Skip to main content

PERSPECTIVE article

Front. Microbiol., 27 July 2021
Sec. Infectious Agents and Disease

Antimicrobial Activity of Non-steroidal Anti-inflammatory Drugs on Biofilm: Current Evidence and Potential for Drug Repurposing

  • 1Laboratório Especial de Microbiologia Clínica (LEMC), Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil
  • 2Department of Infectious Diseases, Centro Universitário de Volta Redonda, Volta Redonda, Brazil
  • 3Department of Internal Medicine, Hospital São Francisco de Assis, Belo Horizonte, Brazil

It has been demonstrated that some non-steroidal anti-inflammatory drugs (NSAIDs), like acetylsalicylic acid, diclofenac, and ibuprofen, have anti-biofilm activity in concentrations found in human pharmacokinetic studies, which could fuel an interest in repurposing these well tolerated drugs as adjunctive therapies for biofilm-related infections. Here we sought to review the currently available data on the anti-biofilm activity of NSAIDs and its relevance in a clinical context. We performed a systematic literature review to identify the most commonly tested NSAIDs drugs in the last 5 years, the bacterial species that have demonstrated to be responsive to their actions, and the emergence of resistance to these molecules. We found that most studies investigating NSAIDs’ activity against biofilms were in vitro, and frequently tested non-clinical bacterial isolates, which may not adequately represent the bacterial populations that cause clinically-relevant biofilm-related infections. Furthermore, studies concerning NSAIDs and antibiotic resistance are scarce, with divergent outcomes. Although the potential to use NSAIDs to control biofilm-related infections seems to be an exciting avenue, there is a paucity of studies that tested these drugs using appropriate in vivo models of biofilm infections or in controlled human clinical trials to support their repurposing as anti-biofilm agents.

Introduction

Biofilm formation is a common mechanism that promotes the survival and persistence of medically important bacteria in different abiotic and biotic niches. Approximately 80% of chronic and recurrent bacterial infections in humans are related to biofilms, some of which associated with high mortality and morbidity (Banerjee et al., 2020; Uruén et al., 2020). Biofilms are defined as a community of sessile microbes held together by a secreted matrix of polysaccharides, proteins, and extracellular DNA, adherent to a surface, interface, or other cells. It serves as a natural physical barrier, and harbor other remarkable functions: they establish a constant flow of nutrients for microbial growth, create conditions for interbacterial interaction, promote the exchange of genetic material, resistance to extreme pH, and can become significantly more resistant to the action of antimicrobial agents and immune defenses, making biofilm-related infections challenging to eradicate (Yin et al., 2019; Salgar-Chaparro et al., 2020; Uruén et al., 2020). As a result, most healthcare-related infections associated with the insertion of an artificial medical device can only be controlled after removing the device, which results in more challenging and expensive care of these patients (Uruén et al., 2020).

An increasing body of evidence suggests that non-steroidal anti-inflammatory drugs (NSAIDs) seem to exert some antimicrobial and anti-biofilm activities against clinically relevant bacteria. Since these drugs are already universally used in medicine, novel indications to overcome currently unmet problems, like augmenting eradication of biofilm-related infections, seem to be attractive. Here, we present a short discussion on the potential of NSAID to control biofilm infections, focusing on Staphylococcus aureus and selected Gram-negative pathogens, as well as currently identified drawbacks that in our opinion hinder the potential to repurposing NSAIDs as anti-biofilm agents. We performed a systematic literature search to identify the most commonly tested NSAIDs drugs in the last 5 years, the bacterial species that have demonstrated to be responsive to their actions, and the emergence of resistance to these molecules.

However, although the available in vitro data can create some enthusiasm on the prospects of repurposing NSAIDs as anti-biofilm agents, these observations can hardly be translated into clinical practice as it stands. Data on NSAIDs’ in vitro activity against planktonic bacteria and biofilms are not consistent (Riordan et al., 2011; Belfield et al., 2017; Abbas et al., 2020; Gerner et al., 2020; Leão et al., 2020). In addition, there is evidence showing the selection of resistant bacterial populations following exposure to NSAIDs (Cohen et al., 1993; Gustafson et al., 1999; Helal, 2012; Belfield et al., 2017; Verma et al., 2018).

Methods

A systematic search in PubMed and Scopus (Supplementary Appendix A), was conducted by one the authors. Subsequently, all authors analyzed the abstracts and extracted data from the manuscripts. This search aimed to quantify the number of studies that tested NSAIDs’ activities against biofilm-forming pathogens in vitro or in vivo and identify the most targeted organisms in reports published in the last 5 years. Thirty two studies were found. They were all experimental in vitro studies, except for one, which investigate whether aspirin (ASA, acetylsalicylic acid) could promote infection control for patients with periprosthetic joint infections (Wei et al., 2020). Most of them tested pathogens that are highly relevant in biofilm-associated infections (Table 1). Moreover, we performed a bibliometric analysis to determine the most studied NSAIDs regarding biofilm activity also in the last 5 years (Table 2). Bibliometric analysis is a universally used instrument that applies mathematical and statistical methods to assess research trends and progress. For citations, the data were analyzed from a Microsoft Excel file containing all Digital Object Identifiers (DOI) using VOS viewer software (version 1.6.15) through exported PubMed citation files. In addition, we manually added other studies from a Scopus systematic search.

TABLE 1
www.frontiersin.org

Table 1. Studied human pathogens in the biofilm state concerning non-steroidal anti-inflammatory drugs (NSAIDs) activity in the last 5 years.

TABLE 2
www.frontiersin.org

Table 2. Frequency of studied NSAIDs in thirty-two articles.

Antibiofilm Activity of NSAIDs Against Staphylococcus Aureus and Selected Pathogenic Gram-Negative Species

Staphylococcus aureus has evolved intricate mechanisms to escape the immune system, efficiently invade and damage the host tissues, causing a multitude of infections (Kuehl et al., 2020). Its ability to form biofilms is a key mechanism that confers a survival advantage under harsh conditions. Attachment of S. aureus to medical implants and host tissues plays an important role in persistent chronic infections. Although biofilm production is a highly conserved mechanism in S. aureus, the composition and thickness of biofilms may differ among different strains (Kiedrowski et al., 2011; Vanhommerig et al., 2014). Methicillin-susceptible S. aureus (MSSA) and methicillin-resistant S. aureus (MRSA) may behave differently during infection, forming biofilms of varying biomass through different pathways (Lamret et al., 2020). Biofilm formation in MSSA is usually dependent on polysaccharide intercellular adhesin or polymeric N-acetyl-glucosamine, whereas MRSA strains frequently produce biofilms with a more proteinaceous matrix (Pozzi et al., 2012; Vanhommerig et al., 2014). Differences in biofilm phenotypes are also seen among the major MRSA clonal complexes that cause human infections, with the USA300 clone producing more robust and abundant biofilms under static and dynamic conditions than the other major clones (Vanhommerig et al., 2014).

Although there is evidence pointing to the activity of NSAIDs against S. aureus biofilms and other anti-virulence properties (e.g., inhibition of hemolysis and staphyloxanthin production; Abbas et al., 2020), most of these data were generated by in vitro studies using control laboratory bacterial strains, which may not necessarily indicate that these observations will be translated in vivo. These findings could also not be extrapolated to infections caused by different clonal complexes, given their significant differences in biofilm phenotypes (Vanhommerig et al., 2014). Thus, data from these in vitro experiments must be interpreted with caution whilst future studies investigate whether results are replicated in clinical isolates and in vivo models.

Studies testing the anti-biofilm potential of NSAIDs against other pathogenic bacteria have also been published in the last 5 years. Pseudomonas aeruginosa was the most studied Gram-negative pathogen (Table 1), with other species being scarcely tested. Gerner et al. conducted an interesting study involving P. aeruginosa clinical strains isolated from chronic wounds with different virulence profiles. Strains were evaluated using in vitro assays to produce biofilm, pyocyanin, siderophores, alkaline protease, elastase, and stapholytic protease. Exposure of these isolates to sodium salicylate, a sodium salt of salicylic acid, resulted in quorum-sensing inhibition and decreased virulence (Gerner et al., 2020). A previous report demonstrated that the growth of P. aeruginosa in the presence of salicylate reduced the production of extracellular polysaccharides required for biofilm formation (Pereira et al., 2018). Belfield et al. did not eradicate a mature biofilm applying ciprofloxacin or gentamicin 100 × MIC, with or without ASA or sodium salicylate or salicylic acid in high concentrations (200, 175, and 150 μg/ml, respectively). However, bacterial growth was inhibited by antibiotic–NSAID combinations (Belfield et al., 2017). Therefore, testing specific NSAIDs alone or associated with antibiotics and employing different concentrations on biofilms produced by high-risk and biofilm-producing bacterial clones could result in additional relevant data regarding biofilm control by NSAIDs (Cohen et al., 1993; Verma et al., 2018; Gerner et al., 2020).

The Concentration of NSAIDs in Experimental Studies and Their Pharmacokinetics (PK) in Humans’ Trials

Because NSAIDs have been extensively tested in humans, pharmacokinetics data of these molecules can point toward whether or not concentrations tested in vitro against biofilms are achievable in the plasma, which would suggest a potential clinical application and drug repurposing. ASA concentrations ranging from 50 to 200 μg/ml, which are in the range frequently achieved by therapeutic ASA doses in humans, produces a significant level of anti-biofilm activity in vitro against Candida albicans (Alem and Douglas, 2004; Wei et al., 2020). Physiological concentrations of the drug was shown to reduce mature biofilms by 20–80% (in vivo), suggesting that ASA could have a significant inhibitory effect (Alem and Douglas, 2004). Salicylic acid at therapeutic concentrations was shown to reduce biofilms produced by Candida spp. (Alem and Douglas, 2004; Stepanović et al., 2004; Carvalho et al., 2010; Zhou et al., 2012), Escherichia coli (Kang et al., 1998; Chubiz and Rao, 2011; Vila and Soto, 2012), P. aeruginosa (Farber et al., 1995; Prithiviraj et al., 2005; El-Mowafy et al., 2014), and Staphylococcus epidermidis (Teichberg et al., 1993; Farber et al., 1995; Muller et al., 1998). Abbas et al. used a sub-MIC of diclofenac (1/4, 62.5 μg/ml) to investigate its inhibitory effect on different virulence factors of MRSA, including biofilm formation. Diclofenac exhibited a substantial reduction in biofilm formation compared to controls; the inhibition varied between 22.67 and 70% (Abbas et al., 2020). Diclofenac when tested against a S. aureus isolate demonstrated in vitro activity with a high MIC (2,000 μg/mL) and no detectable minimal bactericidal concentration (>2,000 μg/mL; Leão et al.). Even by applying a high diclofenac concentration (5 × MIC), anti-S. aureus-biofilm activity was not found. However, this high concentration of diclofenac had an effect in inactivating the S. aureus biofilm metabolism (inactivation percentages were between 80.4 and 86.7%). The authors observed a biofilm killing activity only when diclofenac was associated with kanamycin or tetracycline: at the established MIC (2,000 μg/ml) and 5 × MIC with kanamycin (low removal) and the MIC, 5 × MIC and 10 × MIC for tetracycline (low and moderate removal; Leão et al., 2020). Shah et al. observed a peak plasma concentration (Cmax) of 0.625 μg/ml for diclofenac, lower than the concentrations mentioned above. This pharmacokinetic study was performed with 30 volunteers who received 100 mg of diclofenac sodium (extended-release capsules; Shah et al., 2016). Of note, there is no safety data for doses above 150 mg once daily.

Dai et al. demonstrated positive results regarding the anti-biofilm formation and anti-quorum sensing activity of ibuprofen against P. aeruginosa. Furthermore, they found a concentration-dependent reduction in biofilm formation with rising drug concentrations (0, 50, 75, and 100 μg/mL). A previous PK study concerning intra-venous ibuprofen administration in children showed a mean Cmax = 35.8 μg/mL at a single dose of 8 mg/kg (Kauffman and Nelson, 1992).

Although the current PK data for ASA, diclofenac, and ibuprofen demonstrate that these molecules could achieve serum levels that are shown to have anti-biofilm activities in vitro, drug levels in tissues that are frequently infected by biofilms (bone and soft tissue in chronic osteomyelitis, valvar cardiac tissue in endocarditis, and urine or bladder tissue in recurrent urinary tract infections) need to be further evaluated.

NSAIDs and Their Ability to Affect Tolerance and Antibiotic Resistance

Antibiotic tolerance and resistance are common mechanisms of survival within biofilm-embedded cells (Gerner et al., 2020). A proven unwanted side effect of salicylate, for example, is the modulation of antibiotic resistance (Cohen et al., 1993; Gustafson et al., 1999; Helal, 2012). In a biofilm, the high density of bacteria increases horizontal gene transfers, a common mechanism used by bacteria to disseminate antimicrobial resistance genes located in mobile genetic elements (Verma et al., 2018). Antibiotic tolerance is an ephemeral and reversible phenotypic state associated with differences in the physiology of cells according to their localization in the biofilm structure. Cells closer to the biotic or abiotic surfaces and thus more deeply located within the biofilm tend to be more deprived of oxygen and nutrients and join in a physiologically dormant state for which antimicrobials target key metabolic routes would not be active (Verma et al., 2018).

Experimental observations have demonstrated that exposure to NSAIDs can result in transcriptional adaptions that result in increased antibiotic resistance (Cohen et al., 1993; Gustafson et al., 1999; Helal, 2012; Verma et al., 2018). Verma et al. (2018) demonstrated that exposure of planktonic E. coli to sodium salicylate, ASA, acetaminophen, and ibuprofen could result in increased resistance to ciprofloxacin and tetracycline. Ibuprofen seems to be associated with low-level resistance phenotypes primarily associated with an increase in the efflux of antibiotics via the AcrAB-TolC efflux pump in a manner dependent on the transcriptional regulator MarA (Price et al., 2000; Helal, 2012). Increased antibiotic resistance following exposure to sodium salicylate or ASA, or acetaminophen is relatively higher when compared to ibuprofen and only partially dependent on MarA (Riordan et al., 2011). Other studies have shown that E. coli exhibits increased resistance to several antimicrobials (fluoroquinolones, ampicillin, cephalosporins, tetracycline, and chloramphenicol) in the presence of salicylate (Soheili et al., 2015; Belfield et al., 2017; Fourie et al., 2017; Pereira et al., 2018; She et al., 2018; Abidi et al., 2019; Dai et al., 2019). This drug also increases S. aureus resistance to fluoroquinolones and fusidic acid (Belfield et al., 2017; Cai et al., 2018; Oliveira et al., 2019; Abbas et al., 2020; Leão et al., 2020). Enhanced resistance to antimicrobials in the presence of salicylate has been attributed to the modification of membrane-associated proteins that results in reduced drug accumulation in the intracellular compartment and decreased susceptibility to a range of antimicrobials (Abidi et al., 2019). However, unlike ASA and ibuprofen, diclofenac did not induce antibiotic resistance in previous experimental research. Evidence regarding this successful outcome showed that diclofenac reduced the MIC for fluoroquinolones, oxacillin, and vancomycin against S. aureus (Price et al., 1999). One of the two clinical strains used in the experiment had a high ciprofloxacin MIC in the absence of diclofenac (MIC = 32), but a significant reduction in MIC in the presence of this drug at two different concentrations (MIC = 4; Price et al., 1999).

Discussion

Recently, Leão et al. (2020) published an interesting study evaluating the in vitro activity of NSAIDs against E. coli and S. aureus biofilms. In vitro experiments showed that ASA diclofenac and ibuprofen have anti-biofilm activity in concentrations similar to those found in human pharmacokinetic studies (plasma concentrations). There are multiple suggested mechanisms of anti-bacterial and anti-biofilm actions of NSAIDs and they differ according to the species. For example, in S. aureus, NSAIDs seem to exert an antimicrobial or anti-biofilm activity by blocking AgrA-regulated virulence, inhibiting hemolysis, hindering staphyloxanthin production, and downregulating the expression fnbA and icaA genes, which are crucial in biofilm formation. ASA inhibits quorum-sensing in P. aeruginosa and salicylate reduces the production of extracellular polysaccharides required for biofilm formation (Hendrix et al., 2016; Pereira et al., 2018; Abbas et al., 2020).

Even though the available in vitro data can create some interest in the prospects of repurposing NSAIDs as anti-biofilm agents, more evidence from in vivo studies is required to determine whether the use of NSAIDs would translate into clinical efficacy in treating biofilm infections. Moreover, data regarding in vitro activity of NSAIDs against planktonic bacteria and biofilms are not consistent. According to the performed systematic search, only two studies have assessed NSAID activity against relevant pathogenic coagulase-negative staphylococci in the last 5 years (Pereira et al., 2018; Abidi et al., 2019). These organisms are the most common causes of healthcare-associated biofilm-related infections, like catheter-related bloodstream infections and prosthetic joint infections (De Vecchi et al., 2018; Hebeisen et al., 2019). Species like S. epidermidis, Staphylococcus haemolyticus, Staphylococcus hominis, are also important biofilm-forming microorganisms. Enterococcus spp. and some Gram-negative species have also been poorly studied and constitute a relevant pathogen in community and healthcare environments. Enterococcus spp. have been recognized as an important pathogen in chronic wound infections, mainly in chronic diabetes foot infections (Jneid et al., 2018; Carro et al., 2020). In the future, the NSAIDs could be an alternative tool to treat a delayed healing wound, clinical evidence of an active and mature biofilm. Staphylococcus spp. and Enterococcus spp. are also present in PJI, which could be linked to biofilm formation. Intracellular internalization of staphylococci, particularly “Small colony variant” strains, besides invasion and colonization of the lacuna-canalicular system of cortical bone are other causes of PJIs (Parvizi et al., 2013; Zoller et al., 2020). A clinical study explored if ASA could promote infection control for patients with periprosthetic joint infections (PJIs) (Wei et al., 2020). Regular ASA exposure was characterized as a prescription of ASA for >6 months prior to diagnosis of PJI and uninterrupted use during the PJI therapy at a dose ≧ 100 mg/day. Eighty-eight patients who met the PJI criteria were identified and incorporated in this study. Of these patients, 12 were taking ASA regularly during the infectious events. The Cox proportional hazard model analysis of time-to-infection resolution showed that the ASA was beneficial for PJIs ending (95% CI 1.018–4.757; p = 0.045; Wei et al., 2020). However, we do not know how many PJIs were biofilm-related and whether the anti-inflammatory effect of ASA proportionate a better outcome. Although it may represent an anti-biofilm effect of ASA, data regarding ASA and antimicrobial tissue concentrations, for example, are lacking. So, it is not possible to state that ASA works in this clinical setting. The in vitro experiments showing anti-biofilm inhibition or control with equal plasma serum concentrations identified in human pharmacokinetic studies should be reproduced, as well as new research employing other NSAIDs and distinct microorganisms (pointing out the clones of interest). Importantly, because of the positive data outcomes from PK in vitro studies, trials regarding NSAIDs levels on the tissues of biofilm-associated infections–like bone and soft tissue in chronic osteomyelitis, and valvar cardiac tissue in endocarditis–should be performed.

According to the outcomes of the study of Leão et al. (2020), the association between an antimicrobial and an NSAID–piroxicam or diclofenac or ASA employment with kanamycin or tetracycline–could be an interesting research field since this combination can result in biofilm removal. Unlike the study of Leão et al., Belfield et al. (2017) do not reach biofilm removal employing ciprofloxacin and gentamycin against P. aeruginosa and S. aureus strains isolated from clinical biofilm infections. Thus, the association mechanism of action between those drugs and the determination of the clonal lineages for each microorganism could clarify these discordant results.

Besides NSAIDs, alternative strategies and novel antibiofilm agents have been studied earlier, like nanoparticles, photodynamic therapy, natural compounds, anti-quorum sensing signaling molecules, matrix-degrading enzyme, and gene editing technique (CRISPR-CAS) (Sharma et al., 2019). Most of these tools, however, are also restricted to in vitro studies. So far, there is no place for NSAIDs in managing biofilm-related infections, even in non-life-threatening ones, like mild chronic wounds, despite the availability of topical commercial formulations of NSAIDs in high concentrations. Biofilms may be polymicrobial, which provides several advantages, such as passive resistance, metabolic cooperation, quorum sensing systems, an enlarged gene pool with more efficient DNA sharing, and many other synergies giving them a competitive advantage. Therefore, given the knowledge regarding biofilms in humans, data concerning NSAID concentrations, performance in distinct clones of pathogenic agents, and bacterial resistance is required to obtain sufficient evidence.

Data Availability Statement

The raw data supporting the conclusions of this article will be made available by the authors, without undue reservation.

Author Contributions

Both authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Acknowledgments

We thank Paulo Bispo for useful discussions and suggestions to improve the quality of this manuscript.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fmicb.2021.707629/full#supplementary-material

References

Abbas, H. A., Atallah, H., El-Sayed, M. A., and El-Ganiny, A. M. (2020). Diclofenac mitigates virulence of multidrug-resistant Staphylococcus aureus. Arch. Microbiol. 202, 2751–2760. doi: 10.1007/s00203-020-01992-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Abidi, S., Ahmed, K., and Kazmi, S. (2019). The antibiofilm activity of Acetylsalicylic acid, Mefenamic acid, acetaminophen against biofilms formed by P. aeruginosa and S. epidermidis. J. Pak. Med. Assoc. 1, 1493–1495.

Google Scholar

Alem, M. A. S., and Douglas, L. J. (2004). Effects of aspirin and other nonsteroidal anti-inflammatory drugs on biofilms and planktonic cells of Candida albicans. Antimicrob. Agents Chemother. 48, 41–47. doi: 10.1128/aac.48.1.41-47.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Banerjee, D., Shivapriya, P. M., Gautam, P. K., Misra, K., Sahoo, A. K., and Samanta, S. K. (2020). A review on basic biology of bacterial biofilm infections and their treatments by nanotechnology-based approaches. Proc. Natl. Acad. Sci. India Sect. B Biol. Sci. 90, 243–259. doi: 10.1007/s40011-018-01065-7

CrossRef Full Text | Google Scholar

Belfield, K., Bayston, R., Hajduk, N., Levell, G., Birchall, J. P., and Daniel, M. (2017). Evaluation of combinations of putative anti-biofilm agents and antibiotics to eradicate biofilms of Staphylococcus aureus and Pseudomonas aeruginosa. J. Antimicrob. Chemother. 72, 2531–2538. doi: 10.1093/jac/dkx192

PubMed Abstract | CrossRef Full Text | Google Scholar

Brilhante, R. S. N., Brasil, J. A., de Oliveira, J. S., Pereira, V. S., Pereira-Neto, W. A., Sidrim, J. J. C., et al. (2020). Diclofenac exhibits synergism with azoles against planktonic cells and biofilms of Candida tropicalis. Biofouling 36, 528–536. doi: 10.1080/08927014.2020.1777285

PubMed Abstract | CrossRef Full Text | Google Scholar

Cai, J.-Y., Hou, Y.-N., Li, J., Ma, K., Yao, G.-D., Liu, W.-W., et al. (2018). Prostaglandin E2 attenuates synergistic bactericidal effects between COX inhibitors and antibiotics on Staphylococcus aureus. Prostaglandins Leukot. Essent. Fat. Acids 133, 16–22. doi: 10.1016/j.plefa.2018.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Carro, G. V., Saurral, R., Salvador Sagüez, F., and Witman, E. L. (2020). Diabetic foot infections: bacterial isolates from the centers and hospitals of Latin American countries. Int. J. Low. Extrem. Wounds doi: 10.1177/1534734620976305 Online ahead of print.

CrossRef Full Text | PubMed Abstract | Google Scholar

Carvalho, A. P., Gursky, L. C., Rosa, R. T., Rymovicz, A. U. M., Campelo, P. M. S., Grégio, A. M. T., et al. (2010). Non-steroidal anti-inflammatory drugs may modulate the protease activity of Candida albicans. Microb. Pathog. 49, 315–322. doi: 10.1016/j.micpath.2010.07.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Cattò, C., Grazioso, G., Dell’Orto, S., Gelain, A., Villa, S., Marzano, V., et al. (2017). The response of Escherichia coli biofilm to salicylic acid. Biofouling 33, 235–251. doi: 10.1080/08927014.2017.1286649

PubMed Abstract | CrossRef Full Text | Google Scholar

Chubiz, L. M., and Rao, C. V. (2011). Role of the mar-sox-rob regulon in regulating outer membrane porin expression. J. Bacteriol. 193, 2252–2260. doi: 10.1128/jb.01382-10

PubMed Abstract | CrossRef Full Text | Google Scholar

Cohen, S. P., Levy, S. B., Foulds, J., and Rosner, J. L. (1993). Salicylate induction of antibiotic resistance in Escherichia coli: activation of the mar operon and a mar-independent pathway. J. Bacteriol. 175, 7856–7862. doi: 10.1128/jb.175.24.7856-7862.1993

PubMed Abstract | CrossRef Full Text | Google Scholar

Dai, L., Wu, T., Xiong, Y., Ni, H., Ding, Y., Zhang, W., et al. (2019). Ibuprofen-mediated potential inhibition of biofilm development and quorum sensing in Pseudomonas aeruginosa. Life Sci. 237:116947. doi: 10.1016/j.lfs.2019.116947

PubMed Abstract | CrossRef Full Text | Google Scholar

De Vecchi, E., George, D., Romanò, C. L., Pregliasco, F., Mattina, R., and Drago, L. (2018). Antibiotic sensitivities of coagulase-negative staphylococci and Staphylococcus aureus in hip and knee periprosthetic joint infections: does this differ if patients meet the international consensus meeting criteria? Infect. Drug Resist. 11, 539–546. doi: 10.2147/idr.s151271

PubMed Abstract | CrossRef Full Text | Google Scholar

Dotto, C., Serrat, A. L., Cattelan, N., Barbagelata, M. S., Yantorno, O. M., Sordelli, D. O., et al. (2017). The active component of aspirin, salicylic acid, promotes staphylococcus aureus biofilm formation in a pia-dependent manner. Front. Microbiol. 8:4.

Google Scholar

El-Mowafy, S. A., Abd El Galil, K. H., El-Messery, S. M., and Shaaban, M. I. (2014). Aspirin is an efficient inhibitor of quorum sensing, virulence and toxins in Pseudomonas aeruginosa. Microb. Pathog. 74, 25–32. doi: 10.1016/j.micpath.2014.07.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Farber, B. F., Hsieh, H.-C., Donnenfeld, E. D., Perry, H. D., Epstein, A., and Wolff, A. (1995). A novel antibiofilm technology for contact lens solutions. Ophthalmology 102, 831–836. doi: 10.1016/s0161-6420(95)30949-9

CrossRef Full Text | Google Scholar

Fourie, R., Ells, R., Kemp, G., Sebolai, O. M., Albertyn, J., and Pohl, C. H. (2017). Pseudomonas aeruginosa produces aspirin insensitive eicosanoids and contributes to the eicosanoid profile of polymicrobial biofilms with Candida albicans. Prostaglandins Leukot. Essent. Fat. Acids 117, 36–46. doi: 10.1016/j.plefa.2017.01.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Gerner, E., Almqvist, S., Werthén, M., and Trobos, M. (2020). Sodium salicylate interferes with quorum-sensing-regulated virulence in chronic wound isolates of Pseudomonas aeruginosa in simulated wound fluid. J. Med. Microbiol. 69, 767–780. doi: 10.1099/jmm.0.001188

PubMed Abstract | CrossRef Full Text | Google Scholar

Gomes, I. B., Simões, L. C., and Simões, M. (2018). The effects of emerging environmental contaminants on Stenotrophomonas maltophilia isolated from drinking water in planktonic and sessile states. Sci. Total Environ. 643, 1348–1356. doi: 10.1016/j.scitotenv.2018.06.263

PubMed Abstract | CrossRef Full Text | Google Scholar

Gustafson, J. E., Candelaria, P. V., Fisher, S. A., Goodridge, J. P., Lichocik, T. M., McWilliams, T. M., et al. (1999). Growth in the presence of salicylate increases fluoroquinolone resistance in Staphylococcus aureus. Antimicrob. Agents Chemother. 43, 990–992. doi: 10.1128/aac.43.4.990

PubMed Abstract | CrossRef Full Text | Google Scholar

Hebeisen, U. P., Atkinson, A., Marschall, J., and Buetti, N. (2019). Catheter-related bloodstream infections with coagulase-negative staphylococci: are antibiotics necessary if the catheter is removed? Antimicrob. Resist. Infect. Control 8:21.

Google Scholar

Helal, N. S. (2012). Investigation into the Mechanism of Salicylate-Associated Genotypic Antibiotic Resistance in Staphylococcus aureus. Grad. Theses Diss. Available online at: https://scholarcommons.usf.edu/etd/4069 (accessed October 8, 2020).

Google Scholar

Hendrix, A. S., Spoonmore, T. J., Wilde, A. D., Putnam, N. E., Hammer, N. D., Snyder, D. J., et al. (2016). Repurposing the nonsteroidal anti-inflammatory drug diflunisal as an osteoprotective, antivirulence therapy for Staphylococcus aureus Osteomyelitis. Antimicrob. Agents Chemother. 60, 5322–5330. doi: 10.1128/aac.00834-16

PubMed Abstract | CrossRef Full Text | Google Scholar

Jneid, J., Cassir, N., Schuldiner, S., Jourdan, N., Sotto, A., Lavigne, J.-P., et al. (2018). Exploring the microbiota of diabetic foot infections with culturomics. Front. Cell. Infect. Microbiol. 8:282.

Google Scholar

Kang, G., Balasubramanian, K. A., Koshi, A. R., Mathan, M. M., and Mathan, V. I. (1998). Salicylate inhibits fimbriae mediated HEp-2 cell adherence of and haemagglutination by enteroaggregative Escherichia coli. FEMS Microbiol. Lett. 166, 257–265. doi: 10.1111/j.1574-6968.1998.tb13899.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Kauffman, R. E., and Nelson, M. V. (1992). Effect of age on ibuprofen pharmacokinetics and antipyretic response. J. Pediatr. 121, 969–973. doi: 10.1016/S0022-3476(05)80354-3

CrossRef Full Text | Google Scholar

Kiedrowski, M. R., Kavanaugh, J. S., Malone, C. L., Mootz, J. M., Voyich, J. M., Smeltzer, M. S., et al. (2011). Nuclease modulates biofilm formation in community-associated methicillin-resistant Staphylococcus aureus. PLoS One 6:e26714. doi: 10.1371/journal.pone.0026714

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuehl, R., Morata, L., Meylan, S., Mensa, J., and Soriano, A. (2020). When antibiotics fail: a clinical and microbiological perspective on antibiotic tolerance and persistence of Staphylococcus aureus. J. Antimicrob. Chemother. 75, 1071–1086. doi: 10.1093/jac/dkz559

PubMed Abstract | CrossRef Full Text | Google Scholar

Lamret, F., Colin, M., Mongaret, C., Gangloff, S. C., and Reffuveille, F. (2020). Antibiotic tolerance of Staphylococcus aureus biofilm in periprosthetic joint infections and antibiofilm strategies. Antibiotics 9:547. doi: 10.3390/antibiotics9090547

PubMed Abstract | CrossRef Full Text | Google Scholar

Leão, C., Borges, A., and Simões, M. (2020). NSAIDs as a drug repurposing strategy for biofilm control. Antibiotics 9:591. doi: 10.3390/antibiotics9090591

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, W., Yu, X., Huang, J., Huang, H., and Cheng, J. (2020). A comparative study of relevant drug intervention in Candida albicans biofilm. Lat. Am. J. Pharm. 39, 1065–1075.

Google Scholar

Maitra, A., Evangelopoulos, D., Chrzastek, A., Martin, L. T., Hanrath, A., Chapman, E., et al. (2020). Carprofen elicits pleiotropic mechanisms of bactericidal action with the potential to reverse antimicrobial drug resistance in tuberculosis. J. Antimicrob. Chemother. 75, 3194–3201. doi: 10.1093/jac/dkaa307

PubMed Abstract | CrossRef Full Text | Google Scholar

Muller, E., Al-Attar, J., Wolff, A. G., and Farber, B. F. (1998). Mechanism of salicylate-mediated inhibition of biofilm in Staphylococcus epidermidis. J. Infect. Dis. 177, 501–503. doi: 10.1086/517386

PubMed Abstract | CrossRef Full Text | Google Scholar

Oliveira, I. M., Borges, A., Borges, F., and Simões, M. (2019). Repurposing ibuprofen to control Staphylococcus aureus biofilms. Eur. J. Med. Chem. 166, 197–205. doi: 10.1016/j.ejmech.2019.01.046

PubMed Abstract | CrossRef Full Text | Google Scholar

Parvizi, J., Aggarwal, V., and Rasouli, M. (2013). Periprosthetic joint infection: current concept. Indian J. Orthop. 47:10. doi: 10.4103/0019-5413.106884

PubMed Abstract | CrossRef Full Text | Google Scholar

Pereira, S. G., Domingues, V. S., Theriága, J., Chasqueira, M. J. M., and Paixão, P. (2018). Non-antimicrobial drugs: etodolac as a possible antimicrobial or adjuvant agent against ESKAPE pathogens. Open Microbiol. J. 12, 288–296. doi: 10.2174/1874285801812010288

PubMed Abstract | CrossRef Full Text | Google Scholar

Pozzi, C., Waters, E. M., Rudkin, J. K., Schaeffer, C. R., Lohan, A. J., Tong, P., et al. (2012). Methicillin resistance alters the biofilm phenotype and attenuates virulence in Staphylococcus aureus device-associated infections. PLoS Pathog. 8:e1002626. doi: 10.1371/journal.ppat.1002626

PubMed Abstract | CrossRef Full Text | Google Scholar

Price, C. T. D., O’Brien, F. G., Shelton, B. P., Warmington, J. R., Grubb, W. B., and Gustafson, J. E. (1999). Effects of salicylate and related compounds on fusidic acid MICs in Staphylococcus aureus. J. Antimicrob. Chemother. 44, 57–64. doi: 10.1093/jac/44.1.57

PubMed Abstract | CrossRef Full Text | Google Scholar

Price, C. T., Lee, I. R., and Gustafson, J. E. (2000). The effects of Salicylate on bacteria. Int. J. Biochem. Cell Biol. 32, 1029–1043. doi: 10.1016/s1357-2725(00)00042-x

CrossRef Full Text | Google Scholar

Prithiviraj, B., Bais, H. P., Weir, T., Suresh, B., Najarro, E. H., Dayakar, B. V., et al. (2005). Down regulation of virulence factors of Pseudomonas aeruginosa by salicylic acid attenuates its virulence on Arabidopsis thaliana and Caenorhabditis elegans. Infect. Immun. 73, 5319–5328. doi: 10.1128/iai.73.9.5319-5328.2005

PubMed Abstract | CrossRef Full Text | Google Scholar

Riordan, J. T., Dupre, J. M., Cantore-Matyi, S. A., Kumar-Singh, A., Song, Y., Zaman, S., et al. (2011). Alterations in the transcriptome and antibiotic susceptibility of Staphylococcus aureus grown in the presence of diclofenac. Ann. Clin. Microbiol. Antimicrob. 10:30. doi: 10.1186/1476-0711-10-30

PubMed Abstract | CrossRef Full Text | Google Scholar

Rosato, A., Catalano, A., Carocci, A., Carrieri, A., Carone, A., Caggiano, G., et al. (2016). In vitro interactions between anidulafungin and nonsteroidal anti-inflammatory drugs on biofilms of Candida spp. Bioorg. Med. Chem. 24, 1002–1005. doi: 10.1016/j.bmc.2016.01.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Salgar-Chaparro, S. J., Lepkova, K., Pojtanabuntoeng, T., Darwin, A., and Machuca, L. L. (2020). Nutrient level determines biofilm characteristics and subsequent impact on microbial corrosion and biocide effectiveness. Appl. Environ. Microbiol. 86:e2885-19.

Google Scholar

Santos, F. A. G., dos, Leite-Andrade, M. C., Brandão, I. S., Alves, A. I. S., Buonafina, M. D. S., et al. (2020). Anti-biofilm effect by the combined action of fluconazole and acetylsalicylic acid against species of Candida parapsilosis complex. Infect. Genet. Evol. 84:104378. doi: 10.1016/j.meegid.2020.104378

PubMed Abstract | CrossRef Full Text | Google Scholar

Shah, P. N., Marshall-Batty, K. R., Smolen, J. A., Tagaev, J. A., Chen, Q., Rodesney, C. A., et al. (2018). Antimicrobial activity of ibuprofen against cystic fibrosis-associated gram-negative pathogens. Antimicrob. Agents Chemother. 62:e1574-17.

Google Scholar

Shah, I., Barker, J., Naughton, D. P., Barton, S. J., and Ashraf, S. S. (2016). Determination of diclofenac concentrations in human plasma using a sensitive gas chromatography mass spectrometry method. Chem. Cent. J. 10:52. doi: 10.1186/s13065-016-0199-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, D., Misba, L., and Khan, A. U. (2019). Antibiotics versus biofilm: an emerging battleground in microbial communities. Antimicrob. Resist. Infect. Control 8:76.

Google Scholar

She, P., Wang, Y., Luo, Z., Chen, L., Tan, R., Wang, Y., et al. (2018). Meloxicam inhibits biofilm formation and enhances antimicrobial agents efficacy by Pseudomonas aerugino. Microbiologyopen 7:e00545. doi: 10.1002/mbo3.545

PubMed Abstract | CrossRef Full Text | Google Scholar

Soheili, V., Bazzaz, B. S. F., Abdollahpour, N., and Hadizadeh, F. (2015). Investigation of Pseudomonas aeruginosa quorum-sensing signaling system for identifying multiple inhibitors using molecular docking and structural analysis methodology. Microb. Pathog. 89, 73–78. doi: 10.1016/j.micpath.2015.08.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Stepanović, S., Vuković, D., Ješić, M., and Ranin, L. (2004). Influence of acetylsalicylic acid (Aspirin) on biofilm production by Candida species. J. Chemother. 16, 134–138. doi: 10.1179/joc.2004.16.2.134

PubMed Abstract | CrossRef Full Text | Google Scholar

Teichberg, S., Farber, B. F., Wolff, A. G., and Roberts, B. (1993). Salicylic acid decreases extracellular biofilm production by staphylococcus epidermidis: electron microscopic analysis. J. Infect. Dis. 167, 1501–1503. doi: 10.1093/infdis/167.6.1501

PubMed Abstract | CrossRef Full Text | Google Scholar

Tzeng, S.-R., Huang, Y.-W., Zhang, Y.-Q., Yang, C.-Y., Chien, H.-S., Chen, Y.-R., et al. (2020). A celecoxib derivative eradicates antibiotic-resistant Staphylococcus aureus and biofilms by targeting YidC2 translocase. Int. J. Mol. Sci. 21:9312. doi: 10.3390/ijms21239312

PubMed Abstract | CrossRef Full Text | Google Scholar

Ulusoy, S., Senkardes, S., Coskun, I., Bosgelmez-T|naz, G., Soulere, L., and Queneau, Y. (2017). Quorum sensing inhibitor activities of celecoxib derivatives in Pseudomonas aeruginosa. Lett. Drug Des. Discov. 14, 613–618. doi: 10.2174/1570180814666161121120719

CrossRef Full Text | Google Scholar

Uruén, C., Chopo-Escuin, G., Tommassen, J., Mainar-Jaime, R. C., and Arenas, J. (2020). Biofilms as promoters of bacterial antibiotic resistance and tolerance. Antibiotics 10:3. doi: 10.3390/antibiotics10010003

PubMed Abstract | CrossRef Full Text | Google Scholar

Vanhommerig, E., Moons, P., Pirici, D., Lammens, C., Hernalsteens, J.-P., De Greve, H., et al. (2014). Comparison of biofilm formation between major clonal lineages of methicillin resistant Staphylococcus aureus. PLoS One 9:e104561. doi: 10.1371/journal.pone.0104561

PubMed Abstract | CrossRef Full Text | Google Scholar

Verma, T., Bhaskarla, C., Sadhir, I., Sreedharan, S., and Nandi, D. (2018). Non-steroidal anti-inflammatory drugs, Acetaminophen and Ibuprofen, induce phenotypic antibiotic resistance in Escherichia coli: roles of marA and acrB. FEMS Microbiol. Lett. 365, 1–9.

Google Scholar

Vila, J., and Soto, S. M. (2012). Salicylate increases the expression of marA and reduces in vitro biofilm formation in uropathogenic Escherichia coli by decreasing type 1 fimbriae expression. Virulence 3, 280–285. doi: 10.4161/viru.19205

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, Y. P., Chien, J. C., Hsiang, W. H., Yang, S. W., and Chen, C. Y. (2020). Aspirin administration might accelerate the subsidence of periprosthetic joint infection. Sci. Rep. 10:15967.

Google Scholar

Xu, C.-G., Yang, Y.-B., Zhou, Y.-H., Hao, M.-Q., Ren, Y. Z., Wang, X.-T., et al. (2017). Comparative proteomic analysis provides insight into the key proteins as possible targets involved in aspirin inhibiting biofilm formation of Staphylococcus xylosus. Front. Pharmacol. 8:543.

Google Scholar

Yan, W., Bai, R., Wang, S. S., Tian, X., Li, Y., Wang, S. S., et al. (2020). Antibiotic resistance genes are increased by combined exposure to sulfamethoxazole and naproxen but relieved by low-salinity. Environ. Int. 139:105742. doi: 10.1016/j.envint.2020.105742

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, S., Liao, Y., Cong, L., Lu, X., and Yang, R. (2016). In vitro interactions between non-steroidal anti-inflammatory drugs and antifungal agents against planktonic and biofilm forms of Trichosporon asahii. PLoS One 11:e0157047. doi: 10.1371/journal.pone.0157047

PubMed Abstract | CrossRef Full Text | Google Scholar

Yin, W., Wang, Y., Liu, L., and He, J. (2019). Biofilms: the microbial “protective clothing” in extreme environments. Int. J. Mol. Sci. 20:3423. doi: 10.3390/ijms20143423

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Y., Wang, G., Li, Y., Liu, Y., Song, Y., Zheng, W., et al. (2012). In vitro interactions between aspirin and amphotericin b against planktonic cells and biofilm cells of Candida albicans and C. parapsilosis. Antimicrob. Agents Chemother. 56, 3250–3260. doi: 10.1128/aac.06082-11

PubMed Abstract | CrossRef Full Text | Google Scholar

Zoller, S. D., Hegde, V., Burke, Z. D. C., Park, H. Y., Ishmael, C. R., and Blumstein, G. W. (2020). Evading the host response: Staphylococcus “hiding” in cortical bone canalicular system causes increased bacterial burden. Bone Res. 8:43.

Google Scholar

Keywords: non-steroidal anti-inflammatory drugs, biofilm, Staphylococcus aureus, gram-negative pathogens, antibiotic resistance

Citation: Paes Leme RC and da Silva RB (2021) Antimicrobial Activity of Non-steroidal Anti-inflammatory Drugs on Biofilm: Current Evidence and Potential for Drug Repurposing. Front. Microbiol. 12:707629. doi: 10.3389/fmicb.2021.707629

Received: 10 May 2021; Accepted: 05 July 2021;
Published: 27 July 2021.

Edited by:

Michal Letek, Universidad de León, Spain

Reviewed by:

Katharina Richter, University of Adelaide, Australia
Oana Ciofu, University of Copenhagen, Denmark

Copyright © 2021 Paes Leme and da Silva. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Rodrigo Cuiabano Paes Leme, cuiabano.leme@unifesp.br

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.