Next Article in Journal
Avirulent Isolates of Penicillium chrysogenum to Control the Blue Mold of Apple Caused by P. expansum
Next Article in Special Issue
Metabolome and Transcriptome Combinatory Profiling Reveals Fluconazole Resistance Mechanisms of Trichosporon asahii and the Role of Farnesol in Fluconazole Tolerance
Previous Article in Journal
Prevalence of Diarrhoeagenic Escherichia coli among Children Aged between 0–36 Months in Peri-Urban Areas of Lusaka
Previous Article in Special Issue
Uncovering the Resistance Mechanisms in Extended-Drug-Resistant Pseudomonas aeruginosa Clinical Isolates: Insights from Gene Expression and Phenotypic Tests
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Preliminary SAR of Novel Pleuromutilin–Polyamine Conjugates

by
Kenneth Sue
1,
Melissa M. Cadelis
1,2,
Kerrin Hainsworth
1,
Florent Rouvier
3,
Marie-Lise Bourguet-Kondracki
4,
Jean Michel Brunel
3 and
Brent R. Copp
1,*
1
School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
2
School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
3
Membranes et Cibles Thérapeutiques, INSERM, Aix-Marseille Universite, 27 bd Jean Moulin, 13385 Marseille, France
4
Laboratoire Molécules de Communication et Adaptation des Micro-organismes, UMR 7245 CNRS, Muséum National d’Histoire Naturelle, 57 rue Cuvier (C.P. 54), 75005 Paris, France
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(11), 2791; https://doi.org/10.3390/microorganisms11112791
Submission received: 25 September 2023 / Revised: 3 November 2023 / Accepted: 13 November 2023 / Published: 17 November 2023
(This article belongs to the Special Issue Advances in Antibiotic and Drug-Resistance Mechanisms)

Abstract

:
While pleuromutilin (1) and its clinically available derivatives (26) are highly effective against Gram-positive bacteria, they remain inactive against many pathogenic Gram-negative bacteria due to the efflux pump AcrAB-TolC. In an effort to broaden the spectrum of activity of pleuromutilin (1), we developed a series of novel pleuromutilin–polyamine conjugates (9af) which exhibited promising intrinsic antimicrobial properties, targeting both Gram-positive and Gram-negative bacteria, including Staphylococcus aureus, methicillin-resistant S. aureus (MRSA), and Escherichia coli, along with the fungal strain Cryptococcus neoformans, and were devoid of cytotoxic and hemolytic properties with the exception of one conjugate. Furthermore, this series displayed moderate to low antibiotic potentiation of legacy antibiotics doxycycline and erythromycin, with three conjugates enhancing the activity four-fold in combination with doxycycline. In comparison to pleuromutilin (1) and tiamulin (2), one of the conjugates exhibited an expanded spectrum of activity, including Gram-negative bacteria and fungi, making it a promising option for combating microbial infections.

1. Introduction

The rise in antimicrobial resistance, caused by the excessive use of antibiotics and a lack of investment towards the discovery of new antibiotics, has resulted in a growing number of challenging microbial infections [1,2,3,4,5,6]. The discovery of new antibiotics is one avenue for overcoming resistant microbes, while another is the discovery of compounds with little or no antimicrobial activity which can be used in combination with legacy antibiotics to restore their activities [6].
Upon its discovery in 1951, the diterpenoid antibiotic pleuromutilin (1), from the fungi Pleurotus mutilus (currently known as Clitopilus scyphoides) and P. passeckerianus (C. passeckerianus), was reported to exhibit bactericidal activity against Staphylococcus aureus and moderate activity against Mycobacterium smegmatis but showed no effect against Escherichia coli [7,8,9,10]. However, the exact structure of pleuromutilin (1) (Figure 1) remained unknown until a decade after its initial discovery, after which researchers synthesized derivatives focusing on substituting the hydroxyl group at C-22 [8,10,11]. This led to the development of tiamulin (2), an analogue achieved through substitution with sulphonate esters, which gained approval for veterinary use in 1979. While these derivatives, including the second-generation pleuromutilin antibiotic valnemulin (3), which boasted improved potency, were successful in veterinary applications, pharmaceutical companies aimed to develop pleuromutilin antibiotics for human use [8,10,11]. In 1982, progress was made towards this goal with azamulin (4) entering phase I clinical trials, though it failed to pass due to poor bioavailability [8,12]. Nevertheless, the breakthrough came with retapamulin (5), a potent pleuromutilin analogue approved by the FDA in 2007 for topical infections, sparking renewed interest in pleuromutilin and its derivatives [8,10,13]. More recently, lefamulin (6), another pleuromutilin derivative, successfully passed phase III clinical trials for bacterial pneumonia and improved upon retapamulin (5) as it could be administered orally or intravenously [14,15].
Lefamulin displayed potent antibacterial activity against aerobic Gram-positive bacteria, proving effective against challenging strains such as methicillin-resistant S. aureus (MRSA), vancomycin-intermediate S. aureus (VISA), heterogeneous VISA (hVISA), vancomycin-resistant S. aureus (VRSA), penicillin-resistant Streptococcus pneumoniae (PRSP), multidrug-resistant S. pneumoniae, and vancomycin-resistant Enterococcus faecium (VRE) [10,16,17,18]. Additionally, it exhibited favorable to moderate activity against fastidious Gram-negative bacteria, including Haemophilus influenza and Moraxella catarrhalis. However, like its predecessors, no activity was observed against non-fastidious Gram-negative bacteria [10,16,17,18].
The lack of activity of this structural class against Gram-negative bacteria was attributed to the expression of the efflux pump AcrAB-TolC, a tripartite efflux pump which exports small molecules from the bacterial cell, reducing drug efficacy [10,19,20]. Encouragingly, a derivative of pleuromutilin showed promise against an E. coli strain when tested in the presence of the efflux pump inhibitor Phe-Arg-β-naphthylamide [10], indicating that inhibiting AcrAB-TolC could potentially enable pleuromutilin to be used in the treatment of Gram-negative bacterial infections.
SAR studies show that pleuromutilins with thioether or basic group linkers have enhanced antibacterial activity. Recent research suggests that increasing primary amines and positive charges in antibiotics can improve their uptake and accumulation in Gram-negative bacteria, potentially broadening their spectrum beyond Gram-positives. In our previous work on α,ω polyamines with disubstituted lipophilic head groups, we have successfully demonstrated examples that inhibit AcrAB-TolC [21]. Thus, the present study aims to synthesize and evaluate a series of pleuromutilin–polyamine conjugates bearing varying polyamine chain lengths for their intrinsic antimicrobial activity and their ability to enhance the activity of doxycycline and erythromycin against Gram-negative bacteria.

2. Materials and Methods

2.1. Chemical Synthesis General Methods

2.2. Synthesis of Compounds

2.2.1. Pleuromutilin 22-O-Tosylate (8)

A solution of pleuromutilin (1) (0.50 g, 1.3 mmol), toluenesulfonylchloride (0.30 g, 1.6 mmol), and DMAP (0.48 g, 3.9 mmol) in anhydrous CH2Cl2 was stirred at 0 °C for 4 h under N2 atmosphere. The reaction was quenched with 1 N HCl and extracted twice with EtOAc. The combined organic layers were then washed with sat. aq. NaHCO3, dried with MgSO4, and concentrated under reduced pressure. The crude product was purified by diol-bonded silica gel column chromatography (20–80% EtOAc/petroleum ether) to afford (8) as a white foam (0.39 g, 57%). 1H and 13C NMR data agreed with those reported in the literature [25].

2.2.2. Tiamulin (2)

A solution of pleuromutilin 22-O-tosylate (8) (0.130 g, 0.244 mmol) and KI (0.080 g, 0.482 mmol) in MeCN (10 mL) was stirred at 70 °C under an N2 atmosphere for 30 min. A solution of 2-diethylaminoethane thiol hydrochloride (0.046 g, 0.271 mmol) and DIPEA (0.26 mL, 1.49 mmol) in anhydrous MeCN (2 mL) was then added and the reaction stirred for a further 2 h. The solvent was removed under reduced pressure, to which was added CH2Cl2 (30 mL), and the organic phase was washed with sat. aq. NaHCO3 (2 × 30 mL) and H2O (2 × 30 mL) and dried over anhydrous Na2SO4, and the solvent was removed again under reduced pressure. The crude product was purified by diol-bonded silica gel column chromatography (75–100% EtOAc/hexane followed by 100% MeOH) to afford 2 as a pale orange foam (0.057 g, 47%). Rf (silica gel, 100% EtOAc) 0.44; [ α ] D 23.3 = + 50.6 (c 0.195, CH2Cl2); IR (ATR) νmax 3444, 2929, 1721, 1455, 1277, 1115 cm−1; 1H NMR (CDCl3, 400 MHz) δ 6.47 (1H, dd, J = 17.4, 11.0 Hz, H-19), 5.74 (1H, d, J = 8.5 Hz, H-14), 5.33 (1H, dd, J = 11.0, 1.5 Hz, H2-20a), 5.19 (1H, dd, J = 17.4, 1.5 Hz, H2-20b), 3.35 (1H, d, J = 6.1 Hz, H-11), 3.16 (2H, s, H2-22), 2.68 (4H, s, H2-23, H2-24), 2.53 (4H, q, J = 7.1 Hz, 2H2-25), 2.38–2.32 (1H, m, H-10), 2.25–2.16 (2H, m, H2-2), 2.11–2.05 (2H, m, H-4, H2-13a), 1.79–1.74 (1H, m, H2-8a), 1.69–1.60 (2H, m, H2-1a, H-6), 1.57–1.50 (1H, m, H2-7a), 1.48–1.41 (1H, m, H2-1b), 1.45 (3H, s, H3-15), 1.39–1.32 (2H, m, H2-7b, H2-13b), 1.16 (3H, s, H3-18), 1.15–1.08 (1H, m, H2-8b), 1.02 (6H, t, J = 7.2 Hz, 2H3-26), 0.87 (3H, d, J = 7.2 Hz, H3-17), 0.73 (3H, d, J = 6.9 Hz, H3-16), OH signal not observed; 13C NMR (CDCl3, 100 MHz) δ 217.2 (C-3), 169.1 (C-21), 139.2 (C-19), 117.3 (C-20), 74.7 (C-11), 69.4 (C-14), 58.3 (C-4), 52.0 (C-24), 46.9 (C-25), 45.6 (C-9), 44.9 (C-13), 44.0 (C-12), 41.9 (C-5), 36.9 (C-6), 36.1 (C-10), 34.63 (C-22), 34.58 (C-2), 30.5 (C-8), 29.4 (C-23), 27.0 (C-7), 26.5 (C-18), 25.0 (C-1), 17.0 (C-16), 15.0 (C-15), 11.6 (C-17), 11.2 (C-26); (+)-HRESIMS m/z 494.3287 [M + H]+ (calculated for C28H48NO4S, 494.3299).

2.2.3. N1,N1′-(Butane-1,4-diyl)bis(N3-(2-(((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl)oxy)-2-oxoethyl)propane-1,3-diaminium) 2,2,2-trifluoroacetate (9a)

Following general procedure A, the reaction of pleuromutilin 22-O-tosylate (8) (0.025 g, 0.047 mmol), KI (0.009 g, 0.054 mmol), DIPEA (0.025 mL, 0.144 mmol), and di-tert-butyl butane-1,4-diylbis((3-aminopropyl)carbamate) (7a) (0.009 g, 0.024 mmol) afforded bis((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl) 7,12-bis(tert-butoxycarbonyl)-3,7,12,16-tetraazaoctadecanedioate (0.019 g, 72%) as a yellow oil. Following general procedure B, a sub-sample of the protected product (0.010 g, 0.009 mmol) was reacted with TFA in CH2Cl2 to afford the tetra-TFA salt 9a (0.008 g, 65%) as a yellow oil. [ α ] D 18.8 = + 11 (c 0.1, MeOH); Rf (RP-18, 10% aq HCl:MeOH 1:3) 0.57; IR (ATR) νmax 2922, 1732, 1671, 1420, 1179, 1121, 1020, 914, 833, 798, 720 cm−1; 1H NMR (DMSO-d6, 400 MHz) δ 9.34 (4H, br s, NH2-23), 8.82 (4H, br s, NH2-27), 6.14 (2H, dd, J = 17.8, 11.2 Hz, H-19), 5.63 (2H, d, J = 8.2 Hz, H-14), 5.14 (2H, dd, J = 17.8, 1.4 Hz, H2-20a), 5.06 (2H, dd, J = 11.2, 1.1 Hz, H2-20b), 4.05 (2H, d, J = 17.9 Hz, H2-22a), 3.86 (2H, d, J = 17.4 Hz, H2-22b), 3.45 (2H, d, J = 8.2 Hz, H-11), 3.02–2.95 (8H, m, H2-24, H2-26), 2.95–2.89 (4H, m, H2-28), 2.46 (2H, s, H-4), 2.24–1.95 (12H, m, H2-2, H-10, H2-13a, H2-25), 1.69–1.62 (8H, m, H2-1b, H2-8a, H2-29), 1.56–1.50 (2H, m, H-6), 1.40–1.23 (8H, m, H2-1a, H2-7, H2-13b), 1.38 (6H, s, H3-15), 1.08 (6H, s, H3-18), 1.03–0.99 (2H, m, H2-8b), 0.83 (6H, d, J = 6.8 Hz, H3-17), 0.65 (6H, d, J = 6.8 Hz, H3-16); 13C NMR (DMSO-d6, 100 MHz) δ 217.0 (C-3), 165.6 (C-21), 140.8 (C-19), 115.5 (C-20), 72.4 (C-11), 71.1 (C-14), 57.0 (C-4), 47.4 (C-22), 46.1 (C-28), 44.9 (C-9), 44.2 (C-12), 44.0 (C-24/C-26), 43.8 (C-24/C-26), 43.3 (C-13), 41.5 (C-5), 36.5 (C-10), 36.2 (C-6), 34.0 (C-2), 30.0 (C-8), 28.5 (C-18), 26.6 (C-7), 24.4 (C-1), 22.7 (C-29), 22.1 (C-25), 16.2 (C-16), 14.4 (C-15), 11.6 (C-17); (+)-HRESIMS [M + H]+ m/z 923.6834 (calculated for C54H91N4O8, 923.6831).

2.2.4. N1,N1′-(Hexane-1,6-diyl)bis(N3-(2-(((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl)oxy)-2-oxoethyl)propane-1,3-diaminium) 2,2,2-trifluoroacetate (9b)

Following general procedure A, the reaction of pleuromutilin 22-O-tosylate (8) (0.050 g, 0.094 mmol), KI (0.017 g, 0.103 mmol), DIPEA (0.049 mL, 0.281 mmol), and di-tert-butyl hexane-1,6-diylbis((3-aminopropyl)carbamate) (7b) (0.020 g, 0.046 mmol) afforded bis((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl) 7,14-bis(tert-butoxycarbonyl)-3,7,14,18-tetraazaicosanedioate (0.028 g, 52%) as a yellow oil. Following general procedure B, a sub-sample of the protected product (0.014 g, 0.012 mmol) was reacted with TFA in CH2Cl2 to afford, after chromatography, the tetra-TFA salt 9b (0.009 g, 53%) as a yellow oil. [ α ] D 19.9 = + 13 (c 0.1, MeOH); Rf (RP-18, 10% aq HCl:MeOH 1:3) 0.50; IR (ATR) νmax 2928, 1739, 1671, 1464, 1413, 1199, 1180, 1131, 798, 721 cm−1; 1H NMR (DMSO-d6, 400 MHz) δ 9.32 (4H, br s, NH2-23), 8.69 (4H, br s, NH2-27), 6.15 (2H, dd, J = 17.8, 11.2 Hz, H-19), 5.63 (2H, d, J = 8.3 Hz, H-14), 5.14 (2H, dd, J = 17.5, 1.5 Hz, H2-20a), 5.07 (2H, dd, J = 11.1, 1.3 Hz, H2-20b), 4.61 (2H, br s, OH-11), 4.05 (2H, d, J = 16.9 Hz, H2-22a), 3.85 (2H, d, J = 16.7 Hz, H2-22b), 3.45 (2H, obscured by H2O, H-11), 3.00–2.93 (8H, m, H2-24, H2-26), 2.90–2.84 (4H, m, H2-28), 2.46 (2H, s, H-4), 2.24–1.94 (12H, m, H2-2, H-10, H2-13a, H2-25), 1.70–1.62 (8H, m, H2-1b, H2-8a, H2-29), 1.60–1.51 (2H, m, H-6), 1.46–1.36 (6H, m, H2-7, H2-13b), 1.38 (6H, s, H3-15), 1.33–1.24 (6H, s, H2-1a, H2-30), 1.08 (6H, s, H3-18), 1.03–1.00 (2H, m, H2-8b), 0.83 (6H, d, J = 6.9 Hz, H3-17), 0.65 (6H, d, J = 7.0 Hz, H3-16); 13C NMR (DMSO-d6, 100 MHz) δ 217.0 (C-3), 165.6 (C-21), 140.8 (C-19), 115.5 (C-20), 72.4 (C-11), 71.1 (C-14), 57.0 (C-4), 47.4 (C-22), 46.7 (C-28), 44.9 (C-9), 44.2 (C-12), 44.0 (C-24/C-26), 43.8 (C-24/C-26), 43.2 (C-13), 41.5 (C-5), 36.4 (C-10), 36.2 (C-6), 34.0 (C-2), 30.0 (C-8), 28.5 (C-18), 26.6 (C-7), 25.8 (C-30), 25.4 (C-29), 24.4 (C-1), 22.1 (C-25), 16.2 (C-16), 14.4 (C-15), 11.6 (C-17); (+)-HRESIMS [M + H]+ m/z 951.7142 (calculated for C56H95N4O8, 951.7144).

2.2.5. N1,N1′-(Heptane-1,7-diyl)bis(N3-(2-(((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl)oxy)-2-oxoethyl)propane-1,3-diaminium) 2,2,2-trifluoroacetate (9c)

Following general procedure A, the reaction of pleuromutilin 22-O-tosylate (8) (0.050 g, 0.094 mmol), KI (0.017 g, 0.103 mmol), DIPEA (0.049 mL, 0.281 mmol), and di-tert-butyl heptane-1,7-diylbis((3-aminopropyl)carbamate) (7c) (0.021 g, 0.047 mmol) afforded bis((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl) 7,15-bis(tert-butoxycarbonyl)-3,7,15,19-tetraazahenicosanedioate (0.032 g, 58%) as a yellow oil. Following general procedure B, a sub-sample of the protected product (0.016 g, 0.014 mmol) was reacted with TFA in CH2Cl2 to afford, after chromatography, the tetra-TFA salt 9c (0.014 g, 72%) as a yellow oil. [ α ] D 21.5 = +5 (c 0.1, MeOH); Rf (RP-18, 10% aq HCl:MeOH 1:3) 0.48; IR (ATR) νmax 2944, 1736, 1676, 1460, 1418, 1200, 1178, 1129, 1051, 1026, 799, 721 cm−1; 1H NMR (DMSO-d6, 400 MHz) δ 9.35 (4H, br s, NH2-23), 8.74 (4H, br s, NH2-27), 6.14 (2H, dd, J = 17.8, 11.2 Hz, H-19), 5.63 (2H, d, J = 8.3 Hz, H-14), 5.14 (2H, d, J = 17.8 Hz, H2-20a), 5.06 (2H, d, J = 11.3 Hz, H2-20b), 4.05 (2H, d, J = 17.7 Hz, H2-22a), 3.85 (2H, d, J = 16.7 Hz, H2-22b), 3.45 (2H, d, J = 5.5 Hz, H-11), 3.00–2.94 (8H, m, H2-24, H2-26), 2.90–2.84 (4H, m, H2-28), 2.46 (2H, s, H-4), 2.24–1.95 (12H, m, H2-2, H-10, H2-13a, H2-25), 1.69–1.62 (8H, m, H2-1b, H2-8a, H2-29), 1.59–1.50 (2H, m, H-6), 1.40–1.37 (6H, m, H2-1a, H2-7b, H2-13b), 1.38 (6H, s, H3-15), 1.32–1.24 (8H, m, H2-7a, H2-30, H2-31), 1.08 (6H, s, H3-18), 1.03–1.00 (2H, m, H2-8b), 0.83 (6H, d, J = 6.8 Hz, H3-17), 0.65 (6H, d, J = 6.9 Hz, H3-16); 13C NMR (DMSO-d6, 100 MHz) δ 217.0 (C-3), 165.6 (C-21), 140.8 (C-19), 115.5 (C-20), 72.4 (C-11), 71.1 (C-14), 57.0 (C-4), 47.4 (C-22), 46.7 (C-28), 44.9 (C-9), 44.2 (C-12), 44.0 (C-24/C-26), 43.8 (C-24/C-26), 43.2 (C-13), 41.5 (C-5), 36.4 (C-10), 36.2 (C-6), 34.0 (C-2), 30.0 (C-8), 28.5 (C-18), 28.1 (C-31), 26.6 (C-7), 25.8 (C-30), 25.3 (C-29), 24.4 (C-1), 22.1 (C-25), 16.2 (C-16), 14.4 (C-15), 11.6 (C-17); (+)-HRESIMS [M + 2H]+ m/z 483.3684 (calculated for C57H98N4O8, 483.3687).

2.2.6. N1,N1′-(Octane-1,8-diyl)bis(N3-(2-(((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl)oxy)-2-oxoethyl)propane-1,3-diaminium) 2,2,2-trifluoroacetate (9d)

Following general procedure A, the reaction of pleuromutilin 22-O-tosylate (8) (0.050 g, 0.094 mmol), KI (0.017 g, 0.103 mmol), DIPEA (0.049 mL, 0.281 mmol), and di-tert-butyl octane-1,8-diylbis((3-aminopropyl)carbamate) (7d) (0.022 g, 0.047 mmol) afforded bis((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl) 7,16-bis(tert-butoxycarbonyl)-3,7,16,20-tetraazadocosanedioate (0.031 g, 56%) as a yellow oil. Following general procedure B, a sub-sample of the protected product (0.015 g, 0.013 mmol) was reacted with TFA in CH2Cl2 to afford, after chromatography, the tetra-TFA salt 9d (0.018 g, 99%) as a yellow oil. [ α ] D 20.0 = +5 (c 0.1, MeOH); Rf (RP-18, 10% aq HCl:MeOH 1:3) 0.48; IR (ATR) νmax 2945, 1733, 1670, 1464, 1421, 1202, 1180, 1125, 1025, 798, 720 cm−1; 1H NMR (DMSO-d6, 400 MHz) δ 9.33 (4H, br s, NH2-23), 8.68 (4H, br s, NH2-27), 6.14 (2H, dd, J = 17.8, 11.3 Hz, H-19), 5.63 (2H, d, J = 8.4 Hz, H-14), 5.14 (2H, dd, J = 17.8, 1.7 Hz, H2-20a), 5.06 (2H, dd, J = 11.2, 1.7 Hz, H2-20b), 4.05 (2H, d, J = 17.5 Hz, H2-22a), 3.85 (2H, d, J = 16.9 Hz, H2-22b), 3.45 (2H, d, J = 5.7 Hz, H-11), 3.00–2.94 (8H, m, H2-24, H2-26), 2.90–2.85 (4H, m, H2-28), 2.45 (2H, s, H-4), 2.22–2.03 (12H, m, H2-2, H-10, H2-13a), 1.97 (4H, tt, J = 7.7, 7.6 Hz, H2-25), 1.69–1.62 (4H, m, H2-1b, H2-8a), 1.56–1.50 (6H, m, H-6, H2-29), 1.41–1.37 (4H, m, H2-7b, H2-13b), 1.38 (6H, s, H3-15), 1.30–1.23 (12H, m, H2-1a, H2-7a, H2-30, H2-31), 1.08 (6H, s, H3-18), 1.03–0.98 (2H, m, H2-8b), 0.83 (6H, d, J = 7.0 Hz, H3-17), 0.65 (6H, d, J = 7.0 Hz, H3-16); 13C NMR (DMSO-d6, 100 MHz) δ 217.0 (C-3), 165.6 (C-21), 140.8 (C-19), 115.5 (C-20), 72.4 (C-11), 71.1 (C-14), 57.0 (C-4), 47.4 (C-22), 46.6 (C-28), 44.9 (C-9), 44.2 (C-12), 44.0 (C-24/C-26), 43.8 (C-24/C-26), 43.2 (C-13), 41.5 (C-5), 36.5 (C-10), 36.2 (C-6), 34.0 (C-2), 30.0 (C-8), 28.5 (C-18, C-31), 26.6 (C-7), 25.6 (C-30), 25.3 (C-29), 24.4 (C-1), 22.1 (C-25), 16.2 (C-16), 14.4 (C-15), 11.6 (C-17); (+)-HRESIMS [M + 2H]+ m/z 490.3763 (calculated for C58H100N4O8, 490.3765).

2.2.7. N1,N1′-(Decane-1,10-diyl)bis(N3-(2-(((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl)oxy)-2-oxoethyl)propane-1,3-diaminium) 2,2,2-trifluoroacetate (9e)

Following general procedure A, the reaction of pleuromutilin 22-O-tosylate (8) (0.050 g, 0.094 mmol), KI (0.017 g, 0.103 mmol), DIPEA (0.049 mL, 0.281 mmol), and di-tert-butyl decane-1,10-diylbis((3-aminopropyl)carbamate) (7e) (0.023 g, 0.047 mmol) afforded bis((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl) 7,18-bis(tert-butoxycarbonyl)-3,7,18,22-tetraazatetracosanedioate (0.028 g, 49%) as a yellow oil. Following general procedure B, a sub-sample of the protected product (0.014 g, 0.012 mmol) was reacted with TFA in CH2Cl2 to afford, after chromatography, the tetra-TFA salt 9e (0.011 g, 65%) as a yellow oil. [ α ] D 19.2 = +4 (c 0.1, MeOH); Rf (RP-18, 10% aq HCl:MeOH 1:3) 0.47; IR (ATR) νmax 2922, 1738, 1669, 1417, 1199, 1129, 1019, 915, 835, 797, 721 cm−1; 1H NMR (DMSO-d6, 400 MHz) δ 9.33 (4H, br s, NH2-23), 8.68 (4H, br s, NH2-27), 6.14 (2H, dd, J = 17.8, 11.2 Hz, H-19), 5.63 (2H, d, J = 8.3 Hz, H-14), 5.14 (2H, dd, J = 17.8, 1.6 Hz, H2-20a), 5.06 (2H, dd, J = 11.2, 1.5 Hz, H2-20b), 4.05 (2H, d, J = 16.6 Hz, H2-22a), 3.86 (2H, d, J = 17.4 Hz, H2-22b), 3.45 (2H, d, J = 5.5 Hz, H-11), 3.00–2.93 (8H, m, H2-24, H2-26), 2.90–2.84 (4H, m, H2-28), 2.46 (2H, s, H-4), 2.24–2.01 (8H, m, H2-2, H-10, H2-13a), 1.96 (4H, tt, J = 7.5, 7.5 Hz, H2-25), 1.69–1.60 (8H, m, H2-1b, H2-8a, H2-29), 1.56–1.50 (2H, m, H-6), 1.40–1.36 (4H, m, H2-7b, H2-13b), 1.38 (6H, s, H3-15), 1.30–1.29 (4H, m, H2-1a, H-7a), 1.28–1.24 (12H, m, H2-30, H2-31, H2-32), 1.08 (6H, s, H3-18), 1.03–0.99 (2H, m, H2-8b), 0.83 (6H, d, J = 6.9 Hz, H3-17), 0.65 (6H, d, J = 7.0 Hz, H3-16); 13C NMR (DMSO-d6, 100 MHz) δ 217.0 (C-3), 165.6 (C-21), 140.8 (C-19), 115.5 (C-20), 72.4 (C-11), 71.1 (C-14), 57.0 (C-4), 47.3 (C-22), 46.7 (C-28), 44.9 (C-9), 44.2 (C-12), 44.0 (C-24/C-26), 43.8 (C-24/C-26), 43.2 (C-13), 41.5 (C-5), 36.4 (C-10), 36.2 (C-6), 34.0 (C-2), 30.0 (C-8), 28.7 (C-32), 28.54 (C-18/C-31), 28.51 (C-18/C-31), 26.6 (C-7), 25.9 (C-30), 25.4 (C-29), 24.4 (C-1), 22.1 (C-25), 16.2 (C-16), 14.4 (C-15), 11.6 (C-17); (+)-HRESIMS [M + 2H]+ m/z 504.3922 (calculated for C60H104N4O8, 504.3922).

2.2.8. N1,N1′-(Dodecane-1,12-diyl)bis(N3-(2-(((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl)oxy)-2-oxoethyl)propane-1,3-diaminium) 2,2,2-trifluoroacetate (9f)

Following general procedure A, the reaction of pleuromutilin 22-O-tosylate (8) (0.050 g, 0.094 mmol), KI (0.017 g, 0.103 mmol), DIPEA (0.049 mL, 0.281 mmol), and di-tert-butyl dodecane-1,12-diylbis((3-aminopropyl)carbamate) (7f) (0.024 g, 0.047 mmol) afforded bis((3aR,4R,5R,7S,8S,9R,9aS,12R)-8-hydroxy-4,7,9,12-tetramethyl-3-oxo-7-vinyldecahydro-4,9a-propanocyclopenta [8]annulen-5-yl) 7,20-bis(tert-butoxycarbonyl)-3,7,20,24-tetraazahexacosanedioate (0.023 g, 40%) as a yellow oil. Following general procedure B, a sub-sample of the protected product (0.014 g, 0.011 mmol) was reacted with TFA in CH2Cl2 to afford, after chromatography, the tetra-TFA salt 9f (0.008 g, 47%) as a yellow oil. [ α ] D 19.7 = +5 (c 0.1, MeOH); Rf (RP-18, 10% aq HCl:MeOH 1:3) 0.40; IR (ATR) νmax 2930, 1735, 1671, 1499, 1417, 1199, 1175, 1125, 1018, 915, 834, 797, 721 cm−1; 1H NMR (DMSO-d6, 500 MHz) δ 9.32 (4H, br s, NH2-23), 8.62 (4H, br s, NH2-27), 6.14 (2H, dd, J = 17.7, 11.2 Hz, H-19), 5.63 (2H, d, J = 8.3 Hz, H-14), 5.14 (2H, d, J = 17.8 Hz, H2-20a), 5.06 (2H, d, J = 11.2 Hz, H2-20b), 4.03 (2H, d, J = 17.0 Hz, H2-22a), 3.84 (2H, d, J = 17.1 Hz, H2-22b), 3.45 (2H, obscured by H2O, H-11), 2.96 (8H, t, J = 7.2 Hz, H2-24, H2-26), 2.87 (4H, t, J = 7.8 Hz, H2-28), 2.46 (2H, br s, H-4), 2.23–2.01 (8H, m, H2-2, H-10, H2-13a), 1.95 (4H, tt, J = 7.3, 7.2 Hz, H2-25), 1.69–1.51 (10H, m, H2-1b, H-6, H2-8a, H2-29), 1.39–1.34 (4H, m, H2-7b, H2-13b), 1.38 (6H, s, H3-15), 1.31–1.28 (4H, m, H2-1a, H2-7a), 1.28–1.23 (16H, m, H2-30, H2-31, H2-32, H2-33), 1.08 (6H, s, H3-18), 1.03–1.00 (2H, m, H2-8b), 0.83 (6H, d, J = 6.8 Hz, H3-17), 0.65 (6H, d, J = 7.0 Hz, H3-16); 13C NMR (DMSO-d6, 125 MHz) δ 217.0 (C-3), 165.8 (C-21), 140.8 (C-19), 115.5 (C-20), 72.4 (C-11), 71.0 (C-14), 57.0 (C-4), 47.4 (C-22), 46.7 (C-28), 44.9 (C-9), 44.2 (C-12), 44.0 (C-24/C-26), 43.9 (C-24/C-26), 43.2 (C-13), 41.5 (C-5), 36.5 (C-10), 36.2 (C-6), 34.0 (C-2), 30.0 (C-8), 29.0 (C-33), 28.9 (C-32), 28.6 (C-31), 28.5 (C-18), 26.6 (C-7), 25.9 (C-30), 25.4 (C-29), 24.4 (C-1), 22.2 (C-25), 16.2 (C-16), 14.4 (C-15), 11.6 (C-17); (+)-HRESIMS [M + 2H]+ m/z 518.4083 (calculated for C62H108N4O8, 518.4078).

2.3. Antimicrobial Assays

The susceptibility of bacterial strains S. aureus (ATCC 25923), E. coli (ATCC 25922), and P. aeruginosa (ATCC 27853 or PAO1) to antibiotics and compounds was determined using reported protocols [26]. Additional antimicrobial evaluation against MRSA (ATCC 43300), Klebsiella pneumoniae (ATCC 700603), A. baumannii (ATCC 19606), Candida albicans (ATCC 90028), and Cryptococcus neoformans (ATCC 208821) was undertaken at the Community for Open Antimicrobial Drug Discovery at The University of Queensland (Australia) according to their standard protocols as reported previously [26,27]. (See Supplementary File.)

2.4. Determination of the MICs of Antibiotics in the Presence of Synergizing Compounds

Antibiotic restoring enhancer concentrations were determined using reported protocols (see Supplementary File) [26].

2.5. Nitrocefin Assay

Nitrocefin assays were conducted using reported protocols (see Supplementary File) [28].

2.6. Cytotoxicity Assays

Cytotoxicity assays were conducted using reported protocols (see Supplementary File) [26,27].

2.7. Hemolytic Assays

Hemolysis assays were conducted using reported protocols (see Supplementary File) [26,27].

2.8. Real-Time Growth Curves

Real-time growth curves were determined using reported protocols (see Supplementary File) [26].

2.9. ATP Efflux Assay

ATP efflux assays were conducted using reported protocols (see Supplementary File) [28].

3. Results and Discussion

3.1. Synthesis of Tiamulin (2)

Functionalization at C-22 of pleuromutilin (1) has been extensively explored, with derivatives prepared via nucleophilic substitution of sulphonate esters. Tosylation at O-22 was achieved using a modified procedure by Zhang et al. [29], whereby pleuromutilin (1) in CH2Cl2 was reacted with 4-toluenesulfonylchloride and DMAP for 4 h to afford 8 in 57% yield (Scheme 1).
Reagents and conditions: (i) 4-Toluenesulfonylchloride (1.2 equiv), DMAP (3 equiv.), CH2Cl2, 4 h, 70 °C, N2 (57%); (ii) KI (2 equiv.), MeCN, 70 °C, 30 min, N2, then 2-(diethylamino)ethane-1-thiol hydrochloride (1.1 equiv.), and DIPEA (6 equiv.), 70 °C, 2 h, N2 (47%).
Tiamulin (2) was selected to be used as a positive control in biological assays and was prepared by a one pot, two-step sequence, whereby 22-O-tosylate 8 was preincubated with 2.0 equivalents of KI to give the 22-iodo derivative which was then reacted with 1.1 equivalents of 2-(diethylamino)ethane-1-thiol HCl salt in the presence of excess DIPEA to afford (2) in 47% yield (Scheme 1) (Figure S1).

3.2. Synthesis of Pleuromutilin–Polyamine Conjugates

The target set of conjugates required for the synthesis of Boc-protected polyamine scaffolds 7af (Figure 2), which were prepared according to protocols reported in the literature [22,23,24]. The polyamines (PA) chosen covered a range of overall lengths, from spermine (PA-3-4-3) through to the longer chain length PA-3-12-3. The set was chosen to allow the exploration of chain length, lipophilicity, and positioning of positive charges on antimicrobial and cytotoxicity/hemolytic properties.
The target pleuromutilin–polyamine conjugates were prepared using the same nucleophilic displacement of the iodo-activated pleuromutilin methodology used to prepare tiamulin (2). Thus, activation of pleuromutilin 22-O-tosylate (8) with KI, followed by reaction with Boc-protected polyamines 7af in MeCN with DIPEA, afforded Boc-protected intermediates that were then deprotected with 2,2,2-trifluoroacetic acid (TFA) to afford target compounds 9af as their tetra-TFA salts (Scheme 2) (Figures S2–S7).
Regents and conditions: (i) Pleuromutilin O-22-tosylate (8) (2.0 equiv.), KI (2.2 equiv.), MeCN, 70 °C, 1.5 h, N2, then Boc-protected polyamine 7af (1.0 equiv.), and DIPEA (6 equiv.), 70 °C, 2.5 h, N2 (40–72%); (ii) TFA (0.2 mL), CH2Cl2 (2 mL), r.t., 2 h (47–99%).

3.3. Antimicrobial Activities

The antimicrobial activities of pleuromutilin (1), tiamulin (2), and pleuromutilin–polyamine conjugates 9af were determined as the minimum inhibitory concentration (MIC) against a panel of Gram-positive (S. aureus and MRSA) and Gram-negative (E. coli, P. aeruginosa, K. pneumoniae, and A. baumannii) bacterial strains and two fungal strains (C. albicans and C. neoformans) (Table 1). Tiamulin (2) demonstrated potent activity against both S. aureus and MRSA with an MIC of 3.125 and ≤0.51 μM, respectively, while exhibiting no activity against any of the Gram-negative bacteria or fungi. Intriguingly, all of the conjugates exhibited moderate to good growth inhibition of S. aureus and the Gram-negative bacterium, E. coli, with the conjugates 9bf also demonstrating strong growth inhibition of MRSA. Antifungal activity was observed for all conjugates against C. neoformans, with MIC’s ranging from ≤0.17 to 0.72 μM, while against C. albicans, activity was only observed for the longer chained PA-3-10-3 9e (MIC 10.9 μM) and PA-3-12-3 9f (MIC 21.5 μM) conjugates. Meanwhile, the PA-3-12-3 conjugate 9f was the most active, exhibiting broad spectrum activity against all tested strains, notably against S. aureus, MRSA, E. coli, A. baumannii, and C. neoformans.

3.4. Cytotoxic and Hemolytic Activities

Tiamulin (2) and pleuromutilin–polyamine conjugates 9af were evaluated for cytotoxicity towards HEK293 cells (human kidney epithelial cell line), reported as the concentration of compound at 50% cytotoxicity (IC50), and for hemolytic activity against human red blood cells, reported as the concentration of compound at 10% hemolytic activity (HC10) (Table 2). The only conjugate to exhibit either of these properties was the longest PA-3-12-3 variant (9f), which was both cytotoxic (IC50 8.3 µM) and hemolytic (HC10 13.2 µM).
When taken together, the combination of intrinsic antimicrobial activities and cytotoxicity/hemolytic activities identified the PA-3-10-3 conjugate 9e as being of particular interest. Of note, in addition to the strong activity towards Gram-positive bacteria, the conjugate also exhibited activity towards the Gram-negative bacterium E. coli.

3.5. Real-Time Growth Inhibition Assay

To investigate the kinetics of the antibacterial activity exhibited by 9e and tiamulin (2), real-time growth inhibition curves were determined against S. aureus ATCC 25922 and P. aeruginosa PAO1 via measurement of optical density at 490 nm during an 18 h culturing period. Although appearing to be qualitatively different, both compounds inhibited the growth of S. aureus (Figure 3), with the 18 h time point values in close agreement with the MIC values obtained for the two compounds (Table 1) using classical microdilution techniques. Further investigation is required to determine the factors leading to the differences observed in the sub-MIC growth curves for the two compounds. No such inhibition was observed against P. aeruginosa (Figure S8).

3.6. Antibiotic Enhancement Activities

Tiamulin (2) and polyamine conjugates 9af were also evaluated for their ability to enhance the antibiotic action of doxycycline towards P. aeruginosa ATCC 27853 and of erythromycin towards E. coli ATCC 25922. For the doxycycline assay, the antibiotic was present at a concentration of 4.5 μM (2 μg/mL), 20 times below the MIC value of 90 μM (40 μg/mL). Amongst the test compounds, only modest levels of enhancement were detected, with 4-fold increases in activity observed for 9a, 9b, and 9f, while the remaining compounds, including tiamulin (2), were essentially unable to enhance the antibiotic action of doxycycline (Table 3). For the erythromycin assay, the antibiotic was present at a concentration of 2.7 μM (2 μg/mL), well below the MIC value of 174 μM (128 μg/mL). Insignificant levels of enhancement of the action of the lipophilic antibiotic erythromycin was detected towards E. coli, though of note is the observance of improved activity for three of the conjugates (9a, 9c, and 9d) in comparison to tiamulin (2).

3.7. Membrane Perturbation Activities

Perturbation or disruption of the bacterial membrane is a validated mechanism of action of particular classes of antibiotics, including the polymyxin family of lipopeptides. In addition to being lethal to bacteria, this mechanism of membrane perturbation can also be used to enhance the action of other antibiotics, whereby a sub-MIC dose of the disruptor can facilitate the entry of the antibiotics into the microorganism [30]. Membrane perturbation appears to be one mechanism of both antibiotic activity and antibiotic enhancement activity exhibited by α,ω-disubstituted polyamines, being well documented in a number of studies [21].
The mechanism of antibiotic action of 9e towards the Gram-positive bacterium Staphylococcus aureus was attributed to its ability to disrupt the bacterial membrane and cause the release of intracellular ATP. The test organisms, being S. aureus ATCC 25923 and MRSA, were briefly exposed to 9e at a single dose of 100 µg/mL, with the leakage of ATP determined by the use of a bioluminescence assay (Figure 4). While considered active in the assay, the level of disruption induced by exposure to 9e was significantly less than that induced by the positive control squalamine. Nevertheless, a 40% level of ATP release is detrimental to the survival of the bacteria. After 3 min, it will be clear that the bacteria will be killed.
We also examined the ability of 9e to act as membrane disruptor of the Gram-negative bacteria P. aeruginosa PAO1 (Figure 5), using a nitrocefin colorimetric assay. This assay makes use of a chromogenic cephalosporin derivative, which in the presence of an outer membrane disruptor gains entry to the periplasm, where upon the action of β-lactamases leads to substrate hydrolysis with a detectable color change from yellow to red. While the positive control, polymyxin B, demonstrated potent ability to perturb the outer membrane of P. aeruginosa PAO1, polyamine conjugate 9e was inactive at all test doses. Tiamulin (2) was also evaluated in the same assay, also failing to exhibit any detectable membrane perturbation.

4. Conclusions

Six pleuromutilin–polyamine conjugates 9af were successfully synthesized from pleuromutilin-22-OTs (8) and Boc-protected polyamines 7af, in addition to the veterinary drug tiamulin (2). These compounds primarily inhibited the growth of Gram-positive bacteria, with limited effects on Gram-negative bacteria and two fungal strains. Notably, all polyamine conjugates exhibited anti-E. coli and antifungal activity against C. neoformans, unlike pleuromutilin (1) or tiamulin (2). Cytotoxicity and hemolysis assays showed that all compounds, except the longest polyamine conjugate, 9f, were non-toxic. Moreover, when combined with doxycycline against P. aeruginosa, three conjugates (9a, 9b, and 9f) exhibited better antibiotic potentiation than tiamulin (2). Preliminary investigations suggest that while 9e exhibits intrinsic Gram-positive antibacterial by a mechanism related to membrane perturbation, the ability of the compound class to enhance the action of doxycycline towards Gram-negative bacteria does not appear to be linked to outer membrane disruption. Further studies will be required to determine their precise mechanism.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms11112791/s1, Figure S1: 1H (CDCl3, 400 MHz) and 13C (CDCl3, 100 MHz) NMR spectra for 2; Figure S2: 1H (DMSO-d6, 400 MHz) and 13C (DMSO-d6, 100 MHz) NMR spectra for 9a; Figure S3: 1H (DMSO-d6, 400 MHz) and 13C (DMSO-d6, 100 MHz) NMR spectra for 9b; Figure S4: 1H (DMSO-d6, 400 MHz) and 13C (DMSO-d6, 100 MHz) NMR spectra for 9c; Figure S5: 1H (DMSO-d6, 400 MHz) and 13C (DMSO-d6, 100 MHz) NMR spectra for 9d; Figure S6: 1H (DMSO-d6, 400 MHz) and 13C (DMSO-d6, 100 MHz) NMR spectra for 9e; Figure S7: 1H (DMSO-d6, 500 MHz) and 13C (DMSO-d6, 125 MHz) NMR spectra for 9f; Figure S8: Bacterial growth inhibition exhibited by 2 (left) and 9e (right) against P. aeruginosa PAO1 at different concentrations. Positive control was bacteria only and negative control was media only. The Supplementary File contains full protocols for all bioassays conducted.

Author Contributions

Conceptualization, B.R.C.; methodology, K.S., K.H. and F.R.; formal analysis, B.R.C. and J.M.B.; investigation, K.S., M.M.C., K.H., F.R., M.-L.B.-K., J.M.B. and B.R.C.; resources, B.R.C. and J.M.B.; data curation, B.R.C.; writing—original draft preparation, B.R.C. and M.M.C.; writing—review and editing, B.R.C., M.M.C., M.-L.B.-K. and J.M.B.; supervision, B.R.C., M.M.C. and J.M.B.; project administration, B.R.C. and M.M.C.; funding acquisition, B.R.C., M.M.C., M.-L.B.-K. and J.M.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Catalyst: Seeding Dumont d’Urville NZ-France Science and Technology Support Program (19-UOA-057-DDU), provided by the New Zealand Ministry of Business, Innovation, and Employment and administered by the Royal Society Te Apārangi.

Data Availability Statement

Data are contained within the article or Supplementary Materials.

Acknowledgments

We thank Michael Schmitz and Mansa Nair for their assistance with NMR and mass spectrometric data. Some of the antimicrobial screening was performed by CO-ADD (The Community for Antimicrobial Drug Discovery), funded by the Wellcome Trust (UK) and The University of Queensland (Australia).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mulani, M.S.; Kamble, E.E.; Kumkar, S.N.; Tawre, M.S.; Pardesi, K.R. Emerging Strategies to Combat ESKAPE Pathogens in the Era of Antimicrobial Resistance: A Review. Front. Microbiol. 2019, 10, 539. [Google Scholar] [CrossRef] [PubMed]
  2. Munita, J.M.; Arias, C.A. Mechanisms of Antibiotic Resistance. Microbiol. Spectr. 2016, 4, VMBF-0016-5015. [Google Scholar] [CrossRef] [PubMed]
  3. Tacconelli, E.; Carrara, E.; Savoldi, A.; Harbarth, S.; Mendelson, M.; Monnet, D.L.; Pulcini, C.; Kahlmeter, G.; Kluytmans, J.; Carmeli, Y.; et al. Discovery, Research, and Development of New Antibiotics: The WHO Priority List of Antibiotic-Resistant Bacteria and Tuberculosis. Lancet Infect. Dis. 2018, 18, 318–327. [Google Scholar] [CrossRef] [PubMed]
  4. Bartlett, J.G.; Gilbert, D.N.; Spellberg, B. Seven Ways to Preserve the Miracle of Antibiotics. Clin. Infect. Dis. 2013, 56, 1445–1450. [Google Scholar] [CrossRef]
  5. Piddock, L.J. The Crisis of No New Antibiotics—What Is the Way Forward? Lancet Infect. Dis. 2012, 12, 249–253. [Google Scholar] [CrossRef] [PubMed]
  6. Wright, G.D. Solving the Antibiotic Crisis. ACS Infect. Dis. 2015, 1, 80–84. [Google Scholar] [CrossRef]
  7. Kavanagh, F.; Hervey, A.; Robbins, W.J. Antibiotic Substances from Basidiomycetes: VIII. Pleurotus Multilus (Fr.) Sacc. and Pleurotus Passeckerianus Pilat. Proc. Natl. Acad. Sci. USA 1951, 37, 570–574. [Google Scholar] [CrossRef]
  8. Tang, Y.-Z.; Liu, Y.-H.; Chen, J.-X. Pleuromutilin and Its Derivatives-The Lead Compounds for Novel Antibiotics. Mini-Rev. Med. Chem. 2012, 12, 53–61. [Google Scholar] [CrossRef]
  9. Nigam, P.S.; Singh, A. METABOLIC PATHWAYS|Production of Secondary Metabolites—Fungi. In Encyclopedia of Food Microbiology; Elsevier: Amsterdam, The Netherlands, 2014; pp. 570–578. ISBN 978-0-12-384733-1. [Google Scholar]
  10. Novak, R. Are Pleuromutilin Antibiotics Finally Fit for Human Use? Ann. N. Y. Acad. Sci. 2011, 1241, 71–81. [Google Scholar] [CrossRef]
  11. Lolk, L.; Pøhlsgaard, J.; Jepsen, A.S.; Hansen, L.H.; Nielsen, H.; Steffansen, S.I.; Sparving, L.; Nielsen, A.B.; Vester, B.; Nielsen, P. A Click Chemistry Approach to Pleuromutilin Conjugates with Nucleosides or Acyclic Nucleoside Derivatives and Their Binding to the Bacterial Ribosome. J. Med. Chem. 2008, 51, 4957–4967. [Google Scholar] [CrossRef]
  12. Sevrioukova, I.F. Structural Insights into the Interaction of Cytochrome P450 3A4 with Suicide Substrates: Mibefradil, Azamulin and 6′,7′-Dihydroxybergamottin. Int. J. Mol. Sci. 2019, 20, 4245. [Google Scholar] [CrossRef] [PubMed]
  13. Jones, R.N.; Fritsche, T.R.; Sader, H.S.; Ross, J.E. Activity of Retapamulin (SB-275833), a Novel Pleuromutilin, against Selected Resistant Gram-Positive Cocci. Antimicrob. Agents Chemother. 2006, 50, 2583–2586. [Google Scholar] [CrossRef] [PubMed]
  14. File, T.M.; Alexander, E.; Goldberg, L.; Das, A.F.; Sandrock, C.; Paukner, S.; Moran, G.J. Lefamulin Efficacy and Safety in a Pooled Phase 3 Clinical Trial Population with Community-Acquired Bacterial Pneumonia and Common Clinical Comorbidities. BMC Pulm. Med. 2021, 21, 154. [Google Scholar] [CrossRef]
  15. Watkins, R.R.; File, T.M. Lefamulin: A Novel Semisynthetic Pleuromutilin Antibiotic for Community-Acquired Bacterial Pneumonia. Clin. Infect. Dis. 2020, 71, 2757–2762. [Google Scholar] [CrossRef] [PubMed]
  16. Paukner, S.; Riedl, R. Pleuromutilins: Potent Drugs for Resistant Bugs—Mode of Action and Resistance. Cold Spring Harb. Perspect. Med. 2017, 7, a027110. [Google Scholar] [CrossRef]
  17. Paukner, S.; Sader, H.S.; Ivezic-Schoenfeld, Z.; Jones, R.N. Antimicrobial Activity of the Pleuromutilin Antibiotic BC-3781 against Bacterial Pathogens Isolated in the SENTRY Antimicrobial Surveillance Program in 2010. Antimicrob. Agents Chemother. 2013, 57, 4489–4495. [Google Scholar] [CrossRef]
  18. Veve, M.P.; Wagner, J.L. Lefamulin: Review of a Promising Novel Pleuromutilin Antibiotic. Pharmacotherapy 2018, 38, 935–946. [Google Scholar] [CrossRef]
  19. Nikaido, H. Multidrug Efflux Pumps of Gram-Negative Bacteria. J. Bacteriol. 1996, 178, 5853–5859. [Google Scholar] [CrossRef]
  20. Keeney, D.; Ruzin, A.; McAleese, F.; Murphy, E.; Bradford, P.A. MarA-Mediated Overexpression of the AcrAB Efflux Pump Results in Decreased Susceptibility to Tigecycline in Escherichia coli. J. Antimicrob. Chemother. 2008, 61, 46–53. [Google Scholar] [CrossRef]
  21. Cadelis, M.M.; Li, S.A.; Bourguet-Kondracki, M.-L.; Blanchet, M.; Douafer, H.; Brunel, J.M.; Copp, B.R. Spermine Derivatives of Indole-3-Carboxylic Acid, Indole-3-Acetic Acid and Indole-3-Acrylic Acid as Gram-Negative Antibiotic Adjuvants. ChemMedChem 2021, 16, 513–523. [Google Scholar] [CrossRef]
  22. Pearce, A.N.; Kaiser, M.; Copp, B.R. Synthesis and Antimalarial Evaluation of Artesunate-Polyamine and Trioxolane-Polyamine Conjugates. Eur. J. Med. Chem. 2017, 140, 595–603. [Google Scholar] [CrossRef] [PubMed]
  23. Klenke, B.; Gilbert, I.H. Nitrile Reduction in the Presence of Boc-Protected Amino Groups by Catalytic Hydrogenation over Palladium-Activated Raney-Nickel. J. Org. Chem. 2001, 66, 2480–2483. [Google Scholar] [CrossRef] [PubMed]
  24. Klenke, B.; Stewart, M.; Barrett, M.P.; Brun, R.; Gilbert, I.H. Synthesis and Biological Evaluation of S-Triazine Substituted Polyamines as Potential New Anti-Trypanosomal Drugs. J. Med. Chem. 2001, 44, 3440–3452. [Google Scholar] [CrossRef]
  25. Lemieux, M.R.; Siricilla, S.; Mitachi, K.; Eslamimehr, S.; Wang, Y.; Yang, D.; Pressly, J.D.; Kong, Y.; Park, F.; Franzblau, S.G.; et al. An Antimycobacterial Pleuromutilin Analogue Effective against Dormant Bacilli. Bioorg. Med. Chem. 2018, 26, 4787–4796. [Google Scholar] [CrossRef]
  26. Chen, D.; Cadelis, M.M.; Rouvier, F.; Troia, T.; Edmeades, L.R.; Fraser, K.; Gill, E.S.; Bourguet-Kondracki, M.-L.; Brunel, J.M.; Copp, B.R. α,ω-Diacyl-Substituted Analogues of Natural and Unnatural Polyamines: Identification of Potent Bactericides That Selectively Target Bacterial Membranes. Int. J. Mol. Sci. 2023, 24, 5882. [Google Scholar] [CrossRef] [PubMed]
  27. Blaskovich, M.A.T.; Zuegg, J.; Elliott, A.G.; Cooper, M.A. Helping Chemists Discover New Antibiotics. ACS Infect. Dis. 2015, 1, 285–287. [Google Scholar] [CrossRef]
  28. Troudi, A.; Bolla, J.M.; Klibi, N.; Brunel, J.M. An Original and Efficient Antibiotic Adjuvant Strategy to Enhance the Activity of Macrolide Antibiotics against Gram-Negative Resistant Strains. Int. J. Mol. Sci. 2022, 23, 12457. [Google Scholar] [CrossRef]
  29. Zhang, Z.-S.; Huang, Y.-Z.; Luo, J.; Jin, Z.; Liu, Y.-H.; Tang, Y.-Z. Synthesis and Antibacterial Activities of Novel Pleuromutilin Derivatives Bearing an Aminothiophenol Moiety. Chem. Biol. Drug Des. 2018, 92, 1627–1637. [Google Scholar] [CrossRef]
  30. French, S.; Farha, M.; Ellis, M.J.; Sameer, Z.; Côté, J.-P.; Cotroneo, N.; Lister, T.; Rubio, A.; Brown, E.D. Potentiation of Antibiotics against Gram-Negative Bacteria by Polymyxin B Analogue SPR741 from Unique Perturbation of the Outer Membrane. ACS Infect. Dis. 2020, 6, 1405–1412. [Google Scholar] [CrossRef]
Figure 1. Structures of pleuromutilin (1) and its derivatives (26).
Figure 1. Structures of pleuromutilin (1) and its derivatives (26).
Microorganisms 11 02791 g001
Scheme 1. Synthesis of pleuromutilin O-22-tosylate (8) and tiamulin (2).
Scheme 1. Synthesis of pleuromutilin O-22-tosylate (8) and tiamulin (2).
Microorganisms 11 02791 sch001
Figure 2. Boc-protected polyamines 7af.
Figure 2. Boc-protected polyamines 7af.
Microorganisms 11 02791 g002
Scheme 2. General method for the synthesis of target pleuromutilin–polyamine conjugates (9af).
Scheme 2. General method for the synthesis of target pleuromutilin–polyamine conjugates (9af).
Microorganisms 11 02791 sch002
Figure 3. Bacterial growth inhibition exhibited by 2 (left) and 9e (right) against S. aureus ATCC 25923 at different concentrations. Positive control was bacteria only and negative control was media only.
Figure 3. Bacterial growth inhibition exhibited by 2 (left) and 9e (right) against S. aureus ATCC 25923 at different concentrations. Positive control was bacteria only and negative control was media only.
Microorganisms 11 02791 g003
Figure 4. ATP release in S. aureus ATCC 25923 (left) and MRSA (right) exhibited by polyamine conjugate 9e as determined using ATP efflux assay. Squalamine (100 µg/mL) was the positive control and water was the negative control. Compounds were tested at a final concentration of 100 µg/mL, and results are reported as percentage (%) relative to positive control.
Figure 4. ATP release in S. aureus ATCC 25923 (left) and MRSA (right) exhibited by polyamine conjugate 9e as determined using ATP efflux assay. Squalamine (100 µg/mL) was the positive control and water was the negative control. Compounds were tested at a final concentration of 100 µg/mL, and results are reported as percentage (%) relative to positive control.
Microorganisms 11 02791 g004
Figure 5. The ability of tiamulin (2) (left) and polyamine conjugate 9e (right) to act as membrane disruptors in P. aeruginosa PAO1, as determined using a nitrocefin hydrolysis assay. Polymixin B (PMB) was the positive control (98.3 μM, 128 μg/mL), and the negative control was bacteria with nitrocefin.
Figure 5. The ability of tiamulin (2) (left) and polyamine conjugate 9e (right) to act as membrane disruptors in P. aeruginosa PAO1, as determined using a nitrocefin hydrolysis assay. Polymixin B (PMB) was the positive control (98.3 μM, 128 μg/mL), and the negative control was bacteria with nitrocefin.
Microorganisms 11 02791 g005
Table 1. Antimicrobial (MIC, μM) activities of pleuromutilin (1), tiamulin (2), and conjugates 9af.
Table 1. Antimicrobial (MIC, μM) activities of pleuromutilin (1), tiamulin (2), and conjugates 9af.
CompoundS.a. aMRSA bE.c. cP.a. dK.p. eA.b. fC.a. gC.n. h
13.125n.t. i400800n.t.n.t.n.t.n.t.
23.125≤0.51200800>64.8>64.8>64.8>64.8
9a18.123.218.172.5>23.2>23.2>23.20.72
9b17.8≤0.188.88142>22.7>22.7>22.7≤0.18
9c17.6≤0.1835.2>141>22.5>22.5>22.5≤0.18
9d34.85.5770>139>22.3>22.3>22.3≤0.17
9e4.27≤0.174.27>88>21.9>21.910.9≤0.17
9f2.10≤0.174.1933.521.52.721.5≤0.17
a S. aureus ATCC 25923, streptomycin (MIC 21.5 μM) and chloramphenicol (MIC 1.5–3 μM) used as positive controls and values presented as the mean (n = 3); b MRSA ATCC 43300, vancomycin (MIC 0.7 μM) used as a positive control and values presented as the mean (n = 2); c E. coli ATCC 25922, streptomycin (MIC 21.5 μM) and colistin (MIC 2 μM) used as positive controls and values presented as the mean (n = 3); d P. aeruginosa PAO1, streptomycin (MIC 21.5 μM) and colistin (MIC 1 μM) used as positive controls and values presented as the mean (n = 3); e K. pneumoniae ATCC 700603, colistin (MIC 0.2 µM) used as a positive control and values presented as the mean (n = 2); f A. baumannii ATCC 19606, colistin (MIC 0.2 µM) used as a positive control and values presented as the mean (n = 2); g C. albicans ATCC 90028, fluconazole (MIC 0.4 μM) used as a positive control and values presented as the mean (n = 2); h C. neoformans ATCC 208821, fluconazole (MIC 26 µM) used as a positive control and values presented as the mean (n = 2); i Not tested.
Table 2. Cytotoxicity (IC50, μM) and hemolytic (HC10, μM) activities of tiamulin (2) and conjugates 9af.
Table 2. Cytotoxicity (IC50, μM) and hemolytic (HC10, μM) activities of tiamulin (2) and conjugates 9af.
CompoundCytotoxicity aHemolysis b
2>64.8>64.8
9a>23.2>23.2
9b>22.7>22.7
9c>22.5>22.5
9d>22.3>22.3
9e>21.9>21.9
9f8.313.2
a Concentration of compound at 50% cytotoxicity on HEK293 human embryonic kidney cells with tamoxifen as the positive control (IC50 24 µM) and values presented as the mean (n = 2); b Concentration of compound at 10% hemolytic activity on human red blood cells with melittin as the positive control (HC10 0.95 µM) and values presented as the mean (n = 2).
Table 3. Antibiotic enhancement activity (MIC, µM) of tiamulin (2) and conjugates 9af.
Table 3. Antibiotic enhancement activity (MIC, µM) of tiamulin (2) and conjugates 9af.
CompoundDox/P.a. aErythro/E.c. b
2>405 (2)>405 (0.5)
9a18.1 (4)9.06 (2)
9b35.5 (4)8.88 (1)
9c141 (1)17.6 (2)
9d139 (1)34.8 (2)
9e34.2 (2)8.54 (0.5)
9f8.38 (4)8.38 (0.5)
a Concentration (µM) required to restore doxycycline activity at 4.5 µM against P. aeruginosa ATCC 27853. Fold change shown in parentheses is the ratio between the intrinsic MIC of the test compound and the combination MIC; b Concentration (µM) required to restore erythromycin activity at 2.7 µM against E. coli ATCC 25922. Fold change shown in parentheses is the ratio between the intrinsic MIC of the test compound and the combination MIC.
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

Sue, K.; Cadelis, M.M.; Hainsworth, K.; Rouvier, F.; Bourguet-Kondracki, M.-L.; Brunel, J.M.; Copp, B.R. Preliminary SAR of Novel Pleuromutilin–Polyamine Conjugates. Microorganisms 2023, 11, 2791. https://doi.org/10.3390/microorganisms11112791

AMA Style

Sue K, Cadelis MM, Hainsworth K, Rouvier F, Bourguet-Kondracki M-L, Brunel JM, Copp BR. Preliminary SAR of Novel Pleuromutilin–Polyamine Conjugates. Microorganisms. 2023; 11(11):2791. https://doi.org/10.3390/microorganisms11112791

Chicago/Turabian Style

Sue, Kenneth, Melissa M. Cadelis, Kerrin Hainsworth, Florent Rouvier, Marie-Lise Bourguet-Kondracki, Jean Michel Brunel, and Brent R. Copp. 2023. "Preliminary SAR of Novel Pleuromutilin–Polyamine Conjugates" Microorganisms 11, no. 11: 2791. https://doi.org/10.3390/microorganisms11112791

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