You are on page 1of 17

J Antimicrob Chemother

doi:10.1093/jac/dkz161

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


The use of aminoglycosides in animals within the EU: development of
resistance in animals and possible impact on human and
animal health: a review
Engeline van Duijkeren1*, Christine Schwarz2, Damien Bouchard3, Boudewijn Catry4,5, Constança Pomba6,
Keith Edward Baptiste7, Miguel A. Moreno8, Merja Rantala9, Modestas Ruzauskas10, Pascal Sanders11,
Christopher Teale12, Astrid L. Wester13, Kristine Ignate14, Zoltan Kunsagi14 and Helen Jukes15

1
National Institute for Public Health and the Environment, Bilthoven, The Netherlands; 2Federal Office of Consumer Protection and
Food Safety, Berlin, Germany; 3French Agency for Food, Environmental, and Occupational Safety, National Agency for Veterinary
Medicinal Products, Fougères, France; 4Sciensano, Brussels, Belgium; 5Faculty of Medicine, Université libre de Bruxelles (ULB), Brussels,
Belgium; 6Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal; 7Danish Medicines Agency, Copenhagen, Denmark;
8
Faculty of Veterinary Medicine, Complutense University, Madrid, Spain; 9Faculty of Veterinary Medicine, University of Helsinki, Helsinki,
Finland; 10Lithuanian University of Health Sciences, Kaunas, Lithuania; 11French Agency for Food, Environmental, and Occupational
Safety, Fougères Laboratory, Fougères, France; 12Animal and Plant Health Agency, Weybridge, UK; 13World Health Organization,
Genève, Switzerland; 14European Medicines Agency, Amsterdam, The Netherlands; 15Veterinary Medicines Directorate, Addlestone, UK

*Corresponding author. Tel: !31 30 2743942; Fax: !31 30 2744409; E-mail: engeline.van.duijkeren@rivm.nl

Aminoglycosides (AGs) are important antibacterial agents for the treatment of various infections in humans and
animals. Following extensive use of AGs in humans, food-producing animals and companion animals, acquired
resistance among human and animal pathogens and commensal bacteria has emerged. Acquired resistance
occurs through several mechanisms, but enzymatic inactivation of AGs is the most common one. Resistance
genes are often located on mobile genetic elements, facilitating their spread between different bacterial species
and between animals and humans. AG resistance has been found in many different bacterial species, including
those with zoonotic potential such as Salmonella spp., Campylobacter spp. and livestock-associated MRSA. The
highest risk is anticipated from transfer of resistant enterococci or coliforms (Escherichia coli) since infections
with these pathogens in humans would potentially be treated with AGs. There is evidence that the use of AGs in
human and veterinary medicine is associated with the increased prevalence of resistance. The same resistance
genes have been found in isolates from humans and animals. Evaluation of risk factors indicates that the prob-
ability of transmission of AG resistance from animals to humans through transfer of zoonotic or commensal
foodborne bacteria and/or their mobile genetic elements can be regarded as high, although there are no quanti-
tative data on the actual contribution of animals to AG resistance in human pathogens. Responsible use of AGs is
of great importance in order to safeguard their clinical efficacy for human and veterinary medicine.

Introduction
poorly absorbed from the gut, and penetration of the blood–brain
Aminoglycosides (AGs), introduced in 1944, are among the oldest barrier is minimal.1,2
classes of antimicrobials. AGs are bactericidal antimicrobials that The spectrum of activity of AGs includes Gram-negative bac-
act by impairing bacterial protein synthesis through binding to the teria, staphylococci, mycobacteria, leptospira and certain proto-
30S ribosomal subunit.1 AGs must penetrate into the bacterium to zoa. They have poor efficacy against streptococci, anaerobic
exert their effect, and their uptake in to the bacterial cell is an bacteria and intracellular bacteria. Enterococci and streptococci
oxygen-dependent process. Therefore, the spectrum of activity of generally show a degree of intrinsic resistance to AGs due to imper-
AGs is limited to aerobic and facultative anaerobic bacteria under meability of their cell wall, although penetration into the bacterial
aerobic conditions. AGs are less potent in hyperosmolar environ- cell can be enhanced by other antimicrobials that inhibit cell wall
ments or environments with low pH. In addition, purulent debris at synthesis, such as the b-lactam antibiotics. Therefore, AGs are
the infection site can bind to AGs and inactivate them.1 AGs are often used in combination with b-lactams. This combination also
hydrophilic molecules and relatively insoluble in lipids. They are broadens the spectrum of activity.1

C The Author(s) 2019. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved.
V
For permissions, please email: journals.permissions@oup.com.
1 of 17
Review

In veterinary medicine, AGs are used to treat infections in all Substances used for parenteral applications include (dihydro)-
major food-producing animals and in companion animal species. streptomycin, gentamicin, kanamycin, framycetin, spectinomycin
They are categorized as veterinary critically important antimicro- and neomycin. (Dihydro)streptomycin, neomycin, apramycin, gen-

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


bials (VCIAs) by the World Organisation for Animal Health (OIE)3 tamicin, paromomycin and spectinomycin are used in oral formu-
based on the wide range and nature of the diseases they are used lations.5 The majority of oral formulations (oral solution, oral
to treat in animals and the limited availability of economic alterna- powder, premix) are used for treatments in pigs, calves, sheep,
tives for some infections. Similarly, AGs are classified by WHO as poultry and rabbits. They are generally administered in a once daily
critically important antimicrobials (CIAs-high priority) for human treatment regimen as oral drenches (to neonates) or in feed or
medicine.4 In human medicine in the EU, AGs are important for drinking water/milk over a period of 3–5 (and in exceptional cases
treatment of enterococcal endocarditis, infections with MDR even 7) days. Individual products are authorized for considerably
Gram-negative bacteria (especially Enterobacteriaceae and longer treatment durations, e.g. apramycin for 21 days or up to
Pseudomonas spp.) and MDR TB. Due to their importance in human 28 days. Local applications include ear drops, eye drops, topical ap-
medicine, the public health impact of the use of AGs in animals plication to the skin and intramammary and intrauterine
needs to be assessed. Our objective was to critically review the cur- preparations.5,7
rent knowledge on the usage of AGs, resistance development and
the potential impact of this resistance on animal and human Pharmacokinetic/pharmacodynamic (PK/PD)
health. relationship and dosing regimens
For concentration-dependent antimicrobial agents, optimal dosing
The use of AGs in veterinary medicine involves administration of high doses with long dosing intervals.1
AGs are used extensively in veterinary medicine.5 In 2015, the PK/PD indices have been proposed from in vitro and in vivo
sales of AGs made up 3.5% of the total sales of antimicrobials for infection models and subsequently validated in retrospective or
food-producing species (including horses), in mg/population cor- prospective human clinical trials.8 Two PK/PD indices, Cmax/MIC
rection unit (PCU), aggregated for 30 European countries. AGs are and 24 h AUC/MIC, are the most important PK/PD predictors for
the sixth most commonly used antimicrobial class after tetracy- bacteriological and clinical efficacy of concentration-dependent
clines, penicillins, sulphonamides, macrolides and polymyxins antimicrobials.9–11
(Figure 1).5 There are marked differences in the sales of AGs be- Most authors have proposed the Cmax/MIC ratio as the PK/PD
tween the different EU countries, with the lowest sales in the index of choice for AGs (gentamicin, tobramycin and amikacin).
Scandinavian countries and the highest in Spain (Figure 2); these A Cmax/MIC ratio of 10 had the strongest association, with a posi-
differences are not explained by the differences in overall anti- tive clinical outcome in patients with pneumonia caused by aerobic
microbial use between countries in all cases. The reasons for these Gram-negative rods and with bacteraemia caused by P. aerugi-
differences should be investigated. The most frequently used AGs nosa.12–14 Furthermore, a Cmax/MIC ratio of 10–12 was determined
are neomycin, dihydrostreptomycin and spectinomycin (Figure 3). to minimize the survival and overgrowth of resistant strains.8 If
Other substances from the group that are used in food-producing this preferable Cmax/MIC ratio is obtained, most susceptible bac-
species [where maximum residue limits (MRLs) have been estab- teria die rapidly, and consequently the effect of the duration of
lished] are: apramycin, gentamicin, kanamycin, paromomycin, drug exposure is minimal. Accordingly, in neutropenic and non-
neomycin, framycetin and streptomycin. neutropenic models of infection, significantly more animals sur-
In EU Member States (MSs) the vast majority of available AGs vived a potentially lethal challenge of bacteria when treated with a
are administered to animals for clinical purposes. The most fre- single large dose of an AG rather than with the same dose given on
quent use is therapy for septicaemia, and infections of the digest- an 8 h schedule. A high-dose and infrequent administration of AGs
ive tract, respiratory tract and urinary tract in many animal species has also been shown to reduce the rate of nephrotoxicity.15 These
including cattle, pigs, poultry, sheep, goats, horses, dogs and cats findings, and meta-analyses of different dosing regimens of AGs,
(Table S1 available as Supplementary data at JAC Online). In par- led to a shift in clinical dosing in humans from two to three times
ticular, gentamicin is indicated for Pseudomonas aeruginosa infec- daily administration to once a day treatments.11,16 The actual goal
tions, with few alternative treatments being available.1 AGs have of AG therapy is to maximize peak concentrations to increase effi-
not been authorized as growth promoters in the EU MSs since cacy and, in order to reduce toxicity, to administer once a day to
1976. Before this, neomycin and hygromycin-B were authorized to achieve a low trough concentration, and to reduce treatment
be added to poultry feed for growth promotion only on a national duration.17
level in certain EU MSs.6 Some products containing AGs, especially In veterinary medicine, the situation is more complex because
those with old marketing authorizations, are approved for general of potential interspecies differences in PK and PD as well as differ-
indications such as the treatment of ‘infections caused by suscep- ences in indications and target pathogens.18 In addition, in ani-
tible organisms’ in various animal species.7 mals, AGs are to a large extent administered via the oral route for
the treatment of gastrointestinal infections where they exert their
antibacterial activity in situ without being absorbed. Thus, for veter-
Route of administration in animals inary purposes, human-derived PK/PD concepts cannot be applied
In veterinary medicine, AGs are used for parenteral, oral and topic- for oral applications and may be applied for parenteral applications
al applications. In the EU, approximately half of AG use is as oral by approximation only.
formulations (premix, oral powder or soluble in drinking water) When given via the parenteral route, AGs were traditionally
and about half is as injectable formulations (Figure S1).5 administered every 8–12 h. Recent studies in veterinary patients

2 of 17
Review JAC
2.1% 3.0%
2.8%
3.5%

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


Tetracyclines
32.8% Penicillins
6.8%
Sulphonamides
Trimethoprim
3.2%
Macrolides
Lincosamides
7.2%
Polymyxins
Aminoglycosides

1.9% Pleuromutilins
Fluoroquinolones
Others*

11.8%
25.0%

Figure 1 Sales of antimicrobial agents by antimicrobial class as a percentage of the total sales for food-producing species (including horses), in mg/
PCU, aggregated by 30 European countries, for 2015.5 *Amphenicols, cephalosporins, other quinolones and other antibacterials (classified as such in
the ATCvet system). This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.

likewise support high-dose, once daily therapy with AGs to avoid it should be treated orally with substances such as apramycin that
adaptive resistance and to reduce risks of toxicity. The optimal are considered to have very low oral bioavailability.22 The rationale
doses and the ideal drug monitoring strategy are unknown for for indications for treatment of systemic diseases with AGs that
most target animal species. Dosages have to be modified in neo- have very low oral bioavailability is debatable and the supporting
nates and in animals with impaired liver or kidney function.1 evidence should be reviewed.
In conclusion, dosing regimens should be re-investigated in vet-
Pigs
erinary medicine in order to improve the efficacy and safety of AG
use, and to reduce the risk of antimicrobial resistance. Further re- In pigs, apramycin, gentamicin, paromomycin and neomycin are
search should be conducted into the PK/PD surrogate indices for used for oral treatment of colibacillosis and salmonellosis.23 AGs
treatment of veterinary infections with AGs, as validated parame- are an important alternative to colistin for the treatment of post-
ters are predictive of clinical efficacy and would assist optimization weaning diarrhoea caused by E. coli.24 Dihydrostreptomycin in
of dosing regimens as well as reducing the toxicity of AGs that are combination with benzylpenicillin is authorized for respiratory
administered parenterally. infections caused by Actinobacillus pleuropneumoniae and/or
Pasteurella multocida, and for the treatment of Glässer’s disease
caused by Haemophilus parasuis.
Aminoglycoside use in different animal species
Poultry Cattle
In the EU, neomycin, apramycin, spectinomycin and streptomycin Neomycin, streptomycin, kanamycin and framycetin, in combin-
are authorized.7,19,20 Outside the EU, gentamicin is used as a sub- ation with other antimicrobial agents, are used in preparations for
cutaneous injection in day-old chicks or by in ovo injections. In ovo intramammary administrations to cows with mastitis.7,19,24 In
injection is a route for administration of Marek’s disease vaccin- calves, neomycin and apramycin are used for the treatment of
ation in the USA and, to prevent bacterial contamination of the bacterial enteritis caused by E. coli and salmonellae, and paromo-
eggs, gentamicin is injected in combination with the vaccine.21 In mycin is used for the treatment of enteric infections caused by
ovo injections or other applications of gentamicin in poultry are not E. coli.1,7
authorized in the EU as no MRLs exist for gentamicin in this species. Gentamicin is used to treat respiratory infections caused
Neomycin and apramycin are authorized for oral treatment of en- by Mannheimia haemolytica and P. multocida in calves.7
teric infections, e.g. for the treatment of Escherichia coli and Dihydrostreptomycin or streptomycin is used in the treatment of
Salmonella spp. infections in young chickens. Antimicrobials are leptospirosis in cattle.
not permitted to be used for the specific purpose of control of
Salmonella spp., with certain exceptions [Commission Regulation Horses
(EC) No. 1177/2006]. It is noted that colibacillosis is essentially a AGs (amikacin, neomycin and gentamicin) are commonly used for
systemic infection in poultry and it therefore appears contrary that treatment of bacterial septicaemia, respiratory tract infections,

3 of 17
Review

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


Figure 2 Spatial distribution of veterinary sales of aminoglycosides [amikacin, apramycin, (dihydro)streptomycin, framycetin, gentamicin, kanamycin
and neomycin] for food-producing animals in mg/PCU in 30 European countries for 2015.5 Sales of spectinomycin and paromomycin are not included
here as they are reported under ‘other antimicrobials’ in the ESVAC report. This figure appears in colour in the online version of JAC and in black and
white in the print version of JAC.

peritonitis, osteomyelitis, meningitis, wound infections, joint infec- gentamicin, neomycin and framycetin are used as topical treat-
tions and endometritis, often in combination with other antimicro- ment for infections of the eye (blepharitis, conjunctivitis, kerato-
bials such as b-lactams.25 For infections in neonatal foals, where conjunctivitis and anterior uveitis), ear (otitis externa) and skin.7,19
Gram-negative bacteria are isolated from a high proportion of Gentamicin and tobramycin are effective topically to manage
cases and a bactericidal effect is desirable, there is a preference to otitis externa caused by P. aeruginosa.26 AGs are included for the
select gentamicin or amikacin as empirical therapy whilst awaiting treatment of MDR infections in treatment guidelines for respiratory
culture results.25 Topical application of AGs is recommended for infections in dogs and cats and for deep pyoderma, as third-line
infections of the eye and uterus.25 Amikacin is authorized in some choice.27,28 They are also suggested in guidelines to treat ESBL-
MSs for horses that are kept as companion animals and do not producing E coli urinary tract infections when there is resistance to
enter the food chain. Amikacin cannot be used in food-producing authorized alternatives.29
animals as no MRL has been established.
Combination preparations
Companion animals AGs are often used in combination with other antimicrobials, such
Injections of gentamicin or amikacin are licensed for the treatment as b-lactams, in order to achieve a synergistic effect or to broaden
of septicaemia and respiratory infections. In textbooks, AGs are the spectrum of activity. Streptomycin and neomycin are author-
recommended for the treatment of bacterial peritonitis, metritis, ized in the EU in combination with penicillin for treatment of a
osteomyelitis, leptospirosis and nocardiosis.1 AGs such as broad range of non-specific indications in livestock and companion

4 of 17
Review JAC
3%
3%
6%

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


4%
35% Neomycin
Dihydrostreptomycin
Spectinomycin
18%
Paromomycin
Apramycin
Gentamicin
Streptomycin

30%

Figure 3 Sales of aminoglycosides, spectinomycin and paromomycin in food-producing species, as a percentage of total mg/PCU, aggregated for 30
European countries in 2015 (ESVAC, unpublished data). Minor sales (0.7%) of kanamycin and framycetin were also reported in 2015. This figure
appears in colour in the online version of JAC and in black and white in the print version of JAC.

animals.19 AGs are also used in combination with b-lactams and/ be used against non-tuberculous mycobacterial infections. AGs
or other antimicrobials in intramammary preparations. In pigs and are used for empirical treatment of sepsis, respiratory tract infec-
poultry, spectinomycin/lincomycin combinations are used for the tions, urinary tract infections and some CNS infections if MDR
treatment of various infections.7,19 The rationale for some of these Gram-negative bacteria are suspected to be involved.36
combinations is disputable. Owing to the widespread resistance of Enterococci are intrinsically resistant to low to moderate levels of
many bacterial species to streptomycin, streptomycin/penicillin AGs, but synergism is generally seen when they are combined with
combinations have very limited extra value. In addition, a synergis- a cell wall-active antimicrobial agent. Therefore, AGs are used in
tic effect of this combination has been shown for only a limited combination with a b-lactam or a glycopeptide for the treatment
number of pathogens and only in vitro. The indications for (dihy- of endocarditis caused by Gram-positive cocci without high-level
dro)streptomycin mono-products and its combinations should resistance to AGs. AGs are first-line treatment for plague, brucel-
therefore be re-evaluated. The penicillin/streptomycin combin- losis and tularaemia.37 Aerosolized tobramycin, amikacin and gen-
ation was withdrawn from the US market in 1993 and later by tamicin are used to treat Pseudomonas infections in patients with
Australia since no evidence of clinical synergy was presented.30,31 cystic fibrosis.37,38 Topical applications of various AGs (gentamicin,
tobramycin and neomycin) are utilized for the treatment of ear
Other applications of AGs and eye infections.36,39 Paromomycin is used to treat AIDS patients
AGs are applied in apiculture, aquaculture and in other minor suffering from cryptosporidiosis40 and is an alternative against dif-
species such as rabbits, reptiles and birds, although safety and ferent parasite diseases (amoebiasis and giardiasis) and some-
efficacy have not been established in all cases, with use often times used topically for the treatment of cutaneous leishmaniasis.
being off-label. Gentamicin is utilized as a sperm diluter and as an Spectinomycin is occasionally used for the treatment of gonor-
antimicrobial preservative for vaccines.32 Certain AGs are used rhoea in patients allergic to penicillins (Table 1).
as anthelmintics in animals (destomycin A and hygromycin B). The AGs most used in hospitals are amikacin, gentamicin and
Furthermore, paromomycin, ribostamycin and streptomycin tobramycin.41 The most common route of administration for sys-
are used in horticulture as they have antifungal activity.33 temic infections is parenteral, by intravenous or intramuscular injec-
Streptomycin is also used to control fire blight caused by Erwinia tion. Oral administration is limited to decontamination of the gut
amylovora in orchards.34 in ICUs, since bioavailability following oral administration is low.42
In the European Surveillance of Antimicrobial Consumption
Network (ESAC-Net) survey, including data from 20 European
The use of AGs in human medicine countries, details on the consumption of individual AGs are not
AGs (e.g. tobramycin, gentamicin, amikacin and netilmicin) are reported separately. Available ESAC-Net data from 2015 show
used systemically for treatment of (MDR) Gram-negative infec- that there are large differences in AG consumption between EU
tions such as those caused by Pseudomonas spp., Acinetobacter MSs, consumption being highest in Romania (0.363 DDD per
spp. and Enterobacteriaceae. Kanamycin and amikacin are utilized 1000 inhabitants), Italy (0.289 DDD per 1000 inhabitants) and
for the treatment of MDR TB;35 streptomycin was the first AG to be Bulgaria (0.277 DDD per 1000 inhabitants) whereas consump-
used against TB, but is nowadays rarely used. Amikacin may also tion is much lower in other countries, e.g. in Sweden (0.015 DDD

5 of 17
Review

Table 1 Importance of aminoglycosides in human medicine

Bacterial targets in human medicine (for which Relative frequency

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


availability of class/substance is critically of use in humans in Hazard of resistance transfer between animals
Aminoglycoside important due to few alternatives) the EU and humans

Kanamycin Used for MDR infections including TB. low: rarely used, M. tuberculosis is of limited zoonotic relevance,
generally no plasmid-mediated transfer of resistance
not for first-line determinants in M. tuberculosis.
treatment Enterobacteriaceae—high risk of clonal and
horizontal transfer of resistance genes.
Enterococci—risk of clonal (E. faecalis) and
horizontal (E. faecium and E. faecalis) transfer
of resistance genes.
Gentamicin Gram-negative infections, enterococcal and high see above.
streptococcal endocarditis, brucellosis, tular-
aemia, plague, oral decolonization, impreg-
nated beads to prevent surgical site infections.
Amikacin MDR Gram-negative infections, MDR TB, high see above.
Nocardia spp. infections.
Tobramycin Gram-negative infections, Pseudomonas high see above.
infections in cystic fibrosis.
(Dihydro)streptomycin MDR tuberculosis, but very rarely used. low see above.
Spectinomycin Gonorrhoea in patients allergic to penicillins. low Gonorrhoea is not transmitted to humans from
non-human sources.Transfer of resistance
genes from non-human sources unlikely.
Paromomycin Cryptosporidiosis. low Cryptosporidium parvum is of zoonotic
relevance.
Apramycin No target. not used Selects for gentamicin resistance in
Enterobacteriaceae, such as E. coli and
Salmonella spp.

per 1000 inhabitants) and Finland (0.016 DDD per 1000 inhabi- Resistance mechanisms
tants; Figure 4). In a study of data from the initial ESAC project
describing parenteral antimicrobial treatment in outpatients, Following extensive use of AGs in humans, food-producing animals
AGs were the second most commonly used class (25.27%) after and companion animals, resistance has emerged. Resistance
the cephalosporins (44.58%). At the level of individual mole- occurs through several mechanisms. Resistance genes can be
cules, gentamicin (18.53%) was administered more than the in- located on the chromosome, gene cassettes, plasmids, transpo-
dividual cephalosporins (e.g. ceftriaxone, 17.85%; cefazolin, sons or other mobile elements, thereby increasing the likelihood of
13.16%).43 spread of AG resistance as well as co-resistance to other com-
Consumption data from European countries as outlined above pounds.50 The three main mechanisms of bacterial resistance to
are collected by continuous surveillance data44 aggregated by AGs are the reduction of the intracellular concentration of the anti-
country, although many countries have their own surveillance pro- microbial, the enzymatic modification of the drug and the modifi-
grammes.45,46 Long-term monitoring in the Netherlands has cation of the molecular target.51 Resistance mechanisms are
shown an increase in the systemic use of AGs in Dutch hospitals complex and differ between the AG molecules and between bac-
during the last decade (from 2.5 to 3.7 DDD/100 patient-days be- terial species, and generally there is less cross-resistance when
tween 2006 and 2015). In Norway and Denmark, the use was sta- compared with other classes of antimicrobials.
ble.45,47 Large teaching hospitals tend to have the highest use.41 Decreased intracellular concentrations can result from either
In addition to continuous surveillance as performed by the reduced drug uptake or active efflux mechanisms. Reduced uptake
ESAC survey in outpatients and the hospital sector, targeted point can occur in mutants deficient in components of the electron
prevalence surveys are done in hospitals and long-term care. The transport chain and has been described in Pseudomonas spp.,
latest published data show that on average 34.6% of patients re- E. coli and Staphylococcus aureus.52 Gentamicin resistance by in-
ceive antimicrobial therapy in acute care hospitals versus 4.4% in activation of an outer membrane porin, which serves as an entry
long-term care facilities.48,49 In these settings, the proportion of for gentamicin to the bacterial cell, has also been described.53
AG use was 4.5% and 1.2%, respectively. Considering all agents AG efflux is a significant mechanism in Pseudomonas spp.,
used in acute care, the AGs most used were gentamicin 3.7%, ami- Burkholderia spp. and Stenotrophomonas spp., but has also been
kacin 1.1%, tobramycin 0.4% and netilmicin 0.1%.48 described in other bacteria such as E. coli, Lactococcus lactis and

6 of 17
Review JAC
0.4

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


0.35
DDD per 1000 inhabitants and per day

0.3

0.25

0.2

0.15

0.1

0.05

0
Belgium

United kingdom
Bulgaria

Cyprus*

Denmark

Finland
Croatia

Estonia

France

Greece

Hungary

Ireland

Luxembourg
Italy

Latvia

Lithuania

Malta

Netherlands

Norway

Poland

Portugal

Romania*

Slovakia

Slovenia

Sweden
Figure 4 Total consumption of aminoglycosides expressed as DDD per 1000 inhabitants in European countries in 2015 (source: ESAC-Net142).
*Country provided only total care data.

Acinetobacter baumannii.51,53 There are five families of efflux sys- nucleus or the sugar moieties, preventing ribosomal binding.
tems: the major facilitator superfamily, the ATP-binding cassette Within the three major classes of modifying enzymes, a further
family, the resistance-nodulation division family (RND), the small subdivision can be made based on the target site of the
MDR family as well as the multidrug and toxic compound extrusion enzymes.56 To date, there are four AACs: AAC(1), AAC(20 ), AAC(3)
family.54 The majority of AG transporters belong to the RND fam- and AAC(60 ); five ANTs: ANT(200 ), ANT(300 ), ANT(40 ), ANT(6) and
ily.53 Genes coding for AG efflux mechanisms are most often ANT(9); and seven APHs: APH(200 ), APH(30 ), APH(300 ), APH(4), APH(6),
located on the chromosome, but members of the major facilitator APH(700 ) and APH(9).56 Occasionally several subtypes of these
superfamily can also be located on plasmids.54 The ability of bac- enzymes are present in bacteria. The genes coding for all these
teria to survive antimicrobial challenge without mutation is called enzymes are often located on mobile genetic elements.
adaptive resistance and can be caused by a decreased transport of The AACs catalyse the acetylation of NH2 groups in AGs using
the drug into the bacterial cell.1 Adaptive resistance of P. aerugi- acetyl coenzyme A as donor substrate.51 In addition, the bifunc-
nosa has been shown to be associated with the overproduction of tional enzyme AAC(60 )–APH(200 ) can acetylate and subsequently
the RND efflux system MexXY–OprM.55 The clinical significance of phosphorylate its substrate.51 Among the AACs, AAC(60 ) enzymes
adaptive resistance is that frequent dosing or constant infusion is are the most common and can be found in Gram-positive as well
less effective than high-dose, once daily administration since AGs as Gram-negative bacteria. AAC(1), AAC(2) and AAC(3) are mainly
act in a concentration-dependent manner.1 found in Gram-negative bacteria.51 The substrate profile of AAC(1)
enzymes includes neomycin, apramycin and paromomycin and
that of AAC(20 ) enzymes includes gentamicin, kanamycin, tobra-
Enzymatic drug modification mycin, netilmicin and dibekacin. Enzymes of subclass AAC(3)-I
Roberts et al.56 provide an overview of most acquired resistance confer resistance to fortimicin, sisomicin and gentamicin, while
genes. A few novel spectinomycin resistance genes in staphylo- those of subclass AAC(3)-II confer resistance to gentamicin, tobra-
cocci have subsequently been discovered.57–59 Resistance genes mycin, sisomicin, netilmicin and dibekacin, and AAC(3)-IV to apra-
for AG-modifying enzymes are often found on mobile elements. mycin and gentamicin. AAC(60 ) enzymes cause resistance to
The most common mechanism of resistance to AGs in clinical iso- gentamicin and sometimes amikacin. AAC(60 )-Ib-cr is an enzyme
lates is the production of AG-modifying enzymes such as acetyl- that also confers resistance to selected fluoroquinolones such as
transferases (AACs), phosphotransferases (APHs) and ciprofloxacin.51
nucleotidyltransferases (ANTs).56,60,61 These enzymes modify the The ANTs represent the smallest class of AG-inactivating
AG at the hydroxyl or amino groups of the 2-deoxystreptamine enzymes. These enzymes catalyse the reaction between Mg-ATP

7 of 17
Review

and AGs to form the O-adenylated antimicrobial molecule. To Resistance mechanisms in relevant Gram-negative
date, there are five classes of ANTs categorized depending on the bacteria
position of adenylation on the AG molecule.51 The ANT(200 ) and

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


AG resistance in Enterobacteriaceae relies mainly on the AG-
ANT(300 ) enzymes are more frequent among Gram-negative bac- modifying enzymes (APHs, ANTs and AACs). AAC(3)-II/IV and
teria, whereas the ANT(40 ), ANT(6) and ANT(9) enzymes are most AAC(6)-Ib are the most frequently encountered AACs among E.
often found in Gram-positive bacteria.51,62 ANT(6) enzymes have coli of human and animal origin. Among the ANTs, ANT(200 ) and
streptomycin as their substrate. The ant(6) gene is often found in a ANT(300 ) enzymes are most commonly found in Gram-negative
cluster ant(6)-Ia-sat4-aph(30 )-III that specifies resistance to AGs bacteria.70 The emergence of 16S rRNA methylases in bacteria of
and streptothricin. ANT(9) causes resistance to spectinomycin. animal origin was first identified in Spain in 2005 in an E. coli isolate
ANT(40 ) enzymes confer resistance to tobramycin, amikacin and of pig origin harbouring the armA gene.71 Since then, different
isepamicin. ANT(200 ) mediates resistance to gentamicin, tobra- methylases have been detected in Enterobacteriaceae isolates
mycin, dibekacin, sisomycin and kanamycin. ANT(300 ) are the most from animals in different countries.72–84 The dissemination of the
commonly found ANT enzymes. They specify resistance to spec- armA gene is favoured by its location on a transposon.70 In Spain,
tinomycin and streptomycin.51 seven K. pneumoniae ST11 isolates from dogs and cats were found
APHs catalyse the transfer of a phosphate group to the to be resistant to AGs, and the armA gene was responsible for this
AG molecule. They are widely distributed among bacterial phenotype.84 Although the rmtA gene is rarely reported from ani-
pathogens.51 APH(200 ) plays an important role in Gram-positive mals, it has been found in E. coli isolates from giant pandas in
organisms resistant to gentamicin. APH(30 )-IIIa, generally China.80 In China, the rmtB gene was detected in 69 out of 267
found in Gram-positive bacteria, confers resistance to a broad Enterobacteriaceae isolates collected from pets. The rmtB gene
range of AGs including neomycin, paromomycin, kanamycin was commonly found with ESBL blaCTX-M-9 group genes within the
and amikacin, but not tobramycin or gentamicin. Isolates same incompatibility group (Inc) FII plasmid.74 The rmtB gene was
carrying APH(3) group enzymes show a resistance profile most also found in clinical E. coli isolates originating from bovines in
often including kanamycin, neomycin and paromomycin, and China and France and from healthy pigs in China.72,82,83, rmtD has
APH(30 ) also to amikacin. APH(300 ) mediates resistance to been found in E. coli isolates from diseased chickens in China and a
streptomycin. APH(4) mediates resistance to hygromycin and is horse in Brazil.77,81 The RmtE methylase was reported from E. coli
not clinically relevant. APH(6) enzymes confer resistance to isolates from healthy calves in the USA and from diseased food-
streptomycin. APH(700 ) mediates resistance to hygromycin. producing animals in China.78,79
APH(9) enzymes confer resistance to spectinomycin.51 To date, 16S rRNA methylases do not appear to be common in
veterinary bacteria in EU MSs, but the use of most AGs would select
for resistance as these enzymes result in resistance to almost all
AGs, especially those of clinical relevance in humans.
Target modification AG efflux is a significant mechanism in P. aeruginosa. AG-
Target site modification occurs naturally in AG-producing bacteria: modifying enzymes also confer AG resistance in P. aeruginosa.60 In
the bacterium protects the target by employing enzymes that add A. baumannii, besides AG-modifying enzymes, the armA gene,
a methyl group to specific nucleotides in the 16S rRNA that are es- located on a transposon, is widespread in many countries world-
sential for AG binding, thus inhibiting the antimicrobial action with- wide.60 In addition, rmtB has recently been identified in nine
out interfering with other ribosomal functions. This mechanism A. baumannii isolates from humans in Vietnam.85 To date, no meth-
has been described mainly in different species of the AG-producing ylases have been reported from veterinary Pseudomonas isolates.
genera Streptomyces spp. and Micromonospora spp.62 Nowadays,
the methylation of the ribosomal target responsible for high-level Resistance mechanisms in relevant Gram-positive
AG resistance is an emerging mechanism of great concern in clinic- bacteria
ally relevant Gram-negative bacteria.51,60–65 Resistance to various AGs in staphylococci can be mediated by the
The first plasmid-mediated gene identified was the 16S rRNA genes aac(60 )-Ie-aph(200 )-Ia (kanamycin/gentamicin/tobramycin/
methylase armA.64 To date, 10 additional genes encoding methyl- amikacin resistance), ant(40 )-Ia (kanamycin/neomycin/tobramycin
ases have been reported: rmtA, rmtB, rmtC, rmtD, rmtD2, rmtE, resistance), aph(30 )-III (kanamycin/neomycin/amikacin resist-
rmtF, rmtG, rmtH and npmA.60,65 The genes encoding these deter- ance), apmA (apramycin resistance and decreased susceptibility
minants are usually located on mobile genetic elements and have to gentamicin) and aadE or str (streptomycin resistance).86–89
been associated with genes coding for resistance to other anti- Spectinomycin resistance in staphylococci is mostly mediated by
microbial classes, such as quinolones (Qnr proteins) or b-lactam spectinomycin 9-O-adenyltransferase encoded by the spc gene
antimicrobials (acquired AmpC-b-lactamases or ESBLs). Recently located on a transposon. Resistance in staphylococci to spectino-
these methyltransferases have been found in association with car- mycin can also be due to the plasmid-associated gene spd and the
bapenemases such as NDM-1.66–68 The genes rmtA, rmtB, rmtC, chromosomal- or plasmid-located gene spw.57,59
rmtD, rmtD2, rmtE, rmtF, rmtG and rmtH confer resistance to gen- In Mycobacterium tuberculosis, mutations in the genes rpsL and
tamicin, tobramycin, kanamycin and amikacin, whereas npmA rrs encoding the ribosomal protein S12 and the 16S rRNA, respect-
confers resistance to gentamicin, tobramycin, kanamycin, amika- ively, are responsible for most of the high-level streptomycin resist-
cin, neomycin and apramycin, but not to streptomycin or ance. rrs A1401G is the most frequent mutation conferring
spectinomycin.62,69,70 amikacin and kanamycin resistance.90 Overexpression of the AG

8 of 17
Review JAC
acetyltransferase-encoding gene, eis, has mainly been associated obtained with Vitek 2 and Etest should be interpreted with caution,
with resistance to kanamycin in M. tuberculosis. EIS is a unique en- especially if the EUCAST breakpoints were used. False gentamicin
zyme capable of acetylating multiple positions of any given AG susceptibilities were observed using Vitek 2 and occurred with

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


scaffold.91 This overexpression resulted from either point muta- K. pneumoniae isolates carrying armA.101 Susceptibility testing of
tions in the promoter region of the eis gene or mutations of the Pseudomonas spp. isolates against tobramycin using MALDI-TOF
whiB7 gene, which encodes a putative regulator of the eis gene.92 MS technology has been explored and it was able to distinguish be-
Although the eis gene has been mainly associated with kanamycin tween resistant and susceptible isolates. Therefore, this technique
resistance, amikacin resistance has also been reported.90 The gene has the potential to allow for the susceptibility testing of a much
gidB, when mutated, was found to be associated with low-level wider range of antimicrobial substances in the future.102
streptomycin resistance in M. tuberculosis.93 The gidB gene enco- In conclusion, interpretation of susceptibility testing is impaired
des a 7-methylguanosine methyltransferase that specifically by the lack of veterinary breakpoints for most AGs. Susceptibility
modifies residues in the 16S rRNA (rrs). It is a non-essential gene, testing should be standardized and veterinary clinical breakpoints
and loss-of-function mutations in gidB result in failure to methy- established.
late G527 within the 530 loop of the 16S rRNA molecule. Many dif-
ferent gidB mutations, including deletions, are associated with AG
Occurrence of resistance in bacteria from
resistance, suggesting that loss of function of this gene confers re-
sistance.90 To date, no plasmid-mediated resistance has been animals and humans
reported in M. tuberculosis or M. bovis. Isolates from food-producing animals collected under
EU surveillance programmes
Consideration of susceptibility testing Gram-negatives
Susceptibility data from national monitoring programmes are In the EU monitoring programmes (MARAN, DANMAP and EFSA),
available, and MIC determination via broth microdilution is the ecological cut-off values (ECOFFs) are often used for the interpret-
most frequently used method in these programmes. ation of antimicrobial susceptibility testing in food-producing ani-
Methodologies used differ among countries, since they use differ- mals, and therefore isolates reported as ‘resistant’ in this
ent standards and guidelines (EUCAST, CLSI or country specific), paragraph are not always clinically resistant.103–105 In the
different antimicrobial agents for the same bacteria, different con- Netherlands in 2017, gentamicin resistance in Salmonella spp. iso-
centration ranges for the same antimicrobial agent and different lates from humans, food-producing animals and food products
interpretative criteria.94 In vitro susceptibility testing for many anti- was uncommon (3.4%).103 In Campylobacter spp. isolates from
microbials including AGs is hampered by the fact that standards cattle, pigs and poultry, no gentamicin resistance was found. The
and guidelines for determination of MICs do not include all micro- level of resistance to streptomycin varied between 0% and 2.6%
organisms. Single AGs cannot be used as representatives for the for Campylobacter jejuni isolates from broilers, poultry meat and
whole AG class, since resistance is not a class effect, i.e. there are cattle, and was 8% in Campylobacter coli isolates from broilers and
numerous resistance genes specifying a wide variety of resistance 73% in C. coli isolates from pigs. For E. coli, 5.6%, 0.7%, 0%, 3.8%
mechanisms with different substrate spectra. Resistance to and 2.2% of the isolates originating from broilers, pigs, dairy cattle,
streptomycin and spectinomycin, for example, is distinct from re- white veal calves and rosé veal calves, respectively, were resistant
sistance to gentamicin, kanamycin and/or tobramycin.95 to gentamicin in the Netherlands.103 In Denmark, 5% and 12% of
Alternatively, unrelated enzymes, affecting different sites, can Salmonella Typhimurium isolates from pigs and pork, respectively,
confer the same resistance phenotypes. Despite these difficulties, were resistant to gentamicin. The level of resistance among
the enzymes produced by isolates can sometimes be predicted Danish C. jejuni isolates from broilers and cattle to streptomycin
from susceptibility testing.96 and gentamicin was low (0%–1%).104 The level of resistance to
To date, EUCAST has no veterinary-specific breakpoints. gentamicin of indicator E. coli in Denmark was low in broilers, pigs
VetCAST is the EUCAST subcommittee for Veterinary Antimicrobial and cattle (0%–3%).104 Data for 2016 from the European surveil-
Susceptibility Testing and aims to define clinical breakpoints for
lance monitoring show that resistance to gentamicin in Salmonella
antimicrobial drugs used in veterinary medicine in Europe in the fu-
spp. isolates from broilers, broiler meat, turkeys and turkey meat is
ture.97 CLSI has veterinary-specific breakpoints for amikacin ap-
generally low (5.6%, 2.4%, 7.2% and 4.4%, respectively). There are
plicable to E. coli and P. aeruginosa from dogs, foals and adult
differences between Salmonella serovars: in S. Kentucky from
horses, Staphylococcus spp. from dogs, S. aureus from foals and
broilers and turkeys, resistance to gentamicin was common, 71%
adult horses, Streptococcus spp. from dogs, and Streptococcus equi
and 85% of isolates being non-susceptible.105 Resistance to genta-
subsp. zooepidemicus and subsp. equi from foals and adult
micin in 2016 was rare in C. jejuni from broilers (0.7%), whereas it
horses.98–100
was not found in C. coli isolates.105 Levels of streptomycin resist-
For Enterococcus spp. (E. faecalis, E. faecium, E. gallinarum/
ance were 1.7% for C. jejuni and 12% for C. coli. In indicator E. coli
E. casseliflavus), AGs (except when tested positive for high-level
isolates from broilers, broiler meat, turkeys and turkey meat, resist-
resistance) may appear to be active in vitro, but are not effective
ance to gentamicin was 8.9%, 4.1%, 6.2% and 11%, respectively.
clinically and should not be reported as susceptible. Anaerobic bac-
teria, such as Clostridium spp., Bacteroides spp. and Fusobacterium
canifelinum, are intrinsically resistant to AGs.99 Gram-positives
A recent study demonstrated that results of susceptibility test- In Denmark, gentamicin resistance in E. faecalis was 7% in isolates
ing for gentamicin for K. pneumoniae resistant to carbapenems from pigs, but no high-level resistance was detected.104

9 of 17
Review

Clinical isolates from animals dogs originating from several countries in Europe, the USA and
Gram-negatives Canada, resistance to gentamicin/kanamycin (88.3%), kanamycin
(90.3%), streptomycin (90.3%) and streptothricin (90.3%) was

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


Equine E. coli isolates were generally susceptible to gentamicin, very common.114
the resistance rate being 8.8% in Germany.94 A significant increase
in the percentage of E. coli isolates resistant to gentamicin was Human clinical isolates
identified in equine E. coli isolates from 2007 to 2012 (53.9%) com-
pared with isolates from 1999 to 2004 (28.5%) in the UK.106 In Data from the ECDC EARS-Net surveillance of human clinical cases
France, most equine E. coli isolates were susceptible to strepto- indicate that resistance of Klebsiella pneumoniae isolates to AGs
mycin, neomycin, kanamycin, amikacin and gentamicin (60%– was below 5% in Scandinavian countries and Austria, between 5%
100% susceptibility).107 In France, the percentages of susceptible and 10% in the Netherlands, Germany and the UK, 25%–50% in
feline E. coli were 74% for streptomycin, 92% for kanamycin, 98% France, Italy, Portugal, Lithuania, Hungary, Croatia and the Czech
for gentamicin and 92% for neomycin.107 Canine E. coli isolates Republic and between 50% and 75% in Greece, Bulgaria, Romania,
from urinary tract infections were generally susceptible to genta- Poland and Slovakia.115 For E. coli, AG resistance was ,5% in
micin, neomycin and apramycin (.90% of isolates susceptible) Finland and Iceland, between 5% and 10% in Sweden, Norway,
and 71% were susceptible to streptomycin.107 Integron-borne AG the Netherlands, Germany, France, Belgium, the UK and Austria
resistance was found frequently among avian pathogenic E. coli (among others), 10%–25% in Spain, Italy, Ireland, Greece and
(APEC) in Italy. High levels of resistance were observed for strepto- many eastern European countries and 35% in Bulgaria. High-level
mycin (67.2%), whereas resistance against gentamicin (16.7%), gentamicin resistance in E. faecalis was between 10% and 25% in
kanamycin (14.7%) and apramycin (3.0%) was lower.108 Sweden, Norway, France, Belgium, the Netherlands, Iceland and
Resistance in P. aeruginosa isolates from ear infections of compan- Greece; it was between 25% and 50% in most other EU MSs, while
ion animals was found in 25% of the isolates, while only 41% of a prevalence of 56% was found in Romania. Even higher prevalen-
the isolates were fully susceptible.94 ces of high-level gentamicin resistance were reported for
E. faecium, with levels .70% in the Netherlands, Czech Republic,
Gram-positives Romania, Lithuania and Bulgaria.115 Generally, countries with the
highest human consumption of AGs, such as Bulgaria and
Characterization of 227 Streptococcus suis isolates from pigs dur- Romania, also reported the highest levels of AG resistance.115
ing 2010–13 showed high-level resistance to neomycin (70.0%)
and gentamicin (55.1%), and resistance to AGs was attributed to
aph(30 )-IIIa and aac(60 )Ie-aph(200 )-Ia genes.109 These genes have
Possible links between the use of AGs in animals and
rarely been reported in S. suis.109 Coagulase-positive staphylococci resistance in bacteria of animal origin
isolated from the udder of bovines in France were often susceptible A systematic review on the effect of oral antimicrobials on porcine
to all AGs tested, with 90%–99% of the isolates susceptible to E. coli found that oral administration of AGs increased the preva-
streptomycin, kanamycin, neomycin and gentamicin.107Among S. lence of antimicrobial resistance.116 Sun et al.117 investigated the
aureus originating from infections in dogs, 66%, 71% and 74% effect of treatment of sows with lincomycin, chlortetracycline and
were susceptible to streptomycin, neomycin and kanamycin, re- amoxicillin on resistance development in the intestinal microbiota.
spectively, and 91% were found to be susceptible to gentamicin in The treatment increased the abundance of AG resistance genes,
France.107 Susceptibilities of feline staphylococci were similar. probably due to co-selection. Apramycin and neomycin fed in sub-
Among equine S. aureus isolates, susceptibility to AGs was 80% for therapeutic concentrations to pigs enhanced transfer of an anti-
kanamycin, 82% for gentamicin and 90% for streptomycin.107 microbial resistance-encoding plasmid from commensal E. coli
Resistance to gentamicin, tobramycin and kanamycin was com- organisms to Yersinia and Proteus organisms in an infection model
mon (36%) among MRSA CC398 isolates collected from pigs at using isolated ligated intestinal loops.118 Apramycin consumption
Dutch slaughterhouses.110 Non-susceptibility to gentamicin was at farm level in pigs was most probably driving the increasing oc-
also found among MRSA isolates on broiler farms.88 Non- currence of apramycin/gentamicin cross-resistant E. coli in dis-
susceptibility to gentamicin (40%), neomycin (30%) and amikacin eased pigs and healthy finishers at slaughter in Denmark. The
(1%) was found among 1290 MRSA isolates from pigs, veal calves, duration of use and amounts used both had a significant effect on
poultry and meat in the Netherlands.111 A high prevalence of non- the prevalence of apramycin/gentamicin cross-resistance in dis-
susceptibility to AGs has been reported in MSSA CC398 isolates in eased weaning pigs at the national level.119 Another Danish study
Belgium.112 MRSA CC1 isolates from dairy cattle and humans in investigated the effect of apramycin treatment on transfer and
Italy were often kanamycin resistant and carried aphA3 and sat selection of an MDR E. coli strain in the intestine of pigs, and found
(conferring streptothricin resistance) genes with Tn5405-like ele- that the use of apramycin enhanced the spread of gentamicin-
ments, and contained several markers indicating a human resistant E. coli.120 In a study investigating the influence of oral
origin.113 administration of a fluoroquinolone, an AG and ampicillin on
In Germany, 96% of canine and feline S. aureus isolates from prevalence and patterns of antimicrobial resistance among E. coli
ear infections and 84% of S. aureus from skin infection were sus- and Enterococcus spp. isolated from growing broilers, the overall
ceptible to gentamicin. Gentamicin susceptibility percentages for resistance to all drugs tested reached the highest level among
Staphylococcus pseudintermedius isolates were 87% for isolates enterococci after medication with gentamicin. The frequency of re-
from ear infections and 74% for isolates from skin infections.94 sistance to most antimicrobials tested was significantly higher in
Among 103 methicillin-resistant S. pseudintermedius isolates from E. coli isolated from broilers receiving intermittent antimicrobial

10 of 17
Review JAC
pressure than that from non-medicated broilers.121 On a German fluoroquinolones are one of the few other treatment options be-
broiler farm, resistance to spectinomycin in E. coli isolates sides AGs.1,7,24
increased significantly with age in all three production turns, des- In conclusion, AGs are very important for treatment of a broad

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


pite the fact that the substance was not used on the farm. A pos- range of Gram-negative infections in animals. If AGs were no lon-
sible explanation for this phenomenon was co-selection by the use ger available for veterinary medicine, then it could be speculated
of other antimicrobials.122 Gentamicin resistance increased in clin- that other antimicrobials would replace their use. Alternatives to
ical E. coli isolates from broiler chickens in Québec, despite the fact AGs for the treatment of some MDR Gram-negative infections in
that this antimicrobial was no longer used. This increase coincided animals include antimicrobials that are critically important for the
with the use of a spectinomycin/lincomycin combination for pre- treatment of human infections, such as fluoroquinolones and co-
vention of colibacillosis, including in ovo injection. The major genes listin.126 The consequences of the use of these alternatives instead
identified for resistance to gentamicin and spectinomycin were of AGs should be taken into account in risk assessments.
aac(3)-VI and aadA, respectively. The aadA and aac(3)-VI genes
were located on a modified class 1 integron. Therefore, the use of Impact of resistance on human health
spectinomycin/lincomycin not only resulted in an increase in spec-
tinomycin resistance, but also co-selected for gentamicin All AGs (including streptomycin, neomycin and kanamycin), with
resistance.123 the exception of spectinomycin, are categorized as ‘critically im-
The use of various antimicrobials, including kanamycin, at con- portant’ antimicrobials for human medicine by the WHO, whereas
centrations in vitro far below the MIC, promoted the selection of an spectinomycin is categorized as ‘important’.4 AGs are most often
ESBL-carrying plasmid conferring resistance not only to b-lactams used in combination with b-lactams for the empirical treatment of
but also to AGs, tetracycline, trimethoprim, sulphonamide and a broad range of life-threatening infections in humans.
erythromycin, as well as biocides and heavy metals.124 These find- Nephrotoxicity and ototoxicity and the discovery of less-toxic anti-
ings suggest that low concentrations of antimicrobials present in microbials in recent decades have limited the use of AGs in human
polluted external environments and in the gut of exposed animals medicine.36 However, high levels of resistance to other antimicro-
and humans could allow for selection and enrichment of bacteria bials and MDR in certain bacteria have resulted in renewed interest
with MDR plasmids and thereby contribute to the emergence, in the AGs.
maintenance and transmission of antimicrobial-resistant, disease- Usage of AGs in humans is associated with increased preva-
causing bacteria. lence of resistance in humans. In human isolates from the
In conclusion, there is evidence that the usage of AGs in veterin- Enterobacteriaceae family, there was a significant effect of selec-
ary medicine is associated with the increased prevalence of resist- tion pressure of gentamicin in the selection of resistant K. pneumo-
ance to AGs and other antimicrobial classes in bacteria in animals. niae and E. coli and of amikacin in the selection for resistant E. coli
and Enterobacter cloacae isolates.127 Another study showed that
the abundance of antimicrobial resistance genes more than
Impact of resistance on animal health doubled during selective digestive decontamination with colistin,
Loss of efficacy of AGs could have a serious negative impact on ani- tobramycin and amphotericin B in ICU patients, mainly due to a
mal health and welfare. Although AGs are very important antimi- 6.7-fold increase in AG resistance genes, in particular aph(200 )-Ib
crobials for treatment of animal infections, they are seldom the and an aadE-like gene.128
sole treatment option in the EU. In horses, for example, gentamicin To date, ESBLs conferring resistance to broad-spectrum cepha-
is one of the few available antimicrobials for treating losporins, carbapenemases conferring resistance to carbapenems,
Gram-negative infections, the alternative treatment options being and 16S rRNA methylases conferring resistance to all clinically rele-
trimethoprim/sulphonamide combinations, third- and fourth- vant AGs are the most important causes of concern. In recent
generation cephalosporins and fluoroquinolones.25 Resistance to years, the global dissemination of A. baumannii and
trimethoprim/sulphonamide combinations is very common Enterobacteriaceae, including Salmonella spp., that co-produce
among Gram-negative bacteria, and the latter two mentioned 16S rRNA methylases and carbapenemases such as NDM-1 MBL is
classes are included in Antimicrobial Advice Ad Hoc Expert Group becoming a serious threat to human health.60 The resistance
(AMEG) category 2. Category 2 includes human critically important genes are often co-located on the same plasmid. In addition to
antimicrobials also used in veterinary medicine where the associ- 16S rRNA methylases, resistance to AGs in both Gram-positive and
ated risk for public health is estimated to be high. The use of cat- Gram-negative clinical isolates is often related to the production of
egory 2 antimicrobials should be restricted to conditions where no modifying enzymes of several classes.51 It should be noted that a
alternative treatment options are available.125 In pigs, AGs are im- systematic review assessed mortality, treatment failures and anti-
portant drugs for the treatment of post-weaning diarrhoea.1,24 microbial resistance by comparing b-lactam therapy versus any
Alternatives include tetracycline, trimethoprim/sulphonamide combination of a b-lactam with an AG for human cases of blood-
combinations and ampicillin/amoxicillin, but the prevalence of re- stream infections. The authors concluded that the addition of an
sistance among E. coli to these antimicrobials is high. Other alter- AG to b-lactams for treatment of sepsis should be discouraged,
natives include colistin or quinolones.7,24 With regard to infections since mortality rates were not improved and the addition of AGs
with Pseudomonas spp., AGs are one of the few treatment considerably increased the risk for nephrotoxicity. The subgroup of
options.3 In companion animals, AGs are used to treat ear and eye P. aeruginosa infections was underpowered to examine effects.129
infections caused by Pseudomonas spp. by topical application of Besides infections with MDR Enterobacteriaceae, Pseudomonas
drops or ointments; alternatives include polymyxins and fluoroqui- spp. and Acinetobacter spp., MDR TB and Gram-positive endocardi-
nolones.26 For systemic treatment of Pseudomonas infections, tis are among the diseases for which the availability of AGs is

11 of 17
Review

critically important due to few alternatives.125 For endocarditis identified in E. coli and S. Typhimurium from animals in France and
caused by enterococci without high-level AG resistance, ampicillin the UK.140–142 A high degree of genetic homology between plas-
combined with gentamicin is considered the regimen of first mids harbouring aac(3)IV and hphB of human and animal origin

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


choice. During the last decade, alternative treatment options has been demonstrated.140,141 In a prospective study on a pig
including high-dose daptomycin and the combination of ampicillin farm, Hunter et al.143 demonstrated a widespread dissemination
with ceftriaxone have been explored and have been shown to be of plasmids harbouring aac(3)IV in E. coli from pigs, calves, the
equally effective in certain studies.130–132 farmer and the environment. K. pneumoniae with a slightly smaller
In conclusion, AGs are important drugs in human medicine for conjugative plasmid and a similar resistance pattern was isolated
the treatment of infections with MDR Gram-negative bacteria, from the farmer’s wife, indicative of animal to human transmission
enterococcal endocarditis and MDR TB, but they are seldom the of resistance genes.
only therapeutic option.
Gram-positives
Transmission of resistance and determinants between The risk of transmission of MDR TB from animals to humans is
animals and humans limited, as to date the main resistance mechanism for mycobacte-
AGs are frequently used in veterinary and human medicine, and re- ria is chromosomal mutation. In addition, TB in humans is mostly
sistance has emerged. Resistance can be due to chromosomal caused by Mycobacterium tuberculosis, which is mainly transmit-
mutations, but resistance determinants are often located on mo- ted from humans to humans. Bovine TB (Mycobacterium bovis) is a
bile elements such as transposons, integrons and plasmids. The reportable disease in EU MSs and has been eradicated in many EU
same resistance genes and mobile genetic elements have been MSs. During the years 2006–12, the proportion of cattle herds
found in isolates from animals and humans.88,133–135 In addition, infected or positive for M. bovis in the EU (all MSs) was at a very low
resistance to AGs has been found in bacteria that can cause food- level, ranging from 0.37% in 2007 to 0.67% in 2012.144 The emer-
borne infections in humans, such as Salmonella spp. and gence of MDR M. bovis strains in the EU is regarded as unlikely be-
Campylobacter spp., although AGs are generally not used to treat cause animals infected with TB are not treated but are culled.
Salmonella or Campylobacter infections in humans. Livestock-associated MRSA CC398 (LA-MRSA) isolates from vet-
erinarians in Belgium and Denmark were often resistant to genta-
micin, kanamycin and tobramycin mediated by aac(60 )-aph(2a00 )
Gram-negatives
or aadC, and LA-MRSA carriage was significantly associated with
Antimicrobial resistance in several Salmonella enterica serovars is contact with livestock.145 This indicates that LA-MRSA resistant to
due to genomic islands carrying a class 1 integron, which carries AGs can be transmitted between animals and humans.
the relevant resistance genes. Salmonella genomic island 1 (SGI1) Molecular epidemiological studies based on MLST have
was found in Salmonella enterica serovar Typhimurium Definitive revealed that the vast majority of E. faecium isolates causing clinic-
phage type (DT) 104 ("S. Typhimurium DT 104) isolates, which are al infections and nosocomial outbreaks in humans belong to a glo-
resistant to ampicillin, chloramphenicol, florfenicol, streptomycin, bally dispersed polyclonal subpopulation, genotypically different
spectinomycin, sulphonamides and tetracyclines. Several from E. faecium strains colonizing animals and healthy humans in
Salmonella serovars have been shown to harbour SGI1 or related the community. There was a significant discrepancy in accessory
islands. SGI1 is an integrative mobilizable element and can be gene content between hospital- and community-acquired ampi-
transferred experimentally into E. coli.134 Co-selection of resistance cillin-resistant E. faecium that includes putative virulence and anti-
to all these antimicrobials can potentially result from the use of microbial resistance genes, and indicates that if zoonotic transfer
AGs if SGI1 is present. ESBL or plasmidic AmpC b-lactamase-pro- occurs, it only occurs infrequently.146 Although E. faecium isolates
ducing (ESBL/pAmpC) Enterobacteriaceae are widely distributed from animals do not seem to be a direct hazard to human health
among human and animal populations.135 ESBL- and by clonal transmission, they could act as donors of resistance
carbapenemase-encoding plasmids frequently bear resistance genes to pathogenic enterococci through horizontal gene transfer.
determinants for other antimicrobial classes, including AGs and For E. faecalis, the same MLST types can be detected in isolates
fluoroquinolones, a key feature that fosters the spread of MDR in from food, animals and patients with clinical infections, and there-
Enterobacteriaceae.136 Transmission of ESBL/pAmpC E. coli from fore the zoonotic potential is higher.147
animals to humans can potentially occur by direct contact, Generally, the risk of the emergence and transfer of AG resist-
through the food chain or the environment.135 Evidence for clonal ance resulting from the use of oral products, which are indicated
transmission of ESBL-producing E. coli between humans and mostly to treat enteric infections in pigs, chickens and calves, is
broilers has been found on conventional broiler farms, and hori- anticipated to be much higher than for other formulations as the
zontal gene transfer was suspected on both conventional and or- former products are used as mass medication and, as AGs are not
ganic farms.137,138 absorbed from the gut, the gut flora is exposed to considerable se-
Combined apramycin and hygromycin B resistance in lective pressure.148 The risk for infections with resistant bacteria in
Enterobacteriaceae is mediated by the aac(3)IV and hphB genes. humans resulting from the use of topical products including drops
These genes are part of one resistance gene operon associated used to treat eye and ear infections (mainly infections with
with an insertion sequence, IS140, which is found on plasmids.139 Pseudomonas spp.) in companion animals is generally regarded as
The organization of aac(3)IV and hphB is such that they are co- low, since treatment involves individual animals and this local
transferred.139 Apramycin is used exclusively in animals. The gene route of administration does not result in a selective pressure on
aac(3)IV, conferring cross-resistance to gentamicin, was first the gut flora. This also holds for the use of AGs as intramammary

12 of 17
Review JAC
preparations for the treatment of mastitis in cattle, although their 7 FIDIN 2019. Online FIDIN Repertorium Diergeneesmiddelen. https://reperto
use as dry cow therapy might result in a somewhat higher risk as rium.fidin.nl/.
more individuals are treated (unless selective treatment is prac- 8 Toutain P-L, Del Castillo JR, Bousquet-Mélou A. The pharmacokinetic–

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


tised) and longer-acting preparations are used. The risk for the pharmacodynamic approach to a rational dosage regimen for antibiotics. Res
Vet Sci 2002; 73: 105–14.
emergence of antimicrobial resistance from the use of AGs as in-
jectable formulations will generally be lower if animals are treated 9 Craig WA. Interrelationship between pharmacokinetics and pharmaco-
dynamics in determining dosage regimens for broad-spectrum cephalospor-
individually rather than as a group.
ins. Diagn Microbiol Infect Dis 1995; 22: 89–96.
In humans, AGs are mostly used to treat infections caused by
10 Jacobs M. Optimisation of antimicrobial therapy using pharmacokinetic
bacteria that are not transmitted via food or contact with animals.
and pharmacodynamic parameters. Clin Microbiol Infect 2001; 7: 589–96.
However, they are also used for treatment of zoonotic infections
11 Tulkens PM. The Pharmacological and Microbiological Basis of PK/PD: Why
such as TB (M. bovis), brucellosis and tularaemia. Even bacteria Did We Need to Invent PK/PD in the First Place? 2005. http://www.facm.ucl.ac.
causing human infections not directly originating from animals be/conferences/2005/Manila-24th-ICC-06-05/ISAP-Pharmacodynamics-
may acquire resistance determinants from commensal bacteria why-05-06-05.pdf.
transmitted from animals, such as Enterobacteriaceae and enter- 12 Kashuba AD, Nafziger AN, Drusano GL et al. Optimizing aminoglycoside
ococci, but the extent to which this occurs is unknown. therapy for nosocomial pneumonia caused by gram-negative bacteria.
Antimicrob Agents Chemother 1999; 43: 623–9.
Conclusions 13 Moore RD, Lietman PS, Smith CR. Clinical response to aminoglycoside
therapy: importance of the ratio of peak concentration to minimal inhibitory
These data show that the probability of transfer of AG resistance concentration. J Infect Dis 1987; 155: 93–9.
from animals to humans is high, especially in Enterobacteriaceae, 14 Zelenitsky SA, Harding GK, Sun S et al. Treatment and outcome of
LA-MRSA and enterococci. Further research is needed to elaborate Pseudomonas aeruginosa bacteraemia: an antibiotic pharmacodynamic ana-
on the link between the use of AGs in animals and the impact on lysis. J Antimicrob Chemother 2003; 52: 668–74.
public health. Since very few new and effective antimicrobials for 15 Ambrose PG, Owens RC, Grasela D. Antimicrobial pharmacodynamics.
the treatment of infections due to MDR Gram-negative bacteria Med Clin North Am 2000; 84: 1431–46.
are likely to be launched in the near future, there is an urgent need 16 Frimodt-Møller N. How predictive is PK/PD for antibacterial agents? Int J
to implement strategies that may slow the development of Antimicrob Agents 2002; 19: 333–9.
acquired resistance. Responsible use in both human and veterinary 17 Van Bambeke F, Tulkens PM. Optimizing aminoglycoside dosage based on
medicine is of great importance. PK/PD. 2011. http://www.facm.ucl.ac.be/Advanced-Courses/WBI-Vietnam-
April-2011/5A-Aminoglycosides.pdf.
18 Toutain P-L. Pharmacokinetic/pharmacodynamic integration in drug de-
Transparency declarations velopment and dosage-regimen optimization for veterinary medicine. AAPS J
None to declare. 2002; 4: 160–88.
19 Veterinary Medicines Directorate. Product Information Database. http://
www.vmd.defra.gov.uk/ProductInformationDatabase/.
Supplementary data 20 Norwegian Medicines Agency 2003. Medikamentell behandling av
fjørfe. https://legemiddelverket.no/Documents/Veterin%C3%A6rmedisin/
Table S1 and Figure S1 are available as Supplementary data at JAC Online. Terapianbefalinger/Medikamentell%20behandling%20av%20fj%C3%B8rfe.
pdf.
21 Bailey J, Line E. In ovo gentamicin and mucosal starter culture to control
References Salmonella in broiler production. J Appl Poultry Res 2001; 10: 376–9.
1 Dowling PM. Aminoglycosides. In: S Giguère, JF Prescott, JF Baggot et al., 22 Afifi N, Ramadan A. Kinetic disposition, systemic bioavailability and
eds. Antimicrobial Therapy in Veterinary Medicine. Ames, IA, USA: Blackwell tissue distribution of apramycin in broiler chickens. Res Vet Sci 1997; 62:
Publishing, 2006: 207–29. 249–52.
2 Nau R, Sörgel F, Eiffert H. Penetration of drugs through the blood– 23 Norwegian Medicines Agency 2012. Bruk av antibakterielle midler til pro-
cerebrospinal fluid/blood–brain barrier for treatment of central nervous sys- duksjonsdyr. https://legemiddelverket.no/Documents/Veterin%C3%A6rmedi
tem infections. Clin Microbiol Rev 2010; 23: 858–83. sin/Terapianbefalinger/Terapianbefaling_bruk%20av%20antibakteri
3 OIE 2007. OIE List of Antimicrobials of Veterinary Importance. https://www. ellt%20midler%20til%20produks.pdf.
oie.int/doc/ged/D9840.PDF. 24 FVE 2017. Antimicrobial Use in Food-Producing Animals (Annex A of the
4 WHO 2017. Critically Important Antimicrobials for Human Medicine (5th re- RONAFA opinion). http://www.ema.europa.eu/docs/en_GB/document_library/
vision) 2016. http://www.who.int/foodsafety/publications/antimicrobials- Report/2017/01/WC500220031.pdf.
fifth/en/. 25 Giguère S. Antimicrobial drug use in horses. In: S Giguère, JF Prescott, JD
5 EMA/ESVAC 2017. European Medicines Agency, European Surveillance of Baggot et al., eds. Antimicrobial Therapy in Veterinary Medicine. Ames, IA,
Veterinary Antimicrobial Consumption. Sales of Veterinary Antimicrobial USA: Blackwell Publishing, 2006: 449–62.
Agents in 30 European Countries in 2015 (EMA/184855/2017). Trends from 26 Barnard N, Foster A. Pseudomonas otitis in dogs: a general practitioner’s
2010 to 2015. https://www.ema.europa.eu/documents/report/seventh- guide to treatment. In Pract 2017; 39: 892.
esvac-report-sales-veterinary-antimicrobial-agents-30-european-countries- 27 Lappin M, Blondeau J, Boothe D et al. Antimicrobial use guidelines for
2015_en.pdf. treatment of respiratory tract disease in dogs and cats: antimicrobial guide-
6 Castanon J. History of the use of antibiotic as growth promoters in lines working group of the International Society for Companion Animal
European poultry feeds. Poult Sci 2007; 86: 2466–71. Infectious Diseases. J Vet Intern Med 2017; 31: 279–94.

13 of 17
Review

28 Beco L, Guaguere E, Méndez CL et al. Suggested guidelines for using sys- eu/sites/portal/files/media/en/publications/Publications/0512-TED-PPS-HAI-
temic antimicrobials in bacterial skin infections: part 2—antimicrobial choice, antimicrobial-use-protocol.pdf.
treatment regimens and compliance. Vet Rec 2013; 172: 156–60. 49 ECDC 2014. Point Prevalence Survey of Healthcare Associated Infections

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


29 DSAVA 2015. Antibiotic Use Guidelines for Companion Animal Practice. and Antimicrobial Use in European Long-Term Care Facilities. https://ecdc.eur
https://www.ddd.dk/sektioner/familiedyr/antibiotikavejledning/Documents/ opa.eu/sites/portal/files/media/en/publications/Publications/healthcare-asso
AntibioticGuidelines%20-%20v1.4_jun15.pdf. ciated-infections-point-prevalence-survey-long-term-care-facilities-2013.
30 Gloyd J. Regulatory front: penicillin/streptomycin combinations to dis- pdf.
appear in 1993. J Am Vet Med Assoc 1992; 201: 1826. 50 Ramirez MS, Nikolaidis N, Tolmasky ME. Rise and dissemination of amino-
31 NRA 1999. Review of (Dihydro) Streptomycin/Penicillin Combination glycoside resistance: the aac (60 )-Ib paradigm. Front Microbiol 2013; 4: 121.
Products and (Dihydro) Streptomycin Products. https://apvma.gov.au/sites/de 51 Ramirez MS, Tolmasky ME. Aminoglycoside modifying enzymes. Drug
fault/files/publication/15006-streptomycinpenicillin-review-final-report.pdf. Resist Updat 2010; 13: 151–71.
32 Price S, Aurich J, Davies-Morel M et al. Effects of oxygen exposure and 52 Taber HW, Mueller J, Miller P et al. Bacterial uptake of aminoglycoside
gentamicin on stallion semen stored at 5 and 15 C. Reprod Domest Anim antibiotics. Microbiol Rev 1987; 51: 439.
2008; 43: 261–6. 53 Poole K. Aminoglycoside resistance in Pseudomonas aeruginosa.
33 Lee H, Kim Y, Kim J et al. Activity of some aminoglycoside antibiotics Antimicrob Agents Chemother 2005; 49: 479–87.
against true fungi, Phytophthora and Pythium species. J Appl Microbiol 2005; 54 Poole K. Efflux-mediated multiresistance in Gram-negative bacteria. Clin
99: 836–43. Microbiol Infect 2004; 10: 12–26.
34 Stockwell V, Duffy B. Use of antibiotics in plant agriculture. Rev Sci Tech 55 Hocquet D, Vogne C, El Garch F et al. MexXY–OprM efflux pump is neces-
Off Int Epiz 2012; 31: 199–210. sary for adaptive resistance of Pseudomonas aeruginosa to aminoglycosides.
35 Labby KJ, Garneau-Tsodikova S. Strategies to overcome the action of Antimicrob Agents Chemother 2003; 47: 1371–5.
aminoglycoside-modifying enzymes for treating resistant bacterial infec- 56 Roberts MC, Schwarz S, Aarts HJ. Erratum: acquired antibiotic resistance
tions. Future Med Chem 2013; 5: 1285–309. genes: an overview. Front Microbiol 2012; 3: 384.
36 Poulikakos P, Falagas ME. Aminoglycoside therapy in infectious diseases. 57 Jamrozy D, Coldham N, Butaye P et al. Identification of a novel plasmid-
Expert Opin Pharmacother 2013; 14: 1585–97. associated spectinomycin adenyltransferase gene spd in methicillin-resistant
37 Jackson J, Chen C, Buising K. Aminoglycosides: how should we use them Staphylococcus aureus ST398 isolated from animal and human sources. J
in the 21st century? Curr Opin Infect Dis 2013; 26: 516–25. Antimicrob Chemother 2014; 69: 1193–6.
38 Brodt AM, Stovold E, Zhang L. Inhaled antibiotics for stable non-cystic fi- 58 Wendlandt S, Kadlec K, Schwarz S. Four novel plasmids from
brosis bronchiectasis: a systematic review. Eur Respir J 2014; 44: 382–93. Staphylococcus hyicus and CoNS that carry a variant of the spectinomycin re-
39 Agence française de sécurité sanitaire des produits de santé. Update on sistance gene spd. J Antimicrob Chemother 2014; 70: 948–9.
good use of injectable aminoglycosides, gentamycin, tobramycin, netilmycin, 59 Wendlandt S, Li B, Lozano C et al. Identification of the novel spectino-
amikacin. Pharmacological properties, indications, dosage, and mode of ad- mycin resistance gene spw in methicillin-resistant and methicillin-susceptible
ministration, treatment monitoring. Med Mal Infect 2012; 42: 301. Staphylococcus aureus of human and animal origin. J Antimicrob Chemother
40 Fichtenbaum CJ, Ritchie DJ, Powderly WG. Use of paromomycin for treat- 2013; 68: 1679–80.
ment of cryptosporidiosis in patients with AIDS. Clin Infect Dis 1993; 16: 60 Potron A, Poirel L, Nordmann P. Emerging broad-spectrum resistance in
298–300. Pseudomonas aeruginosa and Acinetobacter baumannii: mechanisms and
41 Ingenbleek A, Van Gastel E, Costers M et al. 2015 National Report on epidemiology. Int J Antimicrob Agents 2015; 45: 568–85.
Consumption of Systemic Antimicrobial Agents in Belgian Hospitals 61 van Hoek AH, Mevius D, Guerra B et al. Acquired antibiotic resistance
2007–2013. https://www.sciensano.be/en/biblio/national-report-consump genes: an overview. Front Microbiol 2011; 2: 203.
tion-systemic-antimicrobial-agents-belgian-hospitals-2007-2013. 62 Wachino J-I, Arakawa Y. Exogenously acquired 16S rRNA methyltransfer-
42 Huttner B, Haustein T, Uckay I et al. Decolonization of intestinal carriage ases found in aminoglycoside-resistant pathogenic Gram-negative bacteria:
of extended-spectrum b-lactamase-producing Enterobacteriaceae with oral an update. Drug Rest Updat 2012; 15: 133–48.
colistin and neomycin: a randomized, double-blind, placebo-controlled trial. J 63 Shaw K, Rather P, Hare R et al. Molecular genetics of aminoglycoside re-
Antimicrob Chemother 2013; 68: 2375–82. sistance genes and familial relationships of the aminoglycoside-modifying
43 Coenen S, Muller A, Adriaenssens N et al. European Surveillance of enzymes. Microbiol Rev 1993; 57: 138.
Antimicrobial Consumption (ESAC): outpatient parenteral antibiotic treat- 64 Galimand M, Courvalin P, Lambert T. Plasmid-mediated high-level resist-
ment in Europe. J Antimicrob Chemother 2009; 64: 200–5. ance to aminoglycosides in Enterobacteriaceae due to 16S rRNA methylation.
44 ECDC 2014. Surveillance of Antimicrobial Consumption in Europe 2012. Antimicrob Agents Chemother 2003; 47: 2565–71.
http://ecdc.europa.eu/en/publications/Publications/antimicrobial-consump 65 O’Hara JA, McGann P, Snesrud EC et al. Novel 16S rRNA methyltransferase
tion-europe-esac-net-2012.pdf. RmtH produced by Klebsiella pneumoniae associated with war-related
45 DANMAP 2017. Use of Antimicrobial Agents and Occurrence of trauma. Antimicrob Agents Chemother 2013; 57: 2413–6.
Antimicrobial Resistance in Bacteria from Food Animals, Food and Humans in 66 Berçot B, Poirel L, Nordmann P. Updated multiplex polymerase chain re-
Denmark. http://www.danmap.org/Downloads/Reports.aspx. action for detection of 16S rRNA methylases: high prevalence among NDM-1
46 Nethmap 2017. Consumption of Antimicrobial Agents and Antimicrobial producers. Diagn Microbiol Infect Dis 2011; 71: 442–5.
Resistance Among Medically Important Bacteria in the Netherlands. https:// 67 Hidalgo L, Hopkins KL, Gutierrez B et al. Association of the novel aminogly-
www.rivm.nl/bibliotheek/rapporten/2017-0056.pdf. coside resistance determinant RmtF with NDM carbapenemase in
47 NORM/NORM-VET 2014. Usage of Antimicrobial Agents and Occurrence of Enterobacteriaceae isolated in India and the UK. J Antimicrob Chemother
Antimicrobial Resistance in Norway. https://www.vetinst.no/en/surveillance- 2013; 68: 1543–50.
programmes/norm-norm-vet-report. 68 Ho PL, Lo WU, Yeung MK et al. Complete sequencing of pNDM-HK encod-
48 ECDC 2012. Point Prevalence Survey of Healthcare-associated Infections ing NDM-1 carbapenemase from a multidrug-resistant Escherichia coli strain
and Antimicrobial Use in European Acute Care Hospitals. https://ecdc.europa. isolated in Hong Kong. PloS One 2011; 6: e17989.

14 of 17
Review JAC
69 Garneau-Tsodikova S, Labby KJ. Mechanisms of resistance to aminogly- 89 Wendlandt S, Kadlec K, Feßler AT et al. Resistance phenotypes and geno-
coside antibiotics: overview and perspectives. Med Chem Commun 2016; 7: types of methicillin-resistant Staphylococcus aureus isolates from broiler
11–27. chickens at slaughter and abattoir workers. J Antimicrob Chemother 2013;

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


70 .Poirel L, Madec JY, Lupo A et al. Antimicrobial resistance in Escherichia 68: 2458–63.
coli. Microbiol Spectr 2018; 6: 1–27. 90 Cohen K, Bishai W, Pym A. Molecular basis of drug resistance in
71 Gonzalez-Zorn B, Teshager T, Casas M et al. armA and aminoglycoside re- Mycobacterium tuberculosis. Microbiol Spectr 2014; 2: 1–16.
sistance in Escherichia coli. Emerg Infect Dis 2005; 11: 954–6. 91 Chen W, Biswas T, Porter VR et al. Unusual regioversatility of acetyltrans-
72 Chen L, Chen ZL, Liu JH et al. Emergence of RmtB methylase-producing ferase Eis, a cause of drug resistance in XDR-TB. Proc Natl Acad Sci USA 2011;
Escherichia coli and Enterobacter cloacae isolates from pigs in China. J 108: 9804–8.
Antimicrob Chemother 2007; 59: 880. 92 Sowajassatakul A, Prammananan T, Chaiprasert A et al. Molecular char-
73 Davis MA, Baker KN, Orfe LH et al. Discovery of a gene conferring multiple- acterization of amikacin, kanamycin and capreomycin resistance in M/XDR-
aminoglycoside resistance in Escherichia coli. Antimicrob Agents Chemother TB strains isolated in Thailand. BMC Microbiol 2014; 14: 165.
2010; 54: 2666–9. 93 Spies FS, Da Silva PEA, Ribeiro MO et al. Identification of mutations related
74 Deng Y, He L, Chen S et al. F33:A-:B- and F2:A-:B- plasmids mediate dis- to streptomycin resistance in clinical isolates of Mycobacterium tuberculosis
semination of rmtB-blaCTX-M-9 group genes and rmtB-qepA in and possible involvement of efflux mechanism. Antimicrob Agents
Enterobacteriaceae isolates from pets in China. Antimicrob Agents Chemother 2008; 52: 2947–9.
Chemother 2011; 55: 4926–9. 94 Schwarz S, Kadlec K, Silley P. Antimicrobial Resistance in Bacteria of
75 Du XD, Wu CM, Liu HB et al. Plasmid-mediated ArmA and RmtB 16S rRNA Animal Origin. Steinen: ZETT-Verlag, 2013.
methylases in Escherichia coli isolated from chickens. J Antimicrob 95 Schwarz S, Silley P, Simjee S et al. Editorial: assessing the antimicrobial
Chemother 2009; 64: 1328–30. susceptibility of bacteria obtained from animals. J Antimicrob Chemother
76 Liu JH, Deng YT, Zeng ZL et al. Coprevalence of plasmid-mediated quin- 2010; 65: 601.
olone resistance determinants QepA, Qnr, and AAC(6’)-Ib-cr among 16S 96 Livermore DM, Winstanley TG, Shannon KP. Interpretative reading: recog-
rRNA methylase RmtB-producing Escherichia coli isolates from pigs. nizing the unusual and inferring resistance mechanisms from resistance phe-
Antimicrob Agents Chemother 2008; 52: 2992–3.
notypes. J Antimicrob Chemother 2001; 48: 87–102.
77 Leigue L, Warth JF, Melo LC et al. MDR ST2179-CTXM-15 Escherichia coli 97 Toutain PL, Bousquet-Mélou A, Damborg P et al. En route towards
co-producing RmtD and AAC(6)-Ib-cr in a horse with extraintestinal infection,
European clinical breakpoints for veterinary antimicrobial susceptibility test-
Brazil. J Antimicrob Chemother 2015; 70: 1263–5.
ing: a position paper explaining the VetCAST approach. Front Microbiol 2017;
78 Lee CS, Hu F, Rivera JI et al. Escherichia coli sequence type354 coproduc- 8: 2344.
ing CMY-2 cephalosporinase and RmtE 16S rRNA methyltransferase.
98 Clinical and Laboratory Standards Institute. Performance Standards for
Antimicrob Agents Chemother 2014; 58: 4246–7.
Antimicrobial Disk and Dilution Susceptibility Tests for Bacteria Isolated from
79 Xia J, Sun J, Li L et al. First report of the IncI1/ST898 conjugative plasmid Animals, 3rd Edition (VET01S-Ed3). CLSI, Wayne, PA, USA, 2015.
carrying rmtE2 16S rRNA methyltransferase gene in Escherichia coli.
99 Clinical and Laboratory Standards Institute. Performance Standards for
Antimicrob Agents Chemother 2015; 59: 7921–2.
Antimicrobial Susceptibility Testing, 26th Edition (M100-S26). CLSI, Wayne, PA,
80 Zou W, Li C, Yang X et al. Frequency of antimicrobial resistance and inte- USA, 2015.
gron gene cassettes in Escherichia coli isolated from giant pandas
100 Clinical and Laboratory Standards Institute. Performance Standards for
(Ailuropoda melanoleuca) in China. Microb Pathog 2018; 116: 173–9.
Antimicrobial Disk and Dilution Susceptibility Tests for Bacteria Isolated from
81 Yang Y, Zhang A, Lei C et al. Characteristics of plasmids coharboring 16S
Animals, 4th Edition (VET01A4E). CLSI, Wayne, PA, USA, 2013.
rRNA methylases, CTX-M, and virulence factors in Escherichia coli and
Klebsiella pneumoniae isolates from chickens in China. Foodborne Pathog Dis 101 Arena F, Giani T, Vaggelli G et al. Accuracy of different methods for sus-
2015; 12: 873–80. ceptibility testing of gentamicin with KPC carbapenemase-producing
Klebsiella pneumoniae. Diagn Microbiol Infect Dis 2015; 81: 132–4.
82 Lupo A, Saras E, Madec JY et al. Emergence of blaCTXM-55 associated
with fosA, rmtB and mcr gene variants in Escherichia coli from various animal 102 Jung J, Eberl T, Sparbier K et al. Rapid detection of antibiotic resistance
species in France. J Antimicrob Chemother 2018; 73: 867–72. based on mass spectrometry and stable isotopes. Eur J Clin Microbiol Infect
Dis 2014; 33: 949–55.
83 Yu T, He T, Yao H et al. Prevalence of 16S rRNA methylases gene rtmB
among Escherichia coli isolated from bovine mastitis in Ningxia, China. 103 MARAN 2018. https://www.wur.nl/upload_mm/7/b/0/5e568649-c674-
Foodborne Pathog Dis 2015; 12: 770–7. 420e-a2ca-acc8ca56f016_Maran%202018.pdf.
84 Hidalgo L, Gutierrez B, Ovejero CM et al. Klebsiella pneumoniae ST11 from 104 DANMAP 2018. Use of Antimicrobial Agents and Occurrence of
companion animals bearing ArmA methyltransferase, DHA-1 b-lactamase Antimicrobial Resistance in Bacteria from Food Animals, Food and Humans in
and QnrB4. Antimicrob Agents Chemother 2013; 57: 4532–4. Denmark. http://www.danmap.org/Downloads/Reports.aspx.
85 Tada T, Miyoshi-Akiyama T, Kato Y et al. Emergence of 16S rRNA 105 EFSA/ECDC 2018. EU Summary Report on Antimicrobial Resistance in
methylase-producing Acinetobacter baumannii and Pseudomonas aerugi- Zoonotic and Indicator Bacteria from Humans, Animals and Food in 2016.
nosa isolates in hospitals in Vietnam. BMC Infect Dis 2013; 13: 251. https://efsa.onlinelibrary.wiley.com/doi/epdf/10.2903/j.efsa.2018.5182.
86 Feßler AT, Kadlec K, Schwarz S. Novel apramycin resistance gene apmA 106 Johns I, Adams E. Trends in antimicrobial resistance in equine bacterial
in bovine and porcine methicillin-resistant Staphylococcus aureus ST398 iso- isolates: 1999–2012. Vet Rec 2015; 176: 334.
lates. Antimicrob Agents Chemother 2011; 55: 373–5. 107 Anses 2017. Résapath - Réseau d’épidémiosurveillance de l’antibiorésist-
87 Wendlandt S, Fessler AT, Monecke S et al. The diversity of antimicrobial ance des bactéries pathogènes animales. https://resapath.anses.fr/SITE_
resistance genes among staphylococci of animal origin. Int J Med Microbiol RESAPATH_WEB/uploadfiles/files/Documents/2017_RESAPATH%20Rapport
2013; 303: 338–49. %20Annuel%20[id_doc"310].pdf.
88 Wendlandt S, Kadlec K, Feßler AT et al. Transmission of methicillin- 108 Cavicchio L, Dotto G, Giacomelli M et al. Class 1 and class 2 integrons in
resistant Staphylococcus aureus isolates on broiler farms. Vet Microbiol 2013; avian pathogenic Escherichia coli from poultry in Italy. Poult Sci 2015; 94:
167: 632–7. 1202–8.

15 of 17
Review

109 Gurung M, Tamang MD, Moon DC et al. Molecular basis of resistance to https://www.ema.europa.eu/documents/other/answers-requests-scientific-
selected antimicrobial agents in the emerging zoonotic pathogen advice-impact-public-health-animal-health-use-antibiotics-animals_en.pdf.
Streptococcus suis. J Clin Microbiol 2015; 53: 2332–6. 126 .Werkgroep Veterinair Antibioticabeleid 2019. Formularia. https://www.

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


110 de Neeling AJ, van den Broek MJ, Spalburg EC et al. High prevalence of wvab.nl/formularia/.
methicillin resistant Staphylococcus aureus in pigs. Vet Microbiol 2007; 122: 127 Sedláková MH, Urbánek K, Vojtová V et al. Antibiotic consumption and
366–72. its influence on the resistance in Enterobacteriaceae. BMC Res Notes 2014; 7:
111 Wagenaar J, Van de Giessen A. Veegerelateerde MRSA: epidemiologie in 454.
dierlijke productieketens, transmissie naar de mens en karakterisatie van de 128 Buelow E, Gonzalez TB, Versluis D et al. Effects of selective digestive de-
kloon. 2010. https://www.rivm.nl/publicaties/veegerelateerde-mrsa-epide contamination (SDD) on the gut resistome. J Antimicrob Chemother 2014; 69:
miologie-in-dierlijke-productieketens-transmissie-naar-mens. 2215–23.
112 Vandendriessche S, Vanderhaeghen W, Larsen J et al. High genetic di- 129 Paul M, Lador A, Grozinsky-Glasberg S et al. Beta lactam antibiotic
versity of methicillin-susceptible Staphylococcus aureus (MSSA) from humans monotherapy versus beta lactam-aminoglycoside antibiotic combination
and animals on livestock farms and presence of SCCmec remnant DNA in therapy for sepsis. Cochrane Database Syst Rev 2014; issue 1: 1–113.
MSSA CC398. J Antimicrob Chemother 2014; 69: 355–62. 130 Falcone M, Russo A, Venditti M. Optimizing antibiotic therapy of bacter-
113 Alba P, Feltrin F, Cordaro G et al. Livestock-associated methicillin resist- emia and endocarditis due to staphylococci and enterococci: new insights
ant and methicillin susceptible Staphylococcus aureus Sequence Type (CC) 1 and evidence from the literature. J Infect Chemother 2015; 21: 330–9.
in European farmed animals: high genetic relatedness of isolates from Italian 131 Fernández-Hidalgo N, Almirante B, Gavaldà J et al. Ampicillin plus cef-
cattle herds and humans. PLoS One 2015; 10: e0137143. triaxone is as effective as ampicillin plus gentamicin for treating Enterococcus
114 Perreten V, Kadlec K, Schwarz S et al. Clonal spread of methicillin- faecalis infective endocarditis. Clin Infect Dis 2013; 56: 1261–8.
resistant Staphylococcus pseudintermedius in Europe and North America: an 132 Carugati M, Bayer A, Miró J et al. High-dose daptomycin therapy for left-
international multicentre study. J Antimicrob Chemother 2010; 65: 1145–54. sided infective endocarditis: a prospective study from the international col-
115 ECDC 2018. Data from the ECDC Surveillance Atlas—Antimicrobial laboration on endocarditis. Antimicrob Agents Chemother 2013; 57: 6213–22.
Resistance. https://ecdc.europa.eu/en/antimicrobial-resistance/surveillance- 133 Garcı́a P, Hopkins KL, Garcı́a V et al. Diversity of plasmids encoding viru-
and-disease-data/data-ecdc. lence and resistance functions in Salmonella enterica subsp. enterica serovar
116 Burow E, Simoneit C, Tenhagen B-A et al. Oral antimicrobials increase Typhimurium monophasic variant 4,[5], 12:i:- strains circulating in Europe.
antimicrobial resistance in porcine E. coli—a systematic review. Prev Vet Med PLoS One 2014; 9: e89635.
2014; 113: 364–75. 134 Hall RM. Salmonella genomic islands and antibiotic resistance in
117 Sun J, Li L, Liu B et al. Development of aminoglycoside and b-lactamase Salmonella enterica. Future Microbiol 2010; 5: 1525–38.
resistance among intestinal microbiota of swine treated with lincomycin, 135 Dorado-Garcı́a A, Smid JH, van Pelt W et al. Molecular relatedness of
chlortetracycline, and amoxicillin. Front Microbiol 2014; 5: 580. ESBL/AmpC-producing Escherichia coli from humans, animals, food and the
118 Brewer MT, Xiong N, Anderson KL et al. Effects of subtherapeutic con- environment: a pooled analysis. J Antimicrob Chemother 2018; 73: 339–47.
centrations of antimicrobials on gene acquisition events in Yersinia, Proteus, 136 Ruppé É, Woerther P-L, Barbier F. Mechanisms of antimicrobial resist-
Shigella, and Salmonella recipient organisms in isolated ligated intestinal ance in Gram-negative bacilli. Ann Intensive Care 2015; 5: 1–15.
loops of swine. Am J Vet Res 2013; 74: 1078–83. 137 Huijbers P, Graat E, Haenen A et al. Extended-spectrum and AmpC b-
119 Jensen VF, Jakobsen L, Emborg H-D et al. Correlation between apramy- lactamase-producing Escherichia coli in broilers and people living and/or
cin and gentamicin use in pigs and an increasing reservoir of gentamicin- working on broiler farms: prevalence, risk factors and molecular characteris-
resistant Escherichia coli. J Antimicrob Chemother 2006; 58: 101–7. tics. J Antimicrob Chemother 2014; 69: 2669–75.
120 Herrero-Fresno A, Zachariasen C, Hansen MH et al. Apramycin treat- 138 Huijbers PM, van Hoek AH, Graat EA et al. Methicillin-resistant
ment affects selection and spread of a multidrug-resistant Escherichia coli Staphylococcus aureus and extended-spectrum and AmpC b-lactamase-pro-
strain able to colonize the human gut in the intestinal microbiota of pigs. Vet ducing Escherichia coli in broilers and in people living and/or working on or-
Res 2016; 47: 1–10. ganic broiler farms. Vet Microbiol 2015; 176: 120–5.
121 Da Costa PM, Belo A, Gonçalves J et al. Field trial evaluating changes in 139 Salauze D, Otal I, Gomez-Lus R et al. Aminoglycoside acetyltransferase
prevalence and patterns of antimicrobial resistance among Escherichia coli 3-IV (aacC4) and hygromycin B 4-I phosphotransferase (hphB) in bacteria iso-
and Enterococcus spp. isolated from growing broilers medicated with enro- lated from human and animal sources. Antimicrob Agents Chemother 1990;
floxacin, apramycin and amoxicillin. Vet Microbiol 2009; 139: 284–92. 34: 1915–20.
122 Schwaiger K, Bauer J, Hölzel CS. Selection and persistence of 140 Chaslus-Dancla E, Pohl P, Meurisse M et al. High genetic homology be-
antimicrobial-resistant Escherichia coli including extended-spectrum b-lacta- tween plasmids of human and animal origins conferring resistance to the
mase producers in different poultry flocks on one chicken farm. Microb Drug aminoglycosides gentamicin and apramycin. Antimicrob Agents Chemother
Resist 2013; 19: 498–506. 1991; 35: 590.
123 Chalmers G, Cormier AC, Nadeau M et al. Determinants of virulence and 141 Chaslus-Dancla E, Martel J, Carlier C et al. Emergence of aminoglycoside
of resistance to ceftiofur, gentamicin, and spectinomycin in clinical 3-N-acetyltransferase IV in Escherichia coli and Salmonella typhimurium iso-
Escherichia coli from broiler chickens in Québec, Canada. Vet Microbiol 2017; lated from animals in France. Antimicrob Agents Chemother 1986; 29:
203: 149–57. 239–43.
124 Gullberg E, Albrecht LM, Karlsson C et al. Selection of a multidrug resist- 142 Wray C, Hedges R, Shannon K et al. Apramycin and gentamicin resist-
ance plasmid by sublethal levels of antibiotics and heavy metals. MBio 2014; ance in Escherichia coli and salmonellas isolated from farm animals. J Hyg
5: e01918–14. (Lond) 1986; 97: 445–56.
125 EMA/AMEG 2014. Answers to the Requests for Scientific Advice on the 143 Hunter J, Bennett M, Hart C et al. Apramycin-resistant Escherichia coli
Impact on Public Health and Animal Health of the Use of Antibiotics in isolated from pigs and a stockman. Epidemiol Infect 1994; 112: 473–80.
Animals—Answer to the Second Request from the EC (Ranking of Antibiotics); 144 EURL for Bovine Tuberculosis 2018. Bovine Tuberculosis Eradication
Answer to the Third Request from the EC (New Antibiotics); Answer to the in Europe. https://www.visavet.es/bovinetuberculosis/bovine-tb/eradication.
Fourth Request from the EC (Risk Mitigation Options) (EMA/381884/2014). php.

16 of 17
Review JAC
145 Garcia-Graells C, Antoine J, Larsen J et al. Livestock veterinarians at high 147 Hammerum A. Enterococci of animal origin and their significance for
risk of acquiring methicillin-resistant Staphylococcus aureus ST398. Epidemiol public health. Clin Microbiol Infect 2012; 18: 619–25.
Infect 2012; 140: 383–9. 148 EFSA/ECDC 2017. EMA and EFSA Joint Scientific Opinion on Measures

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkz161/5475277 by University of South Australia user on 20 April 2019


146 de Regt MJ, van Schaik W, van Luit-Asbroek M et al. Hospital and com- to Reduce the Need to Use Antimicrobial Agents in Animal Husbandry in
munity ampicillin-resistant Enterococcus faecium are evolutionarily closely the European Union, and the Resulting Impacts on Food Safety
linked but have diversified through niche adaptation. PLoS One 2012; 7: (RONAFA). https://efsa.onlinelibrary.wiley.com/doi/epdf/10.2903/j.efsa.2017.
e30319. 4666.

17 of 17

You might also like