Next Article in Journal
Prevalence and Antibiotic Resistance Profile of Clostridium perfringens Isolated from Pork and Chicken Meat in Vietnam
Previous Article in Journal
A Comparison Study of Lymph Node Tuberculosis and Sarcoidosis Involvement to Facilitate Differential Diagnosis and to Establish a Predictive Score for Tuberculosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Comparative Minimum Inhibitory and Mutant Prevention Drug Concentrations for Pradofloxacin and Seven Other Antimicrobial Agents Tested against Bovine Isolates of Mannheimia haemolytica and Pasteurella multocida

by
Joseph M. Blondeau
1,2,* and
Shantelle D. Fitch
1
1
Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada
2
Department of Biochemistry, Microbiology and Immunology, Pathology and Laboratory Medicine and Ophthalmology, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
*
Author to whom correspondence should be addressed.
Pathogens 2024, 13(5), 399; https://doi.org/10.3390/pathogens13050399
Submission received: 16 April 2024 / Revised: 2 May 2024 / Accepted: 6 May 2024 / Published: 9 May 2024
(This article belongs to the Section Bacterial Pathogens)

Abstract

:
Pradofloxacin—a dual-targeting fluoroquinolone—is the most recent approved for use in food animals. Minimum inhibitory and mutant prevention concentration values were determined for pradofloxacin, ceftiofur, enrofloxacin, florfenicol, marbofloxacin, tildipirosin, tilmicosin, and tulathromycin. For M. haemolytica strains, MIC50/90/100 values were ≤0.016/≤0.016/≤0.016 and MPC50/90/100 values were 0.031/0.063/0.063; for P. multocida strains, the MIC50/90/100 values ≤0.016/≤0.016/0.031 and MPC50/90/100 ≤ 0.016/0.031/0.063 for pradofloxacin. The pradofloxacin Cmax/MIC90 and Cmax/MPC90 values for M. haemolytica and P. multocida strains, respectively, were 212.5 and 53.9 and 212.5 and 109.7. Similarly, AUC24/MIC90 and AUC24/MPC90 for M. haemolytica were 825 and 209.5, and for P. multocida, they were 825 and 425.8. Pradofloxacin would exceed the mutant selection window for >12–16 h. Pradofloxacin appears to have a low likelihood for resistance selection against key bovine respiratory disease bacterial pathogens based on low MIC and MPC values.

1. Introduction

Bovine respiratory disease (BRD), or shipping fever, is a complex multi-factorial syndrome in cattle precipitated by viral infection and/or a variety of known stressors (weather, transport, co-mingling, castration, dehorning, weaning, and auction), which, in turn, predispose to secondary bacterial infections prompting treatment with antimicrobial agents [1]. The principal bacterial pathogens include Mannheimia haemolytica and Pasteurella multocida. Morbidity rates have been reported to be 75%, with mortality of 24.5–44.8% in calves [2] and between 50 and 70% in feed lots [3,4]. Economic losses from BRD range from USD 800–900 million annually in the USA alone [5], and treatment costs also exceed USD 54 million annually [6]. Antimicrobial therapy remains an important intervention and may consist of metaphylaxis control, which is the treatment of groups of high-risk animals to minimize disease onset, and for BRD is generally with a macrolide antimicrobial agent [7]. Treatment refers to therapy for infection and may be with any one of a number of approved veterinary antibiotics, including beta-lactam, fluoroquinolone, macrolide, or phenicol agents [8,9]. In human medicine, it is well recognized that prompt antimicrobial therapy (4 h after clinical presentation) in patients with pneumonia impacts morbidity and mortality [10].
The susceptibility or resistance of bacteria to antimicrobial agents is determined by an in vitro measurement called the minimal inhibitory concentration (MIC) which uses a bacterial inoculum of 105 colony-forming units per milliliter (CFU/mL). An alternative measurement called mutant prevention concentration (MPC) defines the antimicrobial drug concentration blocking the growth of the least susceptible cells present in high-density (≥109 CFUs) bacterial populations. High-density bacterial populations have been documented in clinical infections in humans, including pneumonia [11], other respiratory tract infections [12], meningitis [13,14] and urinary tract infections [15,16]. In cattle, McVey and colleagues showed bacterial densities of ≥108 CFUs in experimentally induced respiratory infections in cattle [17]. High density bacterial populations are concerning as organisms with reduced susceptibility, requiring higher drug concentrations for inhibition of growth, may exist as subpopulations and not be detected by standardized susceptibility testing. Antimicrobial therapy with achievable drug concentrations that are insufficient to block the growth of the least susceptible cells allows for selective amplification of the resistant cells in the presence of the drug—regardless of the mechanism of reduced susceptibility [18]. Therapeutic drug concentrations preventing selective amplification of resistant bacterial cells reduce resistance selection from bacterial populations tested susceptible by MIC testing. Dagan and colleagues provided evidence that eradication of bacteria causing respiratory tract infections was necessary for clinical recovery and reducing resistance spread [19].
Pradofloxacin is a third-generation [20] enhanced-spectrum, dual-targeting fluoroquinolone approved for use in veterinary medicine [21] and has broad-spectrum activity against Gram-positive, Gram-negative [22], atypical bacteria [20,23,24] and also against anaerobic organisms [25]. It simultaneously targets DNA gyrase and topoisomerase IV in both Gram-negative and Gram-positive bacteria. These enzymes are critical for bacterial DNA regulation. Pradofloxacin has been argued to have a reduced likelihood for resistance selection based on low MPC values [21,26] and dual enzyme targeting. Previous investigations on companion animal pathogens [18,27,28] reported low MIC and MPC values for pradofloxacin, and we were interested in determining similar measurements for two primary BRD pathogens—M. haemolytica (MIC90 ≤ 0.016, MPC90 0.063) and P. multocida (MIC90 ≤ 0.016, MPC90 0.031) with pradofloxacin compared to ceftiofur, enrofloxacin, florfenicol, marbofloxacin, tilmicosin, tildipirosin, and tulathromycin. All strains tested had pradofloxacin MIC and MPC values below the susceptibility breakpoints established for MIC testing.

2. Materials and Methods

2.1. Bacterial Strains

Clinical isolates of M. haemolytica (n = 34) and P. multocida (n = 40) collected from field trials in the USA were used. Organism identification was by both matrix-assisted laser desorption ionization—time of flight (MALDI-TOF) (BioMerieux, St. Laurent, QC, Canada) and Vitek II (BioMerieux, St. Laurent, QC, Canada). Bacterial strains were cultured on tryptic soy agar containing 5% sheep red blood cells (BA) (Oxoid, Nepean, ON, Canada) for 18–24 h in O2 at 35–37 °C. For storage, single colonies were picked, transferred to skim milk, and stored frozen at −70 °C. Each isolate included in the study had to be susceptible to each agent based on recommended susceptibility MIC breakpoints [29].

2.2. Antimicrobial Compounds

Enrofloxacin and pradofloxacin were obtained in powder form from Bayer Animal Health (Elanco, Greenfield, IN, USA as of 2020) and prepared based on the manufacturers’ instructions. Ceftiofur (Zoetis, Kirkland, QC, Canada), florfenicol (Merck, Kirkland, QC, Canada), marbofloxacin (Vetoquinal, Lavaltrie, QC, Canada), tildipirosin (Merck, Kirkland, QC, Canada), tilmicosin, and tulathromycin (Zoetis, Kirkland, QC, Canada) were purchased in commercial form and prepared as per the manufacturer’s directions. Formulation excipients had no effect on MIC or MPC determination. Fresh stock solutions or samples stored at −70 °C were used for each experiment.

2.3. MIC Testing

MIC testing followed the procedure recommended by the Clinical and Laboratory Standards Institute [30]. Thawed isolates were sub-cultured (×2) on BA plates and incubated for 18–24 h in O2 at 35–37 °C. In 96-well microdilution trays, Mueller–Hinton Broth (MHB) (Difco Laboratories, Detroit, MI, USA) containing 2-fold drug concentration increments was added. A 0.5 McFarland standard was diluted to a final inoculum of 5 × 105 cfu/mL and added to the microtiter trays, incubated for 18–24 h at 35–37 °C in O2. The lowest drug concentration preventing visible bacterial growth was recorded as the MIC. Four American Type Culture Collection (ATCC) control strains—Enterococcus faecalis 29212, Escherichia coli 25922, Staphylococcus aureus 29213, and Pseudomonas aeruginosa 27853—were tested with each MIC assay to ensure the assays were within acceptable quality control performance ranges.

2.4. MPC Testing

Five BA plates per strain were inoculated for confluent growth, incubated for 18–24 h at 35–37 °C in O2, and the next day, the complete contents of the inoculated plates were transferred to 100 mL of MHB and incubated (18–24 h at 35–37C in O2) [31,32]. Cultures were subsequently estimated to have concentrations of ≥3 × 109 cfu/mL by spectrophotometric readings (600 nm) > 0.3 (Thermo Scientific Genesys 10s vis, Mississauga, ON, Canada) and by colony counts. Aliquots (100 µL) containing ≥109 cfu were applied to BA plates containing antimicrobial agent at drug concentrations ranging from one dilution below the measured MIC to seven dilutions above the MIC. BA plates containing antimicrobial agents were used within 1 week of preparation. Inoculated plates were incubated (as described) for a total of 48 h, with examination for growth at 24 and 48 h. The lowest drug concentration preventing all visible growth (48 h) was the MPC. Each experiment included the four aforementioned ATCC control strains.

3. Results

The MIC and MPC data for the M. haemolytica strains tested against eight antimicrobial agents are shown in Table 1. The drug concentrations (µg/mL) inhibiting 50% (MIC50), 90% (MIC90) and 100% (MIC100), respectively, of the strains tested were as follows: ceftiofur ≤0.016, ≤0.016, 0.031; enrofloxacin ≤0.016, ≤0.016, 0.031; florfenicol 1, 2, 2; marbofloxacin ≤0.016, ≤0.016, 0.016; pradofloxacin ≤0.016, ≤0.016, 0.016; tildipirosin 0.5, 1, 1; tilmicosin 0.5, 8, 8; tulathromycin 0.5, 1, 2. The MPC50, MPC90, and MPC100, respectively, were as follows: ceftiofur 0.25, 1, 1; enrofloxacin 0.125, 0.25, 0.5; florfenicol 2, 4, 4; marbofloxacin 0.063, 0.063, 0.125; pradofloxacin 0.031, 0.63, 0.063; tildipirosin 2, 4, 8; tilmicosin 16, ≥32, ≥32; tulathromycin 2, 4, 8.
Table 2 summarizes MIC and MPC data for the P. multocida strains tested. The MIC50, MIC90 and MIC100 values (µg/mL) were as follows, respectively: ceftiofur ≤0.016, ≤0.016, ≤0.016; enrofloxacin ≤0.016, ≤0.016, ≤0.016; florfenicol 0.5, 0.5, 0.5; marbofloxacin ≤0.016, 0.031, 0.031; pradofloxacin ≤0.016, ≤0.016, 0.031; tildipirosin 0.5, 2, 2; tilmicosin 2, 4, 8; tulathromycin 0.25, 0.5, 0.5. The MPC50, MPC90, and MPC100 values were as follows, respectively: ceftiofur ≤0.125, 0.25, 0.5; enrofloxacin 0.063, 0.125, 0.125; florfenicol 1, 1, 1; marbofloxacin 0.063, 0.125, 0.25; pradofloxacin ≤0.016, 0.031, 0.063; tildipirosin 4, 4, 8; tilmicosin 8, 16, 32; tulathromycin 2, 2, 4.
The modal MICs against M. haemolytica/P. multocida strains were ≤0.016 for ceftiofur, enrofloxacin, marbofloxacin, and pradofloxacin; 0.1/0.5 µg/mL for florfenicol; and were 0.5 tildipirosin, 0.5/1 for tilmicosin, and 0.5/2 for tulathromycin.
The modal MPC value against the M. haemolytica strain was lowest for pradofloxacin at 0.031 µg/mL, followed by ceftiofur at 0.125 µg/mL, marbofloxacin at 0.063 µg/mL, enrofloxacin at 0.125 µg/mL, tildipirosin and tulathromycin at 2 µg/mL, and tilmicosin at 4 µg/mL. For all agents—except tilmicosin, MPC values were at or below the susceptibility breakpoints.
For M. haemolytica and P. multocida, the Cmax/MIC90 values were 118.8 for enrofloxacin, 93.8 for marbofloxacin, and 212.5 for pradofloxacin. By comparison, Cmax/MPC90 values were 7.6, 23.8, and 53.9, respectively (Table 3).

4. Discussion

Fluoroquinolones are an important class of bactericidal antimicrobial agents for treating bacterial infections, including BRD, and pradofloxacin is the newest approved veterinary fluoroquinolones and has a number of favorable characteristics. Like other fluoroquinolones, it is active against Gram-positive and Gram-negative bacteria and against atypical bacteria and has favourable pharmacokinetic and pharmacodynamic properties. Pradofloxacin is also active against anaerobic bacteria [25].
Previously approved veterinary fluoroquinolones, including enrofloxacin, marbofloxacin, orbifloxacin, and difloxacin, preferentially target one of two enzymes (e.g., DNA gyrase or topoisomerase IV) critical for bacterial DNA replication. Typically, topoisomerase IV is the target in Gram-positive bacteria and DNA gyrase in Gram-negative bacteria. Pradofloxacin—as a dual-targeting fluoroquinolone—simultaneously inhibits both enzymes. Mutations in either of the parC or gyrA genes elevate MIC values and lessen the activity of single-target drugs. If the MIC is elevated above the susceptibility breakpoint, the organism is then considered non-susceptible or resistant. Pradofloxacin as a dual-targeting drug would require simultaneous mutations in both the parC and gyrA genes for resistance to occur.
The mutant prevention concentration (MPC) defines the antimicrobial drug concentration that blocks the growth of the less susceptible cells present in high-density bacterial populations [18]. MPC testing is similar to MIC testing, with some important differences: first, the inoculum (105 cfu/mL—MIC versus ≥109 CFUs MPC); second, MPC testing is currently conducted by agar dilution with antimicrobial compounds incorporated into the agar plates; and third, the MPC endpoint is 100% inhibition of bacterial growth. In this study, only organisms testing susceptible to each of the antimicrobials were included, as MPC testing is only performed on organisms testing susceptible to recommended breakpoints [18].
The MIC50/90 and MPC50/90 values for enrofloxacin, florfenicol, tilmicosin, and tulathromycin against the M. haemolytica isolates are similar to those previously published on a larger collection of strains [32]. Similarly, the MIC50/90 and MPC50/90 values for the same five drugs against the P. multocida strains in this study are similar to values previously reported for P. multocida strains from swine [38].
In this study, 100% of the strains tested had MPC values of ≤0.063 µg/mL—4-fold below the susceptibility breakpoint of ≤0.25 µg/mL for pradofloxacin. Fluoroquinolones are characterized as concentration-dependent agents, and their activity is based on the maximum serum-to-MIC ratio (Cmax/MIC) and the area of the drug concentration curve to MIC ratio (AUC/MIC). The exact values for the Cmax/MIC or AUC/MIC need to be debated; however, values of 10–12 and ≥125, respectively, have been used to show a more favorable clinical outcome and perhaps to minimize resistance selection. For pradofloxacin the Cmax/MIC ratio was 212.5 and the AUC/MIC was 825 for both organisms.
The free fraction of the drug represents the unbound percentage [39] that is accepted to contribute to the antimicrobial effect of pradofloxacin at approximately 40% protein bound. The Cmax/MIC ratio would be 127.5 and the AUC/MIC would be 495—both ratios well above the aforementioned ratio considered important for minimizing resistance and a favorable clinical outcome.
MIC testing is universally accepted as the principal antibacterial measurement for PK/PD modeling. Zhang and colleagues commented on MIC testing and potential limitations [40]. Indeed, they suggested that MIC test methods may contribute to treatment failure and the emergence of resistant mutants due to the following variables: (1) MIC testing is an all-or-none measurement with antibacterial activity only when the drug concentration is at or exceeds the MIC, but, in fact, a lower drug concentration may exert some level of antibacterial activity; (2) MIC testing is by doubling dilutions, which may elevate drug concentration with prolonged residual time; (3) MIC measurements are with static drug concentration; and (4) bacterial cell densities are 105 cfu/mL, which is well below the organism density that occurs during infection. Based on the above, the authors suggested that PK/PD modeling should also include MPC measurements, as reported here.
Ahmad and colleagues suggested MPC measurements need to be incorporated into PK/PD to assist with dosing guidelines [41]. In support of this argument, Xu et al. reported that Time>MPC was more important than T>MIC in the dosage design for preventing antimicrobial resistance for antimicrobial drug use (enrofloxacin) in aquaculture [42]. Cui and colleagues working with Staphylococcus aureus reported that AUC0–24/MPC above 25 h restricted the acquisition of resistance [43]. Olofsson et al. working with E. coli and ciprofloxacin in an in vitro kinetic model, reported that an AUC/MPC ratio of ≥22 was the single pharmacodynamic index that predicted the prevention of resistant mutant enrichment [44]. Vilalta et al. working with marbofloxacin, Haemophilus parasuis, and Actinobacillus pleuropneumoniae, indicated that marbofloxacin should prevent resistance selection for strains with MPC values up to 1 µg/mL [45]. Additionally, an AUC/MPC >25 h correlated with resistance prevention. Liang et al. investigated levofloxacin-resistant strains of S. aureus—same MIC but different MPC values—and compared differences between AUC24/MIC and AUC24/MPC for inhibiting the emergence of drug-resistant bacteria. Drug-resistant mutants were inhibited with AUC24/MPC values between 22 and 25. AUC24/MPC appeared more suitable than AUC24/MIC due to up to 8-fold differences for strains having the same MIC. Zhang and colleagues are investigating danofloxacin and A. pleuropneumoniae in a porcine tissue cage infection model showed the selection of drug-resistant bacteria could be significantly inhibited when the AUC24h/MPC was >18.58 h [46]. While the above investigations apply to concentration-dependent drugs, the application of time in the mutant selection window and T>MPC can be applied to time-dependent drugs. Alieva et al. investigated linezolid in an in vitro dynamic model [47]. They measured the drug residence time in the mutant selection window and the emergence of drug resistant bacteria. Time within the MSW was an important predictor for the selection and emergence of drug resistant bacteria. Xiong et al. showed drug-resistant bacteria occurred when T>MIC99 > 70% or T>MPC < 58% when studying cefquinome and S. aureus [48]. Zhang et al. investigating cefquinome and E. coli, showed selection and amplification of resistant bacteria when T>MIC90 > 25% or T>MPC was <50% [49]. The above-noted studies for both concentration and time-dependent agents showed MPC-based measurements integrated into PK/PD modeling for resistance prevention.
In considering MPC (versus MIC) values for pradofloxacin in Cmax and AUC calculations based on the data from our study, the Cmax/MPC90 value was 53.9 for M. haemolytica and 109.7 for P. multocida. The AUC24/MPC90 values were 209.5 for M. haemolytica and 425.8 for P. multocida. Drug curves considering MIC90 and MPC90 and the MSW showed pradofloxacin would exceed these values for >12 and <18 h of the drug concentration curve, respectively. For M. haemolytica, the peak pradofloxacin drug concentration was 50× the MIC90 and 12× the MPC90. For P. multocida, those values were 50× and 26×, respectively.
Fluoroquinolones are considered critically important antimicrobial agents in human health. The use of critically important compounds in food animals has been questioned; however, Kasimanickan et al. commented that there is no alternative to antimicrobials to treat non-vaccine preventable infectious diseases [50]. To this point, the authors argue that food animal producers need to play an important role in preventing overuse and misuse of antimicrobial agents. Scott and colleagues commented on the need for research on alternative therapies for infectious diseases in animals and for more judicious use of antibiotics in food animals. Label restrictions are intended to reduce inappropriate and non-approved use of drugs [51].
Pradofloxacin is the newest of the veterinary fluoroquinolones to be approved for use in food animals. The favorable in vitro activity against the principal BRD bacterial pathogens and favorable pharmacological characteristics—including dual targeting against critical enzymes for DNA replication—suggest an agent with a low likelihood for resistance selection against the pathogens tested.

Author Contributions

Conceptualization, J.M.B. and S.D.F.; methodology, J.M.B. and S.D.F.; software, J.M.B.; validation, J.M.B. and S.D.F.; formal analysis, J.M.B.; investigation, J.M.B. and S.D.F.; resources, J.M.B.; data curation, J.M.B.; writing—original draft preparation, J.M.B.; writing—review and editing, J.M.B.; visualization, J.M.B.; supervision, J.M.B.; project administration, J.M.B.; funding acquisition, J.M.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Elanco Animal Health (previously Bayer Animal Health), grant number #205235 10300.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Acknowledgments

We thank Deb Hills for excellent clerical assistance.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Gaudino, M.; Nagamine, B.; Ducatez, M.F.; Meyer, G. Understanding the mechanisms of viral and bacterial coinfections in bovine respiratory disease: A comprehensive literature review of experimental evidence. Vet. Res. 2022, 53, 70. [Google Scholar] [CrossRef]
  2. Callan, R.J.; Garry, F.B. Biosecurity and bovine respiratory disease. Vet. Clin. N. Am. Food Anim. Pract. 2002, 18, 57–77. [Google Scholar] [CrossRef] [PubMed]
  3. Blakebrough-Hall, C.; McMeniman, J.P.; González, L.A. An evaluation of the economic effects of bovine respiratory disease on animal performance, carcass traits, and economic outcomes in feedlot cattle defined using four BRD diagnosis methods. J. Anim. Sci. 2020, 98, skaa005. [Google Scholar] [CrossRef]
  4. Loneragan, G.H.; Dargatz, D.A.; Morley, P.S.; Smith, M.A. Trends in mortality ratios among cattle in US feedlots. J. Am. Vet. Med. Assoc. 2001, 219, 1122–1127. [Google Scholar] [CrossRef]
  5. Preview: Economic Effects of Bovine Respiratory Disease. J. Anim. Sci. 2020, 98, skaa042. [CrossRef] [PubMed]
  6. Johnson, K.K.; Pendell, D.L. Market Impacts of Reducing the Prevalence of Bovine Respiratory Disease in United States Beef Cattle Feedlots. Front. Vet. Sci. 2017, 4, 189. [Google Scholar] [CrossRef]
  7. Abell, K.M.; Theurer, M.E.; Larson, R.L.; White, B.J.; Apley, M. A mixed treatment comparison meta-analysis of metaphylaxis treatments for bovine respiratory disease in beef cattle. J. Anim. Sci. 2017, 95, 626–635. [Google Scholar] [CrossRef]
  8. Booker, C.W.; Lubbers, B.V. Bovine Respiratory Disease Treatment Failure: Impact and Potential Causes. Vet. Clin. N. Am. Food Anim. Pract. 2020, 36, 487–496. [Google Scholar] [CrossRef] [PubMed]
  9. Ball, J.J.; Kegley, E.B.; Sarchet, J.; Powell, J.G. Comparison of treatment protocols for bovine respiratory disease in high-risk, newly received beef calves. Appl. Anim. Sci. 2019, 35, 278–283. [Google Scholar] [CrossRef]
  10. Fally, M.; Israelsen, S.; Benfield, T.; Tarp, B.; Ravn, P. Time to antibiotic administration and patient outcomes in community-acquired pneumonia: Results from a prospective cohort study. Clin. Microbiol. Infect. 2021, 27, 406–412. [Google Scholar] [CrossRef]
  11. Frisch, A.W.; Tripp, J.T.; Barrett, C.D., Jr.; Pidgeon, B.E. The specific polysaccharide content of pneumonic lungs. J. Exp. Med. 1942, 76, 505–510. [Google Scholar] [CrossRef] [PubMed]
  12. Fagon, J.; Chastre, J.; Trouillet, J.L.; Domart, Y.; Dombret, M.C.; Bornet, M.; Gibert, C. Characterization of distal bronchial microflora during acute exacerbation of chronic bronchitis. Use of the protected specimen brush technique in 54 mechanically ventilated patients. Am. Rev. Respir. Dis. 1990, 142, 1004–1008. [Google Scholar] [CrossRef] [PubMed]
  13. Bingen, E.; Lambert-Zechovsky, N.; Leclercq, R.; Doit, C.; Mariani-Kurkdjian, P. Bactericidal activity of vancomycin, daptomycin, ampicillin and aminoglycosides against vancomycin-resistant Enterococcus facecium. J. Antimicrob. Chemother. 1990, 26, 619–626. [Google Scholar] [CrossRef] [PubMed]
  14. Feldman, W. Concentrations of bacteria in cerebrospinal fluid of patients with bacterial meningitis. J. Pediatr. 1976, 88, 549–552. [Google Scholar] [CrossRef] [PubMed]
  15. Matsukawa, M.; Kunishima, Y.; Takahashi, S.; Takeyama, K.; Tsukamoto, T. Time courses of bacterial density in urine during antibacterial chemotherapy and influential factors in patients having positive bacteriuria with a complicated urinary tract. J. Infect. Chemother. 2007, 13, 99–104. [Google Scholar] [CrossRef] [PubMed]
  16. Scott, V.C.; Haake, D.A.; Churchill, B.M.; Justice, S.S.; Kim, J.H. Intracellular Bacterial Communities: A Potential Etiology for Chronic Lower Urinary Tract Symptoms. Urology 2015, 86, 425–431. [Google Scholar] [CrossRef] [PubMed]
  17. McVey, D.S.; Kuszak, J. Bacterial isolates from the lungs of beef calves with bronchopneumonia associated with acute bovine respiratory disease. In Proceedings of the Conference on Research Workers in Animal Diseases (CRWAD), Chicago, IL, USA, 4–5 December 2010. [Google Scholar]
  18. Blondeau, J.M. New concepts in antimicrobial susceptibility testing: The mutant prevention concentration and mutant selection window approach. Vet. Dermatol. 2009, 20, 383–396. [Google Scholar] [CrossRef] [PubMed]
  19. Dagan, R.; Klugman, K.P.; Craig, W.A.; Baquero, F. Evidence to support the rationale that bacterial eradication in respiratory tract infection is an important aim of antimicrobial therapy. J. Antimicrob. Chemother. 2001, 47, 129–140. [Google Scholar] [CrossRef] [PubMed]
  20. Lees, P. Pharmacokinetics, pharmacodynamics and therapeutics of pradofloxacin in the dog and cat. J. Vet. Pharmacol. Therap. 2013, 36, 209–221. [Google Scholar] [CrossRef]
  21. Sykes, J.E.; Blondeau, J.M. Pradofloxacin: A novel veterinary fluoroquinolone for treatment of bacterial infections in cats. Vet. J. 2014, 201, 207–214. [Google Scholar] [CrossRef]
  22. Silley, P.; Stephan, B.; Greife, H.A.; Pridmore, A. Bactericidal properties of pradofloxacin against veterinary pathogens. Vet. Microbiol. 2012, 157, 106–111. [Google Scholar] [CrossRef] [PubMed]
  23. Dowers, K.L.; Tasker, S.; Radecki, S.V.; Lappin, M.R. Use of pradofloxacin to treat experimentally induced Mycoplasma hemofelis infection in cats. Am. J. Vet. Res. 2009, 70, 105–111. [Google Scholar] [CrossRef] [PubMed]
  24. Hartmann, A.D.; Helps, C.R.; Lappin, M.R.; Werckenthin, C.; Hartmann, K. Efficacy of Pradofloxacin in Cats with Feline Upper Respiratory Tract Disease due to Chlamydophila felis or Mycoplasma Infections. J. Vet. Intern. Med. 2008, 22, 44–52. [Google Scholar] [CrossRef] [PubMed]
  25. Silley, P.; Stephan, B.; Greife, H.A.; Pridmore, A. Comparative activity of pradofloxacin against anaerobic bacteria isolated from dogs and cats. J. Antimicrob. Chemother. 2007, 60, 999–1003. [Google Scholar] [CrossRef] [PubMed]
  26. Wetzstein, H.G. Comparative mutant prevention concentrations of pradofloxacin and other veterinary fluoroquinolones indicate differing potentials in preventing selection of resistance. Antimicrob. Agents Chemother. 2005, 49, 4166–4173. [Google Scholar] [CrossRef] [PubMed]
  27. Blondeau, J.M.; Shebelski, S.D. Comparative in vitro killing of canine strains of Staphylococcus pseudintermedius and Escherichia coli by cefovecin, cefazolin, doxycycline and pradofloxacin. Vet. Dermatol. 2016, 27, 267-e63. [Google Scholar] [CrossRef]
  28. Blondeau, J.M.; Fitch, S.D. In Vitro Killing of Canine Urinary Tract Infection Pathogens by Ampicillin, Cephalexin, Marbofloxacin, Pradofloxacin, and Trimethoprim/Sulfamethoxazole. Microorganisms 2021, 9, 2279. [Google Scholar] [CrossRef]
  29. Clinical and Laboratory Standards Institute. Performance Standards for Antimicrobial Disk and Dilution Susceptibility Tests for Bacteria Isolated from Animals: Approved Standard (M31-A6); M31-A6; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2015. [Google Scholar]
  30. Clinical and Laboratory Standards Institute. Performance Standards for Antimicrobial Disk and Dilution Susceptibililty Tests for Bacteria Isolated from Animals; VET01; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2018. [Google Scholar]
  31. Blondeau, J.M.; Zhao, X.; Hansen, G.T.; Drlica, K. Mutant prevention concentrations (MPC) of fluoroquinolones for clinical isolates of Streptococcus pneumoniae. Antimicrob. Agents Chemother. 2001, 45, 433–438. [Google Scholar] [CrossRef]
  32. Blondeau, J.M.; Borsos, S.; Blondeau, L.D.; Blondeau, B.J.; Hesje, C. Comparative minimum inhibitory and mutant prevention drug concentrations of enrofloxacin, ceftiofur, florfenicol, tilmicosin and tulathromycin against bovine isolates of Mannheimia haemolytica. Vet. Microbiol. 2012, 160, 85–90. [Google Scholar] [CrossRef]
  33. Belew, S.; Kim, J.Y.; Hossain, M.A.; Park, J.Y.; Lee, S.J.; Park, Y.S.; Suh, J.W.; Kim, J.C.; Park, S.C. Pharmacokinetics of marbofloxacin after intravenous and intramuscular administration in Hanwoo, Korean native cattle. J. Vet. Med. Sci. 2015, 77, 327–329. [Google Scholar] [CrossRef]
  34. Menge, M.; Rose, M.; Bohland, C.; Zschiesche, E.; Kilp, S.; Metz, W.; Allan, M.; Röpke, R.; Nürnberger, M. Pharmacokinetics of tildipirosin in bovine plasma, lung tissue, and bronchial fluid (from live, nonanesthetized cattle). J. Vet. Pharmacol. Ther. 2012, 35, 550–559. [Google Scholar] [CrossRef] [PubMed]
  35. Modric, S.; Webb, A.I.; Davidson, M. Effect of respiratory tract disease on pharmacokinetics of tilmicosin in rats. Lab. Anim. Sci. 1999, 49, 248–253. [Google Scholar] [PubMed]
  36. Bretzlaff, K.N.; Neff-Davis, C.A.; Ott, R.S.; Koritz, G.D.; Gustafsson, B.K.; Davis, L.E. Florfenicol in non-lactating dairy cows: Pharmacokinetics, binding to plasma proteins, and effects on phagocytosis by blood neutrophils. J. Vet. Pharmacol. Ther. 1987, 10, 233–240. [Google Scholar] [CrossRef] [PubMed]
  37. Ziv, G.; Shem-Tov, M.; Glickman, A.; Winkler, M.; Saran, A. Tilmicosin antibacterial activity and pharmacokinetics in cows. J. Vet. Pharmacol. Ther. 1995, 18, 340–345. [Google Scholar] [CrossRef] [PubMed]
  38. Blondeau, J.M.; Fitch, S.D. Mutant prevention and minimum inhibitory concentration drug values for enrofloxacin, ceftiofur, florfenicol, tilmicosin and tulathromycin tested against swine pathogens Actinobacillus pleuropneumoniae, Pasteurella multocida and Streptococcus suis. PLoS ONE 2019, 14, e0210154. [Google Scholar] [CrossRef] [PubMed]
  39. Toutain, P.L.; Pelligand, L.; Lees, P.; Bousquet-Mélou, A.; Ferran, A.A.; Turnidge, J.D. The pharmacokinetic/pharmacodynamic paradigm for antimicrobial drugs in veterinary medicine: Recent advances and critical appraisal. J. Vet. Pharmacol. Ther. 2021, 44, 172–200. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, L.; Xie, H.; Wang, Y.; Wang, H.; Hu, J.; Zhang, G. Pharmacodynamic Parameters of Pharmacokinetic/Pharmacodynamic (PK/PD) Integration Models. Front. Vet. Sci. 2022, 9, 860472. [Google Scholar] [CrossRef] [PubMed]
  41. Ahmad, I.; Huang, L.; Hao, H.; Sanders, P.; Yuan, Z. Application of PK/PD Modeling in Veterinary Field: Dose Optimization and Drug Resistance Prediction. BioMed Res. Int. 2016, 2016, 5465678. [Google Scholar] [CrossRef] [PubMed]
  42. Xu, L.; Wang, H.; Yang, X.; Lu, L. Integrated pharmacokinetics/pharmacodynamics parameters-based dosing guidelines of enrofloxacin in grass carp Ctenopharyngodon idella to minimize selection of drug resistance. BMC Vet. Res. 2013, 9, 126. [Google Scholar] [CrossRef]
  43. Cui, J.; Liu, Y.; Wang, R.; Weihang, T.; Drlica, K.; Zhao, X. The mutant selection window in rabbits infected with Staphylococcus aureus. J. Infect. Dis. 2006, 194, 1601–1608. [Google Scholar] [CrossRef]
  44. Olofsson, S.K.; Marcusson, L.L.; Komp Lindgren, P.; Hughes, D.; Cars, O. Selection of ciprofloxacin resistance in Escherichia coli in an in vitro kinetic model: Relation between drug exposure and mutant prevention concentration. J. Antimicrob. Chemother. 2006, 57, 1116–1121. [Google Scholar] [CrossRef] [PubMed]
  45. Vilalta, C.; Giboin, H.; Schneider, M.; El Garch, F.; Fraile, L. Pharmacokinetic/pharmacodynamic evaluation of marbofloxacin in the treatment of Haemophilus parasuis and Actinobacillus pleuropneumoniae infections in nursery and fattener pigs using Monte Carlo simulations. J. Vet. Pharmacol. Ther. 2014, 37, 542–549. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, L.; Kang, Z.; Yao, L.; Gu, X.; Huang, Z.; Cai, Q.; Shen, X.; Ding, H. Pharmacokinetic/Pharmacodynamic Integration to Evaluate the Changes in Susceptibility of Actinobacillus pleuropneumoniae After Repeated Administration of Danofloxacin. Front. Microbiol. 2018, 9, 2445. [Google Scholar] [CrossRef] [PubMed]
  47. Alieva, K.N.; Strukova, E.N.; Golikova, M.V.; Portnoy, Y.A.; Firsov, A.A. Concentration-Dependent Enrichment of Linezolid-Resistant Staphylococcus aureus in an in vitro Dynamic Model. Antibiot. Chemother. 2016, 61, 28–32. [Google Scholar]
  48. Xiong, M.; Wu, X.; Ye, X.; Zhang, L.; Zeng, S.; Huang, Z.; Wu, Y.; Sun, J.; Ding, H. Relationship between Cefquinome PK/PD Parameters and Emergence of Resistance of Staphylococcus aureus in Rabbit Tissue-Cage Infection Model. Front. Microbiol. 2016, 7, 874. [Google Scholar] [CrossRef] [PubMed]
  49. Zhang, B.; Gu, X.; Li, Y.; Li, X.; Gu, M.; Zhang, N.; Shen, X.; Ding, H. In vivo evaluation of mutant selection window of cefquinome against Escherichia coli in piglet tissue-cage model. BMC Vet. Res. 2014, 10, 297. [Google Scholar] [CrossRef] [PubMed]
  50. Kasimanickam, V.; Kasimanickam, M.; Kasimanickam, R. Antibiotics Use in Food Animal Production: Escalation of Antimicrobial Resistance: Where Are We Now in Combating AMR? Med. Sci. 2021, 9, 14. [Google Scholar] [CrossRef]
  51. Scott, H.M.; Acuff, G.; Bergeron, G.; Bourassa, M.W.; Gill, J.; Graham, D.W.; Kahn, L.H.; Morley, P.S.; Salois, M.J.; Simjee, S.; et al. Critically important antibiotics: Criteria and approaches for measuring and reducing their use in food animal agriculture. Ann. N. Y. Acad. Sci. 2019, 1441, 8–16. [Google Scholar] [CrossRef]
Table 1. MIC and MPC distribution values for 34 M. haemolytica strains.
Table 1. MIC and MPC distribution values for 34 M. haemolytica strains.
DrugBacteriostatic(S)/
Bactericidal(C)
MIC/MPC Distribution Values (µg/mL)
≤0.0160.0310.0630.1250.250.5124816≥32
MIC MIC BreapointMIC50/90/100
CeftiofurC331 ≤2≤0.016/≤0.016/0.031
EnrofloxacinC331 ≤0.25≤0.016/≤0.016/0.031
FlorfenicolS 2311 ≤21/2/2
MarbofloxacinC34 ≤1 *≤0.016/≤0.016/2
PradofloxacinC34 ≤0.125≤0.016/≤0.016/≤0.016
TildipirosinS 21913 ≤40.5/1/1
TilmicosinS 2182129 ≤80.5/8/8
TulathromycinS 22111 ≤160.5/1/2
MPC MPC50/90/100
Ceftiofur 313639 0.25/1/1
Enrofloxacin 715102 0.125/0.25/0.5
Florfenicol 1258 2/4/4
Marbofloxacin 1321 0.063/0.063/0.125
Pradofloxacin 6226 0.031/0.063/0.063
Tildipirosin 2842 2/4/8
Tilmicosin 103912 16/≥32/≥32
Tulathromycin 18106 2/4/8
MIC = minimum inhibitory concentration; MPC = mutant prevention concentration. * For Enterobacteriales.
Table 2. MIC and MPC distribution values for 41 P. multocida strains.
Table 2. MIC and MPC distribution values for 41 P. multocida strains.
DrugBacteriostatic(S)/
Bactericidal(C)
MIC/MPC Distribution Values (µg/mL)
≤0.0160.0310.0630.1250.250.5124816≥32
MIC MIC BreakpointMPC50/90/100
CeftiofurC41 ≤2≤0.016/≤0.016/≤0.016
EnrofloxacinC41 ≤0.25≤0.016/≤0.016/≤0.016
FlorfenicolS 1823 ≤20.5/0.5/0.5
MarbofloxacinC365 ≤1 *≤0.016/0.031/0.031
PradofloxacinC383 ≤0.125≤0.016/≤0.016/0.031
TildipirosinS 115141010 ≤80.5/2/2
TilmicosinS 2220152 ≤8 **2/4/8
TulathromycinS 8249 ≤160.25/0.5/0.5
MPC MPC50/90/100
Ceftiofur 38512121 0.125/0.25/0.5
Enrofloxacin 4111511 0.063/0.125/0.125
Florfenicol 338 1/1/1
Marbofloxacin 41013122 0.063/0.125/0.25
Pradofloxacin 22181 ≤0.016/0.031/0.063
Tildipirosin 28283 4/4/8
Tilmicosin 282074 8/16/≥32
Tulathromycin 102722 1/2/8
* For Enterobacteriales.** For M. haemolytica.
Table 3. Pharmacokinetic and pharmacodynamic values for eight antimicrobial agents.
Table 3. Pharmacokinetic and pharmacodynamic values for eight antimicrobial agents.
CompoundCmaxTissuemax AUC24Cmax/
MIC90
Cmax/
MPC90
AUC24/
MIC90
AUC24/
MPC90
T>MIC90 T>MPC90 % Protein BindingConcentration (C) or
Time (T) Dependent
M. haemolytica
Ceftiofur *6.92.64376431.36.923,50037610 days4–5 days95 **T>MIC
Enrofloxacin1.94.620.71118.87.61294.480.7>24 h22 h~46AUC/MIC, CMAX/MIC
Florfenicol3.72.94101.91.850.9350.925.524 h2 h~20T>MIC
Marbofloxacin [33]1.5 6.993.823.8431.3109.5>24 h~18 h~30AUC/MIC, CMAX/MIC
Pradofloxacin3.40.8113.2212.553.9825209.5>72 h>24 h~40AUC/MIC, CMAX/MIC
Tildipirosin [34]0.7714.824.90.770.1924.96.200~30AUC/MIC
Tilmicosin [35,36,37]0.87 17.20.110.032.20.5400~25T>MIC
Tulathromycin0.63.263.70.60.0863.715.900~40T>MIC
P. multocida
Ceftiofur6.92.64376431.327.623,500150410 days6 days
Enrofloxacin1.94.620.71118.815.21294.4165.7>24 h>24 h
Florfenicol3.72.94101.97.43.7203.8101.996 h>2 h
Marbofloxacin1.5 6.993.812222.655.220 h~10 h
Pradofloxacin3.40.8113.2212.5109.7825425.8>24 h>12 <18 h
Tildipirosin0.7714.824.90.380.1912.56.200
Tilmicosin0.87 17.20.210.054.31.100
Tulathromycin0.63.263.71.20.3127.431.900
* Based on total drug and does not account for protein binding. ** For desfuroylceftiofur.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Blondeau, J.M.; Fitch, S.D. Comparative Minimum Inhibitory and Mutant Prevention Drug Concentrations for Pradofloxacin and Seven Other Antimicrobial Agents Tested against Bovine Isolates of Mannheimia haemolytica and Pasteurella multocida. Pathogens 2024, 13, 399. https://doi.org/10.3390/pathogens13050399

AMA Style

Blondeau JM, Fitch SD. Comparative Minimum Inhibitory and Mutant Prevention Drug Concentrations for Pradofloxacin and Seven Other Antimicrobial Agents Tested against Bovine Isolates of Mannheimia haemolytica and Pasteurella multocida. Pathogens. 2024; 13(5):399. https://doi.org/10.3390/pathogens13050399

Chicago/Turabian Style

Blondeau, Joseph M., and Shantelle D. Fitch. 2024. "Comparative Minimum Inhibitory and Mutant Prevention Drug Concentrations for Pradofloxacin and Seven Other Antimicrobial Agents Tested against Bovine Isolates of Mannheimia haemolytica and Pasteurella multocida" Pathogens 13, no. 5: 399. https://doi.org/10.3390/pathogens13050399

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop