Pharmacodynamic model of the dynamic response of Pseudomonas aeruginosa biofilms to drug treatments

Chronic infection by gram-negative bacteria such as Pseudomonas aeruginosa is a leading cause of morbidity and mortality in cystic fibrosis patients in whom overabundant mucus and the formation of bacterial biofilms pose barriers to drug delivery and effectiveness. Accurate pharmacokinetic-pharmacodynamic (PK-PD) models of biofilm treatment could be used to guide formulation and administration strategies to better control bacterial lung infections. To this end, we have developed a detailed pharmacodynamic model of P. aeruginosa treatment with the front-line antibiotics, tobramycin and colistin, and validated it on a detailed dataset of killing dynamics. A compartmental model structure was developed in which the key features are diffusion of drug through a boundary layer to the bacteria, concentration dependent interactions with bacteria, and passage of the bacteria through successive transit states before death. The number of transit states employed was greater for tobramycin, which is a ribosomal inhibitor, than for colistin, which disrupts bacterial membranes. For both drugs, the experimentally observed delay in killing of bacteria following drug exposure was replicated and was consistent with the diffusion time, though for tobramycin, there was an additional delay reflected in the model by passage through the transit states. For each drug, the PD model with a single set of parameters described data across a ten-fold range of concentrations and for both continuous and transient exposure protocols. Furthermore, the parameters fit for each drug individually were used to model the response of biofilms to combined treatment with tobramycin and colistin. The ability to predict drug response over a range of administration protocols allows this PD model to be integrated with PK descriptions to describe in vivo antibiotic response dynamics and to predict drug delivery strategies for improved control of bacterial lung infections. Author Summary Biofilms are self-assembling bacterial communities that adhere to a surface and encase themselves in a protective coating. Biofilm infections are notoriously difficult to treat with conventional antibiotic administrations. To understand better the dynamics of bacterial biofilm killing in response to antibiotic treatment, we developed a mathematical model that integrates several features: drug diffusion through a boundary layer that includes the biofilm casing, concentration dependent cell damage, and passage of the cell through damaged states to eventual death. We validated the model by comparison with an extensive published dataset of biofilm response to treatment with the antibiotics, tobramycin and colistin. The model fits to these datasets were able to capture the observed trends for several antibiotic administration protocols, with model parameters reflecting the differences in mechanism of action between the two drugs. This validated model can be integrated with pharmacokinetic descriptions of drug distribution in the body over time to predict dosing and administration protocols for preclinical and clinical studies.

[1]  Vassily Hatzimanikatis,et al.  Spatio-temporal modeling of the crowding conditions and metabolic variability in microbial communities , 2021, PLoS Comput. Biol..

[2]  L. Villarreal-Treviño,et al.  Analysis of biofilm production and expression of adhesion structures of circulating Clostridioides difficile strains from Mexico. , 2021, Enfermedades infecciosas y microbiologia clinica.

[3]  M. Windbergs,et al.  Therapy of infected wounds: overcoming clinical challenges by advanced drug delivery systems , 2021, Drug Delivery and Translational Research.

[4]  M. Ntzouni,et al.  Colistin Dependence in Extensively Drug-Resistant Acinetobacter baumannii Strain Is Associated with ISAjo2 and ISAba13 Insertions and Multiple Cellular Responses , 2021, International journal of molecular sciences.

[5]  Minh Tam Tran Thi,et al.  Pseudomonas aeruginosa Biofilms , 2020, International journal of molecular sciences.

[6]  P. Stewart,et al.  Conceptual Model of Biofilm Antibiotic Tolerance That Integrates Phenomena of Diffusion, Metabolism, Gene Expression, and Physiology , 2019, Journal of bacteriology.

[7]  Andrey G. Cherstvy,et al.  Burst statistics in an early biofilm quorum sensing model: the role of spatial colony-growth heterogeneity , 2019, Scientific Reports.

[8]  Christel A. S. Bergström,et al.  Model-Based Drug Development in Pulmonary Delivery: Pharmacokinetic Analysis of Novel Drug Candidates for Treatment of Pseudomonas aeruginosa Lung Infection , 2019, Journal of pharmaceutical sciences.

[9]  Mathias Müsken,et al.  Breaking the Vicious Cycle of Antibiotic Killing and Regrowth of Biofilm-Residing Pseudomonas aeruginosa , 2018, Antimicrobial Agents and Chemotherapy.

[10]  Robert Tyminski Breaking it , 2018, Male Alienation at the Crossroads of Identity, Culture and Cyberspace.

[11]  L. Frunzo,et al.  Continuum and discrete approach in modeling biofilm development and structure: a review , 2018, Journal of mathematical biology.

[12]  S. Rice,et al.  Mesoscopic Energy Minimization Drives Pseudomonas aeruginosa Biofilm Morphologies and Consequent Stratification of Antibiotic Activity Based on Cell Metabolism , 2018, Antimicrobial Agents and Chemotherapy.

[13]  R. Gehring,et al.  Limitations of MIC as sole metric of pharmacodynamic response across the range of antimicrobial susceptibilities within a single bacterial species , 2016, Scientific Reports.

[14]  P. Stewart,et al.  Reaction–diffusion theory explains hypoxia and heterogeneous growth within microbial biofilms associated with chronic infections , 2016, npj Biofilms and Microbiomes.

[15]  Yue Zheng,et al.  Biofilm formation mechanisms and targets for developing antibiofilm agents. , 2015, Future medicinal chemistry.

[16]  S. Britland,et al.  Strategies for antimicrobial drug delivery to biofilm. , 2014, Current pharmaceutical design.

[17]  I. Banat,et al.  Microbial biofilms: biosurfactants as antibiofilm agents , 2014, Applied Microbiology and Biotechnology.

[18]  J. Knobloch,et al.  Successful treatment of extensively drug-resistant Pseudomonas aeruginosa osteomyelitis using a colistin- and tobramycin-impregnated PMMA spacer. , 2014, International journal of antimicrobial agents.

[19]  D. Thornton,et al.  Cystic fibrosis: an inherited disease affecting mucin-producing organs. , 2014, The international journal of biochemistry & cell biology.

[20]  N. Cogan,et al.  Effect of Periodic Disinfection on Persisters in a One-Dimensional Biofilm Model , 2013, Bulletin of mathematical biology.

[21]  I. Rajman PK/PD modelling and simulations: utility in drug development. , 2008, Drug discovery today.

[22]  B. Ploeger,et al.  Mechanism-based pharmacokinetic-pharmacodynamic (PK-PD) modeling in translational drug research. , 2008, Trends in pharmacological sciences.

[23]  Philip S. Stewart,et al.  Modeling Antibiotic Tolerance in Biofilms by Accounting for Nutrient Limitation , 2004, Antimicrobial Agents and Chemotherapy.

[24]  Philip S. Stewart,et al.  Diffusion in Biofilms , 2003, Journal of bacteriology.

[25]  R. Donlan,et al.  Biofilms: Microbial Life on Surfaces , 2002, Emerging infectious diseases.

[26]  P. Stewart,et al.  Biofilm accumulation model that predicts antibiotic resistance of Pseudomonas aeruginosa biofilms , 1994, Antimicrobial Agents and Chemotherapy.

[27]  P. Stewart,et al.  A model of biofilm detachment , 1993, Biotechnology and bioengineering.

[28]  Nichols,et al.  Inhibition of tobramycin diffusion by binding to alginate , 1988, Antimicrobial Agents and Chemotherapy.