Pharmacokinetics and pharmacodynamics of minocycline against Acinetobacter

Similar documents
ETX2514SUL (sulbactam/etx2514) for the treatment of Acinetobacter baumannii infections

OPTIMIZATION OF PK/PD OF ANTIBIOTICS FOR RESISTANT GRAM-NEGATIVE ORGANISMS

Other β-lactamase Inhibitor (BLI) Combinations: Focus on VNRX-5133, WCK 5222 and ETX2514SUL

Extremely Drug-resistant organisms: Synergy Testing

Antimicrobial Pharmacodynamics

Animal models and PK/PD. Examples with selected antibiotics

Percent Time Above MIC ( T MIC)

Dynamic Drug Combination Response on Pathogenic Mutations of Staphylococcus aureus

Outline. Antimicrobial resistance. Antimicrobial resistance in gram negative bacilli. % susceptibility 7/11/2010

Tel: Fax:

Doripenem: A new carbapenem antibiotic a review of comparative antimicrobial and bactericidal activities

COMMITTEE FOR VETERINARY MEDICINAL PRODUCTS

Building a Better Mousetrap for Nosocomial Drug-resistant Bacteria: use of available resources to optimize the antimicrobial strategy

International Journal of Advances in Pharmacy and Biotechnology Vol.3, Issue-2, 2017, 1-7 Research Article Open Access.

Appropriate antimicrobial therapy in HAP: What does this mean?

VOL. XXIII NO. II THE JOURNAL OF ANTIBIOTICS 559. ANTIBIOTIC 6640.* Ill

Summary of unmet need guidance and statistical challenges

ETX2514: Responding to the global threat of nosocomial multidrug and extremely drug resistant Gram-negative pathogens

The pharmacological and microbiological basis of PK/PD : why did we need to invent PK/PD in the first place? Paul M. Tulkens

A Unique Approach to Managing the Problem of Antibiotic Resistance

CHSPSC, LLC Antimicrobial Stewardship Education Series

DETERMINING CORRECT DOSING REGIMENS OF ANTIBIOTICS BASED ON THE THEIR BACTERICIDAL ACTIVITY*

Int.J.Curr.Microbiol.App.Sci (2017) 6(3):

Introduction to Pharmacokinetics and Pharmacodynamics

DETERMINANTS OF TARGET NON- ATTAINMENT IN CRITICALLY ILL PATIENTS RECEIVING β-lactams

Pierre-Louis Toutain, Ecole Nationale Vétérinaire National veterinary School of Toulouse, France Wuhan 12/10/2015

Detection and Quantitation of the Etiologic Agents of Ventilator Associated Pneumonia in Endotracheal Tube Aspirates From Patients in Iran

ETX0282, a Novel Oral Agent Against Multidrug-Resistant Enterobacteriaceae

Prevalence of Metallo-Beta-Lactamase Producing Pseudomonas aeruginosa and its antibiogram in a tertiary care centre

Antibiotics in vitro : Which properties do we need to consider for optimizing our therapeutic choice?

In vitro pharmacodynamics of colistin against Acinetobacter baumannii clinical isolates

Sustaining an Antimicrobial Stewardship

JAC Bactericidal index: a new way to assess quinolone bactericidal activity in vitro

Antibiotic utilization and Pseudomonas aeruginosa resistance in intensive care units

DRUG-RESISTANT ACINETOBACTER BAUMANNII A GROWING SUPERBUG POPULATION. Cara Wilder Ph.D. Technical Writer March 13 th 2014

Proceedings of the 13th International Congress of the World Equine Veterinary Association WEVA

Jerome J Schentag, Pharm D

Test Method Modified Association of Analytical Communities Test Method Modified Germicidal Spray Products as Disinfectants

Evaluating the Role of MRSA Nasal Swabs

Antibiotic Kinetic and Dynamic Attributes for Community-Acquired Respiratory Tract Infections

Intrinsic, implied and default resistance

Barriers to Intravenous Penicillin Use for Treatment of Nonmeningitis

Update on Resistance and Epidemiology of Nosocomial Respiratory Pathogens in Asia. Po-Ren Hsueh. National Taiwan University Hospital

Scottish Medicines Consortium

Application of Pharmacokinetics/ Pharmacodynamics (PK/PD) in Designing Effective Antibiotic Treatment Regimens

Ceftaroline versus Ceftriaxone in a Highly Penicillin-Resistant Pneumococcal Pneumonia Rabbit Model Using Simulated Human Dosing

SZENT ISTVÁN UNIVERSITY. Doctoral School of Veterinary Science

International Journal of Antimicrobial Agents

Effect of pulmonary surfactant on antimicrobial activity in-vitro

Systemic Antimicrobial Prophylaxis Issues

Journal of Antimicrobial Chemotherapy Advance Access published August 26, 2006

Combating Antimicrobial Resistance with Extended Infusion Beta-lactams. Stephen Andrews, PharmD PGY-1 Pharmacy Practice Resident

Disclosure. Objectives. Combating Antimicrobial Resistance with Extended Infusion Beta-lactams

Evaluation of a computerized antimicrobial susceptibility system with bacteria isolated from animals

MDR Acinetobacter baumannii. Has the post antibiotic era arrived? Dr. Michael A. Borg Infection Control Dept Mater Dei Hospital Malta

Contribution of pharmacokinetic and pharmacodynamic parameters of antibiotics in the treatment of resistant bacterial infections

Recommended for Implementation at Step 7 of the VICH Process on 15 December 2004 by the VICH Steering Committee

COMMITTEE FOR MEDICINAL PRODUCTS FOR VETERINARY USE

Antimicrobial stewardship: Quick, don t just do something! Stand there!

GARY WOODNUTT* AND VALERIE BERRY SmithKline Beecham Pharmaceuticals, Collegeville, Pennsylvania

DR. MICHAEL A. BORG DIRECTOR OF INFECTION PREVENTION & CONTROL MATER DEI HOSPITAL - MALTA

Comparison of Efficacies of Oral Levofloxacin and Oral Ciprofloxacin in a Rabbit Model of a Staphylococcal Abscess

These recommendations were approved for use by the Pharmaceutical and Therapeutics Committee, RCWMCH on 1 February 2017.

Alasdair P. MacGowan*, Mandy Wootton and H. Alan Holt

Pharmacokinetic-pharmacodynamic profiling of four antimicrobials against Gram-negative bacteria collected from Shenyang, China

TREAT Steward. Antimicrobial Stewardship software with personalized decision support

Comparative studies on pulse and continuous oral norfloxacin treatment in broilers and turkeys. Géza Sárközy

EXCEDE Sterile Suspension

Quantification of Chloramphenicol in Chicken Using Xevo TQD with RADAR Technology

A pilot integrative knowledgebase for the characterization and tracking of multi resistant Acinetobacter baumannii in Colombian hospitals

MAGNITUDE OF ANTIMICROBIAL USE. Antimicrobial Stewardship in Acute and Long Term Healthcare Facilities: Design, Implementation and Challenges

Amikacin Inhale shows promising results in Phase II Study

Should we test Clostridium difficile for antimicrobial resistance? by author

CF WELL Pharmacology: Microbiology & Antibiotics

CME/CE QUIZ CME/CE QUESTIONS. a) 20% b) 22% c) 34% d) 35% b) Susceptible and resistant strains of typical respiratory

USA Product Label CLINTABS TABLETS. Virbac. brand of clindamycin hydrochloride tablets. ANADA # , Approved by FDA DESCRIPTION

2. Albany College of Pharmacy and Health Sciences, Albany, NY, USA

Antimicrobial Stewardship Strategy: Intravenous to oral conversion

ANTIBIOTICS USED FOR RESISTACE BACTERIA. 1. Vancomicin

SESSION XVI NEW ANTIBIOTICS

Towards Rational International Antibiotic Breakpoints: Actions from the European Committee on Antimicrobial Susceptibility Testing (EUCAST)

Defining Extended Spectrum b-lactamases: Implications of Minimum Inhibitory Concentration- Based Screening Versus Clavulanate Confirmation Testing

An evaluation of the susceptibility patterns of Gram-negative organisms isolated in cancer centres with aminoglycoside usage

MICHAEL J. RYBAK,* ELLIE HERSHBERGER, TABITHA MOLDOVAN, AND RICHARD G. GRUCZ

UCSF guideline for management of suspected hospital-acquired or ventilatoracquired pneumonia in adult patients

COMMITTEE FOR MEDICINAL PRODUCTS FOR VETERINARY USE (CVMP) REVISED GUIDELINE ON THE SPC FOR ANTIMICROBIAL PRODUCTS

Received 27 August 2002; returned 26 November 2002; revised 8 January 2003; accepted 11 January 2003

* gender factor (male=1, female=0.85)

Antibiotic stewardship in long term care

Use of Pharmacokinetics and Pharmacodynamics to Optimize Antimicrobial Treatment of Pseudomonas aeruginosa Infections

American Association of Feline Practitioners American Animal Hospital Association

In Vivo Efficacy of the Novel Aminoglycoside ACHN-490 in Murine Infection Models

Global Antibiotic Research and Development (GARD) Partnership Pasteur Institute DNDi Meeting February 29, 2016

Effects of Minocycline and Other Antibiotics on Fusobacterium necrophorum Infections in Mice

Pharmaceutical Form Ciprofloxacin 2 mg/ml Solution for infusion. Applicant Name Strength. Ciprofloxacin Nycomed. Ciprofloxacin Nycomed

SUMMARY OF PRODUCT CHARACTERISTICS. NUFLOR 300 mg/ml solution for injection for cattle and sheep

What s next in the antibiotic pipeline?

6.0 ANTIBACTERIAL ACTIVITY OF CAROTENOID FROM HALOMONAS SPECIES AGAINST CHOSEN HUMAN BACTERIAL PATHOGENS

Guidelines for Laboratory Verification of Performance of the FilmArray BCID System

The International Collaborative Conference in Clinical Microbiology & Infectious Diseases

Transcription:

AAC Accepted Manuscript Posted Online 6 March 2017 Antimicrob. Agents Chemother. doi:10.1128/aac.02371-16 Copyright 2017 American Society for Microbiology. All Rights Reserved. 1 2 Pharmacokinetics and pharmacodynamics of minocycline against Acinetobacter baumannii in a neutropenic murine pneumonia model 3 Running title: PK-PD for minocycline 4 5 6 Jian Zhou, a Kimberly R. Ledesma, a Kai-Tai Chang, a Henrietta Abodakpi, a Song Gao, a Vincent H. Tam a,b* 7 8 9 10 Department of Pharmacological and Pharmaceutical Sciences, a and Department of Pharmacy Practice and Translational Research, b University of Houston College of Pharmacy, Houston, TX 11 12 13 14 15 16 17 * Corresponding author and mailing address: University of Houston College of Pharmacy 1441 Moursund Street, Houston, TX 77030 Phone: (832) 842-8316; Fax: (832) 842-8383; E-mail: vtam@uh.edu 18 19 20 1

21 ABSTRACT 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 Multi-drug resistant (MDR) Acinetobacter baumannii is increasingly more prevalent in nosocomial infections. Although in vitro susceptibility of A. baumannii to minocycline is promising, the in vivo efficacy of minocycline has not been well established. In this study, the in vivo activity of minocycline was evaluated in a neutropenic murine pneumonia model. Specifically, we investigated the relationship between minocycline exposure and bactericidal activity using 5 A. baumannii isolates with a broad range of susceptibility (MIC ranged from 0.25 mg/l to 16 mg/l). The pharmacokinetics of minocycline (single dose of 25 mg/kg, 50 mg/kg, 100 mg/kg and a humanized regimen, given intraperitoneally) in serum and epithelial lining fluid (ELF) were characterized. Dose linearity was observed for doses up to 50 mg/kg and pulmonary penetration ratios (AUC ELF 0-24h /AUC serum 0-24h) ranged from 2.5 to 2.8. Pharmacokinetic-pharmacodynamics (PK-PD) indices values in ELF for various dose regimens against different A. baumannii isolates were calculated. Maximum efficacy at 24h was approximately 1.5 log reduction of pulmonary bacterial burdens from baseline. AUC/MIC was the PK- PD index most closely correlating to the bacterial burden (r 2 = 0.81). The required AUC ELF 0-24h /MIC for maintaining stasis and achieving 1 log reduction were 140 and 410, respectively. These findings could guide the treatment of infections caused by A. baumannii using minocycline in the future. Additional studies to examine resistance development during therapy are warranted. 42 2

43 INTRODUCTION 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 Acinetobacter baumannii is an opportunistic pathogen which mostly affects patients with compromised immune function. It is commonly implicated in infections of the respiratory tract, bloodstream, urinary tract and skin. In recent years, the prevalence of multi-drug resistant (MDR) A. baumannii isolates has been increasing (1). Many commonly used antimicrobial agents are ineffective, and increased mortality was reported in patients infected with MDR A. baumannii (2, 3). Minocycline is a semi-synthetic derivative of tetracycline that has a broad spectrum of activity against Gram-positive and Gram-negative bacteria. Compared to other tetracyclines, it has excellent penetration into tissues and a long elimination half-life (4, 5). Conventionally, minocycline has not been used as a first-line agent in Gram-negative bacterial infections. However, the shortage of new effective antibiotics against MDR A. baumannii has motivated us to reevaluate the utility of minocycline. Despite good in vitro results of minocycline against MDR A. baumannii (6), satisfactory clinical response was not consistently seen in patients treated with minocycline (7). 59 60 61 62 63 64 65 Although the typical dosing regimen of minocycline is 200 mg/day, 400 mg IV q12h with a 800 mg loading dose has been used in humans for acute spinal cord injury (8). A higher minocycline daily dose may be necessary for infections caused by MDR A. baumannii. However, the in vivo efficacy of minocycline has not been well established and the rationale of minocycline dosing regimen design needs to be further substantiated. In this study, the relationship between minocycline exposure and bactericidal activity was studied in a neutropenic 3

66 67 68 69 murine pneumonia model. The minocycline exposure at the infection site was correlated to systemic exposure. We expect these findings will be used to optimize the dosing regimens of minocycline for more favorable therapeutic outcomes. 70 71 MATERIALS AND METHODS 72 Chemicals and reagents 73 74 75 76 77 78 79 80 81 82 Liquid chromatography-mass spectrometry (LC-MS)-grade water, methanol and acetonitrile were purchased from EMD Millipore Corporation (Billerica, MA). Methyl-sulfoxide (DMSO) was obtained from EM SCIENCE (Gibbstown, NJ). LC- MS-grade formic acid, minocycline hydrochloride and doxycycline hyclate powder were purchased from Sigma-Aldrich (St. Louis, MO). Mouse serum was obtained from Equitech-Bio, Inc (Kerrville, TX). The Ultra Clean microbial DNA isolation kit was from Mo Bio laboratories, Inc. (Carlsbad, CA); DNA Acinetobacter strain typing kit was a product of Diversilab (Marcy I Etoile, France), and Taq DNA polymerase was purchased from Bioline USA Inc (Randolph, MA). The urea assay kit was from BioAssay Systems (Hayward, CA). 83 Bacterial isolates 84 85 86 Five A. baumannii isolates (1 laboratory wild-type isolate and 4 clinical isolates) with a wide range of minocycline MICs were used. Susceptibilities to minocycline, doxycycline, amikacin and imipenem were determined by the broth dilution 4

87 88 89 90 91 92 93 94 95 method as recommended by CLSI. Pseudomonas aeruginosa strain ATCC 27853 (American Type Culture Collection, Manassas, VA) was used as a control strain. The clonal relatedness of the A. baumannii isolates was assessed by repetitive-element based polymerase chain reaction (rep-pcr) (9, 10). Briefly, genomic DNA was isolated using Ultra Clean microbial DNA isolation kit, and used as the template for the Acinetobacter strain typing kit. The DNA fragments of rep-pcr products were separated by the Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA), and compared by the DiversiLab software using the Pearson correlation coefficient (Bacterial Barcodes, Inc., Athens, GA). 96 Neutropenic murine pneumonia model 97 98 99 100 101 102 103 104 105 A neutropenic murine pneumonia model was used to examine the in vivo bactericidal activity of minocycline. The experimental setup was as previously described (11, 12). Briefly, female Swiss Webster mice between 20 and 25 g (Harlan, Indianapolis, IN) were rendered neutropenic by two doses of intraperitoneal cyclophosphamide prior to the experiment (150 mg/kg in day -4 and 100 mg/kg in day -1). Anesthetized mice were inoculated with approximately 10 7 colony forming unit (CFU) of A. baumannii under laryngoscopic guidance. The protocol was approved by Institutional Animal Care and Use Committee of the University of Houston. 106 107 108 Infected animals were treated with different dosing regimens of minocycline approximately 2 h after infection. For reference, there was a no treatment control group for each bacterial isolate. There were 80 animals given 23 dosing regimen 5

109 110 111 112 113 114 115 - bacterium combinations in total (n 3 for each regimen). For reference, there was a placebo (i.e., no treatment) control group for each bacterial isolate. The bacterial burdens in lung tissues were determined at 0h (baseline) and 24h after first dose of minocycline as described elsewhere (11, 13). Briefly, after the animals were sacrificed by CO 2 asphyxiation, the lung tissues were harvested and homogenized in sterile saline. Pulmonary bacterial burdens were determined by quantitative culture, and normalized by the weights of lung tissues. 116 Minocycline pharmacokinetic study 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 The single-dose pharmacokinetics of minocycline in mouse serum and ELF were characterized. Minocycline was administered intraperitoneally in 67 animals 2 h after infection (by AB 1261). Serum or bronchoalveolar lavage (BAL) samples were collected serially over time (n 3 for each time point). Blood samples were collected by cardiac puncture. BAL samples were recovered through the trachea after 1 ml of saline was injected into the lungs. Minocycline concentrations were determined by the LC-MS/MS method as detailed below. The minocycline concentrations in ELF were calculated by correcting that in BAL for urea concentrations as described previously (14). The serum and ELF concentrationtime profiles were co-modelled by a modified two compartmental model in ADAPT 5 (15) (data not shown). Three regimens were initially evaluated: 25 mg/kg, 50 mg/kg and 100 mg/kg. Based on the preliminary results, a humanized regimen mimicking the human serum concentration-time profile of minocycline (when a clinical dose of 200 mg is given intravenously to humans) was validated. The area under the curve (AUC) of the serum and ELF concentration-time 6

132 133 134 135 136 137 profiles were derived by integrating the best-fit instantaneous concentrations with respect to time. Pulmonary penetration ratio of minocycline was estimated by the AUC ratio of ELF to serum. In addition, using the best-fit parameters, various PK- PD indices (i.e., AUC/MIC, C max /MIC and T%>MIC) for ELF profiles were calculated for different dosing regimens against different A. baumannii isolates (data not shown). 138 Minocycline assay 139 140 141 142 143 144 145 146 147 148 149 150 151 152 Minocycline concentrations in serum and epithelial lining fluid (ELF) samples were determined by a validated liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. The samples for the standard curve were prepared by spiking minocycline into blank serum. Briefly, 10 µl of serum or ELF sample was mixed with 10 µl water (with 0.5% DMSO), 30 µl of internal standard (10 mg/l doxycycline in water) and 170 µl acetonitrile (with 0.03% formic acid). The samples were centrifuged for 15 min at 18,000 g. An aliquot of 40 µl of the supernatants were transferred into a new tube, and evaporated under a stream of ambient air. After being reconstituted with 1 ml of 50% methanol, 5 µl of the samples were injected into the Ultra Performance Liquid Chromatography (UPLC) system. The conditions and parameters of LC-MS/MS were described previously (11). The linear range of quantification was 0.0625-128 mg/l. The intra-day and inter-day variability of the assay method were < 2.5% and < 3.6%, respectively. 153 Data analysis 7

154 155 156 157 158 159 160 The relationships between minocycline PK-PD indices (unbound drug exposures in ELF) and bacterial burden in lung tissues at 24 h were described by an inhibitory sigmoid E max model, as shown previously (16). The coefficient of determination was used to discriminate the PK-PD index most closely correlated to bactericidal activity. In addition, the best-fit parameters were also used to derive the required PK-PD exposures for maintaining stasis or achieving 1 log reduction of bacterial tissue burden at 24h. 161 162 RESULTS 163 Susceptibility and clonality assessment 164 165 166 167 The known tetracycline resistance mechanisms and susceptibilities of the A. baumannii isolates to various antibiotics are shown in Table 1. Cross-resistance was observed between minocycline and doxycycline. The isolates were found to belong to 3 clonally diverse groups (data not shown). 168 Minocycline pharmacokinetic study 169 170 171 172 173 174 The initial estimation of serum AUC 0-24h and the best-fit C max suggested that the pharmacokinetics of 25 mg/kg and 50 mg/kg doses were within the linear range. Also, the daily dose of 50 mg/kg was found to be comparable to the human equivalent AUC reported in a previous study (11). Therefore, the total daily dose was split into 5 doses to mimic a humanized regimen. Briefly, 18 mg/kg of minocycline was given at 0 h to achieve the serum C max similar to that in humans, 8

175 176 177 and 4 supplemental doses (11 mg/kg, 9 mg/kg, 8 mg/kg and 4 mg/kg given at 4 h, 9 h, 14 h and 22 h) were given to maintain the serum concentrations around the target pharmacokinetic profile reported in humans (4). 178 179 180 181 182 183 184 185 The data from these 3 dosing regimens were co-modeled. The best-fit minocycline concentration-time profiles of different dosing regimens are shown in Figure 1. The profiles of minocycline in both serum (r 2 = 0.977) and ELF (r 2 = 0.952) were captured satisfactorily (Figure 2). The elimination half-life in serum was 2.6 h. The serum AUC 0-24h were 34 mg*h/l, 68 mg*h/l and 63 mg*h/l for 25 mg/kg, 50 mg/kg and humanized regimen, respectively; while the ELF AUC 0-24h were 94 mg*h/l, 189 mg*h/l and 175 mg*h/l. The pulmonary penetration ratio of minocycline was 2.8. 186 187 188 189 190 191 192 Compared to 50 mg/kg, the AUC 0-24h of the 100 mg/kg dose observed was more than 3 times higher. Therefore, the concentration-time profiles of 100 mg/kg were analyzed separately (shown in Figure 1). The r 2 were 0.971 and 0.867 for serum and ELF concentration-time profiles (Figure 2). The elimination half-life was prolonged (3.9 h), suggesting saturable clearance for the 100 mg/kg dose. The AUC 0-24h were 227 mg*h/l and 564 mg*h/l for serum and ELF, respectively. The pulmonary penetration ratio was 2.5. 193 Minocycline in vivo PD study 194 195 196 The baseline bacterial burdens in lung tissues ranged from 7.75 to 8.18 log CFU/g. In the absence of treatment, the tissue burdens increased to 8.60 to 9.65 log CFU/g in 24h. All treatment groups infected with the minocycline-resistant 9

197 198 199 isolate (AB7416) showed similar results as the no treatment control group, while the other susceptible / intermediate isolates were suppressed to various extents. PK-PD correlation 200 201 202 203 204 205 Looking at all the data collectively, the relationships between minocycline PK-PD indices in ELF and bacterial tissue burden at 24h are shown in Figure 3. The strongest relationship was observed when the tissue burdens were correlated with AUC ELF 0-24h /MIC (r 2 = 0.81). Using the best-fit parameters, the required AUC ELF 0-24h /MIC for maintaining stasis was 140, and the required AUC ELF 0-24h/MIC for achieving 1 log reduction was 410. 206 207 DISCUSSION 208 209 210 211 212 The shortage of new effective antibiotics against MDR A. baumannii prompted us to maximize the effectiveness of the currently available drugs, such as minocycline. With a good understanding of the drug exposures and bacterial susceptibilities likely to be encountered, a PK-PD model could facilitate the optimal use of minocycline. 213 214 215 216 217 218 In vitro susceptibility results of minocycline are promising. Denys et al. reported the susceptibility data of Gram-negative bacteria from the U.S. between 2005 and 2011. In that study (n=883), the susceptibility rate of MDR Acinetobacter isolates to minocycline was 72.1 %, whereas most of other drugs examined were found to be resistant (17). In addition, the pharmacokinetics of minocycline in animals and humans has been characterized previously. In horses, the 10

219 220 221 222 223 224 225 226 227 228 229 230 231 elimination half-life was 7.7 ± 1.9 h (mean ± SD) after a 2.2 mg/kg single IV injection (18). A prolonged half-life (11.5 ± 3.2 h, mean ± SD) at steady state was observed when the dose was increased to 4 mg/kg q12h (19). The observation of non-linear pharmacokinetics reported with high doses was consistent with our findings from the current study (100 mg/kg in mice), and might be due to saturation of drug metabolism (20). The elimination half-life in humans ranged from 12 h to 18 h, and the AUC 0- of serum concentration-time profile were 70-86 mg*h/l after 200 mg was given intravenously (4). Up to 800 mg of minocycline daily has been given intravenously in a clinical trial for acute spinal cord injury (8), however there is little known to date about the dose linearity of minocycline in humans. A prospective study examining the safety, tolerability, and pharmacokinetics of minocycline (single and multiple ascending doses) in heathy adults has been planned (ClinicalTrials.gov identifier: NCT02802631). 232 233 234 235 236 237 238 239 240 241 Free drug exposures in serum are commonly used in PK-PD studies. In the current study, we found that minocycline concentrations in ELF were higher than those observed in the serum. Subsequently, the PK-PD analysis was performed with unbound minocycline concentrations in ELF (i.e., the site of infection), as they were deemed to be more relevant to therapeutic outcomes of pneumonia. Furthermore, serum protein binding of the tetracyclines may not be as straight forward as one might anticipate. Tigecycline was previously shown to exhibit atypical serum protein binding; the fraction of binding was higher with increasing total drug concentration in serum (21). The serum protein binding of minocycline was found be dependent on the experimental conditions. However, our 11

242 243 244 245 246 247 preliminary data showed that protein bindings of minocycline in human and mouse sera were comparable at different concentrations (Zhou J et al. J Antimicrob Chemother, in press). Since the ELF exposure of minocycline achieved in humans after a clinical dose is not available, the total drug concentration-time profiles in the serum was mimicked and we assumed pulmonary penetration ratios of minocycline in humans and mice were similar. 248 249 250 251 252 253 254 255 Using 3 different bacterial isolates, regimens with the same daily dose (50 mg/kg single dose, 25 mg/kg every 12 h and humanized regimen) showed similar in vivo efficacy. Since these dosing regimens have similar AUC 0-24h but different C max and percentage of time above MIC, AUC ELF 0-24h /MIC was expected be the PK-PD index best correlating to the bacterial burden. Using a wide range of minocycline exposure and bacterial susceptibility (23 dose regimen bacterium combinations in total), our initial finding was subsequently substantiated by the results of the full PK-PD analysis. 256 257 258 259 260 261 262 263 264 The PK-PD model described the relationship between minocycline exposure and bactericidal activity quantitatively. Maximum efficacy observed was approximately 1.5 log reduction of bacterial burdens from baseline. The required AUC ELF 0-24h /MIC values for maintaining stasis or achieving 1 log reduction of bacterial tissue burdens were also estimated. AUC ELF 0-24h of humanized regimen (a clinical dose of 200 mg is given intravenously to humans) was 175 mg*h/l. With a MIC of 0.25 mg/l, the corresponding AUC ELF 0-24h /MIC (700) is expected to achieve more than 1-log kill. However, the therapeutic outcomes will vary when the MICs are different. Using these estimates, minocycline dosing 12

265 266 267 268 269 270 regimens could be optimized when the susceptibility of an A. baumannii isolate is known. Furthermore, it is also important to avoid selective amplification of the resistant subpopulation(s) during treatment. In the future, additional studies are warranted to evaluate resistance development during therapy, and correlating minocycline exposures to therapeutic outcomes of patients with infections caused by A. baumannii. 271 272 273 In conclusion, the pharmacokinetics of minocycline in mice serum and ELF were characterized. We have also identified threshold target exposures for achieving different pharmacodynamic responses in A. baumannii infections. 274 275 276 ACKNOWLEDGEMENT 277 278 279 This study was supported in part by the Medicines Company. The authors thank David Griffith for providing minocycline (pharmaceutical grade) powder and isolate AB 1129. 280 281 DISCLOSURE 282 V.H.T. is a consultant on the Advisory Board of Tetraphase Pharmaceuticals. 13

283 REFERENCES 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 1. Keen EF, 3rd, Murray CK, Robinson BJ, Hospenthal DR, Co EM, Aldous WK. 2010. Changes in the incidences of multidrug-resistant and extensively drug-resistant organisms isolated in a military medical center. Infect Control Hosp Epidemiol 31:728-732. 2. Abbo A, Carmeli Y, Navon-Venezia S, Siegman-Igra Y, Schwaber MJ. 2007. Impact of multi-drug-resistant Acinetobacter baumannii on clinical outcomes. Eur J Clin Microbiol Infect Dis 26:793-800. 3. Zheng YL, Wan YF, Zhou LY, Ye ML, Liu S, Xu CQ, He YQ, Chen JH. 2013. Risk factors and mortality of patients with nosocomial carbapenemresistant Acinetobacter baumannii pneumonia. Am J Infect Control 41:e59-63. 4. Agwuh KN, MacGowan A. 2006. Pharmacokinetics and pharmacodynamics of the tetracyclines including glycylcyclines. J Antimicrob Chemother 58:256-265. 5. Watanabe A, Anzai Y, Niitsuma K, Saito M, Yanase K, Nakamura M. 2001. Penetration of minocycline hydrochloride into lung tissue and sputum. Chemotherapy 47:1-9. 6. Castanheira M, Mendes RE, Jones RN. 2014. Update on Acinetobacter species: mechanisms of antimicrobial resistance and contemporary in vitro activity of minocycline and other treatment options. Clin Infect Dis 59 Suppl 6:S367-373. 14

305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 7. Goff DA, Bauer KA, Mangino JE. 2014. Bad bugs need old drugs: a stewardship program's evaluation of minocycline for multidrug-resistant Acinetobacter baumannii infections. Clin Infect Dis 59 Suppl 6:S381-387. 8. Casha S, Zygun D, McGowan MD, Bains I, Yong VW, Hurlbert RJ. 2012. Results of a phase II placebo-controlled randomized trial of minocycline in acute spinal cord injury. Brain 135:1224-1236. 9. Tam VH, Chang KT, LaRocco MT, Schilling AN, McCauley SK, Poole K, Garey KW. 2007. Prevalence, mechanisms, and risk factors of carbapenem resistance in bloodstream isolates of Pseudomonas aeruginosa. Diagn Microbiol Infect Dis 58:309-314. 10. Syrmis MW, O'Carroll MR, Sloots TP, Coulter C, Wainwright CE, Bell SC, Nissen MD. 2004. Rapid genotyping of Pseudomonas aeruginosa isolates harboured by adult and paediatric patients with cystic fibrosis using repetitive-element-based PCR assays. J Med Microbiol 53:1089-1096. 11. Bowers DR, Cao H, Zhou J, Ledesma KR, Sun D, Lomovskaya O, Tam VH. 2015. Assessment of minocycline and polymyxin B combination against Acinetobacter baumannii. Antimicrob Agents Chemother 59:2720-2725. 12. Tam VH, Ledesma KR, Schilling AN, Lim TP, Yuan Z, Ghose R, Lewis RE. 2009. In vivo dynamics of carbapenem-resistant Pseudomonas aeruginosa selection after suboptimal dosing. Diagn Microbiol Infect Dis 64:427-433. 15

328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 13. Hirsch EB, Guo B, Chang KT, Cao H, Ledesma KR, Singh M, Tam VH. 2013. Assessment of antimicrobial combinations for Klebsiella pneumoniae carbapenemase-producing K. pneumoniae. J Infect Dis 207:786-793. 14. He J, Abdelraouf K, Ledesma KR, Chow DS, Tam VH. 2013. Pharmacokinetics and efficacy of liposomal polymyxin B in a murine pneumonia model. Int J Antimicrob Agents 42:559-564. 15. D'Argenio DZ, Schumitzky A, Wang X. 2009. ADAPT 5 User s Guide: Pharmacokinetic/Pharmacodynamic Systems Analysis Software. Los Angeles: Biomedical Simulations Resource, University of Southern California. 16. Lim TP, Ledesma KR, Chang KT, Hou JG, Kwa AL, Nikolaou M, Quinn JP, Prince RA, Tam VH. 2008. Quantitative assessment of combination antimicrobial therapy against multidrug-resistant Acinetobacter baumannii. Antimicrob Agents Chemother 52:2898-2904. 17. Denys GA, Callister SM, Dowzicky MJ. 2013. Antimicrobial susceptibility among gram-negative isolates collected in the USA between 2005 and 2011 as part of the Tigecycline Evaluation and Surveillance Trial (T.E.S.T.). Ann Clin Microbiol Antimicrob 12:24. 18. Nagata S, Yamashita S, Kurosawa M, Kuwajima M, Hobo S, Katayama Y, Anzai T. 2010. Pharmacokinetics and tissue distribution of minocycline hydrochloride in horses. Am J Vet Res 71:1062-1066. 16

350 351 352 353 354 355 356 357 358 19. Schnabel LV, Papich MG, Divers TJ, Altier C, Aprea MS, McCarrel TM, Fortier LA. 2012. Pharmacokinetics and distribution of minocycline in mature horses after oral administration of multiple doses and comparison with minimum inhibitory concentrations. Equine Vet J 44:453-458. 20. Nelis HJ, De Leenheer AP. 1982. Metabolism of minocycline in humans. Drug Metab Dispos 10:142-146. 21. Mukker JK, Singh RP, Derendorf H. 2014. Determination of atypical nonlinear plasma-protein-binding behavior of tigecycline using an in vitro microdialysis technique. J Pharm Sci 103:1013-1019. 359 360 361 362 363 364 365 366 367 368 17

369 TABLE 1: Susceptibilities of A. baumannii isolates Isolate Source Tetracyline resistance mechanism(s) MIC (mg/l) Minocycline Doxycycline Imipenem Amikacin AB BAA 747 Laboratory Wild-type 0.25 0.25 0.25 2 370 AB 7283 AB 1261 AB 1129 AB 7416 Clinical Clinical Clinical Clinical Moderate overexpression of adeb Moderate overexpression of adeb adeabc and adeijk overexpressed tetb, moderate overexpression of adeb 0.5 0.5 128 128 1 0.5 128 128 4 4 8 32 16 128 16 > 512 371 Note: Bold fonts depict resistant phenotype as defined by the Clinical and Laboratory Standards Institute (CLSI) 372 373 374 18

375 FIGURE 1: Minocycline serum and ELF concentration-time profiles 376 377 378 Concentration (mg/l) 25 20 15 10 5 25mg/kg Observed serum data Observed ELF data Best-fit serum profile Best-fit ELF profile 379 380 0 0 4 8 12 16 20 24 Time (h) ---- Target serum profile in humans 381 382 383 Concentration (mg/l) 60 50 40 30 20 10 50mg/kg 384 385 0 0 4 8 12 16 20 24 Time (h) 386 387 388 Concentration (mg/l) 16 12 8 4 Humanized regimen 389 390 0 0 4 8 12 16 20 24 Time (h) 391 19

392 393 394 395 Concentration (mg/l) 120 100 80 60 40 20 100 mg/kg 396 0 0 4 8 12 16 20 24 Time (h) 397 The observed data were shown as mean ± SD. 398 399 400 401 402 403 404 405 406 407 408 20

409 FIGURE 2: Correlation between observed and best-fit PK data 410 A. 25 mg/kg, 50 mg/kg and humanized regimen 411 412 Serum concentration (mg/l) r 2 = 0.977 16 50 ELF concentration (mg/l) r 2 = 0.952 413 414 415 416 Best fit 12 8 4 0 0 4 8 12 16 Observed Best fit 40 30 20 10 0 0 10 20 30 40 50 Observed 417 B. 100 mg/kg 418 40 Serum concentration (mg/l) r 2 = 0.971 100 ELF concentration (mg/l) r 2 = 0.867 419 30 80 420 Best fit 20 Best fit 60 40 421 10 20 422 423 0 0 10 20 30 40 Observed 0 0 20 40 60 80 100 Observed 424 The dashed line is the line of identity (i.e., y=x). 425 21

426 FIGURE 3: Correlation of PK-PD indices in ELF and tissue burden at 24h 427 428 429 Log10 CFU/g 10 9 8 7 6 R 2 = 0.81 Best-fit profile Observed data 430 5 0 1 2 3 4 Log10 AUC/MIC 431 432 433 434 Log10 CFU/g 10 9 8 7 6 R 2 =0.73 435 5 0 50 100 %T>MIC 436 437 438 439 440 Log10 CFU/g 10 9 8 7 6 5 R 2 = 0.78-1 0 1 2 3 Log10 C max /MIC 441 442 443 Each data point represents an observation from a single animal. In view of the logarithmic scale used, AUC/MIC values were input as 1 and C max /MIC values were input as 0.1 for placebo controls. 22