ABSTRACT
Otilonium bromide is a poorly absorbed oral medication used to control irritable bowel syndrome. It is thought to act as a muscle relaxant in the intestine. Here, we show that otilonium bromide has broad-spectrum antibacterial and antifungal activity, including against multidrug-resistant strains. Our results suggest otilonium bromide acts on enteric pathogens and may offer a new scaffold for poorly absorbed intestinal antimicrobial therapy.
KEYWORDS: otilonium bromide, antibacterial, antifungal
INTRODUCTION
Otilonium bromide (OB) is an effective orally administered therapy for treating irritable bowel syndrome (IBS), and it is considered to act as a smooth-muscle relaxant (1–6). Studies indicate that OB can act on several proteins, including calcium channels, muscarinic receptors, and tachykinin receptors, to affect muscle contraction (3, 7–10). As a poorly absorbed compound, OB is reported to be almost entirely excreted in feces and also largely unchanged during intestinal passage (3, 11). While the mechanism of action of OB is complex, it has clear benefits in alleviating patient symptoms. IBS is a multifaceted disease, and, more recently, changes in the composition of our gut microbiome have been associated with its onset and symptoms (12–14). Antibiotics, typically used to treat traveler’s diarrhea, such as rifaximin, can also control IBS symptoms, supporting a role for bacteria in IBS etiology (15–17).
Our previous studies of quaternary amine compounds led us to discover that OB had antibacterial activity against Acinetobacter baumannii, Staphylococcus aureus, and Clostridioides difficile (18). Further evaluation of OB’s substructure shows that it is related to the salicylanilide class of small molecules (Fig. 1A). Salicylanilide derivatives can have antibacterial activity against Gram-positive bacteria but generally lack activity against Gram-negative bacteria (19–21). The salicylanilide-like core of OB is appended with a quaternary amine and an eight-carbon alkyl chain. Quaternary amine groups are found in many classes of molecules, including neurotransmitters and biocides (22, 23). As biocides, lone quaternary amines require an alkyl chain length typically of ≥12 carbons for antimicrobial activity (24, 25), but this chain length is cytotoxic (26, 27). The eight-carbon chain length of OB is below the typical cytotoxic length and similar in length to the alkyl chains found on daptomycin and polymyxins. Both the quaternary amine and the alkyl chain likely promote bacterial membrane interactions. Thus, OB appears to be a combination of two antimicrobial backbones (salicylanilide and quaternary amine biocide), which could give it broad-spectrum antibacterial activity.
FIG 1.

(A) Structure of OB (top), salicylanilide (blue), and general quaternary amine biocide (red). Counter ions have been excluded for clarity. Related structures are color matched. (B and C) Time-kill curve of untreated or 0.5×, 1×, and 2× OB MIC-treated E. coli H10407 (B) and S. aureus USA100 635 (C). Cultures were serially diluted and spotted to determine viable numbers of CFU/ml at the indicated time points. Plots show the means and standard errors of the means from 6 biological replicates. The limit of detection was 102 CFU/ml.
We tested OB against a range of Gram-negative and Gram-positive bacteria, focusing on enteric pathogens, potentially pathogenic gut colonizers, and drug-resistant strains (Tables 1 and 2). For comparison we included rifaximin, another poorly absorbed broad-spectrum antibiotic of the rifampin family (15–17). We also tested carbenicillin against Gram-negative bacteria and vancomycin against Gram-positive bacteria. MICs were determined using Mueller-Hinton broth dilution methods in at least biological triplicate (28, 29). Changes in growth medium used for some strains are noted in the tables. OB stock solutions were dissolved in water at 5 mg/ml.
TABLE 1.
MIC of otilonium bromide, rifaximin, and carbenicillin toward Gram-negative bacteria
| Strain | Note | MICa (μg/ml) of: |
||
|---|---|---|---|---|
| OB | Rifaximin | Carbenicillin | ||
| Salmonella enterica serovar Typhimurium LT2 | 32 | 8 | 8 | |
| S. Typhimurium 14028S | 32 | 8 | 8 | |
| E. coli B7A | 32 | 8 | 8 | |
| E. coli H10407 | 32 | 8 | 8 | |
| E. coli O157 (CDC 0427) | MDR | 32 | 4 | >256 |
| E. coli O157 (CDC 0429) | MDR | 32 | 4 | 128 |
| C. jejuni CG8421 | 1 | 32 | ND | |
| C. jejuni 81-176 | 4 | >128 | ND | |
| A. baumannii AYE | MDR | 16 | 16 | >256 |
| A. baumannii 5075 | MDR | 16 | 2 | >256 |
| S. flexneri SA100 | 32 | 4 | 4 | |
| S. sonnei (CDC 0422) | MDR | 32 | 8 | >256 |
| S. flexneri (CDC 0423) | MDR | 32 | >256 | >256 |
| K. pneumoniae BAA-1705 | MDR | 64 | 64 | >256 |
| K. pneumoniae MKP103 | 64 | >256 | >256 | |
| K. pneumoniae ATCC 4208 | 64 | 16 | 4 | |
ND not determined.
TABLE 2.
MIC of otilonium bromide, rifaximin, and vancomycin toward Gram-positive bacteria
| Strain | Note | MIC (μg/ml) of: |
||
|---|---|---|---|---|
| OB | Rifaximin | Vancomycin | ||
| S. aureus USA100 635 | MRSA | 4 | <1 | 1 |
| S. aureus USA300 AH1263 | MRSA | 4 | <1 | <1 |
| S. aureus MU50 | MRSA | 4 | >256 | 2 |
| E. faecium 1230933a | VRE | 4 | 8 | >256 |
| E. faecium 1231502a | VRE | 8 | 32 | >256 |
| E. faecalis HIP11704a | VRE | 4 | 1 | >256 |
MIC determined in BHI medium.
OB was broadly active against Gram-negative bacteria, with MICs between 1 and 64 μg/ml (Table 1). The majority of strains tested had a MIC of 16 to 32 μg/ml. Campylobacter jejuni was the most sensitive species, with MICs of 1 and 4 μg/ml. Interestingly, OB was significantly more active against C. jejuni than rifaximin. A recent clinical trial using rifaximin to prevent Campylobacter infection with strain CG8421 was unsuccessful (30), possibly due to the tolerance of this strain. Klebsiella pneumoniae strains were the least sensitive to OB, with MICs of 64 μg/ml. Klebsiella species can express a thick capsule layer that can inhibit the action of antimicrobials (31, 32), which may also lead to decreased activity of OB. OB showed similar activity between multidrug-resistant (MDR) and non-MDR strains, suggesting its action is not affected by traditional mechanisms of drug resistance.
OB was more potent against Gram-positive bacteria than Gram-negative bacteria (Table 2). Nearly all strains tested had a MIC of 4 μg/ml OB. This MIC was quite consistent compared to those of rifaximin and vancomycin, which ranged from <1 μg/ml to >256 μg/ml depending on the strain. Similar to our observations with Gram-negative bacteria, OB activity was largely unaffected by current drug resistance mechanisms present in the Gram-positive bacteria tested.
We performed time-kill studies using representative Gram-negative (Escherichia coli H10407) and Gram-positive (S. aureus USA100 635) strains (Fig. 1B and C). The same medium and inoculum used for determining MICs were used for these time-kill studies. Exponentially growing bacterial cultures were untreated or treated with 0.5×, 1×, or 2× OB MIC. At the indicated time points, samples were drawn, serially diluted, and plated to determine remaining viable CFU numbers (CFU/ml). We found 0.5× MIC had only a slight effect on E. coli viability in the first hour before growth resumed. In contrast, both 1× and 2× OB MIC E. coli cultures quickly lost viability below the level of detection of our assay (102 CFU/ml) within the first 0.5 to 1 h. Membrane-disrupting agents frequently cause rapid loss of Gram-negative viability (33, 34). In comparison, 0.5×, 1×, and 2× MIC treatment of S. aureus cultures resulted in a graded dose-dependent loss in viability over the time course. OB at 2× MIC caused loss of S. aureus viability below our detection limit by 4 h. At 1× MIC, viable S. aureus cells were still observed at 4 h. The chemical nature of OB suggests a membrane lytic mechanism of action, which is supported by our previous studies that showed OB promoted the influx of a cell membrane-impermeable dye (propidium iodide) into the bacterial cytoplasm (18). The difference in MIC and time-kill results between E. coli and S. aureus may be related to OB accumulation at the cytoplasmic membrane, required to induce cell death. A critical OB concentration may be required to first breach the outer membrane of Gram-negative bacteria, like E. coli, leading to an all-or-nothing loss in viability at the MIC, whereas no such barrier is present for Gram-positive bacteria.
Bacteria generally show low rates of spontaneous resistance to membrane-active antimicrobials. Consistent with a membrane-targeting mechanism of action, we observed low spontaneous OB resistance of both E. coli and S. aureus compared to rifaximin (Table 3). Bacteria were plated on solid growth medium plates with 5× MIC OB or rifaximin, and resistant colonies were identified. We have been unable to isolate any OB-resistant strains after repeated attempts.
TABLE 3.
Rate of emergence of mutants spontaneously resistant to rifaximin and OB
| Strain | Concn | Rifaximina | OBa,b |
|---|---|---|---|
| S. aureus USA100 635c | 5× MIC | 2.8 × 10−7 | >10−9 |
| E. coli H10407d | 5× MIC | 2.3 × 10−8 | >10−9 |
The resistance frequency is expressed as the mean from three biological replicates.
The detection limit for this assay was 10−9.
TSA plates were used.
RPMI 1640 plates were used.
While less studied than bacteria, fungi are also important intestinal colonizers that influence our health and disease (35, 36). Colonization of Candida species is associated with their subsequent infection (37, 38). We tested three fungal pathogens and found all were susceptible to OB between 0.5 and 4 μg/ml (Table 4). In each case, OB was much more active than the antifungal fluconazole.
TABLE 4.
MIC of otilonium bromide and fluconazole toward fungi
| Straina | MIC (μg/ml) |
|
|---|---|---|
| OB | Fluconazole | |
| C. albicans SC5314 | 1 | 2 |
| C. parapsilopsis ATCC 22019 | 0.5 | 2 |
| C. krusei ATCC 6258 | 4 | 64 |
MICs performed in YPD medium.
Our study shows that the IBS therapeutic otilonium bromide is a broadly active antibacterial and antifungal. It retains activity against drug-resistant strains and has low spontaneous levels of resistance. Given the importance of our microbiome in IBS, our results suggest that part of the action of OB in these patients is due to the activity against pathogenic gut colonizers. Efforts to elucidate the effect on the commensal gut microbiota are currently in progress. Furthermore, OB may represent an unexplored chemical scaffold for the development of nonabsorbed intestinal antibacterials and antifungals.
ACKNOWLEDGMENTS
We thank Lindsey Shaw and Breck Duerkop for kindly providing bacterial isolates.
This work was supported by the NIH (R01 AI125337, R01 AI148419, and R21 AI159203 to B.W.D.; R01 AI150098, R01 AI129940, and R01 AI138576 to M.S.T.; and F32 GM125264 to B.J.V.), Tito’s Handmade Vodka (to B.W.D.), and the Welch Foundation (F‐1870 to B.W.D.).
REFERENCES
- 1.Clavé P, Acalovschi M, Triantafillidis JK, Uspensky YP, Kalayci C, Shee V, Tack J, OBIS Study Investigators. 2011. Randomised clinical trial: otilonium bromide improves frequency of abdominal pain, severity of distention and time to relapse in patients with irritable bowel syndrome. Aliment Pharmacol Ther 34:432–442. 10.1111/j.1365-2036.2011.04730.x. [DOI] [PubMed] [Google Scholar]
- 2.Clavé P, Tack J. 2017. Efficacy of otilonium bromide in irritable bowel syndrome: a pooled analysis. Ther Adv Gastroenterol 10:311–322. 10.1177/1756283X16681708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Evangelista S, Traini C, Vannucchi MG. 2018. Otilonium bromide: a drug with a complex mechanism of action. Curr Pharm Des 24:1772–1779. 10.2174/1381612824666180507122935. [DOI] [PubMed] [Google Scholar]
- 4.Forte E, Pizzoferrato M, Lopetuso L, Scaldaferri F. 2012. The use of anti-spasmodics in the treatment of irritable bowel syndrome: focus on otilonium bromide. Eur Rev Med Pharmacol Sci 16:25–37. [PubMed] [Google Scholar]
- 5.Sutton JA, Kilminster SG, Mould GP. 1997. The clinical pharmacology of single doses of otilonium bromide in healthy volunteers. Eur J Clin Pharmacol 52:365–369. 10.1007/s002280050302. [DOI] [PubMed] [Google Scholar]
- 6.Triantafillidis JK, Malgarinos G. 2014. Long-term efficacy and safety of otilonium bromide in the management of irritable bowel syndrome: a literature review. Clin Exp Gastroenterol 7:75–82. 10.2147/CEG.S46291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Santicioli P, Zagorodnyuk V, Renzetti AR, Maggi CA. 1999. Antimuscarinic, calcium channel blocker and tachykinin NK2 receptor antagonist actions of otilonium bromide in the circular muscle of guinea-pig colon. Naunyn Schmiedebergs Arch Pharmacol 359:420–427. 10.1007/pl00005370. [DOI] [PubMed] [Google Scholar]
- 8.Evangelista S, Giachetti A, Chapelain B, Neliat G, Maggi CA. 1998. Receptor binding profile of otilonium bromide. Pharmacol Res 38:111–117. 10.1006/phrs.1998.0340. [DOI] [PubMed] [Google Scholar]
- 9.Martínez-Cutillas M, Gil V, Gallego D, Mañé N, Martín MT, Jiménez M. 2013. Mechanisms of action of otilonium bromide (OB) in human cultured smooth muscle cells and rat colonic strips. Neurogastroenterol Motil 25:e803–e812. 10.1111/nmo.12206. [DOI] [PubMed] [Google Scholar]
- 10.Strege PR, Evangelista S, Lyford GL, Sarr MG, Farrugia G. 2004. Otilonium bromide inhibits calcium entry through L-type calcium channels in human intestinal smooth muscle. Neurogastroenterol Motil 16:167–173. 10.1111/j.1365-2982.2004.00517.x. [DOI] [PubMed] [Google Scholar]
- 11.Evangelista S. 1999. Otilonium bromide: a selective spasmolytic for the gastrointestinal tract. J Int Med Res 27:207–222. 10.1177/030006059902700501. [DOI] [PubMed] [Google Scholar]
- 12.Halvorson HA, Schlett CD, Riddle MS. 2006. Postinfectious irritable bowel syndrome–a meta-analysis. Am J Gastroenterol 101:1894–1899. 10.1111/j.1572-0241.2006.00654.x. [DOI] [PubMed] [Google Scholar]
- 13.Klem F, Wadhwa A, Prokop LJ, Sundt WJ, Farrugia G, Camilleri M, Singh S, Grover M. 2017. Prevalence, risk factors, and outcomes of irritable bowel syndrome after infectious enteritis: a systematic review and meta-analysis. Gastroenterology 152:1042–1054. 10.1053/j.gastro.2016.12.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Tap J, Derrien M, Törnblom H, Brazeilles R, Cools-Portier S, Doré J, Störsrud S, Le Nevé B, Öhman L, Simrén M. 2017. Identification of an intestinal microbiota signature associated with severity of irritable bowel syndrome. Gastroenterology 152:111–123. 10.1053/j.gastro.2016.09.049. [DOI] [PubMed] [Google Scholar]
- 15.Pimentel M, Lembo A, Chey WD, Zakko S, Ringel Y, Yu J, Mareya SM, Shaw AL, Bortey E, Forbes WP, TARGET Study Group. 2011. Rifaximin therapy for patients with irritable bowel syndrome without constipation. N Engl J Med 364:22–32. 10.1056/NEJMoa1004409. [DOI] [PubMed] [Google Scholar]
- 16.Taylor DN, Bourgeois AL, Ericsson CD, Steffen R, Jiang Z-D, Halpern J, Haake R, Dupont HL. 2006. A randomized, double-blind, multicenter study of rifaximin compared with placebo and with ciprofloxacin in the treatment of travelers’ diarrhea. Am J Trop Med Hyg 74:1060–1066. 10.4269/ajtmh.2006.74.1060. [DOI] [PubMed] [Google Scholar]
- 17.Adachi JA, DuPont HL. 2006. Rifaximin: a novel nonabsorbed rifamycin for gastrointestinal disorders. Clin Infect Dis 42:541–547. 10.1086/499950. [DOI] [PubMed] [Google Scholar]
- 18.Knauf GA, Cunningham AL, Kazi MI, Riddington IM, Crofts AA, Cattoir V, Trent MS, Davies BW. 2018. Exploring the antimicrobial action of quaternary amines against Acinetobacter baumannii. mBio 9:e02394-17. 10.1128/mBio.02394-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Pauk K, Zadražilová I, Imramovský A, Vinšová J, Pokorná M, Masaříková M, Cížek A, Jampílek J. 2013. New derivatives of salicylamides: preparation and antimicrobial activity against various bacterial species. Bioorg Med Chem 21:6574–6581. 10.1016/j.bmc.2013.08.029. [DOI] [PubMed] [Google Scholar]
- 20.Gooyit M, Janda KD. 2016. Reprofiled anthelmintics abate hypervirulent stationary-phase Clostridium difficile. Sci Rep 6:33642. 10.1038/srep33642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Rajamuthiah R, Fuchs BB, Conery AL, Kim W, Jayamani E, Kwon B, Ausubel FM, Mylonakis E. 2015. Repurposing salicylanilide anthelmintic drugs to combat drug resistant Staphylococcus aureus. PLoS One 10:e0124595. 10.1371/journal.pone.0124595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Maillard J-Y. 2002. Bacterial target sites for biocide action. J Appl Microbiol 92(Suppl):16S–27S. 10.1046/j.1365-2672.92.5s1.3.x. [DOI] [PubMed] [Google Scholar]
- 23.Ghelardoni M, Pestellini V, Pisanti N, Volterra G. 1973. Quaternary salts of substituted 2-aminoethyl N-benzoylaminobenzoate. A new class of smooth muscle relaxant agents. J Med Chem 16:1063–1065. 10.1021/jm00267a029. [DOI] [PubMed] [Google Scholar]
- 24.Daoud NN, Dickinson NA, Gilbert P. 1983. Antimicrobial activity and physico-chemical properties of some alkyldimethylbenzylammonium chlorides. Microbios 37:73–85. [PubMed] [Google Scholar]
- 25.Jono K, Takayama T, Kuno M, Higashide E. 1986. Effect of alkyl chain length of benzalkonium chloride on the bactericidal activity and binding to organic materials. Chem Pharm Bull 34:4215–4224. 10.1248/cpb.34.4215. [DOI] [PubMed] [Google Scholar]
- 26.Cadwallader DE, Ansel HC. 1965. Hemolysis of erythrocytes by antibacterial preservatives. II. Quaternary ammonium salts. J Pharm Sci 54:1010–1012. 10.1002/jps.2600540712. [DOI] [PubMed] [Google Scholar]
- 27.Pantaler E, Kamp D, Haest CW. 2000. Acceleration of phospholipid flip-flop in the erythrocyte membrane by detergents differing in polar head group and alkyl chain length. Biochim Biophys Acta 1509:397–408. 10.1016/s0005-2736(00)00322-9. [DOI] [PubMed] [Google Scholar]
- 28.Wiegand I, Hilpert K, Hancock REW. 2008. Agar and broth dilution methods to determine the minimal inhibitory concentration (MIC) of antimicrobial substances. Nat Protoc 3:163–175. 10.1038/nprot.2007.521. [DOI] [PubMed] [Google Scholar]
- 29.Esarte Palomero O, Cunningham AL, Davies BW, Jones RA. 2021. Antibacterial thiamine inspired silver (I) and gold (I) N-heterocyclic carbene compounds. Inorganica Chim Acta 517:120152. 10.1016/j.ica.2020.120152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Rimmer JE, Harro C, Sack DA, Talaat KR, Gutierrez RL, DeNearing B, Brubaker J, Laird RM, Poly F, Maue AC, Jaep K, Alcala A, Mochalova Y, Gariepy CL, Chakraborty S, Guerry P, Tribble DR, Porter CK, Riddle MS. 2018. Rifaximin fails to prevent campylobacteriosis in the human challenge model: a randomized, double-blind, placebo-controlled trial. Clin Infect Dis 66:1435–1441. 10.1093/cid/cix1014. [DOI] [PubMed] [Google Scholar]
- 31.Fleeman RM, Macias LA, Brodbelt JS, Davies BW. 2020. Defining principles that influence antimicrobial peptide activity against capsulated Klebsiella pneumoniae. Proc Natl Acad Sci U S A 117:27620–27626. 10.1073/pnas.2007036117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Campos MA, Vargas MA, Regueiro V, Llompart CM, Albertí S, Bengoechea JA. 2004. Capsule polysaccharide mediates bacterial resistance to antimicrobial peptides. Infect Immun 72:7107–7114. 10.1128/IAI.72.12.7107-7114.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Dong N, Chou S, Li J, Xue C, Li X, Cheng B, Shan A, Xu L. 2018. Short symmetric-end antimicrobial peptides centered on β-turn amino acids unit improve selectivity and stability. Front Microbiol 9:2832. 10.3389/fmicb.2018.02832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kawamura-Sato K, Wachino J, Kondo T, Ito H, Arakawa Y. 2010. Correlation between reduced susceptibility to disinfectants and multidrug resistance among clinical isolates of Acinetobacter species. J Antimicrob Chemother 65:1975–1983. 10.1093/jac/dkq227. [DOI] [PubMed] [Google Scholar]
- 35.Septimus EJ, Schweizer ML. 2016. Decolonization in prevention of health care-associated infections. Clin Microbiol Rev 29:201–222. 10.1128/CMR.00049-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Huffnagle GB, Noverr MC. 2013. The emerging world of the fungal microbiome. Trends Microbiol 21:334–341. 10.1016/j.tim.2013.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Cole GT, Halawa AA, Anaissie EJ. 1996. The role of the gastrointestinal tract in hematogenous candidiasis: from the laboratory to the bedside. Clin Infect Dis 22(Suppl 2):S73–S88. 10.1093/clinids/22.supplement_2.s73. [DOI] [PubMed] [Google Scholar]
- 38.Miranda LN, van der Heijden IM, Costa SF, Sousa API, Sienra RA, Gobara S, Santos CR, Lobo RD, Pessoa VP, Levin AS. 2009. Candida colonisation as a source for candidaemia. J Hosp Infect 72:9–16. 10.1016/j.jhin.2009.02.009. [DOI] [PubMed] [Google Scholar]
