Sei sulla pagina 1di 7

Review Article

published: 08 December 2010


doi: 10.3389/fmicb.2010.00134

A brief history of the antibiotic era: lessons learned and


challenges for the future
Rustam I. Aminov*
Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, UK

Edited by: This article gives a very brief overview of the antibiotic era, beginning from the discovery of
Jose L. Martinez, Centro Nacional de
first antibiotics until the present day situation, which is marred by the emergence of hard-to-
Biotecnologa, Spain
treat multiple antibiotic-resistant infections. The ways of responding to the antibiotic resistance
Reviewed by:
Morten Otto Alexander Sommer, challenges such as the development of novel strategies in the search for new antimicrobials,
Technical University of Denmark, designing more effective preventive measures and, importantly, better understanding the
Denmark ecology of antibiotics and antibiotic resistance are discussed. The expansion of conceptual
Jose L. Martinez, Centro Nacional de
frameworks based on recent developments in the field of antimicrobials, antibiotic resistance,
Biotecnologa, Spain
and chemotherapy is also discussed.
*Correspondence:
Rustam I. Aminov, Rowett Institute of Keywords: history of antibiotic discovery, antibiotic resistance, novel antimicrobials, prevention of antibiotic resistance,
Nutrition and Health, University of environmental resistome
Aberdeen, Aberdeen AB21 9SB, UK.
e-mail: r.aminov@abdn.ac.uk

Antibiotics and antibiotic resistance in the for treating skin infections have led to the discovery of a number
preantibiotic era of antibiotic-producing bacteria and concomitant antibiotic pro-
Antimicrobials are probably one of the most successful forms of duction in these soils (Falkinham etal., 2009). The actinomycete
chemotherapy in the history of medicine. It is not necessary to bacteria isolated from these soils produced actinomycin C2 and
reiterate here how many lives they have saved and how significantly actinomycin C3, which are polypeptide antibiotics that bind to a
they have contributed to the control of infectious diseases that were pre-melted DNA conformation present within the transcriptional
the leading causes of human morbidity and mortality for most of complex (Sobell, 1985) and, as such, have very little chance to be
human existence. Contrary to the common belief that the exposure preserved in ancient samples.
to antibiotics is confined to the modern antibiotic era, research Another possibility of exposure to antimicrobials in the pre-
has revealed that this is not the case. The traces of tetracycline, for antibiotic era could be through the remedies used for millennia
example, have been found in human skeletal remains from ancient in traditional/alternative medicine, in particular in traditional
Sudanese Nubia dating back to 350550 CE (Bassett etal., 1980; Chinese medicine (TCM). The best-known example is the discov-
Nelson etal., 2010). The distribution of tetracycline in bones is ery of a potent anti-malarial drug, qinghaosu (artemisinin), which
only explicable after exposure to tetracycline-containing materi- was extracted in the 1970s from Artemisia plants, used by Chinese
als in the diet of these ancient people. Another example of ancient herbalists for thousands of years as a remedy for many illnesses (Cui
antibiotic exposure is from a histological study of samples taken and Su, 2009). Antimicrobial activity seems present in a number of
from the femoral midshafts of the late Roman period skeletons other herbs used in TCM (Wong etal., 2010) and the discovery of
from the Dakhleh Oasis, Egypt (Cook etal., 1989). These samples active components in these ancient remedies may enrich the arsenal
showed discrete fluorochrome labeling consistent with the pres- of antimicrobials used by the mainstream medicine. At the same
ence of tetracycline in the diet at that time (Cook etal., 1989). The time, selective pressures imposed by these antimicrobial activities
postulated intake of tetracycline in these populations possibly had a during the long-term history of TCM may have been one of the
protective effect because the rate of infectious diseases documented factors contributing to the accumulation of antibiotic resistance
in the Sudanese Nubian population was low, and no traces of bone genes in human populations.
infection were detected in the samples from the Dakhleh Oasis The natural history of antibiotic resistance genes can be revealed
(Armelagos, 1969; Cook etal., 1989). through the phylogenetic reconstruction and this kind of analysis
Tetracyclines are unique among antibiotics in that they are suggests the long-term presence of genes conferring resistance to
strong chelators and are incorporated into the hydroxyapatite min- several classes of antibiotics in nature well before the antibiotic era
eral portion of bones as well as tooth enamel and thus provide (Aminov and Mackie, 2007; Kobayashi etal., 2007). Structure-based
permanent markers of metabolically active areas during tetracy- phylogeny of serine and metallo--lactamases, for example, estab-
cline exposure. Traces of exposure to other antibiotics in ancient lished that these ancient enzymes originated more than two billion
populations are much more difficult to detect, and only surviving years ago, with some serine -lactamases being present on plasmids
customs and anecdotal evidence may point to these occurrences. for millions of years (Hall and Barlow, 2004; Garau etal., 2005).
For example, anecdotes about the antibiotic-like properties of red Phylogeny of the -lactamase and housekeeping genes is highly
soils in Jordan that were used historically (and are still being used congruent in Klebsiella oxytoca implying that these genes have been
as an inexpensive alternative to pharmaceutical products today) evolving for over 100 million years in this host (Fevreetal.,2005).

www.frontiersin.org December 2010 | Volume 1 | Article 134 | 1


Aminov Lessons and challenges of antibiotic era

The similar phylogenetic analysis of -lactamases in the metage- 1 integrons. Moreover, once the sulfa drug resistance is established
nomic clones derived from the 10,000years old cold-seep sedi- on a mobile genetic element, it may be difficult to eliminate because
ments indicated that most of the diversity of these enzymes is not the resulting construct confers a fitness advantage to the host even
the result of recent evolution, but is that of ancient evolution (Song in the absence of antibiotic selection (Enne etal., 2004). Despite
etal., 2005). this, many continuously modified derivatives of this oldest class of
synthetic antibiotics are still a viable option for therapy, and the
Foundation of the antibiotic era action of and resistance to sulfanilamide is one of the best examples
We usually associate the beginning of the modern antibiotic era for the arms race between man and microbes. Two other classes
with the names of Paul Ehrlich and Alexander Fleming. Ehrlichs of synthetic antibiotics successful in clinical use are the quinolo-
idea of a magic bullet that selectively targets only disease-causing nes, such as ciprofloxacin, and oxazolidinones, such as linezoild
microbes and not the host was based on an observation that aniline (Walsh, 2003).
and other synthetic dyes, which first became available at that time, Probably many of us are familiar with the somewhat seren-
could stain specific microbes but not others. Ehrlich argued that dipitous event on the September 3, 1928 that led to the penicillin
chemical compounds could be synthesized that would be able to discovery by Fleming (1929). Although the antibacterial proper-
exert their full action exclusively on the parasite harbored within ties of mold had been known from ancient times, and researchers
the organism1. This idea led him to begin a large-scale and sys- before him had come upon the similar observations regarding the
tematic screening program (as we would call it today) in 1904 to antimicrobial activity of Penicillium from time to time2, it was his
find a drug against syphilis, a disease that was endemic and almost formidable persistency and his belief in the idea that made the
incurable at that time. This sexually transmitted disease, caused difference. For 12years after his initial observation, A. Fleming
by the spirochete Treponema pallidium, was usually treated with was trying to get chemists interested in resolving persisting prob-
inorganic mercury salts but the treatment had severe side effects lems with purification and stability of the active substance and
and poor efficacy. In his laboratory, together with chemist Alfred supplied the Penicillium strain to anyone requesting it. He finally
Bertheim and bacteriologist Sahachiro Hata, they synthesized hun- abandoned the idea in 1940, but, fortunately, in the same year an
dreds of organoarsenic derivatives of a highly toxic drug Atoxyl Oxford team led by Howard Florey and Ernest Chain published a
and tested them in syphilis-infected rabbits. In 1909 they came paper describing the purification of penicillin quantities sufficient
across the sixth compound in the 600th series tested, thus numbered for clinical testing (Chain et al., 2005). Their protocol eventu-
606, which cured syphilis-infected rabbits and showed significant ally led to penicillin mass production and distribution in 1945.
promise for the treatment of patients with this venereal disease Flemings screening method using inhibition zones in lawns of
in limited trials on humans (Ehrlich and Hata, 1910). Despite the pathogenic bacteria on the surface of agar-medium plates required
tedious injection procedure and side effects, the drug, marketed much less resources than any testing in animal disease models
by Hoechst under the name Salvarsan, was a great success and, and thus became widely used in mass screenings for antibiotic-
together with a more soluble and less toxic Neosalvarsan, enjoyed producing microorganisms by many researchers in academia and
the status of the most frequently prescribed drug until its replace- industry. Fleming was also among the first who cautioned about
ment by penicillin in the 1940s (Mahoney etal., 1943). Amazingly, the potential resistance to penicillin if used too little or for a too
the mode of action of this 100-year-old drug is still unknown, and short period during treatment.
the controversy about its chemical structure has been solved only Unknown to many, however, is the fact that the first hospital
recently (Lloydetal.,2005). use of a drug that we would name an antibiotic today was the so-
The systematic screening approach introduced by Paul Ehrlich called Pyocyanase prepared by Emmerich and Lw (1899) from
became the cornerstone of drug search strategies in the pharma- Pseudomonas aeruginosa (formerly Bacillus pycyaneus). Importantly,
ceutical industry and resulted in thousands of drugs identified Emmerich and Lw noticed that the bacterium as well as the prepared
and translated into clinical practice, including, of course, a vari- extracts were active against a number of pathogenic bacteria and
ety of antimicrobial drugs. During the earlier days of antibiotics thus tried to use the extract for treatment of various diseases. As the
research, this approach led to the discovery of sulfa drugs, namely results of these treatments were not consistent and the preparation
sulfonamidochrysoidine (KI-730, Prontosil), which was synthe- itself was quite toxic for humans, the treatment was eventually aban-
sized by Bayer chemists Josef Klarer and Fritz Mietzsch and tested by doned. Further investigations confirmed the production of antibi-
Gerhard Domagk for antibacterial activity in a number of diseases otic substances by Pseudomonas aeruginosa (Hays etal., 1945), which
(Domagk, 1935). Prontosil, however, appeared to be a precursor appeared to be the quorum sensing molecules, 2-alkyl-4quinolones,
to the active drug, and the active part of it, sulfanilamide, was thus in this bacterium (Dubern and Diggle, 2008). Another quorum sens-
not patentable as it had already been in use in the dye industry for ing molecule of Pseudomonas aeruginosa, N-(3-oxododecanoyl)
some years. As sulfanilamide was cheap to produce and off-patent, homoserine lactone, and its non-enzymatically formed product,
and the sulfanilamide moiety was easy to modify, many companies 3-(1-hydroxydecylidene)-5-(2-hydroxyethyl)pyrrolidine-2,4-dione,
subsequently started mass production of sulfonamide derivatives. also display potent antibacterial activities (Kaufmann etal., 2005).
The legacy of this oldest antibiotic on market is possibly reflected The discovery of these first three antimicrobials, Salvarsan,
in one of the most broadly disseminated cases of drug resistance: Prontosil, and penicillin, was exemplary, as those studies set up the
sulfa drug resistance, which is almost universally linked with class paradigms for future drug discovery research. The paths, followed

1
http://pubs.acs.org/cen/coverstory/83/8325/8325salvarsan.html http://en.wikipedia.org/wiki/Discovery_of_penicillin
2

Frontiers in Microbiology | Antimicrobials, Resistance and Chemotherapy December 2010 | Volume 1 | Article 134 | 2
Aminov Lessons and challenges of antibiotic era

by other researchers, resulted in a number of new antibiotics, some Recent works in the area of antimicrobials and resistance suggest
of which made their way up to the patients bedside. The period that not all interactions of bacteria with antibiotics can be explained
between the 1950s and 1970s was indeed the golden era of discovery within the frames of the classical bullet-target concept. For example,
of novel antibiotics classes, with no new classes discovered since then. a recent work on novel antibiotic resistance mechanism used the
Therefore, with the decline of the discovery rate, the mainstream kin selection concept, since this resistance mechanism operates at
approach for the development of new drugs to combat emerging the population/system level (Lee etal., 2010). This well-established
and re-emerging resistance of pathogens to antibiotics has been the theory for the macro-organismal world seems also applicable to the
modification of existing antibiotics (Chopra etal., 2002). microbial world, where a small number of antibiotic-resistant bacte-
ria provide protection for the antibiotic sensitive cells, thus ensuring
Antibiotic resistance the survival of the whole population under the antibiotic assault.
Even before the extensive use of penicillin, some observations Moreover, in complex biofilm consortia, the protection against
suggested that bacteria could destroy it by enzymatic degrada- antibiotics is offered to all community members, irrespectively of
tion (Abraham and Chain, 1940). In general, though, the outlook the kinship, which requires a conceptual framework operating at
was more or less optimistic. One of the earlier studies of possi- the system level. Thus the conceptual base of microbeantibiotic
ble resistance emergence under laboratory conditions concluded interaction has been broadening beyond the bullet-target model
that: Syphilis has now been treated with arsenicals for about to reflect the complexity of these interactions (Davies etal., 2006;
40 years without any indications of an increased incidence of Fajardo and Martnez, 2008; Aminov, 2009; Lee etal., 2010).
arsenic-resistant infections, and this work gives grounds for hop-
ing that the widespread use of penicillin will equally not result Resistance problem complexity
in an increasing incidence of infections resistant to penicillin The current state in the field of antimicrobials, resistance, and
(Rollo etal., 1952). Surprisingly, this is still true for T. pallidium chemotherapy is certainly not limited to clinical microbiology as
(Cha etal., 2004), but not for many other pathogenic bacteria, it was in the early years of the antibiotic era. Thus, it is not a single
including the Enterobacteriaceae, which have become resistant grand challenge; it is rather a complex problem requiring con-
not only to the original penicillin but also to semi-synthetic peni- certed efforts of microbiologists, ecologists, health care specialists,
cillins, cephalosporins, and newer carbapenems (Kumarasamy educationalists, policy makers, legislative bodies, agricultural and
etal., 2010). pharmaceutical industry workers, and the public to deal with. In
The mortality rates due to multidrug-resistant bacterial infec- fact, this should be of everyones concern, because, in the end, there
tions are high. Each year, about 25,000 patients in the EU die from is always a probability for any of us at some stage to get infected
an infection with the selected multidrug-resistant bacteria (ECDC/ with a pathogen that is resistant to antibiotic treatment. Moreover,
EMEA Joint Working Group, 2009), and more than 63,000 patients even the behavioral patterns, such as hygienic habits or compliance
in the United States die every year from hospital-acquired bac- with antibiotic treatment regimens, may have consequences that are
terial infections3. Estimated economic costs due to infections by not limited only to individual health issues but, on a larger scale,
multidrug-resistant bacteria in the EU result in extra healthcare contribute to the interaction with the resistomes around us. In
costs and productivity losses of at least EUR 1.5 billion each year the following sections I will briefly touch upon some of the areas
(ECDC/EMEA Joint Working Group, 2009). The annual additional ranging from research to regulations to the cultural patterns that
cost of treating hospital-acquired infections from just six species are important in dealing with the challenges of the antimicrobials,
of antibiotic-resistant bacteria was estimated to be at least $1.3 resistance, and therapy fields.
billion in 1992 dollars ($1.87 billion in 2006 dollars) more than
the annual spending on influenza3. Improvement of antimicrobials
There are a number of excellent reviews elsewhere describing a This strategy of modification of the existing antimicrobials was
variety of antibiotic resistance mechanisms and, within the frames initiated (and successfully implemented) during the period, when
of the bullet-target concept, these mechanisms can be classified as the rate of discovery of novel drug classes suddenly dropped in the
a target or bullet-related. Targets can be: (i) protected by modifica- 1970s, and the growing resistance problem enforced researchers
tion (mutations making it insensitive to antibiotic action such as to look into the possible modification of the existing arsenal that
mutations in RNA polymerase conferring resistant to rifampin; (ii) could confer improved activity, less sensitivity toward resistance
modified by an enzyme (such as methylation of an adenine residue mechanisms, and less toxicity (Chopra etal., 2002). Although this
in 23S rRNA making it insensitive to macrolides); (iii) replaced approach still successfully provides effective antimicrobials for the
(for example, ribosomal protection proteins conferring resistance market, one of the lessons learned during this arms race is that
to tetracyclines); and (iv) protected at cellular or population levels sooner or later bacteria will acquire resistance to these modified ver-
(formation of a protective barrier by secretion, for example, of large sions as well through the horizontal acquisition of novel resistance
amounts of exopolysaccharides). The bullet can be: (i) modified mechanisms or rapid molecular evolution of the existing resistances
so the efficiency is lost, as in the case of acetylation of aminogly- to older antibiotics.
cosides, (ii) destroyed (as the -lactam antibiotics by the action
of -lactamases), and (iii) pumped out from the cell as in efflux Novel antimicrobials
pump mechanisms of resistance. The antibiotic treatment choices for already existing or emerging
hard-to-treat multidrug-resistant bacterial infections are limited,
3
www.extendingthecure.org/report resulting in high morbidity and mortality rates. Although there

www.frontiersin.org December 2010 | Volume 1 | Article 134 | 3


Aminov Lessons and challenges of antibiotic era

are some potential alternatives to antibiotic treatment such as pas- Other potential targets for intervention in bacterial metabolism
sive immunization (Keller and Stiehm, 2000) or phage therapy include fatty acid biosynthesis (Su and Honek, 2007), cell divi-
(Levin and Bull, 2004; Monk etal., 2010), the mainstream approach sion (Lock and Harry, 2008), biosynthesis of aminoacyl-tRNAs
relies on the discovery and development of newer, more efficient (Schimmel etal., 1998), quorum sensing (Njoroge and Sperandio,
antibiotics. The vast majority of antimicrobial classes in use today 2009), bacterial two-component signal transduction (Gotoh etal.,
have been isolated in the golden era of antibiotic discovery from a 2010), and proton motive force (Diacon etal., 2009). Antibiotic resist-
limited number of ecological niches and taxonomic groups, mainly ance mechanisms themselves such as efflux pumps or -lactamases
from soil Actinomyces. Further exploration of this ecological niche, can also be targeted to restore the efficacy of antibiotics, which is
coupled with newer technologies such as cell-free assays and high- compromised by the growing resistance problem (Lomovskaya and
throughput screening, however, did not produce any novel drug Bostian, 2006; Bush and Macielag, 2010). It should be noted that,
classes in the past 20+ years. What approaches could be taken to moving along this route, we are cardinally departing from the previ-
uncover the novel antimicrobials diversity that is potentially suit- ously defined classical structural divisions into antibiotic classes. We
able for therapeuticapplications? are only at the beginning here, and not many antibacterial drugs with
Some possible approaches to tap the novel antimicrobial diver- novel mechanisms of action have entered clinical trials yet, but even
sity is the exploration of ecological niches other than soil, such at this stage the majority of them do not belong to the previously
as the marine environment (Hughes and Fenical, 2010; Rahman defined antibiotic classes (Devasahayam etal., 2010).
et al., 2010), borrowing antimicrobial peptides and compounds Also, the intervention strategies aimed not only at the targets but
from animals and plants (Hancock and Sahl, 2006), mimicking rather at biological networks may help to create new antibacterial
the natural lipopeptides of bacteria and fungi (Makovitzki etal., therapies (Kohanski etal., 2010a). Combination therapy coupling
2006), accessing the uncultivated portion of microbiota through antibiotics with an antibiotic-enhancing phage, for example, has
the metagenomic approach (MacNeil etal., 2001), and, finally, the demonstrated the potential to be a promising antimicrobial inter-
use of the complete synthetic route pioneered during the early years vention (Lu and Collins, 2009).
of the antibiotic era. The latter approach becomes dominant in the
search for drugs aimed at the newly identified targets in a bacterial Prevention of antibiotic resistance
cell. Other strategies may include drugs engineered to possess dual The main problem we are facing with antibiotic therapy is that after a
target activities, such as a rifamycinquinolone hybrid antibiotic, new antibiotic is introduced, resistance to it will, sooner or later, arise.
CBR-2092 (Robertson etal., 2008). This scenario has been seen on multiple occasions, and thus there is a
continuing race between the discovery and development of new antibi-
New targets for antimicrobials otics and the bacteria that will respond to this selective pressure by the
The vast majority of current antibiotics, even heavily modified, emergence of resistance mechanisms. So how to protect the power of
target the same cellular processes as their natural or synthetic antibiotics and extend their lifespan? There are many factors contribut-
predecessors. The range of these targets is limited to the compo- ing to the emergence and dissemination of antibiotic resistance and, as
nents of translational machinery, cell wall biosynthesis, DNA/ mentioned before, the problems require a complex approach.
RNA metabolism and some other cellular processes. With the A significant factor to consider apparently is the use of antibiotics
extensive range of genomes sequenced, it becomes possible to by humans. Not surprisingly, the level of antibiotic-resistant infec-
implement the idea of a magic bullet in a more elaborate way, with tions strongly correlates with the level of antibiotic consumption
essential targets defined much more precisely at the molecular (Goossens et al., 2005). There may be requests from patients to
level. This needs to be complemented by the availability of chemi- prescribe antibiotics when there is no need for them, as in the case of
cally diverse compound collections to screen for the target/drug viral infections, and which should be explained to them. Indeed, the
combination (Payne etal., 2007). The comparison of metabolic lack of knowledge about antibiotic resistance positively correlates
pathways in commensal and pathogenic bacteria and drugs tar- with the higher prevalence of resistance (Grigoryan etal., 2007). The
geting the pathogenic traits may help to identify the novel drug/ important part is also to comply with the drug use regimen, which
target combinations in pathogens and thus the novel paradigm of may be difficult in the case of infections requiring long-term therapy
antimicrobial therapy as targeting virulence (Clatworthy, 2007). with multiple antibiotics as in the case of TB. The contributing fac-
Successful implementations of this approach have already been tor to the dissemination of antibiotic resistance, even in the case of
demonstrated in the suppression of an important virulence fac- absolute compliance, may be the practice of empirical prescription
tor, type III secretion system (Negrea et al., 2007), and in the of antibiotics (which accounts for the vast majority of prescriptions).
inhibition of the QseC-mediated activation of virulence gene The development of express ABR profiling tests would be quite help-
expression in several pathogens (Rasko etal., 2008). The drugs ful in the initiation of the most efficient therapy available, avoiding
initially designed for a different purpose may find application as the hurdles associated with a resistant pathogen. In therapy, the use
antimicrobials. For example, BPH-652, a phosphonosulfonate, of a recombinant -lactamase during intravenous administration of
which was previously tested for cholesterol-lowering activity in ampicillin may prevent the emergence of antimicrobial resistance
humans as targeting the enzyme in cholesterol biosynthesis path- in gut microbiota (Tarkkanen etal., 2009).
way, squalene synthase, is also inhibiting an important enzyme The situation is different in countries where the sales of antibiot-
involved in Staphylococcus aureus virulence, dehydrosqualene syn- ics are inadequately regulated, and antibiotics are available without
thase, and thus may be considered as a candidate drug to control prescription. In the absence of regulation, the personal decisions on
MRSA (Liu etal., 2008). antibiotic purchase and use are governed by cultural and economic

Frontiers in Microbiology | Antimicrobials, Resistance and Chemotherapy December 2010 | Volume 1 | Article 134 | 4
Aminov Lessons and challenges of antibiotic era

reasons (Gartin et al., 2010). Self-medication certainly lacks the in DNA metabolism in environmental bacteria also happened to
attributes of a successful therapy, such as proper diagnosis, suitable be protecting against this synthetic antibiotic, and, once the encod-
antibiotic choice, correct usage, compliance, and treatment efficiency ing gene entered into the human/animal microbiota on a mobile
monitoring, thus contributing to the mounting resistance problem. element, a strong selective pressure led to its amplification and
Domesticated animals also get infected and require antibiotic dissemination into the commensal and pathogenic microbiota
therapy. The agricultural use of antibiotics, however, is not limited (Aminov and Mackie, 2007).
exclusively to this use. Antibiotics are also used for the growth Another important aspect is the release of antibiotics and the
promotional and prophylactic purposes in food animals, as well corresponding pre-selected and amplified antibiotic resistance gene
as for a broader and less-targeted treatment in aquaculture and pool from the human and animal ecological compartments back into
horticulture. The experience of the Scandinavian countries, where the environment (Chee-Sanford etal., 2009). What are the conse-
the programs of optimal disease preventive management routines quences of this? Does the resulting multidrug combination in the
and proper use of antimicrobials, combined with the withdrawal environment accelerate the evolution toward antimicrobial resistance
of antibiotic growth promoters, were implemented in food animal (Hegreness etal., 2008)? What are the consequences of the presence
production, is encouraging. These measures resulted in reduction of sublethal concentration antibiotics in the environment (Kohanski
in the use of antimicrobials and prevented the creation of a rela- etal., 2010b)? What is the impact of the environmental stresses, such
tively favorable situation for antimicrobial resistance (Bengtsson as, for example, the SOS response-inducing UV radiation, on the
and Wierup, 2006). With the ban of growth promoting antibiot- horizontal dissemination of antibiotic resistance genes (Beaber etal.,
ics in 2006, other EU countries have been implementing similar 2004)? What is the rate of decay of the released antibiotic resistance
measures to limit the occurrence and dissemination of antibiotic genes in the environment, given the improved survival strategies of
resistance from agricultural sources. mobile genetic elements (de la Cueva-Mndez and Pimentel, 2007)?
Could the environment provide a broader playground for the mobile
Environmental resistome antibiotic resistance encoding elements to promote their own diver-
The history of massive production and use of antibiotics by humans sity (Garriss etal., 2009)? And what are the chances for antibiotic
is very short on the evolutionary scale, but even this short-term resistance genes to re-enter the human and animal food chain? There
(albeit large-scale) practice has produced very interesting results are pressing needs to answer these questions to build the broader
demonstrating the interminable adaptive capabilities of bacteria, strategies that would help to preserve the power of antibiotics.
which allow them to withstand massive antibiotic insults and gen-
erate some formidable examples of hard-to-treat infections that Conclusions
we call superbugs. Although no broad baseline data collection Albeit very brief, both on the global evolutionary and human his-
on antibiotic resistance genes has been performed at the onset of tory scales, the antibiotics era went through many ups and downs,
antibiotic use, recent studies suggest that the most likely source of providing us valuable lessons on many aspects of how the microbial
these genes is the environmental antibiotic resistome (Martnez, world around us functions. The discovery and use of antibiotics, anti-
2008; Wright, 2010). The functional role of antimicrobials and the biotic resistance markers, and mobile elements such plasmids were
corresponding resistance genes in natural ecosystems is hypoth- at the foundation of genetic engineering and molecular biology that
esized as a signaling/regulatory rather than a bullet/target rela- eventually resulted in spectacular successes of the human genome
tionship (Davies etal., 2006; Fajardo and Martnez, 2008; Aminov, and other sequencing projects. These tools were also indispensable
2009). Confirmatory to this suggestion are also the other thera- for shaping modern biotechnology ranging from the production of
peutic properties of antimicrobials, well beyond the initial range recombinant proteins to construction of entire metabolic pathways.
of use as anti-infective agents (Griffin etal., 2010). Bacteria do not Microorganisms, however, use the very same (and probably some
respect the boundaries of ecological compartments, and there is additional but still unidentified) natural mechanisms to protect them-
always a continuous flow by genetic information between different selves against the massive antibiotic assaults continuously launched by
ecological compartments. Once the potential antibiotic resistance the humankind from the time of discovery of antibiotics. Although
genes enter, even in small numbers or low frequencies, into the the majority of infections were placed under control, this equilibrium
commensal/pathogenic human/animal microbiota, the antibiotic in the arm race is fragile, since during the almost four billion years of
selection immediately leads to the amplification and dissemination evolution the microbial world has accumulated an enormous diversity
of these genes. Indeed, there is some evidence on the environmental of metabolic and protective mechanisms than can be mobilized in
origin of some clinically relevant resistance genes (Wright, 2010). response to a strong selection. We need to learn to be more precise
The predecessor/ancestral genes for antibiotic resistance are not in targeting the pathogens and limit the indiscriminate use of anti-
necessarily the genes conferring resistance per se in natural ecosys- microbials and other practices that accelerate the emergence of novel
tems. The recently emerged resistance to synthetic antibiotics of the resistance mechanisms. Investigation of the microbial world around
quinolone class is mediated by the qnr genes that have been acquired us for potential mechanisms of antibiotic resistance and dissemina-
from aquatic bacteria (Poirel etal., 2005). It was suggested that the tion may help to design the early warning and preventive measures
original function of one of its homologs, MfpA, is providing DNA to sustain the efficacy of antimicrobials and chemotherapy.
topological assistance when needed, and maintaining a condensed
chromosome and preventing undesired topological changes dur- Acknowledgment
ing periods of replicative senescence (Hegde etal., 2005). In this I thank the Scottish Government Rural and Environment Research
scenario of quinolone resistance emergence, the enzyme involved and Analysis Directorate for support.

www.frontiersin.org December 2010 | Volume 1 | Article 134 | 5


Aminov Lessons and challenges of antibiotic era

References c ombination therapy of artemisi- Fleming, A. (1929). On antibacterial from Mycobacterium t uberculosis
Abraham, E. P., and Chain, E. (1940). An nin. Expert Rev. Anti. Infect. Ther. 7, action of culture of Penicillium, with that mimics DNA. Science 308,
enzyme from bacteria able to destroy 9991013. special reference to their use in isola- 14801483.
penicillin. Nature 146, 837. Davies, J., Spiegelman, G. B., and Yim, G. tion of B. influenzae. Br. J. Exp. Pathol. Hegreness, M., Shoresh, N., Damian, D.,
Aminov, R. I. (2009). The role of antibiot- (2006). The world of subinhibitory 10, 226236. Hartl, D., and Kishony, R. (2008).
ics and antibiotic resistance in nature. antibiotic concentrations. Curr. Opin. Garau, G., Di Guilmi, A. M., and Hall, B. Accelerated evolution of resistance in
Environ. Microbiol. 11, 29702988. Microbiol. 9, 445453. G. (2005). Structure-based phylog- multidrug environments. Proc. Natl.
Aminov, R. I., and Mackie, R. I. (2007). de la Cueva-Mndez, G., and Pimentel, B. eny of the metallo-beta-lactamases. Acad. Sci. U.S.A. 105, 1397713981.
Evolution and ecology of antibiotic (2007). Gene and cell survival: lessons Antimicrob. Agents Chemother. 49, Hughes, C. C., and Fenical, W. (2010).
resistance genes. FEMS Microbiol. Lett. from prokaryotic plasmid R1. EMBO 27782784. Antibacterials from the sea. Chemistry
271, 147161. Rep. 8, 458464. Garriss, G., Waldor, M. K., and Burrus, 16, 1251212525.
Armelagos, G. J. (1969). Disease in ancient Devasahayam, G., Scheld, W. M., and V. (2009). Mobile antibiotic resistance Kaufmann, G. F., Sartorio, R., Lee, S. H.,
Nubia. Science 163, 225258. Hoffman, P. S. (2010). Newer antibac- encoding elements promote their own Rogers, C. J., Meijler, M. M., Moss, J. A.,
Bassett, E. J., Keith, M. S., Armelagos, G. terial drugs for a new century. Expert diversity. PLoS Genet. 5, e1000775. doi: Clapham, B., Brogan, A. P., Dickerson,
J., Martin, D. L., and Villanueva, A. R. Opin. Investig. Drugs 19, 215234. 10.1371/journal.pgen.1000775. T. J., and Janda, K. D. (2005). Revisiting
(1980). Tetracycline-labeled human Diacon, A. H., Pym, A., and Grobusch, M. Gartin, M., Brewis, A. A., and Schwartz, quorum sensing: discovery of addi-
bone from ancient Sudanese Nubia (2009). The diarylquinoline TMC207 N. A. (2010). Nonprescription anti- tional chemical and biological func-
(A.D. 350). Science 209, 15321534. for multidrug-resistant tuberculosis. biotic therapy: cultural models on tions for 3-oxo-N-acylhomoserine
Beaber, J. W., Hochhut, B., and Waldor N. Engl. J. Med. 360, 23972405. both sides of the counter and both lactones. Proc. Natl. Acad. Sci. U.S.A.
M. K. (2004). SOS response promotes Domagk, G. (1935). Ein Beitrag zur sides of the border. Med. Anthropol. 102, 309314.
horizontal dissemination of antibiotic Chemotherapie der bakteriellen Q. 24, 85107. Keller, M. A., and Stiehm, E. R. (2000).
resistance genes. Nature 427, 7274. Infektionen. Dtsch. Med. Wochenschr. Goossens, H., Ferech, M., Vander Stichele, Passive immunity in prevention and
Bengtsson, B., and Wierup, M. (2006). 61, 250. R., Elseviers, M., and ESAC Project treatment of infectious diseases. Clin.
An t i m i c ro b i a l re s i s t a n ce i n Dubern, J. F., and Diggle, S. P. (2008). Group. (2005). Outpatient antibiotic Microbiol. Rev. 13, 602614.
Scandinavia after ban of antimicrobial Quorum sensing by 2-alkyl-4-qui- use in Europe and association with Kobayashi, T., Nonaka, L., Maruyama,
growth promoters. Anim. Biotechnol. nolones in Pseudomonas aeruginosa resistance: a cross-national database F., and Suzuki, S. (2007). Molecular
17, 147156. and other bacterial species. Mol Biosyst study. Lancet 365, 579587. evidence for the ancient origin of the
Bush, K., and Macielag, M. J. (2010). New 4, 882888. Gotoh, Y., Eguchi, Y., Watanabe, T., ribosomal protection protein that
-lactam antibiotics and -lactamase Ehrlich, P., and Hata, S. (1910). Die Okamoto, S., Doi, A., and Utsumi, mediates tetracycline resistance in
inhibitors. Expert Opin. Ther. Pat. 20, Experimentelle Chemotherapie der R. (2010). Two-component signal bacteria. J. Mol. Evol. 65, 228235.
12771293. Spirilosen. Berlin: Julius Springer. transduction as potential drug targets Kohanski, M. A., Dwyer, D. J., and Collins,
Cha, J. Y., Ishiwata, A., and Mobashery, S. Emmerich, R., and Lw, O. (1899). in pathogenic bacteria. Curr. Opin. J. J. (2010a). How antibiotics kill bac-
(2004). A novel -lactamase activity Bakteriolytische enzyme als Ursache Microbiol. 13, 232239. teria: from targets to networks. Nat.
from a penicillin-binding protein of der erworbenen Immunitt und die Griffin, M. O., Fricovsky, E., Ceballos, G., Rev. Microbiol. 8, 423435.
Treponema pallidum and why syphilis Heilung von Infectionskrankheiten and Villarreal, F. (2010). Tetracyclines: Kohanski, M. A., DePristo, M. A., and
is still treatable with penicillin. J. Biol. durch dieselben. Z. Hyg. 31, 165. a pleitropic family of compounds with Collins, J. J. (2010b). Sublethal anti-
Chem. 279, 1491714921. Enne,V. I., Bennett, P. M., Livermore, D. M., promising therapeutic properties. biotic treatment leads to multidrug
Chain, E., Florey, H. W., Gardner, A. D., and Hall, L. M. (2004). Enhancement Review of the literature. Am. J. Physiol. resistance via radical-induced muta-
Heatley, N. G., Jennings, M. A., Orr- of host fitness by the sul2-coding plas- Cell Physiol. 299, C539C548. genesis. Mol. Cell 37, 311320.
Ewing, J., and Sanders, A. G. (2005). mid p9123 in the absence of selective Grigoryan, L., Burgerhof, J. G., Degener, Kumarasamy, K. K., Toleman, M. A., Walsh,
The classic: penicillin as a chemothera- pressure. J. Antimicrob. Chemother. 53, J. E., Deschepper, R., Lundborg, C. S., T. R., Bagaria, J., Butt, F., Balakrishnan,
peutic agent. 1940. Clin. Orthop. Relat. 958963. Monnet, D. L., Scicluna, E. A., Birkin, R., Chaudhary, U., Doumith, M., Giske,
Res. 439, 2326. European Centre for Disease Prevention J., Haaijer-Ruskamp, F. M., and SAR C. G., Irfan, S., Krishnan, P., Kumar,
Chee-Sanford, J. C., Mackie, R. I., Koike, and Control/European Medicines Consortium. (2007). Attitudes, beliefs A. V., Maharjan, S., Mushtaq, S.,
S., Krapac, I. G., Lin, Y. F., Yannarell, Agency Joint Working Group and knowledge concerning antibiotic Noorie, T., Paterson, D. L., Pearson,
A. C., Maxwell, S., and Aminov, R. I. (ECDC/EMEA). (2009). The Bacterial use and self-medication: a comparative A., Perry, C., Pike, R., Rao, B., Ray, U.,
(2009). Fate and transport of antibi- Challenge: Time to React. Available at European study. Pharmacoepidemiol. Sarma, J. B., Sharma, M., Sheridan,
otic residues and antibiotic resistance www.ecdc.europa.eu/en/publica- Drug. Saf. 16, 12341243. E., Thirunarayan, M. A., Turton, J.,
genes following land application of tions/Publications/0909_TER_The_ Hall, B. G., and Barlow, M. (2004). Upadhyay, S., Warner, M., Welfare,
manure waste. J. Environ. Qual. 38, Bacterial_Challenge_Time_to_React. Evolution of the serine beta-lacta- W., Livermore, D. M., and Woodford,
10861108. pdf. mases: past, present and future. Drug N. (2010). Emergence of a new anti-
Chopra, I., Hesse, L., and ONeill, A. Fajardo, A., and Martnez, J. L. (2008). Resist. Updat. 7, 111123. biotic resistance mechanism in India,
(2002). Discovery and develop- Antibiotics as signals that trigger spe- Hancock, R. E., and Sahl, H. G. (2006). Pakistan, and the UK: a molecular,
ment of new anti-bacterial drugs, cific bacterial responses. Curr. Opin. Antimicrobial and host-defense biological, and epidemiological study.
in Pharmacochemistry Library, Vol. Microbiol. 11, 161167. peptides as new anti-infective thera- Lancet Infect. Dis. 10, 597602.
32, Trends in Drug Research III, ed. H. Falkinham, J. O. 3rd, Wall, T. E., Tanner, J. peutic strategies. Nat. Biotechnol. 24, Lee, H. H., Molla, M. N., Cantor, C. R., and
van der Goot (Amsterdam: Elsevier), R., Tawaha, K., Alali, F. Q., Li, C., and 15511557. Collins, J. J. (2010). Bacterial charity
213225. Oberlies, N. H. (2009). Proliferation Hays, E. E., Wells, I. C., Katzman, P. A., work leads to population-wide resist-
Clatworthy, A. E., Pierson, E., and Hung, of antibiotic-producing bacteria and Cain, C. K., Jacobs, F. A., Thayer, S. ance. Nature 467, 8285.
D. T. (2007). Targeting virulence: a new concomitant antibiotic production A., Doisy, E. A., Gaby, W. L., Roberts, Levin, B. R., and Bull, J. J. (2004).
paradigm for antimicrobial therapy. as the basis for the antibiotic activity E. C., Muir, R. D., Carroll, C. J., Population and evolutionary dynamics
Nat. Chem. Biol. 3, 541548. of Jordans red soils. Appl. Environ. Jones, L. R., and Wade, N. J. (1945). of phage therapy. Nat. Rev. Microbiol.
Cook, M., Molto, E., and Anderson, C. Microbiol. 75, 27352741. Antibiotic substances produced by 2, 166173.
(1989). Fluorochrome labelling in Fevre, C., Jbel, M., Passet, V., Weill, F. X., Pseudomonas aeruginosa. J. Biol. Liu, C. I., Liu, G. Y., Song, Y., Yin, F.,
Roman period skeletons from Dakhleh Grimont, P. A., and Brisse, S. (2005). Chem. 159, 725750. Hensler, M. E., Jeng, W. Y., Nizet, V.,
Oasis, Egypt. Am. J. Phys. Anthropol. Six groups of the OXY -lactamase Hegde, S. S., Vetting, M. W., Roderick, S. L., Wang, A. H., and Oldfield, E. (2008).
80, 137143. evolved over millions of years in Mitchenall, L. A., Maxwell, A., Takiff, A cholesterol biosynthesis inhibitor
Cui, L., and Su, X. Z. (2009). Discovery, Klebsiella oxytoca. Antimicrob. Agents H. E., and Blanchard, J. S. (2005). A blocks Staphylococcus aureus virulence.
m e ch a n i s m s o f a c t i o n a n d Chemother. 49, 34533462. fluoroquinolone resistance protein Science 319, 13911394.

Frontiers in Microbiology | Antimicrobials, Resistance and Chemotherapy December 2010 | Volume 1 | Article 134 | 6
Aminov Lessons and challenges of antibiotic era

Lloyd, N. C., Morgan, H. W., Nicholson, Bacteriophage applications: where Roth, M., Hughes, D. T., Huntley, J. F., Antimicrob. Agents Chemother. 53,
B. K., and Ronimus, R. S. (2005). The are we now? Lett. Appl. Microbiol. 51, Fina, M. W., Falck, J. R., and Sperandio, 24552462.
Composition of Ehrlichs Salvarsan: 363369. V. (2008). Targeting QseC signaling Walsh, C. T. (2003). Antibiotics: Actions,
resolution of a century-old debate. Negrea, A., Bjur, E., and Ygberg, S. E. and virulence for antibiotic develop- Origins, and Resistance. Washington,
Angew. Chem. Int. Ed. 44, 941944. (2007). Salicylidene acylhydrazides ment. Science 321, 10781080. DC: ASM Press.
Lock, R. L., and Harry, E. J. (2008). Cell- that affect type III protein secretion in Robertson, G. T., Bonventre, E. J., and Wong, R. W., Hgg, U., Samaranayake,
division inhibitors: new insights for Salmonella enterica serovar typhimu- Doyle, T. B. (2008). In vitro evalua- L., Yuen, M. K., Seneviratne, C. J., and
future antibiotics. Nat. Rev. Drug rium. Antimicrob. Agents Chemother. tion of CBR-2092, a novel rifamycin- Kao, R. (2010). Antimicrobial activ-
Discov. 7, 324338. 51, 28672876. quinolone hybrid antibiotic: studies of ity of Chinese medicine herbs against
Lomovskaya, O., and Bostian, K. A. (2006). Nelson, M. L., Dinardo, A., Hochberg, the mode of action in Staphylococcus common bacteria in oral biofilm. A
Practical applications and feasibility of J., and Armelagos, G. J. (2010). Brief aureus. Antimicrob. Agents Chemother. pilot study. Int. J. Oral Maxillofac. Surg.
efflux pump inhibitors in the clinic--a communication: mass spectroscopic 52, 23132323. 39, 599605.
vision for applied use. Biochem. characterization of tetracycline in the Rollo, I. M., Williamson, J., and Plackett, Wright, G. D. (2010). Antibiotic resistance
Pharmacol. 71, 910908. skeletal remains of an ancient popula- R. L. (1952). Acquired resistance to in the environment: a link to the clinic?
Lu, T. K., and Collins, J. J. (2009). tion from Sudanese Nubia 350550 CE. penicillin and to neoarsphenamine Curr. Opin. Microbiol.13, 589594.
Engineered bacteriophage targeting Am. J. Phys. Anthropol. 143, 151154. in Spirochaeta recurrentis. Br. J.
gene networks as adjuvants for anti- Njoroge, J., and Sperandio, V. (2009). Pharmacol. Chemother. 7, 3341. Conflict of Interest Statement: The author
biotic therapy. Proc. Natl. Acad. Sci. Jamming bacterial communication: Schimmel, P., Tao, J., and Hill, J. (1998). declares that the research was conducted in
U.S.A. 106, 46294633. new approaches for the treatment of Aminoacyl tRNA synthetases as tar- the absence of any commercial or financial
MacNeil, I. A., Tiong, C. L., Minor, C., infectious diseases. EMBO Mol. Med. gets for new anti-infectives. FASEB J. relationships that could be construed as a
August, P. R., Grossman, T. H., Loiacono, 1, 201210. 12, 15991609. potential conflict of interest.
K. A., Lynch, B. A., Phillips, T., Narula, S., Payne, D. J., Gwynn, M. N., Holmes, D. J., Sobell, H. M. (1985). Actinomycin and
Sundaramoorthi, R., Tyler, A., Aldredge, and Pompliano, D. L. (2007). Drugs for DNA transcription. Proc. Natl. Acad. Received: 29 September 2010; paper pend-
T., Long, H., Gilman, M., Holt, D., and bad bugs: confronting the challenges Sci. U.S.A. 82, 53285331. ing published: 29 October 2010; accepted:
Osburne, M. S. (2001). Expression and of antibacterial discovery. Nat. Rev. Song, J. S., Jeon, J. H., Lee, J. H., Jeong, 17 November 2010; published online: 08
isolation of antimicrobial small mol- Drug Discov. 6, 2940. S. H., Jeong, B. C., Kim, S. J., Lee, J. December 2010.
ecules from soil DNA libraries. J. Mol. Poirel, L., Rodriguez-Martinez, J. H., and Lee, S. H. (2005). Molecular Citation: Aminov RI (2010) A brief history
Microbiol. Biotechnol. 3, 301308. M., Mammeri, H., Liard, A., and characterization of TEM-type beta- of the antibiotic era: lessons learned and
Mahoney, J., Arnold, R., and Harris, A. Nordmann, P. (2005). Origin of plas- lactamases identified in cold-seep challenges for the future. Front. Microbio.
(1943). Penicillin treatment of early mid-mediated quinolone resistance sediments of Edison Seamount (south 1:134. doi: 10.3389/fmicb.2010.00134
syphilis. A preliminary report. Verer. determinant QnrA. Antimicrob. Agents of Lihir Island, Papua New Guinea). J. This article was submitted to Frontiers
Dis. Inform. 24, 355357. Chemother. 49, 35233525. Microbiol. 43, 172178. in Antimicrobials, Resistance and
Makovitzki, A., Avrahami, D., and Shai, Y. Rahman, H., Austin, B., Mitchell, W. J., Su, Z., and Honek, J. F. (2007). Emerging Chemotherapy, a specialty of Frontiers in
(2006). Ultrashort antibacterial and Morris, P. C., Jamieson, D. J., Adams, bacterial enzyme targets. Curr. Opin. Microbiology.
antifungal lipopeptides. Proc. Natl. D. R., Spragg, A. M., and Schweizer, Investig. Drugs 8, 140149. Copyright 2010 Aminov. This is an
Acad. Sci. U.S.A. 103, 1599716002. M. (2010). Novel anti-infective com- Tarkkanen, A. M., Heinonen, T., and open-access article subject to an exclusive
Martnez, J. L. (2008). Antibiotics and pounds from marine bacteria. Mar. Jogi, R. (2009). P1A recombinant license agreement between the authors and
antibiotic resistance genes in natural Drugs 8, 498518. -lactamase prevents emergence of the Frontiers Research Foundation, which
environments. Science 321, 365367. Rasko, D. A., Moreira, C. G., Li de, R., antimicrobial resistance in gut micro- permits unrestricted use, distribution, and
Monk, A. B., Rees, C. D., Barrow, P., Reading, N. C., Ritchie, J. M., Waldor, flora of healthy subjects during intra- reproduction in any medium, provided the
Hagens, S., and Harper, D. R. (2010). M. K., Williams, N., Taussig, R., Wei, S., venous administration of ampicillin. original authors and source are credited.

www.frontiersin.org December 2010 | Volume 1 | Article 134 | 7

Potrebbero piacerti anche