You are on page 1of 16

Biochemical Pharmacology 133 (2017) 4–19

Contents lists available at ScienceDirect

Biochemical Pharmacology
journal homepage: www.elsevier.com/locate/biochempharm

Review

History of antimicrobial drug discovery: Major classes and health impact


Rustam Aminov
School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZD, United Kingdom

a r t i c l e i n f o a b s t r a c t

Article history: The introduction of antibiotics into clinical practice revolutionized the treatment and management of
Received 25 August 2016 infectious diseases. Before the introduction of antibiotics, these diseases were the leading cause of mor-
Accepted 4 October 2016 bidity and mortality in human populations. This review presents a brief history of discovery of the main
Available online 5 October 2016
antimicrobial classes (arsphenamines, b-lactams, sulphonamides, polypeptides, aminoglycosides, tetra-
cyclines, amphenicols, lipopeptides, macrolides, oxazolidinones, glycopeptides, streptogramins, ansamy-
Keywords: cins, quinolones, and lincosamides) that have changed the landscape of contemporary medicine. Given
Antimicrobial drug discovery
within a historical timeline context, the review discusses how the introduction of certain antimicrobial
History
classes affected the morbidity and mortality rates due to bacterial infectious diseases in human popula-
tions. Problems of resistance to antibiotics of different classes are also extensively discussed.
Ó 2016 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4


2. Arsphenamines and the foundation of modern antimicrobial chemotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3. b-Lactams . . . . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
4. Sulphonamides . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
5. Polypeptides . . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
6. Aminoglycosides. . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
7. Tetracyclines. . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
8. Amphenicols . . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
9. Lipopeptides . . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
10. Macrolides . . . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
11. Oxazolidinones . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
12. Glycopeptides . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
13. Streptogramins . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
14. Ansamycins. . . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
15. Quinolones . . . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
16. Lincosamides . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
17. Concluding remarks . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Conflict of interest statement . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
Author contribution statement . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
References . . . . . . . . . . . . . . . . . . . . . . . ....... ........................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14

1. Introduction when the annual mortality rate due to infectious diseases was
rapidly decreasing, by 8.2% per year [16]. Infectious diseases
The highest rate of decline in infectious disease mortality in the that mostly contributed to this sharp decline were pneumonia,
USA was recorded for a period of 15 years, from 1938 to 1952, influenza, and tuberculosis. These declines corresponded to the
introduction into clinical practice of sulphonamides in 1935, peni-
cillin in 1941, and streptomycin in 1943, with a number of other
E-mail address: rustam.aminov@abdn.ac.uk

http://dx.doi.org/10.1016/j.bcp.2016.10.001
0006-2952/Ó 2016 Elsevier Inc. All rights reserved.
R. Aminov / Biochemical Pharmacology 133 (2017) 4–19 5

combination drugs, such as para-aminosalicylic acid in 1944 and the drug, marketed by Hoechst under the trade name Salvarsan,
isoniazid in 1952, introduced for tuberculosis treatment in addi- was a great success and, together with a more soluble and less
tion to streptomycin [21]. This correlation clearly indicates the toxic Neosalvarsan, enjoyed the status of the most frequently pre-
importance of antimicrobials in the control of infectious diseases. scribed drug until its replacement by penicillin in the 1940s [148].
A recent statistics also reflects our success in dealing with infec- Remarkably, the mode of action of this hundred-year-old drug is
tious diseases that now cause much less mortality compared to still unknown, and the controversy about its chemical structure
many other diseases of a non-infectious nature. In the most recent was solved only in 2005 [142]. Other derivatives of the lead com-
National Vital Statistics Reports, among the 15 leading causes of pound, arsanilic acid, however, have been in a much more pro-
death in the USA, infectious diseases, such as influenza and pneu- longed use as feed additives in poultry and swine. The U.S. Food
monia, are superseded by heart disease, cancer, chronic lower res- and Drug Administration (FDA) announced the complete with-
piratory diseases, accidents, stroke, Alzheimer’s disease, and drawal of arsenic-based drugs for use in food-producing animals
diabetes [245]. The foundation for this success in confronting death only by the end of 2015 [83].
from infectious diseases was built by formidable scientists, who Presently, syphilis infections are successfully managed by peni-
made important antimicrobial drug discoveries and are greatly cillin drugs, in particular by intramuscular injection of benzathine
acknowledged for saving numerous lives. benzylpenicillin, which allows reaching a prolonged antibiotic
exposure over a two- to four-week period after a single dose deliv-
ery. Patients with severe allergy to penicillin can be treated with
2. Arsphenamines and the foundation of modern antimicrobial tetracycline or doxycycline. The availability of very efficient thera-
chemotherapy pies resulted in a substantial drop of mortality due to syphilis, from
202,000 in 1990 to 113,000 in 2010 [143]. Still, the number of new
Paul Ehrlich’s idea of a ‘‘magic bullet”, which is highly selective infections remains relatively high, with 315,000 cases reported in
and targets only the disease-causing microorganisms, came to him 2013 [61].
while he was working with an extensive range of aniline and other
synthetic dyes that became available as a result of the rapidly
developing German chemical industry. He noticed that some stains 3. b-Lactams
could be specific for certain microbes but not to others. Ehrlich rea-
soned that chemical compounds could be synthesized in a way that Discovered serendipitously in 1928 by Alexander Fleming [86],
it would be possible ‘‘to exert their full action exclusively on the penicillin did not immediately take off as a clinically useful antibi-
parasite harboured within the organism” (http://pubs.acs. otic. This was hindered by many drawbacks, such as low yield,
org/cen/coverstory/83/8325/8325salvarsan.html). Based on this instability, purification and other problems. In fact, military actions
idea, in 1904, he initiated a large-scale and systematic screening in the 1940s helped to develop it into a valuable treatment of infec-
program for a drug active against syphilis, the disease that had tions, with a considerable production for the treatment of sick and
grown to the epidemic levels in the USA and Europe and was wounded soldiers in the U.S. and Allies’ military forces. Thereafter,
hardly curable at the time. The mainstream treatment for this penicillin became a widely used antibiotic for a broad range of pre-
sexually transmitted disease, which is caused by the spirochete viously untreatable infectious diseases, with a wider range of tar-
Treponema pallidium, involved administration of mercury chloride gets and fewer side effects than sulpha drugs (see the next section).
along with other inorganic mercury salts. Due to the extreme Although the antibacterial properties of mould had been known
toxicity of mercury compounds, the treatment had severe side from ancient times, and researchers before him had come upon
effects and, yet, poor efficacy. Another type of treatment included similar observations regarding the antimicrobial activity of Penicil-
arsenic and inorganic arsenical compounds, but the toxicity and lium fungi from time to time (e.g., Vincenzo Tiberio, see [46]), it
low efficiency remained an issue with this treatment as well. was Alexander Fleming’s firm faith in the idea and his impressive
A less toxic organic arsenical drug, named Atoxyl, was synthe- determination that made the difference. For more than a decade
sized by Antoine Béchamp in 1859 [47,214], initially for the treat- after his initial observation, he tried hard to involve chemists in
ment of African sleeping sickness. This drug attracted the attention resolving the problems of purification and stability of the active
of Paul Ehrlich and Alfred Bertheim, an organic chemist working compound, supplying the producer strain to every request. And
with him. They correctly identified the chemical structure of this finally, in 1940, an Oxford team, led by Howard Florey and Ernest
compound as aminophenyl arsenic acid, thus opening the possibil- Chain, published a paper describing the purification procedure for
ity of synthetizing various derivatives in the search for a more effi- penicillin in quantities sufficient for clinical testing [53]. The fol-
cient and less toxic therapeutic agent. They synthesized hundreds lowing refinements and optimizations of the original procedure,
of arsenobenzene compounds, and the arsphenamine derivative, isolation of more efficient penicillin producer strains, and opti-
the sixth compound in the 600th series (i.e. compound 606), was mization of the strain fermentation procedure eventually led to
synthesized in 1907. Although initially aimed at the treatment of the mass production and distribution of penicillin in 1945 [171].
African sleeping sickness, the drug appeared to be ineffective at The screening procedures in the discovery of Salvarsan and
it, but, in 1909, Ehrlich and Bertheim, together with bacteriologist Prontosil required testing of many compounds using the animal
Sahachiro Hata, established the efficiency of this compound in the models of human disease. The screening method of Alexander
treatment of syphilis-infected rabbits. In subsequent limited trials Fleming used inhibition zones in lawns of pathogenic bacteria on
in humans, the drug demonstrated significant capacity for the the surface of agar-medium plates and, thus, required much less
treatment of patients with this venereal disease [72]. This process time and resources. At least in the initial stages of screening, before
of systematic synthesis and activity check is considered to be the testing in animal disease models, the approach made it possible to
beginning of the modern chemotherapeutic era (Stefan and reasonably inexpensively test a much larger range of compounds
[213]. Almost all further developments in modern pharmaceutical with a potential antimicrobial activity. This method became widely
research followed this route, with systematic chemical modifica- used in mass screenings for antibiotic-producing microorganisms
tions of a lead compound to improve its biological activity and les- by many researchers in academia and industry during the antibi-
sen the side effects. otic discovery programmes.
Despite the problems associated with its stability and storage, Identification of 6-aminopenicillanic acid as the core of peni-
as well as a rather tedious injection procedure and side effects, cillin by scientists of Beecham Research Laboratories in the UK
6 R. Aminov / Biochemical Pharmacology 133 (2017) 4–19

[30] allowed the synthesis and production of numerous semisyn- suggested that ceftriaxone, which increases the expression of
thetic penicillins. The main developments included the glutamate transporter, displays potent neuroprotective and
penicillinase-resistant penicillins, such as methicillin, oxacillin, immunomodulatory effects, and also could be a valuable candidate
and nafcillin; followed by the derivatives active against Gram- for the treatment of alcohol and other drug dependencies due to its
negative bacteria: the aminopenicillins (ampicillin, amoxicillin, capability of normalizing glutamate transmission, which is affected
and bacampicillin), the carboxypenicillins (carbenicillin and ticar- in addiction [10]. Thus, the use of b-lactams that have been already
cillin), and the ureidopenicillins (mezlocillin, azlocillin, and piper- approved for the treatment of infectious diseases could be extended
acillin) [242]. Further developments to overcome resistance and to the therapy of other, non-infectious, diseases and conditions.
extend the range of targeted organisms led to the combination of
a b-lactamase inhibitor (clavulanic acid, sulbactam, or tazobactam)
and an aminopenicillin, ticarcillin, or piperacillin [242]. Although 4. Sulphonamides
unified under the umbrella of the b-lactam structure, this group
of antibiotics is sometimes separated into the penicillin, cephalos- The systematic screening approach suggested by Paul Ehrlich
porin, and carbapenem classes. These are the most frequently pre- was then followed by others in the search for chemotherapy of
scribed broad-spectrum antibiotics in outpatient care [136,202], other infectious diseases. In particular, the first class of antimicro-
and, as such, may have contributed to a significant problem of bials that went into the truly large-scale production was the group
resistance towards b-lactams among pathogenic bacteria [39]. of sulpha drugs. In the late 1920s and early 1930s, the German
Alexander Fleming was also among the first who cautioned chemical industry was experiencing a dramatic increase in the
about the potential resistance to penicillin if used in too little dozes number of newly synthesized compounds that were available for
or for a too short period during treatment. Even before the exten- testing, especially dye compounds, due to a common assumption
sive use of penicillin, some observations suggested that bacteria at the time that dyes, which bind specifically to bacteria and para-
could destroy it by enzymatic degradation [1]. The outlook, how- sites, might then exert lethal effects on them.
ever, was optimistic, largely based on the previous experience with In the laboratories of Bayer AG, hundreds of compounds were
arsenic drugs. One of the earlier studies of possible resistance synthesized and tested before coming across the compound called
emergence under laboratory conditions concluded that: ‘‘Syphilis sulphonamidochrysoidine (KI-730, commercial name Prontosil). It
has now been treated with arsenicals for about 40 years without was synthesized by Bayer chemists Josef Klarer and Fritz Mietzsch
any indications of an increased incidence of arsenic-resistant infec- and tested by Gerhard Domagk for antibacterial activity in several
tions, and this work gives grounds for hoping that the widespread animal disease models [69]. It appeared to be particularly effective
use of penicillin will equally not result in an increasing incidence of against streptococcal infections, while less efficient against other
infections resistant to penicillin” [189]. Surprisingly, this is still cocci. Prontosil itself, however, appeared to be a precursor for the
true for syphilis infections [52], the first line of treatment for which active drug, and only the active part of it, sulphanilamide, was
is benzathine benzylpenicillin, but not for many other pathogenic actually responsible for the antibacterial activity [224]. It was not
bacteria, including the Enterobacteriaceae, which have become patentable, since sulphanilamide patent had already expired, and
resistant not only to the original penicillin but also to semi- these compounds had been in use in the dye industry for some
synthetic penicillins, cephalosporins, and newer carbapenems years. As sulphanilamide was cheap to produce and off patent,
[130]. and the sulphanilamide moiety was easy to modify, many compa-
The rise of ESKAPE pathogens (Enterococcus faecium, Staphylo- nies subsequently started mass production of sulphonamide
coccus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseu- derivatives. Some of the preparations, however, included diethy-
domonas aeruginosa, and Enterobacter spp.) [185,39] represents an lene glycol resulting in the death of more than 100 patients, which
especially worrying trend. Although the majority of these infec- enforced the introduction of a legislative framework, with a set of
tions were seemingly under control in the past, this equilibrium laws called the United States Federal Food, Drug, and Cosmetic Act,
in the arms race between humans and pathogens appeared to be in 1938 (http://www.fda.gov/RegulatoryInformation/Legislation/
fragile. During the almost four billion years of evolution the micro- FederalFoodDrugandCosmeticActFDCAct/default.htm).
bial world has accumulated an enormous diversity of metabolic A recent study has evaluated the effect of the introduction of
and protective mechanisms than can be mobilized in response to sulpha drugs, which, between 1937 and 1943, led to a 24–36%
a strong selection, including the pressure of antibiotics [6,7]. decline in maternal mortality, a 17–32% decline in pneumonia
Despite being obstructed by the growing resistance problem, the mortality, and a 52–65% decline in scarlet fever mortality [198].
b-lactam antimicrobials remain the most widely prescribed anti- Overall, sulpha drugs reduced mortality by 2–3 per cent and
infective agents due to their safety, efficacy, and availability increased life expectancy by 0.4–0.7 years. Presently, sulpha drugs
[136,202]. Strategies for the further development of this class of are mainly used to treat urinary tract infections and as a supportive
antimicrobials may include the novel broad-spectrum b- therapy in HIV/AIDS patients. Besides their antibacterial activity,
lactamase inhibitors that work against many problematic b- exerted via the competitive inhibition of the bacterial enzyme
lactamases such as cephalosporinases and carbapenemases [49]. dihydropteroate synthase, other clinical applications of sulphona-
This could also be the combination of two non-b-lactam families mides include their use as anti-diabetics, diuretics, anticonvul-
of b-lactamase inhibitors, diazabicyclooctanes and boronic acids, sants, and antiretrovirals.
with novel or existing b-lactam antibiotics [119]. Besides, the con- The legacy of this oldest mass-produced antimicrobial class in a
jugation of b-lactams to siderophores may allow drug entry, via poorly regulated market is possibly reflected in one of the most
bacterial iron transport systems, into the Gram-negative bacterial broadly disseminated cases of drug resistance, i.e. sulpha drug
pathogens that are intrinsically resistant due to restricted porin resistance, which is almost universally linked with class 1 inte-
entry and drug efflux [119]. grons. Mobile genetic elements (MGEs) have been instrumental
Another biological activity of b-lactams, not related to the killing in the rapid dissemination of antimicrobial resistance genes [8].
of bacteria, has been discovered during a large-scale screening of Moreover, once the sulpha drug resistance is established on an
the FDA-approved drugs for modulating the activity of the gluta- MGE, it may be difficult to eliminate, because the resulting con-
mate transporter subtype 1 [192]. Glutamate is a principal excita- struct confers a fitness advantage to the host even in the absence
tory neurotransmitter in the brain involved in memory and of antibiotic selection [75]. Despite this, many continuously modi-
learning processes [203]. A number of investigations have fied derivatives of this oldest class of synthetic antibiotics are still a
R. Aminov / Biochemical Pharmacology 133 (2017) 4–19 7

viable option for therapy, while the action of and resistance to sul- tuberculosis have dramatically fallen. In the USA, for example,
phanilamides is one of the best examples of the arms race between the mortality rate decreased more than fourfold in a decade (from
humans and microbes. 1945 to 1955), from 39.9 deaths per 100,000 population to 9.1 [96].
Until the emergence of multidrug-resistant (MDR) M. tuberculosis
strains, the prevailing outlook was extremely confident, as
5. Polypeptides
expressed by the words of the streptomycin discoverer: ‘‘the final
chapter of the battle against tuberculosis appears to be at hand”
It was independently discovered at the end of the 1930s and the
[235].
beginning of the 1940s that various strains of the soil bacterium
Despite some successes in the treatment of tuberculosis,
Brevibacillus brevis (formerly Bacillus brevis) produced substances
which resulted in the drop of TB death rate by 47% between
inhibiting a range of pathogenic bacteria and even fungi. This spe-
1990 and 2015, the situation is deteriorating. In 1991, an MDR
cies appeared to be producing a variety of linear and cyclic pep-
strain of M. tuberculosis (strain W) was isolated in the USA,
tides using nonribosomal protein synthetases [125]. Tyrothricin
which was resistant to isoniazid, rifampin, ethambutol, strepto-
isolated by René Dubos, an American microbiologist of French ori-
mycin, kanamycin, ethionamide, and rifabutin [175]. According
gin, in 1939, appeared to be a mix of cyclic and linear polypeptides
to WHO Fact Sheet N°104, 9.6 million people fell ill with TB
with antimicrobial activity. Its principal component is tyrocidine,
and 1.5 million died from the disease in 2014 (http://www.
which is also a mixture of cyclic decapeptides. The nonribosomal
who.int/mediacentre/factsheets/fs104/en/). The rise of infections
biosynthesis of tyrocidine is via an enzymatic assembly consisting
caused by MDR M. tuberculosis strains that resist the first-line
of 3 peptide synthetase proteins, TycA, TycB, and TycC, which con-
therapy with isoniazid and rifampicin poses a major problem
tain 10 modules [191]. Another cyclic polypeptide, from another
restricting the available therapeutic choices. The situation is fur-
strain of B. brevis, gramicidin S (S stands for Soviet), was reported
ther aggravated by the emergence of extensively drug-resistant
in 1944 [92]. Other gramicidins, A, B, and C, as well as the mix of
tuberculosis (XDR-TB), which, in addition, displays resistance to
these three called gramicidin D, are the linear pentadecapeptides
fluoroquinolones (such as levofloxacin or moxifloxacin) and to
[48]. The structures of gramicidins and tyrocidines were unknown
at least one of the three injectable second-line drugs (amikacin,
at the time, thus a cyclic polypeptide was called gramicidin S,
capreomycin, or kanamycin) (http://www.who.int/entity/tb/ar-
while a more appropriate name would be tyrocidine S. All grami-
eas-of-work/drug-resistant-tb/xdr-tb-faq/en/index.html).
cidins and tyrocidines belong to the group of polypeptide antibiotic
As early as in 2007, two tuberculosis cases that resisted all
compounds, which also include microcystin, bacitracin, and others.
available drugs and long-term (5–8 years) treatments and eventu-
Some of them, such as the amanitins and phallotoxins, are synthe-
ally resulted in the death of both patients were reported in Italy
sized ribosomally by various species of mushrooms, and can be
[154]. In 2009, the emergence of new forms of resistant M. tubercu-
extremely toxic [98].
losis strains, totally drug-resistant (TDR), was also documented in
The use of gramicidins and other polypeptides, however, is
Iran [231]. These strains were resistant to all first- and second-
limited to topical applications. The mechanism of action of these
line drugs tested (isoniazid, rifampicin, streptomycin, ethambutol,
antimicrobials is as follows: they act as channels and increase
pyrazinamide, ethionamide, para-aminosalicylic acid, cycloserine,
the permeability of the bacterial cell membrane when incorpo-
ofloxacin, amikacin, ciprofloxacin, capreomycin, and kanamycin).
rated, thus destroying the ion gradient between the cytoplasm
TDR tuberculosis cases have been also described in India and South
and the extracellular environment [227,179]. In animals and
Africa [226,123].
humans, this activity, at concentrations lower than needed to
Although presently relatively rare, these cases demand imme-
achieve the bacterial killing effect, induces hemolysis. Thus, topi-
diate actions geared towards the rapid identification and con-
cal applications require the skin or mucosa surface to be intact
tainment of these dangerous strains to prevent further
to prevent systemic entry. Other polypeptide antibiotics also dis-
outbreaks of untreatable TB [232]. Secondly, new antibiotics that
play substantial side effects and are thus mostly used as topical
are effective against these extremely problematic infections have
applications. In exceptional cases, however, they can be used
to be urgently sought. Bedaquiline is the first new medicine for
systemically as last resort antibiotics when other options are
TB in more than forty years and received a fast-track approval
exhausted, for instance in the case of resistance or contraindica-
by the FDA for use only in cases of MDR and TDR tuberculosis
tions such as allergy [18].
that resist the first and second lines of treatment [81]. Unlike
other quinolones, the molecular target of this antibiotic is
6. Aminoglycosides mycobacterial ATP synthase, not DNA gyrase. Targeting energy
metabolism may thus represent a new, promising approach for
In 1943, Selman Abraham Waksman headed a research team at antibacterial drug discovery [13,126]. There are a number of
Rutgers University in the search for new antibiotics [235]. This compounds at various stages of clinical and preclinical trials
effort was fuelled by the success of penicillin that was discovered for the treatment of TB, especially its drug-resistant forms
in 1928 but did not evolve into a useful treatment until the (http://www.newtbdrugs.org/pipeline.php).
1940s. The same year, his student Albert Schatz isolated two Aminoglycosides were extensively used in the early days of
strains of Streptomyces active against tubercle bacillus and Gram- antimicrobial chemotherapy but were largely replaced in the
negative bacteria resistant to penicillin and purified the active 1980s with more efficient antimicrobials, with lesser side effects,
compound, streptomycin. The authorship issue in streptomycin such as cephalosporins, carbapenems, and fluoroquinolones
discovery, however, was surrounded by considerable controversies [127]. There is, however, a renewed interest in aminoglycosides
[233,133,120,234]. Clinical trials in the following year demon- due to the ever-increasing problem of antibiotic resistance. In par-
strated that streptomycin is effective against infectious diseases ticular, gentamicin is still widely used in hospital settings for treat-
caused by Gram-negative bacteria and Mycobacterium tuberculosis. ment of serious infections [131]. Amikacin is commonly used in
Despite the toxicity and an already present resistance problem, the intensive care units for the treatment of patients with life-
drug soon became the cornerstone for multidrug therapies of threatening Gram-negative infections [150]. Aminoglycosides dis-
tuberculosis. play synergistic activities in combination with other antibiotic
Following the discovery and introduction of streptomycin and classes, while the safety and efficacy issues can be potentially
other antibiotics into clinical practice, the mortality rates due to solved via PK/PD-based optimised therapy regimens.
8 R. Aminov / Biochemical Pharmacology 133 (2017) 4–19

7. Tetracyclines the diet of these ancient people. The second case of tetracycline
incorporation in bones of Sudanese Nubians is documented for
The first antibiotic of this class, chlortetracycline, was discov- the remains from the medieval period (550–1450 CE) [110].
ered in 1945 by Benjamin Minge Duggar under the supervision Although there have been some doubts regarding the identity of
of Yellapragada Subbarow at Lederle Laboratory Division of Amer- the fluorescent compound(s) in the ancient bones, the results of
ican Cyanamid Company [71]. The antibiotic-producing soil bac- mass spectroscopic characterization conclusively identified them
terium was named Streptomyces aureofaciens, and the antibiotic as tetracyclines [162].
purified from it was marketed under the trade name Aureomycin, Another example of ancient tetracycline exposure came from a
to reflect the golden colour of both the producer colonies and the histological study of samples taken from the femoral midshafts of
product purified. Aureomycin was almost immediately examined the late Roman period skeletons from the Dakhleh Oasis, Egypt
in the treatments of various human infections, and the first pre- [62]. The samples demonstrated discrete fluorochrome labelling
scriptions were issued within a timeframe that is almost unimag- consistent with the presence of tetracycline, thus suggesting its
inable for the tempo of current drug development. The clinical intake with the diet in the late Roman period [62]. The hypothe-
evaluations demonstrated a broad clinical applicability of the drug, sized intake of tetracycline may potentially have had a protective
estimated to be equal in value to penicillin [241]. effect because the rate of infectious diseases documented in the
The interesting twist in the tetracycline story is that the animal Sudanese Nubian population was low, and no traces of bone infec-
growth promoting properties of antibiotics were for the first time tion were detected in the samples from the Dakhleh Oasis [15,62].
demonstrated namely with this antibiotic. Colleagues of Benjamin There was no correlation, however, between the exposure to low
Minge Duggar at Lederle Laboratory Division, animal nutritionist concentrations of tetracycline and disease incidence in the medie-
Robert Stokstad and biochemist Thomas Jukes, accidentally came val population of Sudanese Nubia [110].
across the growth promoting effects of the biomass of S. aureofa- As mentioned before, the extensive use of tetracyclines in clin-
ciens, left after the fermentation and extraction of Aureomycin, ical medicine and in production of food animals started from the
on chickens. This discovery occurred during their search for 1950s. In 2013, the leading antibiotics by use in food animals in
sources of vitamin B-12, and the debris of S. aureofaciens contained the USA were tetracyclines (6,514,779 kg of active ingredient
a substantial amount of it. It appeared, however, that the growth annually), which accounted for 71% of all antibiotics sold for use
promoting property was not due to the vitamin but because of in food-producing animals [84]. Tetracyclines administered via
the low residual antibiotic left in the producer biomass after medicated feed accounted for the majority of domestic sales and
antibiotic extraction. Ensuing the discovery, American Cyanamid distribution of medically important antimicrobials approved for
Company swiftly initiated the development of antibiotic feed addi- use in food-producing animals, and they were also the leading
tives for growth promotion of food animals, and this exemplar was antibiotics for administration via drinking water [84]. Besides,
promptly followed by many other companies and countries around the non-clinical use of tetracyclines was extended to aquaculture
the globe. It is highly likely that the extremely rapid onset and glo- and horticulture [57]. The clinical use of the first-generation tetra-
bal dissemination of tetracycline resistance among pathogens was cyclines, however, progressively decreased due to the widespread
(and is) mainly due to the massive use of tetracyclines by the food resistance towards this antibiotic class. The most common mecha-
animal industry. It took a considerable amount of time and efforts nisms of resistance are the efflux of tetracycline from the cell and
to reveal this link, and the corresponding legislative measures to ribosomal protection, where tetracycline is prevented from bind-
limit and ban the use of growth-promoting antibiotics were first ing to the ribosome because of the synthesis of alternative elonga-
implemented in the European Union (EU) countries. Sweden pro- tion factors [57]. The less conspicuous mechanisms of tetracycline
hibited them in 1986, while other EU countries banned specific resistance included the enzymatic degradation of tetracycline
antibiotics in feedstuffs prior to January 1, 2006, when all these [209] and, presumably, a metabolic mechanism protecting against
antibiotics were deleted from the Community Register of autho- the entry of the antibiotic into the cell [117]. The wide dissemina-
rized feed additives [76,50]. tion of antibiotic resistance genes is facilitated by their location on
In a parallel drug discovery programme, which was a collabora- MGEs, allowing an almost indiscriminate exchange among a
tive effort between Pfizer and Harvard University, the research variety of taxonomic entities [8].
team discovered and resolved the chemical structure of another The rapid expansion of tetracycline resistance among the
tetracycline antibiotic, oxytetracycline (trade named Terramycin) human and animal pathogens prompted the development of the
[109]. With the determination of the chemical structure of other second-generation tetracyclines such as minocycline (available
tetracyclines, this group of antibiotics is currently defined as: ‘‘A from 1966 [184]) and doxycycline (available from 1967 [60]). They
subclass of polyketides having an octahydrotetracene-2-carboxa are used to treat many different infectious diseases, such as urinary
mide skeleton, substituted with many hydroxy and other groups” and intestinal tract infections, respiratory infections, skin infec-
[113]. Resolving the chemical structure of natural tetracyclines laid tions, acne, gonorrhoea, tick fever, chlamydia, eye infections, peri-
the foundation for the development of the second-generation odontitis, and others. Besides the antibacterial effects, they also
tetracyclines, such as doxycycline and minocycline, to combat display other potent activities directed towards the eukaryotic cell
the ever-growing resistance problem [163]. targets [10]. In particular, minocycline displays strong anti-
Another interesting aspect of tetracyclines is that there are indi- inflammatory, neuroprotective, anti-proteolytic, and anti-
cations of their ancient consumption well before their discovery in apoptotic properties, as well as inhibits angiogenesis and meta-
the modern era. The traces of tetracycline have been found in static growth [91]. In addition, it displays antioxidant activity, inhi-
human skeletal remains from ancient Sudanese Nubia dating back bits several enzyme activities, and modulates immune cell
to 350–550 CE [28]. The efficient incorporation and binding of activation and proliferation [10].
tetracycline to the mineralizing surface of the bone is well known In June 2005, tigecycline, the first representative of the third-
in clinical research and histomorphometry applications. It is a generation tetracyclines (also called glycylcyclines), was approved
standard method for determining the rate of bone formation in by the FDA [190]. It is administered intravenously to treat compli-
all bone compartments, including cancellous, endocortical, intra- cated skin and intra-abdominal infections, as well as respiratory
cortical, and periosteal bone [139]. The presence of tetracycline infections. It is active against many MDR pathogens including S.
incorporated in bones of ancient Sudanese Nubians is only explica- aureus, A. baumannii, K. pneumoniae, and Escherichia coli. After a
ble if there was exposure to tetracycline-containing materials in decade of clinical use, tigecycline remains relatively active towards
R. Aminov / Biochemical Pharmacology 133 (2017) 4–19 9

infections caused by the Enterobacteriaceae, with resistance rates riaceae expressing the novel NDM-1 metallo-b-lactamase still
largely <10% in most of the wide-scale surveillance studies per- remain susceptible to colistin [130].
formed [248]. Among the mechanisms that may confer resistance Colistin targets the bacterial cell membrane of Gram-negative
to tigecycline, RND-type transporters, such as the AcrAB efflux bacteria. It initially associates with the anionic lipopolysaccharide
pump, may play a major role. The extensive dissemination of the (LPS) molecules in the outer membrane of Gram-negative bacteria
tet(X) gene, which confers resistance to all tetracyclines, including and displaces magnesium and calcium, which stabilize the nega-
tigecycline, to pathogenic microbiota may also represent a signifi- tively charged LPS molecules [78]. This results in a local distur-
cant problem in controlling resistance to this antibiotic [11]. bance of the outer membrane, with increased permeability,
leakage of the cell content, and, eventually, cell death. Although
presently rare, resistance to colistin is emerging. It is encoded on
8. Amphenicols
a plasmid with a very high conjugative transfer frequency and a
potential for rapid dissemination to key pathogenic species of the
Chronologically, the next antibiotic in the pipeline of the antibi-
Enterobacteriaceae [140]. The authors suggest that one of the main
otic drug discovery programmes was chloramphenicol, which was
causes of the emerging resistance could be its large-scale use in
discovered and isolated from Streptomyces venezuelae by David
agriculture. Another worrying finding is that colistin therapy may
Gottlieb in 1947. It belongs to the class of amphenicols, with a
increase pathogen resistance to host cationic antimicrobials
phenylpropanoid structure. Although it was initially isolated from
[161]. Colistin selection for pathogen resistance against the first
a natural source, chemical synthesis appeared to be more advanta-
line of host’s innate immunity defence may potentially facilitate
geous, and chloramphenicol became the first antibiotic produced
the infection process and thus increase the infection rates.
synthetically on an industrial scale [68]. Chloramphenicol easily
Another cyclic lipopeptide antibiotic, daptomycin, is produced
crosses the haematoencephalic barrier and, in some cases, could
by Streptomyces roseosporus [218]. It was discovered by researchers
be the drug of choice for the treatment of bacterial meningitis
at Eli Lilly and Company in early 1980s but did not enter the clin-
caused by Neisseria meningitidis, Streptococcus pneumoniae, and
ical practice until 2003 [212]. It is active only against Gram-
Haemophilus influenza, especially in patients with an allergy to b-
positive bacteria, including vancomycin-resistant enterococci
lactams or in patients with other serious infections when alterna-
(VRE) and methicillin-resistant S. aureus (MRSA). The mechanism
tive medications are contraindicated or inefficient [73]. The main
of action includes primarily targeting bacterial cell membranes
disadvantage of the antibiotic is in its side effects, which are quite
leading to the mislocalization of essential cell division proteins
common after a prolonged use, and may include haematological
and causing severe cell wall and membrane defects, with an even-
disorders such as aplastic anaemia, bone marrow suppression,
tual breach in the cell membrane integrity and cell death [176].
and leukaemia. It may cause neurotoxicity and Grey syndrome in
The antibiotic is well tolerated, with the frequency and distribution
infants because of the accumulation of toxic chloramphenicol
of adverse effects similar to comparator drugs [194]. Resistance to
metabolites in patients of this age group [45].
daptomycin remains rare. In S. aureus, it is the result of incremental
To circumvent the toxicity and side effect problems, a number
accumulation of point mutations in genes encoding a lysylphos-
of other amphenicol derivatives have been synthesized [68], and
phatidylglycerol synthetase, a histidine kinase, and RpoB and RpoC
some of them, such as thiamphenicol, azidamfenicol, and florfeni-
subunits of RNA polymerase [89]. There is a good potential for the
col, have even entered into clinical and veterinary practice. The rel-
development of various daptomycin derivatives using a biosyn-
atively simple molecule of chloramphenicol opened the possibility
thetic engineering approach [23].
of constructing hybrid antibiotics, where the segments of two
A group of lipopeptides called echinocandins display
drugs are covalently connected into one molecule [68]. Besides
potent antifungal activities [65]. They noncompetitively inhibit
antimicrobial activities, this class of drugs may find potential
b-(1,3)-D-glucan synthase, an essential enzyme complex for the
applications in cancer therapy [68].
synthesis of glucan in the fungal cell wall [156]. They were discov-
The most frequently encountered mechanism of resistance
ered in 1970s during extensive screening programmes for antifun-
towards chloramphenicol is its enzymatic inactivation by acetyla-
gals with a broad-spectrum activity against the species of Candida.
tion, via different types of chloramphenicol acetyltransferases
Although the natural products appeared to be toxic, the synthetic
[197]. Less frequent are the efflux of chloramphenicol, its inactiva-
modifications allowed lowering the toxicity, and the approved
tion by phosphotransferases, target mutations, and permeability
drugs among the semi-synthetic echinocandins include caspo-
barriers.
fungin, micafungin, and anidulafungin [156]. In a recent report
by the Centers for Disease Control and Prevention regarding the
9. Lipopeptides microorganisms with a serious threat level, the only fungal patho-
gen among the 12 most serious threats is a fluconazole-resistant
The structure of this class of antimicrobials includes a cyclic Candida [51]. Thus the echinocandins serve as a valuable first-
peptide with a hydrophobic tail. Of significant clinical relevance line treatment option against these serious fungal infections. Resis-
is colistin (also known as polymyxin E), a polypeptide antibiotic tance to the echinocandins is rare, essentially limited to the case
produced by certain strains of Paenibacillus polymyxa (formerly studies with the resistance emerging during the treatment [77].
Bacillus polymyxa var. colistinus). It was discovered in Japan in
1947 and entered the clinical practice in 1949 [215]. It fell out of
favour, however, because of a variety of adverse effects, such as 10. Macrolides
nephrotoxicity, ototoxicity, and neuromuscular blockade, and
when other, less toxic, antimicrobials became available [249]. With The first antibiotic of the macrolide class, pikromycin, was iso-
the emergence and dissemination of Gram-negative bacterial lated from Streptomyces venezuelae by Brockmann and Henkel in
pathogens that are resistant to the mainstream treatment with 1950 [42,43]. It has not advanced to clinical use but still remains
the aminoglycosides, b-lactams, and quinolones, there is a renewed an important precursor for the synthesis of other macrolides and
interest in the use of colistin. It can be used as a last resort antibi- derivatives [121]. The first commercially successful macrolide, ery-
otic against serious and difficult-to-treat infections such as those thromycin (also known as Ilosone or Ilotycin to reflect the soil sam-
caused by multidrug-resistant P. aeruginosa, K. pneumoniae, and ple collection place in the Iloilo region in the Philippines), was
A. baumannii [79]. Multidrug-resistant isolates of the Enterobacte- discovered by the team of scientists led by J. M. McGuire at Eli Lilly
10 R. Aminov / Biochemical Pharmacology 133 (2017) 4–19

[153]. Acting within the antibiotic discovery programme there, the against the antibiotic action [237]. But since telithromycin has
team tested many soil samples, including the one sent in 1949 by the capability to bind strongly to another site, it overcomes the
Abelardo Aguilar, a Filipino scientist employed by the company. resistance conferred by this mechanism.
The genome sequence of the erythromycin producer, Saccha- The only fluoroketolide drug, solithromycin (oral formulation
ropolyspora erythraea (formerly Streptomyces erythraeus), was pub- called Solithera), is currently undergoing the final stages of clinical
lished in 2007 [167]. trials and has not yet been approved. With its three binding sites
Structurally, the natural macrolides consist of a large macro- on the ribosome target, it is an important improvement compared
cyclic lactone ring, which is typically 14-, 15-, or 16-membered. to the ketolides [138]. It also lacks the pyridine-imidazole group
One or more deoxy sugars may be attached to the lactone ring. present on the side chain of telithromycin, which acts as an antag-
Their natural synthesis is performed by polyketide synthases, onist of various nicotinic acetylcholine receptors and thus causes
which is a family of multi-domain enzymes or enzyme complexes off-target undesirable side effects [34].
that can be found in the representatives of bacterial and eukaryotic Besides the antimicrobial use, the macrolides have demon-
domains [118,115]. Besides the macrolide biosynthesis, polyketide strated very good anti-inflammatory and pro-kinetic properties
synthases are also involved in the biosynthesis of many other bio- in the treatment of various chronic respiratory diseases of non-
logically active secondary metabolites. These include a broad range infectious nature. It was demonstrated, almost three decades ago,
of compounds covering other antibiotic classes such as tetracycli- that the long-term administration of low-dose erythromycin had
nes, as well as immunosuppressants/immunomodulators, and a positive effect on patients with diffuse panbronchiolitis [129].
anticholesterol and anticancer drugs [124]. In terms of infection From this point on, the use of macrolides in the therapy of chronic
control, the macrolides are possibly the second most prescribed respiratory diseases of non-infectious nature became one of the
antibiotic class after the b-lactams, targeting a similarly broad mainstream options in the management of this group of diseases
range of bacterial pathogens, although with a lesser efficiency [160]. Other pulmonary diseases, for which the non-antimicrobial
against Gram-negative bacteria. The obvious advantage of the therapeutic effect of macrolides has been demonstrated, include
macrolides over the b-lactams is in the activity against bacteria cystic fibrosis [144], asthma [93], and chronic obstructive pul-
lacking the cell wall target such as the mycoplasmas. Macrolides monary disease [199]. Furthermore, the positive non-
are also among the first-line treatment options for patients with antimicrobial therapeutic effect of macrolides has been demon-
penicillin allergy. The subgroup of macrolides, polyene antimy- strated in cardiovascular diseases, autoimmunity, prophylaxis of
cotics, consists of antibiotics active against fungal infections, and transplant rejection, and a number of other conditions [10]. Alleg-
these include amphotericin B, nystatin, and others [99]. edly, these therapeutic effects of macrolides are due to their anti-
Various modifications of natural macrolides have been per- inflammatory properties [10].
formed to improve the pharmacokinetic properties of antibiotics
in this class, such as the use of the azalide scaffold [112,216]. This
approach was initially implemented in the synthesis of 9-dihydro-
9-deoxo-9a-methyl-9a-aza-9a-homoerythromycin A (azithromy- 11. Oxazolidinones
cin) by the Croatian pharmaceutical company Pliva in 1980 [95].
The antibiotic displays outstanding pharmacokinetic properties, The oxazolidinones class of antimicrobials is characterised by
including the lack of inhibition of cytochrome P450 3A4 [12,159]. the presence of 2-oxazolidone in their chemical structure. There
It was the most prescribed antibiotic for outpatients in the US in are two groups in this class that differ in their mechanism of
2010 [107]. With other macrolides, this group of antibiotics is antimicrobial activity. The first is represented by cycloserine ((R)-
among the most frequently prescribed anti-infective drugs in out- 4-amino-1,2-oxazolidin-3-one), which was initially isolated from
patient care [136,202]. Streptomyces K-300 strain by Kurosawa in 1952 and named orien-
Further efforts to improve the antibacterial activities of macro- tomycin [27]. The antimicrobials isolated later from S. lavendulae, S.
lides have been focused on increasing the affinity to the target and orchidaceus, S. garyphalus, and other Streptomyces species appeared
the capability to bind to a larger number of sites on the ribosome to be identical in structure and were given the generic name
target. These works have led to the development of ketolides and cycloserine or D-cycloserine. It can also be synthesized chemically
fluoroketolides. The first and the only approved ketolide drug, teli- [211]. The antibacterial action of D-cycloserine is mainly due to the
thromycin, was developed by the French pharmaceutical company inhibition of D-Ala-D-Ala ligase activity, thus interfering with cell-
Hoechst Marion Roussel (later a subsidiary of Sanofi-Aventis and wall biosynthesis [180]. The drug is a second-line antibiotic used
then Sanofi) and approved by the European Commission in 2001 in the treatment of tuberculosis, particularly of MDR M. tuberculo-
and by the FDA in 2004. The most important modifications of the sis infections [250]. The recently uncovered resistance mechanism
precursor erythromycin molecule included the removal of the neu- of M. tuberculosis towards D-cycloserine involves the loss-of-
tral sugar L-cladinose from position 3 of the macrolide ring and the function mutations in ald (Rv2780), which encodes the L-alanine
subsequent oxidation of the 3-hydroxyl to a 3-keto functional dehydrogenase [66].
group, thus also giving the ketolides name for this group of drugs The mechanism of the antibacterial activity in the second group
[196]. Other modifications included the addition of a carbamate of oxazolidinones involves the inhibition of protein synthesis, by
ring in the lactone ring, attachment of an alkyl-aryl moiety to the targeting an early step involving the binding of N-
carbamate ring, and methylation of carbon at position 6. The affin- formylmethionyl-tRNA to the ribosome [204]. The antibacterial
ity of the resulting compound, telithromycin, to the 50S subunit of properties of compounds in this group were already known in
the ribosome is more than by an order of magnitude higher than the 1970s, and a number of derivatives were synthesized by
that of erythromycin. Besides, telithromycin displays a strong DuPont Pharmaceuticals researchers as potential antimicrobials
binding capability to two domains in the 23S RNA molecule, while for use in mammals [206,41]. While active in vitro, they were, how-
a strong binding of older macrolides is limited only to one domain, ever, too toxic, affecting in particular the liver, in order to be useful
with a weak binding to the second domain. The significant advan- in clinical applications. Finally, the oxazolidinone research pro-
tage of this design is that telithromycin is also active against bac- gramme at Pharmacia & Upjohn (now part of Pfizer) resulted in a
teria resistant to older macrolides. Resistance to macrolides is relatively safe antimicrobial, linezolid [155,88], which was
frequently due to the dimethylation of the adenine residue approved by the FDA in 2000 and, shortly thereafter, by the corre-
A2058 in the 23S rRNA molecule, which protects the ribosome sponding regulatory bodies in a number of other countries.
R. Aminov / Biochemical Pharmacology 133 (2017) 4–19 11

Linezolid is active against Gram-positive bacteria and especially and four minor components (RS-1 through RS-4) [33]. The compo-
useful for the treatment of skin, pulmonary and other infections nents from A2-1 to A2-5 contain the same teicoplanin glycopeptide
such as those caused by MDR streptococci, VRE, and MRSA [174]. core, and they differ only by the lengths and branching of their
It is not clinically effective against Gram-negative pathogens as a acyl-aliphatic side-chains. Compared to vancomycin teicoplanin
result of endogenous efflux activity in these organisms [141]. demonstrates better pharmacokinetics allowing once-daily dosing,
Although relatively safe during short-term treatments, a more pro- and it is also a safer drug, with a lower incidence of nephrotoxicity
longed use may result in peripheral or optical neuropathy, most or ototoxicity [158]. Thus teicoplanin may have pharmacoeco-
likely due to the mitochondrial toxicity of the drug [207,26]. Resis- nomic advantages over vancomycin in at least of treatment of sim-
tance to linezolid remains low, in a survey of clinical isolated from ilar Gram-positive bacterial infections [210,239]. Teicoplanin
23 countries all streptococci were found to be susceptible, and the remains active against vancomycin resistance caused by VanB
resistance was rare among S. aureus (0.03%), coagulase-negative and VanC, but is not active against VanA resistant strains [173].
staphylococci (0.28%), and the enterococci (0.11%, 0.55% intermedi- According to a large European survey, however, the resistance
ate) [116]. Other drugs in this group include tedizolid, which was levels among Gram-positive bacterial pathogens during 1995 were
approved by the FDA in 2014 [82], and a number of other oxazo- similar towards the two antibiotics [97].
lidinones are currently under the various stages of development. The currently approved second-generation glycopeptides
include telavancin (approved by the [80], and oritavancin and dal-
bavancin (both approved in 2014) [35]. In addition to the ‘‘classi-
12. Glycopeptides
cal” activity of glycopeptide, such as the inhibition of cell wall
biosynthesis, telavancin and oritavancin also disrupt bacterial
This class consists of antibiotics composed of glycosylated cyclic
membrane integrity and increase membrane permeability, while
or polycyclic peptides synthesized nonribosomally. The first repre-
oritavancin also inhibits RNA synthesis [247]. The multiplicity of
sentative of the class, vancomycin, was discovered in 1953 by
targets makes them extremely potent, surpassing the potency of
Edmund Kornfeld and the team at Eli Lilly in the producer bac-
vancomycin by 4- to 8-fold [122]. Another advantage of some
terium Amycolatopsis orientalis (formerly Streptomyces orientalis
second-generation glycopeptides is excellent pharmacokinetics
and Nocardia orientalis), which was isolated from the soil sample
allowing less frequent administrations such as once-daily for tela-
brought from Borneo [205,137]. Vancomycin and resistance to it
vancin, once-weekly for dalbavancin, and one dose per treatment
has received considerable media attention, because the drug is
for oritavancin [228,247]. These antimicrobials are also called the
not a first-line antibiotic but is reserved as a last resort option
lipoglycopeptides to reflect the synthetic modifications made to
for life-threatening conditions, such as septicemia, and compli-
the parent glycopeptides by the addition of lipophilic tails
cated infections of the lower respiratory tract, skin, and bones
[247,187].
caused by Gram-positive bacteria [38]. The emergence and dissem-
ination of VRE [14], vancomycin-intermediate S. aureus (VISA)
[108], and vancomycin-resistant S. aureus (VRSA) [54,90] may rep-
13. Streptogramins
resent a significant problem for healthcare facilities such as hospi-
tals. There are suggestions that the agricultural growth-promoting
Streptogramins have been isolated from many different species
use of another glycopeptide antibiotic, avoparcin, may have con-
of Streptomyces in the course of numerous antibiotic discovery pro-
tributed to the wide dissemination of vancomycin resistance
grammes in many companies, so it is hard to tell which strep-
[19,59,2].
togramin was discovered first [58]. According to the available
The mechanisms of vancomycin action and the corresponding
literature, the first description of streptogramin was published by
resistance mechanisms are largely well understood, especially that
Charney and others [55]. Among streptogramins, probably the
of the acquired resistance. The drug inhibits cell wall biosynthesis
most known is virginiamycin, which was (and still is) used exten-
in Gram-positive bacteria by tightly binding to the terminal dipep-
sively in food animals for growth-promoting purposes [25,246]. In
tide D-Ala-D-Ala on the end of the precursor peptide chains, thus
pigs, addition of virginiamycin to feed is superior compared to
preventing transpeptidation and transglycosylation, which halts
chlortetracycline in terms of growth promotion (9% faster) and
the cross-linking of the cell wall, and leads to cell lysis and death
feed conversion efficiency (5% more efficient) [152]. The former
[114]. Acquired resistance to vancomycin is mainly caused by the
is also used in fuel ethanol production to prevent and treat bacte-
modification in the terminal amino acid residues of the precursor
rial contamination during the fermentation process [37]. Only con-
peptide. The van genes, which usually reside on MGEs, encode
siderably later, the streptogramins, such as pristinamycin and
the set of enzymes that alter these residues to D-Ala-D-Lac, with a
quinupristin/dalfopristin, started to be marketed for human con-
dramatic loss of the target affinity to vancomycin, which makes
sumption as well [100,4]. The targeted pathogens are mainly the
the cell wall biosynthesis insensitive to the inhibitory action of
strains of VRE and MRSA [64,74].
the drug [17]. Another modification of the terminal dipeptide to
Streptogramin antibiotics are unique in the sense that the pro-
D-Ala-D-Sermay also offer protection against the drug but at a ducer strains synthetize two structurally unrelated antibiotics,
much lower level [178]. An interesting and valuable approach streptogramin A, which is a cyclic hybrid peptide-polyketide macro-
has been proposed to overcome this type of resistance by reengi- lactone compound, and streptogramin B, which is a cyclic depsipep-
neering vancomycin to confer a dual binding capability to A-Ala- tide compound [58]. The combination of these two results in a
A-Ala and A-Ala-A-Lac, with a resulting significant activity against potent synergistic antibacterial action. In particular, binding of
VanA VRE [63]. The subsequent vancomycin reengineering works streptogramin A to the bacterial ribosome facilitates binding of
resulted in derivatives with a remarkable spectrum of extremely streptogramin B to the same target, and the synergy of actions
potent activities against both vancomycin-sensitive and results in rapid bacterial cell death [67]. X-ray structural analyses
vancomycin-resistant pathogenic bacteria [243,244,165,166]. suggests that streptogramin B, which is a cyclic depsipeptide com-
The second natural antibiotic of this class, teicoplanin (formerly pound, acts analogously to the macrolides by binding to the riboso-
named teichomycin) was isolated from Actinoplanes teichomyceti- mal exit tunnel and blocking it [102]. Streptogramin A binds close to
cus and described in 1978 [172,24]. Teicoplanin actually represents streptogramin B within the peptidyl transferase centre, thus affect-
a mixture of several compounds consisting of five major compo- ing both A- and P-site occupation by tRNA molecules. The conforma-
nents (A2-1 through A2-5), one hydrolysis component (A3-1), tional changes of the ribosome imposed by the streptogramins,
12 R. Aminov / Biochemical Pharmacology 133 (2017) 4–19

therefore, may be responsible for their bactericidal activity and the changes that significantly decrease the affinity of the b-subunit
post-antibiotic inhibition of protein synthesis [102]. to the drug [182,87]. Other mechanisms of resistance include
Although streptogramin B is a cyclic hexadepsipeptide, which is duplication of the target, action of RNAP-binding proteins, various
structurally different from the macrolides and lincosamides, these enzymatic modifications of rifamycins, and alteration of cell per-
three classes of antibiotics share the similar functionality of bind- meability [225]. Similar to quinolone resistance (see the next sec-
ing to the same site on the 50S ribosomal subunit. Structural stud- tion), some of the rifamycin resistance mechanisms may have the
ies suggest that macrolide–lincosamide–streptogramin B classes environmental origin [225,208].
(MLSB) of antibiotics share a common mode of action, which is
modulated by the space available between the peptidyl transferase
centre and the drug [219]. Thus, it is not surprising that there is 15. Quinolones
overlapping resistance to these three classes of antibiotics. In par-
ticular, these are the erm genes that encode methylases, which This group of antimicrobials is unique in the sense that its first
dimethylate the adenine residue A2058 in the 23S rRNA compo- representative, nalidixic acid, was discovered during an attempt of
nent of the 50S large subunit of the bacterial ribosome [237]. The chloroquine synthesis [238]. After the introduction of nalidixic acid
residue is located within the conserved region of domain V, which in 1962 for the treatment of urinary tract infections, the synthesis
is a binding site for MLSB antibiotics. As a consequence, this results of other derivatives to broaden the range of targeted bacterial
in cross-resistance to all three classes of antibiotics, MLSB resis- pathogens resulted in tens of thousands of new compounds, few
tance, towards macrolides, lincosamides, and depsipeptides of which have entered clinical practice and represent the currently
[135]. Chimeric streptogramin-tyrocidine antibiotics could be defined four generations of quinolones. The division into the gen-
potentially helpful in overcoming streptogramin resistance [157]. erations is fairly arbitrary, except for the first generation, which
is represented by the nonfluorinated drugs, while all later genera-
tions are exclusively fluorinated compounds (usually fluorinated at
14. Ansamycins the C-6 or C-7 position) and, thus, are commonly called fluoro-
quinolones. The most known antimicrobial activity of quinolones
A representative of this class, rifamycin (more precisely a mix of is due to the formation of a DNA gyrase-quinolone-DNA complex,
rifamycins A, B, C, D, and E), was first obtained in 1957 by Piero which blocks the bacterial chromosome replication leading to cell
Sensi, Maria Teresa Timbal and Pinhas Margalith, while working death [106]. Besides bacterial DNA gyrase, quinolones may also
at Gruppo Lepetit SpA in Milan. The producer strain, named at have other targets in the bacterial cell. Bedaquiline, for example,
the time Streptomyces mediterranei, was isolated from the soil sam- targets energy metabolism by inactivating ATP synthase [13,126].
ple collected in southern France [149]. The producer strain was In addition to the antibacterial activities, the 4-quinolone scaffolds
renamed several times, to reflect more accurately its taxonomy exhibit many other pharmacological properties such as antiviral,
according to various taxonomic criteria and phylogeny. Thus it antitumor, antiischemic, and anxiolytic activities [3,29].
went from the original naming as S. mediterranei to Nocardia Since the synthesis and testing of fluoroquinolones were aimed
mediterranei [220], to Amycolatopsis mediterranei [134] and, finally, at targeting a broad range of bacteria, the approved drugs are suit-
to Amycolatopsis rifamycinica [20]. able for the treatment of a variety of infections, with no obvious
The rifamycins appeared to be rather unstable to purify from Gram-positive vs. Gram-negative bias. They were the third in
the culture broth, except for Rifamycin B, which, unfortunately, terms of outpatient prescription frequency, after the b-lactams
was practically inactive [200]. However, it is spontaneously oxi- and macrolides, in the period from 2000 to 2010 [136]. Within a
dized and hydrolysed in aqueous solutions to produce the highly narrower and more recent timeframe (2007–2009), however,
active Rifamycin S. Further chemical modifications yielded Rifamy- ambulatory prescriptions of quinolones were the highest (25% of
cin SV, which became the first member of the ansamycins class to antibiotics), followed by macrolides (20%), and aminopenicillins
enter clinical use. An additional modification yielded an improved (12%) [202]. Thus in recent years the quinolones became a pre-
derivative, Rifamide, which entered the clinical practice, but was ferred choice for physicians to treat respiratory and skin/mucosal
limited to intravenous use. Among further modifications that conditions and urinary tract infections in outpatient care. This
included several hydrazones of 3-formylrifamycin SV, the hydra- trend, however, is highly undesirable because the frequent use of
zone with N-amino-N’-methylpiperazine (rifampin or rifampicin) these broad-spectrum antibiotics as a first-line treatment for many
appeared to be suitable for peroral use and, ensuing successful infectious diseases may lead to a widespread resistance problem.
clinical trials, was introduced into therapeutic use in 1968 [200]. Recently the FDA issued a recommendation to restrict the use of
Other semisynthetic derivatives include rifapentine, rifabutin, fluoroquinolones for treatment of uncomplicated infections so they
rifalazil, and rifaximin. The latter has a poor oral bioavailability can ‘‘. . .be reserved for those who do not have alternative treat-
and is used for the prevention of travellers’ diarrhoea [151]. It is ment options” [85].
also currently investigated for the treatment of other non- The mechanism of antimicrobial action of fluoroquinolones is
infectious pathologies of the gastrointestinal tract such as IBS mediated via the formation of DNA gyrase-quinolone-DNA com-
and IBD [177,195]. Rifalazil is used for the treatment of persistent plex blocking the replication process and thus leading to cell death
chlamydia infections [193]. But other rifamycins, including the [106]. The mechanisms of resistance to this class of antimicrobials
original drug, remain the first-line treatment for tuberculosis, are of great interest because these are completely synthetic com-
leprosy, and other mycobacterial infections, where they are usually pounds. It is generally thought that one of the main contributors
used in combination with other antimicrobials [201]. Other drugs of antibiotic resistance genes for dissemination to other bacteria,
among ansamycins include antimicrobials, such as naphthomycins including pathogens, are the producers of antibiotics, which thus
[22] and streptovaricins [186], as well as an antitumor antibiotic protect themselves from the lethal action of antibiotics they pro-
geldanamycin [32]. duce [240]. Consequently, in the absence of natural producers of
The mechanism of antimicrobial activity of rifamycins is unique quinolones, the presence of acquired resistance among pathogens
among other antibiotics in that it targets bacterial DNA-dependent is unlikely. Nevertheless, the acquired resistance to quinolones
RNA polymerase, with no cross-resistance with other antibiotics exists and is due to the qnr genes, which encode a 218-aa protein
[236]. Resistance to rifamycins develops quickly; in mycobacteria belonging to the pentapeptide repeat family, with a sequence
these are point mutations in the target gene, with amino acid homology to the immunity protein McbG [222]. This quinolone
R. Aminov / Biochemical Pharmacology 133 (2017) 4–19 13

Table 1 space available between the peptidyl transferase centre and the
History of discovery of main drug [219]. Thus, the modification and protection of the target site
antimicrobial classes.
by the erm gene-encoded methylases confer resistance to all these
Arsphenamines 1910 classes of drugs, including lincosamides [237].
b-Lactams 1929
Sulphonamides 1935
Polypeptides 1939
Aminoglycosides 1943 17. Concluding remarks
Tetracyclines 1945
Amphenicols 1947 Currently we are facing a global antibiotic crisis because of the
Lipopeptides 1947
alarmingly growing antimicrobial resistance among many human
Macrolides 1950
Oxazolidinones 1952 pathogens The mortality rate due to antimicrobial resistant infec-
Glycopeptides 1953 tions is at least 50,000 each year across Europe and the US alone,
Streptogramins 1953 with many hundreds of thousands more dying in other areas of
Ansamycins 1957 the world [168]. If no immediate actions are taken, the estimated
Quinolones 1962
Lincosamides 1963
death toll due to the antimicrobial resistance will reach 10 million
by the year 2050, surpassing the mortality rate, for example, of
cancer. It has to be emphasized here that all major classes of
antimicrobials were discovered during the golden age of antibiotic
resistance mechanism is via the binding of Qnr to DNA gyrase in discovery, which came to the end more than 50 years ago (Table 1).
the early stages of gyrase-DNA complex formation, and, by lower- Since then, principal activities in the new antimicrobial drug devel-
ing gyrase binding to DNA, Qnr may reduce the amount of opment have been focused, largely, on extensive modifications of
holoenzyme-DNA targets for quinolone inhibition [223]. The genes existing natural drugs, and also performed by reengineering and
encoding the pentapeptide repeat family proteins are common complete chemical synthesis, if cost-effective. The older antimicro-
among bacteria in many ecosystems, and the phylogenetic analysis bials can still be useful and their therapeutic use optimised, which
suggests that the qnr genes in pathogens have been probably allows extracting their antimicrobial potential to the full. In partic-
acquired from marine bacteria [5]. It has been suggested that one ular, one of the pharmacokinetic and pharmacodynamics (PK/PD)
of the pentapeptide repeat family proteins, MfpA, provides a topo- strategies, front-loading, may allow a more therapeutically effec-
logical assistance to DNA when needed, but also helps to maintain tive use of some ‘‘old” antibiotics such as colistin [183]. More PK/
it in a condensed state, hence preventing undesired topological PD data on the efficacy and toxicity ranges may provide recom-
changes during the replicative senescence periods [105]. Thus, mendations for the optimal use of this antimicrobial as well
the probable scenario for the acquired resistance to quinolones is [169]. Combining antibiotics that display synergistic interactions
that the DNA metabolism enzyme of environmental bacteria is also a promising strategy for the extension of the useful life of
appeared to be also protective against this synthetic antibiotic. some older antibiotics [40].
The frequency of horizontal gene exchange among bacteria of dif- The strategies for other potential natural antimicrobials discov-
ferent ecological compartments is high [8,9]. Once transferred to ery routes could be the exploitation of other ecological compart-
other, probably commensal, microbiota and subjected to a strong ments and taxonomic groups besides the well-known soil
selective pressure, the corresponding qnr genes disseminated, Actinomycetes. These could include the isolation of antibiotic pro-
eventually entering the pathogenic microbiota [5]. ducer strains, for example, from the marine environment
Despite being a product of an entirely chemical synthesis, struc- [181,111] or retrieving the antimicrobial biosynthetic pathways
turally similar compounds can be found in natural ecosystems as from the uncultivated part of environmental microbiota via the
well. The species of Pseudomonas and some other bacteria produce metagenomic approach [146]. The source of antimicrobials could
a quorum-signaling molecule, 2-heptyl-3-hydroxy-4-quinolone, be also enriched by antimicrobial peptides and compounds from
which belongs to the family of 2-alkyl-4-quinolones [70]. Initially, animals and plants [101] or by mimicking the natural bacterial
however, these compounds were defined as antimicrobials for and fungal lipopeptides [147]. Finally, it could be the use of the
their corresponding activities [103]. The abundance of natural qui- complete synthetic routes pioneered during the early years of the
nolones appeared to be underestimated: several plant, animal, and antibiotic era, as in the case of sulphonamides, but with the current
microbial species may produce them [104]. Then, potentially, qui- knowledge of bacterial targets and possibilities of in silico modeling
nolone resistance may have appeared as a protective mechanism of drug-target interaction. The latter approach becomes dominant
against these naturally produced substances. in the search for drugs aimed at the newly identified targets in a
bacterial cell. Other strategies may include drugs engineered to
16. Lincosamides possess dual target activities, such as a rifamycin-quinolone hybrid
antibiotic, CBR-2092 [188], or the previously mentioned chimeric
The first antibiotic of this class discovered was lincomycin, streptogramin-tyrocidine antibiotics [157] and the combination
which is produced by Streptomyces lincolnensis var. lincolnensis of other antimicrobials with amphenicols [68].
strain and was made available for clinical studies in January The need for new antibiotics is urgent [164,217]. For a variety of
1963 by Upjohn [145]. It has a rather narrow range of bacterial reasons, however, big pharmaceutical companies have largely
pathogens targeted, has considerable side effects, and is presently abandoned the antimicrobial research area [31]. Thus the level of
rarely used in the therapy of infectious diseases. Much broader are investment into the development of novel antimicrobials is not
the applications of clindamycin, which is obtained from lincomycin sufficient to meet the growing and urgent demands. In this regard,
by introducing a chlorine atom with the inversion of chirality [36]. the change of paradigms under which the regulatory agencies and
It has lesser side effects compared to the original drug and is used pharmaceutical industry operate can help to address the unmet
for the treatment of many anaerobic infections [44]. Its use, how- demands for new antimicrobials [31]. The suggestions proposed
ever, may be associated with the hospital-acquired Clostridium dif- include the development of a new business model for antimicro-
ficile-associated diarrhoea [221]. As mentioned before, the bials, which is considerably different from that for other pharma-
structurally unrelated macrolides, lincosamides and streptogramin ceuticals, and will separate the return on investment from
B, share a common mode of action, which is modulated by the antibiotic sales volume [56]. To compensate for potential business
14 R. Aminov / Biochemical Pharmacology 133 (2017) 4–19

deficiency implemented under this model, it calls for a much Author contribution statement
broader involvement of governments in financial support of antibi-
otic R&D, in particular by a broad menu of incentives across the The corresponding author contributed 100% of the submission.
antibiotic life-cycle, with the highest incentives targeted at the
development of antibiotics directed at the greatest health threats
arising from antibiotic resistance. It is envisaged that contributions References
from the governments of different countries should be coordinated
within a globally agreed framework [56]. The first practical steps in [1] E.P. Abraham, E. Chain, An enzyme from bacteria able to destroy penicillin,
this direction can be already seen in the form of the establishment Nature 146 (1940) 837, http://dx.doi.org/10.1038/146837a0.
[2] J. Acar, M. Casewell, J. Freeman, C. Friis, H. Goossens, Avoparcin and
of a new global public–private partnership to support antibiotic
virginiamycin as animal growth promoters: a plea for science in decision-
innovations called CARB-X [170]. CARB-X will invest more than making, Clin. Microbiol. Infect. 6 (9) (2000) 477–482, http://dx.doi.org/
US$350 million in the next 5 years to accelerate the advancement 10.1046/j.1469-0691.2000.00128.x.
[3] A. Ahmed, M. Daneshtalab, Nonclassical biological activities of quinolone
of a diverse range of innovative antimicrobials into clinical trials.
derivatives, J. Pharm. Pharm. Sci. 15 (1) (2012) 52–72, http://dx.doi.org/
We have to keep in mind, however, that even with newer and 10.18433/J3302N.
better antimicrobials introduced, there is always a risk that the [4] D.R. Allington, M. Rivey, Quinupristin/dalfopristin: a therapeutic review, Clin.
microbes may eventually develop resistance against them. More- Ther. 23 (1) (2001) 24–44, http://dx.doi.org/10.1016/S0149-2918(01)80028-
X.
over, no exception to this scenario has ever been seen: the devel- [5] R.I. Aminov, R.I. Mackie, Evolution and ecology of antibiotic resistance genes,
opment of resistance is just a question of time and the FEMS Microbiol. Lett. 271 (2007) 147–161, http://dx.doi.org/10.1111/j.1574-
antimicrobial use practices involved. Thus, limiting the emergence 6968.2007.00757.x.
[6] R.I. Aminov, The role of antibiotics and antibiotic resistance in nature,
and dissemination of antimicrobial resistance remains one of the Environ. Microbiol. 11 (2009) 2970–2988, http://dx.doi.org/10.1111/j.1462-
important cornerstones of antimicrobial stewardship to prolong 2920.2009.01972.x.
the useful lifespan of these drugs. [7] R.I. Aminov, A brief history of the antibiotic era: lessons learned and
challenges for the future, Front. Microbiol. 1 (2010) 134, http://dx.doi.org/
There is a direct correlation between the level of antibiotic use 10.3389/fmicb.2010.00134.
and corresponding resistance [94], which is perfectly understand- [8] R.I. Aminov, Horizontal gene exchange in environmental microbiota, Front.
able from the biology point of view: the broader a selection is, the Microbiol. 2 (2011) 158, http://dx.doi.org/10.3389/fmicb.2011.00158.
[9] R.I. Aminov, The extent and regulation of lateral gene transfer in natural
higher is the chance for the trait selected, that is antibiotic resis- microbial ecosystems, in: M.P. Francino (Ed.), Horizontal Gene Transfer in
tance, to appear [5]. The human antibiotic consumption and the Microorganisms, Horizon Scientific Press, Norwich NR9 3DB, UK, 2012, ISBN
use of last-resort antibiotics are on the rise [229]. While we cannot 978-1-908230-10-2. Chapter 6.
[10] R.I. Aminov, Biotic acts of antibiotics, Front. Microbiol. 4 (2013) 241, http://
easily judge what is right and what is wrong in the use of antibiotics
dx.doi.org/10.3389/fmicb.2013.00241.
in humans for treatment of infectious diseases, the other uses may [11] R.I. Aminov, Evolution in action: dissemination of tet(X) into pathogenic
be confronted and addressed to. It seems obvious that the use of microbiota, Front. Microbiol. 4 (2013) 192, http://dx.doi.org/10.3389/
antibiotics has to be limited to the approved and controlled treat- fmicb.2013.00192.
[12] G.W. Amsden, Advanced-generation macrolides: tissue-directed antibiotics,
ment of bacterial infectious diseases, both in humans and animals, Int. J. Antimicrob. Agents 18 (Suppl. 1) (2001) S11–S15, http://dx.doi.org/
but presently this is not the case. Several estimates, for example, 10.1016/S0924-8579(01)00410-1.
suggest that a substantial proportion of antibiotics produced are [13] K. Andries, P. Verhasselt, J. Guillemont, H.W. Göhlmann, J.M. Neefs, H.
Winkler, J. Van Gestel, P. Timmerman, M. Zhu, E. Lee, P. Williams, D. de
used in food animals for non-therapeutic purposes [132,128]. Chaffoy, E. Huitric, S. Hoffner, E. Cambau, C. Truffot-Pernot, N. Lounis, V.
Extensive literature suggesting an epidemiologic link between the Jarlier, A diarylquinoline drug active on the ATP synthase of Mycobacterium
antibiotic use in food animals and antibiotic resistance in humans tuberculosis, Science 307 (5707) (2005) 223–227, http://dx.doi.org/
10.1126/science.1106753.
has been also compiled [132]. Unfortunately, however, the global [14] C.A. Arias, B.E. Murray, The rise of the Enterococcus: beyond vancomycin
use of antimicrobials in food animals is predicted to grow rapidly, resistance, Nat. Rev. Microbiol. 10 (4) (2012) 266–278, http://dx.doi.org/
by at least 67% from 2010 to 2030 [230]. In the BRICS countries (Bra- 10.1038/nrmicro2761.
[15] G.J. Armelagos, Disease in ancient Nubia, Science 163 (1969) 225–258, http://
zil, Russia, India, China, and South Africa) the antimicrobial con- dx.doi.org/10.1126/science.163.3864.255.
sumption by livestock will be doubled. The total amount of [16] G.L. Armstrong, L.A. Conn, R.W. Pinner, Trends in infectious disease mortality
antibiotics produced for this purpose will reach 105,596 ± 3605 tons in the United States during the 20th century, JAMA 281 (1) (1999) 61–66,
http://dx.doi.org/10.1001/jama.281.1.61.
in 2030 from 63,151 ± 1560 tons in 2010 [230]. It is estimated that
[17] M. Arthur, P.E. Reynolds, F. Depardieu, S. Evers, S. Dutka-Malen, R. Quintiliani
about a third of this increase will be due to the switch from exten- Jr, P. Courvalin, Mechanisms of glycopeptide resistance in enterococci, J.
sive farming systems to large-scale intensive farming operations Infect. 32 (1) (1996) 11–16, http://dx.doi.org/10.1016/S0163-4453(96)
that routinely use antimicrobials at sub-therapeutic concentrations 80003-X.
[18] Bacitracin. 2010. Available at: http://labeling.pfizer.com/ShowLabeling.aspx?
[230]. It is clear that there are certain economic drivers for this id=529.
development in livestock production. But should these considera- [19] F. Bager, M. Madsen, J. Christensen, F.M. Aarestrup, Avoparcin used as a
tions also include the costs that are eventually paid by the society growth promoter is associated with the occurrence of vancomycin-resistant
Enterococcus faecium on Danish poultry and pig farms, Prev. Vet. Med. 31 (1–
for the development of new antimicrobials, for more expensive 2) (1997) 95–112, http://dx.doi.org/10.1016/S0167-5877(96)01119-1.
treatment of MDR infections and, finally, for the higher morbidity [20] S. Bala, R. Khanna, M. Dadhwal, S.R. Prabagaran, S. Shivaji, J. Cullum, R. Lal,
and mortality rates imposed by MDR infections? These extremely Reclassification of Amycolatopsis mediterranei DSM 46095 as Amycolatopsis
rifamycinica sp. nov., Int. J. Syst. Evol. Microbiol. 54 (4) (2004) 1145–1149,
worrying trends dictate the need for global and coordinated actions http://dx.doi.org/10.1099/ijs.0.02901-0.
to prevent the overuse of antibiotics and the subsequent rise of [21] P. Baldry, The Battle Against Bacteria: A Fresh Look, Cambridge University
antibiotic resistance. Otherwise, the newly developed antimicro- Press, New York, NY, 1976. ISBN-13: 978-0521212687, ISBN-10:
0521212685.
bials may succumb to the same destiny as those ‘‘old” antimicro- [22] M. Balerna, W. Keller-Schierlein, C. Martius, H. Wolf, H. Zähner, Metabolic
bials described here. products of microorganisms. 72. Naphthomycin, an antimetabolite of vitamin
K, Arch. Mikrobiol. 65 (4) (1969) 303–317.
[23] R.H. Baltz, Daptomycin: mechanisms of action and resistance, and
biosynthetic engineering, Curr. Opin. Chem. Biol. 13 (2) (2009) 144–151,
Conflict of interest statement http://dx.doi.org/10.1016/j.cbpa.2009.02.031.
[24] M.R. Bardone, M. Paternoster, C. Coronelli, Teichomycins, new antibiotics
The author declares that the work was conducted in the absence from Actinoplanes teichomyceticus nov. sp. II. Extraction and chemical
characterization, J. Antibiot. (Tokyo) 31 (3) (1978) 170–177.
of any commercial or financial relationships that could be con- [25] C.E. Barnhart, J.C. Robertson, H.W. Miller, Virginiamycin, a new antibiotic for
strued as a potential conflict of interest. growing swine, J. Anim. Sci. 19 (1960) 9.
R. Aminov / Biochemical Pharmacology 133 (2017) 4–19 15

[26] A.E. Barnhill, M.T. Brewer, S.A. Carlson, Adverse effects of antimicrobials via [54] S. Chang, D.M. Sievert, J.C. Hageman, M.L. Boulton, F.C. Tenover, F.P. Downes,
predictable or idiosyncratic inhibition of host mitochondrial components, S. Shah, J.T. Rudrik, G.R. Pupp, W.J. Brown, D. Cardo, S.K. Fridkin, Infection
Antimicrob. Agents Chemother. 56 (8) (2012) 4046–4051, http://dx.doi.org/ with vancomycin-resistant Staphylococcus aureus containing the vanA
10.1128/AAC.00678-12. resistance gene, N. Engl. J. Med. 348 (14) (2003) 1342–1347, http://dx.doi.
[27] K. Bartmann, H. Iwainsky, H.H. Kleeberg, P. Mison, H.A. Offe, H. Otten, D. org/10.1056/NEJMoa025025.
Tettenborn, L. Trnka, Antituberculosis Drugs, in: K. Bartman (Ed.), Springer [55] J. Charney, W.P. Fisher, C. Curran, R.A. Machlowitz, A.A. Tytell, Streptogramin,
Science Business Media, 2013, p. 566. ISBN 3642728731, 9783642728730. a new antibiotic, Antibiot. Chemother. (Northfield) 3 (12) (1953)
[28] E.J. Bassett, M.S. Keith, G.J. Armelagos, D.L. Martin, A.R. Villanueva, 1283–1286.
Tetracycline-labelled human bone from ancient Sudanese Nubia (A.D. 350), [56] Chatham House Report, 2015. Towards a New Global Business Model for
Science 209 (1980) 1532–1534. Antibiotics: Delinking Revenues from Sales. Edited by C. Clift, U. Gopinathan,
[29] P.N. Batalha, M.C. de Souza, E. Peña-Cabrera, D.C. Cruz, F. da Costa Santos C. Morel, K. Outterson, J.-A. Røttingen, A. So. Available at: https://www.
Boechat, Quinolones in the search for new anticancer agents, Curr. Pharm. chathamhouse.org/sites/files/chathamhouse/field/field_document/20151009
Des. (2016). Jul 15 [Epub ahead of print]. NewBusinessModelAntibioticsCliftGopinathanMorelOuttersonRottingenSo.
[30] F.R. Batchelor, F.P. Doyle, J.H.C. Nayler, G.N. Rolinson, Synthesis of penicillin: pdf.
6-aminopenicillanic acid in penicillin fermentations, Nature 183 (1959) 257– [57] I. Chopra, M. Roberts, Tetracycline antibiotics: mode of action, applications,
258, http://dx.doi.org/10.1038/183257b0. molecular biology, and epidemiology of bacterial resistance, Microbiol. Mol.
[31] R. Bax, S. Green, Antibiotics: the changing regulatory and pharmaceutical Biol. Rev. 65 (2) (2001) 232–260, http://dx.doi.org/10.1128/MMBR.65.2.232-
industry paradigm, J. Antimicrob. Chemother. 70 (5) (2015) 1281–1284, 260.2001.
http://dx.doi.org/10.1093/jac/dku572. [58] C. Cocito, Antibiotics of the virginiamycin family, inhibitors which contain
[32] M. Bedin, A.M. Gaben, C.C. Saucier, J. Mester, Geldanamycin, an inhibitor of synergistic components, Microbiol. Rev. 43 (2) (1979) 145–192.
the chaperone activity of HSP90, induces MAPK-independent cell cycle arrest, [59] P.J. Collignon, Vancomycin-resistant enterococci and use of avoparcin in
Int. J. Cancer 109 (5) (2004) 643–652, http://dx.doi.org/10.1002/ijc.20010. animal feed: is there a link? Med. J. Aust. 171 (3) (1999) 144–146.
[33] A. Bernareggi, A. Borghi, M. Borgonovi, L. Cavenaghi, P. Ferrari, K. Vékey, M. [60] E.J. Corey, Drug Discovery Practices, Processes, and Perspectives, John Wiley &
Zanol, L.F. Zerilli, Teicoplanin metabolism in humans, Antimicrob. Agents Sons, Hoboken, N.J., 2013. p. 406. ISBN 9781118354469.
Chemother. 36 (8) (1992) 1744–1749. [61] CollaboratorsGlobal Burden of Disease Study 2013, Global, regional, and
[34] D. Bertrand, S. Bertrand, E. Neveu, P. Fernandes, Molecular characterization of national incidence, prevalence, and years lived with disability for 301 acute
off-target activities of telithromycin: a potential role for nicotinic and chronic diseases and injuries in 188 countries, 1990–2013: a systematic
acetylcholine receptors, Antimicrob. Agents Chemother. 54 (12) (2010) analysis for the Global Burden of Disease Study 2013, Lancet 386 (9995)
599–5402, http://dx.doi.org/10.1128/AAC.00840-10. (2013) 743–800, http://dx.doi.org/10.1016/S0140-6736(15)60692-4.
[35] E. Binda, F. Marinelli, G.L. Marcone, Old and new glycopeptide antibiotics: [62] M. Cook, E. Molto, C. Anderson, Fluorochrome labelling in Roman period
action and resistance, Antibiotics (Basel) 3 (4) (2014) 572–594, http://dx.doi. skeletons from Dakhleh Oasis, Egypt, Am. J. Phys. Anthropol. 80 (1989) 137–
org/10.3390/antibiotics3040572. 143, http://dx.doi.org/10.1002/ajpa.1330800202.
[36] R.D. Birkenmeyer, F. Kagan, Lincomycin. XI. Synthesis and structure of [63] B.M. Crowley, D.L. Boger, Total synthesis and evaluation of [Psi[CH2NH]
clindamycin, a potent antibacterial agent, J. Med. Chem. 13 (4) (1970) 616– Tpg4]vancomycin aglycon: reengineering vancomycin for dual D-Ala-D-Ala
619, http://dx.doi.org/10.1021/jm00298a007. and D-Ala-D-Lac binding, J. Am. Chem. Soc. 128 (9) (2006) 2885–2892, http://
[37] K.M. Bischoff, Y. Zhang, J.O. Rich, Fate of virginiamycin through the fuel dx.doi.org/10.1021/ja0572912.
ethanol production process, World J. Microbiol. Biotechnol. 32 (5) (2016) 76, [64] G. Delgado Jr., M.M. Neuhauser, D.T. Bearden, L.H. Danziger, Quinupristin-
http://dx.doi.org/10.1007/s11274-016-2026-3. dalfopristin: an overview, Pharmacotherapy 20 (2000) 1469–1485.
[38] I.G. Boneca, G. Chiosis, Vancomycin resistance: occurrence, mechanisms and [65] D.W. Denning, Echinocandins: a new class of antifungal, J. Antimicrob.
strategies to combat it, Expert Opin. Ther. Targets 7 (3) (2003) 311–328, Chemother. 49 (6) (2002) 889–891, http://dx.doi.org/10.1093/jac/dkf045.
http://dx.doi.org/10.1517/14728222.7.3.311. [66] C.A. Desjardins, K.A. Cohen, V. Munsamy, T. Abeel, K. Maharaj, B.J. Walker, T.P.
[39] H.W. Boucher, G.H. Talbot, J.S. Bradley, J.E. Edwards, D. Gilbert, L.B. Rice, M. Shea, D.V. Almeida, A.L. Manson, A. Salazar, N. Padayatchi, M.R. O’Donnell, K.
Scheld, B. Spellberg, J. Bartlett, Bad bugs, no drugs: no ESKAPE! An update P. Mlisana, J. Wortman, B.W. Birren, J. Grosset, A.M. Earl, A.S. Pym, Genomic
from the Infectious Diseases Society of America, Clin. Infect. Dis. 48 (2009) 1– and functional analyses of Mycobacterium tuberculosis strains implicate ald
12, http://dx.doi.org/10.1086/595011. in D-cycloserine resistance, Nat. Genet. 48 (5) (2016) 544–551, http://dx.doi.
[40] T. Bollenbach, Antimicrobial interactions: mechanisms and implications for org/10.1038/ng.3548.
drug discovery and resistance evolution, Curr. Opin. Microbiol. 27 (2015) 1–9, [67] M. Di Giambattista, G. Chinali, C. Cocito, The molecular basis of the inhibitory
http://dx.doi.org/10.1016/j.mib.2015.05.008. activities of type A and type B synergimycins and related antibiotics on
[41] S.J. Brickner, Oxazolidinone antibacterial agents, Curr. Pharm. Des. 2 (2) ribosomes, J. Antimicrob. Chemother. 4 (1989) 485–507, http://dx.doi.org/
(1996) 175–194. 10.1093/jac/24.4.485.
[42] H. Brockmann, W. Henkel, Pikromycin, ein neues Antibiotikum aus [68] G.P. Dinos, C.M. Athanassopoulos, D.A. Missiri, P.C. Giannopoulou, I.A.
Actinomycetes, Naturwissenschaften 37 (1950) 138–139. Vlachogiannis, G.E. Papadopoulos, D. Papaioannou, D.L. Kalpaxis,
[43] H. Brockmann, W. Henkel, Pikromycin, ein bitter schmeckendes Antibioticum Chloramphenicol derivatives as antibacterial and anticancer agents: historic
aus Actinomyceten, Antibiotica aus Actinomyceten 84 (1951) 184–288, problems and current solutions, Antibiotics (Basel) 5 (2) (2016). pii: E20. DOI:
http://dx.doi.org/10.1002/cber.19510840306. 10.3390/antibiotics5020020.
[44] I. Brook, Antimicrobials therapy of anaerobic infections, J. Chemother. 28 (3) [69] G. Domagk, Ein Beitrag zur Chemotherapie der bakteriellen Infektionen,
(2016) 143–150, http://dx.doi.org/10.1179/1973947815Y.0000000068. Deutsch. Med. Wschr. 61 (1935) 250, http://dx.doi.org/10.1055/s-0028-
[45] L.L. Brunton, J.S. Lazo, K. Parker, Protein synthesis inhibitors and 1129486.
miscellaneous antibacterial agents, in: Chapter 46. Goodman & Gilman’s [70] J.F. Dubern, S.P. Diggle, Quorum sensing by 2-alkyl-4-quinolones in
The Pharmacological Basis of Therapeutics, 11th ed., McGraw-Hill, New York, Pseudomonas aeruginosa and other bacterial species, Mol. BioSyst. 4 (2008)
2005, p. 2021. ISBN 0-07-142280-3. 882–888, http://dx.doi.org/10.1039/b803796p.
[46] R. Bucci, P. Galli, Vincenzo Tiberio: a misunderstood researcher, Ital. J. Public [71] B.M. Duggar, Aureomycin: a product of the continuing search for new
Health 8 (4) (2011) 404–406, http://dx.doi.org/10.2427/5688. antibiotics, Ann. N. Y. Acad. Sci. 51 (1948) 177–181, http://dx.doi.org/
[47] E.T. Burke, The arseno-therapy of syphilis; stovarsol, and tryparsamide, Br. J. 10.1111/j.1749-6632.1948.tb27262.x.
Vener. Dis. 1 (4) (1925) 321–338, http://dx.doi.org/10.1136/sti.1.4.321. [72] P. Ehrlich, S. Hata, Die Experimentelle Chemotherapie der Spirilosen, Julius
[48] B.M. Burkhart, R.M. Gassman, D.A. Langs, W.A. Pangborn, W.L. Duax, V. Springer, Berlin, 1910.
Pletnev, Gramicidin D conformation, dynamics and membrane ion transport, [73] N. Eliakim-Raz, A. Lador, Y. Leibovici-Weissman, M. Elbaz, M. Paul, L.
Biopolymers 51 (2) (1999) 129–144. DOI: 10.1002/(SICI)1097-0282(1999) Leibovici, Efficacy and safety of chloramphenicol: joining the revival of old
51:2<129::AID-BIP3>3.0.CO;2-Y. antibiotics? Systematic review and meta-analysis of randomized controlled
[49] K. Bush, P.A. Bradford, b-Lactams and b-lactamase inhibitors: an overview, in: trials, J. Antimicrob. Chemother. 70 (4) (2015) 979–996, http://dx.doi.org/
Cold Spring Harb. Perspect. Med., 2016, p. a025247, http://dx.doi.org/ 10.1093/jac/dku530.
10.1101/cshperspect.a025247. [74] G.M. Eliopoulos, Quinupristin-dalfopristin and linezolid: evidence and
[50] J.I. Castanon, History of the use of antibiotic as growth promoters in European opinion, Clin. Infect. Dis. 36 (2003) 473–481, http://dx.doi.org/10.1086/
poultry feeds, Poult. Sci. 86 (2007) 2466–2471, http://dx.doi.org/10.3382/ 367662.
ps.2007-00249. [75] V.I. Enne, P.M. Bennett, D.M. Livermore, L.M. Hall, Enhancement of host
[51] CDC, Centers for Disease Control and Prevention. Antibiotic resistance threats fitness by the sul2-coding plasmid p9123 in the absence of selective pressure,
in the United States, 2013. Available at: http://www. cdc.gov/drugresistance/ J. Antimicrob. Chemother. 53 (2004) 958–963, http://dx.doi.org/10.1093/jac/
pdf/ar-threats-2013-508.pdf. dkh217.
[52] J.Y. Cha, A. Ishiwata, S. Mobashery, A novel b-lactamase activity from a [76] EPC, Ban on Antibiotics as Growth Promoters in Animal Feed Enters into
penicillin-binding protein of Treponema pallidum and why syphilis is still Effect, European Commission – IP/05/1687, 2005. Available at: http://europa.
treatable with penicillin, J. Biol. Chem. 279 (2004) 14917–14921, http://dx. eu/rapid/press-release_IP-05-1687_en.htm.
doi.org/10.1074/jbc.M400666200. [77] G. Eschenauer, D.D. Depestel, P.L. Carver, Comparison of echinocandin
[53] E. Chain, H.W. Florey, A.D. Gardner, N.G. Heatley, M.A. Jennings, J. Orr-Ewing, antifungals, Ther. Clin. Risk Manag. 3 (1) (2007) 71–97, http://dx.doi.org/
A.G. Sanders, THE CLASSIC: penicillin as a chemotherapeutic agent. 1940, 10.2147/tcrm.2007.3.1.71.
Clin. Orthop. Relat. Res. 439 (2005) 23–26.
16 R. Aminov / Biochemical Pharmacology 133 (2017) 4–19

[78] M.E. Falagas, S.K. Kasiakou, Colistin: the revival of polymyxins for the [105] S.S. Hegde, M.W. Vetting, S.L. Roderick, L.A. Mitchenall, A. Maxwell, H.E.
management of multidrug-resistant gram-negative bacterial infections, Clin. Takiff, J.S. Blanchard, A fluoroquinolone resistance protein from
Infect. Dis. 40 (9) (2005) 1333–1341. Mycobacterium tuberculosis that mimics DNA, Science 308 (2005) 1480–
[79] M.E. Falagas, A.P. Grammatikos, A. Michalopoulos, Potential of old-generation 1483.
antibiotics to address current need for new antibiotics, Expert Rev. Anti- [106] H. Hiasa, M.E. Shea, DNA gyrase-mediated wrapping of the DNA strand is
infective Ther. 6 (5) (2008) 593–600, http://dx.doi.org/10.1586/ required for the replication fork arrest by the DNA gyrase-quinolone-DNA
14787210.6.5.593. ternary complex, J. Biol. Chem. 275 (2000) 34780–34786, http://dx.doi.org/
[80] FDA, Summary Report on Antimicrobials Sold or Distributed for Use in Food- 10.1074/jbc.M001608200.
Producing Animals. Department of Health and Human Services, 2009. [107] L.A. Hicks, T.H. Taylor Jr., R.J. Hunkler, U.S. outpatient antibiotic prescribing,
Available at: www.fda.gov/downloads/ForIndustry/UserFees/ 2010, N. Engl. J. Med. 368 (15) (2013) 1461–1462, http://dx.doi.org/10.1056/
AnimalDrugUserFeeActADUFA/UCM231851.pdf. NEJMc1212055.
[81] FDA, FDA News Release, 2012. Available at: (http://www.fda.gov/ [108] K. Hiramatsu, H. Hanaki, T. Ino, K. Yabuta, T. Oguri, F.C. Tenover, Methicillin-
NewsEvents/Newsroom/PressAnnouncements/ucm333695.htm). resistant Staphylococcus aureus clinical strain with reduced vancomycin
[82] FDA, FDA approves Sivextro to treat skin infections, 2014. Available at: http:// susceptibility, J. Antimicrob. Chemother. 40 (1) (1997) 135–136, http://dx.
www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm402174. doi.org/10.1093/jac/40.1.135.
htm. [109] F.A. Hochstein, C.R. Stephens, L.H. Conover, P.P. Regna, R. Pasternack, P.N.
[83] FDA, FDA Announces Pending Withdrawal of Approval of Nitarsone, 2015. Gordon, F.J. Pilgrim, K.J. Brunings, R.B. Woodward, The structure of
Available at: www.fda.gov/AnimalVeterinary/NewsEvents/CVMUpdates/ terramycin, J. Am. Chem. Soc. 75 (22) (1953) 5455–5475, http://dx.doi.org/
ucm440668.htm. 10.1021/ja01118a001.
[84] FDA, Summary Report on Antimicrobials Sold or Distributed for Use in Food- [110] J. Hummert, D. Van Gerven, Tetracycline-labeled human bone from a
Producing Animals, 2015. Available at: https://docs.google.com/viewer?url= medieval population in Nubia’s Batn El Hajar (550–1450 A.D.), Hum. Biol.
http%3A%2F%2Fwww.fda.gov%2Fdownloads%2FForIndustry%2FUserFees% 54 (2) (1982) 355–363.
2FAnimalDrugUserFeeActADUFA%2FUCM440584.pdf. [111] C.C. Hughes, W. Fenical, Antibacterials from the sea, Chemistry 16 (42) (2010)
[85] FDA, Fluoroquinolone Antibacterial Drugs: Drug Safety Communication – FDA 12512–12525, http://dx.doi.org/10.1002/chem.201001279.
Advises Restricting Use for Certain Uncomplicated Infections, 2016. Available [112] Z.M. Ištuk, A. Cikoš, D. Gembarovski, G. Lazarevski, I. Dilović, D. Matković-
at: http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsfor Čalogović, G. Kragol, Novel 9a,11-bridged azalides: One-pot synthesis of N0 -
HumanMedicalProducts/ucm500665.htm. substituted 2-imino-1,3-oxazolidines condensed to an azalide aglycone,
[86] A. Fleming, On antibacterial action of culture of penicillium, with special Bioorg. Med. Chem. 19 (2011) 556–566, http://dx.doi.org/10.1016/j.
reference to their use in isolation of B. influenzae, Br. J. Exp. Pathol. 10 (1929) bmc.2010.10.061.
226–236. [113] IUPAC, Compendium of Chemical Terminology, 2nd ed. (the ‘‘Gold Book”).
[87] H.G. Floss, T.W. Yu, Rifamycin-mode of action, resistance, and biosynthesis, Compiled by A.D. McNaught, A. Wilkinson. Blackwell Scientific Publications,
Chem. Rev. 105 (2) (2005) 621–632, http://dx.doi.org/10.1021/cr030112j. Oxford (1997). XML on-line corrected version: http://goldbook.iupac.org
[88] G. French, Safety and tolerability of linezolid, J. Antimicrob. Chemother. 51 (2006-) created by M. Nic, J. Jirat, B. Kosata; updates compiled by A. Jenkins.
(Suppl. 2) (2003) ii45–ii53, http://dx.doi.org/10.1093/jac/dkg253. ISBN 0-9678550-9-8. doi: 10.1351/goldbook.
[89] L. Friedman, J.D. Alder, J.A. Silverman, Genetic changes that correlate with [114] R.C. James, J.G. Pierce, A. Okano, J. Xie, D.L. Boger, Redesign of glycopeptide
reduced susceptibility to daptomycin in Staphylococcus aureus, Antimicrob. antibiotics: back to the future, ACS Chem. Biol. 7 (5) (2012) 797–804, http://
Agents Chemother. 50 (6) (2006) 2137–2145, http://dx.doi.org/10.1128/ dx.doi.org/10.1021/cb300007j.
AAC.00039-06. [115] H. Jenke-Kodama, A. Sandmann, R. Müller, E. Dittmann, Evolutionary
[90] S. Gardete, A. Tomasz, Mechanisms of vancomycin resistance in implications of bacterial polyketide synthases, Mol. Biol. Evol. 22 (10)
Staphylococcus aureus, J. Clin. Invest. 124 (7) (2014) 2836–2840, http://dx. (2005) 2027–2039, http://dx.doi.org/10.1093/molbev/msi193.
doi.org/10.1172/JCI68834. [116] R.N. Jones, S. Kohno, Y. Ono, J.E. Ross, K. Yanagihara, ZAAPS International
[91] N. Garrido-Mesa, A. Zarzuelo, J. Gálvez, Minocycline: far beyond an antibiotic, Surveillance Program (2007) for linezolid resistance: results from 5591 g-
Br. J. Pharmacol. 169 (2013) 337–352, http://dx.doi.org/10.1111/bph.12139. positive clinical isolates in 23 countries, Diagn. Microbiol. Infect. Dis. 64 (2)
[92] G.F. Gause, M.G. Brazhnikova, Gramicidin S and its use in the Treatment of (2009) 191–201, http://dx.doi.org/10.1016/j.diagmicrobio.2009.03.001.
Infected Wounds, Nature 154 (3918) (1944) 703, http://dx.doi.org/10.1038/ [117] K.A. Kazimierczak, K.P. Scott, D. Kelly, R.I. Aminov, Tetracycline resistome of
154703a0. the organic pig gut, Appl. Environ. Microbiol. 75 (2009) 1717–1722, http://dx.
[93] J.T. Good Jr, D.R. Rollins, R.J. Martin, Macrolides in the treatment of asthma, doi.org/10.1128/AEM.02206-08.
Curr. Opin. Pulm. Med. 18 (2012) 76–84, http://dx.doi.org/10.1097/ [118] C. Khosla, R.S. Gokhale, J.R. Jacobsen, D.E. Cane, Tolerance and specificity of
MCP.0b013e32834daff8. polyketide synthases, Annu. Rev. Biochem. 68 (1999) 219–253, http://dx.doi.
[94] H. Goossens, M. Ferech, R. Vander Stichele, Elseviers M, ESAC Project Group, org/10.1146/annurev.biochem.68.1.219.
Outpatient antibiotic use in Europe and association with resistance: a cross- [119] D.T. King, S. Sobhanifar, N.C. Strynadka, One ring to rule them all: current
national database study, Lancet 365 (2005) 579–587, http://dx.doi.org/ trends in combating bacterial resistance to the b-lactams, Protein Sci. 25 (4)
10.1016/S0140-6736(05)17907-0. (2016) 787–803, http://dx.doi.org/10.1002/pro.2889.
[95] D. Greenwood, Antimicrobial Drugs: Chronicle of a Twentieth Century [120] W. Kingston, Streptomycin, Schatz v. Waksman, and the balance of credit for
Medical Triumph, Oxford University Press, 2008, p. 464. ISBN discovery, J. Hist. Med. Allied Sci. 59 (3) (2004) 441–462, http://dx.doi.org/
9780199534845. 10.1093/jhmas/jrh091.
[96] R.D. Groves, A.M. Hetzel, Vital Statistics Rates in the United States, 1940- [121] J.D. Kittendorf, D.H. Sherman, The methymycin/pikromycin biosynthetic
1960, National Center for Health Statistics, Washington, DC, 1968, p. 1968 pathway: a model for metabolic diversity in natural product, Bioorg. Med.
(PHS publication no. 1677). Chem. 17 (2009) 2137–2146, http://dx.doi.org/10.1016/j.bmc.2008.10.082.
[97] R.N. Grüneberg, W. Hryniewicz, Clinical relevance of a European [122] K.P. Klinker, S.J. Borgert, Beyond vancomycin: the tail of the
collaborative study on comparative susceptibility of Gram-positive clinical lipoglycopeptides, Clin. Ther. 37 (12) (2015) 2619–2636, http://dx.doi.org/
isolates to teicoplanin and vancomycin, Int. J. Antimicrob. Agents 10 (4) 10.1016/j.clinthera.2015.11.007.
(1998) 271–277. [123] M. Klopper, R.M. Warren, C. Hayes, N.C. Gey van Pittius, E.M. Streicher, B.
[98] H. Hallen, H. Luo, J.S. Scott-Craig, J.D. Walton, A gene family encoding the Müller, F.A. Sirgel, M. Chabula-Nxiweni, E. Hoosain, G. Coetzee, P. David van
major toxins of lethal Amanita mushrooms, Proc. Natl. Acad. Sci. U.S.A. 104 Helden, T.C. Victor, A.P. Trollip, Emergence and spread of extensively and
(48) (2007) 19097–19101, http://dx.doi.org/10.1073/pnas.0707340104. totally drug-resistant tuberculosis, South Africa, Emerg. Infect. Dis. 9 (3)
[99] J.M. Hamilton-Miller, Chemistry and biology of the polyene macrolide (2013) 449–455, http://dx.doi.org/10.3201//EID1903.120246.
antibiotics, Bacteriol. Rev. 37 (2) (1973) 166–196. [124] F.E. Koehn, G.T. Carter, The evolving role of natural products in drug
[100] J. Hamilton-Miller, From foreign pharmacopoeias: ‘new’ antibiotics from old? discovery, Nat. Rev. Drug Discov. 4 (3) (2005) 206–220, http://dx.doi.org/
J. Antimicrob. Chemother. 27 (6) (1991) 702–705, http://dx.doi.org/ 10.1038/nrd1657.
10.1093/jac/27.6.702. [125] F. Kopp, M.A. Marahiel, Macrocyclization strategies in polyketide and
[101] R.E. Hancock, H.G. Sahl, Antimicrobial and host-defence peptides as new anti- nonribosomal peptide biosynthesis, Nat. Prod. Rep. 24 (4) (2007) 735–749,
infective therapeutic strategies, Nat. Biotechnol. 24 (2006) 1551–1557, http://dx.doi.org/10.1039/b613652b.
http://dx.doi.org/10.1038/nbt1267. [126] A. Koul, N. Dendouga, K. Vergauwen, B. Molenberghs, L. Vranckx, R.
[102] J.M. Harms, F. Schluenzen, P. Fucini, H. Bartels, A.E. Yonath, Alterations at the Willebrords, Z. Ristic, H. Lill, I. Dorange, J. Guillemont, D. Bald, K. Andries,
peptidyl transferase centre of the ribosome induced by the synergistic action Diarylquinolines target subunit c of mycobacterial ATP synthase, Nat. Chem.
of the streptogramins dalfopristin and quinupristin, BMC Biol. 2 (2004) 4, Biol. 3 (6) (2007) 323–324, http://dx.doi.org/10.1038/nchembio884.
http://dx.doi.org/10.1186/1741-7007-2-4. [127] K.M. Krause, A.W. Serio, T.R. Kane, L.E. Connolly, Aminoglycosides: an
[103] E.E. Hays, I.C. Wells, P.A. Katzman, C.K. Cain, F.A. Jacobs, S.A. Thayer, E.A. overview, Cold Spring Harb. Perspect. Med. 6 (6) (2016) a027029, http://dx.
Doisy, W.L. Gaby, E.C. Roberts, R.D. Muir, C.J. Carroll, L.R. Jones, N.J. Wade, doi.org/10.1101/cshperspect.a027029.
Antibiotic substances produced by Pseudomonas aeruginosa, J. Biol. Chem. 159 [128] V. Krishnasamy, J. Otte, E. Silbergeld, Antimicrobial use in Chinese swine and
(1945) 725–750. broiler poultry production, Antimicrob. Resist. Infect. Control 4 (2015) 17,
[104] S. Heeb, M.P. Fletcher, S.R. Chhabra, S.P. Diggle, P. Williams, M. Cámara, http://dx.doi.org/10.1186/s13756-015-0050-y.
Quinolones: from antibiotics to autoinducers, FEMS Microbiol. Rev. 35 (2) [129] S. Kudoh, T. Uetake, K. Hagiwara, M. Hirayama, L.H. Hus, H. Kimura, Y.
(2011) 247–274, http://dx.doi.org/10.1111/j.1574-6976.2010.00247.x. Sugiyama, Clinical effects of low-dose long-term erythromycin
R. Aminov / Biochemical Pharmacology 133 (2017) 4–19 17

chemotherapy on diffuse panbronchiolitis, Nihon Kyobu Shikkan Gakkai [144] L. Lutz, D.C. Pereira, R.M. Paiva, A.P. Zavascki, A.L. Barth, Macrolides decrease
Zasshi 25 (1987) 632–642. the minimal inhibitory concentration of anti-pseudomonal agents against
[130] K.K. Kumarasamy, M.A. Toleman, T.R. Walsh, J. Bagaria, F. Butt, R. Pseudomonas aeruginosa from cystic fibrosis patients in biofilm, BMC
Balakrishnan, U. Chaudhary, M. Doumith, C.G. Giske, S. Irfan, P. Krishnan, A. Microbiol. 12 (2012) 196, http://dx.doi.org/10.1186/1471-2180-12-196.
V. Kumar, S. Maharjan, S. Mushtaq, T. Noorie, D.L. Paterson, A. Pearson, C. [145] A.J. MacLeod, H.B. Ross, R.L. Ozere, Geo. Digout, C.E. van Rooyen, Lincomycin:
Perry, R. Pike, B. Rao, U. Ray, J.B. Sarma, M. Sharma, E. Sheridan, M.A. a new antibiotic active against staphylococci and other gram-positive cocci,
Thirunarayan, J. Turton, S. Upadhyay, M. Warner, W. Welfare, D.M. Livermore, Can. Med. Assoc. J. 91 (20) (1964) 1056–1060.
N. Woodford, Emergence of a new antibiotic resistance mechanism in India, [146] I.A. MacNeil, C.L. Tiong, C. Minor, P.R. August, T.H. Grossman, K.A. Loiacono, B.
Pakistan, and the UK: a molecular, biological, and epidemiological study, A. Lynch, T. Phillips, S. Narula, R. Sundaramoorthi, A. Tyler, T. Aldredge, H.
Lancet Infect. Dis. 10 (2010) 597–602, http://dx.doi.org/10.1016/S1473-3099 Long, M. Gilman, D. Holt, M.S. Osburne, Expression and isolation of
(10)70143-2. antimicrobial small molecules from soil DNA libraries, J. Mol. Microbiol.
[131] B. Kushner, P.D. Allen, B.T. Crane, Frequency and demographics of gentamicin Biotechnol. 3 (2001) 301–308.
use, Otol. Neurotol. 37 (2) (2016) 190–195, http://dx.doi.org/10.1097/ [147] A. Makovitzki, D. Avrahami, Y. Shai, Ultrashort antibacterial and antifungal
MAO.0000000000000937. lipopeptides, Proc. Natl. Acad. Sci. U.S.A. 103 (2006) 15997–16002, http://dx.
[132] T.F. Landers, B. Cohen, T.E. Wittum, E.L. Larson, A review of antibiotic use in doi.org/10.1073/pnas.0606129103.
food animals: perspective, policy, and potential, Public Health Rep. 127 (1) [148] J. Mahoney, R. Arnold, A. Harris, Penicillin treatment of early syphilis: a
(2012) 4–22. preliminary report, Am. J. Public Health Nations Health 33 (12) (1943) 1387–
[133] P.A. Lawrence, Rank injustice, Nature 415 (6874) (2002) 835–836, http://dx. 1391.
doi.org/10.1038/415835a. [149] P. Margalith, G. Beretta, Rifomycin. XI. Taxonomic study on Streptomyces
[134] M.P. Lechevalier, H. Prauser, D.P. Labeda, J.S. Ruan, Two new genera of mediterranei nov. sp., Mycopathol. Mycol. Appl. 13 (4) (1960) 321–330,
nocardioform actinomycetes: Amycolata gen. nov. and Amycolatopsis gen. http://dx.doi.org/10.1007/BF02089930.
nov, Int. J. Syst. Evol. Microbiol. 36 (1) (1986) 29–37, http://dx.doi.org/ [150] A. Marsot, R. Guilhaumou, C. Riff, O. Blin, Amikacin in critically ill patients: a
10.1099/00207713-36-1-29. review of population pharmacokinetic studies, Clin. Pharmacokinet. (2016).
[135] R. Leclercq, Mechanisms of resistance to macrolides and lincosamides: nature Jun 21. [Epub ahead of print].
of the resistance elements and their clinical implications, Clin. Infect. Dis. 34 [151] F. Martinez-Sandoval, C.D. Ericsson, Z.D. Jiang, P.C. Okhuysen, J.H. Romero, N.
(4) (2002) 482–492, http://dx.doi.org/10.1086/324626. Hernandez, W.P. Forbes, A. Shaw, E. Bortey, H.L. DuPont, Prevention of
[136] G.C. Lee, K.R. Reveles, R.T. Attridge, K.A. Lawson, I.A. Mansi, J.S. Lewis 2nd, C.R. travelers’ diarrhea with rifaximin in US travelers to Mexico, J. Travel Med. 17
Frei, Outpatient antibiotic prescribing in the United States: 2000 to 2010, (2) (2010) 111–117, http://dx.doi.org/10.1111/j.1708-8305.2009.00385.x.
BMC Med. 12 (2014) 96, http://dx.doi.org/10.1186/1741-7015-12-96. [152] C.V. Maxwell, D.S. Buchanan, W.G. Luce, D. McLaren, R. Vend, Direct
[137] D. Levine, Vancomycin: a history, Clin. Infect. Dis. 42 (2006) S5–S12, http:// Comparisons of Antibiotics for Growing-Finishing Swine. Animal Science
dx.doi.org/10.1086/491709. Research Report, Oklahoma Agricultural Experiment Station, 235-240, 1981.
[138] B. Llano-Sotelo, J. Dunkle, D. Klepacki, W. Zhang, P. Fernandes, J.H. Cate, A.S. Available at: http://beefextension.com/research_reports/research_56_94/
Mankin, Binding and action of CEM-101, a new fluoroketolide antibiotic that rr82/rr82_55.pdf.
inhibits protein synthesis, Antimicrob. Agents Chemother. 54 (12) (2010) [153] J.M. McGuire, R.L. Bunch, R.C. Anderson, H.E. Boaz, E.H. Flynn, H.M. Powell, J.
4961–4970, http://dx.doi.org/10.1128/AAC.00860-10. W. Smith, Ilotycin, a new antibiotic, Antibiot. Chemother. (Northfield) 2 (6)
[139] R. Lindsay, H. Zhou, F. Cosman, J. Nieves, D. Dempster, Double and quadruple (1952) 281–283.
tetracycline labelling of bone: impact of the label itself, J. Bone Miner. Res. 28 [154] G.B. Migliori, G. De Iaco, G. Besozzi, R. Centis, D.M. Cirillo, First tuberculosis
(1) (2013) 222–223, http://dx.doi.org/10.1002/jbmr.1818. cases in Italy resistant to all tested drugs, Euro. Surveill. 12 (20) (2007).
[140] Y.Y. Liu, Y. Wang, T.R. Walsh, L.X. Yi, R. Zhang, J. Spencer, Y. Doi, G. Tian, B. pii=3194. Available at: http://www.eurosurveillance.org/ViewArticle.aspx?
Dong, X. Huang, L.F. Yu, D. Gu, H. Ren, X. Chen, L. Lv, D. He, H. Zhou, Z. Liang, J. ArticleId=3194.
H. Liu, J. Shen, Emergence of plasmid-mediated colistin resistance [155] R.C. Moellering, Linezolid: the first oxazolidinone antimicrobial, Ann. Intern.
mechanism MCR-1 in animals and human beings in China: a Med. 138 (2) (2003) 135–142, http://dx.doi.org/10.7326/0003-4819-138-2-
microbiological and molecular biological study, Lancet Infect. Dis. 16 (2) 200301210-00015.
(2016) 161–168, http://dx.doi.org/10.1016/S1473-3099(15)00424-7. [156] M.I. Morris, M. Villmann, Echinocandins in the management of invasive
[141] D.M. Livermore, Linezolid in vitro: mechanism and antibacterial spectrum, J. fungal infections, part 1, Am. J. Health Syst. Pharm. 63 (18) (2006) 1693–
Antimicrob. Chemother. 51 (Suppl. 2) (2003) ii9–ii16, http://dx.doi.org/ 1703, http://dx.doi.org/10.2146/ajhp050464.p1.
10.1093/jac/dkg249. [157] T.A. Mukhtar, K.P. Koteva, G.D. Wright, Chimeric streptogramin-tyrocidine
[142] N.C. Lloyd, H.W. Morgan, B.K. Nicholson, R.S. Ronimus, The composition of antibiotics that overcome streptogramin resistance, Chem. Biol. 12 (2) (2005)
Ehrlich’s salvarsan: resolution of a century-old debate, Angew. Chem. Int. Ed. 229–235, http://dx.doi.org/10.1016/j.chembiol.2004.12.009.
Engl. 44 (2005) 941–944, http://dx.doi.org/10.1002/anie.200461471. [158] S. Murphy, R.J. Pinney, Teicoplanin or vancomycin in the treatment of gram-
[143] R. Lozano, M. Naghavi, K. Foreman, S. Lim, K. Shibuya, V. Aboyans, J. positive infections? J. Clin. Pharm. Ther. 20 (1) (1995) 5–11.
Abraham, T. Adair, R. Aggarwal, S.Y. Ahn, M. Alvarado, H.R. Anderson, L.M. [159] S. Mutak, Azalides from azithromycin to new azalide derivatives, J. Antibiot.
Anderson, K.G. Andrews, C. Atkinson, L.M. Baddour, S. Barker-Collo, D.H. (Tokyo) 60 (2007) 85–122, http://dx.doi.org/10.1038/ja.2007.10.
Bartels, M.L. Bell, E.J. Benjamin, D. Bennett, K. Bhalla, B. Bikbov, A. Bin [160] H. Nagai, H. Shishido, R. Yoneda, E. Yamaguchi, A. Tamura, A. Kurashima,
Abdulhak, G. Birbeck, F. Blyth, I. Bolliger, S. Boufous, C. Bucello, M. Burch, P. Long-term low-dose administration of erythromycin to patients with diffuse
Burney, J. Carapetis, H. Chen, D. Chou, S.S. Chugh, L.E. Coffeng, S.D. Colan, S. panbronchiolitis, Respiration 58 (1991) 145–149.
Colquhoun, K.E. Colson, J. Condon, M.D. Connor, L.T. Cooper, M. Corriere, M. [161] B.A. Napier, E.M. Burd, S.W. Satola, S.M. Cagle, S.M. Ray, P. McGann, J. Pohl, E.
Cortinovis, K.C. de Vaccaro, W. Couser, B.C. Cowie, M.H. Criqui, M. Cross, K. P. Lesho, D.S. Weiss, Clinical use of colistin induces cross-resistance to host
C. Dabhadkar, N. Dahodwala, D. De Leo, L. Degenhardt, A. Delossantos, J. antimicrobials in Acinetobacter baumannii, MBio 4 (3) (2013) e00021 13,
Denenberg, D.C. Des Jarlais, S.D. Dharmaratne, E.R. Dorsey, T. Driscoll, H. http://dx.doi.org/10.1128/mBio.00021-13.
Duber, B. Ebel, P.J. Erwin, P. Espindola, M. Ezzati, V. Feigin, A.D. Flaxman, [162] M.L. Nelson, A. Dinardo, J. Hochberg, G.J. Armelagos, Brief communication:
M.H. Forouzanfar, F.G. Fowkes, R. Franklin, M. Fransen, M.K. Freeman, S.E. mass spectroscopic characterization of tetracycline in the skeletal remains of
Gabriel, E. Gakidou, F. Gaspari, R.F. Gillum, D. Gonzalez-Medina, Y.A. an ancient population from Sudanese Nubia 350–550 CE, Am. J. Phys.
Halasa, D. Haring, J.E. Harrison, R. Havmoeller, R.J. Hay, B. Hoen, P.J. Hotez, Anthropol. 143 (2010) 151–154, http://dx.doi.org/10.1002/ajpa.21340.
D. Hoy, K.H. Jacobsen, S.L. James, R. Jasrasaria, S. Jayaraman, N. Johns, G. [163] M.L. Nelson, S.B. Levy, The history of the tetracyclines, Ann. N. Y. Acad. Sci.
Karthikeyan, N. Kassebaum, A. Keren, J.P. Khoo, L.M. Knowlton, O. 1241 (2011) 17–32, http://dx.doi.org/10.1111/j.1749-6632.2011.06354.x.
Kobusingye, A. Koranteng, R. Krishnamurthi, M. Lipnick, S.E. Lipshultz, S.L. [164] S.R. Norrby, C.E. Nord, R. Finch, European Society of Clinical Microbiology and
Ohno, J. Mabweijano, M.F. MacIntyre, L. Mallinger, L. March, G.B. Marks, R. Infectious Diseases, Lack of development of new antimicrobial drugs: a
Marks, A. Matsumori, R. Matzopoulos, B.M. Mayosi, J.H. McAnulty, M.M. potential serious threat to public health, Lancet Infect Dis. 5 (2005) 115–119,
McDermott, J. McGrath, G.A. Mensah, T.R. Merriman, C. Michaud, M. Miller, http://dx.doi.org/10.1016/S1473-3099(05)01283-1.
T.R. Miller, C. Mock, A.O. Mocumbi, A.A. Mokdad, A. Moran, K. Mulholland, [165] A. Okano, A. Nakayama, A.W. Schammel, D.L. Boger, Total synthesis of [W[C
M.N. Nair, L. Naldi, K.M. Narayan, K. Nasseri, P. Norman, M. O’Donnell, S.B. (@NH)NH]Tpg(4)]vancomycin and its (4-chlorobiphenyl)methyl derivative:
Omer, K. Ortblad, R. Osborne, D. Ozgediz, B. Pahari, J.D. Pandian, A.P. Rivero, impact of peripheral modifications on vancomycin analogues redesigned for
R.P. Padilla, F. Perez-Ruiz, N. Perico, D. Phillips, K. Pierce, C.A. Pope 3rd, E. dual D-Ala-D-Ala and D-Ala-D-Lac binding, J. Am. Chem. Soc. 136 (39) (2014)
Porrini, F. Pourmalek, M. Raju, D. Ranganathan, J.T. Rehm, D.B. Rein, G. 13522–13525, http://dx.doi.org/10.1021/ja507009a.
Remuzzi, F.P. Rivara, T. Roberts, F.R. De León, L.C. Rosenfeld, L. Rushton, R.L. [166] A. Okano, A. Nakayama, K. Wu, E.A. Lindsey, A.W. Schammel, Y. Feng, K.C.
Sacco, J.A. Salomon, U. Sampson, E. Sanman, D.C. Schwebel, M. Segui- Collins, D.L. Boger, Total syntheses and initial evaluation of [W[C(@S)NH]
Gomez, D.S. Shepard, D. Singh, J. Singleton, K. Sliwa, E. Smith, A. Steer, J.A. Tpg4]vancomycin, [W[C(@NH)NH]Tpg4]vancomycin, [W[CH₂NH]
Taylor, B. Thomas, I.M. Tleyjeh, J.A. Towbin, T. Truelsen, E.A. Undurraga, N. Tpg4]vancomycin, and their (4-chlorobiphenyl)methyl derivatives:
Venketasubramanian, L. Vijayakumar, T. Vos, G.R. Wagner, M. Wang, W. synergistic binding pocket and peripheral modifications for the
Wang, K. Watt, M.A. Weinstock, R. Weintraub, J.D. Wilkinson, A.D. Woolf, S. glycopeptide antibiotics, J. Am. Chem. Soc. 137 (10) (2015) 3693–3704,
Wulf, P.H. Yeh, P. Yip, A. Zabetian, Z.J. Zheng, A.D. Lopez, C.J. Murray, M.A. http://dx.doi.org/10.1021/jacs.5b01008.
AlMazroa, Z.A. Memish, Global and regional mortality from 235 causes of [167] M. Oliynyk, M. Samborskyy, J.B. Lester, T. Mironenko, N. Scott, S. Dickens, S.F.
death for 20 age groups in 1990 and 2010: a systematic analysis for the Haydock, P.F. Leadlay, Complete genome sequence of the erythromycin-
Global Burden of Disease Study 2010, Lancet 380 (9859) (2012) 2095– producing bacterium Saccharopolyspora erythraea NRRL23338, Nat.
2128, http://dx.doi.org/10.1016/S0140-6736(12)61728-0. Biotechnol. 25 (2007) 447–453, http://dx.doi.org/10.1038/nbt1297.
18 R. Aminov / Biochemical Pharmacology 133 (2017) 4–19

[168] J. O’Neill (Chair), The Review on Antimicrobial Resistance, 2014. Available at: [192] J.D. Rothstein, S. Patel, M.R. Regan, C. Haenggeli, Y.H. Huang, D.E. Bergles, L.
http://amr-review.org/sites/default/files/AMR%20Review%20Paper%20-% Jin, M. Dykes Hoberg, S. Vidensky, D.S. Chung, S.V. Toan, L.I. Bruijn, Z.Z. Su, P.
20Tackling%20a%20crisis%20for%20the%20health%20and%20wealth%20of% Gupta, P.B. Fisher, Beta-lactam antibiotics offer neuroprotection by
20nations_1.pdf. increasing glutamate transporter expression, Nature 433 (2005) 73–77.
[169] J.K. Ortwine, K.S. Kaye, J. Li, J.M. Pogue, Colistin: understanding and applying [193] D.M. Rothstein, J. van Duzer, A. Sternlicht, S.C. Gilman, Rifalazil and other
recent pharmacokinetic advances, Pharmacotherapy 35 (1) (2015) 11–16, benzoxazinorifamycins in the treatment of chlamydia-based persistent
http://dx.doi.org/10.1002/phar.1484. infections, Arch. Pharm. (Weinheim) 340 (10) (2007) 517–529.
[170] K. Outterson, J.H. Rex, T. Jinks, P. Jackson, J. Hallinan, S. Karp, D.T. Hung, F. [194] M.J. Rybak, The efficacy and safety of daptomycin: first in a new class of
Franceschi, T. Merkeley, C. Houchens, D.M. Dixon, M.G. Kurilla, R. Aurigemma, antibiotics for Gram-positive bacteria, Clin. Microbiol. Infect. 12 (Suppl. 1)
J. Larsen, Accelerating global innovation to address antibacterial resistance: (2006) 24–32.
introducing CARB-X, Nat. Rev. Drug Discov. 15 (9) (2016) 589–590, http://dx. [195] R.B. Sartor, Review article: the potential mechanisms of action of rifaximin in
doi.org/10.1038/nrd.2016.155. the management of inflammatory bowel diseases, Aliment. Pharmacol. Ther.
[171] J. Parascandola, The history of antibiotics: a symposium. American Institute 43 (Suppl. 1) (2016) 27–36, http://dx.doi.org/10.1111/apt.13436.
of the History of Pharmacy No. 5, 1980. ISBN 0-931292-08-5. [196] N. Scheinfeld, Telithromycin: a brief review of a new ketolide antibiotic, J.
[172] F. Parenti, G. Beretta, M. Berti, V. Arioli, Teichomycins, new antibiotics from Drug Dermatol. 3 (2004) 409–413.
Actinoplanes teichomyceticus Nov. Sp. I. Description of the producer strain, [197] S. Schwarz, C. Kehrenberg, B. Doublet, A. Cloeckaert, Molecular basis of
fermentation studies and biological properties, J. Antibiot. (Tokyo) 31 (4) bacterial resistance to chloramphenicol and florfenicol, FEMS Microbiol. Rev.
(1978) 276–283. 28 (5) (2004) 519–542.
[173] F. Parenti, G.C. Schito, P. Courvalin, Teicoplanin chemistry and microbiology, [198] J. Seema, A. Lleras-Muney, K.V. Smith, Modern medicine and the twentieth
J. Chemother. 12 (Suppl. 5) (2000) 5–14. century decline in mortality: evidence on the impact of sulfa drugs, Am.
[174] Pfizer, 2010. Available at: https://docs.google.com/viewer?url=http%3A%2F% Econ. J.: Appl. Econ. 2 (2) (2010) 118–146, http://dx.doi.org/10.1257/
2Fwww.accessdata.fda.gov%2Fdrugsatfda_docs%2Flabel%2F2010% app.2.2.118.
2F021130s022lbl.pdf. [199] T.A.R. Seemungal, T.M.A. Wilkinson, J.R. Hurst, W.R. Perera, R.J. Sapsford, J.A.
[175] B.B. Plikaytis, J.L. Marden, J.T. Crawford, C.L. Woodley, W.R. Butler, T.M. Wedzicha, Long-term erythromycin therapy is associated with decreased
Shinnick, Multiplex PCR assay specific for the multidrug-resistant strain W of chronic obstructive pulmonary disease exacerbations, Am. J. Respir. Crit. Care
Mycobacterium tuberculosis, J. Clin. Microbiol. 32 (1994) 1542–1546. Med. 178 (2008) 1139–1147, http://dx.doi.org/10.1164/rccm.200801-145OC.
[176] J. Pogliano, N. Pogliano, J.A. Silverman, Daptomycin-mediated reorganization [200] P. Sensi, History of the development of rifampin, Rev. Infect. Dis. 5 (Suppl. 3)
of membrane architecture causes mislocalization of essential cell division (1983) S402–S406, http://dx.doi.org/10.1093/clinids/5.Supplement_3.S402.
proteins, J. Bacteriol. 194 (17) (2012) 4494–4504, http://dx.doi.org/10.1128/ [201] K.A. Sepkowitz, J. Rafalli, L. Riley, T.E. Kiehn, D. Armstrong, Tuberculosis in the
JB.00011-12. AIDS era, Clin. Microbiol. Rev. 8 (2) (1995) 180–199.
[177] F.R. Ponziani, S. Pecere, L. Lopetuso, F. Scaldaferri, G. Cammarota, A. [202] D.J. Shapiro, L.A. Hicks, A.T. Pavia, A.L. Hersh, Antibiotic prescribing for adults
Gasbarrini, Rifaximin for the treatment of irritable bowel syndrome – a in ambulatory care in the USA, 2007–09, J. Antimicrob. Chemother. 69 (1)
drug safety evaluation, Expert Opin. Drug Saf. 15 (7) (2016) 983–991, http:// (2014) 234–240, http://dx.doi.org/10.1093/jac/dkt301.
dx.doi.org/10.1080/14740338.2016.1186639. [203] Y. Shigeri, R.P. Seal, K. Shimamoto, Molecular pharmacology of glutamate
[178] J. Pootoolal, J. Neu, G.D. Wright, Glycopeptide antibiotic resistance, Annu. transporters, EAATs and VGLUTs, Brain Res. Rev. 45 (2004) 250–265, http://
Rev. Pharmacol. Toxicol. 42 (2002) 381–408, http://dx.doi.org/10.1146/ dx.doi.org/10.1016/j.brainresrev.2004.04.004.
annurev.pharmtox.42.091601.142813. [204] D. Shinabarger, Mechanism of action of the oxazolidinone antibacterial
[179] E.J. Prenner, R.N. Lewis, K.C. Neuman, S.M. Gruner, L.H. Kondejewski, R.S. agents, Expert Opin. Investig. Drugs 8 (8) (1999) 1195–1202, http://dx.doi.
Hodges, R.N. McElhaney, Nonlamellar phases induced by the interaction of org/10.1517/13543784.8.8.1195.
gramicidin S with lipid bilayers. A possible relationship to membrane- [205] M. Shnayerson, M. Plotkin, The Killers Within: The Deadly Rise of Drug-
disrupting activity, Biochemistry 36 (25) (1997) 7906–7916, http://dx.doi. Resistant Bacteria, Back Bay Books, 2003. ISBN 978-0-316-73566-7.
org/10.1021/bi962785k. [206] A.M. Slee, M.A. Wuonola, R.J. McRipley, I. Zajac, M.J. Zawada, P.T.
[180] G.A. Prosser, L.P. de Carvalho, Kinetic mechanism and inhibition of Bartholomew, W.A. Gregory, M. Forbes, Oxazolidinones, a new class of
Mycobacterium tuberculosis d-alanine: D-alanine ligase by the antibiotic d- synthetic antibacterial agents: in vitro and in vivo activities of DuP 105 and
cycloserine, FEBS J. 280 (4) (2013) 1150–1166, http://dx.doi.org/10.1111/ DuP 721, Antimicrob. Agents Chemother. 31 (11) (1987) 1791–1797, http://
febs.12108. dx.doi.org/10.1128/AAC.31.11.1791.
[181] H. Rahman, B. Austin, W.J. Mitchell, P.C. Morris, D.J. Jamieson, D.R. Adams, A. [207] A. Soriano, O. Miró, J. Mensa, Mitochondrial toxicity associated with linezolid,
M. Spragg, M. Schweizer, Novel anti-infective compounds from marine N. Engl. J. Med. 353 (21) (2005) 2305–2306, http://dx.doi.org/10.1056/
bacteria, Mar. Drugs 8 (2010) 498–518, http://dx.doi.org/10.3390/ NEJM200511243532123.
md8030498. [208] P. Spanogiannopoulos, N. Waglechner, K. Koteva, G.D. Wright, A rifamycin
[182] S. Ramaswamy, J.M. Musser, Molecular genetic basis of antimicrobial agent inactivating phosphotransferase family shared by environmental and
resistance in Mycobacterium tuberculosis: 1998 update, Tuber. Lung Dis. 79 pathogenic bacteria, Proc. Natl. Acad. Sci. U.S.A. 111 (19) (2014) 7102–
(1) (1998) 3–29, http://dx.doi.org/10.1054/tuld.1998.0002. 7107, http://dx.doi.org/10.1073/pnas.1402358111.
[183] G.G. Rao, N.S. Ly, C.E. Haas, S. Garonzik, A. Forrest, J.B. Bulitta, P.A. Kelchlin, P. [209] B.S. Speer, L. Bedzyk, A.A. Salyers, Evidence that a novel tetracycline
N. Holden, R.L. Nation, J. Li, B.T. Tsuji, New dosing strategies for an old resistance gene found on two Bacteroides transposons encodes an NADP-
antibiotic: pharmacodynamics of front-loaded regimens of colistin at requiring oxidoreductase, J. Bacteriol. 173 (1) (1991) 176–183.
simulated pharmacokinetics in patients with kidney or liver disease, [210] C.M. Spencer, H.M. Bryson, Teicoplanin. A pharmacoeconomic evaluation of
Antimicrob. Agents Chemother. 58 (3) (2014) 1381–1388, http://dx.doi.org/ its use in the treatment of gram-positive infections, Pharmacoeconomics 7
10.1128/AAC.00327-13. (4) (1995) 357–374.
[184] G.S. Redin, Antibacterial activity in mice of minocycline, a new tetracycline, [211] C.H. Stammer, A.N. Wilson, F.W. Holly, K. Folkers, Synthesis of D-4-amino-3-
Antimicrob. Agents Chemother. 6 (1966) 371–376. isoxazolidinone, J. Am. Chem. Soc. 77 (1955) 2346–2347, http://dx.doi.org/
[185] L.B. Rice, Federal funding for the study of antimicrobial resistance in 10.1021/ja01613a107.
nosocomial pathogens: no ESKAPE, J. Infect. Dis. 197 (2009) 1079–1081, [212] J.N. Steenbergen, J. Alder, G.M. Thorne, F.P. Tally, Daptomycin: a lipopeptide
http://dx.doi.org/10.1086/533452. antibiotic for the treatment of serious Gram-positive infections, J.
[186] Kenneth L. Rinehart Jr., Waltraut M. Knoll, Katsumi Kakinuma, Frederick J. Antimicrob. Chemother. 55 (3) (2005) 283–288, http://dx.doi.org/
Antosz, Iain C. Paul, Andrew H.J. Wang, Fritz Reusser, L.H. Li, William C. 10.1093/jac/dkh546.
Krueger, Chemistry and biochemistry of the streptovaricins. XII. [213] S.H.E. Kaufmann, Paul Ehrlich: founder of chemotherapy, Nat. Rev. Drug
Atropisomeric streptovaricins, J. Am. Chem. Soc. 97 (1) (1975) 196–198, Discov. 7 (2008) 373, http://dx.doi.org/10.1038/nrd2582.
http://dx.doi.org/10.1021/ja00834a039. [214] D. Steverding, The development of drugs for treatment of sleeping sickness: a
[187] K.D. Roberts, R.M. Sulaiman, M.J. Rybak, Dalbavancin and oritavancin: an historical review, Parasit. Vectors 3 (1) (2010) 15, http://dx.doi.org/10.1186/
innovative approach to the treatment of gram-positive infections, 1756-3305-3-15.
Pharmacotherapy 35 (10) (2015) 935–948, http://dx.doi.org/10.1002/phar. [215] D.R. Storm, K.S. Rosenthal, P.E. Swanson, Polymyxin and related peptide
[188] G.T. Robertson, E.J. Bonventre, T.B. Doyle, In vitro evaluation of CBR-2092, a antibiotics, Annu. Rev. Biochem. 46 (1977) 723–763, http://dx.doi.org/
novel rifamycin-quinolone hybrid antibiotic: studies of the mode of action in 10.1146/annurev.bi.46.070177.003451.
Staphylococcus aureus, Antimicrob. Agents Chemother. 52 (2008) [216] T. Sugimoto, T. Tanikawa, K. Suzuki, Y. Yamasaki, Synthesis and structure-
2313–2323. activity relationship of a novel class of 15-membered macrolide antibiotics
[189] I.M. Rollo, J. Williamson, R.L. Plackett, Acquired resistance to penicillin and to known as ‘11a-azalides’, Bioorg. Med. Chem. 20 (2012) 5787–5801, http://dx.
neoarsphenamine in Spirochaeta recurrentis, Br. J. Pharmacol. Chemother. 7 doi.org/10.1016/j.bmc.2012.08.007.
(1952) 33–41. [217] G.H. Talbot, J. Bradley, J.E. Edwards Jr, D. Gilbert, M. Scheld, J.G. Bartlett, Bad
[190] W. Rose, M. Rybak, Tigecycline: first of a new class of antimicrobial agents, bugs need drugs: an update on the development pipeline from the
Pharmacotherapy 26 (8) (2006) 1099–1110, http://dx.doi.org/10.1592/ Antimicrobial Availability Task Force of the Infectious Diseases Society of
phco.26.8.1099. America, Clin. Infect. Dis. 42 (2006) 657–668, http://dx.doi.org/10.1086/
[191] R. Roskoski, W. Gevers, H. Kleinkauf, F. Lipmann, Tyrocidine biosynthesis by 499819.
three complementary fractions from Bacillus brevis (ATCC 8185), [218] F.P. Tally, M.F. DeBruin, Development of daptomycin for gram-positive
Biochemistry 9 (25) (1970) 4839–4845, http://dx.doi.org/10.1021/ infections, J. Antimicrob. Chemother. 46 (4) (2000) 523–526, http://dx.doi.
bi00827a002. org/10.1093/jac/46.4.523.
R. Aminov / Biochemical Pharmacology 133 (2017) 4–19 19

[219] T. Tenson, M. Lovmar, M. Ehrenberg, The mechanism of action of macrolides, [234] M.A. Wainwright, Response to William Kingston, ‘‘Streptomycin, Schatz
lincosamides and streptogramin B reveals the nascent peptide exit path in versus Waksman, and the balance of credit for discovery”, J. Hist. Med. Allied
the ribosome, J. Mol. Biol. 330 (5) (2003) 1005–1014, http://dx.doi.org/ Sci. 60 (2) (2005) 218–220, http://dx.doi.org/10.1093/jhmas/jri024.
10.1016/S0022-2836(03)00662-4. [235] S. Waksman, The Conquest of Tuberculosis, University of California Press,
[220] J.E. Thiemann, G. Zucco, G. Pelizza, A proposal for the transfer of Streptomyces Berkeley, CA, 1964, p. 241.
mediterranei Margalith and Beretta 1960 to the genus Nocardia as Nocardia [236] W. Wehrli, M. Staehelin, Actions of the rifamycins, Bacteriol. Rev. 35 (3)
mediterranea (Margalith and Beretta) comb. nov, Arch. Mikrobiol. 67 (2) (1971) 290–309.
(1969) 147–155. [237] B. Weisblum, Erythromycin resistance by ribosome modification, Antimicrob.
[221] C. Thomas, M. Stevenson, T.V. Riley, Antibiotics and hospital-acquired Agents Chemother. 39 (1995) 577–585, http://dx.doi.org/10.1128/
Clostridium difficile-associated diarrhoea: a systematic review, J. Antimicrob. AAC.39.3.577.
Chemother. 51 (6) (2003) 1339–1350, http://dx.doi.org/10.1093/jac/dkg254. [238] M.P. Wentland, In memoriam: George Y. Lesher, Ph.D., in: D.C. Hooper, J.S.
[222] J.H. Tran, G.A. Jacoby, Mechanism of plasmid-mediated quinolone resistance, Wolfson (Eds.), Quinolone Antimicrobial Agents, 2nd ed., American Society
Proc. Natl. Acad. Sci. U.S.A. 99 (2002) 5638–5642, http://dx.doi.org/10.1073/ for Microbiology, Washington DC, 1993, pp. XIII–XIV.
pnas.082092899. [239] M.J. Wood, The comparative efficacy and safety of teicoplanin and
[223] J.H. Tran, G.A. Jacoby, D.C. Hooper, Interaction of the plasmid-encoded vancomycin, J. Antimicrob. Chemother. 37 (2) (1996) 209–222.
quinolone resistance protein Qnr with Escherichia coli DNA gyrase, [240] G.D. Wright, The origins of antibiotic resistance, Handb. Exp. Pharmacol. 211
Antimicrob. Agents Chemother. 49 (2005) 118–125, http://dx.doi.org/ (2012) 13–30, http://dx.doi.org/10.1007/978-3-642-28951-4_2.
10.1128/AAC.49.1.118-125.2005. [241] L.T. Wright, H. Schreiber, The clinical value of aureomycin: a review of
[224] J. Tréfouël, T. Tréfouël, F. Nitti, D. Bovet, Activité du p-aminophénylsulfamide current literature and some unpublished data, J. Natl Med. Assoc. 41 (5)
sur l’infection streptococcique expérimentale de la souris et du lapin, C. R. (1949) 195–201.
Soc. Biol. 120 (1935) 756. [242] A.J. Wright, The penicillins, Mayo Clin. Proc. 74 (3) (1999) 290–307, http://dx.
[225] A. Tupin, M. Gualtieri, F. Roquet-Banères, Z. Morichaud, K. Brodolin, J.P. doi.org/10.4065/74.3.290.
Leonetti, Resistance to rifampicin: at the crossroads between ecological, [243] J. Xie, J.G. Pierce, R.C. James, A. Okano, D.L. Boger, A redesigned vancomycin
genomic and medical concerns, Int. J. Antimicrob. Agents 35 (6) (2010) 519– engineered for dual D-Ala-D-ala And D-Ala-D-Lac binding exhibits potent
523, http://dx.doi.org/10.1016/j.ijantimicag.2009.12.017. antimicrobial activity against vancomycin-resistant bacteria, J. Am. Chem.
[226] Z.F. Udwadia, R.A. Amale, K.K. Ajbani, C. Rodriguez, Totally drug-resistant Soc. 133 (35) (2011) 13946–13949, http://dx.doi.org/10.1021/ja207142h.
tuberculosis in India, Clin. Infect. Dis. 54 (2012) 579–581, http://dx.doi.org/ [244] J. Xie, A. Okano, J.G. Pierce, R.C. James, S. Stamm, C.M. Crane, D.L. Boger, Total
10.1093/cid/cir889. synthesis of [W[C(@S)NH]Tpg4]vancomycin aglycon, [W[C(@NH)NH]
[227] D.W. Urry, The gramicidin A transmembrane channel: a proposed pi(L, D) Tpg4]vancomycin aglycon, and related key compounds: reengineering
helix, Proc. Natl. Acad. Sci. U.S.A. 68 (3) (1971) 672–676. vancomycin for dual D-Ala-D-Ala and D-Ala-D-Lac binding, J. Am. Chem.
[228] F. Van Bambeke, Glycopeptides and glycodepsipeptides in clinical Soc. 134 (2) (2012) 1284–1297, http://dx.doi.org/10.1021/ja209937s.
development: a comparative review of their antibacterial spectrum, [245] J.Q. Xu, S.L. Murphy, K.D. Kochanek, B.A. Bastian, Deaths: final data for 2013,
pharmacokinetics and clinical efficacy, Curr. Opin. Investig. Drugs 7 (8) Natl. Vital Stat. Rep. 64 (2) (2016) 1–119.
(2006) 740–749. [246] J. Yates, P. Schaible, Virginiamycin as an antibiotic for poultry feeds, Nature
[229] T.P. Van Boeckel, S. Gandra, A. Ashok, Q. Caudron, B.T. Grenfell, S.A. Levin, R. 194 (1962) 183–184.
Laxminarayan, Global antibiotic consumption 2000 to 2010: an analysis of [247] G.G. Zhanel, D. Calic, F. Schweizer, S. Zelenitsky, H. Adam, P.R. Lagacé-Wiens,
national pharmaceutical sales data, Lancet Infect. Dis. 14 (8) (2014) 742–750, E. Rubinstein, A.S. Gin, D.J. Hoban, J.A. Karlowsky, New lipoglycopeptides: a
http://dx.doi.org/10.1016/S1473-3099(14)70780-7. comparative review of dalbavancin, oritavancin and telavancin, Drugs 70 (7)
[230] T.P. Van Boeckel, C. Brower, M. Gilbert, B.T. Grenfell, S.A. Levin, T.P. Robinson, (2010) 859–886, http://dx.doi.org/10.2165/11534440-000000000-00000.
A. Teillant, R. Laxminarayan, Global trends in antimicrobial use in food [248] S. Pournaras, V. Koumaki, N. Spanakis, V. Gennimata, A. Tsakris, Current
animals, Proc. Natl. Acad. Sci. U.S.A. 112 (18) (2015) 5649–5654, http://dx. perspectives on tigecycline resistance in Enterobacteriaceae: susceptibility
doi.org/10.1073/pnas.1503141112. testing issues and mechanisms of resistance, Int. J. Antimicrob. Agents 48 (1)
[231] A.A. Velayati, M.R. Masjedi, P. Farnia, P. Tabarsi, J. Ghanavi, A.H. Ziazarifi, S.E. (2016) 11–18, http://dx.doi.org/10.1016/j.ijantimicag.2016.04.017.
Hoffner, Emergence of new forms of totally drug-resistant tuberculosis [249] M.E. Evans, D.J. Feola, R.P. Rapp, Polymyxin B sulfate and colistin: old
bacilli: super extensively drug-resistant tuberculosis of totally drug-resistant antibiotics for emerging multiresistant gram-negative bacteria, Ann.
strain in Iran, Chest 136 (2009) 420–425, http://dx.doi.org/10.1378/chest.08- Pharmacother. 33 (9) (1999) 960–967.
2427. [250] WHO, Guidelines of the Programmatic Management of Drug-Resistant
[232] A.A. Velayati, P. Farnia, M.R. Masjedi, The totally drug resistant tuberculosis Tuberculosis, 2006. Available at: http://whqlibdoc.who.int/publications/
(TDR-TB), Int. J. Clin. Exp. Med. 6 (4) (2013) 307–309. 2006/9241546956_eng.pdf.
[233] M. Wainwright, Streptomycin: discovery and resultant controversy, Hist.
Philos. Life Sci. 13 (1) (1991) 97–124.

You might also like