678
Views
0
CrossRef citations to date
0
Altmetric
Special Report

PROTAC antibiotics: the time is now

ORCID Icon, , & ORCID Icon
Pages 363-370 | Received 11 Oct 2022, Accepted 06 Feb 2023, Published online: 21 Feb 2023

References

  • Munita JM, Arias CA. Mechanisms of antibiotic resistance. Microbiol Spectr. 2016;4(2):VMBF-0016–2015.
  • Interagency Coordination Group on antimicrobial resistance. No time to wait: securing the future from drug-resistant infections. Geneva: World Health Organization; 2019.
  • Cowman S, van Ingen J, Griffith DE, et al. Non-tuberculous mycobacterial pulmonary disease. Eur Respir J. 2019;54(1):1900250.
  • Tortoli E, Fedrizzi T, Meehan CJ, et al. The new phylogeny of the genus Mycobacterium: the old and the news. Infect Genet Evol. 2017;56:19–25.
  • Griffith DE. Therapy of nontuberculous mycobacterial disease. Curr Opin Infect Dis. 2007;20(2):198–203.
  • Wu M-L, Aziz DB, Dartois V, et al. NTM drug discovery: status, gaps and the way forward. Drug Discov Today. 2018;23(8):1502–1519.
  • Cohen J, Powderly WG, Opal SM. Infectious Diseases. Third ed. Edinburgh: Mosby/Elsevier; 2010.
  • Pankey GA, Sabath L. Clinical relevance of bacteriostatic versus bactericidal mechanisms of action in the treatment of Gram-positive bacterial infections. Clin Infect Dis. 2004;38(6):864–870.
  • Urbaniec J, Xu Y, Hu Y, et al. Phenotypic heterogeneity in persisters: a novel ‘hunker’ theory of persistence. FEMS Microbiol Rev. 2022;46(1):fuab042.
  • Lai AC, Crews CM. Induced protein degradation: an emerging drug discovery paradigm. Nat Rev Drug Discov. 2017;16(2):101–114.
  • Cook MA, Wright GD. The past, present, and future of antibiotics. Sci Transl Med. 2022;14(657):eabo7793.
  • Bondeson DP, Crews CM. Targeted protein degradation by small molecules. Annu Rev Pharmacol Toxicol. 2017;57:107–123.
  • Paiva S-L, Crews CM. Targeted protein degradation: elements of PROTAC design. Curr Opin Chem Biol. 2019;50:111–119.
  • Békés M, Langley DR, Crews CM. PROTAC targeted protein degraders: the past is prologue. Nat Rev Drug Discov. 2022;21(3):181–200.
  • Bond MJ, Crews CM. Proteolysis targeting chimeras (PROTACs) come of age: entering the third decade of targeted protein degradation. RSC Chem Biol. 2021;2:725–742.
  • Izert MA, Klimecka MM, Górna MW. Applications of bacterial degrons and degraders—Toward targeted protein degradation in bacteria. Front Mol Biosci. 2021;8:669762.
  • Grohmann C, Marapana DS, Ebert G. Targeted protein degradation at the host–pathogen interface. Mol Microbiol. 2022;117(3):670–681.
  • Gopal P, Dick T. Targeted protein degradation in antibacterial drug discovery? Prog Biophys Mol Biol. 2020;152:10–14.
  • Powell M, Blaskovich MA, Hansford KA. Targeted protein degradation: the new frontier of antimicrobial discovery? ACS Infect Dis. 2021;7(8):2050–2067.
  • Scorpio A, Zhang Y. Mutations in pncA, a gene encoding pyrazinamidase/nicotinamidase, cause resistance to the antituberculous drug pyrazinamide in tubercle bacillus. Nat Med. 1996;2(6):662–667.
  • Gopal P, Yee M, Sarathy J, et al. Pyrazinamide resistance is caused by two distinct mechanisms: prevention of coenzyme A depletion and loss of virulence factor synthesis. ACS Infect Dis. 2016;2(9):616–626.
  • Gopal P, Tasneen R, Yee M, et al. In vivo-selected pyrazinoic acid-resistant Mycobacterium tuberculosis strains harbor missense mutations in the aspartate decarboxylase PanD and the unfoldase ClpC1. ACS Infect Dis. 2017;3(7):492–501.
  • Gopal P, Nartey W, Ragunathan P, et al. Pyrazinoic acid inhibits mycobacterial coenzyme A biosynthesis by binding to aspartate decarboxylase PanD. ACS Infect Dis. 2017;3(11):807–819.
  • Gopal P, Sarathy JP, Yee M, et al. Pyrazinamide triggers degradation of its target aspartate decarboxylase. Nat Commun. 2020;11(1):1661.
  • Yee M, Gopal P, Dick T. Missense mutations in the unfoldase ClpC1 of the caseinolytic protease complex are associated with pyrazinamide resistance in Mycobacterium tuberculosis. Antimicrob Agents Chemother. 2017;61(2):e02342–16.
  • Akopian T, Kandror O, Raju RM, et al. The active ClpP protease from M. tuberculosis is a complex composed of a heptameric ClpP1 and a ClpP2 ring. EMBO J. 2012;31(6):1529–1541.
  • Osborne C, Wakeling A, Nicholson R. Fulvestrant: an oestrogen receptor antagonist with a novel mechanism of action. Br J Cancer. 2004;90(Suppl 1):S2–S6.
  • Morreale FE, Kleine S, Leodolter J, et al. BacPROTACs mediate targeted protein degradation in bacteria. Cell. 2022;185(13):2338–2353.e18.
  • Trentini DB, Suskiewicz MJ, Heuck A, et al. Arginine phosphorylation marks proteins for degradation by a Clp protease. Nature. 2016;539(7627):48–53.
  • DeMonte D, Drake EJ, Lim KH, et al. Structure‐based engineering of streptavidin monomer with a reduced biotin dissociation rate. Proteins. 2013;81(9):1621–1633.
  • Schmidt A, Trentini DB, Spiess S, et al. Quantitative phosphoproteomics reveals the role of protein arginine phosphorylation in the bacterial stress response. Mol Cell Proteomics. 2014;13(2):537–550.
  • Vasudevan D, Rao SP, Noble CG. Structural basis of mycobacterial inhibition by cyclomarin A. J Biol Chem. 2013;288(43):30883–30891.
  • Winter GE, Buckley DL, Paulk J, et al. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science. 2015;348(6241):1376–1381.
  • Zengerle M, Chan K-H CA. Selective small molecule induced degradation of the BET bromodomain protein BRD4. ACS Chem Biol. 2015;10(8):1770–1777.
  • Chen J, Zhao L, Mao Y, et al. Clinical efficacy and adverse effects of antibiotics used to treat Mycobacterium abscessus pulmonary disease. Front Microbiol. 2019;10:1977.
  • Balavoine C, Blanc F-X, Lanotte P, et al. Adverse events during treatment of nontuberculous mycobacterial lung disease: do they really matter? Eur Respir J. 2018;52(Suppl 62): PA2664.
  • Forget EJ, Menzies D. Adverse reactions to first-line antituberculosis drugs. Expert Opin Drug Saf. 2006;5(2):231–249.
  • Burslem GM, Crews CM. Proteolysis-targeting chimeras as therapeutics and tools for biological discovery. Cell. 2020;181(1):102–114.
  • Kaya F, Ernest JP, LoMauro K, et al. A rabbit model to study antibiotic penetration at the site of infection for nontuberculous mycobacterial lung disease: macrolide case study. Antimicrob Agents Chemother. 2022;66(3):e02212–21.
  • Fields FR, Lee SW, McConnell MJ. Using bacterial genomes and essential genes for the development of new antibiotics. Biochem Pharmacol. 2017;134:74–86.
  • Jiang B, Gao Y, Che J, et al. Discovery and resistance mechanism of a selective CDK12 degrader. Nat Chem Biol. 2021;17(6):675–683.
  • Zhang L, Riley-Gillis B, Vijay P, et al. Acquired resistance to BET-PROTACs (proteolysis-targeting chimeras) caused by genomic alterations in core components of E3 ligase complexes. Mol Cancer Ther. 2019;18(7):1302–1311.
  • Ramirez J, Guarner F, Bustos Fernandez L, et al. Antibiotics as major disruptors of gut microbiota. Front Cell Infect Microbiol. 2020;10:572912.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.