Next Article in Journal
Immunological Mechanisms behind Anti-PD-1/PD-L1 Immune Checkpoint Blockade: Intratumoral Reinvigoration or Systemic Induction?
Previous Article in Journal
USP7 Deregulation Impairs S Phase Specific DNA Repair after Irradiation in Breast Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunomodulatory Effects of Fluoroquinolones in Community-Acquired Pneumonia-Associated Acute Respiratory Distress Syndrome

by
Resti Yudhawati
1,2,3,* and
Nisrina Fitriyanti Wicaksono
4
1
Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
2
Department of Pulmonology and Respiratory Medicine, Universitas Airlangga Teaching Hospital, Surabaya 60015, Indonesia
3
Department of Pulmonology and Respiratory Medicine, Dr. Soetomo General Hospital, Surabaya 60286, Indonesia
4
Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(4), 761; https://doi.org/10.3390/biomedicines12040761
Submission received: 26 February 2024 / Revised: 18 March 2024 / Accepted: 26 March 2024 / Published: 29 March 2024
(This article belongs to the Section Immunology and Immunotherapy)

Abstract

:
Community-acquired pneumonia is reported as one of the infectious diseases that leads to the development of acute respiratory distress syndrome. The innate immune system is the first line of defence against microbial invasion; however, its dysregulation during infection, resulting in an increased pathogen load, stimulates the over-secretion of chemokines and pro-inflammatory cytokines. This phenomenon causes damage to the epithelial–endothelial barrier of the pulmonary alveoli and the leakage of the intravascular protein into the alveolar lumen. Fluoroquinolones are synthetic antimicrobial agents with immunomodulatory properties that can inhibit bacterial proliferation as well as exhibit anti-inflammatory activities. It has been demonstrated that the structure of fluoroquinolones, particularly those with a cyclopropyl group, exerts immunomodulatory effects. Its capability to inhibit phosphodiesterase activity leads to the accumulation of intracellular cAMP, which subsequently enhances PKA activity, resulting in the inhibition of transcriptional factor NF-κB and the activation of CREB. Another mechanism reported is the inhibition of TLR and ERK signalling pathways. Although the sequence of events has not been completely understood, significant progress has been made in comprehending the specific mechanisms underlying the immunomodulatory effects of fluoroquinolones. Here, we review the indirect immunomodulatory effects of FQs as an alternative to empirical therapy in patients diagnosed with community-acquired pneumonia.

1. Introduction

Community-acquired pneumonia (CAP) is one of the most common infectious diseases, contributing significantly to reported rates of mortality and morbidity around the world [1,2]. Pathogens that cause CAP are classified into two types: ‘typical’ agents, including Gram-positive organisms (such as Streptococcus pneumoniae, Staphylococcus aureus, Haemophilus influenza. Group A Streptococcus spp.; anaerobes), and Gram-negative organisms (such as Klebsiella pneumoniae, Streptococcus pyogenes, Pseudomonas aeruginosa, Escherichia coli, Acinetobacter baumannii and Stenotrophomonas maltophilia); ‘atypical’ agents include Legionella pneumophila, Mycoplasma pneumoniae, Chlamydophila pneumoniae, Chlamydophila psittaci, influenza viruses (A, B), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and other respiratory viruses [3,4,5]. Co-infection with bacteria is a frequent phenomenon observed in respiratory viral infections, leading to an elevation in both morbidity and mortality rates [6,7]. Co-infection, also commonly referred to as “superinfection”, is frequently found during the pandemic of viruses [8,9,10].
Complications may arise as a result of pneumonia, leading to the development of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS): a condition that is associated with significant rates of morbidity and mortality [11,12]. Pneumonia and sepsis are globally recognised as the main risk factor (~75% of cases) for ARDS [13]. A quantitative model study generated data from 13 countries across the world and concluded that ~22.15% of pneumonia patients developed ARDS [14].
The innate immune system serves as the first barrier of defence in the host’s response to pathogens by identifying their pathogen-associated molecular patterns (PAMPs) or microbe-associated molecular patterns (MAMPs). However, during infection, the innate immune response experiences dysregulation, aggravating the severity of illness by increasing the pathogen load as the consequence of inefficient pathogen clearance or irreversibly damaging the organs of patients with sepsis, who subsequently may die as a result of multi-organ failure [15,16]. The innate immune response specific to a particular organ determines the infection severity. The lungs exhibit a robust innate immune response during localised lung infections associated with severe ALI or ARDS, which significantly influences the outcome of the disease [15,17].
The administration of anti-inflammatory agents such as steroids to patients with CAP (with or without shock) remains controversial, although many studies have demonstrated a significant decrease in hospitalisation duration and time to reach clinical stability [18,19,20,21]. Patients with significant inflammatory responses, such as those with a high level of c-reactive proteins (CRPs), may constitute a subset of severe CAP patients who benefit from such corticosteroid therapy, according to accumulated published data [19]. While many studies suggest the benefit of steroids, one study showed increased mortality [22]. Another study also suggested that corticosteroid treatment did not improve survival in CAP patients, while nosocomial infections were increased [23].
Fluoroquinolones (FQs) are a class of synthetic antimicrobial agents that inhibit DNA synthesis by targeting DNA gyrase and topoisomerase IV enzymes [24]. One of FQ’s properties are its broad spectrum; therefore, these compounds are highly active in combating Gram-positive and Gram-negative bacteria, and even anaerobes, mycobacteria and atypical pathogens [24,25,26,27]. Besides the above-mentioned properties, these drugs have been reported to exert anti-oxidative effects both in vitro and in vivo [28,29]. In addition, FQs is also reported to block pro-inflammatory cytokines and chemokines, leading to the disruption of neutrophil chemotaxis [25]. According to the most recent guidelines and the literature available, FQs have been accurately proven to exert immunomodulatory effects, which are clinically advantageous for the treatment of CAP. Immunomodulatory effects in FQs have been described as beneficial to reducing lung damage due to bacterial, viral, and fungal infections in animal models [27,29,30,31,32,33].
The main obstacle to improving the outcome of CAP is the excessive pro-inflammatory response [34]. Several therapeutic options have been tested to improve the outcome of CAP using different strategies. FQs have immunomodulatory effects beyond their antibacterial effects that might be beneficial for patients with CAP. Hence, the present review article focuses on the indirect immunomodulatory effects of FQs, in addition to their direct antibacterial effects, which have been utilised as an alternative to empirical therapy in patients diagnosed with CAP.

2. Immunopathogenesis of Pneumonia-Associated ALI/ARDS

ALI/ARDS is a condition that results from heterogeneous aetiologies, with bacterial pneumonia being the dominant cause [11,35]. The disruption in the blood–air barrier due to the infiltration of innate immune cells, the release of inflammatory mediators, and other injury pathways leads to further lung damage and the influx of protein-rich pulmonary oedema [25,36].
Bacteria, both Gram-positive and Gram-negative, viruses, and fungi have uniform molecular patterns known as PAMPs, which are identified by pattern recognition receptors (PRRs) on the surface of the organism’s immune cells, one of which is Toll-like receptor (TLR), before finally binding to them [17,37]. Following the successful entrance of bacteria to the lower airway, bacteria interact with macrophages—PRRs—of the pulmonary innate immune system via their cell walls and intracellular components (lipoteichoic acid, peptidoglycan, nucleic acids, pneumolysin, and other pore-forming toxins), and consequently, transcription factors, such as nuclear factor κappa-B (NF-κB) are activated [17,25,38].
NF-κB represents a family of five transcription factors that play crucial roles in various biological processes that support aspects of differentiation and development, immune response modulation, cell growth, proliferation, apoptosis, and phenotypic outcomes associated with inflammation [39,40]. Notably conserved in all mammalian cells, NF-κB plays a pivotal role in the transcription of genes encoding numerous cytokines and chemokines, including those with pro-inflammatory properties [41]. NF-κB is bound to the inhibitory protein, inhibitory κappa B (IκB), and retained in the cytoplasm of the resting, non-stimulated cell. NF-κB proteins are commonly activated and released in response to various extracellular ligands, including agents that induce a DNA damage response (DDR), leading to the translocation of DNA-binding protein dimers to the nucleus after dissociating from IκB molecules [39,40,42].
Signal transduction pathways triggered by cell stimulation with multiple agonists initiate the activation of IκB kinases (IKK). IKK induces the phosphorylation of IκB, which is subsequently followed by a rapid degradation of the IκB proteins, leading to the liberation of NF-κB. Consequently, NF-κB translocates into the nucleus, where it binds to DNA and initiates the process of transcriptional activation [43]. The nuclear translocation of the activated transcription factor triggers the induction of genes encoding various pro-inflammatory cytokines (interleukin (IL)-1β, IL-6, IL-8, IL-17, IL-18, tumour necrosis factor (TNF)) and chemokines (CCL8, monocyte chemotactic protein 1 (MCP-1), macrophage inflammatory protein 1α (MIP-1α)) [38,44]. High levels of cytokines and chemokines in plasma and bronchoalveolar lavage (BAL) fluids are related to poor clinical outcomes in ARDS, including a high mortality rate [44,45]. A study observing ARDS patients showed that Staphylococcus, Streptococcus, and Enterobacteriaceae were identified as the specific bacteria associated with elevated levels of IL-6 in non-surviving patients with ARDS. Streptococcus secretes pneumolysin and MUC5B, both of which have been found to be closely associated with lung cell fibrosis and lung inflammation [46].
The activation of protein-1 (AP-1) is observed to have a role in ALI/ARDS pathogenesis by trans-activating pro-inflammatory cytokines and other genes that lead to lung damage [47]. In response to various stimuli, TLR4 induces and recruits intracellular adaptor proteins, resulting in a signalling cascade that involves similar signalling molecules to NF-κB signalling on the TLR4/TRAF6 axis [48,49]. TRAF6 activates TAK1 (transforming growth factor-activated kinase 1) and subsequently initiates a MAPK (mitogen-activated protein kinase) cascade that includes ERK (extracellular signal-regulated kinases), JNK (c-Jun N-terminal kinases), and p38, leading to the activation and nuclear translocation of AP-1. The activated AP-1 interacts with the promoters of pro-inflammatory cytokines, increasing their expression [47,48].
p38MAPK, an essential signalling protein, has been established in the literature to play a major pro-inflammatory role in the development of ARDS at both the transcriptional and post-transcriptional levels. Evidence has suggested that the activation of p38MAPK plays a critical role in the synthesis of inflammatory cytokines [50]. Among its four isoforms, p38α MAPK was the first to be identified for its function in the regulation of pro-inflammatory cytokines. IL-8 and IL-6 production in response to IL-1 and TNF-α, respectively, were then associated with p38α MAPK. As the initiation factors, IL-1β and TNF-α have the ability to directly injure vascular endothelial cells as well as activate a series of effector cells [51].
Following damage-associated molecular pattern molecules (DAMPs) or alarmins derived from the host, which further bind with TLR on the lung epithelium and alveolar macrophages, the polarisation of alveolar macrophages (AMs), neutrophil extracellular trap (NET)osis, the pro-inflammatory response exhibited by T helper 17 subsets, and the anti-inflammatory and regenerative functions performed by T regulatory cell subsets occur [52,53,54]. During infections, DAMPs and PAMPs synergistically stimulate the synthesis and secretion of pro-inflammatory cytokines and chemokines, as well as induce cell differentiation and cell death [52,53]. Pro-inflammatory cytokines have double effects on the host defence mechanisms; on the one hand, they promote the activation of adaptive immunity that releases multiple mediators such as prostaglandins, leukotrienes, and proteases; however, on the other hand, they induce direct and indirect injury to the microvasculature of the host [37].
The secreted pro-inflammatory cytokines stimulate the activation of localised vascular endothelia cells; while the release of chemo-attractants attracts more monocytes and neutrophils, as well as the exudation of pro-inflammatory complement proteins and acute-phase reactants [38]. The recruitment of neutrophils is a defining feature of ARDS and is considered to play a critical role in the course of the disease [55]. When the neutrophils migrate to the epithelium, these cells elicit toxic mediators, including proteases, nitric oxide (NO), reactive oxygen species (ROS) and NET, which are essential host defence mechanisms [32,56]. Neutrophils release extracellular fibres, myeloperoxidase, DNA, and neutrophil elastase into the extracellular environment during pathogen invasion as a defensive mechanism, referred to as NETosis, to create a network for microbe entrapment. However, excessive neutrophil activation and unbalanced inflammatory responses may result in further tissue damage, including endothelial and epithelial damage, which may subsequently lead to an elevation in the permeability of these cells [55].
The increase in endothelial and epithelial permeability enables the transmigration of leucocytes and ultimately results in the influx of oedematous fluid and red blood cells (RBCs). RBCs produce cell-free haemoglobin, which aggravates damage through oxidant-dependent pathways. The airspace is filled with oedematous fluid, subsequently resulting in impaired gas exchange and profound hypoxemia. Vascular injury and alveolar oedema also participate in the decrease in CO2’s excretion ability (hypercapnia), causing an increase in pulmonary dead space in ARDS. Furthermore, hypoxaemia and hypercapnia reduce alveolar oedema clearance by impairing vectorial sodium transport [45,55].
Figure 1 illustrates the immunopathogenesis of ARDS (black arrow).

3. Specific Features of Quinolone Molecules Related to Immunomodulatory Effects

Quinolone is a class of antibiotics with a bicyclic structure derived from the 4-quinolone compound [57]. The carboxylic acid group at position 3 and the carbonyl at position 4 appear to have a significant role in determining the activity of quinolones. In addition, bulky substituents on one face of the bicyclic core are permitted, specifically at positions 1 and 7 and/or 8, and they are likely important in determining the spectrum of quinolone antibiotics. Regarding these substituents, most quinolones can be classified into three primary types based on their sidechains: piperazinyl-, pyrrolidinyl-, and piperidinyl-types [58]. A class of 6-fluoro-7-piperazinyl-4-quinolones, or fluoroquinolones, are synthetic antimicrobial agents that have a wide range of activity. These agents are derived from quinolones and have a fluorine atom attached to the central ring [59].
FQs have indirect antibacterial effects in addition to their intrinsic antibacterial activity, which may be due to their immunomodulatory activity [41]. Based on their pharmacokinetic profile and antimicrobial activity, fluoroquinolones are classified into four generations. Nalidixic acid was the first within the quinolone class that was discovered to have antibacterial activity. The first generation of quinolones was retracted from the market shortly after their introduction. Besides nalidixic acid, the first generation included cinoxacin [60]. The second generation of quinolones began with the formation of fluoroquinolones by fluoridating the quinolone molecule at position C6, which enhanced the compounds’ activity against Gram-negative bacteria. The addition of a cyclopropyl group at position R1 further improved the compounds’ overall activity [61,62]. Examples of second-generation drugs include ciprofloxacin, enoxacin, norfloxacin, ofloxacin and lomefloxacin. However, lomefloxacine was withdrawn from the market after a few years of approval for clinical use [63]. The third generation of quinolones was initiated with the development of fleroxacin. In this generation, more powerful FQs, including levofloxacin, sparfloxacin, grepafloxacin and gatifloxacin, were developed [64]. According to the WHO’s list of essential medications, ciprofloxacin and levofloxacin are the most commonly prescribed drugs [65]. Third-generation compounds also have an additional incorporation of new substituents, namely a chloro group (Cl) at the R8 position, which demonstrates enhanced bactericidal action against Gram-positive bacteria and atypical bacteria [62]. In the current market, sparfloxacin, gatifloxacin, and grepafloxacin were discontinued for clinical use [63]. The specific insertion of a cyclic diamine piperazine molecule at position C-7 and a fluorine atom at position C-6 have synthesised present-day FQs, which exhibit notable efficacy against anaerobic, Gram-positive, and Gram-negative bacteria. The third and fourth generations of FQs contain a methoxy group at the C-8 position and are currently licensed for the treatment of respiratory tract infections, including lethal pulmonary tuberculosis [66]. The fourth generation is represented by trovafloxacin, moxifloxacin, and gemifloxacin, with trovafloxacin being withdrawn from the market [63]. Some FQs were withdrawn from the market after a few years of approval for use due to an increased risk of various severe adverse effects that were associated with FQ administration [67,68].
FQs were initially optimised and developed as antimicrobial agents, and each generation appeared to confer increased potency. However, according to a review article by Anderson and Osheroff [69], multiple reports have indicated that their capabilities may extend beyond antimicrobial effects only. The extensive utilisation of quinolone derivatives in clinical applications has contributed to the identification of their immunomodulatory effects [70].
Several studies have reported how certain FQs exhibit in vitro anti-proliferative properties through various mechanisms, including the induction of apoptosis, the disruption of the biochemical transformation of potentially cancerous cells, the enhancement of other chemotherapeutic agents’ uptake, and/or mediation of immunomodulatory responses [71,72,73]. Quinolones, in general, exert their modulating effects only when combined with a co-stimulant. Nevertheless, it has been widely observed that quinolone compounds have a tendency to reduce the production of pro-inflammatory cytokines. The induction of cytokines has been observed only in a subset of FQs, and this effect seems to be related to the presence of the cyclopropyl moiety at the N1 position [74].
Quinolones are comprised a bicyclic ring structure with a substitution at position N-1 containing various moieties. The majority of current agents contain a fluorine atom at position 6 and a nitrogen heterocycle moiety at position C7 [75]. The precise mechanism remains unclear, but it is conceivable that certain FQs may activate transcription factors, such as AP-1, which are known to be associated with elevated cytokine levels [76]. The observed variations in outcomes across multiple studies can be attributed to the distinct chemical structures and different pathogens involved. The outcomes of these studies were likely influenced by the timing and frequency of administered doses [31,32,77].
Immunomodulatory effects are especially evident in FQs with a cyclopropyl-moiety at position N1 of their quinolone ring, such as ciprofloxacin and moxifloxacin [78]. Substituents located at position 1 of the basic quinolone structure affect antibacterial activity potency. The presence of a cyclopropyl substituent at this specific position, which is present on all of the new FQs besides levofloxacin and trovafloxacin, is regarded as the most optimal for activity [79]. Several quinolones with and without the cyclopropyl group on N1 are presented in Figure 2.
The presence of a cyclopropyl group at position N1 of the quinolone molecule has been shown to exhibit superior antibacterial action against Gram-negative bacteria, as demonstrated by Chu et al. [80] and Domagala [81]. Exposure to ciprofloxacin and CP-115,953, which contain a cyclopropyl group at position N1, significantly boosted the release of interferon (IFN)-y from human peripheral blood cells in vitro compared to compounds without this moiety [82,83]. Similarly, certain structural properties of FQs are also associated with these agents’ non-antibacterial activity against eucaryotic topoisomerase II. The study conducted by Yamashita et al. [84] presented the augmented anti-leukemic effects observed in a murine leukaemia model, which were accompanied by an increased inhibitory activity against topoisomerase II. This enhanced activity was observed specifically in quinolones that possessed the same cyclopropyl group attached to the N1 position of the quinolone ring. A comparable discovery was documented in a comparative study examining the effect of six quinolones on the production of IL-3 and the granulocyte–macrophage colony-stimulating factor (GM-CSF) by stimulated murine splenocytes [74]. Under the same experimental conditions, the production of these aforementioned cytokines was found to be enhanced exclusively by quinolones that incorporated the cyclopropyl group at position N1. Conversely, quinolones lacking in this moiety had either no impact or an inhibitory effect. The data presented in these studies provide clear evidence that specific FQs containing an N1-cyclopropyl group exhibit immunomodulatory effects [78].
Another study revealed that gemifloxacin, a fourth-generation fluoroquinolone drug, has significant immunomodulatory potential. Due to the presence of a cyclopropyl N-1 group in its structure, gemifloxacin may have the potential to influence both innate and adaptive immune systems. This drug has demonstrated a dual effect on both innate and adaptive immune systems, whereby it enhances the activity of the innate system while suppressing the adaptive immune system. The humoral immune response is produced through the synthesis of antibodies that target the specific epitope of the antigen employed to induce the immune response [85].

4. Immunomodulatory Activity of Fluoroquinolone in ARDS

The immunomodulatory effects of quinolones are mostly anti-inflammatory and have been widely documented in in vitro models; however, the precise mechanisms by which quinolones act as immunomodulators are not yet fully comprehended [65,86,87]. FQs are suggested to exhibit their immunomodulatory activities via influencing phosphodiesterase activity and promoting the production of intracellular cyclic adenosine monophosphate (cAMP) [88,89]. Presumably, quinolones elevate intracellular levels of cAMP by inhibiting the activity of phosphodiesterase enzymes [90]. cAMP is essential for the regulation of various inflammatory responses in innate immune cells [91]. The intracellular levels of these cyclic nucleotides are primarily regulated by enzymes called phosphodiesterases (PDEs), which catalyse the hydrolysis of a cyclic phosphate bond in cAMP and cyclic guanosine monophosphate (cGMP) to produce the inactive 5′-AMP and 5′-GMP [92]. The predominant subtype of PDE in neutrophils is PDE4, which is involved in the pathogenesis of inflammatory diseases [93]. Bailly et al. suggested that the inhibitory effect of ciprofloxacin on TNF-α and IL-1 may possibly be attributed to its inhibitory effect on phosphodiesterase, resulting in the accumulation of intracellular cAMP [94]. The accumulation of cAMP levels leads to an augmentation in the activity of protein kinase A (PKA), which is known to decrease the TNF-α expression [92]. Several investigations have indicated that this accumulation inhibits TNF-α and IL-1 production in mononuclear phagocytes [94,95,96]. A study conducted by Blaine et al. [97] provided evidence of the phosphodiesterase inhibitory effect of ciprofloxacin, which caused cAMP to accumulate in the cells and increase PKA activity, which, in turn, is known to inhibit the production of TNF-α in stimulated monocytes. There is a proposition that cAMP, which functions as a second messenger, has the potential to function as an anti-inflammatory agent through its ability to stimulate downstream signalling pathways, including PKA and the exchange protein directly activated by cAMP (Epac), while also reducing the secretion of cytokines [88]. PKA then subsequently activates CREB (cAMP response element-binding protein), which is a primary regulator of anti-inflammatory and immune response [98]. Furthermore, recent results present compelling evidence suggesting that certain cellular properties associated with cell motility can also be regulated by modulating cAMP levels [99,100].
The mechanisms by which quinolones exert their effects on various cytokines and chemokines involve the regulation of certain key cellular transcription factors. The transcription factor known as NF-κB is one of the key factors in cellular signals [41,42]. It is crucial to emphasise that the modulatory effects of quinolones are not observed when they are used alone, and an extra stimulating effect is required. The stimulators independently induce an intracellular stress response, such as lipopolysaccharide, which may interact synergistically with the inhibitory effects of topoisomerase-II to induce the augmented immunomodulatory event [41]. The inhibition of topoisomerase II impacts protein C kinase, resulting in enhanced AP-1 activity, which has been associated with increased cytokine levels [101,102]. The precise sequence of events that underlie the effect of quinolones on the above transcription factors and their activators, including Iκβ and potentially IKK, remains unknown and requires further studies.
It is known that MAPK, ERK, and JNK are involved in the activation of the transcription factor NF-κB, which, in turn, modulates immunological and inflammatory genes [103]. A biological molecule with a short lifespan called NO is considered a key marker of inflammatory lung diseases, including ARDS, asthma and lung fibrosis. The respiratory epithelium appears to serve as its primary source, subsequently to inducible NO synthase (iNOS) activation [104]. The production of iNOS in humans, as well as various inflammatory cytokines in the lung, are reliant on the MAPK and NF-κB signalling pathways [105]. Multiple in vitro studies have demonstrated that moxifloxacin inhibits the activation of NF-kB and MAP kinases (ERK1/2, p38, and JNK), thereby attenuating the inflammatory response induced by microbial stimuli and inflammatory mediators in various cell types (e.g., respiratory epithelial cells, monocytes) [103,104,105,106]. Moxifloxacin inhibited nitric oxide synthesis and the cytokine-induced activation of NF-κB and MAP kinases in the A549 alveolar epithelial cell line [104]; meanwhile, the expression of inflammatory mediators (IL-6, IL-8) that are dependent on NF-κB- and MAP-kinase and induced by TNF-α was inhibited in cystic fibrosis epithelial cells [106]. The activation of NF-κB and MAP kinases, as well as the release of inflammatory mediators, was also inhibited by moxifloxacin in human monocytes upon bacterial stimuli [103,107].
Quinolones eliminate bacterial cells by increasing the intracellular levels of covalent topoisomerase-cleaved DNA complexes, which serve as intermediates in these enzymes’ DNA strand-passing reactions instead of inhibiting the critical functions of type II topoisomerases. This activity elicits a high number of double-stranded breaks inside the chromosomes of treated bacteria, which induces the SOS (‘Save Our Souls’) response and, eventually, cell death [108]. Riesbeck et al. [109] also demonstrated that ciprofloxacin elicits a stress response in mammals that has a resemblance to the SOS signal response observed in bacteria. This study has shown a strong similarity between the human PBL response to topoisomerase II inhibition and the bacterial SOS response. Thus, the immunomodulatory effects of quinolones were suggested as the result of fluoroquinolones inhibiting topoisomerase II, thereby inducing a stress response in mammals that is comparable to the bacterial quinolones-induced SOS response [110]. Therefore, it is conceivable that cytosolic activation or the inhibition of NF-κB may be influenced by intra-nuclear processes involving the interaction between quinolone and topoisomerase II. A study by Zusso et al. [111] using molecular docking methods provides evidence that FQs inhibit LPS to bond with TLR4-MD-2 complex; hence, the activation of the TLR4/NF-κB signalling pathway is inhibited.
Figure 1 illustrates FQs’ mechanisms and immunomodulatory effects on ARDS (red arrow).

5. Evidence of FQ’s Immunomodulatory Effects Documented in Preclinical and Clinical Studies

A randomised controlled trial (RCT) conducted in Egypt revealed that treatment with 750 mg of levofloxacin once daily for 10 days affected the production of IL-10 as an anti-inflammatory cytokine and TNF-α as a pro-inflammatory cytokine, which may offer additional benefits in the treatment of respiratory tract infections irrespective of their antibacterial properties [112]. A recent in silico study provided evidence that ciprofloxacin and moxifloxacin have a potent ability to bind the main protease (Mpro) of SARS-CoV2, indicating that fluoroquinolone can inhibit SARS-CoV2 replication [113]. According to the current guidelines and literature, fluoroquinolone has an immunomodulatory effect that is clinically beneficial for the treatment of severe pneumonia.
Several studies have reported that FQs can reduce the synthesis of pro-inflammatory cytokines. As demonstrated in an in vitro study, FQs can reduce pro-inflammatory cytokine levels in human peripheral blood mononuclear cells (PBMCs) [114]. Levofloxacin has also been shown to inhibit the secretion of TNF-α, IL-6, and IL-8 by human bronchial epithelial cells [115]. Several other studies suggest that FQs inhibit the production of IL-1 and TNF, which are pro-inflammatory cytokines [86,111,116]. Furthermore, the inhibitory effects of ciprofloxacin and levofloxacin on the NF-κB-mediated microglial inflammatory response have been reported. These effects are attained by the inhibition of lipopolysaccharide (LPS) signalling via TLR4 [107,111].
Moxifloxacin has been shown to effectively decrease the release of IL-8, IL-1b, and TNF-a, which were produced in response to Aspergillus fumigatus infection in human peripheral blood monocytes. The findings of this investigation showed that moxifloxacin inactivates the MAP-kinase ERK1/2, p38 and p65-NF-κB signalling pathways [114]. Ciprofloxacin has been demonstrated to significantly reduce the levels of TNF-α, IL-1β, and CXCL2/MIP-2a and improve the severity of lung damage and overall survival in cases of lung damage induced by LPS [32]. The study by Bailly et al. [94] revealed that both ofloxacin and norfloxacin can inhibit cytokine synthesis. Similar to ciprofloxacin, ofloxacin and grepafloxacin also inhibit the synthesis of IL-1α and IL-1β in LPS-stimulated human peripheral blood lymphocytes (hPBLs).
Another study demonstrated that the activation of TLRs on alveolar cell type II (ATII) in ARDS induces the migration of neutrophils into the epithelium, subsequently leading to the release of toxic mediators, such as proteases and ROS. FQs exhibited antioxidant activity against these conditions and suppressed pneumonia-related pulmonary inflammation [117]. Moxifloxacin has been reported to reduce neutrophil influx and pro-inflammatory cytokines levels, including keratinocytes-derived chemokine (KC), IL-1β, and IL-17A in experimental mice with lung infections induced by Streptococcus pneumoniae and P. aeruginosa [31]. Levofloxacin has also been shown to suppress oxidative and nitrative stress in mice models with ARDS induced by H1N1 influenza. In addition, levofloxacin demonstrated scavenging activity on neutrophil-derived ROS, resulting in a significant reduction in lung injury and an improvement in survival rates [29].
According to recent evidence, the pathogenesis of ARDS may be influenced by several immune cell types, including AMs, as mentioned before. In a healthy state, the fundamental function of AMs in tissue homeostasis is the scavenging and removal of cellular debris and apoptotic cells without inducing an inflammatory response [53]. Three typical quinolone antibiotics, ciprofloxacin, norfloxacin, and pipemidic acid, were investigated for their effects on the polarisation of macrophage RAW264.7 cells in an experimental study conducted by Lang et al. The results suggest that exposure to quinolone at environmentally relevant residual concentrations can lead to the polarisation of macrophages [118].
The immunomodulatory effect of FQ on Th cells was also documented in a study demonstrating that ciprofloxacin induced an immunomodulatory stress response in human T lymphocytes [119]. These cells are essential for humoral and cellular immunity, and the nature of the immune response is regulated by various effector Th cells (Th1, Th2, Th9, Th17, and Th22), which differentiate from naïve T cells in response to antigen stimulation [120]. The activation of T cells is facilitated by the concentration and volume of cytokine secretion in response to infection, leading to the subsequent clearance of the infection [121].
According to Kamiński et al. [122], pre-activated T cells treated with ciprofloxacin exhibited an immunosuppressed phenotype as the result of lower activation-induced ROS production, leading to the reduced expression of IL-2 and IL-4. These findings suggest that ciprofloxacin treatment could have significant implications for the management of inflammatory diseases. Furthermore, several studies in the literature demonstrate that FQs have pharmacodynamic interactions with other drugs, implying that FQs may have immunomodulatory effects, particularly following T-cell activation [123,124,125]. Findings from other studies also suggest that ciprofloxacin, at a dose of more than 20 μg/mL, is capable of counteracting cytokine production inhibition as one of cyclosporine A’s immunosuppressive effects [126]. Additionally, another study on the human leukaemia cell line (HL-60) revealed that levofloxacin-treated cells exhibited an upregulation in the mRNA expression of cytokines and chemokines (e.g., CCL2 and CXCL8) [127].
The administration of gemifloxacin resulted in the inhibition of the immune response in both the 25 mg/kg and 75 mg/kg treatment groups after a 24 h period. The study assessed the effect of gemifloxacin on the humoral component of the immune system at three different doses (25 mg/kg, 50 mg/kg, and 75 mg/kg) using heamagglutination and pneumonia plaque formation assays. The humoral immune response is produced through the synthesis of antibodies that target the specific epitope of the antigen employed to induce the immune response [85].
The summary of the role of FQs as an immunomodulator based on the findings of in vitro, in vivo and ex vivo studies both in humans and animal models is presented in Table 1.

6. Future Perspectives

ALI/ARDS induced by pneumonia is a heterogeneous syndrome with significant variability in pathophysiology, severity, and clinical outcomes. Infection induces an inflammatory response and initiates a complex cytokine network. Heterogeneity contributes to the inconsistent immunomodulatory effects observed in clinical studies. Immunomodulatory effects are likely to occur in the event of severe injury; however, some of the effects identified in preclinical studies are contradicting with clinical observations. A comprehensive understanding of the activation of the inflammatory cascade and the biological phenotype of ARDS patients is required in order to obtain consistent data for performing clinical trials. Phenotypically, the role of fluoroquinolones compared to other antimicrobial agents is considerably well described; however, certain features remain incompletely elucidated at present. Variations in outcomes among studies were associated with the different protocol designs and methodologies used. Although the sequence of events has not been completely understood, significant progress has been made in comprehending the specific mechanisms underlying the immunomodulatory effects of FQs, which involved the kinetic production of cytokines and chemokines, early gene transcription, and the activation or inhibition of transcription factors in cells. Therefore, further comprehensive investigation is necessary to ascertain the precise pathway that ensures FQ interacts with the cellular target topoisomerase II, as well as the subsequent impacts on the transcription mechanism.
In addition, further studies that can effectively identify the clinical characteristics of ARDS and the immunological phenotypes that are likely to respond to immunomodulatory therapy are also needed. Antibiotics administered via aerosol delivery offer higher doses that improve bacterial eradication and lower bacterial resistance. A study suggests that higher concentrations of levofloxacin administered via aerosol may provide immunomodulatory properties independent of its antimicrobial effects; however, further research on in vivo models and in patients is recommended [115].
It should be noted that not every fluoroquinolone demonstrates immunomodulatory properties. The results of several studies reported that differences in time, the frequency of administration and chemical structure may influence it. The development of study models that aim to predict desirable pharmacological properties and facilitate structural modifications of FQs will shape future generations of quinolones. The modification of the chemical structure of FQs may be necessary in order to enhance their capacity to effectively target immune function as immunomodulatory agents in addition to their ability to inhibit bacterial proliferation. The production of FQs should focus on reducing unfavourable features, including developing molecules capable of minimising off-target and drug–drug interactions. On the other side, it is critical to develop novel compounds that are capable of overcoming drug resistance. Drug resistance may be caused by protein post-translational modifications (PTMs), which are enzymatic or chemical reactions that insert covalent groups into the side chains or terminals of amino acids in proteins. Disruptive PTMs can lead to alterations in protein functions and properties that are strongly associated with the development and occurrence of numerous diseases. Targeting PTMs and associated regulatory enzymes may be highly desirable to overcome resistance to FQs and establish therapeutic prospects for various diseases, including CAP.

7. Conclusions

Fluoroquinolones are a class of synthetic antimicrobial agents known for their broad-spectrum activity and have been reported to exhibit immunomodulatory properties. Various pathogens, including both Gram-negative and Gram-positive bacteria, induce cytokine production through different signal transduction pathways, consequently leading to the development of CAP-associated ARDS. In vitro, in vivo, and ex vivo studies have demonstrated the modulation of innate and adaptive immune responses by FQs, which have elucidated the involvement of intracellular signal transduction pathways. According to the evidence demonstrated by these studies, the immunomodulatory activity of FQs was proven to provide indirect antibacterial effects and exhibit anti-inflammatory properties.

Author Contributions

R.Y. conceived the idea and drafted the manuscript; N.F.W. contributed to searching the literature data; R.Y. edited and revised the manuscript; R.Y. and N.F.W. designed figures and tables. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to express our gratitude to the members of the Department of Pulmonology and Respiratory Medicine, Medical Faculty of Airlangga University, for their helpful discussions and contributions. We are also immensely grateful to Tjip S. van der Werf from Department of Lung Diseases and Tuberculosis, University of Groningen, Netherland as our mentor for the insightful feedback, corrections, and suggestions which really impactful in improving the quality of this article. We would also like to show our gratitude to Nimas Roro Gayatri for helping the editing and proofreading processes. Figures were created in Biorender© at the Soetomo General Hospital, Department of Pulmonology and Respiratory Medicine, Surabaya, Indonesia.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ALIAcute lung injury
AMsAlveolar macrophages
AP-1Activator protein-1
ARDSAcute respiratory distress syndrome
ATIIAlveolar cell type II
BALBronchoalveolar lavage
cAMPCyclic adenosine monophosphate
CAPCommunity-acquired pneumonia
cGMPCyclic guanosine monophosphate
CREBcAMP response element-binding protein
CRPC-reactive protein
DAMPsDamage-associated molecular pattern molecules
ERKExtracellular signal-regulated kinases
FQsFluoroquinolones
GM-CSFGranulocyte–macrophage colony-stimulating factor
hPBLHuman peripheral blood lymphocytes
IFNInterferon
ILInterleukin
iNOSInducible NO synthase
IκBInhibitory κappa B
JNKc-Jun N-terminal kinases
KCKeratinocytes-derived chemokine
LPSLipopolysaccharide
MAMPsMicrobe-associated molecular patterns
MAPKMitogen-activated protein kinase
MCP-1Monocyte chemotactic protein
MIP-1αMacrophage inflammatory protein 1α
MproMain protease
NETNeutrophil extracellular trap
NF-κBNuclear factor κappa-B
NONitric oxide
PAMPsPathogen-associated molecular patterns
PBMCsPeripheral blood mononuclear cells
PDEsPhosphodiesterases
PKAProtein kinase A
PRRsPattern recognition receptors
RBCsRed blood cells
RCTRandomised controlled trial
ROSReactive oxygen species
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
TAK1Transforming growth factor-activated kinase 1
TLRToll-like receptor
TNFTumour necrosis factor

References

  1. Rider, A.C.; Frazee, B.W. Community-Acquired Pneumonia. Emerg. Med. Clin. N. Am. 2018, 36, 665–683. [Google Scholar] [CrossRef] [PubMed]
  2. Alshammari, M.K.; Alotaibi, M.A.; Alotaibi, A.S.; Alosaime, H.T.; Aljuaid, M.A.; Alshehri, B.M.; AlOtaibi, Y.B.; Alasmari, A.A.; Alasmari, G.A.; Mohammed, M.H.; et al. Prevalence and Etiology of Community- and Hospital-Acquired Pneumonia in Saudi Arabia and Their Antimicrobial Susceptibility Patterns: A Systematic Review. Medicina 2023, 59, 760. [Google Scholar] [CrossRef] [PubMed]
  3. Regunath, H.; Oba, Y. Community-Acquired Pneumonia. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK430749/ (accessed on 25 May 2023).
  4. Cillóniz, C.; Dominedò, C.; Torres, A. Multidrug Resistant Gram-Negative Bacteria in Community-Acquired Pneumonia. Crit. Care 2019, 23, 79. [Google Scholar] [CrossRef] [PubMed]
  5. Kishimbo, P.; Sogone, N.M.; Kalokola, F.; Mshana, S.E. Prevalence of gram negative bacteria causing community acquired pneumonia among adults in Mwanza City, Tanzania. Pneumonia 2020, 12, 7. [Google Scholar] [CrossRef] [PubMed]
  6. Morris, D.E.; Cleary, D.W.; Clarke, S.C. Secondary bacterial infections associated with influenza pandemics. Front. Microbiol. 2017, 8, 1041. [Google Scholar] [CrossRef] [PubMed]
  7. Wang, H.; Anthony, D.; Selemidis, S.; Vlahos, R.; Bozinovski, S. Resolving viral-induced secondary bacterial infection in COPD: A concise review. Front. Immunol. 2018, 9, 2345. [Google Scholar] [CrossRef] [PubMed]
  8. Oliva, J.; Terrier, O. Viral and Bacterial Co-Infections in the Lungs: Dangerous Liaisons. Viruses 2021, 13, 1725. [Google Scholar] [CrossRef] [PubMed]
  9. Kuek, L.E.; Lee, R.J. First contact: The role of respiratory cilia in host-pathogen interactions in the airways. Am. J. Physiol. Cell. Mol. Physiol. 2020, 319, L603–L619. [Google Scholar] [CrossRef] [PubMed]
  10. Avadhanula, V.; Wang, Y.; Portner, A.; Adderson, E. Nontypeable Haemophilus influenzae and Streptococcus pneumoniae bind respiratory syncytial virus glycoprotein. J. Med. Microbiol. 2007, 56, 1133–1137. [Google Scholar] [CrossRef]
  11. Long, M.E.; Mallampalli, R.K.; Horowitz, J.C. Pathogenesis of Pneumonia and Acute Lung Injury. Clin. Sci. 2022, 136, 747–769. [Google Scholar] [CrossRef]
  12. Odeyemi, Y.; Moraes, A.G.D.; Gajic, O. What factors predispose patients to acute respiratory distress syndrome? Evid.-Based Pract. Crit. Care 2020, 103–108.e1. [Google Scholar] [CrossRef]
  13. Bellani, G.; Laffey, J.G.; Pham, T.; Fan, E.; Brochard, L.; Esteban, A.; Gattinoni, L.; van Haren, F.; Larsson, A.; McAuley, D.F.; et al. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA 2016, 315, 788–800. [Google Scholar] [CrossRef] [PubMed]
  14. Hsu, C.Y.; Lai, C.C.; Yeh, Y.P.; Chang-Chuan, C.; Chen, H.H. Progression from Pneumonia to ARDS as a Predictor for Fatal COVID-19. J. Infect. Public Health 2021, 14, 504–507. [Google Scholar] [CrossRef] [PubMed]
  15. Kumar, V.; Chhibber, S. Acute lung inflammation in Klebsiella pneumonia B5055-induced pneumonia and sepsis in BALB/c mice: A comparative study. Inflammation 2011, 34, 452–462. [Google Scholar] [CrossRef] [PubMed]
  16. Guirgis, F.W.; Khadpe, J.D.; Kuntz, G.M.; Wears, R.L.; Kalynych, C.J.; Jones, A.E. Persistent organ dysfunction after severe sepsis: A systematic review. J. Crit. Care 2014, 29, 320–326. [Google Scholar] [CrossRef] [PubMed]
  17. Kumar, V. Pulmonary Innate Immune Response Determines the Outcome of Inflammation During Pneumonia and Sepsis-Associated Acute Lung Injury. Front. Immunol. 2020, 11, 1722. [Google Scholar] [CrossRef] [PubMed]
  18. Torres, A.; Chalmers, J.D.; Cruz, C.S.D.; Dominedò, C.; Kollef, M.; Martin-Loeches, I.; Niederman, M.; Wunderink, R.G. Challenges in severe community-acquired pneumonia: A point-of-view review. Intensive Care Med. 2019, 45, 159–171. [Google Scholar] [CrossRef] [PubMed]
  19. Torres, A.; Sibila, O.; Ferrer, M.; Polverino, E.; Menendez, R.; Mensa, J.; Gabarrus, A.; Sellares, J.; Restrepo, M.I.; Anzueto, A.; et al. Effect of corticosteroids on treatment failure among hospitalized patients with severe community-acquired pneumonia and high inflammatory response: A randomized clinical trial. JAMA 2015, 313, 677–686. [Google Scholar] [CrossRef] [PubMed]
  20. Jiang, S.; Liu, T.; Hu, Y.; Li, R.; Di, X.; Jin, X.; Wang, Y.; Wang, K. Efficacy and safety of glucocorticoids in the treatment of severe community-acquired pneumonia: A meta-analysis. Medicine 2019, 98, e16239. [Google Scholar] [CrossRef] [PubMed]
  21. Harris, L.K.; Crannage, A.J. Corticosteroids in Community-Acquired Pneumonia: A Review of Current Literature. J. Pharm. Technol. 2021, 37, 152–160. [Google Scholar] [CrossRef]
  22. Rodrigo, C.; Leonardi-Bee, J.; Nguyen-Van-Tam, J.; Lim, W.S. Corticosteroids as adjunctive therapy in the treatment of influenza. Cochrane Database Syst. Rev. 2016, 3, Cd010406. [Google Scholar] [CrossRef]
  23. Mc Mahon, A.; Martin-Loeches, I. The pharmacological management of severe influenza infection—‘existing and emerging therapies’. Expert. Rev. Clin. Pharmacol. 2017, 10, 81–95. [Google Scholar] [CrossRef] [PubMed]
  24. Akinjogunla, O.J.; Odeyemi, A.T.; Alozie, M.F.; Ehinmore, I.; Ukpong, U.E.; Ediomo, J.; Akpanson, E.K. Fluoroquinolone antibiotics: In vitro antibacterial and time-kill bazhaNEctericidal evaluation against etiology of bacteremia in human immunodeficiency virus (HIV)-infected patients. Bull. Natl. Res. Cent. 2022, 46, 135. [Google Scholar] [CrossRef]
  25. Sauer, A.; Peukert, K.; Putensen, C.; Bode, C. Antibiotics as immunomodulators: A potential pharmacologic approach for ARDS treatment. Eur. Respir. Rev. 2021, 30, 210093. [Google Scholar] [CrossRef] [PubMed]
  26. Brar, R.K.; Jyoti, U.; Patil, R.K.; Patil, H.C. Fluoroquinolone antibiotics: An overview. Adesh Univ. J. Med. Sci. Res. 2020, 2, 26–30. [Google Scholar] [CrossRef]
  27. Metlay, J.P.; Waterer, G.W.; Long, A.C.; Anzueto, A.; Brozek, J.; Crothers, K.; Cooley, L.A.; Dean, N.C.; Fine, M.J.; Flanders, S.A.; et al. Diagnosis and Treatment of Adults with Community-acquired Pneumonia. An Official Clinical Practice Guideline of the American Thoracic Society and Infectious Diseases Society of America. Am. J. Respir. Crit. Care Med. 2019, 200, e45–e67. [Google Scholar] [CrossRef]
  28. Akamatsu, H.; Niwa, Y.; Sasaki, H.; Matoba, Y.; Asada, Y.; Horio, T. Effect of pyridone carboxylic acid anti-microbials on the generation of reactive oxygen species in vitro. J. Int. Med. Res. 1996, 24, 345–351. [Google Scholar] [CrossRef] [PubMed]
  29. Enoki, Y.; Ishima, Y.; Tanaka, R.; Sato, K.; Kimachi, K.; Shirai, T.; Watanabe, H.; Chuang, V.T.; Fujiwara, Y.; Takeya, M.; et al. Pleiotropic effects of levofloxacin, fluoroquinolone antibiotics, against influenza virus-induced lung injury. PLoS ONE 2015, 10, e0130248. [Google Scholar] [CrossRef] [PubMed]
  30. Chou, C.C.; Shen, C.F.; Chen, S.J.; Chen, H.M.; Wang, Y.C.; Chang, W.S.; Chang, Y.T.; Chen, W.Y.; Huang, C.Y.; Kuo, C.C.; et al. Recommendations and guidelines for the treatment of pneumonia in Taiwan. J. Microbiol. Immunol. Infect. 2019, 52, 172–199. [Google Scholar] [CrossRef]
  31. Beisswenger, C.; Honecker, A.; Kamyschnikow, A.; Bischoff, M.; Tschernig, T.; Bals, R. Moxifloxacin modulates inflammation during murine pneumonia. Respir. Res. 2014, 15, 82. [Google Scholar] [CrossRef]
  32. Huang, H.C.; Shieh, C.C.; Yu, W.L.; Cheng, K.C.; Chen, C.C.; Chang, S.T.; Chuang, Y.C. Comparing the protective effects of ciprofloxacin, moxifloxacin and levofloxacin in mice with lipopolysaccharide-induced acute lung injuries. Respirology 2008, 13, 47–52. [Google Scholar] [CrossRef]
  33. Shalit, I.; Horev-Azaria, L.; Fabian, I.; Blau, H.; Kariv, N.; Shechtman, I.; Alteraz, H.; Kletter, Y. Immunomodulatory and protective effects of moxifloxacin against Candida albicans-induced bronchopneumonia in mice injected with cyclophosphamide. Antimicrob. Agents Chemother. 2002, 46, 2442–2449. [Google Scholar] [CrossRef] [PubMed]
  34. Mantero, M.; Tarsia, P.; Gramegna, A.; Henchi, S.; Vanoni, N.; Di Pasquale, M. Antibiotic therapy, supportive treatment and management of immunomodulationinflammation response in community acquired pneumonia: Review of recommendations. Multidiscip. Respir. Med. 2017, 12, 26. [Google Scholar] [CrossRef]
  35. Lee, K.Y. Pneumonia, Acute Respiratory Distress Syndrome, and Early Immune-Modulator Therapy. Int. J. Mol. Sci. 2017, 18, 388. [Google Scholar] [CrossRef] [PubMed]
  36. Zemans, R.L.; Matthay, M.A. What drives neutrophils to the alveoli in ARDS? Thorax 2017, 72, 1–3. [Google Scholar] [CrossRef]
  37. Bozoky, G.; Eva, R. Community-acquired pneumonia as a cause of sepsis. Trends Med. 2019, 19, 1–4. [Google Scholar] [CrossRef]
  38. Feldman, C.; Anderson, R. Community-Acquired Pneumonia Pathogenesis of Acute Cardiac Events and Potential Adjunctive Therapies. Chest 2015, 148, 523–532. [Google Scholar] [CrossRef] [PubMed]
  39. Paul, A.; Edwards, J.; Pepper, C.; Mackay, S. Inhibitory-κB Kinase (IKK) α and Nuclear Factor-κB (NFκB)-Inducing Kinase (NIK) as Anti-Cancer Drug Targets. Cells 2018, 7, 176. [Google Scholar] [CrossRef]
  40. Sun, S.C. The non-canonical NF-κB pathway in immunity and inflammation. Nat. Rev. Immunol. 2017, 17, 545–558. [Google Scholar] [CrossRef] [PubMed]
  41. Dalhoff, A.; Shalit, I. Immunomodulatory effects of quinolones. Lancet Infect. Dis. 2003, 3, 359–371. [Google Scholar] [CrossRef]
  42. Ghosh, S.; Karin, M. Missing pieces in the NF-kappaB puzzle. Cell 2002, 109, S81–S96. [Google Scholar] [CrossRef]
  43. Nagafuji, T.; Matsumoto, T.; Takahashi, K.; Kubo, S.; Haraoka, M.; Tanaka, M.; Ogata, N.; Kumazawa, J. Enhancement of superoxide production of polymorphonuclear neutrophils by ofloxacin and the effects of inhibitors of protein kinase C. Chemotherapy 1993, 39, 70–76. [Google Scholar] [CrossRef] [PubMed]
  44. Kosyreva, A.; Dzhalilova, D.; Lokhonina, A.; Vishnyakova, P.; Fatkhudinov, T. The Role of Macrophages in the Pathogenesis of SARS-CoV-2-Associated Acute Respiratory Distress Syndrome. Front. Immunol. 2021, 12, 682871. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, J.; Dean, D.A. Gene Therapy for Acute Respiratory Distress Syndrome. Front. Physiol. 2022, 12, 786255. [Google Scholar] [CrossRef] [PubMed]
  46. Kyo, M.; Nishioka, K.; Nakaya, T.; Kida, Y.; Tanabe, Y.; Ohshimo, S.; Shime, N. Unique patterns of lower respiratory microbiota are associated with inflammation and hospital mortality acute respiratory distress syndrome. Respir. Res. 2019, 20, 246. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, A.; Al-Kuhlani, M.; Johnston, S.C.; Ojcius, D.M.; Chou, J.; Dean, D. Transcription factor complex AP-1 mediates inflammation initiated by chlamydia pneumoniae infection. Cell. Microbiol. 2013, 15, 779–794. [Google Scholar] [CrossRef] [PubMed]
  48. Yang, Y.; Lv, J.; Jiang, S.; Ma, Z.; Wang, D.; Hu, W.; Deng, C.; Fan, C.; Di, S.; Sun, Y.; et al. The emerging role of toll-like receptor 4 in myocardial inflammation. Cell Death Dis. 2016, 7, e2234. [Google Scholar] [CrossRef]
  49. Chen, Z.; Hua, S. Transcription factor-mediated signaling pathways’ contribution to the pathology of acute lung injury and acute respiratory distress syndrome. Am. J. Transl. Res. 2020, 12, 5608–5618. [Google Scholar] [PubMed]
  50. Cuenda, A.; Rousseau, S. p38 MAP-kinases pathway regulation, function and role in human diseases. Biochim. Biophys. Acta 2007, 1773, 1358–1375. [Google Scholar] [CrossRef] [PubMed]
  51. Feng, Y.; Fang, Z.; Liu, B.; Zheng, X. p38MAPK plays a pivotal role in the development of acute respiratory distress syndrome. Clinics 2019, 74, e509. [Google Scholar] [CrossRef]
  52. Steel, H.C.; Cockeran, R.; Anderson, R.; Feldman, C. Overview of Community-Acquired Pneumonia and the Role of Inflammatory Mechanisms in the Immunopathogenesis of Severe Pneumococcal Disease. Mediat. Inflamm. 2013, 2013, 490346. [Google Scholar] [CrossRef]
  53. Huppert, L.A.; Matthay, M.A.; Ware, L.B. Pathogenesis of Acute Respiratory Distress Syndrome. Semin. Respir. Crit. Care Med. 2019, 40, 31–39. [Google Scholar] [CrossRef] [PubMed]
  54. Wong, J.J.M.; Leong, J.Y.; Lee, J.H.; Albani, S.; Yeo, J.G. Insights into the immuno-pathogenesis of acute respiratory distress syndrome. Ann. Transl. Med. 2019, 7, 504. [Google Scholar] [CrossRef] [PubMed]
  55. Thiam, H.R.; Wong, S.L.; Wagner, D.D.; Waterman, C.M. Cellular mechanisms of NETosis. Annu. Rev. Cell Dev. Biol. 2020, 36, 191–218. [Google Scholar] [CrossRef] [PubMed]
  56. Matthay, M.A.; Zemans, R.L.; Zimmerman, G.A.; Arabi, Y.M.; Beitler, J.R.; Mercat, A.; Herridge, M.; Randolph, A.G.; Calfee, C.S. Acute respiratory distress syndrome. Nat. Rev. Dis. Primers 2019, 5, 18. [Google Scholar] [CrossRef] [PubMed]
  57. Ruiz, J. Transferable mechanisms of quinolone resistance from 1998 onward. Clin. Microbiol. Rev. 2019, 32, e00007-19. [Google Scholar] [CrossRef]
  58. Hu, X.E.; Kim, N.K.; Gray, J.L.; Almstead, J.I.K.; Seibel, W.L.; Ledoussal, B. Discovery of (3S)-Amino-(4R)-ethylpiperidinyl quinolones as potent antibacterial agents with a broad spectrum of activity against resistant pathogens. J. Med. Chem. 2003, 46, 3655–3661. [Google Scholar] [CrossRef] [PubMed]
  59. Andersson, M.I.; MacGowan, A.P. Development of the quinolones. J. Antimicrob. Chemother. 2003, 51 (Suppl. 1), 1–11. [Google Scholar] [CrossRef] [PubMed]
  60. Yadav, V.; Talwar, P. Repositioning of fluoroquinolones from antibiotic to anti-cancer agents: An underestimated truth. Biomed. Pharmacother. 2019, 111, 934–946. [Google Scholar] [CrossRef] [PubMed]
  61. Paton, J.H.; Reeves, D.S. Fluoroquinolone antibiotics. Microbiology, pharmacokinetics and clinical use. Drugs 1988, 36, 193–228. [Google Scholar] [CrossRef]
  62. Pham, T.D.M.; Ziora, Z.M.; Blaskovich, M.A.T. Quinolone antibiotics. Med. Chem. Commun. 2019, 10, 1719–1739. [Google Scholar] [CrossRef]
  63. Redgrave, L.S.; Sutton, S.B.; Webberm, M.A.; Piddock, L.J. Fluoroquinolone resistance: Mechanisms, impact on bacteria, and role in evolutionary success. Trends Microbiol. 2014, 22, 438–445. [Google Scholar] [CrossRef] [PubMed]
  64. Perry, C.M.; Balfour, J.A.B.; Lamb, H.M. Gatifloxacin. Drugs 1999, 58, 683–696. [Google Scholar] [CrossRef]
  65. Millanao, A.R.; Mora, A.Y.; Villagra, N.A.; Bucarey, S.A.; Hidalgo, A.A. Biological Effects of Quinolones: A Family of Broad-Spectrum Antimicrobial Agents. Molecules 2021, 26, 7153. [Google Scholar] [CrossRef] [PubMed]
  66. Tartaglione, T.A.; Hooton, T.M. The role of fluoroquinolones in sexually transmitted diseases. Pharmacotherpay 1993, 13, 189–201. [Google Scholar] [CrossRef]
  67. Gatti, M.; Bianchin, M.; Raschi, E.; De Ponti, F. Assessing the Association between Fluoroquinolones and Emerging Adverse Drug Reactions Raised by Regulatory Agencies: An Umbrella Review. Eur. J. Intern. Med. 2020, 75, 60–70. [Google Scholar] [CrossRef]
  68. Roberts, J.R. InFocus: Fluoroquinolone Side Effects Just Got Scarier. Emerg. Med. News 2018, 40, 26–27. [Google Scholar] [CrossRef]
  69. Anderson, V.E.; Osheroff, N. Type II topoisomerases as targets for quinolone antibacterials: Turning Dr. Jekyll into Mr. Hyde. Curr. Pharm. Des. 2001, 7, 337–353. [Google Scholar] [CrossRef]
  70. Stein, G.E.; Goldstein, E.J.C. Fluoroquinolones and anaerobes. Clin. Infect. Dis. 2006, 42, 1598–1607. [Google Scholar] [CrossRef]
  71. El-Rayes, B.F.; Grignon, R.; Aslam, N.; Aranha, O.; Sarkar, F.H. Ciprofloxacin inhibits cell growth and synergises the effect of etoposide in hormone resistant prostate cancer cells. Int. J. Oncol. 2002, 21, 207–211. [Google Scholar] [CrossRef]
  72. Noris, M.D.; Madafiglio, J.; Gilbert, J.; Marshall, G.M.; Haber, M. Reversal of multidrug resistance-associated protein-mediated drug resistance in cultured human neuroblastoma cells by the quinolone antibiotic difloxacin. Med. Pediatr. Oncol. 2001, 36, 177–180. [Google Scholar] [CrossRef]
  73. Herold, C.; Ocker, M.; Ganslmayer, M.; Gerauer, H.; Hahn, E.G.; Schuppan, D. Ciprofloxacin induces apoptosis and inhibits proliferation of human colorectal carcinoma cells. Br. J. Cancer 2002, 86, 443–448. [Google Scholar] [CrossRef] [PubMed]
  74. Shalit, I.; Kletter, Y.; Weiss, K.; Gruss, T.; Fabian, I. Enhanced hematopoiesis in sublethally irradiated mice treated with various quinolones. Eur. J. Haematol. 1997, 58, 92–98. [Google Scholar] [CrossRef] [PubMed]
  75. De Sarro, A.; De Sarro, G. Adverse reactions to fluoroquinolones. an overview on mechanistic aspects. Curr. Med. Chem. 2001, 8, 371–384. [Google Scholar] [CrossRef] [PubMed]
  76. Riesbeck, K. Immunomodulating activity of quinolones: Review. J. Chemother. 2002, 14, 3–12. [Google Scholar] [CrossRef] [PubMed]
  77. Müller-Redetzky, H.; Lienau, J.; Suttorp, N.; Witzenrath, M. Therapeutic strategies in pneumonia: Going beyond antibiotics. Eur. Respir. Rev. 2015, 24, 516–524. [Google Scholar] [CrossRef] [PubMed]
  78. Dalhoff, A. Immunomodulatory activities of fluoroquinolones. Infection 2005, 33 (Suppl. 2), 55–70. [Google Scholar] [CrossRef] [PubMed]
  79. Zhanel, G.G.; Walkty, A.; Vercaigne, L.; Karlowsky, J.A.; Embil, J.; Gin, A.S.; Hoban, D.J. The new fluoroquinolones: A critical review. Can. J. Infect. Dis. 1999, 10, 207–238. [Google Scholar] [CrossRef] [PubMed]
  80. Chu, D.T.W.; Fernandes, P.B. Structure–activity relationships of the fluoroquinolones. Antimicrob. Agents Chemother. 1989, 33, 131–135. [Google Scholar] [CrossRef]
  81. Domagala, J.M. Structure–activity and structure–side-effect relationships for the quinolone antibacterials. J. Antimicrob. Chemother. 1994, 33, 685–706. [Google Scholar] [CrossRef]
  82. DeSimone, C.; Bandinelli, L.; Ferrazzi, M.; De Santis, S.; Pugnaloni, L.; Sorice, F. Influence of ofloxacin, norfloxacin, malidixic acid, pyromidic acid and pipemidic acid on human gamma-interferon production and blastogenesis. J. Antimicrob. Chemother. 1986, 17, 811–814. [Google Scholar] [CrossRef]
  83. Zehavi-Wilner, T.; Shalit, I. Enhancement of interleukin-2 production in human lymphocytes by two new quinolone derivatives. Lymph. Res. 1989, 8, 35–46. [Google Scholar]
  84. Yamashita, Y.; Ashizawa, T.; Morimoto, M.; Hosomi, J.; Makano, H. Antitumor quinolones with mammalian topoisomerase II mediated DNA cleavage activity. Cancer Res. 1992, 52, 2818–2822. [Google Scholar]
  85. Umair, M.; Javeed, A.; Ghafoor, A.; Ashraf, M. Immunomodulatory activities of gemifloxacin in mice. Iran. J. Basic Med. Sci. 2016, 19, 985–992. [Google Scholar]
  86. Ogino, H.; Fujii, M.; Ono, M.; Maezawa, K.; Hori, S.; Kizu, J. In Vivo and in Vitro Effects of Fluoroquinolones on Lipopolysaccharide-Induced pro-Inflammatory Cytokine Production. J. Infect. Chemother. 2009, 15, 168–173. [Google Scholar] [CrossRef]
  87. Zhang, J.Z.; Ward, K.W. Besifloxacin, a Novel Fluoroquinolone Antimicrobial Agent, Exhibits Potent Inhibition of pro-inflammatory Cytokines in Human THP-1 Monocytes. J. Antimicrob. Chemother. 2008, 61, 111–116. [Google Scholar] [CrossRef] [PubMed]
  88. Raker, V.K.; Becker, C.; Esteinbrink, K. The cAMP Pathway as Therapeutic Target in Autoimmune and Inflammatory Diseases. Front. Immunol. 2016, 7, 123. [Google Scholar] [CrossRef] [PubMed]
  89. Wang, J.P.; Raung, S.L.; Huang, L.J.; Kuo, S.-C. Involvement of cyclic AMP generation in the inhibition of respiratory burst by 2-phenyl-4-quinolone (YT-1) in rat neutrophils. Biochem. Pharmacol. 1998, 56, 1505–1514. [Google Scholar] [CrossRef]
  90. Stockley, R.A.; Rennard, S.I.; Rabe, K.; Celli, B. Chronic Obstructive Pulmonary Disease; John Wiley & Sons: Hoboken, NJ, USA, 2008; p. 206. [Google Scholar]
  91. Tsai, Y.F.; Chen, C.Y.; Yang, S.C.; Syu, Y.T.; Hwang, T.L. Apremilast ameliorates acute respiratory distress syndrome by inhibiting neutrophil-induced oxidative stress. Biomed. J. 2023, 46, 100560. [Google Scholar] [CrossRef]
  92. Mokra, D.; Mokry, J. Phosphodiesterase Inhibitors in Acute Lung Injury: What Are the Perspectives? Int. J. Mol. Sci. 2021, 22, 1929. [Google Scholar] [CrossRef]
  93. Wells, J.M.; Jackson, P.L.; Viera, L.; Bhatt, S.P.; Gautney, J.; Handley, G.; King, R.W.; Xu, X.; Gaggar, A.; Bailey, W.C.; et al. A Randomized, Placebo-controlled Trial of Roflumilast. Effect on Proline-Glycine-Proline and Neutrophilic Inflammation in Chronic Obstructive Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2015, 192, 934–942. [Google Scholar] [CrossRef]
  94. Bailly, S.; Fay, M.; Gougerot-Pocidalo, J.J. Effects of quinolones on tumor necrosis factor production by human monocytes. Int. J. Immunopharmacol. 1990, 12, 31–36. [Google Scholar] [CrossRef]
  95. Shames, B.D.; McIntyre, R.C., Jr.; Bensard, D.D.; Pulido, E.J.; Selzman, C.H.; Reznikov, L.L.; Harken, A.H.; Meng, X. Suppression of tumor necrosis factor alpha production by cAMP in human monocytes: Dissociation with mRNA level and independent of interleukin-10. J. Surg. Res. 2001, 99, 187–193. [Google Scholar] [CrossRef]
  96. Ghosh, M.; Garcia-Castillo, D.; Aguirre, V.; Golshani, R.; Atkins, C.M.; Bramlett, H.M.; Dietrich, W.D.; Pearse, D.D. Proinflammatory cytokine regulation of cyclic AMP-phosphodiesterase 4 signaling in microglia in vitro and following CNS injury. Glia 2012, 60, 1839–1859. [Google Scholar] [CrossRef] [PubMed]
  97. Blaine, T.A.; Police, P.F.; Rosier, R.N.; Reynolds, P.R.; Puzas, J.E.; O’Keefe, R.J. Modulation of the production of cytokines in titanium-stimulated human peripheral blood monocytes by pharmacological agents. The role of cAMP-mediated signaling mechanisms. J. Bone Jt. Surg. 1997, 79, 1519–1528. [Google Scholar] [CrossRef]
  98. Naqvi, S.; Martin, K.J.; Arthur, J.S. CREB phosphorylation at Ser133 regulates transcription via distinct mechanisms downstream of cAMP and MAPK signalling. Biochem. J. 2014, 458, 469–479. [Google Scholar] [CrossRef] [PubMed]
  99. Santibáñez, J.F.; Olivares, D.; Guerrero, J.; Martínez, J. Cyclic AMP inhibits TGFβ1-induced cell-scattering and invasiveness in murine-transformed keratinocytes. Int. J. Cancer 2003, 107, 715–720. [Google Scholar] [CrossRef]
  100. Lee, S.Y.; Kim, H.J.; Lee, W.J.; Joo, S.H.; Jeon, S.-J.; Kim, J.W.; Han, S.-H.; Lee, J.; Park, S.H.; Cheong, J.H.; et al. Differential Regulation of Matrix Metalloproteinase-9 and Tissue Plasminogen Activator Activity by the Cyclic-AMP System in Lipopolysaccharide-stimulated Rat Primary Astrocytes. Neurochem. Res. 2008, 33, 2324–2334. [Google Scholar] [CrossRef]
  101. Perez, C.; Vilaboa, N.E.; Garcia-Bermejo, L.; de-Blas, E.; Creighton, A.M.; Aller, P. Differentiation of U-937 promonocytic cells by etoposide and ICRF-193, two antitumour DNA topoisomerase inhibitors with different mechanisms of action. J. Cell Sci. 1997, 110, 337–343. [Google Scholar] [CrossRef]
  102. Testolin, L.; Carson, C.; Wang, Y.; Walker, P.R.; Armato, U.; Sikorska, M. Jun and JNK kinase are activated in thymocytes in response to VM26 and radiation but not glucocorticoids. Exp. Cell Res. 1997, 230, 220–232. [Google Scholar] [CrossRef] [PubMed]
  103. Weiss, T.; Shalit, I.; Blau, H.; Werber, S.; Halperin, D.; Levitov, A.; Fabian, I. Anti-inflammatory effects of moxifloxacin on activated human monocytic cells: Inhibition of NF-kappaB and mitogen-activated protein kinase activation and of synthesis of proinflammatory cytokines. Antimicrob. Agents Chemother. 2004, 48, 1974–1982. [Google Scholar] [CrossRef]
  104. Werber, S.; Shalit, I.; Fabian, I.; Steuer, G.; Weiss, T.; Blau, H. Moxifloxacin inhibits cytokine-induced MAP kinase and NF-kappaB activation as well as nitric oxide synthesis in a human respiratory epithelial cell line. J. Antimicrob. Chemother. 2005, 55, 293–300. [Google Scholar] [CrossRef] [PubMed]
  105. Ganster, R.W.; Taylor, B.S.; Shao, L.; Geller, D.A. Complex regulation of human inducible nitric oxide synthase gene transcription by Stat 1 and NF-κB. Proc. Natl. Acad. Sci. USA 2001, 98, 8638–8643. [Google Scholar] [CrossRef]
  106. Blau, H.; Klein, K.; Shalit, I.; Halperin, D.; Fabian, I. Moxifloxacin but not ciprofloxacin or azithromycin selectively inhibits IL-8, IL-6, ERK1/2, JNK, and NF-kappaB activation in a cystic fibrosis epithelial cell line. Am. J. Physiol. Lung Cell. Mol. Physiol. 2007, 292, L343–L352. [Google Scholar] [CrossRef] [PubMed]
  107. Araujo, F.G.; Slifer, T.L.; Remington, J.S. Effect of moxifloxacin on secretion of cytokines by human monocytes stimulated with lipopolysaccharide. Clin. Microbiol. Infect. 2002, 8, 26–30. [Google Scholar] [CrossRef] [PubMed]
  108. Anderson, V.E.; Zaniewski, R.P.; Kaczmarek, F.S.; Gootz, T.D.; Osheroff, N. Quinolones inhibit DNA religation mediated by Staphylococcus aureus topoisomerase IV. Changes in drug mechanism across evolutionary boundaries. J. Biol. Chem. 1999, 274, 35927–35932. [Google Scholar] [CrossRef]
  109. Riesbeck, K.; Forsgren, A.; Henriksson, A.; Bredberg, A. Ciprofloxacin induces an immunomodulatory stress response in human T lymphocytes. Antimicrob. Agents Chemother. 1998, 8, 1923–1930. [Google Scholar] [CrossRef] [PubMed]
  110. Serebryakova, V.A.; Urazova, O.I.; Novitsky, V.V.; Vengerovskii, A.I.; Kononova, T.E. In Vitro Study of the Modulatory Effects of Levofloxacin and BCG on Secretion of Proinflammatory Cytokines in Infiltrative Pulmonary Tuberculosis. Bull. Exp. Biol. Med. 2018, 166, 182–185. [Google Scholar] [CrossRef]
  111. Zusso, M.; Lunardi, V.; Franceschini, D.; Pagetta, A.; Lo, R.; Stifani, S.; Frigo, A.C.; Giusti, P.; Moro, S. Ciprofloxacin and levofloxacin attenuate microglia inflammatory response via TLR4/NF-kB pathway. J. Neuroinflamm. 2019, 16, 148. [Google Scholar] [CrossRef] [PubMed]
  112. Badari, M.S.; Elgendy, S.G.; Mohamed, A.S.; Hassan, A.T. Immunomodulatory Effects of Levofloxacin on Patients with Pneumonia in Assiut University Hospitals. Egypt. J. Immunol. 2015, 22, 79–85. [Google Scholar]
  113. Marciniec, K.; Beberok, A.; Pęcak, P.; Boryczka, S.; Wrześniok, D. Ciprofloxacin and moxifloxacin could interact with SARS-CoV-2 protease: Preliminary in silico analysis. Pharmacol. Rep. 2020, 72, 1553–1561. [Google Scholar] [CrossRef]
  114. Shalit, I.; Halperin, D.; Haite, D.; Levitov, A.; Romano, J.; Osherov, N.; Fabian, I. Anti-inflammatory effects of moxifloxacin on IL-8, IL-1β and TNF-α secretion and NFκB and MAP-kinase activation in human monocytes stimulated with Aspergillus fumigatus. J. Antimicrob. Chemother. 2006, 57, 230–235. [Google Scholar] [CrossRef] [PubMed]
  115. Tsivkovskii, R.; Sabet, M.; Tarazi, Z.; Griffith, D.C.; Lomovskaya, O.; Dudley, M.N. Levofloxacin reduces inflammatory cytokine levels in human bronchial epithelia cells: Implications for aerosol MP-376 (levofloxacin solution for inhalation) treatment of chronic pulmonary infections. FEMS Immunol. Med. Microbiol. 2011, 61, 141–146. [Google Scholar] [CrossRef]
  116. Khan, A.A.; Slifer, T.R.; Araujo, F.G.; Suzuki, Y.; Remington, J.S. Protection against lipopolysaccharide-induced death by fluoroquinolones. Antimicrob. Agents Chemother. 2000, 44, 3169–3173. [Google Scholar] [CrossRef]
  117. Vlahos, R.; Stambas, J.; Selemidis, S. Suppressing production of reactive oxygen species (ROS) for influenza A virus therapy. Trends Pharmacol. Sci. 2012, 33, 3–8. [Google Scholar] [CrossRef]
  118. Lang, L.; Zhang, Y.; Yang, A.; Dong, J.; Li, W.; Zhang, G. Macrophage polarization induced by quinolone antibiotics at environmental residue level. Int. Immunopharmacol. 2022, 106, 108596. [Google Scholar] [CrossRef]
  119. Fukumoto, R.; Cary, L.H.; Gorbunov, N.V.; Lombardini, E.D.; Elliott, T.B.; Kiang, J.G. Ciprofloxacin modulates cytokine/chemokine profile in serum, improves bone marrow repopulation, and limits apoptosis and autophagy in ileum after whole body ionizing irradiation combined with skin-wound trauma. PLoS ONE 2013, 8, e58389. [Google Scholar] [CrossRef] [PubMed]
  120. Mosmann, T.R.; Sad, S. The expanding universe of T-cell subsets: Th1, Th2 and more. Immunol. Today 1996, 3, 138–146. [Google Scholar] [CrossRef]
  121. Assar, S.; Nosratabadi, R.; Azad, H.K.; Masoumi, J.; Mohamadi, M.; Hassanshahi, G. A Review of Immunomodulatory Effects of Fluoroquinolones. Immunol. Investig. 2020, 50, 1007–1026. [Google Scholar] [CrossRef] [PubMed]
  122. Kamiński, M.M.; Sauer, S.W.; Klemke, C.-D.; Süss, D.; Okun, J.G.; Krammer, P.H.; Gülow, K. Mitochondrial reactive oxygen species control T cell activation by regulating IL-2 and IL-4 expression: Mechanism of ciprofloxacin-mediated immunosuppression. J. Immunol. 2010, 184, 4827–4841. [Google Scholar] [CrossRef] [PubMed]
  123. Wrishko, R.E.; Levine, M.; Primmett, D.R.; Kim, S.; Partovi, N.; Lewis, S.; Landsberg, D.; Keown, P.A. Investigation of a possible interaction between ciprofloxacin and cyclosporine in renal transplant patients. Transplantation 1997, 7, 996–999. [Google Scholar] [CrossRef]
  124. Federico, S.; Carrano, R.; Capone, D.; Gentile, A.; Palmiero, G.; Basile, V. Pharmacokinetic Interaction between Levofloxacin and Ciclosporin or Tacrolimus in Kidney Transplant Recipients. Clin. Pharmacokinet. 2006, 45, 169–175. [Google Scholar] [CrossRef] [PubMed]
  125. Stein, G.E. Drug interactions with fluoroquinolones. Am. J. Med. 1991, 91 (Suppl. 6A), S81–S86. [Google Scholar] [CrossRef] [PubMed]
  126. Riesbeck, K.; Gullberg, M.; Forsgren, A. Evidence that the antibiotic ciprofloxacin counteracts cyclosporine-dependent suppression of cytokine production. Transplantation 1994, 2, 267–272. [Google Scholar] [CrossRef] [PubMed]
  127. Nakajima, A.; Sato, H.; Oda, S.; Yokoi, T. Fluoroquinolones and propionic acid derivatives induce inflammatory responses in vitro. Cell Biol. Toxicol. 2018, 1, 65–77. [Google Scholar] [CrossRef]
  128. Gupta, P.V.; Nirwane, A.M.; Bellubi, T.; Nagarsenker, M.S. Pulmonary Delivery of Synergistic Combination of Fluoroquinolone Antibiotic Complemented with Proteolytic Enzyme: A Novel Antimicrobial and Antibiofilm Strategy. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 2371–2384. [Google Scholar] [CrossRef] [PubMed]
  129. Saini, H.; Chhibber, S.; Harjai, K. Azithromycin and ciprofloxacin: A possible synergistic combination against Pseudomonas aeruginosa biofilm-associated urinary tract infections. Int. J. Antimicrob. Agents 2015, 45, 359–367. [Google Scholar] [CrossRef] [PubMed]
  130. Badari, M.S.; El-Fatah, S.G.A.; Kamel, S.I.; Mohamed, A.S. Immunomodulatory Action of Levofloxacin on Cytokine Production in Adults with Community-Acquired Pneumonia. Med. J. Cairo Univ. 2014, 82, 127–132. [Google Scholar]
  131. Blasi, F.; Tarsia, P.; Mantero, M.; Morlacchi, L.C.; Piffer, F. Cefditoren versus levofloxacin in patients with exacerbations of chronic bronchitis: Serum inflammatory biomarkers, clinical efficacy, and microbiological eradication. Ther. Clin. Risk Manag. 2013, 9, 55–64. [Google Scholar] [CrossRef]
  132. Calbo, E.; Alsina, M.; Rodríguez-Carballeira, M.; Lite, J.; Garau, J. Systemic expression of cytokine production in patients with severe pneumococcal pneumonia: Effects of treatment with a beta-lactam versus a fluoroquinolone. Antimicrob. Agents Chemother. 2008, 52, 239–402. [Google Scholar] [CrossRef]
  133. Remund, K.; Rechsteiner, T.; Rentsch, K.; Vogt, P.; Russi, E.W.; Boehler, A. Attenuation of airway obliteration by ciprofloxacin in experimental posttransplant bronchiolitis obliterans. Transplantation 2008, 85, 726–731. [Google Scholar] [CrossRef]
  134. Kitazawa, T.; Nakayama, K.; Okugawa, S.; Koike, K.; Shibasaki, Y.; Ota, Y. Biphasic regulation of levofloxacin on lipopolysaccharide-induced IL-1β production. Life Sci. 2007, 80, 1572–1577. [Google Scholar] [CrossRef] [PubMed]
  135. Araujo, F.; Slifer, T.; Li, S.; Kuver, A.; Fong, L.; Remington, J. Gemifloxacin inhibits cytokine secretion by lipopolysaccharide stimulated human monocytes at the post-transcriptional level. Clin. Microbiol. Infect. 2004, 10, 213–219. [Google Scholar] [CrossRef] [PubMed]
  136. Gogos, C.A.; Skoutelis, A.; Lekkou, A.; Drosou, E.; Starakis, I.; Marangos, M.N.; Bassaris, H.P. Comparative effects of ciprofloxacin and ceftazidime on cytokine production in patients with severe sepsis caused by gram-negative bacteria. Antimicrob. Agents Chemother. 2004, 48, 2793–2798. [Google Scholar] [CrossRef] [PubMed]
  137. Uriarte, S.M.; Molestina, R.E.; Miller, R.D.; Bernabo, J.; Farinati, A.; Eiguchi, K.; Ramirez, J.A.; Summersgill, J.T. Effects of Fluoroquinolones on the Migration of Human Phagocytes through Chlamydia pneumoniae-Infected and Tumor Necrosis Factor Alpha-Stimulated Endothelial Cells. Antimicrob. Agents Chemother. 2004, 48, 2538–2543. [Google Scholar] [CrossRef] [PubMed]
  138. König, B.; König, W. Moxifloxacin inhibits staphylococcal superantigen induced apoptosis in T-lymphocytes. In Proceedings of the 12th European Congress of Clinical and Microbiological Infectious Diseases (ECCMID), Milan, Italy, 21–24 April 2002. Abstr. p. 793. [Google Scholar]
  139. Ono, Y.; Ohmoto, Y.; Ono, K.; Sakata, Y.; Murata, K. Effect of grepafloxacin on cytokine production in vitro. J. Antimicrob. Chemother. 2000, 46, 91–94. [Google Scholar] [CrossRef] [PubMed]
  140. Khan, A.A.; Slifer, T.R.; Remington, J.S. Effect of trovafloxacin on production of cytokines by human monocytes. Antimicrob. Agents Chemother. 1998, 42, 1713–1717. [Google Scholar] [CrossRef]
  141. Nwariaku, F.R.; McIntyre, K.L.; Sikes, P.J.; Mileski, W.J. The effect of antimicrobial agents on the induction of tumour necrosis factor by alveolar macrophages in vitro in response to endotoxin. J. Antimicrob. Chemother. 1997, 39, 265–267. [Google Scholar] [CrossRef]
Figure 1. (Black arrow) PAMPs of microorganisms and DAMPs released by the infected or injured cells interact with PRRs on the surface of the organism’s immune cells, one of which is TLR. The stimulation of the macrophage TLR receptor rapidly activates not only the NF-κB pathway but also the ERK pathway. The TLR-activated ERK pathway regulates AP-1 transcriptional activity. The nuclear translocation of the activated transcription factor (NF-κB and AP-1) triggers the induction of genes encoding various pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, etc.)/chemokines (MCP-1, MIP-α, etc.) which stimulate the transepithelial migration of neutrophils, oxidative damage, and increases in NO and ROS. All of these mechanisms result in a cytokine storm and then subsequently lead to ARDS. Pro-inflammatory cytokines and environmental stress cause p38MAPK activation, which plays a role in the regulation of the transcriptional activity of NF-κB. (red arrow) Fluoroquinolones exert their immunomodulatory activity by inhibiting the TLR and ERK signalling pathways. FQs also inhibit the activity of phosphodiesterase activity and result in the accumulation of intracellular levels of cAMP. The accumulation of cAMP levels leads to an augmentation in the activity of PKA, which is known to inhibit the transcription factor of NF-κB, thereby inhibiting further lung damage by reducing pro-inflammatory cytokines and chemokine production, neutrophil influx, oxidative damage, as well as NO and ROS. In addition, PKA, in turn, activate CREB as a primary regulator of anti-inflammatory and immune response. We created this figure using the BioRender online app and license.
Figure 1. (Black arrow) PAMPs of microorganisms and DAMPs released by the infected or injured cells interact with PRRs on the surface of the organism’s immune cells, one of which is TLR. The stimulation of the macrophage TLR receptor rapidly activates not only the NF-κB pathway but also the ERK pathway. The TLR-activated ERK pathway regulates AP-1 transcriptional activity. The nuclear translocation of the activated transcription factor (NF-κB and AP-1) triggers the induction of genes encoding various pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, etc.)/chemokines (MCP-1, MIP-α, etc.) which stimulate the transepithelial migration of neutrophils, oxidative damage, and increases in NO and ROS. All of these mechanisms result in a cytokine storm and then subsequently lead to ARDS. Pro-inflammatory cytokines and environmental stress cause p38MAPK activation, which plays a role in the regulation of the transcriptional activity of NF-κB. (red arrow) Fluoroquinolones exert their immunomodulatory activity by inhibiting the TLR and ERK signalling pathways. FQs also inhibit the activity of phosphodiesterase activity and result in the accumulation of intracellular levels of cAMP. The accumulation of cAMP levels leads to an augmentation in the activity of PKA, which is known to inhibit the transcription factor of NF-κB, thereby inhibiting further lung damage by reducing pro-inflammatory cytokines and chemokine production, neutrophil influx, oxidative damage, as well as NO and ROS. In addition, PKA, in turn, activate CREB as a primary regulator of anti-inflammatory and immune response. We created this figure using the BioRender online app and license.
Biomedicines 12 00761 g001
Figure 2. Structure of fluoroquinolones with and without cyclopropyl moiety. We created this figure using the BioRender online app and license.
Figure 2. Structure of fluoroquinolones with and without cyclopropyl moiety. We created this figure using the BioRender online app and license.
Biomedicines 12 00761 g002
Table 1. Immunomodulatory effects of FQs with their mechanism of action in in vitro, in vivo, ex vivo, and human models.
Table 1. Immunomodulatory effects of FQs with their mechanism of action in in vitro, in vivo, ex vivo, and human models.
NoAuthorAgentSubject/Study Design/Model DiseaseOutcomeStudy DesignReferences
1Zusso et al., 2019Ciprofloxacin, LevofloxacinLPS-induced primary microgliaIL-1β, TNF-α, NFkB translocation ↓In vitro[111]
2Serebryakova et al., 2018LevofloxacinInfiltrative pulmonary tuberculosisIL-12 in drug-sensitive tuberculosis ↓,
TNFα in drug-resistant pulmonary tuberculosis ↓, IFNγ in drug-sensitive tuberculosis ↓
In vitro[110]
3Gupta et al., 2017LevofloxacinStaphylococcus aureusIL-10 ↑,
TNF-α, PCT, IL-1β, and IL-6 ↓
In vitro and in vivo in rats[128]
4Saini et al., 2015CiprofloxacinPseudomonas aeruginosaMDA, NO, MIP and IL-6 ↓,
IL-10 ↑
In vitro and in vivo in mice[129]
5Enoki et al., 2015LevofloxacinH1N1 influenza virus A/PR/8/34Oxidative stress, nitrative marker, NO metabolites, ROS, and IFN-γ ↓In vitro and in vivo in mice[29]
6Badari et al., 2015LevofloxacinTNF-α and IL-10 in the serum of pneumonic patientsTNF-α ↓
IL-10 ↓ in control, IL-10 ↑ in pneumonic patients
RCT in human[112]
7Müller-Redetzky
et al., 2015
MoxifloxacinStreptococcus pneumoniaeIL-6, IL-8, IL-1β, KC/CXCL1 ⇔Ex vivo in human[77]
8Badari et al., 2014LevofloxacinCommunity-Acquired PneumoniaTNF-α ↓ and IL-10 ↑RCT in human[130]
9Beisswenger et al., 2014MoxifloxacinBacterial pneumonia (S. pneumoniae, Pseudomonas aeruginosa)KC, IL-1β, IL-17A, TNF-α-expressing cells, neutrophil influx ↓In vivo in mice[31]
10Blasi et al., 2013LevofloxacinChronic bronchitisKL-6 and IL-6 ↓Open-label, randomised study in human[131]
11Tsivkovskii et al., 2011LevofloxacinHuman bronchial epithelial cellsTNF-α, IL-6, IL-8 ↓In vitro[115]
12Kamiński et al., 2010CiprofloxacinPre-activated primary human T cellsTCR-induced generation ROS, IL-2 and IL-4 ↓In vitro[122]
13Huang et al., 2008Ciprofloxacin, levofloxacin, moxifloxacinLPSTNF-α, IL-1β, CXCL2/MIP-2α ↓ (ciprofloxacin)In vivo in mouse[32]
14Calbo et al., 2008LevofloxacinSevere pneumococcal pneumoniaTNF-α, IL-1β, IL-6, IL-8, IL-10, and IL-1 receptor agonist ↓Open-label, randomised study in human[132]
15Remund et al., 2008CiprofloxacinPost-transplant bronchiolitis obliteransTGF-β ↓ and IFN-γ ↑In vivo in rats[133]
16Zhang and Ward 2007BesifloxacinLPS-stimulated human THP-1 monocytes (ophthalmic infections)IL-1α, G-CSF, IL-1rα, IL-6, GM-CSF, IL-12p40, IL-1β, IL-8, IP-10, MCP-1 and MIP-1α ↓In vitro[87]
17Blau et al., 2007MoxifloxacinCystic fibrosis in IB3 and corrected C38 cellsIL-6, IL-8, MAPK ERK1/2, JNK, and NF-κB ↓In vitro[106]
18Kitazawa et al., 2007LevofloxacinLPS-induced
IL-1β production
pre-synthesised IL-1β, p38 ↑
IL-1β ↓
In vitro[134]
19Shalit et al., 2006MoxifloxacinHuman monocytes stimulated with Aspergillus fumigatusIL-8, IL-1β, TNF-α,
MAPK ERK 1/2, p38, p65-NF-κB ↓
In vitro[114]
20Werber et al., 2005MoxifloxacinHuman respiratory epithelial cell lineMAP kinase, NF-κB, NO ↓In vitro[104]
21Araujo et al., 2004GemifloxacinLPS-stimulated human monocytesNF- κB, IL1α, IL-1β, IL-6, IL-10 and TNF-α ↓In vitro[135]
22Gogos et al., 2004CiprofloxacinGram-negative bacteria (Escherichia coli, P. aeurginosa, Proteus spp., Klebsiella pneumonia)IL-10 alongside the IL-10/TNF-α ratio ↑,
TNF-α and IL-6 ↓
RCT in human[136]
23Weiss et al., 2004MoxifloxacinActivated human peripheral blood monocytes and THP-1 cellsIL-8, TNF-α, IL-1β, MAPK ERK 1/2, NF-κB translocation, JNK ↓In vitro[103]
24Uriarte et al., 2004Levofloxacin, moxifloxacin, gatifloxacinHUVEC infected with Chlamydophila pneumoniae or stimulated with TNF-αNeutrophil and monocyte TEM ↓
IL-8 ↓ (MOX and GTFX)
MCP-1 ↓ (MOX)
In vitro[137]
25Shalit et al., 2002MoxifloxacinCandida albicans, cyclophosphamideTNF-α, KC/CXCL1 ↓,
IL-2, IL-10, IFN-γ ⇔
In vivo in mice[33]
26König et al., 2002MoxifloxacinStaphylococcal superantigen-induced apoptosisFas, FasL, and TNF- RI ↓In vitro[138]
27Araujo et al., 2002MoxifloxacinLPS-stimulated monocytesIL-1α, IL-1β, IL-6, IL-10, IL-12 (p70), TNF-α ↓In vitro[107]
28Ono et al., 2000GrepafloxacinLPS-stimulated human peripheral blood cellsIL-2 ↑,
TNF-α, IL-8, IL-1α, and IL-1β ↓
In vitro[139]
29Khan et al., 1998TrovafloxacinHuman monocytes stimulated by LPS or S. aureus heat-killed cellsIL-1α, IL-1β, IL-6, IL-10, GM-CSF, and TNF-α ↓In vitro[140]
30Nwariak FE et al., 1997CiprofloxacinP. aeruginosaTNF response ↓In vitro samples obtained from rabbits[141]
31Riesbek et al., 1994CiprofloxacinLymphocytes incubated with cyclosporineIFN-γ, IL-2 ↑In vitro[126]
32Bailly et al., 1990Ciprofloxacin, Ofloxacin, Grepafloxacinin LPS-stimulated hPBLTNF, IL-1α, IL-1β ↓In vitro[94]
↓: Decreased/downregulated/inhibited; ↑: increase/upregulated/stimulated; ⇔: no significant difference; CXCL: C-X-C motif chemokine ligand; GM-CSF: granulocyte–macrophage colony-stimulating factor; GTFX: gatifloxacin; IFN: interferon; IL: interleukin; KC: keratinocytes-derived chemokine; KL: Krebs von den Lungen; LPS: lipopolysaccharide; MAPK: MAP-Kinases; MDA: malondialdehyde; MIP: macrophage inflammatory protein; MOX: moxifloxacin; NF-κB: NF-kappaB; NO: nitric oxide; P. aeruginosa: Pseudomonas aeruginosa; ROS: reactive oxygen species; S.: Streptococcus; TGF-β: transforming growth factor-β; TNF-α: tumour necrosis factor-α.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yudhawati, R.; Wicaksono, N.F. Immunomodulatory Effects of Fluoroquinolones in Community-Acquired Pneumonia-Associated Acute Respiratory Distress Syndrome. Biomedicines 2024, 12, 761. https://doi.org/10.3390/biomedicines12040761

AMA Style

Yudhawati R, Wicaksono NF. Immunomodulatory Effects of Fluoroquinolones in Community-Acquired Pneumonia-Associated Acute Respiratory Distress Syndrome. Biomedicines. 2024; 12(4):761. https://doi.org/10.3390/biomedicines12040761

Chicago/Turabian Style

Yudhawati, Resti, and Nisrina Fitriyanti Wicaksono. 2024. "Immunomodulatory Effects of Fluoroquinolones in Community-Acquired Pneumonia-Associated Acute Respiratory Distress Syndrome" Biomedicines 12, no. 4: 761. https://doi.org/10.3390/biomedicines12040761

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop