Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

High prevalence of antimicrobial resistance and multidrug resistance among bacterial isolates from diseased pets: Retrospective laboratory data (2015–2017)

Abstract

Laboratory surveillance and the monitoring of antimicrobial resistance (AMR) trends and patterns among local isolates have been highly effective in providing comprehensive information for public health decision-making. A total of 396 cases along with 449 specimens were received for antibiotic susceptibility testing at a public university veterinary diagnostic laboratory in Malaysia between 2015 and 2017. Escherichia coli was the most frequently isolated (n = 101, 13%) bacteria, followed by Staphylococcus pseudintermedius (n = 97, 12%) and Streptococcus canis (n = 62, 8%). In cats, S. pseudintermedius isolates were highly resistant to azithromycin (90%), while the E. coli isolates were highly resistant to doxycycline (90%), tetracycline (81%), and cephalexin (75%). About 55% of S. pseudintermedius and 82% of E. coli were multi-drug resistant (MDR). In dogs, S. intermedius isolates were highly resistant to aminoglycosides neomycin (90.9%) and gentamicin (84.6%), and tetracycline (75%). Whereas the E. coli isolates were highly resistant to cephalexin (82.1%) and amoxicillin/clavulanic acid (76.5%). MDR was observed in 60% of S. intermedius and 72% of E. coli from dogs. Generally, the bacterial isolates from cats demonstrated higher levels of resistance to multiple antibiotics compared to those from dogs.

Introduction

Antimicrobial resistance (AMR) among bacterial isolates from pets has not received as much attention as AMR among bacteria in livestock. However, AMR among bacterial isolates such as MRSA and MDR E. coli and Klebsiella pneumoniae in pets is an emerging issue of veterinary and public health significance [1]. In addition to AMR complicating the treatment of sick pets, close relationships between pets and their human counterparts can result in the transmission of the resistant pathogens between the two [2, 3]. The transmission of antibiotic-resistant bacteria such as the zoonotic transmission of opportunistic pathogens like Staphylococcus spp. between dogs and their owners have been reported in many studies [4, 5]. In fact, several studies have reported that the AMR level is higher among animals living in close association with humans than the animals residing in remote areas [6]. Studies have found that pets may acquire resistant pathogens from their owners [79].

In the past decade, the use of antibiotics in animals reared for meat has been broadly scrutinized [10, 11]. Multiple reports have also established that the volume and frequency of antibiotics used in livestock production settings are reducing the effectiveness of the antibiotics when used for medicinal purposes in humans [12]. In fact, it is among the major drivers of the increasing level of AMR among bacterial pathogens [13, 14]. In contrast, there have been lesser attention paid to the use of antibiotics in pets and especially, the antibiotic resistance among bacteria from pets. Pets are normally exposed to antibiotics when they are treated for bacterial infections [15], or prevented from secondary bacterial infection during episodes of viral infections [16]. Antibiotic prescriptions often include critically important antibiotic groups used in human medicine including the heavy usage of broad-spectrum agents such as fluoroquinolones, cephalosporins, and penicillins [17]. These antibiotics are categorized as veterinary critically important antibiotics by the OIE [18] as well as critically important for human medicine by the WHO [19]. Veterinarians are often inclined to use broad-spectrum antibiotics as the first-line treatment for bacterial infection due to its convenience [20, 21] and as blanket coverage against a broad range of bacterial diseases. However, such usage increases selective pressure, leading to the development of multidrug resistance in bacteria such as Escherichia coli, Pseudomonas aeruginosa, and other commensal bacteria in pets [2224].

Sporadic reports have suggested an increasing level of resistance in important pathogens in cats and dogs [25] and this situation escalates the complexity of treating bacterial infections such as urinary tract infections and cystitis in these species [2628]. The findings from this study will highlight the significance of AMR among clinically important bacterial pathogens in small animal medicine. In line with Malaysia Strategic Action Plan for AMR (MyAP-AMR) [29] and Global Action Plan of AMR, this study aims to describe the antimicrobial resistance patterns of clinically important bacterial pathogens from diagnostic cases in pets. This study will also determine the differences in resistance profiles between isolates recovered from diseased cats and dogs between 2015 and 2017.

Material and methods

Source of data

The design of this study has been described in Haulisah et al., [30]. The data originated from the veterinary diagnostic cases that were received from various veterinary health premises and animal facilities in Peninsular Malaysia by the accredited Bacteriology Laboratory, Faculty of Veterinary Medicine of Universiti Putra Malaysia (UPM) Serdang, Selangor. Diagnostic reports (paper format) of cases from 1 January 2015 to 31 December 2017 were compiled and transferred into WHONET (vers 5.6, Boston, MA). Relevant data compiled included information concerning animal species, clinical history, sex, type of specimens (urine, wound swab, lavage fluid, etc.), clinical signs, isolated bacteria, and findings from antibiotic susceptibility testing (AST).

The laboratory performed culture and identification of received samples using standard microbiological procedures [31]. The antimicrobial sensitivity was performed using the Kirby Bauer’s disc diffusion on Mueller Hinton Agar (MHA) method following the Clinical Laboratory Standards Institute (CLSI) guidelines [32]. The AST data was grouped into three categories: Resistant (R), Intermediate (I), and Susceptible (S) using the established clinical breakpoints. The antimicrobial agent tested varied depending on the request of the clinician, type of the organism, and the availability of the antimicrobial agents.

Data received from the 396 cases by the laboratory were entered into Excel 2007 (Microsoft Office, 2007) and then transferred into WHONET 5.6 [33]. Data related to cats and dogs were analyzed separately. A total of 780 isolates were recovered from the various clinical samples of dogs and cats over a period of three years (2015–2017).

Data analysis

Descriptive data analysis was performed in WHONET 5.6 as described in WHO [34, 35]. Trends and patterns of tested antibiotics and the multidrug resistance (MDR) levels among the clinical isolates were analyzed based on the species of the pets followed by interspecies comparison. The difference in the level of resistance based on the antimicrobial agents for the 3-year study period was tested using the Chi-square test at 95% confidence intervals using SPSS 22 (IBM, Armonk, NY: IBM Corp.) at α = 0.05. Fisher’s exact test was applied when the expected numbers per cell were small. For the purpose of this analysis, the isolates categorized as ‘intermediate’ and ‘resistant’ were grouped as ‘non-susceptible’. In addition, an isolate was marked as ‘multi-drug resistant’ if it were resistant to at least one agent in three or more classes of antibiotics [36].

Results

Descriptive analysis of samples

Between 2015 and 2017, 396 cases with accompanying 449 specimens were received by the laboratory for antibiotic susceptibility testing (AST) (cats n = 271, 68.4% and dogs n = 125, 31.6%). A total of 780 isolates were obtained out of which more than half were recovered from cats (n = 467, 59.9%) and the rest from dogs (n = 313, 40.1%).

A total of 65 bacterial species were identified from the specimens. The most common species were Escherichia coli (n = 101, 13%), followed by Staphylococcus pseudintermedius (n = 97, 12%), and Streptococcus canis (n = 62, 8%). The majority of Enterococcus spp. were Enterococcus faecalis (n = 48, 72.7%) and the remaining 18 were non-faecalis Enterococcus spp. (Fig 1).

thumbnail
Fig 1. The distribution of bacterial species (Gram-negative and gram-positive bacteria) from diseased pets isolated between January 2015 and December 2017.

Numbers inside brackets ‘()’ indicate the total number of bacterial isolates.

https://doi.org/10.1371/journal.pone.0277664.g001

Antimicrobial resistance of clinical isolates from cats

A total of 289 clinical samples were received (Table 1) from which 467 isolates were recovered. Staphylococcus pseudintermedius was the most common bacteria from wounds/abscesses (61.2%) and ear infections (13.4%). Overall, S. pseudintermedius (n = 67, 14.3%), E. coli (n = 65, 13.9%), S. canis (n = 44, 9.4%), and K. pneumoniae (n = 37, 7.9%) were the most frequently isolated. Klebsiella pneumoniae was found to be more commonly isolated from urine (51.4%) while Escherichia coli, Staphylococcus pseudintermedius, and Streptococcus canis were most commonly isolated from wounds/abscess (Table 1). More than 75.8% (354/467) of the bacteria isolated were MDR out of which 20.6% (96/467) were resistant to one or two antibiotic classes and only 3.6% (17/467) were susceptible to all tested antibiotics.

thumbnail
Table 1. Species of bacteria isolated from diagnostic samples from diseased cats received by the Bacteriology Laboratory between 2015 and 2017.

https://doi.org/10.1371/journal.pone.0277664.t001

Antibiogram based on species of bacteria isolated from cats

Antibiogram of the bacteria isolated is presented in Figs 25. The MDR levels for the selected bacteria for 2015–2017 are shown in Fig 6.

thumbnail
Fig 2. Antibiotic susceptibility pattern from Staphylococcus pseudintermedius isolates from diseased cats (January 2015–December 2017).

R–resistance; I-intermediate; S–susceptible; AMC–amoxicillin/clavulanic acid; AMX–amoxicillin; AZM–azithromycin; LEX–cephalexin; GEN–gentamicin; ENX–enrofloxacin; MBX–marbofloxacin; TET–tetracycline; SXT–trimethoprim-sulfamethoxazole. Numbers inside brackets ‘()’ indicate the total number of tested isolates for each antibiotic.

https://doi.org/10.1371/journal.pone.0277664.g002

thumbnail
Fig 3. Antibiotic susceptibility pattern from Escherichia coli isolates from diseased cats (January 2015–December 2017).

R–resistance; I-intermediate; S–susceptible; AMC–amoxicillin/clavulanic acid; LEX–cephalexin; GEN–gentamicin; CRO–ceftriaxone; DOX–doxycycline; ENX–enrofloxacin; MBX–marbofloxacin; TET–tetracycline; SXT–trimethoprim-sulfamethoxazole. Numbers inside brackets ‘()’ indicate the total number of tested isolates for each antibiotic.

https://doi.org/10.1371/journal.pone.0277664.g003

thumbnail
Fig 4. Antibiotic susceptibility pattern from Streptococcus canis isolates from diseased cats (January 2015–December 2017).

R–resistance; I-intermediate; S–susceptible; AMC–amoxicillin/clavulanic acid; LEX–cephalexin; GEN–gentamicin; ENX–enrofloxacin; MBX–marbofloxacin; TET–tetracycline. Numbers inside brackets ‘()’ indicate the total number of tested isolates for each antibiotic.

https://doi.org/10.1371/journal.pone.0277664.g004

thumbnail
Fig 5. Antibiotic susceptibility pattern from Klebsiella pneumoniae isolates from diseased cats (January 2015–December 2017).

R–resistance; I-intermediate; S–susceptible; AMC–amoxicillin/clavulanic acid; LEX–cephalexin; GEN–gentamicin; CRO–ceftriaxone; ENX–enrofloxacin; MBX–marbofloxacin; TET–tetracycline; SXT–trimethoprim-sulfamethoxazole. Numbers inside brackets ‘()’ indicate the total number of tested isolates for each antibiotic.

https://doi.org/10.1371/journal.pone.0277664.g005

thumbnail
Fig 6. Multidrug resistance of clinically important bacterial pathogens from diseased cats between 2015 and 2017.

Numbers inside brackets ‘()’ indicate the number of isolates detected; those on bars indicate percentage per organism; Non-MDR–Resistant to 1 or 2 classes; MDR–multi-drug-resistant, resistant to 3 or more classes.

https://doi.org/10.1371/journal.pone.0277664.g006

Staphylococcus pseudintermedius.

Fig 2 shows that the highest percentage of resistance was against azithromycin (90%; 95% CI = 54.1–99.5), followed by trimethoprim/sulfamethoxazole (57.1%; 95% CI = 29.6–81.2), and amoxicillin (53.3%; 95% CI = 27.4–77.7). There was no significant change in resistance trends for all tested antibiotics. More than 40.3% (27/67) of the S. pseudintermedius were resistant to multiple classes of antibiotics (MDR) (Fig 6).

Escherichia coli.

The proportion of E. coli isolates resistant to doxycycline was 90% (95% CI = 54.1–99.5), 81% to tetracycline (95% CI = 57.5–93.7), and 75% to cephalexin (95% CI = 60.1–85.9) (Fig 3). There was no significant change in resistance level over the three-year study period. MDR (Fig 6) was observed among 70.8% (46/65) of E. coli isolates.

Streptococcus canis.

Fig 4 shows that the resistance levels were high for gentamicin (66.7%; 95% CI = 35.5–88.7) and enrofloxacin (58.8%; 95% CI = 40.8–74.9). A significant increase in isolates grouped as ‘intermediate’ was observed for amoxicillin/clavulanic acid (p = 0.000, from 13.3% in 2015 to 22.2% in 2017). However, S. canis remained highly susceptible (90.7%) to amoxicillin/clavulanic acid over the three years. The MDR of S. canis (Fig 6) was relatively low at 9.1% (4/44).

Klebsiella pneumoniae.

It was found that K. pneumoniae isolates were highly resistant to fluoroquinolones marbofloxacin (87.5%; 95% CI = 70.1–95.9), enrofloxacin (82.1; 95% CI = 62.4–93.2), and amoxicillin/clavulanic acid (81.1%; 95% CI = 64.3–91.5) (Fig 5). No significant change was observed in the resistance trends for all the tested antimicrobial agents over the three years. MDR was found in 86.5% of K. pneumoniae isolates from cats (86.5%, 32/37). None of the isolates was susceptible to all the classes of tested antimicrobial agents (Fig 6).

Antimicrobial resistance patterns of clinical isolates from dogs

Between 2015 and 2017, 160 clinical samples were analyzed from a total of 125 cases (Table 2). From these samples, 313 isolates were recovered. Staphylococcus intermedius (n = 37, 11.8%), Escherichia coli (n = 36, 11.5%), Proteus mirabilis (n = 31, 9.9%), and Staphylococcus pseudintermedius (n = 30, 9.58%) were the most frequently isolated bacterial species (Table 2). MDR was identified among 71.6% (224 /313) of the isolated bacteria.

thumbnail
Table 2. Species of bacteria isolated from diagnostic samples from diseased dogs received by the Bacteriology Laboratory between 2015 and 2017.

https://doi.org/10.1371/journal.pone.0277664.t002

Antibiogram based on the species of bacteria isolated from dogs

Antibiogram of the bacteria isolated from dogs is presented in Figs 710. The MDR levels for the selected bacterial species (2015–2017) are shown in Fig 11.

thumbnail
Fig 7. Antibiotic susceptibility pattern from Staphylococcus intermedius isolates from diseased dogs (January 2015-December 2017).

R–resistance; I-intermediate; S–susceptible; AMC–amoxicillin/clavulanic acid; LEX–cephalexin; GEN–gentamicin; ENX–enrofloxacin; MBX–marbofloxacin; NEO-neomycin; TET–tetracycline. Numbers inside brackets ‘()’ indicate the total number of tested isolates for each antibiotic.

https://doi.org/10.1371/journal.pone.0277664.g007

thumbnail
Fig 8. Antibiotic susceptibility pattern from Escherichia coli isolates from diseased dogs (January 2015-December 2017).

R–resistance; I-intermediate; S–susceptible; AMC–amoxicillin/clavulanic acid; LEX–cephalexin; ENX–enrofloxacin; MBX–marbofloxacin; TET–tetracycline; SXT–trimethoprim-sulfamethoxazole. Numbers inside brackets ‘()’ indicate the total number of tested isolates for each antibiotic.

https://doi.org/10.1371/journal.pone.0277664.g008

thumbnail
Fig 9. Antibiotic susceptibility pattern from Proteus mirabilis isolates from diseased dogs (January 2015-December 2017).

R–resistance; I-intermediate; S–susceptible; AMC–amoxicillin/clavulanic acid; LEX–cephalexin; ENX–enrofloxacin; MBX–marbofloxacin; TET–tetracycline; SXT–trimethoprim-sulfamethoxazole. Numbers inside brackets ‘()’ indicate the total number of tested isolates for each antibiotic.

https://doi.org/10.1371/journal.pone.0277664.g009

thumbnail
Fig 10. Antibiotic susceptibility pattern from Staphylococcus pseudintermedius isolates from diseased dogs (January 2015-December 2017).

R–resistance; I-intermediate; S–susceptible; AMC–amoxicillin/clavulanic acid; LEX–cephalexin; ENX–enrofloxacin; MBX–marbofloxacin; TET–tetracycline. Numbers inside brackets ‘()’ indicate the total number of tested isolates for each antibiotic.

https://doi.org/10.1371/journal.pone.0277664.g010

thumbnail
Fig 11. Multidrug resistance of clinically important bacterial pathogens from diseased dogs between 2015 and 2017.

Numbers inside brackets ‘()’ indicate number of isolates detected; those on bars indicate percentages per organism; Non-MDR–Resistant to 1 or 2 classes; MDR–multi-drug resistant, resistant to 3 or more classes.

https://doi.org/10.1371/journal.pone.0277664.g011

Staphylococcus intermedius.

The AST results for S. intermedius are presented in Fig 7. The highest resistance level was against neomycin (90.9%; 95% CI = 57.1–99.5), gentamicin (84.6%; 95% CI = 53.6–97.3), and tetracycline (75%; 95% CI = 47.4–91.7). It was also noted that about 60% (22/37) of the S. intermedius isolates were MDR (Fig 11). The resistance trend for the bacteria did not change significantly over time (2015–2017).

Escherichia coli.

Fig 8 depicts the AMR levels of E. coli (2015–2017). The resistance was the highest for cephalexin (82.1%; 95% CI = 62.4–93.2) and amoxicillin/clavulanic acid (76.5%; 95% CI = 58.5–88.6). The resistance trend for E. coli did not change considerably over the study period. It was also noted that the proportion of MDR (Fig 11) among E. coli was 72.2% (26/36).

Proteus mirabilis.

The frequency of AMR of P. mirabilis to all the tested antimicrobial agents (2015–2017) is presented in Fig 9. P. mirabilis isolates were completely resistant to tetracycline (100%; 95% CI = 59.8–100), followed by cephalexin (79.2%; 95% CI = 57.3–92.1), and trimethoprim/sulfamethoxazole (75%; 95% CI = 35.6–95.5). A significant decrease in resistance level was observed for marbofloxacin (p = 0.013, from 87.5% in 2015 to 36.4% in 2017). The MDR of P. mirabilis (Fig 11) was 51.6% (16/31).

Staphylococcus pseudintermedius.

The resistance level was highest for tetracycline at 54.5% (95% CI = 24.5–81.8) (Fig 10). A significantly decreased resistance level to cephalexin (from 53.9% in 2015 to 0% in 2017) and to tetracycline (from 100% in 2015 to 0% in 2017) was observed over the three-year study period. Forty percent (12/30) of the S. pseudintermedius isolates were MDR (Fig 11).

Differences between the profile of resistances of isolates from cats and dogs

Staphylococcus pseudintermedius and Escherichia coli were the most commonly encountered bacterial species from both cats and dogs. In addition, most antibiotic classes used to treat E. coli and S. pseudintermedius infections are shared between these two species. Comparison of the AMR profiles among S. pseudintermedius and E. coli isolates are presented in Tables 3 and 4 respectively.

thumbnail
Table 3. Difference between E. coli isolates from diseased cats and dogs between 2015 and 2017.

https://doi.org/10.1371/journal.pone.0277664.t003

thumbnail
Table 4. Differences between S. pseudintermedius isolates from diseased cats and dogs between 2015 and 2017.

https://doi.org/10.1371/journal.pone.0277664.t004

Escherichia coli.

E. coli isolates from dogs showed high levels of non-susceptibility towards all the tested antimicrobial agents with higher levels to trimethoprim/sulfamethoxazole, amoxicillin/clavulanic acid, and cephalexin. In contrast, E. coli isolates from cats showed high proportions of non-susceptibility towards tetracycline (95.2%). Significant differences were observed between the E. coli isolates from cats and dogs to tetracycline (95.2% versus 64.7%) (Table 3). It was observed that the frequency of MDR E. coli isolates from cats and dogs were similar (70.8% versus 72.2%; χ2 = 0.024, p = 0.877).

Staphylococcus pseudintermedius.

The resistance patterns of S. pseudintermedius isolates to commonly used antimicrobial agents were similar between dogs and cats (Table 4). The frequency of MDR also shared similarities between S. pseudintermedius isolates from cats and dogs (40.3% versus 40%; χ2 = 0.001, p = 1.000).

Discussion

AMR is an ongoing global public health challenge that negatively impacts animals, humans as well as environmental health. Data from veterinary diagnostic laboratories provide vital information about trends and patterns of AMR among disease-causing agents circulating in the local animal population. In this study, the analyses of the data supplies important snapshot information on the AMR situation to shape treatment regime and to guide policy on the use of antibiotics in pets [3739], consequently prolonging the usefulness of commonly prescribed antibiotics.

Our previous study reported a very high AMR prevalence among major disease-causing bacteria in livestock [30]. In this study, we report that S. pseudintermedius, E. coli, S. intermedius, P. mirabilis, K. pneumoniae, and S. canis, which are the most common pathogens isolated from diseased cats and dogs (2015–2017), have comparatively high levels of resistance to commonly used antibiotics for therapy. The resistance pattern against antibiotics for these pathogens underscores increasing challenges in treating common infections in cats and dogs as these bacterial species have also been identified as most clinically relevant in pets elsewhere [40]. Unfortunately, there are very few local or regional AMR information on pathogens causing diseases in pets to enable a more geographically contextual discussion. Therefore comparisons were made based on other available literatures.

Antimicrobial resistance of clinical isolates from cats

Overall, the trend did not change significantly over the study period for all the antibiotics tested. The majority of the S. canis isolates remained sensitive to amoxicillin/clavulanic acid. However, the increasing level of S. canis isolates grouped as ‘intermediate’ for amoxicillin/clavulanic acid from 13.3% in 2015 to 22.2% in 2017 is a note of concern. The level of susceptibility to this antibiotic is markedly higher than observed in a study by Awosile et al., [41], which reported a very low level (0.6%) of resistance from samples received over a ten-year study period at the Diagnostic Services Bacteriology Laboratory, Canada [41]. Additionally, in the study conducted by Ludwig et al., [42] across Europe, AMR was reported at 4% to amoxicillin/clavulanic acid over the entire study period.

Fluoroquinolones enrofloxacin and marbofloxacin are specific formulations for veterinary use [43, 44]. Marbofloxacin, a broad-spectrum antibiotic, is a drug of choice for the exclusive treatment of chronic bacterial infections affecting the urinary and respiratory tracts as well as skin and ear infections [45]. We observed high levels of resistance to enrofloxacin and marbofloxacin for K. pneumoniae (>80%) and E. coli (>40%) isolates recovered from cats. These findings corroborate with other similar studies conducted around the world. For instance, Marques et al., [46] reported a 72% resistance level of K. pneumoniae isolates recovered from urine samples of diseased cats and dogs in Lisbon. Other researchers report a lower resistance level (39.3%) of E. coli isolates against enrofloxacin in Poland [22]. In contrast, lower resistance levels (range: 0–5%) were observed for Enterobacteriaceae against marbofloxacin from stray cats between 2017 and 2018 in South Korea [47]. The observed high resistance level against fluoroquinolones antibiotics is not surprising as they are frequently prescribed in veterinary production and clinical practice [48]. Our finding should provide evidence for precautionary actions from the veterinary authority and other sectors to better strategize its usage to ensure the antibiotic’s continuing efficacy. Moreover, the overuse of fluoroquinolones in the veterinary application has been linked to cross-resistance with other fluoroquinolones such as ciprofloxacin and norfloxacin used for human therapy [19]. These are listed as critically important antimicrobials for humans and are often used for treating human bacterial infections [19, 49].

High resistance levels for S. pseudintermedius isolates from cats were recorded against azithromycin (90%). Azithromycin is a broad-spectrum macrolide used for bacterial, rickettsial, and parasitic infections [50, 51] and has been used in both veterinary and human medicine [19]. The resistance to this antibiotic is not commonly reported in pet animals. However, multiple reports of high-level resistance of azithromycin in animals are available amongst pathogens of public health significance in livestock [52] and human-specific pathogens such as Salmonella typhi [53] and Neiserria [54]. The high level of resistance points to the over-prescription of azithromycin and therefore must be re-examined as the antibiotic is preferred for treatment against rickettsial and hemotropica parasites [5557].

In Finland [37] and Australia [58], the resistance levels for gentamicin and trimethoprim/sulfamethoxazole (30.8%-44.8%) for S. pseudintermedius isolates from cats were slightly lower than the findings of this study for the two antibiotics (>50%). Antibiotic-resistant S. pseudintermedius is an emerging veterinary clinical pathogen with some reports highlighting its increasing level of multi-drug resistance [59]. Although the zoonotic transmission of S. pseudintermedius remains rare [60], a few reports have linked infection from cats to their owners [61, 62].

Antimicrobial resistance of clinical isolates from dogs

The resistance trends for cephalexin and tetracycline in S. pseudintermedius and marbofloxacin in P. mirabilis isolates from dogs significantly declined between 2015 and 2017. P. mirabilis isolates were highly resistant to tetracycline (100%) and cephalexin (79.2%) which is concordant with the findings documented in Beijing [63] and Granada [64] where the isolates from dogs were 100% and 61.7% resistant against tetracycline, respectively. Auwaerter [65] reported the high resistance to tetracycline and cephalosporins by P. mirabilis isolates from dogs to be a common finding. Our findings of 79.2% resistance against cephalexin are higher than that reported in Canada (48.9%) by Awosile et al., [41]. The frequency of prescription of this antibiotic in small animal clinics [66] has been linked to the increasing level of resistance worldwide. Our study shows that more than half of S. intermedius and S. pseudintermedius isolates were resistant against tetracycline and is thus consistent with the findings from Kalhoro et al., [67] in sick dogs in China. A lower resistance level (less than 30%) of tetracycline by S. pseudintermedius isolates was reported from healthy dogs in Canada [68] and Australia [69].

E. coli isolates showing higher resistance levels against cephalexin (82.1%) and amoxicillin/clavulanic acid (76.5%) recorded in this study are similar to those reported among ESBL-E. coli isolates against amoxicillin/clavulanic acid (86.8%) from dogs and cats at the Veterinary Diagnostic Laboratory, United States [70]. Similar findings were also documented from dogs in Canada [41], Europe [71], the United States [72], and New Zealand [73]. Furthermore, in the present study, the E. coli isolates from dogs show high resistance levels against enrofloxacin, marbofloxacin, tetracycline, and trimethoprim/sulfamethoxazole, thus signifying an impending difficulty of treating E. coli infection in local dogs.

Comparison of antimicrobial resistance of clinical isolates from dogs and cats

E. coli and S. pseudintermedius are regarded as an excellent sentinel for AMR in a wide range of companion animals and as part of the their normal microbiota [1, 74]. These opportunistic bacteria can cause a wide range of infections. Hence, they are suitable candidates for comparing resistance profiles between cats and dogs. Our study shows interspecies variation in resistance patterns to tetracycline for E. coli isolates. E. coli isolates originating from cats had significantly higher resistance levels than those from dogs, which is consistent with the report from Australia [75]. However, more research is needed to explain this finding.

When we compare our findings on E. coli isolates from pets for the purpose of this study to that from local diseased livestock [30], we found that the level of resistance to tetracycline is very similar to that from diseased ruminants (cattle and goat) but significantly lower compared to that from diseased non-ruminants (chicken and pig). Our finding supports the conjecture relating to the key role of the environment and frequency of antibiotic usage in the trends of AMR. The finding for tetracycline resistance of E. coli is similar to that of diseased ruminants in Malaysia [30] that are raised extensively or semi-intensively with limited usage of antibiotics. Non-ruminants have a significantly higher level of resistance to tetracycline [30] as this antibiotic is one of the most commonly used as feed additives in intensive farming systems [76, 77]. In contrast, tetracycline is used only for treatment for conditions such as eye infection and bacterial gastrointestinal infection in cats and dogs [78].

Multi-drug resistance of clinical isolates from dogs and cats

Several bacteria are shared between companion animals and humans and this creates opportunities for interspecies transmission of AMR-bacteria [1]. In the current study, isolates from diseased dogs and cats exhibited high levels of MDR with the majority of the isolates being resistant to 3–4 antimicrobial agents. For example, we reported a high frequency of MDR among E. coli isolates from diseased dogs (72.2%) and cats (70.8%). We found a similar level of MDR among E. coli isolates from diseased livestock (both ruminant and non-ruminants) in our previous study [30]. However, with the exception of a study in Poland [22], which reported a 66.8% MDR among E. coli isolates from sick pets, many reports documented lower MDR levels. In the northern part of Peninsular Malaysia by Shahaza et al., [79] reported MDR E. coli isolates in only 0.58% of E. coli isolated from pets. Similarly, in Portugal [39] and Canada [41], a relatively lower MDR level (range: 13%–32%) was found among clinical E. coli isolates from sick cats and dogs presented to the Veterinary Diagnostic Laboratory. The level of MDR of other pathogens such as S. pseudintermedius from dogs and cats (>40%) is similar to those reported from a convenient sample of dogs (47.5% MDR) in the USA [80] but lower than that reported in another local study on S. pseudintermedius (n = 23) isolates from stray and pet dogs and cats (100% MDR) [81]. In contrast, Australia [59], France [25], and the United Kingdom [82] reported MDR levels of between 4.7% and 20.7%. The variations in the MDR levels could reflect the choice or preferences of some antimicrobials used for the treatment of bacterial diseases in pets [83] and also on the frequency of antimicrobial usage in livestock production settings as well as in human medicine within a locality or geographic region [84].

Limitations of the study

The present study shares similar inherent biases as stated by Hayer, et al., [85] about the study limitations using veterinary diagnostic laboratory data. The data used for this study are laboratory reports generated from diagnostics cases; therefore, the findings may not represent the AMR situation in the general pet population in peninsular Malaysia and may not reflect the overall picture of AMR levels among bacteria in healthy pets. Additionally, the samples received by the lab were from diseased pets which may have been treated with antibiotics prior to sample collection, thereby altering the antibiogram profiles. Unfortunately, the histories regarding their previous antimicrobial therapy were mostly not available. In addition, the bacteria isolated could be a part of the normal flora or contaminants rather than the disease-causing agent. Finally, the panels of antibiotics used in the analyses were not consistent because they were dependent on several factors such as the request by the clinician, availability of antibiotics in the laboratory, and the type of organism isolated.

Conclusion

This study presents a snapshot of the AMR among important bacterial pathogens such as Staphylococci (S. pseudintermedius and S. intermedius), E. coli, K. pneumoniae, P. mirabilis, and S. canis recovered from diseased pets. A similar AMR pattern was observed for S. pseudintermedius and E. coli isolates recovered from sick cats and dogs, except for their resistance to tetracycline—where E. coli isolates from cats show higher AMR levels than those from dogs. The majority of the aforementioned bacterial isolates also demonstrated high levels of MDR, thereby undermining or complicating treatment options for bacterial infections in pets. Despite the limitation in inferencing the findings of this study to larger populations, the high MDR levels observed alludes to the growing challenges concerning the treatment of pets with common bacterial infections. The findings from this study provide important baseline measurements for the ongoing AMR surveillance and provide a foundation to better frame the antimicrobial stewardship efforts in pets.

Acknowledgments

We thank the technical staff of the Bacteriology Laboratory, Faculty of Veterinary Medicine, UPM Serdang, Malaysia for their time and for sharing knowledge from the inception to the end of this research project.

References

  1. 1. Pomba C, Rantala M, Greko C, Baptiste KE, Catry B, Van Duijkeren E, et al. Public health risk of antimicrobial resistance transfer from companion animals. J. Antimicrob. Chemother. 2017 April; 72(4): 957–968. pmid:27999066
  2. 2. Chomel BB. Emerging and re-emerging zoonoses of dogs and cats. Animals. 2014 July; 4(3): 434–445. pmid:26480316
  3. 3. Ghasemzadeh I, Namazi SH. Review of bacterial and viral zoonotic infections transmitted by dogs. J. Med. Life. 2015; 8, 1–5. pmid:28316698
  4. 4. Walther B, Hermes J, Cuny C, Wieler LH, Vincze S, Elnaga YA, et al. Sharing more than friendship—nasal colonization with Coagulase-positive Staphylococci (CPS) and co-habitation aspects of dogs and their owners. PLoS One. 2012 April; 7(4): e35197. pmid:22529990
  5. 5. Robb AR, Wright ED, Foster AME, Walker R, Malone C. Skin infection caused by a novel strain of Staphylococcus pseudintermedius in a Siberian husky dog owner. JMM Case Rep. 2017 March; 4(3): jmmcr005087. pmid:28663821
  6. 6. Köck R, Daniels-Haardt I, Becker K, Mellmann A, Friedrich AW, Mevius D, et al. Carbapenem-resistant Enterobacteriaceae in wildlife, food-producing, and companion animals: a systematic review. Clin. Microbiol. Infec. 2018 December; 24(12): 1241–1250. pmid:29654871
  7. 7. Lloyd DH. Reservoirs of antimicrobial resistance in pet animals. Clin. Infect. Dis. 2007 September; 45 Suppl 2: S148–152. pmid:17683019
  8. 8. Rutland BE, Weese JS, Bolin C, Au J, Malani AN. Human-to-dog transmission of methicillin-resistant Staphylococcus aureus. Emerg. Infect. Dis. 2009 August; 15(8): 1328–1330. pmid:19751611
  9. 9. Van Duijkeren E, Wolfhagen MJHM, Box ATA, Heck MEOC, Wannet WJB, Fluit AC. Human-to-dog transmission of methicillin-resistant Staphylococcus aureus. Emerg. Infect. Dis. 2004 December; 10(12): 2235–2237. pmid:15663871
  10. 10. Van Boeckel TP, Brower C, Gilbert M, Grenfell BT, Levin SA, Robinson TP, et al. Global trends in antimicrobial use in food animals. P. Natl. Acad. Sci. USA. 2015 March; 112(18): 5649–5654. pmid:25792457
  11. 11. Marshall BM, Levy SB. Food animals and antimicrobials: Impacts on human health. Clin. Microbiol. Rev. 2011 October; 24(4): 718–733. pmid:21976606
  12. 12. Marquardt RR, Li S. Antimicrobial resistance in livestock: advances and alternatives to antibiotics. Anim. Front. 2018 April; 8(2): 30–37. pmid:32002216
  13. 13. Suriyasathaporn W, Chupia V, Sing-Lah T, Wongsawan K, Mektrirat R, Chaisri W. Increases of antibiotic resistance in excessive use of antibiotics in smallholder dairy farms in northern Thailand. Asian-Australas. J. Anim. Sci. 2012 September; 25(9): 1322–1328. pmid:25049697
  14. 14. Economou V, Gousia P. Agriculture and food animals as a source of antimicrobial-resistant bacteria. Infect. Drug Resist. 2015 April; 8: 49–61. pmid:25878509
  15. 15. Guardabassi L, Schwarz S, Lloyd DH. Pet animals as reservoirs of antimicrobial-resistant bacteria. J. Antimicrob. Chemoth. 2004 August; 54(2): 321–322. pmid:15254022
  16. 16. Lappin MR, Blondeau J, Boothe D, Breitschwerdt EB, Guardabassi L, Lloyd DH, et al. Antimicrobial use guidelines for treatment of respiratory tract disease in dogs and cats: antimicrobial guidelines working group of the international society for companion animal infectious diseases. J. Vet. Intern. Med. 2017 February; 31(2): 279–294. pmid:28185306
  17. 17. Moulin G, Cavalié P, Pellanne I, Chevance A, Laval A, Millemann Y, et al. A comparison of antimicrobial usage in human and veterinary medicine in France from 1999 to 2005. J. Antimicrob. Chemoth. 2008 September; 62(3): 617–625. pmid:18490374
  18. 18. World Organisation for Animal Health. OIE List of antimicrobials of veterinary importance. 2007 [cited 2022 January 5]. https://www.oie.int/fileadmin/Home/eng/Our_scientific_expertise/docs/pdf/AMR/A_OIE_List_antimicrobials_May2018.pdf.
  19. 19. World Health Organisation. WHO list of critically important antimicrobials for human medicine: ranking of antimicrobial agents for risk management of antimicrobial resistance due to non-human use. 2017 [cited 2022 January 5]. https://apps.who.int/iris/bitstream/handle/10665/255027/9789241512220-eng.pdf.
  20. 20. Guardabassi L, Houser GA, Frank LA, Papich MG. Guidelines for antimicrobial use in dogs and cats, in: Guardabassi L., Jensen L.B., Kruse H., (Ed), Guide to antimicrobial use in animals. Blackwell Publishing, Oxford, UK, 2009; pp. 183–206. https://doi.org/10.1002/9781444302639.ch11
  21. 21. Beco L, Guaguère E, Lorente Méndez C, Noli C, Nuttall T, Vroom M. Suggested guidelines for using systemic antimicrobials in bacterial skin infections (2): Antimicrobial choice, treatment regimens and compliance. Vet. Rec. 2013 February; 172(6): 156–160. pmid:23292948
  22. 22. Rzewuska M, Czopowicz MB, U MK, Chrobak D, Borys BB, Binek M. Multidrug resistance in Escherichia coli strains isolated from infections in dogs and cats in Poland (2007–2013). Sci. World J. 2015 January; 2015:1–8. pmid:25667937
  23. 23. Ferreira TSP, Moreno LZ, Felizardo MR, de Gobbi DDS, de LN Filsner PH, de Moura Gomes VT, et al. Pheno- and genotypic characterization of Pasteurella multocida isolated from cats, dogs and rabbits from Brazil. Comp. Immunol. Microbiol. Infect. Dis. 2016 April; 45: 48–52. pmid:27012921
  24. 24. Moyaert H, de Jong A, Simjee S, Rose M, Youala M, El Garch F, et al. Erratum: Survey of antimicrobial susceptibility of bacterial pathogens isolated from dogs and cats with respiratory tract infections in Europe: ComPath results. J. Appl. Microbiol. 2019 November; 127(5): 1594. pmid:31606951
  25. 25. Bourély C, Cazeau G, Jarrige N, Leblond A, Madec JY, Haenni M, et al. Antimicrobial resistance patterns of bacteria isolated from dogs with otitis. Epidemiol. Infect. 2019 March;147: e121. pmid:30868979
  26. 26. Jessen LR, Sørensen TM, Bjornvad CR, Nielsen SS, Guardabassi L. Effect of antibiotic treatment in canine and feline urinary tract infections: A systematic review. Vet. J. 2015 March: 203(3): 270–277. pmid:25634080
  27. 27. Dorsch R, Von Vopelius-Feldt C, Hartmann K, Wolf G, Straubinger RK. Feline urinary tract pathogens: Prevalence of bacterial species and antimicrobial resistance over a 10-year period. Vet. Rec. 2015 February;176(8): 201–201. pmid:25351232
  28. 28. Teichmann-Knorrn S, Reese S, Wolf G, Hartmann K, Dorsch R. Prevalence of feline urinary tract pathogens and antimicrobial resistance over five years. Vet. Rec. 2018 July;183(1): 21–21. pmid:29622684
  29. 29. Ministry of Health Malaysia. Malaysian action plan on antimicrobial resistance (MyAP-AMR) 2017–2021. Ministry of Health Malaysia. 2017 [cited 2022 January 5]. https://rr-asia.oie.int/wp-content/uploads/2020/03/malaysia_malaysia-national_action_plan_-
  30. 30. Haulisah NA, Hassan L, Bejo SK, Jajere SM. High levels of antibiotic resistance in isolates from diseased livestock. Front. Vet. Sci. 2021 April; 8, 300. pmid:33869326
  31. 31. Jang SS, Biberstein EL, Hirsh DC. A diagnostic: manual of veterinary clinical bacteriology and mycology. Iowa, University of California Davis County; 2008.
  32. 32. Clinical and Laboratory Standard Institute. Performance standards for antimicrobial disk and dilution susceptibility tests for bacteria isolated from animals: approved standard-third edition. M31A3. CLSI: Wayne, PA. Clinical Laboratory Standards Institute. 2015.
  33. 33. Agarwal A, Kapila K, Kumar S. WHONET software for the surveillance of antimicrobial susceptibility. Med. J. Armed Forces India. 2009 July; 65(3): 264–266. pmid:27408261
  34. 34. World Health Organization. WHONET tutorial data analysis 1 for the surveillance of antimicrobial resistance. 2006a [cited 2022 January 5]. https://whonet.org/Docs/WHONET%204.Data%20analysis%201.doc.
  35. 35. World Health Organization. WHONET tutorial data analysis 2 for the surveillance of antimicrobial resistance. 2006b [cited 2022 January 5]. https://whonet.org/Docs/WHONET%205.Data%20analysis%202.doc.
  36. 36. Magiorakos AP, Srinivasan A, Carey RB, Carmeli Y, Falagas ME, Giske CG, et al. Multidrug-resistant, extensively drug-resistant and pandrug-resistant bacteria: An international expert proposal for interim standard definitions for acquired resistance. Clin. Microbiol. Infec. 2012 March;18(3): 268–281. pmid:21793988
  37. 37. Grönthal T, Eklund M, Thomson K, Piiparinen H, Sironen T, Rantala M. Antimicrobial resistance in Staphylococcus pseudintermedius and the molecular epidemiology of methicillin-resistant S. pseudintermedius in small animals in Finland. J. Antimicrob. Chemoth. 2017 January;72(4): 1021–1030. pmid:28065889
  38. 38. Cummings KJ, Aprea VA, Altier C. Antimicrobial resistance trends among canine Escherichia coli isolates obtained from clinical samples in the northeastern USA, 2004–2011. Can. Vet. J. 2015 April; 56(4): 393–398. pmid:25829560.
  39. 39. Marques C, Belas A, Franco A, Aboim C, Gama LT, Pomba C. Increase in antimicrobial resistance and emergence of major international high-risk clonal lineages in dogs and cats with urinary tract infection: 16 year retrospective study. J. Antimicrob. Chemoth. 2018 February; 73(2): 377–384. pmid:29136156
  40. 40. Normand EH, Gibson NR, Reid SWJ, Carmichael S, Taylor DJ. Antimicrobial-resistance trends in bacterial isolates from companion-animal community practice in the UK. Prev. Vet. Med. 2000 September; 46(4): 267–278. pmid:10960713
  41. 41. Awosile BB, Mcclure JT, Saab ME, Heider LC. Antimicrobial resistance in bacteria isolated from cats and dogs from the Atlantic Provinces, Canada from 1994–2013. Can. Vet. J. 2018 August; 59(8): 885–893. pmid:30104781.
  42. 42. Ludwig C, de Jong A, Moyaert H, El Garch F, Janes R, Klein U, et al. Antimicrobial susceptibility monitoring of dermatological bacterial pathogens isolated from diseased dogs and cats across Europe (ComPath results). J. Appl. Microbiol. 2016 September; 121(5): 1254–1267. pmid:27593360
  43. 43. Ihrke PJ, Papich MG, Demanuelle TC. The use of fluoroquinolones in veterinary dermatology. Vet. Dermatol. 1999; 10(3): 193–204. pmid:34644924
  44. 44. Park Y, Oh J, Park S, Sum S, Song W, Chae J, et al. Antimicrobial resistance and novel mutations detected in the gyrA and parC genes of Pseudomonas aeruginosa strains isolated from companion dogs. BMC Vet. Res. 2020 April; 16(1): 1–8. pmid:32293442
  45. 45. Yohannes S, Awji EG, Lee SJ, Park SC. Pharmacokinetics and pharmacokinetic/pharmacodynamic integration of marbofloxacin after intravenous and intramuscular administration in beagle dogs. Xenobiotica. 2015 December; 45(3): 264–269. pmid:25470431
  46. 46. Marques C, Menezes J, Belas A, Aboim C, Cavaco-Silva P, Trigueiro G, et al. Klebsiella pneumoniae causing urinary tract infections in companion animals and humans: Population structure, antimicrobial resistance and virulence genes. J. Antimicrob. Chemoth. 2019 March; 74(3): 594–602. pmid:30535393
  47. 47. Jung WK, Shin S, Park YK, Lim SK, Moon DC, Park KT, et al. Distribution and antimicrobial resistance profiles of bacterial species in stray cats, hospital-admitted cats, and veterinary staff in South Korea. BMC Vet. Res. 2020 April;16(1): 1–14. pmid:32272916
  48. 48. Pedersen K, Pedersen K, Jensen H, Finster K, Jensen VF, Heuer OE. Occurrence of antimicrobial resistance in bacteria from diagnostic samples from dogs. J. Antimicrob. Chemoth. 2007 October; 60(4): 775–781. pmid:17644533
  49. 49. Heuer OE, Jensen VF, Hammerum AM. Antimicrobial drug consumption in companion animals. Emerg. Infect. Dis. 2005 February; 11(2): 344–345. pmid:15759339.
  50. 50. Bakheit AHH, Al-Hadiya BMH, Abd-Elgalil AA. Chapter One–Azithromycin. Profiles Drug Subst. Excip. Relat. Methodol. 2014 May; 39: 1–40. pmid:24794904
  51. 51. Mitchell MA. Azithromycin. J. Exot. Pet. Med. 2005 July; 14(3): 212–214.
  52. 52. Abdus Sobur M, Al Momen Sabuj A, Sarker R, Taufiqur Rahman AMM, Lutful Kabir SM, Tanvir Rahman M. Antibiotic-resistant Escherichia coli and Salmonella spp. Associated with dairy cattle and farm environment having public health significance. Vet. World. 2019 July; 12(7): 984–993. pmid:31528022.
  53. 53. Duy PT, Dongol S, Giri A, Nguyen To NT, Dan Thanh HN, Nhu Quynh NP, et al. The emergence of azithromycin-resistant Salmonella typhi in Nepal. JAC Antimicrob. Resist. 2020 December; 2(4): dlaa109. pmid:34223059
  54. 54. Eyre DW, Sanderson ND, Lord E, Regisford-Reimmer N, Chau K, Barker L, et al. Gonorrhoea treatment failure caused by a Neisseria gonorrhoeae strain with combined ceftriaxone and high-level azithromycin resistance, England, February 2018. Eurosurveillance. 2018; 23(27): 1800323. pmid:29991383
  55. 55. El Sayed I, Liu Q, Wee I, Hine P. Antibiotics for treating scrub typhus. Cochrane Db. Syst. Rev. 2018 September; 3(9): 1–28. pmid:30246875
  56. 56. Fang Y, Huang Z, Tu C, Zhang L, Ye D, Zhu BP. Meta-analysis of drug treatment for scrub typhus in Asia. Internal Med. 2012; 51(17), 2313–2320. pmid:22975540
  57. 57. Breitschwerdt EB. Feline bartonellosis and cat scratch disease. Vet. Immunol. Immunop. 2008 May; 123(1–2): 167–171. pmid:18295347
  58. 58. Saputra S, Jordan D, Worthing KA, Norris JM, Wong HS, Abraham R, et al. Antimicrobial resistance in Coagulase-positive Staphylococci isolated from companion animals in Australia: a one year study. PLoS One. 2017 April; 12(4): e0176379. pmid:28430811
  59. 59. Pomba C, Couto N, Moodley A. Treatment of a lower urinary tract infection in a cat caused by a multi-drug methicillin-resistant Staphylococcus pseudintermedius and Enterococcus faecalis. J. Feline Med. Surg. 2010 October; 12(10): 802–806.
  60. 60. Kuan EC, Yoon AJ, Vijayan T, Humphries RM, Suh JD. Canine Staphylococcus pseudintermedius sinonasal infection in human hosts. Int. Forum Allergy Rh. 2016 February; 6: 710–715. pmid:26880481
  61. 61. Van Duijkeren E, Kamphuis M, Van Der Mije IC, Laarhoven LM, Duim B, Wagenaar JA, et al. Transmission of methicillin-resistant Staphylococcus pseudintermedius between infected dogs and cats and contact pets, humans and the environment in households and veterinary clinics. Vet. Microbiol. 2011 June; 150(3–4): 338–343. pmid:21420256
  62. 62. Worthing KA, Brown J, Gerber L, Trott DJ, Abraham S, Norris JM. Methicillin-resistant Staphylococci amongst veterinary personnel, personnel-owned pets, patients and the hospital environment of two small animal veterinary hospitals. Vet. Microbiol. 2018 September; 223: 79–85. pmid:30173756
  63. 63. Han P, Luo L, Shahid M, Li M, Wang S, Nobrega DB, et al. Prevalence, genetic diversity and antimicrobial resistance of Proteus mirabilis isolated from dogs hospitalized in Beijing. Pak. Vet. 2020 September; 40(1), 61–66.
  64. 64. Irizarry R, Amadi V, Brathwaite-Sylvester E, Nicholas-Thomas R, Sharma R, Hariharan H. Update on urinary tract infections in dogs in a tropical island and antimicrobial susceptibility of Escherichia coli isolates for the period 2010–2016. Vet. Med. 2016 December; 1(3): 56–61.
  65. 65. Auwaerter P. Proteus species. In John Hopkins ABX guide. The John Hopkins university. 2019 [cited 2022 January 5]. https://www.hopkinsguides.com/hopkins/view/Johns_Hopkins_ABX_Guide/540454/all/Pro teus_species.
  66. 66. Wong C, Epstein SE, Westropp JL. Antimicrobial susceptibility patterns in urinary tract infections in dogs (2010–2013). J. Vet. Intern. Med. 2015 July; 29(4): 1045–1052. pmid:26133165
  67. 67. Kalhoro DH, Kalhoro MS, Mangi MH, Jahejo AR, Kumbhar S, Lochi GM, et al. Antimicrobial resistance of Staphylococci and Streptococci isolated from dogs. Trop. Biomed. 2019; 36(2): 468–474. pmid:33597408
  68. 68. Priyantha R, Gaunt MC, Rubin JE. Antimicrobial susceptibility of Staphylococcus pseudintermedius colonizing healthy dogs in Saskatoon, Canada. Can. Vet. J. 2016 January; 57(1): 65–69. pmid:26740701
  69. 69. Bean DC, Wigmore SM. Carriage rate and antibiotic susceptibility of Coagulase-positive Staphylococci isolated from healthy dogs in Victoria, Australia. Aust. Vet. J. 2016 November; 94(12): 456–460. pmid:27804112
  70. 70. Liu X, Thungrat K, Boothe DM. Occurrence of OXA-48 carbapenemase and other β- Lactamase genes in ESBL-producing multidrug resistant: Escherichia coli from dogs and cats in the United States, 2009–2013. Front. Microbiol. 2016; 7: 1057. pmid:27462301
  71. 71. Moyaert H, Morrissey I, De Jong A, El Garch F, Klein U, Ludwig C, et al. Antimicrobial susceptibility monitoring of bacterial pathogens isolated from urinary tract infections in dogs and cats across Europe: ComPath results. Microb. Drug Resist. 2017; 23(3): 391–403. pmid:28384093
  72. 72. KuKanich K, Lubbers B, Salgado B. Amoxicillin and amoxicillin-clavulanate resistance in urinary Escherichia coli antibiograms of cats and dogs from the Midwestern United States. J. Vet. Intern. Med. 2020; 34(1): 227–231. pmid:31777977
  73. 73. McMeekin CH, Hill KE, Gibson IR, Bridges JP, Benschop J. Antimicrobial resistance patterns of bacteria isolated from canine urinary samples submitted to a New Zealand veterinary diagnostic laboratory between 2005–2012. N. Z. Vet. J. 2017; 65(2): 99–104. pmid:27842208
  74. 74. Joosten P, Ceccarelli D, Odent E, Sarrazin S, Graveland H, Van Gompel L, et al. Antimicrobial usage and resistance in companion animals: A cross-sectional study in three european countries. Antibiotics. 2020 February; 9(2): 87. pmid:32079072
  75. 75. Bourne JA, Chong WL, Gordon DM. Genetic structure, antimicrobial resistance and frequency of human associated Escherichia coli sequence types among faecal isolates from healthy dogs and cats living in Canberra, Australia. PLoS One. 2019 March; 14(3): e0212867. pmid:30830915
  76. 76. Anadón A, Martínez-Larrañaga MR. Residues of antimicrobial drugs and feed additives in animal products: regulatory aspects. Livest. Prod. Sci. 1999 June; 59(2–3): 183–198.
  77. 77. Castanon JIR. History of the use of antibiotic as growth promoters in European poultry feeds. Poult. Sci. J. 2007 November; 86(11): 2466–2471. pmid:17954599
  78. 78. Sykes JE. Feline chlamydiosis. Clin. Tech. Small Anim. Pract. 2005 May; 20(2): 129–134. pmid:15948428
  79. 79. Shahaza O, Zakirah S, Muhammad A, Syamsyul A, Maswati MA. Antimicrobial resistance in veterinary clinical isolates of Escherichia coli from northern region of Peninsular Malaysia. Malays. J. Vet. Res. 2017 July; 8: 1–7.
  80. 80. Videla R, Solyman SM, Brahmbhatt A, Sadeghi L, Bemis DA, Kania SA. Clonal complexes and antimicrobial susceptibility profiles of Staphylococcus pseudintermedius isolates from dogs in the United States. Microb. Drug Resist. 2018 January; 24(1): 83–88. pmid:28504897
  81. 81. Mohamed MA, Abdul-Aziz S, Dhaliwal GK, Bejo SK, Goni MD, Bitrus AA, et al. Antibiotic resistance profiles of Staphylococcus pseudintermedius isolated from dogs and cats. Malays. J. Microbiol. 2017; 13: 180–186.
  82. 82. Beever L, Bond R, Graham PA, Jackson B, Lloyd DH, Loeffler A. Increasing antimicrobial resistance in clinical isolates of Staphylococcus intermedius group bacteria and emergence of MRSP in the UK. Vet. Rec. 2015 February; 176(7): 172–172. pmid:25376505
  83. 83. Ayukekbong JA, Ntemgwa M, Atabe AN. The threat of antimicrobial resistance in developing countries: causes and control strategies. Antimicrob. Resist. Infect. Control. 2017 May; 6(1): 1–8. pmid:28515903
  84. 84. Manyi-Loh C, Mamphweli S, Meyer E, Okoh A. Antibiotic use in agriculture and its consequential resistance in environmental sources: Potential public health implications. Molecules. 2018 March; 23(4): 795. pmid:29601469
  85. 85. Hayer SS, Rovira A, Olsen K, Johnson TJ, Vannucci F, Rendahl A, et al. Prevalence and trend analysis of antimicrobial resistance in clinical Escherichia coli isolates collected from diseased pigs in the USA between 2006 and 2016. Transbound. Emerg. Dis. 2020 February; 67(5): 1930–41. pmid:32097517