1
|
He X, Karlsson PA, Xiong R, Moodie LWK, Wang H, Bergström CAS, Hubert M. Liquid crystal nanoparticles for oral combination antibiotic therapies: A strategy towards protecting commensal gut bacteria during treatment. J Colloid Interface Sci 2025; 678:287-300. [PMID: 39245019 DOI: 10.1016/j.jcis.2024.08.230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
Antibiotics are essential for treating infections and reducing risks during medical interventions. However, many commonly used antibiotics lack the physiochemical properties for an efficient oral administration when treating systemic infection. Instead, we are reliant on intravenous delivery, which presents complications outside of clinical settings. Developing novel formulations for oral administration is a potential solution to this problem. We engineered hexosome and cubosome liquid crystal nanoparticles (LCNPs) characterized by small-angle X-ray scattering and cryogenic transmission electron microscopy, and could encapsulate the antibiotics vancomycin (VAN) and clarithromycin (CLA) with high loading efficiencies. By rationally choosing stable lipid building blocks, the loaded LCNPs demonstrated excellent resilience against enzymatic degradation in an in vitro gut model LCNP stability is crucial as premature antibiotic leakage can negatively impact the gut microbiota. In screens against the representative gut bacteria Enterococcus faecalis and Escherichia coli, our LCNPs provided a protective effect. Furthermore, we explored co-administration and dual loading strategies of VAN and CLA, and demonstrated effective loading, stability and protection for E. faecalis and E. coli. This work represents a proof of concept for the early-stage development of antibiotic-loaded LCNPs to treat systemic infection via oral administration, opening opportunities for combination antibiotic therapies.
Collapse
Affiliation(s)
- Xiguo He
- Department of Pharmacy, Uppsala University, Uppsala, Sweden; The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Uppsala, Sweden; Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden
| | - Philip A Karlsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Ruisheng Xiong
- Department of Chemistry, Uppsala University, Uppsala, Sweden
| | - Lindon W K Moodie
- Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden; Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Helen Wang
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Christel A S Bergström
- Department of Pharmacy, Uppsala University, Uppsala, Sweden; The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Uppsala, Sweden; Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden
| | - Madlen Hubert
- Department of Pharmacy, Uppsala University, Uppsala, Sweden; Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
2
|
Xu T, Yan X, Kang A, Yang L, Li X, Tian Y, Yang R, Qin S, Guo Y. Development of Membrane-Targeting Fluorescent 2-Phenyl-1 H-phenanthro[9,10- d]imidazole-Antimicrobial Peptide Mimic Conjugates against Methicillin-Resistant Staphylococcus aureus. J Med Chem 2024; 67:9302-9317. [PMID: 38491982 DOI: 10.1021/acs.jmedchem.4c00436] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2024]
Abstract
The escalation of multidrug-resistant bacterial infections, especially infections caused by methicillin-resistant Staphylococcus aureus (MRSA), underscores the urgent need for novel antimicrobial drugs. Here, we synthesized a series of amphiphilic 2-phenyl-1H-phenanthro[9,10-d]imidazole-antimicrobial peptide (AMP) mimic conjugates (III1-30). Among them, compound III13 exhibited excellent antibacterial activity against G+ bacteria and clinical MRSA isolates (MIC = 0.5-2 μg/mL), high membrane selectivity, and low toxicity. Additionally, compared with traditional clinical antibiotics, III13 demonstrated rapid bactericidal efficacy and was less susceptible to causing bacterial resistance. Mechanistic studies revealed that III13 targets phosphatidylglycerol (PG) on bacterial membranes to disrupt membrane integrity, leading to an increase in intracellular ROS and leakage of proteins and DNA, ultimately causing bacterial cell death. Furthermore, III13 possessed good fluorescence properties with potential for further dynamic monitoring of the antimicrobial process. Notably, III13 showed better in vivo efficacy against MRSA compared to vancomycin, suggesting its potential as a promising candidate for anti-MRSA medication.
Collapse
Affiliation(s)
- Ting Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
| | - Xiaoting Yan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Ayue Kang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Longhua Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Xinhui Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Yue Tian
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Ruige Yang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Yong Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| |
Collapse
|
3
|
Dey R, Mukherjee S, Mukherjee R, Haldar J. Small molecular adjuvants repurpose antibiotics towards Gram-negative bacterial infections and multispecies bacterial biofilms. Chem Sci 2023; 15:259-270. [PMID: 38143555 PMCID: PMC10739173 DOI: 10.1039/d3sc05124b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/12/2023] [Indexed: 12/26/2023] Open
Abstract
Gram-negative bacterial infections pose a significant challenge due to two major resistance elements, including the impermeability of the outer membrane and the overexpression of efflux pumps, which contribute to antibiotic resistance. Additionally, the coexistence of multispecies superbugs in mixed species biofilms further complicates treatment, as these infections are refractory to most antibiotics. To address this issue, combining obsolete antibiotics with non-antibiotic adjuvants that target bacterial membranes has shown promise in combating antibacterial resistance. However, the clinical translation of this cocktail therapy has been hindered by the toxicity associated with these membrane active adjuvants, mainly due to a limited understanding of their structure and mechanism of action. Towards this goal, herein, we have designed a small molecular adjuvant by tuning different structural parameters, such as the balance between hydrophilic and hydrophobic groups, spatial positioning of hydrophobicity and hydrogen bonding interactions, causing moderate membrane perturbation in bacterial cells without any toxicity to mammalian cells. Moderate membrane perturbation not only enhances the internalization of antibiotics, but also increases the intracellular concentration of drugs by hampering the efflux machinery. This revitalises the efficacy of various classes of antibiotics by 32-512 fold, without inducing toxicity. The leading combination not only exhibits potent bactericidal activity against A. baumannii biofilms but also effectively disrupts mature multispecies biofilms composed of A. baumannii and methicillin-resistant Staphylococcus aureus (MRSA), which is typically resistant to most antibiotics. Importantly, the combination therapy demonstrates good biocompatibility and excellent in vivo antibacterial efficacy (>99% reduction) in a skin infection model of A. baumannii. Interestingly, A. baumannii shows reduced susceptibility to develop resistance against the leading combination, underscoring its potential for treating multi-drug resistant infections.
Collapse
Affiliation(s)
- Rajib Dey
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur Bengaluru 560064 Karnataka India
| | - Sudip Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur Bengaluru 560064 Karnataka India
| | - Riya Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur Bengaluru 560064 Karnataka India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur Bengaluru 560064 Karnataka India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur Bengaluru 560064 Karnataka India
| |
Collapse
|
4
|
Update on the Discovery of Efflux Pump Inhibitors against Critical Priority Gram-Negative Bacteria. Antibiotics (Basel) 2023; 12:antibiotics12010180. [PMID: 36671381 PMCID: PMC9854755 DOI: 10.3390/antibiotics12010180] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Antimicrobial resistance (AMR) has become a major problem in public health leading to an estimated 4.95 million deaths in 2019. The selective pressure caused by the massive and repeated use of antibiotics has led to bacterial strains that are partially or even entirely resistant to known antibiotics. AMR is caused by several mechanisms, among which the (over)expression of multidrug efflux pumps plays a central role. Multidrug efflux pumps are transmembrane transporters, naturally expressed by Gram-negative bacteria, able to extrude and confer resistance to several classes of antibiotics. Targeting them would be an effective way to revive various options for treatment. Many efflux pump inhibitors (EPIs) have been described in the literature; however, none of them have entered clinical trials to date. This review presents eight families of EPIs active against Escherichia coli or Pseudomonas aeruginosa. Structure-activity relationships, chemical synthesis, in vitro and in vivo activities, and pharmacological properties are reported. Their binding sites and their mechanisms of action are also analyzed comparatively.
Collapse
|
5
|
Melander RJ, Mattingly AE, Nemeth AM, Melander C. Overcoming intrinsic resistance in gram-negative bacteria using small molecule adjuvants. Bioorg Med Chem Lett 2023; 80:129113. [PMID: 36566797 PMCID: PMC9885958 DOI: 10.1016/j.bmcl.2022.129113] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Gram-negative bacteria are intrinsically resistant to many classes of antibiotics, predominantly due to the impermeability of the outer membrane and the presence of efflux pumps. Small molecule adjuvants that circumvent these resistance mechanisms have the potential to expand therapeutic options for treating Gram-negative infections to encompass antibiotic classes that are otherwise limited to treating Gram-positive infections. Adjuvants that effect increased antibiotic permeation, either by physical disruption of the outer membrane or through interference with synthesis, transport, or assembly of membrane components, and adjuvants that limit efflux, are discussed as potential avenues to overcoming intrinsic resistance in Gram-negative bacteria.
Collapse
Affiliation(s)
- Roberta J Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Anne E Mattingly
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Ansley M Nemeth
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Christian Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States.
| |
Collapse
|
6
|
Kumar D, Singhal C, Yadav M, Joshi P, Patra P, Tanwar S, Das A, Kumar Pramanik S, Chaudhuri S. Colistin potentiation in multidrug-resistant Acinetobacter baumannii by a non-cytotoxic guanidine derivative of silver. Front Microbiol 2023; 13:1006604. [PMID: 36687622 PMCID: PMC9846554 DOI: 10.3389/fmicb.2022.1006604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/23/2022] [Indexed: 01/06/2023] Open
Abstract
A novel nano-formulation (NF) that sensitizes Acinetobacter baumannii (AB) to otherwise ineffective colistin is described in the present study. Infections due to multidrug resistant (MDR) AB represent a major therapeutic challenge, especially in situations of pre-existing colistin resistance (colR). Subsequently, boosting the effectiveness of colistin would be a better alternative tactic to treat AB infections rather than discovering a new class of antibiotics. We have previously demonstrated an NF comprising self-assembled guanidinium and ionic silver nanoparticles [AD-L@Ag(0)] to have anti-biofilm and bactericidal activity. We report NF AD-L@Ag(0) for the very first time for the potentiation of colistin in Gram-negative colistin-resistant bacteria. Our results implied that a combination of clinically relevant concentrations of colistin and AD-L@Ag(0) significantly decreased colistin-resistant AB bacterial growth and viability, which otherwise was elevated in the presence of only colistin. In this study, we have described various combinations of minimum inhibitory concentration (MIC) of colistin (MICcol, 1/2 MICcol, and 1/4 MICcol) and that of AD-L@Ag(0) [MICAD-L@Ag(0), 1/2 MICAD-L@Ag(0), and 1/4 MICAD-L@Ag(0)] and tested them against MDR AB culture. The results (in broth as well as in solid media) signified that AD-L@Ag(0) was able to potentiate the anti-microbial activity of colistin at sub-MIC concentrations. Furthermore, the viability and metabolic activity of bacterial cells were also measured by CTC fluorescence assay and ATP bioluminescence assay. The results of these assays were in perfect concordance with the scores of cultures (colony forming unit and culture turbidity). In addition, quantitative real-time PCR (qRT-PCR) was performed to unveil the expression of selected genes, DNAgyrA, DNAgyrB, and dac. These genes introduce negative supercoiling in the DNA, and hence are important for basic cellular processes. These genes, due to mutation, modified the Lipid A of bacteria, further resisting the uptake of colistin. Therefore, the expression of these genes was upregulated when AB was treated with only colistin, substantiating that AB is resistant to colistin, whereas the combinations of MICcol + MICAD-L@Ag(0) downregulated the expression of these genes, implying that the developed formulation can potentiate the efficiency of colistin. In conclusion, AD-L@Ag(0) can potentiate the proficiency of colistin, further enhancing colistin-mediated death of AB by putatively disrupting the outer membrane (OM) and facilitating bacterial death.
Collapse
Affiliation(s)
- Deepak Kumar
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Chaitali Singhal
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Manisha Yadav
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Pooja Joshi
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Priyanka Patra
- CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, India
| | - Subhash Tanwar
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Amitava Das
- Indian Institute of Science Education and Research Kolkata, Mohanpur, India,*Correspondence: Amitava Dasc,
| | - Sumit Kumar Pramanik
- CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, India,Sumit Kumar Pramanikb,
| | - Susmita Chaudhuri
- Translational Health Science and Technology Institute (THSTI), Faridabad, India,Susmita Chaudhuria,
| |
Collapse
|
7
|
Wesseling CJ, Martin NI. Synergy by Perturbing the Gram-Negative Outer Membrane: Opening the Door for Gram-Positive Specific Antibiotics. ACS Infect Dis 2022; 8:1731-1757. [PMID: 35946799 PMCID: PMC9469101 DOI: 10.1021/acsinfecdis.2c00193] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
New approaches to target antibacterial agents toward Gram-negative bacteria are key, given the rise of antibiotic resistance. Since the discovery of polymyxin B nonapeptide as a potent Gram-negative outer membrane (OM)-permeabilizing synergist in the early 1980s, a vast amount of literature on such synergists has been published. This Review addresses a range of peptide-based and small organic compounds that disrupt the OM to elicit a synergistic effect with antibiotics that are otherwise inactive toward Gram-negative bacteria, with synergy defined as a fractional inhibitory concentration index (FICI) of <0.5. Another requirement for the inclusion of the synergists here covered is their potentiation of a specific set of clinically used antibiotics: erythromycin, rifampicin, novobiocin, or vancomycin. In addition, we have focused on those synergists with reported activity against Gram-negative members of the ESKAPE family of pathogens namely, Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, and/or Acinetobacter baumannii. In cases where the FICI values were not directly reported in the primary literature but could be calculated from the published data, we have done so, allowing for more direct comparison of potency with other synergists. We also address the hemolytic activity of the various OM-disrupting synergists reported in the literature, an effect that is often downplayed but is of key importance in assessing the selectivity of such compounds for Gram-negative bacteria.
Collapse
|
8
|
Yu B, Choudhury MR, Yang X, Benoit SL, Womack E, Van Mouwerik Lyles K, Acharya A, Kumar A, Yang C, Pavlova A, Zhu M, Yuan Z, Gumbart JC, Boykin DW, Maier RJ, Eichenbaum Z, Wang B. Restoring and Enhancing the Potency of Existing Antibiotics against Drug-Resistant Gram-Negative Bacteria through the Development of Potent Small-Molecule Adjuvants. ACS Infect Dis 2022; 8:1491-1508. [PMID: 35801980 PMCID: PMC11227883 DOI: 10.1021/acsinfecdis.2c00121] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The rapid and persistent emergence of drug-resistant bacteria poses a looming public health crisis. The possible task of developing new sets of antibiotics to replenish the existing ones is daunting to say the least. Searching for adjuvants that restore or even enhance the potency of existing antibiotics against drug-resistant strains of bacteria represents a practical and cost-effective approach. Herein, we describe the discovery of potent adjuvants that extend the antimicrobial spectrum of existing antibiotics and restore their effectiveness toward drug-resistant strains including mcr-1-expressing strains. From a library of cationic compounds, MD-100, which has a diamidine core structure, was identified as a potent antibiotic adjuvant against Gram-negative bacteria. Further optimization efforts including the synthesis of ∼20 compounds through medicinal chemistry work led to the discovery of a much more potent compound MD-124. MD-124 was shown to sensitize various Gram-negative bacterial species and strains, including multidrug resistant pathogens, toward existing antibiotics with diverse mechanisms of action. We further demonstrated the efficacy of MD-124 in an ex vivo skin infection model and in an in vivo murine systemic infection model using both wild-type and drug-resistant Escherichia coli strains. MD-124 functions through selective permeabilization of the outer membrane of Gram-negative bacteria. Importantly, bacteria exhibited low-resistance frequency toward MD-124. In-depth computational investigations of MD-124 binding to the bacterial outer membrane using equilibrium and steered molecular dynamics simulations revealed key structural features for favorable interactions. The very potent nature of such adjuvants distinguishes them as very useful leads for future drug development in combating bacterial drug resistance.
Collapse
Affiliation(s)
- Bingchen Yu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303 USA
| | - Manjusha Roy Choudhury
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303 USA
| | - Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303 USA
| | | | - Edroyal Womack
- Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | | | - Atanu Acharya
- School of Physics and School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332 United States
| | - Arvind Kumar
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303 USA
| | - Ce Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303 USA
| | - Anna Pavlova
- School of Physics and School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332 United States
| | - Mengyuan Zhu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303 USA
| | - Zhengnan Yuan
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303 USA
| | - James C. Gumbart
- School of Physics and School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332 United States
| | - David W. Boykin
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303 USA
| | - Robert J. Maier
- Department of Microbiology, University of Georgia, Athens, GA 30602 USA
| | - Zehava Eichenbaum
- Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303 USA
| |
Collapse
|
9
|
Zhang Y, Rosado-Lugo JD, Datta P, Sun Y, Cao Y, Banerjee A, Yuan Y, Parhi AK. Evaluation of a Conformationally Constrained Indole Carboxamide as a Potential Efflux Pump Inhibitor in Pseudomonas aeruginosa. Antibiotics (Basel) 2022; 11:716. [PMID: 35740123 PMCID: PMC9220351 DOI: 10.3390/antibiotics11060716] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
Efflux pumps in Gram-negative bacteria such as Pseudomonas aeruginosa provide intrinsic antimicrobial resistance by facilitating the extrusion of a wide range of antimicrobials. Approaches for combating efflux-mediated multidrug resistance involve, in part, developing indirect antimicrobial agents capable of inhibiting efflux, thus rescuing the activity of antimicrobials previously rendered inactive by efflux. Herein, TXA09155 is presented as a novel efflux pump inhibitor (EPI) formed by conformationally constraining our previously reported EPI TXA01182. TXA09155 demonstrates strong potentiation in combination with multiple antibiotics with efflux liabilities against wild-type and multidrug-resistant (MDR) P. aeruginosa. At 6.25 µg/mL, TXA09155, showed ≥8-fold potentiation of levofloxacin, moxifloxacin, doxycycline, minocycline, cefpirome, chloramphenicol, and cotrimoxazole. Several biophysical and genetic studies rule out membrane disruption and support efflux inhibition as the mechanism of action (MOA) of TXA09155. TXA09155 was determined to lower the frequency of resistance (FoR) to levofloxacin and enhance the killing kinetics of moxifloxacin. Most importantly, TXA09155 outperformed the levofloxacin-potentiation activity of EPIs TXA01182 and MC-04,124 against a CDC/FDA panel of MDR clinical isolates of P. aeruginosa. TXA09155 possesses favorable physiochemical and ADME properties that warrant its optimization and further development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ajit K. Parhi
- TAXIS Pharmaceuticals, Inc., 9 Deer Park Drive, Suite J-15, Monmouth Junction, NJ 08852, USA; (Y.Z.); (J.D.R.-L.); (P.D.); (Y.S.); (Y.C.); (A.B.); (Y.Y.)
| |
Collapse
|
10
|
Evaluation of Heterocyclic Carboxamides as Potential Efflux Pump Inhibitors in Pseudomonas aeruginosa. Antibiotics (Basel) 2021; 11:antibiotics11010030. [PMID: 35052908 PMCID: PMC8772707 DOI: 10.3390/antibiotics11010030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/19/2022] Open
Abstract
The ability to rescue the activity of antimicrobials that are no longer effective against bacterial pathogens such as Pseudomonas aeruginosa is an attractive strategy to combat antimicrobial drug resistance. Herein, novel efflux pump inhibitors (EPIs) demonstrating strong potentiation in combination with levofloxacin against wild-type P. aeruginosa ATCC 27853 are presented. A structure activity relationship of aryl substituted heterocyclic carboxamides containing a pentane diamine side chain is described. Out of several classes of fused heterocyclic carboxamides, aryl indole carboxamide compound 6j (TXA01182) at 6.25 µg/mL showed 8-fold potentiation of levofloxacin. TXA01182 was found to have equally synergistic activities with other antimicrobial classes (monobactam, fluoroquinolones, sulfonamide and tetracyclines) against P. aeruginosa. Several biophysical and genetic studies rule out membrane disruption and support efflux inhibition as the mechanism of action (MOA) of TXA01182. TXA01182 was determined to lower the frequency of resistance (FoR) of the partner antimicrobials and enhance the killing kinetics of levofloxacin. Furthermore, TXA01182 demonstrated a synergistic effect with levofloxacin against several multidrug resistant P. aeruginosa clinical isolates.
Collapse
|
11
|
New potentiators of ineffective antibiotics: Targeting the Gram-negative outer membrane to overcome intrinsic resistance. Curr Opin Chem Biol 2021; 66:102099. [PMID: 34808425 DOI: 10.1016/j.cbpa.2021.102099] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 12/20/2022]
Abstract
Because of the rise in antibiotic resistance and the dwindling pipeline of effective antibiotics, it is imperative to explore avenues that breathe new life into existing drugs. This is particularly important for intrinsically resistant Gram-negative bacteria, which are exceedingly difficult to treat. The Gram-negative outer membrane (OM) prevents the entry of a plethora of antibiotics that are effective against Gram-positive bacteria, despite the presence of the targets of these drugs. Uncovering molecules that increase the permeability of the OM to sensitize Gram-negative bacteria to otherwise ineffective antibiotics is an approach that has recently garnered increased attention in the field. In this review, we survey chemical matter which has been shown to potentiate antibiotics against Gram-negative bacteria by perturbing the OM. These include peptides, nanoparticles, macromolecules, antibiotic conjugates, and small molecules.
Collapse
|
12
|
Salem MA, Abu‐Hashem AA, Abdelgawad AAM, Gouda MA. Synthesis and reactivity of thieno[2,3‐ b]quinoline derivatives (Part II). J Heterocycl Chem 2021; 58:1705-1740. [DOI: 10.1002/jhet.4269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 03/31/2021] [Indexed: 11/12/2022]
Abstract
AbstractAs a continuation of our previous review entitled “Vilsmeier‐Haack cyclisation as a facile synthetic route to thieno[2,3‐b]quinolines (Part I).” This review describes the methods of preparation and the chemical reactivity of thieno[2,3‐b]quinolines, which might show interesting biological activities.
Collapse
Affiliation(s)
- Mohammed A. Salem
- Department of Chemistry, Faculty of Science and Arts King Khalid University Mohail Assir Saudi Arabia
- Department of Chemistry, Faculty of Science Al‐Azhar University Cairo Egypt
| | - Ameen A. Abu‐Hashem
- Photochemistry Department (Heterocyclic Unit) National Research Centre Giza Egypt
- Chemistry Departments, Faculty of Science Jazan University Jazan Saudi Arabia
| | - Ahmed A. M. Abdelgawad
- Chemistry Departments, Faculty of Science Jazan University Jazan Saudi Arabia
- Medicinal and Aromatic Plants Department Desert Research Center Cairo Egypt
| | - Moustafa A. Gouda
- Department of Chemistry, Faculty of Science and Arts Taibah University, Ulla Medina Saudi Arabia
- Department of Chemistry Faculty of Science, Mansoura University, Mansoura Egypt
| |
Collapse
|
13
|
Roy MMD, Baird SR, Dornsiepen E, Paul LA, Miao L, Ferguson MJ, Zhou Y, Siewert I, Rivard E. A Stable Homoleptic Divinyl Tetrelene Series. Chemistry 2021; 27:8572-8579. [PMID: 33848023 PMCID: PMC8252546 DOI: 10.1002/chem.202100969] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Indexed: 12/16/2022]
Abstract
The synthesis of the new bulky vinyllithium reagent (Me IPr=CH)Li, (Me IPr=[(MeCNDipp)2 C]; Dipp=2,6-iPr2 C6 H3 ) is reported. This vinyllithium precursor was found to act as a general source of the anionic 2σ, 2π-electron donor ligand [Me IPr=CH]- . Furthermore, a high-yielding route to the degradation-resistant SiII precursor Me IPr⋅SiBr2 is presented. The efficacy of (Me IPr=CH)Li in synthesis was demonstrated by the generation of a complete inorganic divinyltetrelene series (Me IPrCH)2 E: (E=Si to Pb). (Me IPrCH)2 Si: represents the first two-coordinate acyclic silylene not bound by heteroatom donors, with dual electrophilic and nucleophilic character at the SiII center noted. Cyclic voltammetry shows this electron-rich silylene to be a potent reducing agent, rivalling the reducing power of the 19-electron complex cobaltocene (Cp2 Co).
Collapse
Affiliation(s)
- Matthew M. D. Roy
- Department of ChemistryUniversity of Alberta11227 Saskatchewan Dr.EdmontonAlbertaT6G 2G2Canada
| | - Samuel R. Baird
- Department of ChemistryUniversity of Alberta11227 Saskatchewan Dr.EdmontonAlbertaT6G 2G2Canada
| | - Eike Dornsiepen
- Department of ChemistryUniversity of Alberta11227 Saskatchewan Dr.EdmontonAlbertaT6G 2G2Canada
| | - Lucas A. Paul
- Universität GöttingenInstitut für Anorganische ChemieTammannstr. 437077GöttingenGermany
| | - Linkun Miao
- Department of ChemistryUniversity of Alberta11227 Saskatchewan Dr.EdmontonAlbertaT6G 2G2Canada
| | - Michael J. Ferguson
- Department of ChemistryUniversity of Alberta11227 Saskatchewan Dr.EdmontonAlbertaT6G 2G2Canada
| | - Yuqiao Zhou
- Department of ChemistryUniversity of Alberta11227 Saskatchewan Dr.EdmontonAlbertaT6G 2G2Canada
| | - Inke Siewert
- Universität GöttingenInstitut für Anorganische ChemieTammannstr. 437077GöttingenGermany
| | - Eric Rivard
- Department of ChemistryUniversity of Alberta11227 Saskatchewan Dr.EdmontonAlbertaT6G 2G2Canada
| |
Collapse
|
14
|
Sadeer NB, Mahomoodally MF. Antibiotic Potentiation of Natural Products: A Promising Target to Fight Pathogenic Bacteria. Curr Drug Targets 2021; 22:555-572. [PMID: 32972338 DOI: 10.2174/1389450121666200924113740] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/29/2020] [Accepted: 07/08/2020] [Indexed: 01/13/2023]
Abstract
Pathogenic microorganisms should be considered as the number one foe of human, as witnessed by recent outbreaks of coronavirus disease (COVID-19) and with bacteria no longer sensitive to existing antibiotics. The resistance of pathogenic bacteria and deaths attributable to bacterial infections is increasing exponentially. Bacteria used different mechanisms to counterattack to existing antibiotics, namely (i) enzymatic inhibition, (ii) penicillin-binding protein modification, (iii) porin mutations, (iv) efflux pumps and (v) molecular modifications of antibiotic targets. Developing new antibiotics would be time-consuming to address such a situation, thus one of the promising approaches is by potentiating existing antibiotics. Plants used synergism to naturally defend and protect themselves from microbes. Using the same strategy, several studies have shown that the combinations of natural products and antibiotics could effectively prolong the lifespan of existing antibiotics and minimize the impact and emergence of antibiotic resistance. Combining essential oils constituents, namely uvaol, ferruginol, farnesol and carvacrol, with antibiotics, have proved to be efficient efflux pump inhibitors. Plant-derived compounds such as gallic acid and tannic acid are effective potentiators of various antibiotics, including novobiocin, chlorobiocin, coumermycin, fusidic acid, and rifampicin, resulting in a 4-fold increase in the potencies of these antibiotics. Several lines of research, as discussed in this review, have demonstrated the effectiveness of natural products in potentiating existing antibiotics. For this reason, the search for more efficient combinations should be an ongoing process with the aim to extend the life of the ones that we have and may preserve the life for the ones that are yet to come.
Collapse
Affiliation(s)
- Nabeelah Bibi Sadeer
- Department of Health Sciences, Faculty of Science, University of Mauritius, 230Reduit, Mauritius
| | - Mohamad Fawzi Mahomoodally
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam.,Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| |
Collapse
|
15
|
Spósito L, Fortunato GC, de Camargo BAF, Ramos MADS, Souza MPCD, Meneguin AB, Bauab TM, Chorilli M. Exploiting drug delivery systems for oral route in the peptic ulcer disease treatment. J Drug Target 2021; 29:1029-1047. [PMID: 33729081 DOI: 10.1080/1061186x.2021.1904249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Peptic ulcer disease (PUD) is a common condition that is induced by acid and pepsin causing lesions in the mucosa of the duodenum and stomach. The pathogenesis of PUD is a many-sided scenario, which involves an imbalance between protective factors, such as prostaglandins, blood flow, and cell renewal, and aggressive ones, like alcohol abuse, smoking, Helicobacter pylori colonisation, and the use of non-steroidal anti-inflammatory drugs. The standard oral treatment is well established; however, several problems can decrease the success of this therapy, such as drug degradation in the gastric environment, low oral bioavailability, and lack of vectorisation to the target site. In this way, the use of strategies to improve the effectiveness of these conventional drugs becomes interesting. Currently, the use of drug delivery systems is being explored as an option to improve the drug therapy limitations, such as antimicrobial resistance, low bioavailability, molecule degradation in an acid environment, and low concentration of the drug at the site of action. This article provides a review of oral drug delivery systems looking for improving the treatment of PUD.
Collapse
Affiliation(s)
- Larissa Spósito
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Giovanna Capaldi Fortunato
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Bruna Almeida Furquim de Camargo
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | | | | | - Andréia Bagliotti Meneguin
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Taís Maria Bauab
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Marlus Chorilli
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| |
Collapse
|
16
|
Janas A, Pecyna P, Gajecka M, Bartl F, Przybylski P. Synthesis and Antibacterial Activity of New
N
‐Alkylammonium and Carbonate‐Triazole Derivatives within Desosamine of 14‐ and 15‐Membered Lactone Macrolides. ChemMedChem 2020; 15:1529-1551. [DOI: 10.1002/cmdc.202000273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/21/2020] [Indexed: 02/03/2023]
Affiliation(s)
- Anna Janas
- Faculty of ChemistryAdam Mickiewicz University Uniwersytetu Poznańskiego 8 61-614 Poznań Poland
| | - Paulina Pecyna
- Chair and Department of Genetics and Pharmaceutical MicrobiologyPoznań University of Medical Sciences (PUMS) Święcickiego 4 60-781 Poznań Poland
| | - Marzena Gajecka
- Chair and Department of Genetics and Pharmaceutical MicrobiologyPoznań University of Medical Sciences (PUMS) Święcickiego 4 60-781 Poznań Poland
- Institute of Human GeneticsPolish Academy of Sciences Strzeszynska 32 60-479 Poznań Poland
| | - Franz Bartl
- Lebenswissenschaftliche Fakultät, Institut für Biologie Biophysikalische ChemieHumboldt-Universität zu Berlin Invalidenstrasse 42 10099 Berlin Germany
| | - Piotr Przybylski
- Faculty of ChemistryAdam Mickiewicz University Uniwersytetu Poznańskiego 8 61-614 Poznań Poland
| |
Collapse
|
17
|
Advances in the structural studies of antibiotic potentiators against Escherichia coli. Bioorg Med Chem 2019; 27:3254-3278. [PMID: 31235265 DOI: 10.1016/j.bmc.2019.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 05/28/2019] [Accepted: 06/01/2019] [Indexed: 11/24/2022]
|