2018
DOI: 10.1101/495630
|View full text |Cite
Preprint
|
Sign up to set email alerts
|

Antibiotic usage promotes the evolution of resistance against gepotidacin, a novel multi-targeting drug

Abstract: Multi-targeting antibiotics, i.e. single compounds capable to inhibit two or more bacterial targets offer a promising therapeutic strategy, but information on resistance evolution against such drugs is scarce.Gepotidacin is an antibiotic candidate that selectively inhibits both bacterial DNA gyrase and topoisomerase IV. In a susceptible organism, Klebsiella pneumoniae, a combination of two specific mutations in these target proteins provide an over 2000-fold increment in resistance, while individually none of … Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1
1
1

Citation Types

0
4
0

Year Published

2020
2020
2022
2022

Publication Types

Select...
3
1

Relationship

3
1

Authors

Journals

citations
Cited by 4 publications
(4 citation statements)
references
References 73 publications
0
4
0
Order By: Relevance
“…By playing with a stringent expression system for rec2, applying multiple cycles of recombinase production/oligonucleotide transformation, and reversibly inhibiting the MMR system during a limited time window we report in the following discussion high-fidelity recombination frequencies that approach those achieved with the archetypal Red-based system (Datsenko and Wanner, 2000). This opens genome editing possibilities in this environmental bacterium that were thus far limited to strains of E. coli, closely related enteric species (Nyerges et al, 2018;Szili et al, 2019), and some lactic acid bacteria (van Pijkeren et al, 2012).…”
Section: Introductionmentioning
confidence: 87%
“…By playing with a stringent expression system for rec2, applying multiple cycles of recombinase production/oligonucleotide transformation, and reversibly inhibiting the MMR system during a limited time window we report in the following discussion high-fidelity recombination frequencies that approach those achieved with the archetypal Red-based system (Datsenko and Wanner, 2000). This opens genome editing possibilities in this environmental bacterium that were thus far limited to strains of E. coli, closely related enteric species (Nyerges et al, 2018;Szili et al, 2019), and some lactic acid bacteria (van Pijkeren et al, 2012).…”
Section: Introductionmentioning
confidence: 87%
“…Compared with fluoroquinolones, gepotidacin has a unique binding site on GyrA and topoisomerase IV (subunit ParC). Nevertheless, early findings indicate that fluoroquinolone resistance might be a stepping stone for gepotidacin resistance as the target sites on topoisomerase IV are in close proximity to each other (preprint: Szili et al , 2018 ; World Health Organization, 2022 ).…”
Section: Analysis Of the Clinical And Preclinical Antibacterial Pipelinementioning
confidence: 99%
“…genome editing (i.e., MAGE and DIvERGE) holds the promise of enabling rapid analysis of genotype-to-phenotype relationships and predicting future mechanisms of antimicrobial resistance (13,46). In contrast, C. freundii is an intriguing biomanufacturing host in which the optimization of metabolic pathways has remained challenging (47,48).…”
Section: Screening Diverse Rect Homologs Identifies a Highly Efficienmentioning
confidence: 99%
“…138474) were constructed by replacing the Redβ ORF of pORTMAGE311B plasmid (Addgene accession no. 120418) (46) with PapRecT and CspRecT, respectively. pORTMAGE-Pa1 was constructed in many steps: 1) The Kanamycin resistance cassette and the RSF1010 origin-of-replication on pORTMAGE312B with Gentamicin resistance marker and pBBR1 origin-of-replication, amplified from pSEVA631 (75) (gift from Victor de Lorenzo, Consejo Superior de Investigaciones Cientificas, Madrid, Spain); 2) optimization of RBSs in pORTMAGE-Pa1 was done by designing a 30-nt optimal RBS in front of the SSAP ORF and in between the SSAP and MutL ORFs with an automated design program, De Novo DNA (76); 3) PaMutL was amplified from P. aeruginosa genomic DNA and cloned in place of EcMutL_E32K; and finally 4) PaMutL was mutated by site-directed mutagenesis to encode E36K.…”
Section: Seermentioning
confidence: 99%