Elsevier

Biochemical Pharmacology

Volume 133, 1 June 2017, Pages 43-62
Biochemical Pharmacology

Review
Antibiotics: Pharmacokinetics, toxicity, resistance and multidrug efflux pumps

https://doi.org/10.1016/j.bcp.2016.10.005Get rights and content

Abstract

The discovery of penicillin followed by streptomycin, tetracycline, cephalosporins and other natural, semi-synthetic and synthetic antimicrobials completely revolutionized medicine by reducing human morbidity and mortality from most of the common infections. However, shortly after they were introduced to clinical practice, the development of resistance was emerged. The decreasing interest from antibiotic industry in spite of rapid global emergence of antibiotic resistance is a tough dilemma from the pointview of public health. The efficiency of antimicrobial treatment is determined by both pharmacokinetics and pharmacodynamics. In spite of their selective toxicity, antibiotics still cause severe, life-threatening adverse reactions in host body mostly due to defective drug metabolism or excessive dosing regimen. The present article aims at updating current knowledge on pharmacokinetics/pharmacodynamics concepts and models, toxicity of antibiotics as well as antibiotic resistance mechanisms, resistome analyses and search for novel antibiotic resistance determinants with special emphasis given to the-state-of-the-art regarding multidrug efflux pumps and their additional physiological functions in stress adaptation and virulence of bacteria. All these issues are highly linked to each other and not only important for most efficient and prolonged use of current antibiotics, but also for discovery and development of new antibiotics and novel inhibitors of antibiotic resistance determinants of pathogens.

Introduction

The identification of penicillin by Alexander Fleming in 1928 [1] is the cornerstone discovery in the history of the ‘antibiotic era’. Still, the efforts of Paul Ehrlich and his co-workers to establish a systematic screening approach for discovery of antimicrobials took place much earlier and introduced the ‘magic bullet’ Salvarsan [2], [3]. Salvarsan remained the most populously prescribed drug against syphilis till the first use of penicillin in 1941 [3], [4]. Prontosil as the first sulfa drug was next discovered by the use of same approach [5] and followed by discovery of streptomycin which was the first antibiotic used for the treatment of tuberculosis [6]. In 1945, the fungus Cephalosporium acremonium was shown to produce an “antibiotic principle” effective against staphylococcal, streptococcal infections, typhoid fever and brucellosis [7]. Later on, the principle was demonstrated to represent a group of natural compounds called cephalosporins, and N-phenylacetyl derivative of cephalosporin C being the most effective against Staphylococcus aureus [8], [9]. These investigations led to the production of new generation cephalosporin compounds and saved many lives as the former ones. However, resistance has eventually appeared for nearly all antibiotics, shortly after they were introduced to clinical practice [10].

According to the estimates of Bérdy (2012) as based on Bioactive MICrobial Metabolite Database of his own, of 60–80 thousand natural metabolites produced by microbes, 47% exhibit bioactivity [11]. On the other hand, when it comes to the total number of drugs in market for use in human therapy, there are ca. 3500 such compounds 200–220 of which include antibiotics made up of direct natural products, more than 250 being semisynthetic/modified derivatives of them, and synthetic antimicrobials (especially quinolones and oxazolidinones) do also have a role in the antimicrobial market. As headed by the problem of increasing resistance to antibiotics creating clinical and economic burden, the search for novel antibiotics (from nature, combinational biosynthesis, hybrid antibiotics, discovery of new molecular targets, screening of unculturable microorganisms, combinatorial chemistry as well as computerized drug design) should constitute a very hot research area for finding new antibacterial drugs. Indeed, in between late 1960s and mid 1980s, the pharmaceutical industry introduced many new antibiotics to solve resistance problem, but since then there appears only a very limited number of new antibiotics reported. To exemplify, some new antibiotics including synthetic (e.g. besifloxacin, doripenem, radezolid), and semi-synthetic (e.g. cethromycin; derived from erythromycin A) compounds were recently approved by FDA or in clinical trials [12]. In a recent study, Ling and his co-workers (2015) announced the discovery of a new class of natural antibiotic namely teixobactin which inhibits cell wall synthesis by a novel mechanism after the screening of a previously uncultured bacterium namely Eleftheria terrae [13]. Another research group suggested a new antimicrobial agent called lugdunin produced by Staphylococcus lugdunensis which is an inhabitant of human nares. They demonstrated the potential killing effect of lugdunin against many Gram-positive bacteria including methicillin-resistant S. aureus (MRSA) and vancomycin-resistant Enterococcus (VRE) isolates [14]. Also, a team at Harvard University proposed a new platform for the production of new macrolides which is based on the synthesis of different compounds that cannot be produced by traditional semi-synthetic methods [15]. The novel targets in different bacterial processes such as quorum sensing (QS) and biofilm formation are being investigated for development of new antibacterial agents mainly by academia [16]. The Infectious Diseases Society of America (IDSA) provided a platform called ‘the 10 × 20 initiative’ for development of ten new antibacterial drugs including new chemical classes and modified versions of current classes till 2020 [17]. ‘Return On Investment’ considerations and challenging regulatory requirements unfortunately restricted the attempts of antibiotic industry to discover and develop novel classes of agents against pathogenic bacteria which instead preferred to focus on altering existing ones or development of antiviral agents [18], [19]. Thus, the decreasing interest from antibiotic industry in spite of worldwide rapid emergence of antibiotic resistance is a tough dilemma from the pointview of public health. Maintaining and prolonging the useful life span of existing antibiotics must have a high priority under these circumstances [20]. In this respect, computer-aided screening to identify potential inhibitors of the antibiotic resistance and also preferentially quorum sensing and virulence has received great attention in recent years [21].

Pharmacokinetic properties of antibiotics are mainly based on their chemical structure which absolutely affects their bioavailability, half-life, tissue penetration, distribution, degradation and elimination [22]. For each class of antibiotics, dosage application and duration of exposure have been critical issues to obtain optimum outcomes in patients while minimizing the risk of resistance development and toxicity. Expanding knowledge on the interaction between antibiotic pharmacokinetics, toxicity and resistance provided better understanding of individualized therapy [22], [23]. Pharmacodynamic factors include antimicrobial activity against the pathogen, drug stability in the case of resistance and absence of organ toxicity [24]. The present article aims at overviewing pharmacokinetics/pharmacodynamics concepts and models, toxicity of antibiotics as well as antibiotic resistance mechanisms with special emphasis to multi-drug transporters, all of which are highly linked to most efficient and prolonged use of antibiotics.

Section snippets

Pharmacokinetics/pharmacodynamics concepts and models

Since the emergence of antibiotic resistance is mostly attributed to drug overuse, inappropriate prescribing and suboptimal dosing, certain measures must be taken for dose optimization of current antibiotics [25]. Optimization of antibiotic usage requires well-understood criteria that can be simplified as the relationships between concentration, dose and both desirable and side effects. This requirement has emerged a well established and authorities-recognized field called

General outlook to antibiotic-resistant pathogens, mechanisms of resistance and transfer

As declared and warned by many authorities, we are now in the “post-antibiotic era,” exposed to a global “antibiotic resistance (AR) crisis” [10], [160]. With respect to their clinical impact, economic impact, incidence, 10-year projection of incidence, transmissibility, availability of effective antibiotics, and barriers to prevention, the Centers for Disease Control and Prevention classified the pathogens concerning the degree of threat (Table 1) [161].

Nosocomial infections caused by AR

Conclusion

Antibiotics have been savior of humankind since their first discovery but increasing AR which is a natural ecological phenomenon of billions of years evolution and a result of escape of resistance genes from environmental ‘resistome’ to human pathogens created a fearful and alarming situation in recent years. Moreover, individualized therapy became the forefront of providing most proper use of antibiotics. Knowledge about evolution and mechanisms of resistance can pave way to reduce resistance

Disclosures

There are no conflicts of interest.

Acknowledgement

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

References (320)

  • P. Kontou et al.

    Pharmacokinetics of moxifloxacin and high-dose levofloxacin in severe lower respiratory tract infections

    Int. J. Antimicrob. Agents

    (2013)
  • S. Tafelski et al.

    Observational clinical study on the effects of different dosing regimens on vancomycin target levels in critically ill patients: continuous versus intermittent application

    J. Infect. Public Health

    (2015)
  • E. Ampe et al.

    Implementation of a protocol for administration of vancomycin by continuous infusion: pharmacokinetic, pharmacodynamic and toxicological aspects

    Int. J. Antimicrob Agents

    (2013)
  • C. Scheerans et al.

    Concentration–response studies and modelling of the pharmacodynamics of linezolid: Staphylococcus aureus versus Enterococcus faecium

    Int. J. Antimicrob. Agents

    (2015)
  • F. Van Bambeke et al.

    Macrolides: pharmacokinetics and pharmacodynamics

    Int. J. Antimicrob. Agents

    (2001)
  • J. Li et al.

    Colistin: the re-emerging antibiotic for multidrug-resistant Gram-negative bacterial infections

    Lancet Infect. Dis.

    (2006)
  • A. Dhand et al.

    Daptomycin in combination with other antibiotics for the treatment of complicated methicillin-resistant Staphylococcus aureus bacteremia

    Clin. Ther.

    (2014)
  • A. Fleming

    On the antibacterial action of cultures of a penicillium, with special reference to their use in the isolation of B. influenzæ

    Br. J. Exp. Pathol.

    (1929)
  • P. Ehrlich et al.

    Die Experimentelle Chemotherapie der Spirilosen

    (1910)
  • R.I. Aminov

    A brief history of the antibiotic era: lessons learned and challenges for the future

    Front. Microbiol.

    (2010)
  • G. Domagk

    Ein Beitrag zur Chemotherapie der bakteriellen Infektionen

    Dtsch. Med. Wochenschr.

    (1935)
  • A. Schatz et al.

    Effect of streptomycin and other antibiotic substances upon Mycobacterium tuberculosis and related organisms

    Exp. Biol. Med. Nov.

    (1944)
  • G. Brotzu

    Richerche su di un nuovo antibiotico

    Lavori Istituto Igiene Cagliari

    (1948)
  • E.P. Abraham et al.

    The structure of cephalosporin C

    Biochem. J.

    (1961)
  • C.L. Ventola

    The antibiotic resistance crisis. Part 1: causes and threats

    P.T.

    (2015)
  • J. Bérdy

    Thoughts and facts about antibiotics: where we are now and where we are heading

    J. Antibiot.

    (2012)
  • M. Bassetti et al.

    New antibiotics for bad bugs: where are we?

    Ann. Clin. Microbiol. Antimicrob.

    (2013)
  • L.L. Ling et al.

    A new antibiotic kills pathogens without detectable resistance

    Nature

    (2015)
  • A. Zipperer et al.

    Human commensals producing a novel antibiotic impair pathogen colonization

    Nature

    (2016)
  • I.B. Seiple et al.

    A platform for the discovery of new macrolide antibiotics

    Nature

    (2016)
  • N. Rabin et al.

    Agents that inhibit bacterial biofilm formation

    Future Med. Chem.

    (2015)
  • Infectious Diseases Society of America, The 10 x ′20 Initiative: pursuing a global commitment to develop 10 new...
  • L.L. Silver

    Challenges of antibacterial discovery

    Clin. Microbiol. Rev.

    (2011)
  • T. Fowler et al.

    The risk/benefit of predicting a post-antibiotic era: is the alarm working?

    Ann. N. Y. Acad. Sci.

    (2014)
  • G.F. Ahmed et al.

    Application of pharmacokinetics/pharmacodynamics (PK/PD) in designing effective antibiotic treatment regimens

  • P.A. Smith et al.

    Combating bacteria and drug resistance by inhibiting mechanisms of persistence and adaptation

    Nat. Chem. Biol.

    (2007)
  • P.S. McKinnon et al.

    Pharmacokinetic and pharmacodynamic issues in the treatment of bacterial infectious diseases

    Eur. J. Clin. Microbiol. Infect. Dis.

    (2004)
  • L. Baietto et al.

    A 30-years review on pharmacokinetics of antibiotics: is the right time for pharmacogenetics?

    Curr. Drug. Metab.

    (2014)
  • Y. Valcke et al.

    Pharmacokinetics of antibiotics in the lungs

    Eur. Respir. J.

    (1990)
  • S.K.B. Sy et al.

    Pharmacokinetics and pharmacodynamics in antibiotic dose optimization

    Expert Opin. Drug Metab. Toxicol.

    (2016)
  • H. Eagle et al.

    The effective concentrations of penicillin in vitro and in vivo for streptococci, pneumococci, and Treponema pallidum

    J. Bacteriol.

    (1950)
  • W.A. Craig

    Pharmacokinetic/pharmacodynamic parameters: rationale for antibacterial dosing of mice and men

    Clin. Infect. Dis.

    (1998)
  • G.L. Drusano

    Antimicrobial pharmacodynamics: critical interactions of ’bug and drug’

    Nat. Rev. Microbiol.

    (2004)
  • E.I. Nielsen et al.

    Pharmacokinetic-pharmacodynamic modeling of antibacterial drugs

    Pharmacol. Rev.

    (2013)
  • N. Petrosillo et al.

    Treatment of carbapenem-resistant Klebsiella pneumoniae: the state of the art

    Expert. Rev. Anti Infect. Ther.

    (2013)
  • F. Van Bambeke et al.

    Cellular pharmacodynamics and pharmacokinetics of antibiotics: current views and perspectives

    Curr. Opin. Drug Discov. Devel.

    (2006)
  • E.I. Nielsen et al.

    Pharmacokinetic/pharmacodynamic (PK/PD) indices of antibiotics predicted by a semimechanistic PKPD model: a step toward model-based dose optimization

    Antimicrob. Agents Chemother.

    (2011)
  • T.H. Steinberg

    Cellular transport of drugs

    Clin. Infect. Dis.

    (1994)
  • F. Van Bambeke et al.

    Antibiotic efflux pumps in eukaryotic cells: occurrence and impact on antibiotic cellular pharmacokinetics, pharmacodynamics and toxicodynamics

    J. Antimicrob. Chemother.

    (2003)
  • P.M. Tulkens

    Intracellular distribution and activity of antibiotics

    Eur. J. Clin. Microbiol. Infect. Dis.

    (1991)
  • Cited by (108)

    • Biopharmaceutical Classification System: a strategic tool in pharmaceutical formulation

      2023, Dosage Forms, Formulation Developments and Regulations: Recent and Future Trends in Pharmaceutics, Volume 1
    • Antibiotics, antibiotic-resistant bacteria, and the environment

      2023, Degradation of Antibiotics and Antibiotic-Resistant Bacteria From Various Sources
    View all citing articles on Scopus
    View full text