Directions in the development of modern and promising antimicrobial agents

Tetiana S. Todosiichuk1,

Serhii O. Soloviov1,3,

Lin Wu1,2,

Iryna V. Dzyublyk3,

Olena P. Trokhimenko3,

Magdalena Dudek4,

Artem Symchuk4,

Volodymyr Vasylenko5*

1 National Technical University of Ukraine ‘Igor Sikorsky Kyiv Polytechnic Institute’, Prospect Peremogy 37, Kyiv 03056, Ukraine

2 Hainan Medical University, Haikou, Xueyuan Road 3, Longhua District, Haikou City, Hainan Province, China

3 Shupyk National Healthcare University of Ukraine, Dorohozhytska St 9, Kyiv 04112, Ukraine

4 University Hospital of Lord’s Transfiguration, Dluga 1/2, 61-848, Poznań, Poland

5 Vytautas Magnus University, K. Donelaičio St. 58, Kaunas 44248, Lithuania

The use of antibiotics is becoming increasingly limited. This is mainly due to the development of resistance to pathogenic bacteria, and, over time, more and more bacteria will become resistant to antimicrobials. This problem inevitably leads to the conclusion that studies into alternative methods of combating pathogens, which are necessary to develop sufficiently reliable and effective therapies for bacterial infections, are indispensable. This review highlights some recent developments in conventional antibiotic and non-antibiotic treatment strategies. It has been shown that traditional antibacterial targets include derivatives of known antibiotic classes, new chemical classes with new targets, as well as unknown or undefined agents with unclear targets. Promising strategies for combating microbial pathogens have been identified, including new targets, namely, toxin secretion systems, biofilm formation, and adhesion mechanisms that affect quorum sensing of microbial populations. In addition, it is important to use new antimicrobial agents with other, non-antibiotic, mechanisms of action: phage and phage-derived peptides, microbiotamodulating therapies, and enhancing immune response.

Keywords: conventional antibiotic targets, nonantibiotic targets, nonantibiotic treatment, infections, resistance

INTRODUCTION

The advent of antibiotics in the  first half of the  twentieth century catalysed a  medical revolution by drastically reducing mortality from bacterial infections. Antibiotics contributed significantly to an extension in the average life expectancy in the USA from 59.7 years in 1930 to 78.7 years in 2010 (Arias et al., 2016), providing clinicians with a  wide range of tools to prevent and fight bacterial infections. The mechanisms of action of these substances are mainly related to blocking the development and reproduction of bacterial cells, which leads to the disruption of the synthesis of their cell wall and cytoplasmic membrane. Very quickly, however, due to the high adaptability of pathogens and the  acquisition of resistance, these substances lose their relevance as antimicrobial substances. To solve this problem, the existing classes of antibiotics with limited cross-resistance to existing drugs have been modified and new classes of antibiotics have been introduced (Aminov, 2010).

However, the result of uncontrolled or widespread use of antimicrobials is microbial resistance to the use of each new drug, and the proliferation of resistant pathogens is becoming a major problem (Palumbi, 2001; Tsiodras et al., 2001; Lewis et al., 2005; Clatworthy et al., 2007; Gentry et al., 2008; Morgan et al., 2011). Moreover, due to the high doses of antibiotics required to trigger efflux mechanisms in gram-negative bacteria, intoxication of the  body can occur (Baker et al., 2018). This threatens with the situation when microbial pathogens do not respond to the  existing antibiotics, and therefore there is a need to develop new effective strategies for treating infectious diseases with substances of a different nature. These methods include treatment aimed at modeling the microbiota, namely, the  use of antibacterial vaccines and antibodies; phages or phage-derived proteins , antiviral agents, potentiators that enhance, augment, or transform other agents, and the  use of immunomodulators developed to treat bacterial diseases (Tenson, Mankin, 2006; Motley, Fries, 2017; Theuretzbacher, Piddock, 2019; Kortright et al., 2019; Miró-Canturri, 2019; Rello  et  al., 2019; Monserrat-Martinez  et  al., 2019; Vaccines to tackle drug resistant infections 2019).

In this review, we explain some traditional targets to fight given the development of antibiotic resistance of bacteria, and discuss alternative ways to control microbial pathogens.

The main effect of antibiotics is achieved through direct destruction of the  pathogenic flora. The  vast majority of antibacterial molecules are artificially synthesised; they are not natural compounds. Their registration by regulatory authorities occurs according to simple and well-known algorithms. The development of this trend in pharmacology takes place in several directions: derivatisation of well-studied molecules; research into new known molecules with new target organisms and into innovative unexplored molecules with undefined targets.

DISCUSSION

Derivatives of the  classes of known antibiotics. Most modern antibiotics are derivatives of natural substances that have been used for many years (Fernebro, 2011). The  development of resistance of pathogens to antibiotics can be slowed down by modifying known and already used substances. Such modification may include increased intermolecular and intramolecular interactions and is considered an effective way to combat resistant forms of pathogens (Fernebro, 2011; Silver, 2007). For example, vancomycin could interrupt bacterial cell wall synthesis by binding with high proximity to peptidoglycan and preventing cell wall assembly (Perkins, 1982; Romaniuk, Cegelski, 2015). The emergence of vancomycin-resistant enterococci (VRE), which was associated with their ability to disrupt the factors of interaction with the antibiotic (repeats d-Ala-d-Ala), was recorded after several decades of use of this antibiotic (Noble et al., 1992; Murray, 2000; McKessar et al., 2000). Modified vancomycin was developed to avoid this ability of pathogenic enterococci (Xie et al., 2011; Okano et al., 2017), but this did not stop the growth of bacteria and they soon became resistant again.

New targets for new chemical groups. New target molecules contain new sites for interactions in the area of ribosomes, membranes, gene interference, and metabolism (Ma et al., 2016; Silver, 2016; Xie et al., 2018; Ciumac et al., 2019). Some of these approaches have already been studied, but clinical development has not begun.

Unknown or undefined agents with unclear targets. This group includes several basic groups of antimicrobial molecules depending on their origin and structure: synthetic or natural antimicrobial peptides and proteins (AMPs), natural products, and UDP-3-O-(R3-hydroxymyristoyl)-N-acetylglucosamine deacetylase (LpxC) inhibitors. AMP is a wellstudied natural compound with clear antibacterial effects. These molecules are the basis for the synthesis of peptides and peptide-like molecules (Kang  et  al., 2017; Molchanova  et  al., 2017; Moravej et al., 2018; Torres et al., 2019). The  main goals for research in this direction consist in reducing the  cost of production of the substances, reducing the impact of protolytic degradation to increase the half-life in vivo, and improving safety (Kuppusamy et al., 2019). The basis of the modern market for antibiotics is precisely such natural substances. Developments in the  field of genomics and chemical technologies enable increasing the  effectiveness of research in the  field of pharmacology of natural molecules (Fedorenko  et  al., 2015; Wright, 2017; Rossiter  et  al., 2017). LpxC inhibitors targeting the first stage of lipid A production have been studied exclusively prior to phase I clinical trials. This is due to toxicity, which was identified in a mouse model. In addition, significant local inflammatory reactions were noted after injections (Erwin, 2016; Liu, Ma, 2018; Cohen et al., 2019). The toxicity of the molecules used to inhibit LpxC is the main obstacle to the development of this direction. A growing body of knowledge in this area, such as recent CARB-X-supported studies related to LpxC inhibition (the Shared Platform for Antibiotic Research and Knowledge (SPARK), Pew Trusts), may provide an impetus to overcome some of the barriers to the use of this group of antibiotics. This instance highlights the general difficulties associated with toxicity in transferring molecular safety models from preclinical trials to clinical use.

New nonantibiotic target. In this section, we present an overview of several ‘nonantibiotic’ approaches aimed at the  treatment and prevention of bacterial infections. When bacteria-caused infections occur, pathogens produce virulence factors. The  molecules allow pathogens to resist the  body’s self-cleansing, gain access to deeper tissues, penetrate them, and damage host cells. For the productive treatment of bacterial infections, agents have been developed that block the activity of virulence factors and therefore halt pathogenesis until the host’s immune response limits or kills the bacteria.

Targeting toxins and secretion systems. Secreted toxins play a significant role in the pathogenesis of many medically necessary bacteria, and several of these have been targeted with the aim of blocking infection. Usually, antibodies and antibody-drug conjugates are the inhibitors of a toxin. Currently, only three antibodies against bacterial infections have been approved for use (Lowy et al., 2010; López et al., 2015; Wilcox et al., 2017). Their main effect is the neutralisation of toxins that affect virulence and, consequently, the pathological process. To date, studies into the antibodies with multiple virulence determinants have not yielded positive results.

Some of the  antibody problems described also arise in the  development of vaccines against multi-resistant pathogens. The  lack of preclinical predictive models has led to the failure of a number of bacterial vaccines in late stages of clinical trials. The absence of such models may lead to poor vaccine efficacy in the future (Priebe, Goldberg, 2014; Redi et al., 2018). Published studies on the evaluation of promising vaccines showed the inconsistency of the target pathogens used for development or the uncertainty of development results (https:// vaccinesforamr.org). In order to affect human cells, toxins must overcome the  membranes of the bacterial cell, since they are synthesised directly in the bacterial cytosol. A number of specific secretory systems acquired by bacteria are associated with this. These systems can be used as targets for pharmaceuticals. An example is type III secretion systems (T3SSs) inherent in gram-negative bacteria. Type III secretion (T3S) is a multiprotein needle apparatus with which the bacterium can inject the produced compounds directly into the  cells of the host. It is known that T3S of different types of bacteria are similar to each other, despite differences in the secreted toxins. This leads to the conclusion that T3S inhibitors can be used to combat a wide range of pathogens (Swietnicki et al., 2011).

Targeting biofilms and adherence. Biofilms that form on inert surfaces such as catheters, prosthetic joints, or heart valves can interfere with the penetration of antibiotics and are major sources of infection (Kester, Fortune, 2013; Ribeiro et al., 2016). To prevent the occurrence of microbial infections both in the  biofilm and in the planktonic phase of growth, targeting the adhesins is used. With the help of such adhesins the pathogen is fixed on the mucosal surface (Krachler, Orth, 2013). In the process of infection, microbial pathogens change the internal environment of the host cell by attaching to cellular receptors of their own proteins, which leads to the disruption of the structure of cell membranes and stimulation of various intracellular processes (Shaw  et  al., 2001; Ide  et  al., 2001; Pinchuk  et  al., 2010; Wilson, 2014; Jimenez et al., 2016; Gilbert et al., 2017). One of these processes is the  inhibition of the activity of the membrane enzyme sortase A, which is a determining factor in the interaction of antibiotics with the peptidoglycan of the cell wall of gram-positive pathogens (Mazmanian  et  al., 2001; Ton-That, Schneewind, 2004; Zhang et al., 2014; Thappeta et al., 2020).

Quorum sensing. Quorum sensing is one of the  leading mechanisms of microorganism interaction within the  population thanks to which they can sense the influence of the external environment as a  single organism and produce a collective response. Such a response is often the activation or expression of genes, which depends on the concentration of autoin-ducers, hormone-like molecules that are synthesised because of intercellular interactions (Vasil, 2003; Waters, Bassler, 2005).

The critical concentration of autoinducers is a  signal to the  microorganism population to suppress the  manufacture of its own virulence factors in order to conceal and postpone the  host immune system’s reaction. (Deep, 2011). The  restoration of the  biosynthesis of substances responsible for the pathogenesis of inflammation occurs after the  accumulation of a high concentration of pathogens that can neutralize protective mechanisms, and thus the inflammatory process begins.

In the  regulation of the  synthesis of virulence factors by pathogens, their formation in the form of a biofilm also plays a role.

Among other factors, the  functioning of the quorum sensing (QS) system is ensured by the  biosynthesis of peptides that are specific to each microorganism, so they can be effective targets for the  action of new antiseptics. For example, gram-negative bacteria are dependent on acyl-homoserine lactones (AHL) (Pearson  et  al., 1994; Whitehead  et  al., 2001; Njoroge. Sperandio, 2009), the  precursor of which is S-adenosylmethionine (SAM) (Papenfort, Bassler, 2016), and oligopeptides are the signaling compounds for the QS system of gram-positive bacteria. The  latter have an individual sequence for each species of bacteria, and their synthesis is genetically determined (Ng, Bassler, 2009).

However, the  protein nature of these peptides (AHL, SAM) allows the use of appropriate enzymes (lactonases, acylases, or oxidoreductases) for their degradation, and thus inactivation of QS signals of pathogens and its inhibition (Leadbetter, Greenberg, 2000; Dong et al., 2000; Dong et al., 2001; Uroz et al., 2005; LaSarre, Federle, 2013). The  possibility of nonenzymatic inactivation of these peptides is also being studied. Another approach is based on blocking autoinducers of pathogens to deprive them of the opportunity to disguise themselves and become immediately ‘visible’ to the host’s immune system. By removing or inactivating autoinducers, it is possible to achieve high specificity of action of such antimicrobial drugs, which will also minimise the  development of pathogen resistance.

Phage and phage-derived peptides. Bacteriophages were used for antibacterial therapy before the  advent of antibiotics, and the  rise in infections caused by multidrug-resistent (MDR) strains of bacteria has sparked renewed interest in this therapy (Knoll, Mylonakis, 2014). Compared to traditional antibiotics, phages have important advantages: they do not infect human cells and practically do not affect the normal microbial flora (Rohde et al., 2018; Kortright  et  al., 2019; McCallin  et  al., 2019). In the past, phages were used in preparations only for external use (Morozova et al., 2018); currently, preparations for intravenous administration and aerosols are being developed (Chang et al., 2018).

To improve phage therapy, phages are developed to increase their infectivity and host range, and individual phage components are purified to target bacteria (Nobrega  et  al., 2015). Particular attention should be paid to endolysins, which are obtained from phages (Fischetti, 2018; Abdelkader  et  al., 2019). This group of substances is characterised by a highly specific bacteriolytic effect that occurs upon direct contact. There are reports of the  development of drugs against Staphylococcus aureus infections based on endolysins (Gentry et al., 2008). Molecular methods, namely protein engineering, lead to the emergence of new methods of combating gram-negative microorganisms (Oliveira et al., 2018), although such projects are not numerous and require in-depth study (Briers, Lavigne, 2015).

Microbiota-modulating therapies. Over the  past decades, we have become increasingly aware of the  importance of the  microbiome for human health and disease. A  link has been established between the human microbiota and disease condition, including diabetes, cardiovascular disease, and even mental health. The  main target of preclinical studies is Clostridium difficile, a representative of the intestinal microbiota. There is practically no mention of the microbiota from the lungs, sinuses, or the  skin. The  results of the  study of metagenomic, computational, and synthetic biology helped to renew the interest of researchers in the  human microbiota (Falony et al., 2019). In order to reduce the occurrence of C. difficile relapses and to correct other disorders, a  microbiota modification was used to study and test its effectiveness (Cammarota et al., 2017; Iqbal et al., 2018; Kellermayer, 2019). The  accuracy of genetic engineering using known probiotics is improved by such approaches (Waters, Bassler, 2005). Thus, it is possible to increase the expression of specific antimicrobials (Fehér  et  al., 2017; Ozdemir et al., 2018; Ghosh et al., 2019) and at the same time reduce the likelihood of transfer of potential pathogens (Ramachandran, Bikard, 2019).

Modulation of the  immune response. The  ability to adjust the  mechanisms of the  host’s immune response to infection can be considered one of the  important methods of combating pathogens that will prevent the  spread of infection in the  early stage. Directing macrophages to the site of infection and enhancing the immune response may eliminate the need for antibiotics altogether.

A number of authors demonstrated the role of antimicrobial peptides and proteins in eukaryotic and prokaryotic organisms in the formation of the immune response at the first line of infection (Zasloff, 2002; Ganz, 2003; Reddy  et  al., 2004; Bahar, Ren, 2013). Containing up to 60 amino acids, these molecules, act as antibiotics under physiological conditions and their antimicrobial activity is mainly due to membrane-lytic action. However, the concomitant effect of AMPs has been established, which consist in influencing the processes of cytokine activity, chemotaxis, antigen detection (Territo et al., 1989; Yang et al., 2000; Niyonsaba et al., 2002; Yang et al., 2002; Kurosaka  et  al., 2005), increased protection and inhibition of the infectious process, as well as wound healing (Heilborn  et  al., 2003; Tokumaru et al., 2005; Baroni et al., 2009), complementing the bactericidal effect of other antimicrobials (Lai, Gallo, 2009).

The latest research has developed vitaminbased nanoparticles. These nanoparticles are particularly good at delivering messenger RNA, while constructing messenger RNA encoding antimicrobial peptides and signal proteins and transporting them to macrophages produced by donor monocytes in the  cell. Since macrophages naturally have antibacterial activity, adding other antibacterial peptides to the cells facilitates further enhancement of the antibacterial activity and helps the macrophages eliminate bacteria (Hou et al., 2020).

Classical antibacterial drugs act on target organisms by altering cellular processes. As a result of many years of treatment of the population with high doses of antibiotics and their misuse, we have entered the ‘postantibiotic era’. Many microorganisms have become resistant to a  wide range of antibacterial drugs. New, modern antibiotics are being developed, but the same problem arises: the body is able to develop resistance to them.

CONCLUSIONS

The control of pathogenic microorganisms, which has been going on since the discovery of antibiotics, has led to the  emergence of super pathogens capable of withstanding high concentrations of antibiotics and their combinations. It is said that it is possible to return to the pre-antibiotic era, when there were no means to combat infectious diseases; a future is possible, however, when there really will be no such means, because human activities will lead to the emergence of resistant forms of pathogens.

Obviously, the  control strategy must be radically changed and focused not on killing the  pathogens themselves but on influencing the factors of their pathogenicity and virulence. Numerous attempts to find such approaches can be seen in recent decades. They are generally targeted at inhibiting host-pathogen contact, cell adhesion, or immune system response regulation, which opens up the  possibility of developing medications that can prevent disease progression even after the  pathogen has entered the body. The development of new antimicrobials should be based on an in-depth study of the pathways and mechanisms of pathogen virulence, and therefore the choice of targets that are associated with non-vital cell processes and are least protected by pathogen cells. The development of such new generation drugs has the prospect of long-term effectiveness in overcoming the  current problem of pathogen resistance and can provide a chance for the host organism with a low rate of evolution to successfully resist infectious diseases.

Author contributions: Conceptualisation, investigation, and visualisation, idea for the article, TST; methodology, VV; literature search, IVD and SOS; data analysis, SM; resources and data curation, LW; writing – original draft preparation, OPT and AS; writing – review and editing, MS and MD; supervision, SOS; software, KG. All authors read and approved the final manuscript.

Statements and declarations

Partial financial support was received from Vytautas Magnus University.

The authors have no relevant financial or non-financial interests to disclose.

The authors declare no conflict of interest.

Received 26 September 2022

Accepted 11 October 2022

References

1. Abdelkader K, Gerstmans H, Saafan A, Dishisha  T, Briers  Y. The  preclinical and clinical progress of bacteriophages and their lytic enzymes: the parts are easier than the whole. Viruses 2019; 11: 96.

2. Aminov  RI. A  brief history of the  antibiotic era: lessons learned and challenges for the future. Frontiers microbiol. 2010; 1: 134.

3. Arias E, Heron M, Xu J. United States life tables 2012. Natl Vital Stat Rep. 2016; 65: 1–65.

4. Bahar  AA, Ren  D. Antimicrobial peptides. Pharmaceut. 2013; 6: 1543–75.

5. Baker SJ, Payne DJ, Rappuoli R, Gregorio ED. Technologies to address antimicrobial resistance. Proc National Acad Sci. 2018; 115: 12887–95.

6. Baroni  A, Donnarumma  G, Paoletti  I, Longanesi-Cattani I, Bifulco K, Tufano MA, Carriero  MV. Antimicrobial human beta-defensin-2 stimulates migration, proliferation and tube formation of human umbilical vein endothelial cells. Peptides. 2009; 30: 267–72.

7. Briers Y, Lavigne R. Breaking barriers: expansion of the use of endolysins as novel antibacterials against gram-negative bacteria. Future Microbiol. 2015; 10: 377–90.

8. Cammarota  G, Ianiro  G, Tilg  H, RajilićStojanović M, Kump P, Satokari R, et al. European consensus conference on faecal microbiota transplantation in clinical practice. Gut. 2017; 66: 569–80.

9. Chang  RYK, Wallin  M, Lin  Y, Leung  SSY, Wang H, Morales S, Chan HK. Phage therapy for respiratory infections. Adv Drug Deliv Rev. 2018; 133: 76–86.

10. Ciumac D, Gong H, Hu X, Lu JR. Membrane targeting cationic antimicrobial peptides. J Colloid Interface Sci. 2019; 537: 163–85.

11. Clatworthy AE, Pierson E, Hung DT. Targeting virulence: a new paradigm for antimicrobial therapy. Nat Chem Biol. 2007; 3: 541–8.

12. Cohen  F, Aggen  JB, Andrews  LD, Assar  Z, Boggs J, Choi T, et al. Optimization of LpxC inhibitors for antibacterial activity and cardiovascular safety. Chem Med Chem. 2019; 14: 1560–1572.

13. Deep  A, Chaudhary  U, Gupta  V. Quorum sensing and bacterial pathogenicity: from molecules to disease. J Lab Physicians. 2011; 3: 4–11.

14. Dong YH, Xu JL, Li XZ, Zhang LH. AiiA, an enzyme that inactivates the  acylhomoserine lactone quorum-sensing signal and attenuates the virulence of Erwinia carotovora. Proc Natl Acad Sci. 2000; 97: 3526–31.

15. Dong  YH, Wang  LH, Xu  JL, Zhang  HB, Zhang  XF, Zhang  LH. Quenching quorumsensing-dependent bacterial infection by an N-acyl homoserine lactonase. Nature. 2001; 411: 813–7.

16. Erwin  AL. Antibacterial drug discovery targeting the lipopolysaccharide biosynthetic enzyme LpxC. Cold Spring Harb Perspect Med. 2016; 6: a025304.

17. Falony  G, Vandeputte  D, Caenepeel  C, Viera-Silva  S, Daryoush  T, Vermiere  S, Raes  J. The human microbiome in health and disease: hype or hope. Acta Clin Belg. 2019; 74: 53–64.

18. Fedorenko  V, Genilloud  O, Horbal  L, Marcone GL, Marinelli F, Paitan Y, Ron EZ. Antibacterial discovery and development: from gene to product and back. Biomed Res Int. 2015: 591349.

19. Fehér C, Soriano A, Mensa J. A review of experimental and off-label therapies for Clostridium difficile infection. Infect Dis Ther. 2017; 6: 1–35.

20. Fernebro  J. Fighting bacterial infections–future treatment options. Drug Resist Updates. 2011; 14: 125–39.

21. Fischetti VA. Development of phage lysins as novel therapeutics: a  historical perspective. Viruses. 2018; 10: 310.

22. Ganz T. Defensins: antimicrobial peptides of innate immunity. Nature Rev Immunol. 2003; 3: 710–20.

23. Gentry  DR, McCloskey  L, Gwynn  MN, Rittenhouse  SF, Scangarella  N, Shawar  R,  et  al. Genetic characterization of Vga ABC proteins conferring reduced susceptibility to pleuromutilins in Staphylococcus aureus. Antimicrob Agents Chemother. 2008; 52: 4507–9.

24. Ghosh C, Sarkar P, Issa R, Haldar J. Alternatives to conventional antibiotics in the era of antimicrobial resistance. Trends Microbiol. 2019; 27: 323–38.

25. Gilbert  RJC, Bayley  H, Anderluh  G. Membrane pores: from structure and assembly, to medicine and technology. Phil Trans R Soc. 2017; 372: 20160208.

26. Heilborn JD, Nilsson MF, Kratz G, Sørensen O, Borregaard N, Ståhle-Bäckdahl M. The cathelicidin anti-microbial peptide LL-37 is involved in re-epithelialization of human skin wounds and is lacking in chronic ulcer epithelium. J Investig Dermatol. 2003; 120: 379–89.

27. Hou X, Zhang X, Zhao W, Zeng C, Deng B, McComb DW, et al. Vitamin lipid nanoparticles enable adoptive macrophage transfer for the treatment of multidrug-resistant bacterial sepsis. Nat Nanotechnol. 2020; 15: 41–6.

28. Ide  T, Laarmann  S, Greune  L, Schillers  H, Oberleithner  H, Schmid  MA. Characterization of translocation pores inserted into plasma membranes by type III‐secreted Esp proteins of enteropathogenic Escherichia coli. Cellular Microbiol. 2001; 3: 669–79.

29. Iqbal U, Anwar H, Karim MA. Safety and efficacy of encapsulated fecal microbiota transplantation for recurrent Clostridium difficile infection: a systematic review. Eur J Gastroenterol Hepatol. 2018; 30: 730–4.

30. Jimenez A, Chen D, Alto NM. How bacteria subvert animal cell structure and function. Annu Rev Cell Dev Biol. 2016; 32: 373–97.

31. Kang HK, Kim C, Seo CH, Park Y. The therapeutic applications of antimicrobial peptides (AMPs): a patent review. J Microbiol. 2017; 55: 1–12.

32. Kellermayer R. Fecal microbiota transplantation: great potential with many challenges. Transl Gastroenterol Hepatol 4. 2019.

33. Kester JC, Fortune SM. Persisters and beyond: mechanisms of phenotypic drug resistance and drug tolerance in bacteria. Crit Rev Biochem Mol Biol. 2013; 49: 91–101.

34. Knoll BM, Mylonakis E. Antibacterial bioagents based on principles of bacteriophage biology: an overview. Clin Infect Dis. 2014; 58: 528–34.

35. Kortright KE, Chan BK, Koff JL, Turner PE. Phage therapy: a  renewed approach to combat antibiotic-resistant bacteria. Cell Host Mi-crobe. 2019; 25: 219–232.

36. Krachler AM, Orth K. Targeting the bacteriahost interface: strategies in anti-adhesion therapy. Virulence. 2013; 4: 284–94.

37. Kuppusamy  R, Willcox  M, Black  DSC, Kumar N. Short cationic peptidomimetic antimicrobials. Antibiotics. 2019; 8: 44.

38. Kurosaka  K, Chen  Q, Yarovinsky  F, Oppenheim  JJ, Yang  D. Mouse cathelin-related antimicrobial peptide chemoattracts leukocytes using formyl peptide receptor-like 1/mouse formyl peptide receptor-like 2 as the receptor and acts as an immune adjuvant. J Immunol. 2005; 174: 6257–65.

39. Lai  Y, Gallo  RL. AMPed up immunity: how antimicrobial peptides have multiple roles in immune defense. Trends Immunol. 2009; 30: 131–41.

40. LaSarre  B, Federle  MJ. Exploiting quorum sensing to confuse bacterial pathogens. Microbiol Mol Biol Rev. 2013; 77: 73–111.

41. Leadbetter  JR, Greenberg  EP. Metabolism of acyl-homoserine lactone quorum-sensing signals by Variovorax paradoxus. J  Bacteriol. 2000; 182: 6921–6.

42. Lewis JS, Owens A, Cadena J, Sabol K, Patterson JE, Jorgense JH. Emergence of daptomycin resistance in Enterococcus faecium during daptomycin therapy. Antimicrob Agents Chemother. 2005; 49: 1664–5.

43. Lin J, Zhou D, Steitz TA, Polikanov YS, Gagnon  MG. Ribosome-targeting antibiotics: modes of action, mechanisms of resistance, and implications for drug design. Annu Rev Biochem. 2018; 87: 451–78.

44. Liu F, Ma S. Recent process in the inhibitors of UDP-3-O-(R-3-hydroxyacyl)-nacetylglucosamine deacetylase (LpxC) against gramnegative bacteria. Mini-Rev Med Chem. 2018; 18: 310–23.

45. López EL, Contrini MM, Falaschi A. A phase II study of chimeric monoclonal antibodies to Shiga toxins 1 and 2 (Shigamabs) administered concomitantly to children with Shiga toxin–producing Escherichia coli infection and bloody diarrhea (SHIGATEC trial). VTEC Meeting, Boston, MA. 2015.

46. Lowy I, Molrine DC, Leav BA, Blair BM, Baxter R, Gerding DN, et al. Treatment with monoclonal antibodies against Clostridium difficile toxins. New Engl J Med. 2010; 362: 197–205.

47. Ma C, Yang X, Lewis PJ. Bacterial transcription as a target for antibacterial drug development. Microbiol Mol Biol Rev. 2016; 80: 139–60. https://doi.org/10.1128/MMBR.00055-15

48. Mazmanian SK, Ton‐That H, Schneewind O. Sortase‐catalysed anchoring of surface proteins to the cell wall of Staphylococcus aureus. Mol Microbiol. 2001; 40: 1049–57.

49. McCallin  S, Sacher  JC, Zheng  J, Chan  BK. Current state of compassionate phage therapy. Viruses. 2019; 11: 343.

50. McKessar SJ, Berry AM, Bell JM, Turnidge JD, Paton  JC. Genetic characterization of vanG, a novel vancomycin resistance locus of Enterococcus faecalis. Antimicrob Agents Chemother. 2000; 44: 3224–8.

51. Miró-Canturri A, Ayerbe-Algaba R, Smani Y. Drug repurposing for the treatment of bacterial and fungal infections. Frontiers Microbiol. 2019; 10: 41.

52. Molchanova  N, Hansen  PR, Franzyk  H. Advances in development of antimicrobial peptidomimetics as potential drugs. Molecules. 2017; 22: 1430.

53. Monserrat-Martinez A, Gambin Y, Sierecki E. Thinking outside the  bug: molecular targets and strategies to overcome antibiotic resistance. Int J Mol Sci. 2019; 20: 1255.

54. Moravej H, Moravej Z, Yazdanparast M, Heiat M, Mirhosseini A, Moghaddam MM, et al. Antimicrobial peptides: features, action, and their resistance mechanisms in bacteria. Microbial Drug Resist. 2018; 24: 747–767.

55. Morgan DJ, Okeke IN, Laxminarayan R, Perencevich  EN, Weisenberget  S. Non-prescription antimicrobial use worldwide: a systematic review. Lancet Infect Dis. 2011; 11: 692–701.

56. Morozova VV, Vlassov VV, Tikunova NV. Applications of bacteriophages in the treatment of localized infections in humans. Frontiers Microbiol. 2018; 9: 1696.

57. Motley MP, Fries BC. A new take on an old remedy: generating antibodies against multidrug-resistant gram-negative bacteria in a postantibiotic world. MSphere. 2017; 2: 5.

58. Murray BE. Vancomycin-resistant enterococcal infections. New England J Med. 2000; 342: 710–21.

59. Ng WL, Bassler BL. Bacterial quorum-sensing network architectures. Annu Rev Genet. 2009; 43: 197–222.

60. Niyonsaba  F, Iwabuchi  K, Matsuda  H, Ogawa  H, Nagaoka  I. Epithelial cell‐derived human β‐defensin‐2 acts as a  chemotaxin for mast cells through a pertussis toxin‐sensitive and phospholipase C‐dependent pathway. Int Immunol. 2002; 14: 421–6.

61. Njoroge  J, Sperandio  V. Jamming bacterial communication: new approaches for the treatment of infectious diseases. EMBO Mol Med. 2009; 1: 201–10.

62. Noble WC, Virani Z, Cree RGA. Co-transfer of vancomycin and other resistance genes from Enterococcus faecalis NCTC 12201 to Staphylococcus aureus. FEMS Microbiol Lett. 1992; 93: 195–8.

63. Nobrega FL, Costa AR, Kluskens LD, Azeredo  J. Revisiting phage therapy: new applications for old resources. Trends Microbiol. 2015; 23: 185–91.

64. Okano A, Isley NA, Boger DL. Total syntheses of vancomycin-related glycopeptide antibiotics and key analogues. Chemical Rev. 2017; 117: 11952–93.

65. Oliveira H, São-José C, Azeredo J. Phage-derived peptidoglycan degrading enzymes: challenges and future prospects for in vivo therapy. Viruses. 2018; 10: 292.

66. Ozdemir  T, Fedorec  AJH, Danino  T, Barnes CP. Synthetic biology and engineered live biotherapeutics: toward increasing system complexity. Cell Syst. 2018; 7: 5–16.

67. Palumbi  SR. Humans as the  world’s greatest evolutionary force. Sci. 2001; 293: 1786–90.

68. Papenfort K, Bassler BL. Quorum sensing signal–response systems in gram-negative bacteria. Nature Rev Microbiol. 2016; 14: 576.

69. Pearson JP, Gray KM, Passador L, Tucker KD, Eberhard  A, Iglewski  BH, Greenberg  EP. Structure of the autoinducer required for expression of Pseudomonas aeruginosa virulence genes. Proc Natl Acad Sci. 1994; 91: 197–201.

70. Perkins HR. Vancomycin and related antibiotics. Pharmacol Ther. 1982; 16: 181–97.

71. Pinchuk IV, Beswick EJ, Reyes VE. Staphylococcal enterotoxins. Toxins. 2010; 2: 2177–97.

72. Priebe  GP, Goldberg  JB. Vaccines for Pseudomonas aeruginosa: a long and winding road. Expert Rev Vaccines. 2014; 13: 507–19.

73. Ramachandran G, Bikard D. Editing the microbiome the CRISPR way. Philos Trans R Soc Lond B Biol Sci. 2019; 374: 20180103.

74. Reddy KVR, Yedery RD, Aranha C. Antimicrobial peptides: premises and promises. Int J Antimicrob Agents. 2004; 24: 536–47.

75. Redi D, Raffaelli CS, Rossetti B, Luca AD, Montagnani  F. Staphylococcus aureus vaccine preclinical and clinical development: current state of the art. New Microbiol. 2018; 41: 208–13.

76. Rello J, Parisella FR, Perez A. Alternatives to antibiotics in an era of difficult-to-treat resistance: new insights. Expert Rev Clin Pharmacol. 2019; 12: 635–42.

77. Ribeiro  SM, Felício  MR, Boas  EV, Goncalves S, Costa FF, Samy RP, Santos NC, Franco  OL. New frontiers for anti-biofilm drug development. Pharmacol Therapeut. 2016; 160: 133–44.

78. Rohde C, Wittmann J, Kutter E. Bacteriophages: a  therapy concept against multi-drug-resistant bacteria. Surg Infect. 2018; 19: 737–44.

79. Romaniuk JAH, Cegelski L. Bacterial cell wall composition and the  influence of antibiotics by cell-wall and whole-cell NMR. Philos Trans Royal Soc B: Biol Sci. 2015; 370: 20150024.

80. Rossiter SE, Fletcher MH, Wuest WM. Natural products as platforms to overcome antibiotic resistance. Chem Rev. 2017; 117: 12415–74.

81. Shaw RK, Daniell S, Ebel F, Frankel G, Knutton S. EspA filament‐mediated protein translocation into red blood cells. Cellular Microbiol. 2001; 3: 213–22.

82. Silver LL. Multi-targeting by monotherapeutic antibacterials. Nature Rev Drug Discov. 2007; 6: 41–55.

83. Silver LL. Appropriate targets for antibacterial drugs. Cold Spring Harb Perspect Med. 2016; 6: a030239.

84. Swietnicki W, Carmany D, Retford M, Guelta M, Dorsey R, Bozue J, Lee MS, Olson MA. Identification of small-molecule inhibitors of Yersinia pestis Type III secretion system YscN ATPase. PLoS One. 2011; 6: e19716.

84. Tenson T, Mankin A. Antibiotics and the ribosome. Mol Microbiol. 2006; 59: 1664–77.

86. Territo  MC, Ganz  T, Selsted  ME, Lehrer  R. Monocyte-chemotactic activity of defensins from human neutrophils. J Clin Investig. 1989; 84: 2017–20.

87. Theuretzbacher  U, Piddock  LJV. Non-traditional antibacterial therapeutic options and challenges. Cell Host Microbe. 2019; 26: 61–72.

88. Tokumaru S, Sayama K, Shirakata Y, Komatsuzawa H, Ouhara K, Hanakawa Y, et al. Induction of keratinocyte migration via transactivation of the epidermal growth factor receptor by the antimicrobial peptide LL-37. J Immunol. 2005; 175: 4662–8.

89. Torres  MDT, Sothiselvam  S, Lu  TK, de la Fuente-Nunez C. Peptide design principles for antimicrobial applications. J Mol Biol. 2019; 431: 3547–67.

90. Thappeta  KRV, Zhao  LN, Nge  CE, Crasta  S, Leong CY, Ng V, Kanagasundaram Y, Fan H, Ng SB. In-silico identified new natural sortase a  inhibitors disrupt S.  aureus biofilm formation. Int J Mol Sci. 2020; 21: 8601.

91. Ton-That H, Schneewind O. Assembly of pili in gram-positive bacteria. Trends Microbiol. 2004; 12: 228–34.

92. Tsiodras  S, Gold  HS, Sakoulas  G, Eliopoulos GM, Wennersten C, Venkataraman L, et al. Linezolid resistance in a clinical isolate of Staphylococcus aureus. Lancet. 2001; 358: 207–8.

93. Uroz S, Chhabra SR, Camara M, Williams P, Oger P, Dessaux Y. N-Acylhomoserine lactone quorum-sensing molecules are modified and degraded by Rhodococcus erythropolis W2 by both amidolytic and novel oxidoreductase activities. Microbiol. 2005; 151: 3313–22.

94. Vaccines to tackle drug resistant infections. An evaluation of R&D opportunities. 2019. https://vaccinesforamr.org. Accessed 12 Apr 2021

95. Vasil  ML. DNA microarrays in analysis of quorum sensing: strengths and limitations. J Bacteriol. 2003; 185: 2061–5.

96. Waters CM, Bassler BL. Quorum sensing: cellto-cell communication in bacteria. Annu Rev Cell Dev Biol. 2005; 21: 319–46.

97. Whitehead NA, Barnard AML, Slater H, Simpson NJL, Salmond GPC. Quorum-sensing in gram-negative bacteria. FEMS Microbiol Rev. 2001; 25: 365–404.

98. Wilcox MH, Gerding DN, Poxton IR, Kelly C, Nathan  R, Birch  T,  et  al. Bezlotoxumab for prevention of recurrent Clostridium difficile infection. N Engl J Med. 2017; 376(4): 305–17.

99. Wilson  DN. Ribosome-targeting antibiotics and mechanisms of bacterial resistance. Nat Rev Microbiol. 2014; 12: 35–48.

100. Wright  GD. Opportunities for natural products in 21st century antibiotic discovery. Nat Prod Rep. 2017; 34: 694–701.

101. Xie J, Pierce JG, James RC, Okano A, Boger DL. A redesigned vancomycin engineered for dual D-Ala-D-ala and D-Ala-D-Lac binding exhibits potent antimicrobial activity against vancomycin-resistant bacteria. J American Chemical Soc. 2011; 133: 13946–9.

102. Yang D, Chen Q, Schmidt AP, Anderson GM, Wang  JM, Wooters  J, Oppenheim  JJ, Chertov  O. LL-37, the  neutrophil granuleand epithelial cellderived cathelicidin, utilizes formyl peptide receptor–like 1 (FPRL1) as a receptor to chemoattract human peripheral blood neutrophils, monocytes, and T cells. J Exp Med. 2000; 192: 1069–74.

103. Yang  D, Biragyn  A, Kwak  LW, Oppenheim JJ. Mammalian defensins in immunity: more than just microbicidal. Trends Immunol. 2002; 23: 291–6.

104. Zasloff M. Antimicrobial peptides of multicellular organisms. Nature. 2002; 415: 389–95.

105. Zhang J, Liu H, Zhu K, Yang CG. Antiinfective therapy with a small molecule inhibitor of Staphylococcus aureus sortase. Proc Natl Acad Sci. 2014; 111: 13517–22.


*  Corresponding author. Email: volodymyr.vasylenko@vdu.lt

Tetiana S. Todosiichuk, Serhii O. Soloviov, Lin Wu, Iryna V. Dzyublyk, Olena P. Trokhimenko, Magdalena Dudek, Artem Symchuk, Volodymyr Vasylenko

ŠIUOLAIKINIŲ IR PERSPEKTYVIŲ ANTIMIKROBINIŲ MEDŽIAGŲ KŪRIMO KRYPTYS

Santrauka

Antibiotikų vartojimas vis labiau ribojamas dėl išsivystančio atsparumo patogeninėms bakterijoms. Laikui bėgant vis daugiau bakterijų taps atsparios antimikrobinėms medžiagoms, todėl būtina ištirti alternatyvius kovos su patogenais metodus. Tai padėtų sukurti pakankamai patikimą ir veiksmingą infekcinių ligų terapiją. Šioje apžvalgoje aptariami kai kurie naujausi gydymo įprastiniais antibiotikais ir neantibiotikais strategijų pokyčiai. Įrodyta, kad tradiciniai antibakteriniai taikiniai apima žinomų antibiotikų klasių darinius, naujas chemines klases su naujais taikiniais, taip pat nežinomus ar neapibrėžtus agentus. Buvo nustatytos daug žadančios kovos su mikrobų patogenais strategijos, įskaitant naujus taikinius, būtent – toksinų sekrecijos sistemas, bioplėvelės formavimąsi ir sukibimo mechanizmus, turinčius įtakos Quorum Sensing mikrobų populiacijoms. Be to, svarbu naujas antimikrobines medžiagas naudoti su kitais, ne antibiotiniais, veikimo mechanizmais – fagais ir fagų kilmės peptidais, mikrobiotą moduliuojančias terapijas, stiprinančias imuninį atsaką.

Raktažodžiai: įprastiniai antibiotikų taikiniai, neantibiotikų taikiniai, gydymas neantibiotikais, infekcijos, pasipriešinimas