1.11; P = 0.58). Incident PASC by diagnosis code was 1.6% metformin vs. 2.0% comparator in the N3C, and 2.1% metformin vs. 2.5% comparator in PCORnet. By computable phenotype, incidence was 4.8% metformin and 5.2% comparator in the N3C and 24.7% metformin vs. 26.1% comparator in PCORnet. CONCLUSIONS Prevalent metformin use is associated with a slightly lower incidence of death or PASC after SARS-CoV-2 infection. PASC incidence by computable phenotype is higher than by EHR code, especially in PCORnet. These data are consistent with other observational analyses showing prevalent metformin is associated with favorable outcomes after SARS-CoV-2 infection in adults with T2DM.
Article
Background: Inconsistent results have been reported regarding the association between the use of antidiabetic drugs and the clinical outcomes of coronavirus disease 2019 (COVID-19). This study aimed to investigate the effect of antidiabetic drugs on COVID-19 outcomes in patients with diabetes using data from the National Health Insurance Service (NHIS) in South Korea.Methods: We analyzed the NHIS data of patients aged ≥20 years who tested positive for COVID-19 and were taking antidiabetic drugs between December 2019 and June 2020. Multiple logistic regression analysis was performed to analyze the clinical outcomes of COVID-19 based on the use of antidiabetic drugs.Results: A total of 556 patients taking antidiabetic drugs tested positive for COVID-19, including 271 male (48.7%), most of whom were in their sixties. Of all patients, 433 (77.9%) were hospitalized, 119 (21.4%) received oxygen treatment, 87 (15.6%) were admitted to the intensive care unit, 31 (5.6%) required mechanical ventilation, and 61 (11.0%) died. Metformin was significantly associated with the lower risks of mechanical ventilation (odds ratio [OR], 0.281; 95% confidence interval [CI], 0.109 to 0.720; P =0.008), and death (OR, 0.395; 95% CI, 0.182 to 0.854; P =0.018). Dipeptidylpeptidase-4 inhibitor (DPP-4i) were significantly associated with the lower risks of oxygen treatment (OR, 0.565; 95% CI, 0.356 to 0.895; P =0.015) and death (OR, 0.454; 95% CI, 0.217 to 0.949; P =0.036). Sulfonylurea was significantly associated with the higher risk of mechanical ventilation (OR, 2.579; 95% CI, 1.004 to 6.626; P =0.049).Conclusion: In patients with diabetes and COVID-19, metformin exhibited reduced risks of mechanical ventilation and death, DPP- 4i was linked with lower risks of oxygen treatment and death, while sulfonylurea was related to the increased risk of mechanical ventilation.
Article
It has been four years since long COVID—the protracted consequences that survivors of COVID-19 face—was first described. Yet, this entity continues to devastate the quality of life of an increasing number of COVID-19 survivors without any approved therapy and a paucity of clinical trials addressing its biological root causes. Notably, many of the symptoms of long COVID are typically seen with advancing age. Leveraging this similarity, we posit that Geroscience—which aims to target the biological drivers of aging to prevent age-associated conditions as a group—could offer promising therapeutic avenues for long COVID. Bearing this in mind, this review presents a framework for studying long COVID as a state of effectively accelerated biological aging, identifying research gaps and offering recommendations for future preclinical and clinical studies.
Article
Full-text available
Coronavirus Disease 2019 (COVID-19), caused by a new strain of coronavirus called Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), was declared a pandemic by WHO on March 11, 2020. Soon after its emergence in late December 2019, it was noticed that diabetic individuals were at an increased risk of COVID-19–associated complications, ICU admissions, and mortality. Maintaining proper blood glucose levels using insulin and/or other oral antidiabetic drugs (such as Metformin) reduced the detrimental effects of COVID-19. Interestingly, in diabetic COVID-19 patients, while insulin administration was associated with adverse outcomes, Metformin treatment was correlated with a significant reduction in disease severity and mortality rates among affected individuals. Metformin was extensively studied for its antioxidant, anti-inflammatory, immunomodulatory, and antiviral capabilities that would explain its ability to confer cardiopulmonary and vascular protection in COVID-19. Here, we describe the various possible molecular mechanisms that contribute to Metformin therapy’s beneficial effects and lay out the scientific basis of repurposing Metformin for use in COVID-19 patients.
Article
Full-text available
SARS-CoV-2 infections present with increased disease severity and poor clinical outcomes in diabetic patients compared with their non-diabetic counterparts. Diabetes/hyperglycemia-triggered endothelial dysfunction and hyperactive inflammatory and immune responses are correlated to two- to three-fold higher intensive care hospitalizations and more than twice the mortality among diabetic COVID-19 patients. While comorbidities such as obesity, cardiovascular disease, and hypertension worsen the prognosis of diabetic COVID-19 patients, COVID-19 infections are also associated with new-onset diabetes, severe metabolic complications, and increased thrombotic events in the backdrop of aberrant endothelial function. While several antidiabetic medications are used to manage blood glucose levels, we discuss the multi-faceted ability of metformin to control blood glucose levels, attenuate endothelial dysfunction, inhibit viral entry, and infection and modify inflammatory and immune responses during SARS-CoV-2 infections. These actions make it a viable candidate for drug repurposing and the higher ground against the SARS-CoV-2 induced tsunami in diabetic COVID-19 patients.
Article
Full-text available
The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has prompted an urgent need to identify effective medicines for the prevention and treatment of the disease. A comparative analysis between SARS-CoV-2 and Hepatitis C Virus (HCV) can expand the available knowledge regarding the virology and potential drug targets against these viruses. Interestingly, comparing HCV with SARS-CoV-2 reveals major similarities between them, ranging from the ion channels that are utilized, to the symptoms that are exhibited by patients. Via this comparative analysis, and from what is known about HCV, the most promising treatments for COVID-19 can focus on the reduction of viral load, treatment of pulmonary system damages, and reduction of inflammation. In particular, the drugs that show most potential in this regard include ritonavir, a combination of peg-IFN, and lumacaftor-ivacaftor. This review anaylses SARS-CoV-2 from the perspective of the role of ion homeostasis and channels in viral pathomechanism. We also highlight other novel treatment approaches that can be used for both treatment and prevention of COVID-19. The relevance of this review is to offer high-quality evidence that can be used as the basis for the identification of potential solutions to the COVID-19 pandemic.
Article
Full-text available
Introducción. El nuevo coronavirus causante de un brote de enfermedad respiratoria aguda en China en diciembre de 2019 se identificó como SARS-CoV-2. La enfermedad, denominada COVID-19, fue declarada pandemia por la Organización Mundial de la Salud (OMS). El primer caso de COVID-19 en Colombia se reportó el 6 de marzo de 2020; en este estudio se caracterizó un aislamiento temprano del virus SARS-CoV-2 de una muestra ecolectada en abril de 2020. Objetivos. Describir y caracterizar una cepa temprana a partir de un aislamiento de SARSCoV-2 durante la pandemia en Colombia. Materiales y métodos. Se obtuvo una muestra de un paciente con COVID-19 confirmada por qRT-PCR; la muestra fue inoculada en diferentes líneas celulares hasta la aparición del efecto citopático. Para confirmar la presencia de SARS-CoV-2 en el cultivo, se utilizó la qRT-PCR a partir de los sobrenadantes, la inmunofluorescencia indirecta (IFI) en células Vero-E6, así como microscopía electrónica y secuenciación de nueva generación (nextgeneration sequencing). Resultados. Se confirmó el aislamiento de SARS-CoV-2 en células Vero-E6 por la aparición del efecto citopático tres días después de la infección, así como mediante la qRT-PCR y la IFI positiva con suero de paciente convaleciente positivo para SARS-CoV-2. Además, en las imágenes de microscopía electrónica de trasmisión y de barrido de células infectadas se observaron estructuras compatibles con viriones de SARS-CoV-2. Por último, se obtuvo la secuencia completa del genoma, lo que permitió clasificar el aislamiento como linaje B.1.5. Conclusiones. La evidencia presentada en este artículo permite confirmar el primer aislamiento de SARS-CoV-2 en Colombia. Además, muestra que esta cepa se comporta en cultivo celular de manera similar a lo reportado en la literatura para otros aislamientos y que su composición genética está acorde con la variante predominante en el mundo. Finalmente, se resalta la importancia que tiene el aislamiento viral para la detección de anticuerpos, para la caracterización genotípica y fenotípica de la cepa y para probar compuestos con potencial antiviral.
Article
Full-text available
Some plus-stranded RNA viruses generate double-membrane vesicles (DMVs), one type of the membrane replication factories, as replication sites. Little is known about the lipid components involved in the biogenesis of these vesicles. Sphingomyelin (SM) is required for hepatitis C virus (HCV) replication, but the mechanism of SM involvement remains poorly understood. SM biosynthesis starts in the endoplasmic reticulum (ER) and gives rise to ceramide, which is transported from the ER to the Golgi by the action of ceramide transfer protein (CERT), where it can be converted to SM. In this study, inhibition of SM biosynthesis, either by using small-molecule inhibitors or by knockout (KO) of CERT, suppressed HCV replication in a genotype-independent manner. This reduction in HCV replication was rescued by exogenous SM or ectopic expression of the CERT protein, but not by ectopic expression of nonfunctional CERT mutants. Observing low numbers of DMVs in stable replicon cells treated with a SM biosynthesis inhibitor or in CERT-KO cells transfected with either HCV replicon or with constructs that drive HCV protein production in a replication-independent system indicated the significant importance of SM to DMVs. The degradation of SM of the in vitro-isolated DMVs affected their morphology and increased the vulnerability of HCV RNA and proteins to RNase and protease treatment, respectively. Poliovirus, known to induce DMVs, showed decreased replication in CERT-KO cells, while dengue virus, known to induce invaginated vesicles, did not. In conclusion, these findings indicated that SM is an essential constituent of DMVs generated by some plus-stranded RNA viruses. Copyright
Article
Full-text available
Background: Over 5,488,000 cases of coronavirus disease-19 (COVID-19) have been reported since December 2019. We aim to explore risk factors associated with mortality in COVID-19 patients and assess the use of D-dimer as a biomarker for disease severity and clinical outcome. Methods: We retrospectively analyzed the clinical, laboratory, and radiological characteristics of 248 consecutive cases of COVID-19 in Renmin Hospital of Wuhan University, Wuhan, China from January 28 to March 08, 2020. Univariable and multivariable logistic regression methods were used to explore risk factors associated with in-hospital mortality. Correlations of D-dimer upon admission with disease severity and in-hospital mortality were analyzed. Receiver operating characteristic curve was used to determine the optimal cutoff level for D-dimer that discriminated those survivors versus non-survivors during hospitalization. Results: Multivariable regression that showed D-dimer > 2.0 mg/L at admission was the only variable associated with increased odds of mortality [OR 10.17 (95% CI 1.10-94.38), P = 0.041]. D-dimer elevation (≥ 0.50 mg/L) was seen in 74.6% (185/248) of the patients. Pulmonary embolism and deep vein thrombosis were ruled out in patients with high probability of thrombosis. D-dimer levels significantly increased with increasing severity of COVID-19 as determined by clinical staging (Kendall's tau-b = 0.374, P = 0.000) and chest CT staging (Kendall's tau-b = 0.378, P = 0.000). In-hospital mortality rate was 6.9%. Median D-dimer level in non-survivors (n = 17) was significantly higher than in survivors (n = 231) [6.21 (3.79-16.01) mg/L versus 1.02 (0.47-2.66) mg/L, P = 0.000]. D-dimer level of > 2.14 mg/L predicted in-hospital mortality with a sensitivity of 88.2% and specificity of 71.3% (AUC 0.85; 95% CI = 0.77-0.92). Conclusions: D-dimer is commonly elevated in patients with COVID-19. D-dimer levels correlate with disease severity and are a reliable prognostic marker for in-hospital mortality in patients admitted for COVID-19.
Article
Full-text available
The outbreak of COVID‐19 is an emerging global health threat. The healthcare workers are facing challenges in reducing the severity and mortality of COVID‐19 across the world. Severe patients with COVID‐19 are generally treated in the intensive care unit (ICU), while mild or non‐severe patients treated in the usual isolation ward of the hospital. This article is protected by copyright. All rights reserved.
Article
Full-text available
Background The ongoing outbreak of the recently emerged novel coronavirus (2019-nCoV) poses a challenge for public health laboratories as virus isolates are unavailable while there is growing evidence that the outbreak is more widespread than initially thought, and international spread through travellers does already occur.AimWe aimed to develop and deploy robust diagnostic methodology for use in public health laboratory settings without having virus material available.Methods Here we present a validated diagnostic workflow for 2019-nCoV, its design relying on close genetic relatedness of 2019-nCoV with SARS coronavirus, making use of synthetic nucleic acid technology.ResultsThe workflow reliably detects 2019-nCoV, and further discriminates 2019-nCoV from SARS-CoV. Through coordination between academic and public laboratories, we confirmed assay exclusivity based on 297 original clinical specimens containing a full spectrum of human respiratory viruses. Control material is made available through European Virus Archive - Global (EVAg), a European Union infrastructure project.Conclusion The present study demonstrates the enormous response capacity achieved through coordination of academic and public laboratories in national and European research networks.
Article
Aims: We compared the association of metformin use and COVID-19 outcomes in a cohort of 31 966 patients with diabetes in Lombardy. Methods: We used a COVID-19 linkable administrative regional database to select diabetic patients over 40 years old. They had at least two prescriptions of antidiabetic drugs in 2019 and a positive test for SARS-CoV-2 between February 15, 2020 and March 15, 2021. The association of metformin use and clinical outcomes was assessed by multivariable logistic regression analyses and after propensity score matching. Clinical outcomes were all-cause mortality, in-hospital mortality, hospitalization for COVID-19 and admission to an intensive care unit (ICU). Results: In multivariable models metformin use was associated with a significantly lower risk of total mortality (OR 0.70; 95% CI 0.66-0.75), in-hospital mortality (OR 0.68; 95% CI 0.63-0.73), hospitalization for COVID-19 (OR 0.86; 95% CI 0.81-0.91) and ICU admission (OR 0.81; 95% CI 0.69-0.94) compared with metformin non-users. Results were similar in propensity score analysis; metformin was associated with significantly lower risk of total mortality (OR 0.79; 95% CI, 0.73-0.86), in-hospital mortality (OR 0.74; 95% CI, 0.67-0.81) and ICU admission (OR 0.77; 95% CI, 0.63-0.95). Conclusions: In this large cohort, metformin use was associated with a protective effect in COVID-19 clinical outcomes, suggesting that it might be a potentially useful drug to prevent severe COVID-19 disease, although randomized clinical trials (RCT) are needed to confirm this. While awaiting the results of RCT, we suggest continuing prescribing metformin to diabetic patients with COVID-19. This article is protected by copyright. All rights reserved.