Invest Clin 65(1): 83 - 98, 2024 https://doi.org/10.54817/IC.v65n1a08
Corresponding Author: Halis Suleyman, Department of Pharmacology, Institute of Health Sciences, Erzincan Bi-
nali Yildirim University, Erzincan, Turkey, Postcode: 24100; Phone: +90 530 9211909; Fax: +90 446 2261819.
E-mail: halis.suleyman@gmail.com; hsuleyman@erzincan.edu.tr
Pulmonary toxicity associated with
high-dose favipiravir and treatment
options: biochemical and histopathological
evaluation.
Bekir Elma 1, Bahadir Suleyman 2, Renad Mammadov 2, Bulent Yavuzer 3, Edhem Unver 4,
Durdu Altuner 2, Taha A. Coban 5, Behzad Mokhtare 6 and Halis Suleyman 3
1 Department of Thoracic Surgery, Faculty of Medicine, Division of Surgical Medical
Sciences, Gaziantep University, 27310, Gaziantep, Turkey.
2 Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University,
24100, Erzincan, Turkey.
3 Department of Pharmacology, Institute of Health Sciences, Erzincan Binali Yıldırım
University, 24100, Erzincan, Turkey.
4 Department of Chest Diseases, Faculty of Medicine, Erzincan Binali Yıldırım University,
24100, Erzincan, Turkey.
5 Department of Medical Biochemistry, Faculty of Medicine, Erzincan Binali Yildirim
University, 24100, Erzincan, Turkey.
6 Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, 25240,
Erzurum, Turkey.
Keywords: Favipiravir; lacidipine; thiamine pyrophosphate; adenosine triphosphate;
oxidative stress.
Abstract. Favipiravir is a broad-spectrum antiviral drug that is a viral RNA-
dependent RNA polymerase inhibitor. Favipiravir is used in high doses to treat
COVID-19 but has a side effect on humans at high doses. The side effects of
favipiravir have been associated with oxidative stress in the literature. In this
trial, we investigated the biochemical and histopathological effects of lacidip-
ine, thiamine pyrophosphate (TTP), and adenosine triphosphate (ATP), drugs
with antioxidant properties, on the lung toxicity caused by high-dose favipiravir
in rats. The rats were classified into five groups: healthy (HG), favipiravir alone
(Fav), lacidipine+favipiravir (LFav), TPP+favipiravir (TFav), and ATP+favipiravir
(AFav). Favipiravir (800 mg/kg) was administered twice daily for seven days. Laci-
dipine (4 mg/kg), TPP (20 mg/kg), and ATP (25 mg/kg) were administered once
daily for seven days. Oxidant (malondialdehyde), non-enzymatic (total glutathi-
one), and enzymatic (superoxide dismutase and catalase) antioxidant levels were
measured in the excised lung tissues. Furthermore, the tissues were histopatho-
logically examined. The systemic administration of high doses of favipiravir in-
creased oxidant levels and decreased antioxidant levels in the lung tissue of rats.
84 Elma et al.
Investigación Clínica 65(1): 2024
Toxicidad pulmonar asociada a altas dosis de favipiravir y
opciones de tratamiento: Evaluación bioquímica e histopatológica.
Invest Clin 2024; 65 (1): 83 – 98
Palabras clave: Favipiravir; lacidipino; pirofosfato de tiamina; trifosfato de adenosina;
estrés oxidativo.
Resumen. El Favipiravir es un fármaco antiviral de amplio espectro que es un
inhibidor de la ARN polimerasa viral dependiente de ARN. El Favipiravir se usa en do-
sis altas para tratar el COVID-19, pero tiene efectos secundarios en humanos a estas
dosis. Los efectos secundarios del favipiravir se han asociado con el estrés oxidativo
en la literatura. En este trabajo experimental, investigamos los efectos bioquímicos
e histopatológicos de lacidipina, pirofosfato de tiamina (TTP) y trifosfato de adeno-
sina (ATP), fármacos con propiedades antioxidantes, sobre la toxicidad pulmonar
causada por altas dosis de favipiravir en ratas. Las ratas se clasificaron en cinco gru-
pos: sanas (HG), favipiravir solo (Fav), lacidipina+favipiravir (LFav), TPP+favipiravir
(TFav) y ATP+favipiravir (AFav). Se administró favipiravir (800 mg/kg) dos veces al
día durante siete días. Se administraron lacidipina (4 mg/kg), TPP (20 mg/kg) y
ATP (25 mg/kg) una vez al día durante siete días. Se midieron los niveles de antioxi-
dantes oxidantes (malondialdehído), no enzimáticos (glutatión total) y enzimáticos
(superóxido dismutasa y catalasa) en los tejidos pulmonares disecados. Además, los
tejidos fueron examinados histopatológicamente. La administración sistémica de
altas dosis de favipiravir aumentó los niveles de oxidantes y disminuyó los niveles
de antioxidantes en el tejido pulmonar de ratas. Paralelamente, el examen histopa-
tológico del tejido pulmonar reveló la presencia de graves infiltraciones de células
mononucleares en las zonas intersticiales y una pronunciada hiperplasia linfoide.
Lacidipina mostró una eficacia superior para mitigar el estrés oxidativo y prevenir
la disminución de antioxidantes inducida por favipiravir en comparación con TPP y
ATP. Histopatológicamente, la administración de lacidipina redujo significativamen-
te el daño oxidativo pulmonar. La TTP redujo moderadamente la lesión pulmonar
grave asociada al favipiravir. Sin embargo, el ATP fue ineficaz contra la lesión pul-
monar asociada al favipiravir. Lacidipina ofrece más beneficios terapéuticos que el
TPP en el tratamiento de la lesión pulmonar oxidativa causada por altas dosis de
favipiravir.
Received: 16-07-2023 Accepted: 28-10-2023
In parallel, the histopathological examination of the lung tissue revealed
the presence of severe mononuclear cell infiltrations in interstitial areas and
pronounced lymphoid hyperplasia. Lacidipine exhibited superior efficacy in mit-
igating oxidative stress and preventing the decline of antioxidants induced by
favipiravir compared with TPP and ATP. Histopathologically, the lacidipine admin-
istration significantly reduced lung oxidative damage. TTP moderately reduced
severe favipiravir-associated lung injury. However, ATP was ineffective against fa-
vipiravir-associated lung injury. Lacidipine offers more therapeutic benefits than
TPP in treating oxidative lung injury caused by high doses of favipiravir.
Pulmonary toxicity associated with high-dose favipiravir and treatment options 85
Vol. 65(1): 83 - 98, 2024
INTRODUCTION
Favipiravir is a nucleoside-derived pro-
drug with a wide range of antiviral activity.
It acts by inhibiting the viral RNA-dependent
RNA polymerase (RdRp) 1 and has been used
to treat viral infections, such as Ebola and
severe acute respiratory syndrome (SARS-
CoV-2) 2. Furthermore, favipiravir is approved
to be used as an antiviral medication in Ja-
pan to treat influenza virus infections 3. The
effectiveness of favipiravir against influenza
has been verified via cell cultures, animal
studies, and clinical trials 4. Additionally, fa-
vipiravir has been undergoing clinical trials
as an investigational drug owing to its poten-
tial application in treating the novel corona-
virus disease 2019 (COVID-19) 5. The results
of preclinical and clinical investigations in-
dicate that favipiravir shows promise as a
potential treatment option for severe infec-
tions caused by human rhinovirus, respira-
tory syncytial virus, metapneumovirus, para-
influenza viruses, and hantavirus pulmonary
syndrome 4. In addition, favipiravir has been
reported to be a promising and effective an-
tiviral medication for treating patients with
COVID-19 3. For COVID-19 treatment, a
loading dose of 2400–3000 mg (given in two
doses) every 12 h followed by a maintenance
dose of 1200–1800 mg every 12 h was rec-
ommended 6,7. Numerous studies have evalu-
ated favipiravir’s effectiveness and potential
side effects in treating patients with COV-
ID-19 8. High doses of favipiravir have been
associated with severe side effects in humans
9. Additionally, signs of toxicity have been
observed in animals administered with high
doses of favipiravir 10. Although the lethal
dose of favipiravir in animals is >2000 mg/
kg, it is administered to patients at higher
doses, such as 6000 mg/day on the first day
and 2400 mg/day on the second and subse-
quent days 11. Furthermore, the use of favipi-
ravir has been linked to toxic side effects in
humans, including diarrhea, nephrotoxicity,
elevated serum uric acid and transaminase
levels, and reduced white blood cell and neu-
trophil counts alongside symptoms such as
nausea, vomiting, abdominal pain, skin rash,
itching, delirium, hallucinations, convul-
sions, and potential teratogenicity 12-14. A re-
cent experimental study revealed that favipi-
ravir administration increased the levels of
malondialdehyde (MDA), a toxic byproduct
of lipid peroxidation (LPO), decreased the
levels of the non-enzymatic endogenous an-
tioxidant glutathione (GSH), and inhibited
activities of enzymatic antioxidants such as
superoxide dismutase (SOD), and catalase
(CAT), in liver tissue 15. Considering the
documented side effects of favipiravir, its
safety profile remains a concern based on a
pooled analysis of extensive studies 9. To the
best of our knowledge, no literature studies
have specifically investigated the impact of
high-dose favipiravir on lung function.
Herein, we investigated the therapeutic
effect of lacidipine, a drug derived from di-
hydropyridine and classified as an L-type cal-
cium (Ca2+) channel blocker, in mitigating
the potential pulmonary toxicity caused by
high-dose favipiravir 16. Lacidipine has been
primarily indicated for hypertension treat-
ment 17. Moreover, lacidipine exhibits anti-
oxidant activity by inhibiting the increase in
MDA levels and decrease in enzymatic and
non-enzymatic antioxidant levels in organs
and tissues 18. In addition, lacidipine re-
portedly inhibits acute and chronic inflam-
mation phases 19. Thiamine pyrophosphate
(TPP) is the active metabolite of thiamine,
and we investigated the therapeutic effect
of TPP against potential lung toxicity due to
high-dose favipiravir 20. Previous studies re-
port that TPP exerts a protective effect by
inhibiting elevated oxidant and proinflam-
matory parameters 21,22.
Moreover, we examined the therapeu-
tic effect of adenosine triphosphate (ATP)
against potential lung toxicity associated
with high-dose favipiravir administration.
ATP is a nucleoside triphosphate comprising
adenine, ribose sugar, and three phosphate
groups 23. ATP also synthesizes reactive
oxygen species (ROS)-scavenging antioxi-
86 Elma et al.
Investigación Clínica 65(1): 2024
dants24. In addition, ATP is an energy source
for synthesizing low molecular weight anti-
oxidants 25. This study aimed to biochemical-
ly investigate and histopathologically evalu-
ate the effects of lacidipine, TPP, and ATP on
the possible oxidative lung damage caused
by favipiravir in rats.
MATERIALS AND METHODS
Animals
In total, 36 albino Wistar-type male rats
weighing 285–297 g were obtained from Er-
zincan Binali Yıldırım University Experimen-
tal Animal Research and Application Center.
The experimental rats were subjected to
standard environmental conditions, main-
tained at an ambient temperature of 22°C
and a 12/12 h light–dark cycle. The rats had
ad libitum access to animal feed and tap wa-
ter. The study was approved by the local Ani-
mal Experiments Ethics Committee (Meet-
ing Date: 25.08.2022; Meeting Number:
2022/08; Decision Number: 37).
Chemicals
Thiopental sodium was obtained from
IE Ulagay (Turkey), favipiravir was obtained
from the Ministry of Health Training and
Research Hospital (Turkey), lacidipine was
obtained from GlaxoSmithKline (Turkey),
TPP was obtained from Biofarma (Russia),
and ATP was obtained from Zdorove Narodu
(Ukraine).
Experimental groups
The rats were classified into five
groups: healthy (HG), favipiravir alone (Fav),
lacidipine+favipiravir (LFav), TPP+favipiravir
(TFav), and ATP+favipiravir (AFav).
Experimental procedure
Lacidipine (4 mg/kg orally), TPP (20
mg/kg IP), and ATP (25 mg/kg IP) were ad-
ministered to the LFav (n = 6), TFav (n =
6), and AFav (n = 6) groups, respectively,
to initiate the experiment. The HG (n = 6)
and Fav (n = 6) groups were administered
with distilled water. Following the 1-h inter-
val following the administration of drugs and
distilled water, favipiravir was orally adminis-
tered at a dosage of 800 mg/kg twice daily
for seven days to all the animal groups ex-
cept the HG group. Favipiravir is known to
cause oxidative and inflammatory damage at
high doses 26. Lacidipine, TPP, and ATP were
administered once daily for seven days. TTP
and ATP have been investigated before in
these doses and found to be effective against
oxidative stress 27,28. Upon completion of this
timeframe, the animals were euthanized via
a high dose of thiopental sodium anesthesia
(50 mg/kg), following which their lung tis-
sues were extracted. MDA, tGSH, SOD, and
CAT levels were measured in the excised
lung tissues. Furthermore, the tissues were
histopathologically examined. The biochem-
ical and histopathological findings obtained
from all the animal groups were compared
and assessed for intergroup differences.
Biochemical analyzes
Preparation of samples
At this stage, 0.2 g of each removed tis-
sue was weighed for biochemical examina-
tion. Tissue samples were washed with cold
(+4°C) 0.15 M potassium chloride (KCl).
Tissue samples were homogenized in liquid
nitrogen. They were then passed into an ice-
cold phosphate buffer solution (50 mM, pH
7.4). The tissue homogenates were centri-
fuged at 5000 rpm for 20 min at +4°C, and
the supernatants were extracted to analyze
MDA, tGSH, SOD, and CAT.
Determination of MDA, GSH, SOD, CAT,
and protein
The levels of MDA, GSH, and SOD in
the supernatants derived from the lung tis-
sue samples from experimental animals were
measured using commercially available en-
zyme-linked immunosorbent assay (ELISA)
kits according to the manufacturer’s in-
structions (MDA catalogue no: 10009055;
tGSH catalogue no: 703002; SOD catalogue
no: 706002, Cayman Chemical Company).
Pulmonary toxicity associated with high-dose favipiravir and treatment options 87
Vol. 65(1): 83 - 98, 2024
CAT levels were determined according to
the method proposed by Goth 29. Protein
content was determined spectrophotometri-
cally at 595 nm according to the Bradford
method 30.
Histopathological examination
The rats underwent necropsy for his-
topathological assessment, and their lung
tissue samples were subsequently fixed in a
10% formalin solution. The tissues were sub-
jected to a series of alcohol–xylene solutions
and then embedded in paraffin blocks, from
which 5-µm thick sections were obtained for
histopathological evaluation. Then, these
sections were stained using hematoxylin–
eosin stain. The lung tissues were examined
using a light microscope (Olympus BX51,
Japan), and photographs were captured us-
ing a digital camera (Olympus DP 71) by a
pathologist blinded to the treatment proto-
col. Histopathological damage in each tissue
section was graded on a scale of 0–3 (0 =
normal, 1 = mild, 2 = moderate, and 3 = se-
vere) and assessed for mononuclear cell in-
filtrations in interstitial areas and lymphoid
hyperplasia.
Statistical analysis
All statistical analyses for the biochemi-
cal findings of the experiment were conduct-
ed using IBM SPSS® Statistics for Windows,
version 22.0 (IBM Corp, Armonk, NY, USA,
released in 2013). A significance level of p
<0.05 was considered statistically signifi-
cant. The biochemical results are expressed
as mean ± standard error ( ± SEM). The
normality of distribution for continuous
variables in the biochemical test results was
assessed using the Shapiro–Wilk test. The
significance level of the difference between
the groups was determined using a one-way
analysis of variance, as the distribution was
normal. The Levene’s test was performed to
determine whether the homogeneity of vari-
ances was ensured. Following the assump-
tion of homogeneity of variances, either the
Tukey Honest Significant Differences test or
the Games–Howell test was employed as a
post hoc test. The histopathological findings
were analyzed via the IBM SPSS® Statistics
program for Windows®, version 20.0 (IBM
Corp, Armonk, NY, USA, released in 2011). A
nonparametric Kruskal–Wallis test was per-
formed to assess between-group differences.
Subsequently, the group responsible for the
observed differences was identified using the
Mann–Whitney U test at a significance level
of p <0.05.
RESULTS
Biochemical results
MDA analysis results of lung tissue
The lung tissue of the high-dose favipi-
ravir–treated group exhibited higher MDA
levels than those in the lung tissue of the
HG, which was statistically significant (Table
1). Lacidipine, TPP, and ATP significantly
suppressed the increase in MDA levels in-
duced by high-dose favipiravir administra-
tion in lung tissue. However, ATP prevented
the increase of MDA in lung tissue weaker
than lacidipine and TPP. There was no signif-
icant difference in the MDA levels between
the HG, lacidipine, and TPP groups.
tGSH analysis results of lung tissue
The lung tissue of the high-dose favipi-
ravir–treated group exhibited lower tGSH
levels than the lung tissue of the HG, and
the difference was statistically significant
(Table 1). Lacidipine and TTP significantly
suppressed the decrease in tGSH levels in-
duced by high-dose favipiravir administra-
tion in lung tissue. However, ATP adminis-
tration did not exhibit a significant effect
on preventing a decrease in tGSH levels. A
statistically significant difference was de-
tected in the tGSH levels between the lung
tissue of the ATP-treated group and the HG.
There was no significant difference in the
tGSH levels between the HG, lacidipine, and
TPP groups.
88 Elma et al.
Investigación Clínica 65(1): 2024
SOD analysis results of lung tissue
The lung tissue of the high-dose favipi-
ravir–treated group exhibited lower SOD
activity compared with the lung tissue of
the HG, and the difference was statistically
significant (Table 1). Lacidipine and TPP
significantly alleviated the decrease in SOD
activity induced by high-dose favipiravir ad-
ministration in lung tissue; however, ATP
did not exhibit a similar effect. A statisti-
cally significant difference was found in the
SOD activities between the lung tissue of
the TPP and ATP groups and the HG. There
was no significant difference observed in
SOD activity between the HG and the laci-
dipine group.
Table 1
Effect of lacidipine, TPP, and ATP on oxidant and antioxidant levels in lung tissue
of rats administered with high doses of favipiravir.
Mean ± Standard error
MDA tGSH SOD CAT
Groups (nmol/mg protein) (nmol/mg protein) (U/mg protein) (U/mg protein)
HG 1.65±0.07 6.63±0.08 9.55±0.05 8.51±0.06
Fav 5.80±0.06 2.56±0.05 3.56±0.06 3.53±0.09
LFav 1.82±0.03 6.31±0.15 9.19±0.15 8.14±0.12
TFav 2.72±0.34 5.34±0.34 6.49±0.34 6.18±0.36
AFav 3.84±0.23 2.79±0.19 3.84±0.11 3.66±0.06
Comparison of p-values
Groups MDA tGSH SOD CAT
HG vs Fav <0.001 <0.001 <0.001 <0.001
HG vs LFav 0.215 0.386 0.302 0.116
HG vs TFav 0.118 0.056 0.001 0.006
HG vs AFav 0.001 <0.001 <0.001 <0.001
Fav vs LFav <0.001 <0.001 <0.001 <0.001
Fav vs TFav 0.001 0.002 0.001 0.003
Fav vs AFav 0.002 0.784 0.235 0.714
LFav vs TFav 0.201 0.163 0.001 0.011
LFav vs AFav 0.002 <0.001 <0.001 <0.001
TFav vs AFav 0.131 0.001 0.002 0.004
Abbreviations: TPP: thiamine pyrophosphate; ATP: adenosine triphosphate; HG: healthy group; Fav: alone favipiravir ad-
ministered group; LFav: lacidipine + favipiravir group; TFav: TPP + favipiravir group; AFav: ATP + favipiravir group; MDA:
malondialdehyde; tGSH: total glutathione; SOD: superoxide dismutase; CAT: catalase.
Footnotes: The Games-Howell test was applied as a post-hoc test after one-way ANOVA for all statistical evaluations.
Pulmonary toxicity associated with high-dose favipiravir and treatment options 89
Vol. 65(1): 83 - 98, 2024
CAT analysis results of lung tissue
CAT activity was lower in the lung tis-
sue of the high-dose favipiravir–treated group
than in the lung tissue of the HG, and the dif-
ference was statistically significant (Table 1).
Lacidipine and TPP significantly suppressed
the decrease in CAT activity induced by high-
dose favipiravir administration in lung tissue;
however, ATP did not suppress this decrease.
A statistically significant difference was de-
tected in the CAT activities between the lung
tissue of the TPP and ATP groups and those
of the HG. No significant difference was ob-
served in CAT activity between the HG and
the lacidipine group.
Histopathological results
Histopathologically significant differ-
ences were detected between the HG, Fav,
LFav, TFav, and AFav groups (Table 2; p<
0.05). The lung tissue samples of the HG ex-
hibited a normal histologic appearance (Fig.
1). Nevertheless, the rats in the high-dose fa-
vipiravir group exhibited severe mononucle-
ar cell infiltrations in interstitial areas (Fig.
2A) and significant lymphoid hyperplasia in
the lung tissue (Fig. 2B). In contrast, mono-
nuclear cell infiltrations in interstitial areas
(Fig. 2C) and lymphoid hyperplasia (Fig. 2D)
were mild in the lacidipine group. In the TTP
group, mononuclear cell infiltrations in inter-
stitial areas (Fig. 3A) and lymphoid hyperpla-
sia (Fig. 3B) were moderate. However, mono-
nuclear cell infiltrations in interstitial areas
(Fig. 3C) and lymphoid hyperplasia (Fig. 3D)
were severe in the ATP group.
DISCUSSION
This study investigated the protective
effects of lacidipine, TPP, and ATP against
lung injury induced by high-dose favipira-
vir administration in rats. The investigation
involved biochemical and histopathological
analyses. Our biochemical experiments re-
vealed that the MDA levels increased in the
lung tissue of high-dose favipiravir–treated
animals, whereas the tGSH, SOD, and CAT
levels decreased significantly.
Our experimental results indicate that
high doses of favipiravir may lead to severe
oxidative damage. Herein, we measured MDA
levels as it is a toxic byproduct of LPO and a
significant indicator of oxidative stress 31. A re-
cent experimental study with results that align
with our biochemical findings reported that
favipiravir administration increased MDA levels
in liver tissue. Additionally, the drug decreased
endogenous antioxidant levels 15. Similarly, Bi-
lici et al. reported that rats treated with a high
dose of favipiravir exhibited increased oxidant
and decreased antioxidant levels 26.
Table 2
Effect of lacidipine, TPP, and ATP on histopathological scoring ndings
in lung tissue of rats administered with high doses of favipiravir.
Groups MNC infiltrations in interstitial areas Lymphoid hyperplasia
HG 0.00 ± 0.00a0.00 ± 0.00a
FAV 2.83 ± 0.40b2.83 ± 0.40b
LFav 0.16 ± 0.00a0.16 ± 0.00a
TFav 1.16 ± 0.40c1.16 ± 0.40c
AFav 2.66 ± 0.81b2.56 ± 0.80b
Abbreviations: TPP: thiamine pyrophosphate; ATP: adenosine triphosphate; MNC: mononuclear cell; HG: healthy group;
Fav: alone favipiravir administered group; LFav: lacidipine + favipiravir group; TFav: TPP+ favipiravir group; AFav: ATP+
favipiravir group.
Footnotes: The values given are mean ± standard deviation values. a, b, c: Groups marked with the same letter are statistically
similar, but there is a statistically signicant difference at the level of p < 0.05 among groups with different letters.
90 Elma et al.
Investigación Clínica 65(1): 2024
Fig. 1. Normal histological appearance of lung tissue belonging to the HG group (H&E x10).
Fig. 2. (A) Severe mononuclear cell infiltrations in interstitial areas () appearance in the lung tissue belon-
ging to the Fav group (H&E x20). (B) Severe lymphoid hyperplasia (arrowheads) appearance in the
lung tissue belonging to the Fav group (H&E x10). (C) Mild mononuclear cell infiltrations in inters-
titial areas () appearance in the lung tissue belonging to the LFav group (H&E x20). (D) Mild lym-
phoid hyperplasia (arrowheads) appearance in the lung tissue belonging to the LFav group (H&E x10).
Pulmonary toxicity associated with high-dose favipiravir and treatment options 91
Vol. 65(1): 83 - 98, 2024
Conversely, lacidipine, TPP, and ATP
administration significantly suppressed in-
creased MDA levels in the studied subjects.
In particular, it was observed that lacidipine
suppressed the increase in MDA levels to a
greater extent than TPP and ATP, bringing
them closer to the levels of the healthy con-
trol group. To the best of our knowledge,
there is no information in the literature
regarding the effect of lacidipine, TPP, and
ATP on lung injury induced by high-dose fa-
vipiravir. Previous studies have reported that
lacidipine significantly decreased MDA lev-
els and exerted a nephroprotective effect in
cyclosporine-induced nephrotoxicity 32. Pre-
vious research has also demonstrated that
lacidipine exhibits the highest potency as
an antioxidant among calcium channel an-
tagonists, effectively inhibiting membrane
LPO 33. In addition, TPP exerts an antioxi-
dant effect by significantly suppressing the
cyclophosphamide-induced increase in MDA
levels in female rats 21. Similarly, a previous
study reported that ATP protects kidney tis-
sue from bevacizumab-induced oxidative
damage by significantly inhibiting increased
MDA levels 34.
In addition, enzymatic and non-enzy-
matic antioxidant parameters were mea-
sured to evaluate the possible oxidative
damage of favipiravir in lung tissue. It is
well-known that exposure to ROS from vari-
ous sources activates a cascade of defense
mechanisms in organs and tissues 35, such
as endogenous antioxidants 36. In case of in-
sufficient antioxidant levels to counteract
Fig. 3. (A) Moderate mononuclear cell infiltrations in interstitial areas () appearance in the lung tissue belon-
ging to the TFav group (H&E x20). (B) Moderate lymphoid hyperplasia (arrowheads) appearance in the
lung tissue belonging to the TFav group (H&E x10). (C) Severe mononuclear cell infiltrations in inters-
titial areas () appearance in the lung tissue belonging to the AFav group (H&E x20). (D) Severe lym-
phoid hyperplasia (arrowheads) appearance in the lung tissue belonging to the AFav group (H&E x10).
92 Elma et al.
Investigación Clínica 65(1): 2024
the oxidant accumulation, oxidative stress
occurs, leading to tissue damage 37,38. As re-
vealed by our experimental results, tGSH,
SOD, and CAT levels decreased in lung tis-
sue. It is widely recognized that GSH ex-
ists in two primary forms: the thiol-reduced
form and the disulfide-oxidized form known
as GSSG 39. GSH has several functions, in-
cluding antioxidant defense, detoxification,
maintenance of thiol status, and modulation
of cell proliferation 40. The protective effect
of GSH is attributed to its ability to react
with ROS and effectively perform detoxifi-
cation 41. SODs are enzymatic antioxidants
that play a role in the protection of cells
against oxygen toxicity 42. Likewise, CAT is
a significant antioxidant enzyme in various
cells that facilitates the breakdown of H2O2
into H2O and O2 42. Numerous prior studies
have assessed the levels of oxidants and anti-
oxidants above-mentioned to investigate the
occurrence of lung damage 43.
Our study revealed that lacidipine and
TPP administration effectively mitigated
the decline in tGSH, SOD, and CAT levels.
However, ATP did not demonstrate the same
suppressive effect on these antioxidant
markers. Moreover, lacidipine effectively
reduced the decline in these antioxidant
levels, restoring them to levels comparable
with those observed in the HG. To the best
of our knowledge, there is no data in the
literature regarding the antioxidant effect
of lacidipine against oxidative lung injury
due to favipiravir. Several studies have re-
ported that lacidipine protects heart tissue
from oxidant damage 44. In another study,
lacidipine treatment reportedly attenuated
the decrease in tGSH, SOD, and CAT lev-
els 45. It was determined that another drug,
TPP, suppressed the decrease in tGSH lev-
els and provided similar values as those of
the healthy control group; however, there
was a significant difference in SOD and CAT
activities between the healthy control and
TPP group. To the best of our knowledge,
there was no data in the literature regard-
ing the antioxidant effect of TTP against
favipiravir-induced oxidative lung injury. In
a previous study, TPP reportedly increased
tGSH levels in oxidative optic nerve damage
to levels comparable to those in the healthy
group 46. Demiryilmaz et al. reported that
SOD and CAT activities increased in rats
treated with TPP following oxidative liver
damage 47. Reportedly, ATP was ineffective
in preventing the decline of enzymatic and
non-enzymatic antioxidant levels and exhib-
ited a noticeable difference compared to
the values observed in the healthy control
group. To the best of our knowledge, there
were no reports in the literature investigat-
ing the antioxidant effect of ATP against
favipiravir-induced oxidative lung injury.
While Ozer et al. reported that ATP ad-
ministration increased the enzymatic and
non-enzymatic antioxidant levels in oxida-
tive ovarian damage, our study did not yield
similar findings 48.
Our study revealed a correlation be-
tween the biochemical results obtained from
the lung tissues of the animals and histo-
pathological findings. Severe mononuclear
cell infiltrations and lymphoid hyperplasia
in interstitial areas were observed in the
lung tissue of the favipiravir group, wherein
the oxidant levels were increased, and an-
tioxidant levels were decreased. However,
histopathological damage was observed to
be alleviated in the lacidipine group, which
best antagonized the effect of favipiravir on
oxidant and antioxidant parameters. While
histopathologic damage was moderate in
the TPP group, which prevented oxidant in-
crease and antioxidant decrease at a mod-
erate level, severe histopathologic damage
was detected in the ATP group, which could
not significantly prevent the decrease of an-
tioxidants caused by favipiravir. This is con-
sistent with the literature reporting that
histopathologic damage in lung tissue is as-
sociated with oxidant/antioxidant levels 49,50.
Pneumonia is a widely recognized condition
characterized by inflammation of the lungs,
typically due to an infection 51. There is cur-
rently no information available in the litera-
Pulmonary toxicity associated with high-dose favipiravir and treatment options 93
Vol. 65(1): 83 - 98, 2024
ture suggesting that favipiravir induces in-
terstitial inflammation via oxidative stress in
the lungs. However, Tomoda Y et al. reported
the development of drug-induced intersti-
tial pneumonia in a patient receiving clopi-
dogrel 52. Jo T et al. reported that class III
antiarrhythmic drugs, epidermal growth fac-
tor receptor inhibitors, and numerous other
drugs can cause interstitial pneumonia 53.
Reportedly, the broncho-alveolar lavage fluid
of a patient with drug-associated pneumo-
nia contained abundant lymphocytes, mac-
rophages, neutrophils, and eosinophils 52. A
study reported that ROS-induced oxidative
stress may be one of the underlying factors
in interstitial pneumonia development 54,
supporting our biochemical and histopatho-
logical findings.
In conclusion, the systemic adminis-
tration of high doses of favipiravir increased
oxidant levels and decreased antioxidant
levels in the lung tissue of rats. In parallel,
the histopathological examination of the
lung tissue revealed the presence of severe
mononuclear cell infiltrations in interstitial
areas and pronounced lymphoid hyperpla-
sia. To the best of our knowledge, this was
the first study focusing on the impact of
high-dose systemic administration of favipi-
ravir on lung tissue in terms of biochemical
alterations and histopathological changes.
Lacidipine exhibited superior efficacy in
mitigating oxidative stress and preventing
the decline of antioxidants induced by fa-
vipiravir compared with TPP and ATP. His-
topathologically, lacidipine administration
significantly reduced lung oxidative dam-
age. TTP moderately reduced severe favip-
iravir-associated lung injury. However, ATP
was ineffective against favipiravir-associat-
ed lung injury. Based on our findings, it can
be concluded that lacidipine offers more
therapeutic benefits than TPP in treating
oxidative lung injury caused by high doses
of favipiravir. We believe investigating pro-
inflammatory and inflammatory cytokines
can provide valuable insights into further
understanding this issue.
ACKNOWLEDGMENTS
The authors thank Enago – https://
www.enago.com.tr/ceviri/ for their manu-
script translation and editing assistance.
Conflict of interest
There are no conflicts of interest be-
tween the authors or between family mem-
bers of the scientific and medical com-
mittees. The authors do not have any
consultancy, expertise, working conditions,
shareholdings or similar situations that
could lead to potential conflicts of interest
in any company.
ORCID authors
Bekir Elma(BE):
0000-0001-5083-8088
Bahadir Suleyman (BS):
0000-0001-5795-3177
Renad Mammadov (RM):
0000-0002-5785-1960
Bulent Yavuzer (BY):
0000-0001-7576-0678
Edhem Unver(EU):
0000-0002-0322-8102
Durdu Altuner(DA):
0000-0002-5756-3459
Taha A. Coban (TAC):
0000-0003-1711-5499
Behzad Mokhtare(BM):
0000-0002-9075-7239
Halis Suleyman(HS):
0000-0002-9239-4099
Authorships contribution statement
BE: conceptualization, methodology,
software, formal analysis, resources, data
curation, writing original draft preparation,
writing-review & editing, supervision, proj-
ect administration; BS: conceptualization,
94 Elma et al.
Investigación Clínica 65(1): 2024
methodology, validation, formal analysis,
investigation, resources, writing original
draft preparation, writing-review & editing;
RM: methodology, validation, formal analy-
sis, investigation, resources, writing-review
& editing; BY: methodology, software, for-
mal analysis, investigation, resources, data
curation, writing-review & editing, visual-
ization; EU: methodology, validation, inves-
tigation, resources, writing-review & edit-
ing; DA: methodology, validation, formal
analysis, writing original draft preparation,
writing-review & editing; TAC: conceptu-
alization, methodology, validation, formal
analysis, resources, writing original draft
preparation, writing-review & editing; BM:
conceptualization, methodology, validation,
formal analysis, resources, writing original
draft preparation, writing-review & edit-
ing; HS: conceptualization, methodology,
validation, formal analysis, investigation,
writing original draft preparation, writing-
review & editing, visualization, supervision,
project administration.
REFERENCES
1. García-Lledó A, Gómez-Pavón J, Gonzá-
lez del Castillo J, Hernández-Sampelayo
T, Martín-Delgado MC, Martín Sánchez
FJ, Martínez-Sellés M, Molero García JM,
Moreno Guillén S, Rodríguez-Artalejo F,
Ruiz-Galiana J, Cantón R, De Lucas Ra-
mos P, García-Botella A, Bouza E. Phar-
macological treatment of COVID-19: an
opinion paper. Rev Esp Quimioter 2021;
35(2): 115-130. https://doi.org/10.37201/
req/158.2021
2. Łagocka R, Dziedziejko V, Kłos P, Pawlik
A. Favipiravir in therapy of viral infections.
J Clin Med 2021; 10(2): 273. https://doi.
org/10.3390/jcm10020273
3. Pogue JM, McCreary EK. Coronavirus
disease 2019 treatment: a review of early
and emerging options. Open Forum In-
fect Dis 2020; 7(4): ofaa105. https://doi.
org/10.1093/ofid/ofaa105
4. Majewska A, Smyk JM. Favipiravir in the
battle with respiratory viruses. Mini Rev
Med Chem 2022; 22(17): 2224-2236.
https://doi.org/10.2174/1389557522666
220218122744
5. Favipiravir. National Institute of Child
Health and Human Development; 2006.
Bethesda (MD). Updated 15 February
2023. PMID: 32401463. https://www.ncbi.
nlm.nih.gov/pubmed/32401463
6. Lipsitch M, Sissoko D, Laouenan C, Fo-
lkesson E, M’Lebing A-B, Beavogui A-H,
Baize S, Camara A-M, Maes P, Shepherd
S, Danel C, Carazo S, Conde MN, Gala
J-L, Colin G, Savini H, Bore JA, Le Mar-
cis F, Koundouno FR, Petitjean F, Lamah
M-C, Diederich S, Tounkara A, Poelart
G, Berbain E, Dindart J-M, Duraffour S,
Lefevre A, Leno T, Peyrouset O, Irenge
L, Bangoura NF, Palich R, Hinzmann J,
Kraus A, Barry TS, Berette S, Bongo-
no A, Camara MS, Chanfreau Munoz V,
Doumbouya L, Souley H, Kighoma PM,
Koundouno FR, Réné L, Loua CM, Mas-
sala V, Moumouni K, Provost C, Samake
N, Sekou C, Soumah A, Arnould I, Ko-
mano MS, Gustin L, Berutto C, Camara
D, Camara FS, Colpaert J, Delamou L,
Jansson L, Kourouma E, Loua M, Mal-
me K, Manfrin E, Maomou A, Milinouno
A, Ombelet S, Sidiboun AY, Verreckt I,
Yombouno P, Bocquin A, Carbonnelle C,
Carmoi T, Frange P, Mely S, Nguyen V-K,
Pannetier D, Taburet A-M, Treluyer J-M,
Kolie J, Moh R, Gonzalez MC, Kuisma E,
Liedigk B, Ngabo D, Rudolf M, Thom R,
Kerber R, Gabriel M, Di Caro A, Wölfel
R, Badir J, Bentahir M, Deccache Y, Du-
mont C, Durant J-F, El Bakkouri K, Ga-
sasira Uwamahoro M, Smits B, Toufik N,
Van Cauwenberghe S, Ezzedine K, Dor-
tenzio E, Pizarro L, Etienne A, Guedj J,
Fizet A, Barte de Sainte Fare E, Murgue
B, Tran-Minh T, Rapp C, Piguet P, Pon-
cin M, Draguez B, Allaford Duverger T,
Barbe S, Baret G, Defourny I, Carroll M,
Raoul H, Augier A, Eholie SP, Yazdanpa-
nah Y, Levy-Marchal C, Antierrens A, Van
Herp M, Günther S, de Lamballerie X,
Keïta S, Mentre F, Anglaret X, Malvy D.
Experimental Treatment with Favipiravir
for Ebola Virus Disease (the JIKI Trial):
Pulmonary toxicity associated with high-dose favipiravir and treatment options 95
Vol. 65(1): 83 - 98, 2024
A Historically Controlled, Single-Arm
Proof-of-Concept Trial in Guinea. PLoS
Med 2016; 13(3): e1001967. https://doi.
org/10.1371/journal.pmed.1001967
7. Mentre F, Taburet AM, Guedj J, Anglaret
X, Keita S, de Lamballerie X, Malvy D.
Dose regimen of favipiravir for Ebola virus
disease. Lancet Infect Dis 2015; 15(2):
150-151. https://doi.org/10.1016/S1473-
3099(14)71047-3
8. Hung DT, Ghula S, Aziz JMA, Makram
AM, Tawfik GM, Abozaid AA-F, Pancha-
ratnam RA, Ibrahim AM, Shabouk MB,
Turnage M, Nakhare S, Karmally Z,
Kouz B, Le TN, Alhijazeen S, Phuong
NQ, Ads AM, Abdelaal AH, Nam NH, Ii-
yama T, Kita K, Hirayama K, Huy NT.
The efficacy and adverse effects of fa-
vipiravir on patients with COVID-19: A
systematic review and meta-analysis of
published clinical trials and observatio-
nal studies. Int J Infect Dis 2022; 120:
217-227. https://doi.org/10.1016/j.ijid.
2022.04.035
9. Pilkington V, Pepperrell T, Hill A. A re-
view of the safety of favipiravir - a potential
treatment in the COVID-19 pandemic? J
Virus Erad 2020; 6(2): 45-51. https://doi.
org/10.1016/S2055-6640(20)30016-9
10. Driouich J-S, Cochin M, Lingas G, Mou-
reau G, Touret F, Petit P-R, Piorkowski
G, Barthélémy K, Laprie C, Coutard B,
Guedj J, de Lamballerie X, Solas C, Nou-
gairède A. Favipiravir antiviral efficacy
against SARS-CoV-2 in a hamster model.
Nat Commun 2021; 12(1): 1735. https://
doi.org/10.1038/s41467-021-21992-w
11. Yamazaki S, Suzuki T, Sayama M, Naka-
da T-a, Igari H, Ishii I. Suspected choles-
tatic liver injury induced by favipiravir in
a patient with COVID-19. J Infect Che-
mother 2021; 27(2): 390-392. https://doi.
org/10.1016/j.jiac.2020.12.021
12. Chen C, Zhang Y, Huang J, Yin P, Cheng Z,
Wu J, Chen S, Zhang Y, Chen B, Lu M, Luo
Y, Ju L, Zhang J, Wang X. Favipiravir versus
arbidol for clinical recovery rate in mode-
rate and severe adult COVID-19 patients:
a prospective, multicenter, open-label,
randomized controlled clinical trial. Front
Pharmacol 2021; 12: 683296. https://doi.
org/10.3389/fphar.2021.683296
13. Dogan E, Alkan-Çeviker S, Vurucu S,
Sener A, Yüksel B, Gönlügür U, Simsek
T, Hakan Ulusoy M. Investigation of the
frequency of adverse effects in patients
treated with favipiravir as SARS-CoV-2
treatment. Klimik Dergisi/Klimik J 2021;
34(2): 95-98. https://doi.org/10.36519/
kd.2021.3563
14. Hayden FG, Shindo N. Influenza virus
polymerase inhibitors in clinical deve-
lopment. Curr Opin Infect Dis 2019;
32(2): 176-186. https://doi.org/10.1097/
qco.0000000000000532
15. Kara A, Yakut S, Caglayan C, Atçalı T, Ulu-
can A, Kandemir FM. Evaluation of the to-
xicological effects of favipiravir (T-705) on
liver and kidney in rats: biochemical and
histopathological approach. Drug Chem
Toxicol 2022; 46(3): 546-556. https://doi.
org/10.1080/01480545.2022.2066116
16. McCormack PL, Wagstaff AJ. Lacidipine.
Drugs 2003; 63(21): 2327-2356. https://
doi.org/10.2165/00003495-200363210-
00008
17. Sai Chebrolu T, Kumar L, Verma R. Laci-
dipine: review of analytical methods deve-
loped for pharmaceutical dosage forms and
biological fluids. Bioanalysis 2021; 13(12):
1011-1024. https://doi.org/10.4155/bio-
2021-0024
18. Suleyman B, Halici Z, Odabasoglu F,
Gocer F. The effect of lacidipine on in-
domethacin induced ulcers in rats. Int J
Pharmacol 2012; 8(2): 115-121. https://
doi.org/10.3923/ijp.2012.115.121
19. Suleyman H, Halici Z, Hacimuftuoglu
A, Gocer F. Role of adrenal gland hormo-
nes in antiinflammatory effect of calcium
channel blockers. Pharmacol Rep 2006;
58(5): 692-699. PMID: 17085861
20. Sica DA. Loop diuretic therapy,thiamine
balance, and heart failure. Congestive Heart
Fail 2007; 13(4): 244-247. https://doi.
org/10.1111/j.1527-5299.2007.06260.x
21. Ozer M, Ince S, Gundogdu B, Aktas M,
Uzel K, Gursul C, Suleyman H, Suleyman
Z. Effect of thiamine pyrophosphate on
cyclophosphamide-induced oxidative ova-
96 Elma et al.
Investigación Clínica 65(1): 2024
rian damage and reproductive dysfunction
in female rats. Adv Clin Exp Med 2022;
31(2): 129-137. https://doi.org/10.17219/
acem/142535
22. Turan MI, Cayir A, Cetin N, Suleyman
H, Turan IS, Tan H. An investigation
of the effect of thiamine pyrophospha-
te on cisplatin-induced oxidative stress
and DNA damage in rat brain tissue
compared with thiamine. Hum Exp To-
xicol 2013; 33(1): 14-21. https://doi.
org/10.1177/0960327113485251
23. Dunn J, Grider MH. Physiology, Adeno-
sine Triphosphate. 2023 Feb 13. In: Stat-
Pearls [Internet]. Treasure Island (FL):
StatPearls Publishing; 2023 Jan–.PMID:
31985968. https://www.ncbi.nlm.nih.gov/
pubmed/31985968
24. Saquet AA, Streif J, Bangerth F. Changes
in ATP, ADP and pyridine nucleotide levels
related to the incidence of physiological
disorders in ‘Conference’ pears and ‘Jona-
gold’ apples during controlled atmosphe-
re storage. J Hortic Sci Biotechnol 2015;
75(2): 243-249. https://doi.org/10.1080/
14620316.2000.11511231
25. Yi C, Jiang Y, Shi J, Qu H, Xue S, Duan
X, Shi J, Prasad NK. ATP-regulation of
antioxidant properties and phenolics in
litchi fruit during browning and patho-
gen infection process. Food Chem 2010;
118(1): 42-47. https://doi.org/10.1016/j.
foodchem.2009.04.074
26. Bilici S, Altuner D, Suleyman Z, Bulut
S, Sarigul C, Gulaboglu M, Altindag F,
Ozcicek A, Gursul C, Suleyman H. Fa-
vipiravir-induced inflammatory and hy-
dropic degenerative liver injury in rats.
Adv Clin Exp Med 2023; 32(8): 881-887.
https://doi.org/10.17219/acem/159089
27. Polat B, Suleyman H, Sener E, Akcay
F. Examination of the effects of thia-
mine and thiamine pyrophosphate on
doxorubicin-induced experimental car-
diotoxicity. J Cardiovasc Pharmacol
Ther 2014; 20(2): 221-229. https://doi.
org/10.1177/1074248414552901
28. Koç A, Gazi M, Sayar AC, Onk D, Arı MA,
Süleyman B, Ağgül AG, Altındağ F, Süle-
yman DAH. Molecular mechanism of the
protective effect of adenosine triphospha-
te against paracetamol-induced liver to-
xicity in rats. Gen Physiol Biophys 2023;
42(02): 201-208. https://doi.org/10.4149/
gpb_2022055
29. Góth L. A simple method for determina-
tion of serum catalase activity and revi-
sion of reference range. Clin Chim Acta
1991; 196(2-3): 143-151. https://doi.
org/10.1016/0009-8981(91)90067-m
30. Bradford M. A rapid and sensitive method
for the quantitation of microgram quan-
tities of protein utilizing the principle
of protein-dye binding. Anal Biochem
1976; 72(1-2): 248-254. https://doi.
org/10.1016/0003-2697(76)90527-3
31. Balcı S, Çöllüoğlu Ç, Yavuzer B, Bulut S,
Altındağ F, Akbaş N, Süleyman H. Effect
of low and high dose of favipiravir on ova-
rian and reproductive function in female
rats: Biochemical and histopathological
evaluation. Gen Physiol Biophys 2022;
41(05): 457-463. https://doi.org/10.4149/
gpb_2022036
32. Naidu MU, Kumar KV, Shifow AA, Prayag
A, Ratnakar KS. Lacidipine protects
against cyclosporine-induced nephrotox-
icity in rats. Nephron 1999; 81(1): 60-66.
https://doi.org/10.1159/000045247
33. van Amsterdam FTM, Roveri A, Maiori-
no M, Ratti E, Ursini F. Lacidipine:
A dihydropyridine calcium antagonist
with antioxidant activity. Free Radic Biol
Med 1992; 12(3): 183-187. https://doi.
org/10.1016/0891-5849(92)90025-c
34. Suleyman H, Kocaturk H, Bedir F,
Turangezli O, Arslan R, Coban T, Altun-
er D. Effect of adenosine triphosphate,
benidipine and their combinations on
bevacizumab-induced kidney damage in
rats. Adv Clin Exp Med 2021; 30(11):
1175-1183. https://doi.org/10.17219/
acem/140440
35. Cadenas E. Basic mechanisms of anti-
oxidant activity. Biofactors 1997; 6(4):
391-397. https://doi.org/10.1002/
biof.5520060404
36. Valko M, Leibfritz D, Moncol J, Cronin
MTD, Mazur M, Telser J. Free radicals and
antioxidants in normal physiological func-
Pulmonary toxicity associated with high-dose favipiravir and treatment options 97
Vol. 65(1): 83 - 98, 2024
tions and human disease. Int J Biochem
Cell Biol 2007; 39(1): 44-84. https://doi.
org/10.1016/j.biocel.2006.07.001
37. Clarkson PM, Thompson HS. Antioxi-
dants: what role do they play in physi-
cal activity and health? Am J Clin Nutr
2000; 72(2): 637S-646S. https://doi.
org/10.1093/ajcn/72.2.637S
38. Kisaoglu A, Borekci B, Yapca OE, Bilen H,
Suleyman H. Tissue damage and oxidant/
antioxidant balance. Eurasian J Med 2013;
45(1): 47-49. https://doi.org/10.5152/eaj
m.2013.08
39. Kaplowitz N, Aw TY, Ookhtens M. The
regulation of hepatic glutathione. Annu
Rev Pharmacol Toxicol 1985; 25(1): 715-
744. https://doi.org/10.1146/annurev.pa.
25.040185.003435
40. Lu SC. Regulation of glutathione synthe-
sis. Mol Aspects Med 2009; 30(1-2): 42-59.
https://doi.org/10.1016/j.mam.2008.05.005
41. Owen JB, Butterfield DA. Measurement of
oxidized/reduced glutathione ratio. Meth-
ods Mol Biol 2010; 648: 269-277. https://
doi.org/10.1007/978-1-60761-756-3_18
42. Ali SS, Ahsan H, Zia MK, Siddiqui T,
Khan FH. Understanding oxidants and an-
tioxidants: Classical team with new play-
ers. J Food Biochem 2020; 44(3): e13145.
https://doi.org/10.1111/jfbc.13145
43. Ermis H, Parlakpinar H, Elbe H, Vardi
N, Polat A, Gulbas G. Effects of vareni-
cline on lung tissue in the animal model.
J Bras Pneumol 2020; 46(2): e20180406-
e20180406. Efeitos da vareniclina no teci-
do pulmonar em modelo animal. https://
doi.org/10.36416/1806-3756/e20180406
44. Khan NA, Chattopadhyay P, Abid M,
Pawdey A, Kishore K, Wahi AK. Protective
effects of amlodipine on mitochondrial
injury in ischemic reperfused rat heart. J
Environ Biol 2012; 33(3): 591-595. PMID:
23029908
45. Khurana K, Bansal N. Lacidipine attenu-
ates reserpine-induced depression-like be-
havior and oxido-nitrosative stress in mice.
Naunyn Schmiedebergs Arch Pharmacol
2019; 392(10): 1265-1275. https://doi.
org/10.1007/s00210-019-01667-6
46. Ucak T, Karakurt Y, Tasli G, Cimen FK,
Icel E, Kurt N, Ahiskali I, Süleyman H.
The effects of thiamine pyrophosphate
on ethanol induced optic nerve damage.
BMC Pharmacol Toxicol 2019; 20(1): 40.
https://doi.org/10.1186/s40360-019-
0319-5
47. Demiryilmaz I, Sener E, Cetin N, Altuner
D, Suleyman B, Albayrak F, Akcay F, Su-
leyman H. Biochemically and histopatho-
logically comparative review of thiamine’s
and thiamine pyrophosphate’s oxidative
stress effects generated with methotrexate
in rat liver. Med Sci Monit 2012; 18(12):
BR475-BR481. https://doi.org/10.12659/
msm.883591
48. Ozer M, Ince S, Altuner D, Suleyman Z,
Cicek B, Gulaboglu M, Mokhtare B, Gur-
sul C, Suleyman H. Protective rffect of
adenosine triphosphate against 5-fluorou-
racil-induced oxidative ovarian damage in
vivo. Asian Pac J Cancer Prev 2023; 24(3):
1007-1013. https://doi.org/10.31557/APJ
CP.2023.24.3.1007
49. Tosun M, Olmez H, Unver E, Arslan Y,
Cimen F, Ozcicek A, Aktas M, Suleyman
H. Oxidative and pro-inflammatory lung
injury induced by desflurane inhalation
in rats and the protective effect of rutin.
Adv Clin Exp Med 2021; 30(9): 941-948.
https://doi.org/10.17219/acem/136194
50. Dhouib H, Jallouli M, Draief M, Bouraoui
S, El-Fazâa S. Oxidative damage and histo-
pathological changes in lung of rat chro-
nically exposed to nicotine alone or asso-
ciated to ethanol. Pathol Biol (Paris) 2015;
63(6): 258-267. https://doi.org/10.1016/j.
patbio.2015.10.001
51. Stotts C, Corrales-Medina VF, Rayner KJ.
Pneumonia-induced inflammation, reso-
lution and cardiovascular disease: causes,
consequences and clinical opportunities.
Circ Res 2023; 132(6): 751-774. https://
doi.org/10.1161/circresaha.122.321636
52. Tomoda Y, Miyajima T, Nagasawa C, Awa-
ya Y. Clopidogrel-induced pneumonia. BMJ
Case Rep 2021; 14(6): e244564. https://
doi.org/10.1136/bcr-2021-244564
98 Elma et al.
Investigación Clínica 65(1): 2024
53. Jo T, Michihata N, Yamana H, Morita K,
Ishimaru M, Yamauchi Y, Hasegawa W,
Urushiyama H, Uda K, Matsui H, Fushimi
K, Yasunaga H, Nagase T. Risk of drug-in-
duced interstitial lung disease in hospitali-
sed patients: a nested case–control study.
Thorax 2021; 76(12): 1193-1199. https://
doi.org/10.1136/thoraxjnl-2020-215824.
54. De AK, Rajan RR, Krishnamoorthy L,
Bhatt MB, Singh BB. Oxidative stress in
radiation-induced interstitial pneumo-
nitis in the rat. Int J Radiat Biol 2009;
68(4): 405-409. https://doi.org/10.1080/
09553009514551351.