SlideShare a Scribd company logo
1 of 22
Download to read offline
1
Title: Evaluation of Pharmacokinetic/Pharmacodynamic Relationships of PD-0162819, a Biotin1
Carboxylase Inhibitor Representing a New Class of Antibacterial Compounds, Using In Vitro2
Infection Models3
4
5
Running Title: PK/PD Characterization: Novel Biotin Carboxylase Inhibitor6
7
8
Authors: Adam Ogden*, Michael Kuhn, Michael Dority, Susan Buist, Shawn Mehrens, Tong9
Zhu, Deqing Xiao, J. Richard Miller, and Debra Hanna10
11
Pfizer Global Research and Development12
Ann Arbor, MI, and Groton, CT13
14
15
*Corresponding author. Mailing address: Pfizer Global Research and Development, Eastern16
Point Rd., Groton, CT 06340. Phone: (860) 686-9197. Fax: (860) 441-4036. E-mail:17
adam.ogden@pfizer.com.18
Copyright © 2011, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved.
Antimicrob. Agents Chemother. doi:10.1128/AAC.00090-11
AAC Accepts, published online ahead of print on 10 October 2011
2
Abstract19
The present study investigated the pharmacokinetic/pharmacodynamic (PK/PD)20
relationships of a prototype biotin carboxylase (BC) inhibitor, PD-0162819, against H.21
influenzae 3113 (HI-3113) in static concentration time-kill (SCTK) and one-compartment22
chemostat in vitro infection models. HI-3113 was exposed to PD-0162819 concentrations of23
0.5-16x the minimal inhibitory concentration (MIC; 0.125 µg/mL) and area-under-the-curve24
(AUC)/MIC ratio of 1-1100 in SCTK and chemostat experiments, respectively. Serial samples25
were collected over 24 h. For efficacy driver analysis, a sigmoid Emax model was fit to the26
relationship between bacterial density changes over 24 h and corresponding PK/PD indices. A27
semi-mechanistic PK/PD model describing the time-course of bacterial growth and death was28
developed. AUC/MIC best-explained efficacy (r2
=0.95) compared to peak drug concentration29
(Cmax)/MIC (r2
=0.76) and time above MIC (T>MIC; r2
=0.88). Static effects and 99.9% killing30
were achieved at AUC/MIC values of 500 and 600, respectively. For time-course analysis, the31
net bacterial growth rate constant, maximum bacteria density, and maximum kill rate constant32
were similar in SCTK and chemostat studies, but PD-0162819 was more potent in SCTK as33
compared to the chemostat (EC50=0.046 vs. 0.34 µg/mL). In conclusion, basic PK/PD34
relationships for PD-0162819 were established using in vitro dynamic systems. Although the35
bacterial growth parameters and maximum drug effect were similar in SCTK and chemostat, PD-36
0162819 appeared to be more potent in SCTK, illustrating the importance of understanding37
differences of preclinical models. Additional studies are needed to determine the in vivo38
relevance of these results.39
3
Introduction40
Steadily increasing bacterial resistance to existing antibiotics continues to be a major41
public health concern (3, 8). Because most new antibacterial agents represent chemical42
modification of existing chemical classes of antibacterial agents (5), it is suspected that the43
limited options of chemically-distinct antibiotics has led to extensive drug resistance among44
bacterial pathogens. Therefore, it is of utmost importance to identify novel, safe, and effective45
antibacterial agents which work through unique antibacterial biologic mechanisms. The46
discovery of a new chemical class of antibacterial compounds, the pyridopyrimidines, targeting47
bacterial biotin carboxylase (BC) was recently reported (14, 15) and offers the potential that this48
novel chemical class, targeting a unique antibacterial mechanism, can be developed into drugs49
effective against multi-drug resistant bacteria.50
As compared to development of drugs from an existing chemical class, the discovery of a51
novel class of compounds presents extra challenges (1, 5). The translation of52
pharmacokinetic/pharmacodynamic (PK/PD) relationships between animal infection models and53
human patients has been well-established for several existing chemical classes across a variety of54
indications (1), but for novel chemical classes, PK/PD relationships, and the translation of these55
relationships between in vitro systems, animals, and humans, are not known. Moreover, the56
physicochemical and pharmacokinetic properties of compounds at early stages of the drug57
discovery process are often not optimized for extensive concentration-response testing in animal58
models (21). Thus, as an alternative, in vitro infection models offer a rapid and resource-sparing59
method to determine PK/PD relationships. Building and applying mathematical PK/PD models60
that quantitatively describe the time-course of bacterial replication/death and drug effects enables61
the construction of a more efficient drug discovery process. Furthermore, these quantitative62
4
PK/PD relationships derived from in vitro data can inform future in vivo testing as to optimal63
dose selection and dosing intervals and thereby reduce resources necessary to perform adequate64
in vivo experiments. They can also provide the framework for understanding knowledge gaps65
and for determining optimal drug properties (e.g., pharmacokinetics) required for a successful66
drug candidate (6, 7, 9).67
The present study investigated the PK/PD relationships of a prototype BC inhibitor, PD-68
0162819, against H. influenzae 3113 (HI-3113) in static concentration time-kill (SCTK) and one-69
compartment chemostat in vitro infection models. The objectives of this study were to a)70
establish a basic understanding of concentration-response relationships in vitro for a prototype71
BC inhibitor and b) use in vitro and mathematical modeling tools to guide the drug discovery72
program by understanding the translation among in vitro infection models.73
74
Materials and Methods75
Compound, microorganism, and susceptibility studies. PD-0162819 was synthesized76
by Pfizer chemists (14). Broth micro-dilution susceptibility testing was performed using a77
BioMek FX robotic workstation (Beckman-Coulter, Fullerton, CA). A β-lactamase producing78
clinical isolate of H. influenzae, HI-3113, was tested using Haemophilus Test Medium (HTM;79
PMLMicrobiologicals, Wilsonville, OR) and incubated at 35°C in ambient atmosphere as80
described by the Clinical and Laboratory Standards Institute (CLSI) (17).81
SCTK experiments. In vitro SCTK testing was performed following CLSI methodology82
(17). Specifically, testing was carried out in 10 mL of HTM and incubated at 35°C with 5% CO283
atmosphere. PD-0162819 concentrations ranged from 0.06-2 µg/mL (0.5-16× MIC; MIC= 0.12584
µg/mL) and were determined by LC-MS/MS to remain constant during the course of the85
5
experiment. Serial media samples (100 µL/sample) were collected at t=0, 2, 4, 6, and 24 h.86
Post-exposure MIC testing was performed, and changes in MICs were not observed.87
One-compartment chemostat infection model. In vitro dynamic concentration studies88
were performed using a one-compartment chemostat system as previously described (12, 23).89
The chemostat system consisted of a 250 mL glass chamber with ports for the addition and90
removal of test media via polyethylene tubes connected to peristaltic pumps, injection of drug91
solution, and removal of media samples. Single-dose and dose fractionation experiments were92
performed. Prior to each experiment, colonies from an overnight growth of HI-3113 on93
chocolate agar were added to HTM as necessary to obtain a suspension of 108
CFU/mL. To94
produce a starting inoculum of ~106
CFU/mL, 2.5 mL of this suspension were added to each95
flask. A drug stock solution of PD-0162819 was prepared at the start of each experiment, and96
this solution was diluted with saline such that a dosing volume of 0.5 mL was added to each97
flask via bolus dosing. To increase the solubility of PD-0162819, 3% SBE-β-cyclodextrin was98
added to the dosing solution. Because PD-0162819 is projected to have a short half-life in vivo99
(data not shown), pump rates for all chemostat experiments were set to achieve a drug100
elimination half-life of four hours. A dose escalation study was performed to determine the101
efficacious exposure range using AUC/MIC of 1 to 1100 (MIC=0.125 µg/mL). To determine the102
efficacy driver, a dose fractionation study was performed using exposures corresponding to the103
ED20, ED50, and ED80 from the dose escalation study divided into 1×/day (dosed at t=0 h),104
2×/day (t=0 and 12 h), or 3×/day (t=0, 6, and 12 h) dosing regimens. Media samples were105
collected at 0, 0.5, 1, 2, 4, 6, 12, and 24 h after dosing for analysis of drug concentrations and106
determination of bacteria densities. Post-exposure MIC testing was performed, and changes in107
MICs were not observed.108
6
Determination of bacteria density. Media samples were washed with saline to remove109
any drug carry-over. Samples were then reconstituted, diluted 10-fold, and 100 µL of diluted110
and undiluted samples were plated onto chocolate agar. After 24 h of incubation, colony111
forming units (CFU/mL) were counted and corrected for dilution. Because an undiluted 100 µL112
sample was used for determining bacteria density in samples with low bacteria counts, the limit113
of detection was 10 CFU/mL.114
Drug concentration analysis. Media samples were stored at −20°C until analysis. A115
freeze-thaw study conducted during method development indicated that PD-0162819 was stable116
at −20°C for >4 weeks and <5% degradation occurred after repeated freeze-thaw cycles. The117
samples were thawed at room temperature, spiked with 200 µL of internal standard (200 ng/mL118
of PF-01135527 in acetonitrile), and vortexed for 1 min. Following centrifugation at 5000 rpm119
for 5 min, 3 µL of the supernatants were injected into a validated LC-MS/MS system. A Varian120
Polaris C-18-A column (50 × 2.0 mm) was employed and run under ambient reversed-phase121
conditions. The mobile phases were A) 100% water with 0.1% formic acid and B) 100%122
acetonitrile with 0.1% formic acid. The gradient was programmed with a flow rate of123
250 µL/min starting at 10% mobile phase B to reach 70% mobile phase B in 1.5 min and 90%124
mobile phase B in an additional 1 min. At the end of the program, the analytical column was re-125
equilibrated with 10% mobile phase B for 1 min. The total cycle time for the assay was 3.6126
min/sample. Molecular ion precursor-product transitions of 393.7→ 298.1 and 382→ 271.1127
were used for PD-0162819 and internal standard, respectively, in multiple-reaction monitoring128
(MRM) mode. Data acquisition, integration of peak areas, regression analysis, and sample129
quantification were performed using the standard quantitative analysis package of Analyst 1.4130
(Applied Biosystems, Foster City, CA). A 12-point standard curve consisting of PD-0162819131
7
concentrations ranging from 5 – 10,000 ng/mL was used to quantify PD-0162819 concentrations132
in samples. Where appropriate, media samples were diluted for analysis. The lower limit of133
quantitation (LLOQ) was 5 ng/mL. Using standards of known concentration, accuracy was134
determined to be ±20%, and intra-day and inter-day precision was ±20%.135
Endpoint efficacy driver analysis. Data collected using the one-compartment136
chemostat system was used to determine the efficacy driver. The change of bacteria density over137
24 h (∆log CFU/mL) was calculated and plotted against corresponding endpoint PK/PD138
parameters (AUC/MIC, Cmax/MIC, and T>MIC). Observed drug concentration data was used to139
calculate the pharmacokinetic parameters. A sigmoid Emax model was fit to the relationship140
between ∆log CFU/mL and corresponding PK/PD indices. This model was used to determine141
the endpoint PK/PD index that best-explained efficacy and the magnitude of each parameter to142
achieve bacterial killing ranging from 0-log to ≥3-log killing.143
Time-course PK/PD data analysis. SCTK and chemostat PK/PD data were modeled144
via a two-step process using NONMEM version V (GloboMax LLC, Ellicott City, MD). Based145
on a study to assess the chemical and metabolic stability of PD-0162819 in HTM, drug146
concentrations were determined by LC-MS/MS to be constant throughout the course of SCTK147
studies. Less than 5% degradation occurred over 24 h at 35°C. A population pharmacokinetic148
one-compartment open model with first-order elimination (kelim) and proportional error was used149
to fit the drug-concentration-time profiles. The population pharmacokinetic model incorporated150
inter-individual (i.e., inter-flask) variability of clearance (CL) and volume of distribution (Vd) to151
describe the observed drug concentration data in each flask. The pharmacokinetic parameters152
were then fixed in the PK/PD model, and the bacteria density time-course profiles were modeled153
using a modified net effect mathematical model that is outlined in Figure 1. Similar154
8
mathematical models have been previously described (e.g., 2, 10, 13, 16, 18, 20, 22). In this155
model, changes in drug concentrations over time were described using Equation 1:156
melim
m
Ck
C
×−=
dt
d
(1)157
where Cm is the PD-0162819 concentration in HTM. kelim is the first-order drug elimination rate158
constant, which is determined by the pump rate of the chemostat system. kelim can also be159
represented as CL/Vd. kelim was 0 h-1
in SCTK because drug was not flushed out of the system.160
In the chemostat, kelim was approximately 0.17 h-1
to achieve a drug half-life of approximately161
four hours. For chemostat data, kelim was estimated using the previously-described population162
pharmacokinetic model.163
The bacterial population was hypothesized to be composed of three bacterial populations164
of non-replicating, replicating, and damaged bacterial cells. To model the time-course of165
bacterial density, Equations 2-5 were applied:166








++
+
×
−= flushglagγ
50
γ
m
γ
mkill
1
1
kk
ECC
Ck
CFU
CFU
dt
d
(2)167
( ) 







−
+
×
−
−
×+= flushγ
50
γ
m
γ
mkill
max
totalmax
rep2glag1
2
k
ECC
Ck
N
CFUN
kCFUkCFU
CFU
dt
d
(3)168
( ) ( )flushdlag3γ
50
γ
m
γ
mkill
21
3
kkCFU
ECC
Ck
CFUCFU
CFU
+−







+
×
+=
dt
d
(4)169
321total CFUCFUCFUCFU ++= (5)170
where, as illustrated in Figure 1, CFU1, CFU2, and CFU3 represent the density of non-replicating,171
replicating, and damaged bacterial cells, respectively. CFUtotal is the sum of all bacterial cells in172
the system. For the drug effect, kkill is the rate constant for maximal bacterial killing, EC50 is the173
drug concentration at which 50% of the maximal killing occurs, and γ is the Hill coefficient174
9
corresponding to the degree of sigmoidicity of the concentration-response curve. For the175
replication rate, krep is the net growth rate constant, and Nmax is the maximum number of bacteria176
in the in vitro system. As CFUtotal approaches Nmax, the total bacterial population reaches a177
stationary phase and was observed as a plateau of the bacteria density-time profile. Where178
appropriate, a bacterial growth lag rate constant (kglag) and bacterial death lag rate constant (kdlag)179
were incorporated into the model to describe lags in bacterial growth and death. For the dynamic180
chemostat data, kflush is the rate constant associated with flushing HTM, bacteria, and drug out of181
the in vitro system. Because HTM, bacteria, and drug were flushed out of the system at a rate182
determined by the pump rate, kflush has the same magnitude as kelim. For modeling SCTK data,183
kflush was excluded from the model because HTM, bacteria, and drug were not flushed out of the184
system. Best-fit parameters of the PK/PD model were obtained using first-order conditional185
estimation, and the goodness of fit of the models to the experimental data was assessed by visual186
inspection of observed versus predicted plots, coefficient of determination, evaluation of187
weighted residuals (versus time and concentration), and comparison of objective function values188
for nested models.189
190
Results191
SCTK of PD-0162819 against HI-3113. PD-0162819 demonstrated concentration-192
dependent killing achieving a 3-log kill after 6 h. PD-0162819 was bactericidal at concentrations193
of ≥2× the MIC. The PK/PD model (Figure 1) adequately described the observed bacteria194
density data (r2
=0.91). Observed data and model-predicted fits to the data are shown in Figure195
2A. The maximum bacterial net growth rate constant in this system was 2.04 ± 0.14 h-1
, and the196
death lag rate constant was 1.91 ± 0.20 h-1
. A growth lag rate constant was not estimated due to197
10
the absence of data between 0 and 2 h. The maximum number of bacteria in the system was198
estimated to be 108.80 ± 0.15
CFU/mL. The maximum drug effect (kkill) was 2.53 ± 0.14 h-1
, and199
the EC50 of PD-0162819 against HI-3113 was estimated to be 0.0456 ± 0.0043 µg/mL. Model200
parameter estimates and 90% confidence intervals are shown in Table 1.201
Dynamic concentration time-kill study in chemostat. The PK/PD model outlined in202
Figure 1 adequately described the observed time-course of bacteria density data (r2
=0.98).203
Observed data and model-predicted fits to the pharmacodynamic data are shown in Figure 2B.204
The maximum bacterial net growth rate constant in this system was 1.80 ± 0.10 h-1
, and the death205
lag rate constant was 3.06 ± 1.10 h-1
. A growth lag was observed over the first hour of the206
experiment, and the growth lag rate constant was estimated to be 0.171 ± 0.065 h-1
. The207
maximum number of bacteria in the system was estimated to be 109.28 ± 0.04
CFU/mL. The208
maximum drug effect (kkill) was 2.02 ± 0.11 h-1
, and the EC50 of PD-0162819 against HI-3113209
was estimated to be 0.340 ± 0.041 µg/mL. Model parameter estimates and 90% confidence210
intervals are shown in Table 1.211
Efficacy driver analysis in chemostat. The one-compartment chemostat system was212
used to determine the efficacy driver of PD-0162819 against HI-3113. Although all three PK/PD213
indices exhibited relationships with efficacy (Figure 3), AUC/MIC was the best predictor of214
antibacterial efficacy (r2
=0.95) compared to Cmax/MIC (r2
=0.76) and T>MIC (r2
=0.88). A static215
effect over 24 h was achieved at an AUC/MIC value of 500. An AUC/MIC magnitude of 600216
was required to achieve 3-log killing (i.e., 99.9% killing) against HI-3113.217
218
219
220
11
Discussion221
The data and modeling presented in this report represent initial studies to characterize the222
PK/PD relationships of PD-0162819, a prototype BC inhibitor. Using two different in vitro223
infection systems and mathematical modeling tools, a basic understanding of endpoint efficacy224
driver and time-course concentration-response relationships were established for this novel225
compound which works through a unique mechanism of action. The results of this study indicate226
there is a significant need for understanding the quantitative translation of this data from in vitro227
systems to in vivo systems and, ultimately, to humans.228
In this study, both endpoint efficacy drivers and time-course PK/PD relationships were229
evaluated. Typically, endpoint efficacy driver approaches have been utilized for understanding230
antibacterial PK/PD (4). However, endpoint data often minimizes the importance of the time-231
course of drug effects. Endpoint efficacy driver studies typically assess the change in bacterial232
densities between time=0 h and time=24 h, which essentially ignores the time-course of drug233
effects over the 24 h period. By understanding the drug effects of the entire time-course of the234
experiment, a more thorough understanding of PK/PD relationships can be achieved. Even so,235
endpoint efficacy driver analyses are traditionally used for developing an understanding of236
PK/PD relationships for antibacterial agents. Thus, this study was designed to enable analysis of237
both the endpoint efficacy driver and time-course concentration-response relationships.238
Efficacy driver analysis results indicate that all three PK/PD indices exhibit some trend239
with efficacy. However, based on visual inspection of the plots and the coefficient of240
determination, AUC/MIC appears to be a better predictor of efficacy compared to Cmax/MIC and241
T>MIC. Although T>MIC exhibits a strong coefficient of determination, it is not likely that242
12
T>MIC is a good predictor of efficacy due to the observed variation of approximately 5-logs of243
bacterial killing among the different dosing regimens that achieved 100% T>MIC.244
The mathematical model presented in Figure 1 is the most parsimonious model that was245
fit to the observed data. It is similar to previously published models (e.g., 2, 10, 13, 16, 18, 20,246
22), but the basic structure of the model is most similar to the net effect model (2). Comparisons247
of the different mathematical models showed that potency estimates produced by the various248
mathematical models were similar (analysis not shown). An additional compartment was added249
to the basic net effect model to more accurately describe the apparent growth lag phase over the250
first hour of the chemostat experiments but was not incorporated for SCTK data due to lack of251
data in this early phase. This growth lag phase is hypothesized to be the result of non-replicating252
cells during the initial stages of the experiments. After this early phase, the cells are assumed to253
begin replicating. In addition, an extra compartment was added to describe the apparent death254
lag, which is hypothesized to be caused by damaged cells, that was more obvious in SCTK255
experiments. Because bacterial death was delayed relative to drug concentrations, application of256
a transit compartment model was useful for describing this observation (19). This model257
assumes that bacterial cells are damaged by drug, slow their proliferation rates, and eventually258
die. In addition, because bacteria were constantly being flushed out the chemostat system, a259
parameter (kflush) was added to the model to account for this. Without this parameter, the260
potency estimate would have been overestimated. This parameter was not needed when261
modeling the SCTK data because bacteria were not flushed from the SCTK system.262
Even though HI-3113 replication rates were similar among the two infection systems, the263
potency of PD-0162819 was ~7× more potent in SCTK than in the chemostat system.264
Differences in experimental conditions between the two study designs can account for this265
13
finding. In the chemostat system, nutrients are replenished via fresh media while drug, wastes,266
and old media are being removed from the system. This differs from the SCTK design where267
less optimal growth conditions exist (i.e., nutrients are fixed and used up during the course of the268
experiment). Because the chemostat system provides more optimal bacterial growth conditions269
compared to SCTK, it is not surprising that PD-0162819 appears less potent in the chemostat.270
Rebound of bacteria counts over the course of the experiment was not observed in SCTK271
studies. Although collecting more extensive samples between 6 and 24 h would allow for a more272
definitive determination of potential resistance development in SCTK studies, the lack of273
resistant subpopulations was supported by the low frequency of resistance (~1 resistant cell in274
>108
cells) (14), which is well-above the starting inoculum of 106
CFU/mL used in these studies.275
Thus, the observed experimental data did not warrant the inclusion of additional276
“subpopulations” of resistant bacteria into the mathematical model, which have been277
incorporated into previously published models (e.g., 2, 13, 20, 22). In addition, the278
pyridopyrimidine inhibitors are the only antibacterial agents known to target BC, and, therefore,279
no pre-existing clinical resistance is expected. Although rebound was observed in the chemostat280
studies, this rebound was likely caused by reductions in drug concentrations over the course of281
the experiments and not by the emergence of resistant bacterial subpopulations.282
Although this study focused on in vitro PK/PD relationships, simply understanding in283
vitro PK/PD of a compound is not sufficient. To be useful, the in vivo relevance, and ultimately284
the clinical relevance, of in vitro relationships need to be determined. In addition, only one285
bacterial isolate was tested in this study. The AUC/MIC exposure targets presented in this report286
will likely be modified when additional clinical isolates are taken into consideration (11). At this287
point, the results of this study have been used to guide the discovery project team in the process288
14
of synthesizing a more optimal compound. Attempts to optimize the molecule have included289
improving the physicochemical properties, potency, pharmacokinetics, and other parameters.290
In conclusion, mathematical modeling of data generated in in vitro infection systems was291
successfully employed to evaluate the PK/PD relationships of a novel BC inhibitor early in the292
drug discovery program. Although HI-3113 replication rates were similar among the two293
systems, the potency of PD-0162819 was ~7× more potent in SCTK than in the chemostat294
system, illustrating the importance of understanding differences of preclinical models when295
making decisions about drug exposures required for efficacy. Thus, the results of this study296
indicate that PK/PD relationships need to be interpreted with respect to the system in which the297
data is generated. Different in vitro and in vivo systems are likely to produce different results,298
which underlies the importance of understanding the quantitative translation of these systems to299
humans. The relevance of this modeling approach can be enhanced by translating this approach300
to in vivo systems. Thus, additional studies with additional strains of bacteria are required to301
determine the in vivo relevance of these results.302
303
15
References304
1. Ambrose, P.G., S.M. Bhavnani, C.M. Rubino, A. Louie, T. Gumbo, A. Forrest, and305
G.L. Drusano. 2007. Pharmacokinetics-pharmacodynamics of antimicrobial therapy:306
It's not just for mice anymore. Clin. Infect. Dis. 44:79-86.307
2. Campion, J.J., P.J. McNamara, and M.E. Evans. 2005. Pharmacodynamic modeling308
of ciprofloxacin resistance in Staphylococcus aureus. Antimicrob. Agents Chemother.309
49:209-219.310
3. Charles, P.G.P. and M.L. Grayson. 2004. The dearth of new antibiotic development:311
Why we should be worried and what we can do about it. Med. J. of Australia. 181:549-312
553.313
4. Craig, W.A. 1998. Pharmacokinetic/pharmacodynamic parameters: Rationale for314
antibacterial dosing of mice and men. Clin. Infect. Dis. 26:1-10.315
5. Fernandes, P. 2006. Antibacterial discovery and development – The failure of success?316
Nature Biotechnology 24:1497-1503.317
6. Gabrielsson, J., A.R. Green, and P.H. Van der Graaf. 2010. Optimising in vivo318
pharmacology studies – Practical PKPD considerations. J. Pharmacol. Toxicol. Methods.319
61:146-56.320
7. Gabrielsson J., and A.R. Green. 2009. Quantitative pharmacology or321
pharmacokinetic- pharmacodynamic integration should be a vital component in322
integrative pharmacology. J. Pharmacol. Exp. Ther. 331:767-74.323
8. Levy, S.B., and B. Marshall. 2004. Antibacterial resistance worldwide: Causes,324
challenges and responses Nature Medicine. 10:S122 - S129.325
16
9. Liu, K.K., P. Cornelius, T.A. Patterson, Y. Zeng, S. Santucci, E. Tomlinson, C.326
Gibbons, T.S. Maurer, R. Marala, J. Brown, J.X. Kong, E. Lee, W. Werner, Z.327
Wenzel, and C. Vage. 2010. Design and synthesis of orally-active and selective328
azaindane 5HT2c agonist for the treatment of obesity. Bioorg. Med. Chem. Lett. 20:266-329
71.330
10. Liu, P., K.H. Rand, B. Obermann, and H. Derendorf. 2005. Pharmacokinetic-331
pharmacodynamic modeling of antibacterial activity of cefpodoxime and cefixime in in332
vitro kinetic models. Int. J. Antimicrob. Agents 25:120-129.333
11. MacGowan, A.P., R. Reynolds, A.R. Noel, and K.E. Bowker. 2009. Bacterial strain-334
to-strain variation in pharmacodynamic index magnitude, a hitherto unconsidered factor335
in establishing antibiotic clinical breakpoints. Antimicrob. Agents Chemother. 53:5181-336
5184.337
12. McGrath, B., K.C. Lamp, and M.J. Rybak. 1993. Pharmacodynamic effects of338
extended dosing intervals of imipenem alone and in combination with amikacin against339
Pseudomonas aeruginosa in an in vitro model. Antimicrob. Agents Chemother. 37:1931-340
1937.341
13. Meagher, A.K., A. Forrest, A. Dalhoff, H. Stass, and J.J. Schentag. 2004. Novel342
pharmacokinetic-pharmacodynamic model for prediction of outcomes with an extended-343
release formulation of ciprofloxacin. Antimicrob. Agents Chemother. 48:2061-2068.344
14. Miller, J.R., S. Dunham, I. Mochalkin, C. Banotai, M. Bowman, S. Buist, B. Dunkle,345
D. Hanna, J.H. Harwood, M.D. Huband, A. Karnovsky, M. Kuhn, C. Limberakis,346
J.Y. Liu, S. Mehrens, T.W. Mueller, L. Narasimhan, A. Ogden, J. Ohren, V.J.V.N.347
Prasad, J.A. Shelly, L. Skerlos, M. Sulavik, H.V. Thomas, S. VanderRoest, L. Wang,348
17
Z. Wang, A. Whitton, T. Zhu, and C.K. Stover. 2009. A class of selective349
antibacterials derived from a protein kinase inhibitor pharmacophore. Proc.Nat. Acad.350
Sci. USA 106:1737-42.351
15. Mochalkin, I., J.R. Miller, L. Narasimhan, V. Thanabal, P. Erdman, P.B. Cox,352
V.J.V.N. Prasad, S. Lightle, M.D. Huband, and C.K. Stover. 2009. Discovery of353
antibacterial biotin carboxylase inhibitors by virtual screening and fragment-based354
approaches. ACS Chem. Bio. 4:473-83.355
16. Mouton, J.W., and A.A. Vinks. 2005. Pharmacokinetic/pharmacodynamic modeling of356
antibacterials in vitro and in vivo using bacterial growth and kill kinetics. Clin.357
Pharmacokinet. 44:201-210.358
17. National Committee for Clinical Laboratory Standards. 1999. Methods for359
determining bactericidal activity of antimicrobial agents. Approved Guideline M26-A.360
National Committee for Clinical Laboratory Standards, Wayne, PA.361
18. Schaper, K.J., S. Schubert, and A. Dalhoff. 2005. Kinetics and quantification of362
antibacterial effects of beta-lactams, macrolides, and quinolones against gram-positive363
and gram-negative RTI pathogens. Infection. 33(Suppl 2): 3-14.364
19. Simeoni, M., P. Magni, C. Cammia, G. De Nicolao, V. Croci, E. Pesenti, M.365
Germani, I. Poggesi, and M. Rocchetti. 2004. Predictive pharmacokinetic-366
pharmacodynamic modeling of tumor growth kinetics in xenograft models after367
administration of anticancer agents. Cancer Res. 64:1094-1101.368
20. Tam, V.H., A.N. Schilling, K. Poole, and M. Nikolaou. 2007. Mathematical modeling369
response of Pseudomonas aeruginosa to meropenem. J. Antimicrob. Chemother.370
60:1302-1309.371
18
21. van de Waterbeemd, H. 2009. Improving compound quality through in vitro and in372
silico physicochemical profiling. Chem. Biodivers. 6:1760-6.373
22. Yano, Y., T. Oguma, H. Nagata, and S. Sasaki. 1998. Application of logistic growth374
model to pharmacodynamic analysis of in vitro bactericidal kinetics. J. Pharm. Sci.375
87:1177-1183.376
23. Zinner, S. H., M. Dudley, and J. Blaser. 1986. In vitro models for the study of377
combination antibiotic therapy in neutropenic patients. Am. J. Med. 80:156-160.378
19
379
380
Figure 1. Schematic representation of the mathematical PK/PD model. The total bacteria381
density (CFUtotal) is the combination of non-replicating (CFU1), replicating (CFU2), and damaged382
cells (CFU3). Bacterial replication eventually results in bacterial counts reaching a stationary383
phase, which is determined by the total amount of bacteria in the system (Nmax). The replication384
rate (krep) is modified as bacterial counts increase [krep×((Nmax−CFUtotal)/Nmax)] and, ultimately,385
achieve a stationary phase. The maximum drug effect (kkill), drug concentration associated with386
50% of the maximum effect (EC50), drug concentration in the media (Cm), and Hill coefficient (γ)387
determine the magnitude of the drug effect [kkill×Cm
γ
/( Cm
γ
+ EC50
γ
)]. Rate constants associated388
with a growth lag (kglag), death lag (kdlag), and flushing of bacteria out of the in vitro system389
(kflush) are also included in the model. See the Materials and Methods section for equations and a390
more complete description of the model.391
392
kelim
CmPK:
krep
kglag
kdlag
Drug
Effect
Drug Effect
kflush kflush kflush
PD: CFU1 CFU2 CFU3
20
393
0
2
4
6
8
10
0 6 12 18 24
BacteriaDensity(logCFU/mL)
Time (h)
0
0.5x
1x
2x
4x
8x
16x
x MIC
394
0
2
4
6
8
10
0 6 12 18 24
BacteriaDensity(logCFU/mL)
Time (h)
0
5
8
11
26
46
130
260
530
1100
AUC/MIC
395
Figure 2. PK/PD modeling of PD-0162819 against H. influenzae 3113 in A) SCTK and B)396
chemostat. Symbols represent observed data. Lines represent model predictions.397
398
A
B
21
399
-6
-4
-2
0
2
4
6
0 200 400 600 800 1000 1200
BacteriaDensityChangeOver24h
AUC/MIC
r2 = 0.95
-6
-4
-2
0
2
4
6
0 50 100 150 200
Cmax/MIC
r2 = 0.76
-6
-4
-2
0
2
4
6
0 20 40 60 80 100
T>MIC (%)
r2 = 0.88
400
401
Figure 3. Efficacy driver analysis of PD-0162819 against H. influenzae 3113 in the chemostat system. Closed circles represent402
observed data, and solid lines represent model-predicted values.403
22
Table 1. PK/PD modeling results of the time-course effects of PD-0162819 against H.404
influenzae 3113 in SCTK and chemostat systems.405
406
Parameter Units
Static
Concentration
Time Kill
Dynamic Concentration
Time Kill
krep h-1 2.04 ± 0.14a
(1.8 – 2.3)b
1.80 ± 0.10
(1.6 – 2.0)
Nmax CFU/mL
108.80 ± 0.15
(108.6
– 109.0
)
109.28± 0.04
(109.2
– 109.3
)
kkill h-1 2.53 ± 0.14
(2.3 – 2.8)
2.02 ± 0.11
(1.8 – 2.2)
EC50 µg/mL
0.0456 ± 0.0043
(0.039 – 0.053)
0.340 ± 0.041
(0.27 – 0.41)
γ 
1.80 ± 0.15
(1.6 – 2.1)
1.88 ± 0.42
(1.2 – 2.6)
kglag h-1

0.171 ± 0.065
(0.07 – 0.28)
kdlag h-1 1.91 ± 0.20
(1.6 – 2.2)
3.06 ± 1.10
(1.2 – 4.9)
r2
 0.91 0.98
a
NONMEM estimate ± standard error.407
b
90% confidence intervals in parentheses.408

More Related Content

What's hot

Phase clinicaltrial
Phase clinicaltrialPhase clinicaltrial
Phase clinicaltriallillibabu
 
Pharmacokinetics And Pharmacodynamic of Biotechnology Drugs - Trilok Shahare
Pharmacokinetics And Pharmacodynamic of Biotechnology Drugs - Trilok ShaharePharmacokinetics And Pharmacodynamic of Biotechnology Drugs - Trilok Shahare
Pharmacokinetics And Pharmacodynamic of Biotechnology Drugs - Trilok ShahareTrilok Shahare
 
Clinical pharmacology.Basics.
Clinical pharmacology.Basics.Clinical pharmacology.Basics.
Clinical pharmacology.Basics.Eugene Shorikov
 
Importance of clinical pharmacokinetic studies
Importance of clinical pharmacokinetic studiesImportance of clinical pharmacokinetic studies
Importance of clinical pharmacokinetic studiesJohn Douglas
 
Repurposed drugs and safety monitoring
Repurposed drugs and safety monitoring Repurposed drugs and safety monitoring
Repurposed drugs and safety monitoring PHARMAQUEST Vydehi
 
Pharmacodynamics, ADRs & Preclinical drug testing
Pharmacodynamics, ADRs & Preclinical drug testingPharmacodynamics, ADRs & Preclinical drug testing
Pharmacodynamics, ADRs & Preclinical drug testingYogendra Nayak
 
Intelligent Drug Delivery System
Intelligent Drug Delivery SystemIntelligent Drug Delivery System
Intelligent Drug Delivery Systemmayurkhivansara
 
Pharmacokinetic Drug-Drug interactions
Pharmacokinetic Drug-Drug interactionsPharmacokinetic Drug-Drug interactions
Pharmacokinetic Drug-Drug interactionsAreej Abu Hanieh
 
Allometric scaling
Allometric scaling Allometric scaling
Allometric scaling kafle03
 
Pharmacodynamics for BPH
Pharmacodynamics for BPHPharmacodynamics for BPH
Pharmacodynamics for BPHPravin Prasad
 
Pharmacotherapeutics
PharmacotherapeuticsPharmacotherapeutics
Pharmacotherapeuticsatharshamim
 
Drug transport and drug targeting - rumana hameed
Drug transport  and drug targeting - rumana hameedDrug transport  and drug targeting - rumana hameed
Drug transport and drug targeting - rumana hameedRumana Hameed
 
Pharmacokinetic drug interaction
Pharmacokinetic drug interactionPharmacokinetic drug interaction
Pharmacokinetic drug interactionDr. Ramesh Bhandari
 
Pharmacokinetics and Pharmacodynamics
Pharmacokinetics and PharmacodynamicsPharmacokinetics and Pharmacodynamics
Pharmacokinetics and PharmacodynamicsBhaswat Chakraborty
 
Application of biopharmaceutics in pharmaceutical field.siam(pdf file)
Application of biopharmaceutics in pharmaceutical field.siam(pdf file)Application of biopharmaceutics in pharmaceutical field.siam(pdf file)
Application of biopharmaceutics in pharmaceutical field.siam(pdf file)Kamruzzaman Siam
 
Pharmacokinetic Drug Interactions
Pharmacokinetic Drug InteractionsPharmacokinetic Drug Interactions
Pharmacokinetic Drug InteractionsSiddiquaParveen
 
Applications of bio-pharmaceutics in new drug delivery
Applications of bio-pharmaceutics in new drug  deliveryApplications of bio-pharmaceutics in new drug  delivery
Applications of bio-pharmaceutics in new drug deliveryAkshata shettar
 

What's hot (20)

Phase clinicaltrial
Phase clinicaltrialPhase clinicaltrial
Phase clinicaltrial
 
Pharmacokinetics And Pharmacodynamic of Biotechnology Drugs - Trilok Shahare
Pharmacokinetics And Pharmacodynamic of Biotechnology Drugs - Trilok ShaharePharmacokinetics And Pharmacodynamic of Biotechnology Drugs - Trilok Shahare
Pharmacokinetics And Pharmacodynamic of Biotechnology Drugs - Trilok Shahare
 
Clinical pharmacology.Basics.
Clinical pharmacology.Basics.Clinical pharmacology.Basics.
Clinical pharmacology.Basics.
 
Importance of clinical pharmacokinetic studies
Importance of clinical pharmacokinetic studiesImportance of clinical pharmacokinetic studies
Importance of clinical pharmacokinetic studies
 
Repurposed drugs and safety monitoring
Repurposed drugs and safety monitoring Repurposed drugs and safety monitoring
Repurposed drugs and safety monitoring
 
Pharmacodynamics, ADRs & Preclinical drug testing
Pharmacodynamics, ADRs & Preclinical drug testingPharmacodynamics, ADRs & Preclinical drug testing
Pharmacodynamics, ADRs & Preclinical drug testing
 
Intelligent Drug Delivery System
Intelligent Drug Delivery SystemIntelligent Drug Delivery System
Intelligent Drug Delivery System
 
Pharmacokinetic Drug-Drug interactions
Pharmacokinetic Drug-Drug interactionsPharmacokinetic Drug-Drug interactions
Pharmacokinetic Drug-Drug interactions
 
Drug development
Drug developmentDrug development
Drug development
 
Allometric scaling
Allometric scaling Allometric scaling
Allometric scaling
 
Pharmacodynamics for BPH
Pharmacodynamics for BPHPharmacodynamics for BPH
Pharmacodynamics for BPH
 
Pharmacotherapeutics
PharmacotherapeuticsPharmacotherapeutics
Pharmacotherapeutics
 
Drug transport and drug targeting - rumana hameed
Drug transport  and drug targeting - rumana hameedDrug transport  and drug targeting - rumana hameed
Drug transport and drug targeting - rumana hameed
 
Biopharmaceutics complete notes
Biopharmaceutics complete notes Biopharmaceutics complete notes
Biopharmaceutics complete notes
 
Pharmacokinetic drug interaction
Pharmacokinetic drug interactionPharmacokinetic drug interaction
Pharmacokinetic drug interaction
 
Pharmacokinetics and Pharmacodynamics
Pharmacokinetics and PharmacodynamicsPharmacokinetics and Pharmacodynamics
Pharmacokinetics and Pharmacodynamics
 
Application of biopharmaceutics in pharmaceutical field.siam(pdf file)
Application of biopharmaceutics in pharmaceutical field.siam(pdf file)Application of biopharmaceutics in pharmaceutical field.siam(pdf file)
Application of biopharmaceutics in pharmaceutical field.siam(pdf file)
 
Pharmacokinetic Drug Interactions
Pharmacokinetic Drug InteractionsPharmacokinetic Drug Interactions
Pharmacokinetic Drug Interactions
 
PKPD seminar
PKPD seminarPKPD seminar
PKPD seminar
 
Applications of bio-pharmaceutics in new drug delivery
Applications of bio-pharmaceutics in new drug  deliveryApplications of bio-pharmaceutics in new drug  delivery
Applications of bio-pharmaceutics in new drug delivery
 

Viewers also liked (15)

臺中市政府社會局
臺中市政府社會局臺中市政府社會局
臺中市政府社會局
 
teletrabajo en la Gerencia
 teletrabajo en la Gerencia  teletrabajo en la Gerencia
teletrabajo en la Gerencia
 
Guía de estudio de la biblia
Guía de estudio de la bibliaGuía de estudio de la biblia
Guía de estudio de la biblia
 
感覺統合應用於自閉症兒童之評估與治療研習簡章
感覺統合應用於自閉症兒童之評估與治療研習簡章感覺統合應用於自閉症兒童之評估與治療研習簡章
感覺統合應用於自閉症兒童之評估與治療研習簡章
 
Nsn 789 1
Nsn 789 1Nsn 789 1
Nsn 789 1
 
Nsn 790 1
Nsn 790 1Nsn 790 1
Nsn 790 1
 
Presentación1
Presentación1Presentación1
Presentación1
 
Matematikte süreç var mıdır?
Matematikte süreç var mıdır? Matematikte süreç var mıdır?
Matematikte süreç var mıdır?
 
402516475577213cd38990
402516475577213cd38990402516475577213cd38990
402516475577213cd38990
 
105年0 3歲兒童發展早期介入活動彙整表
105年0 3歲兒童發展早期介入活動彙整表105年0 3歲兒童發展早期介入活動彙整表
105年0 3歲兒童發展早期介入活動彙整表
 
Instaemi service suite
Instaemi   service suiteInstaemi   service suite
Instaemi service suite
 
[簡章]20161029 銀髮智慧生活與健康照護s
[簡章]20161029 銀髮智慧生活與健康照護s[簡章]20161029 銀髮智慧生活與健康照護s
[簡章]20161029 銀髮智慧生活與健康照護s
 
El conde lucanor
El conde lucanorEl conde lucanor
El conde lucanor
 
TCA for Aminoglycoside Compounds
TCA for  Aminoglycoside CompoundsTCA for  Aminoglycoside Compounds
TCA for Aminoglycoside Compounds
 
Geografía joaquin zanetti y pierino actis caporade
Geografía joaquin zanetti y pierino actis caporadeGeografía joaquin zanetti y pierino actis caporade
Geografía joaquin zanetti y pierino actis caporade
 

Similar to AAC.00090-11v1

Bioequivalence
BioequivalenceBioequivalence
BioequivalenceGaurav Kr
 
Bioequivalence
BioequivalenceBioequivalence
BioequivalenceGaurav Kr
 
1. Unit I - new drug discovery and development.
1. Unit I - new drug discovery and development.1. Unit I - new drug discovery and development.
1. Unit I - new drug discovery and development.Audumbar Mali
 
Applying cheminformatics and bioinformatics approaches to neglected tropical ...
Applying cheminformatics and bioinformatics approaches to neglected tropical ...Applying cheminformatics and bioinformatics approaches to neglected tropical ...
Applying cheminformatics and bioinformatics approaches to neglected tropical ...Sean Ekins
 
Univ bacteriav PCR Primer
Univ bacteriav PCR PrimerUniv bacteriav PCR Primer
Univ bacteriav PCR PrimerZulkifli As
 
A COMPENDIUM ON IMMUNOBIOMARKERS IN PATHOLOGYpptx
A COMPENDIUM ON IMMUNOBIOMARKERS IN PATHOLOGYpptxA COMPENDIUM ON IMMUNOBIOMARKERS IN PATHOLOGYpptx
A COMPENDIUM ON IMMUNOBIOMARKERS IN PATHOLOGYpptxAYODEJI BLESSING AJILEYE
 
Combining Metabolite-Based Pharmacophores with Bayesian Machine Learning Mode...
Combining Metabolite-Based Pharmacophores with Bayesian Machine Learning Mode...Combining Metabolite-Based Pharmacophores with Bayesian Machine Learning Mode...
Combining Metabolite-Based Pharmacophores with Bayesian Machine Learning Mode...Sean Ekins
 
FORMULATION AND EVALUATION OF FLOATING- PULSATILE DRUG DELIVERY SYSTEM OF ACE...
FORMULATION AND EVALUATION OF FLOATING- PULSATILE DRUG DELIVERY SYSTEM OF ACE...FORMULATION AND EVALUATION OF FLOATING- PULSATILE DRUG DELIVERY SYSTEM OF ACE...
FORMULATION AND EVALUATION OF FLOATING- PULSATILE DRUG DELIVERY SYSTEM OF ACE...ANURAG GROUP OF INSTITUTIONS
 
Formulation and Evaluation of Moxifloxacin Loaded Alginate Chitosan Nanoparti...
Formulation and Evaluation of Moxifloxacin Loaded Alginate Chitosan Nanoparti...Formulation and Evaluation of Moxifloxacin Loaded Alginate Chitosan Nanoparti...
Formulation and Evaluation of Moxifloxacin Loaded Alginate Chitosan Nanoparti...pharmaindexing
 

Similar to AAC.00090-11v1 (20)

Bioequivalence
BioequivalenceBioequivalence
Bioequivalence
 
Bioequivalence
BioequivalenceBioequivalence
Bioequivalence
 
Plos
PlosPlos
Plos
 
824746
824746824746
824746
 
Artigo
ArtigoArtigo
Artigo
 
paper
paperpaper
paper
 
1. Unit I - new drug discovery and development.
1. Unit I - new drug discovery and development.1. Unit I - new drug discovery and development.
1. Unit I - new drug discovery and development.
 
Applying cheminformatics and bioinformatics approaches to neglected tropical ...
Applying cheminformatics and bioinformatics approaches to neglected tropical ...Applying cheminformatics and bioinformatics approaches to neglected tropical ...
Applying cheminformatics and bioinformatics approaches to neglected tropical ...
 
A0320106
A0320106A0320106
A0320106
 
Cytokines and Homeopathy
Cytokines and HomeopathyCytokines and Homeopathy
Cytokines and Homeopathy
 
2014 molecular mechanisms involved in the cytotoxicity induced
2014 molecular mechanisms involved in the cytotoxicity induced2014 molecular mechanisms involved in the cytotoxicity induced
2014 molecular mechanisms involved in the cytotoxicity induced
 
M0557376
M0557376M0557376
M0557376
 
Nanotech_Dubai_2013
Nanotech_Dubai_2013Nanotech_Dubai_2013
Nanotech_Dubai_2013
 
Univ bacteriav PCR Primer
Univ bacteriav PCR PrimerUniv bacteriav PCR Primer
Univ bacteriav PCR Primer
 
Progress Seminiar.ppt
Progress Seminiar.pptProgress Seminiar.ppt
Progress Seminiar.ppt
 
A COMPENDIUM ON IMMUNOBIOMARKERS IN PATHOLOGYpptx
A COMPENDIUM ON IMMUNOBIOMARKERS IN PATHOLOGYpptxA COMPENDIUM ON IMMUNOBIOMARKERS IN PATHOLOGYpptx
A COMPENDIUM ON IMMUNOBIOMARKERS IN PATHOLOGYpptx
 
Combining Metabolite-Based Pharmacophores with Bayesian Machine Learning Mode...
Combining Metabolite-Based Pharmacophores with Bayesian Machine Learning Mode...Combining Metabolite-Based Pharmacophores with Bayesian Machine Learning Mode...
Combining Metabolite-Based Pharmacophores with Bayesian Machine Learning Mode...
 
FORMULATION AND EVALUATION OF FLOATING- PULSATILE DRUG DELIVERY SYSTEM OF ACE...
FORMULATION AND EVALUATION OF FLOATING- PULSATILE DRUG DELIVERY SYSTEM OF ACE...FORMULATION AND EVALUATION OF FLOATING- PULSATILE DRUG DELIVERY SYSTEM OF ACE...
FORMULATION AND EVALUATION OF FLOATING- PULSATILE DRUG DELIVERY SYSTEM OF ACE...
 
Formulation and Evaluation of Moxifloxacin Loaded Alginate Chitosan Nanoparti...
Formulation and Evaluation of Moxifloxacin Loaded Alginate Chitosan Nanoparti...Formulation and Evaluation of Moxifloxacin Loaded Alginate Chitosan Nanoparti...
Formulation and Evaluation of Moxifloxacin Loaded Alginate Chitosan Nanoparti...
 
Drug delivery
Drug deliveryDrug delivery
Drug delivery
 

AAC.00090-11v1

  • 1. 1 Title: Evaluation of Pharmacokinetic/Pharmacodynamic Relationships of PD-0162819, a Biotin1 Carboxylase Inhibitor Representing a New Class of Antibacterial Compounds, Using In Vitro2 Infection Models3 4 5 Running Title: PK/PD Characterization: Novel Biotin Carboxylase Inhibitor6 7 8 Authors: Adam Ogden*, Michael Kuhn, Michael Dority, Susan Buist, Shawn Mehrens, Tong9 Zhu, Deqing Xiao, J. Richard Miller, and Debra Hanna10 11 Pfizer Global Research and Development12 Ann Arbor, MI, and Groton, CT13 14 15 *Corresponding author. Mailing address: Pfizer Global Research and Development, Eastern16 Point Rd., Groton, CT 06340. Phone: (860) 686-9197. Fax: (860) 441-4036. E-mail:17 adam.ogden@pfizer.com.18 Copyright © 2011, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved. Antimicrob. Agents Chemother. doi:10.1128/AAC.00090-11 AAC Accepts, published online ahead of print on 10 October 2011
  • 2. 2 Abstract19 The present study investigated the pharmacokinetic/pharmacodynamic (PK/PD)20 relationships of a prototype biotin carboxylase (BC) inhibitor, PD-0162819, against H.21 influenzae 3113 (HI-3113) in static concentration time-kill (SCTK) and one-compartment22 chemostat in vitro infection models. HI-3113 was exposed to PD-0162819 concentrations of23 0.5-16x the minimal inhibitory concentration (MIC; 0.125 µg/mL) and area-under-the-curve24 (AUC)/MIC ratio of 1-1100 in SCTK and chemostat experiments, respectively. Serial samples25 were collected over 24 h. For efficacy driver analysis, a sigmoid Emax model was fit to the26 relationship between bacterial density changes over 24 h and corresponding PK/PD indices. A27 semi-mechanistic PK/PD model describing the time-course of bacterial growth and death was28 developed. AUC/MIC best-explained efficacy (r2 =0.95) compared to peak drug concentration29 (Cmax)/MIC (r2 =0.76) and time above MIC (T>MIC; r2 =0.88). Static effects and 99.9% killing30 were achieved at AUC/MIC values of 500 and 600, respectively. For time-course analysis, the31 net bacterial growth rate constant, maximum bacteria density, and maximum kill rate constant32 were similar in SCTK and chemostat studies, but PD-0162819 was more potent in SCTK as33 compared to the chemostat (EC50=0.046 vs. 0.34 µg/mL). In conclusion, basic PK/PD34 relationships for PD-0162819 were established using in vitro dynamic systems. Although the35 bacterial growth parameters and maximum drug effect were similar in SCTK and chemostat, PD-36 0162819 appeared to be more potent in SCTK, illustrating the importance of understanding37 differences of preclinical models. Additional studies are needed to determine the in vivo38 relevance of these results.39
  • 3. 3 Introduction40 Steadily increasing bacterial resistance to existing antibiotics continues to be a major41 public health concern (3, 8). Because most new antibacterial agents represent chemical42 modification of existing chemical classes of antibacterial agents (5), it is suspected that the43 limited options of chemically-distinct antibiotics has led to extensive drug resistance among44 bacterial pathogens. Therefore, it is of utmost importance to identify novel, safe, and effective45 antibacterial agents which work through unique antibacterial biologic mechanisms. The46 discovery of a new chemical class of antibacterial compounds, the pyridopyrimidines, targeting47 bacterial biotin carboxylase (BC) was recently reported (14, 15) and offers the potential that this48 novel chemical class, targeting a unique antibacterial mechanism, can be developed into drugs49 effective against multi-drug resistant bacteria.50 As compared to development of drugs from an existing chemical class, the discovery of a51 novel class of compounds presents extra challenges (1, 5). The translation of52 pharmacokinetic/pharmacodynamic (PK/PD) relationships between animal infection models and53 human patients has been well-established for several existing chemical classes across a variety of54 indications (1), but for novel chemical classes, PK/PD relationships, and the translation of these55 relationships between in vitro systems, animals, and humans, are not known. Moreover, the56 physicochemical and pharmacokinetic properties of compounds at early stages of the drug57 discovery process are often not optimized for extensive concentration-response testing in animal58 models (21). Thus, as an alternative, in vitro infection models offer a rapid and resource-sparing59 method to determine PK/PD relationships. Building and applying mathematical PK/PD models60 that quantitatively describe the time-course of bacterial replication/death and drug effects enables61 the construction of a more efficient drug discovery process. Furthermore, these quantitative62
  • 4. 4 PK/PD relationships derived from in vitro data can inform future in vivo testing as to optimal63 dose selection and dosing intervals and thereby reduce resources necessary to perform adequate64 in vivo experiments. They can also provide the framework for understanding knowledge gaps65 and for determining optimal drug properties (e.g., pharmacokinetics) required for a successful66 drug candidate (6, 7, 9).67 The present study investigated the PK/PD relationships of a prototype BC inhibitor, PD-68 0162819, against H. influenzae 3113 (HI-3113) in static concentration time-kill (SCTK) and one-69 compartment chemostat in vitro infection models. The objectives of this study were to a)70 establish a basic understanding of concentration-response relationships in vitro for a prototype71 BC inhibitor and b) use in vitro and mathematical modeling tools to guide the drug discovery72 program by understanding the translation among in vitro infection models.73 74 Materials and Methods75 Compound, microorganism, and susceptibility studies. PD-0162819 was synthesized76 by Pfizer chemists (14). Broth micro-dilution susceptibility testing was performed using a77 BioMek FX robotic workstation (Beckman-Coulter, Fullerton, CA). A β-lactamase producing78 clinical isolate of H. influenzae, HI-3113, was tested using Haemophilus Test Medium (HTM;79 PMLMicrobiologicals, Wilsonville, OR) and incubated at 35°C in ambient atmosphere as80 described by the Clinical and Laboratory Standards Institute (CLSI) (17).81 SCTK experiments. In vitro SCTK testing was performed following CLSI methodology82 (17). Specifically, testing was carried out in 10 mL of HTM and incubated at 35°C with 5% CO283 atmosphere. PD-0162819 concentrations ranged from 0.06-2 µg/mL (0.5-16× MIC; MIC= 0.12584 µg/mL) and were determined by LC-MS/MS to remain constant during the course of the85
  • 5. 5 experiment. Serial media samples (100 µL/sample) were collected at t=0, 2, 4, 6, and 24 h.86 Post-exposure MIC testing was performed, and changes in MICs were not observed.87 One-compartment chemostat infection model. In vitro dynamic concentration studies88 were performed using a one-compartment chemostat system as previously described (12, 23).89 The chemostat system consisted of a 250 mL glass chamber with ports for the addition and90 removal of test media via polyethylene tubes connected to peristaltic pumps, injection of drug91 solution, and removal of media samples. Single-dose and dose fractionation experiments were92 performed. Prior to each experiment, colonies from an overnight growth of HI-3113 on93 chocolate agar were added to HTM as necessary to obtain a suspension of 108 CFU/mL. To94 produce a starting inoculum of ~106 CFU/mL, 2.5 mL of this suspension were added to each95 flask. A drug stock solution of PD-0162819 was prepared at the start of each experiment, and96 this solution was diluted with saline such that a dosing volume of 0.5 mL was added to each97 flask via bolus dosing. To increase the solubility of PD-0162819, 3% SBE-β-cyclodextrin was98 added to the dosing solution. Because PD-0162819 is projected to have a short half-life in vivo99 (data not shown), pump rates for all chemostat experiments were set to achieve a drug100 elimination half-life of four hours. A dose escalation study was performed to determine the101 efficacious exposure range using AUC/MIC of 1 to 1100 (MIC=0.125 µg/mL). To determine the102 efficacy driver, a dose fractionation study was performed using exposures corresponding to the103 ED20, ED50, and ED80 from the dose escalation study divided into 1×/day (dosed at t=0 h),104 2×/day (t=0 and 12 h), or 3×/day (t=0, 6, and 12 h) dosing regimens. Media samples were105 collected at 0, 0.5, 1, 2, 4, 6, 12, and 24 h after dosing for analysis of drug concentrations and106 determination of bacteria densities. Post-exposure MIC testing was performed, and changes in107 MICs were not observed.108
  • 6. 6 Determination of bacteria density. Media samples were washed with saline to remove109 any drug carry-over. Samples were then reconstituted, diluted 10-fold, and 100 µL of diluted110 and undiluted samples were plated onto chocolate agar. After 24 h of incubation, colony111 forming units (CFU/mL) were counted and corrected for dilution. Because an undiluted 100 µL112 sample was used for determining bacteria density in samples with low bacteria counts, the limit113 of detection was 10 CFU/mL.114 Drug concentration analysis. Media samples were stored at −20°C until analysis. A115 freeze-thaw study conducted during method development indicated that PD-0162819 was stable116 at −20°C for >4 weeks and <5% degradation occurred after repeated freeze-thaw cycles. The117 samples were thawed at room temperature, spiked with 200 µL of internal standard (200 ng/mL118 of PF-01135527 in acetonitrile), and vortexed for 1 min. Following centrifugation at 5000 rpm119 for 5 min, 3 µL of the supernatants were injected into a validated LC-MS/MS system. A Varian120 Polaris C-18-A column (50 × 2.0 mm) was employed and run under ambient reversed-phase121 conditions. The mobile phases were A) 100% water with 0.1% formic acid and B) 100%122 acetonitrile with 0.1% formic acid. The gradient was programmed with a flow rate of123 250 µL/min starting at 10% mobile phase B to reach 70% mobile phase B in 1.5 min and 90%124 mobile phase B in an additional 1 min. At the end of the program, the analytical column was re-125 equilibrated with 10% mobile phase B for 1 min. The total cycle time for the assay was 3.6126 min/sample. Molecular ion precursor-product transitions of 393.7→ 298.1 and 382→ 271.1127 were used for PD-0162819 and internal standard, respectively, in multiple-reaction monitoring128 (MRM) mode. Data acquisition, integration of peak areas, regression analysis, and sample129 quantification were performed using the standard quantitative analysis package of Analyst 1.4130 (Applied Biosystems, Foster City, CA). A 12-point standard curve consisting of PD-0162819131
  • 7. 7 concentrations ranging from 5 – 10,000 ng/mL was used to quantify PD-0162819 concentrations132 in samples. Where appropriate, media samples were diluted for analysis. The lower limit of133 quantitation (LLOQ) was 5 ng/mL. Using standards of known concentration, accuracy was134 determined to be ±20%, and intra-day and inter-day precision was ±20%.135 Endpoint efficacy driver analysis. Data collected using the one-compartment136 chemostat system was used to determine the efficacy driver. The change of bacteria density over137 24 h (∆log CFU/mL) was calculated and plotted against corresponding endpoint PK/PD138 parameters (AUC/MIC, Cmax/MIC, and T>MIC). Observed drug concentration data was used to139 calculate the pharmacokinetic parameters. A sigmoid Emax model was fit to the relationship140 between ∆log CFU/mL and corresponding PK/PD indices. This model was used to determine141 the endpoint PK/PD index that best-explained efficacy and the magnitude of each parameter to142 achieve bacterial killing ranging from 0-log to ≥3-log killing.143 Time-course PK/PD data analysis. SCTK and chemostat PK/PD data were modeled144 via a two-step process using NONMEM version V (GloboMax LLC, Ellicott City, MD). Based145 on a study to assess the chemical and metabolic stability of PD-0162819 in HTM, drug146 concentrations were determined by LC-MS/MS to be constant throughout the course of SCTK147 studies. Less than 5% degradation occurred over 24 h at 35°C. A population pharmacokinetic148 one-compartment open model with first-order elimination (kelim) and proportional error was used149 to fit the drug-concentration-time profiles. The population pharmacokinetic model incorporated150 inter-individual (i.e., inter-flask) variability of clearance (CL) and volume of distribution (Vd) to151 describe the observed drug concentration data in each flask. The pharmacokinetic parameters152 were then fixed in the PK/PD model, and the bacteria density time-course profiles were modeled153 using a modified net effect mathematical model that is outlined in Figure 1. Similar154
  • 8. 8 mathematical models have been previously described (e.g., 2, 10, 13, 16, 18, 20, 22). In this155 model, changes in drug concentrations over time were described using Equation 1:156 melim m Ck C ×−= dt d (1)157 where Cm is the PD-0162819 concentration in HTM. kelim is the first-order drug elimination rate158 constant, which is determined by the pump rate of the chemostat system. kelim can also be159 represented as CL/Vd. kelim was 0 h-1 in SCTK because drug was not flushed out of the system.160 In the chemostat, kelim was approximately 0.17 h-1 to achieve a drug half-life of approximately161 four hours. For chemostat data, kelim was estimated using the previously-described population162 pharmacokinetic model.163 The bacterial population was hypothesized to be composed of three bacterial populations164 of non-replicating, replicating, and damaged bacterial cells. To model the time-course of165 bacterial density, Equations 2-5 were applied:166         ++ + × −= flushglagγ 50 γ m γ mkill 1 1 kk ECC Ck CFU CFU dt d (2)167 ( )         − + × − − ×+= flushγ 50 γ m γ mkill max totalmax rep2glag1 2 k ECC Ck N CFUN kCFUkCFU CFU dt d (3)168 ( ) ( )flushdlag3γ 50 γ m γ mkill 21 3 kkCFU ECC Ck CFUCFU CFU +−        + × += dt d (4)169 321total CFUCFUCFUCFU ++= (5)170 where, as illustrated in Figure 1, CFU1, CFU2, and CFU3 represent the density of non-replicating,171 replicating, and damaged bacterial cells, respectively. CFUtotal is the sum of all bacterial cells in172 the system. For the drug effect, kkill is the rate constant for maximal bacterial killing, EC50 is the173 drug concentration at which 50% of the maximal killing occurs, and γ is the Hill coefficient174
  • 9. 9 corresponding to the degree of sigmoidicity of the concentration-response curve. For the175 replication rate, krep is the net growth rate constant, and Nmax is the maximum number of bacteria176 in the in vitro system. As CFUtotal approaches Nmax, the total bacterial population reaches a177 stationary phase and was observed as a plateau of the bacteria density-time profile. Where178 appropriate, a bacterial growth lag rate constant (kglag) and bacterial death lag rate constant (kdlag)179 were incorporated into the model to describe lags in bacterial growth and death. For the dynamic180 chemostat data, kflush is the rate constant associated with flushing HTM, bacteria, and drug out of181 the in vitro system. Because HTM, bacteria, and drug were flushed out of the system at a rate182 determined by the pump rate, kflush has the same magnitude as kelim. For modeling SCTK data,183 kflush was excluded from the model because HTM, bacteria, and drug were not flushed out of the184 system. Best-fit parameters of the PK/PD model were obtained using first-order conditional185 estimation, and the goodness of fit of the models to the experimental data was assessed by visual186 inspection of observed versus predicted plots, coefficient of determination, evaluation of187 weighted residuals (versus time and concentration), and comparison of objective function values188 for nested models.189 190 Results191 SCTK of PD-0162819 against HI-3113. PD-0162819 demonstrated concentration-192 dependent killing achieving a 3-log kill after 6 h. PD-0162819 was bactericidal at concentrations193 of ≥2× the MIC. The PK/PD model (Figure 1) adequately described the observed bacteria194 density data (r2 =0.91). Observed data and model-predicted fits to the data are shown in Figure195 2A. The maximum bacterial net growth rate constant in this system was 2.04 ± 0.14 h-1 , and the196 death lag rate constant was 1.91 ± 0.20 h-1 . A growth lag rate constant was not estimated due to197
  • 10. 10 the absence of data between 0 and 2 h. The maximum number of bacteria in the system was198 estimated to be 108.80 ± 0.15 CFU/mL. The maximum drug effect (kkill) was 2.53 ± 0.14 h-1 , and199 the EC50 of PD-0162819 against HI-3113 was estimated to be 0.0456 ± 0.0043 µg/mL. Model200 parameter estimates and 90% confidence intervals are shown in Table 1.201 Dynamic concentration time-kill study in chemostat. The PK/PD model outlined in202 Figure 1 adequately described the observed time-course of bacteria density data (r2 =0.98).203 Observed data and model-predicted fits to the pharmacodynamic data are shown in Figure 2B.204 The maximum bacterial net growth rate constant in this system was 1.80 ± 0.10 h-1 , and the death205 lag rate constant was 3.06 ± 1.10 h-1 . A growth lag was observed over the first hour of the206 experiment, and the growth lag rate constant was estimated to be 0.171 ± 0.065 h-1 . The207 maximum number of bacteria in the system was estimated to be 109.28 ± 0.04 CFU/mL. The208 maximum drug effect (kkill) was 2.02 ± 0.11 h-1 , and the EC50 of PD-0162819 against HI-3113209 was estimated to be 0.340 ± 0.041 µg/mL. Model parameter estimates and 90% confidence210 intervals are shown in Table 1.211 Efficacy driver analysis in chemostat. The one-compartment chemostat system was212 used to determine the efficacy driver of PD-0162819 against HI-3113. Although all three PK/PD213 indices exhibited relationships with efficacy (Figure 3), AUC/MIC was the best predictor of214 antibacterial efficacy (r2 =0.95) compared to Cmax/MIC (r2 =0.76) and T>MIC (r2 =0.88). A static215 effect over 24 h was achieved at an AUC/MIC value of 500. An AUC/MIC magnitude of 600216 was required to achieve 3-log killing (i.e., 99.9% killing) against HI-3113.217 218 219 220
  • 11. 11 Discussion221 The data and modeling presented in this report represent initial studies to characterize the222 PK/PD relationships of PD-0162819, a prototype BC inhibitor. Using two different in vitro223 infection systems and mathematical modeling tools, a basic understanding of endpoint efficacy224 driver and time-course concentration-response relationships were established for this novel225 compound which works through a unique mechanism of action. The results of this study indicate226 there is a significant need for understanding the quantitative translation of this data from in vitro227 systems to in vivo systems and, ultimately, to humans.228 In this study, both endpoint efficacy drivers and time-course PK/PD relationships were229 evaluated. Typically, endpoint efficacy driver approaches have been utilized for understanding230 antibacterial PK/PD (4). However, endpoint data often minimizes the importance of the time-231 course of drug effects. Endpoint efficacy driver studies typically assess the change in bacterial232 densities between time=0 h and time=24 h, which essentially ignores the time-course of drug233 effects over the 24 h period. By understanding the drug effects of the entire time-course of the234 experiment, a more thorough understanding of PK/PD relationships can be achieved. Even so,235 endpoint efficacy driver analyses are traditionally used for developing an understanding of236 PK/PD relationships for antibacterial agents. Thus, this study was designed to enable analysis of237 both the endpoint efficacy driver and time-course concentration-response relationships.238 Efficacy driver analysis results indicate that all three PK/PD indices exhibit some trend239 with efficacy. However, based on visual inspection of the plots and the coefficient of240 determination, AUC/MIC appears to be a better predictor of efficacy compared to Cmax/MIC and241 T>MIC. Although T>MIC exhibits a strong coefficient of determination, it is not likely that242
  • 12. 12 T>MIC is a good predictor of efficacy due to the observed variation of approximately 5-logs of243 bacterial killing among the different dosing regimens that achieved 100% T>MIC.244 The mathematical model presented in Figure 1 is the most parsimonious model that was245 fit to the observed data. It is similar to previously published models (e.g., 2, 10, 13, 16, 18, 20,246 22), but the basic structure of the model is most similar to the net effect model (2). Comparisons247 of the different mathematical models showed that potency estimates produced by the various248 mathematical models were similar (analysis not shown). An additional compartment was added249 to the basic net effect model to more accurately describe the apparent growth lag phase over the250 first hour of the chemostat experiments but was not incorporated for SCTK data due to lack of251 data in this early phase. This growth lag phase is hypothesized to be the result of non-replicating252 cells during the initial stages of the experiments. After this early phase, the cells are assumed to253 begin replicating. In addition, an extra compartment was added to describe the apparent death254 lag, which is hypothesized to be caused by damaged cells, that was more obvious in SCTK255 experiments. Because bacterial death was delayed relative to drug concentrations, application of256 a transit compartment model was useful for describing this observation (19). This model257 assumes that bacterial cells are damaged by drug, slow their proliferation rates, and eventually258 die. In addition, because bacteria were constantly being flushed out the chemostat system, a259 parameter (kflush) was added to the model to account for this. Without this parameter, the260 potency estimate would have been overestimated. This parameter was not needed when261 modeling the SCTK data because bacteria were not flushed from the SCTK system.262 Even though HI-3113 replication rates were similar among the two infection systems, the263 potency of PD-0162819 was ~7× more potent in SCTK than in the chemostat system.264 Differences in experimental conditions between the two study designs can account for this265
  • 13. 13 finding. In the chemostat system, nutrients are replenished via fresh media while drug, wastes,266 and old media are being removed from the system. This differs from the SCTK design where267 less optimal growth conditions exist (i.e., nutrients are fixed and used up during the course of the268 experiment). Because the chemostat system provides more optimal bacterial growth conditions269 compared to SCTK, it is not surprising that PD-0162819 appears less potent in the chemostat.270 Rebound of bacteria counts over the course of the experiment was not observed in SCTK271 studies. Although collecting more extensive samples between 6 and 24 h would allow for a more272 definitive determination of potential resistance development in SCTK studies, the lack of273 resistant subpopulations was supported by the low frequency of resistance (~1 resistant cell in274 >108 cells) (14), which is well-above the starting inoculum of 106 CFU/mL used in these studies.275 Thus, the observed experimental data did not warrant the inclusion of additional276 “subpopulations” of resistant bacteria into the mathematical model, which have been277 incorporated into previously published models (e.g., 2, 13, 20, 22). In addition, the278 pyridopyrimidine inhibitors are the only antibacterial agents known to target BC, and, therefore,279 no pre-existing clinical resistance is expected. Although rebound was observed in the chemostat280 studies, this rebound was likely caused by reductions in drug concentrations over the course of281 the experiments and not by the emergence of resistant bacterial subpopulations.282 Although this study focused on in vitro PK/PD relationships, simply understanding in283 vitro PK/PD of a compound is not sufficient. To be useful, the in vivo relevance, and ultimately284 the clinical relevance, of in vitro relationships need to be determined. In addition, only one285 bacterial isolate was tested in this study. The AUC/MIC exposure targets presented in this report286 will likely be modified when additional clinical isolates are taken into consideration (11). At this287 point, the results of this study have been used to guide the discovery project team in the process288
  • 14. 14 of synthesizing a more optimal compound. Attempts to optimize the molecule have included289 improving the physicochemical properties, potency, pharmacokinetics, and other parameters.290 In conclusion, mathematical modeling of data generated in in vitro infection systems was291 successfully employed to evaluate the PK/PD relationships of a novel BC inhibitor early in the292 drug discovery program. Although HI-3113 replication rates were similar among the two293 systems, the potency of PD-0162819 was ~7× more potent in SCTK than in the chemostat294 system, illustrating the importance of understanding differences of preclinical models when295 making decisions about drug exposures required for efficacy. Thus, the results of this study296 indicate that PK/PD relationships need to be interpreted with respect to the system in which the297 data is generated. Different in vitro and in vivo systems are likely to produce different results,298 which underlies the importance of understanding the quantitative translation of these systems to299 humans. The relevance of this modeling approach can be enhanced by translating this approach300 to in vivo systems. Thus, additional studies with additional strains of bacteria are required to301 determine the in vivo relevance of these results.302 303
  • 15. 15 References304 1. Ambrose, P.G., S.M. Bhavnani, C.M. Rubino, A. Louie, T. Gumbo, A. Forrest, and305 G.L. Drusano. 2007. Pharmacokinetics-pharmacodynamics of antimicrobial therapy:306 It's not just for mice anymore. Clin. Infect. Dis. 44:79-86.307 2. Campion, J.J., P.J. McNamara, and M.E. Evans. 2005. Pharmacodynamic modeling308 of ciprofloxacin resistance in Staphylococcus aureus. Antimicrob. Agents Chemother.309 49:209-219.310 3. Charles, P.G.P. and M.L. Grayson. 2004. The dearth of new antibiotic development:311 Why we should be worried and what we can do about it. Med. J. of Australia. 181:549-312 553.313 4. Craig, W.A. 1998. Pharmacokinetic/pharmacodynamic parameters: Rationale for314 antibacterial dosing of mice and men. Clin. Infect. Dis. 26:1-10.315 5. Fernandes, P. 2006. Antibacterial discovery and development – The failure of success?316 Nature Biotechnology 24:1497-1503.317 6. Gabrielsson, J., A.R. Green, and P.H. Van der Graaf. 2010. Optimising in vivo318 pharmacology studies – Practical PKPD considerations. J. Pharmacol. Toxicol. Methods.319 61:146-56.320 7. Gabrielsson J., and A.R. Green. 2009. Quantitative pharmacology or321 pharmacokinetic- pharmacodynamic integration should be a vital component in322 integrative pharmacology. J. Pharmacol. Exp. Ther. 331:767-74.323 8. Levy, S.B., and B. Marshall. 2004. Antibacterial resistance worldwide: Causes,324 challenges and responses Nature Medicine. 10:S122 - S129.325
  • 16. 16 9. Liu, K.K., P. Cornelius, T.A. Patterson, Y. Zeng, S. Santucci, E. Tomlinson, C.326 Gibbons, T.S. Maurer, R. Marala, J. Brown, J.X. Kong, E. Lee, W. Werner, Z.327 Wenzel, and C. Vage. 2010. Design and synthesis of orally-active and selective328 azaindane 5HT2c agonist for the treatment of obesity. Bioorg. Med. Chem. Lett. 20:266-329 71.330 10. Liu, P., K.H. Rand, B. Obermann, and H. Derendorf. 2005. Pharmacokinetic-331 pharmacodynamic modeling of antibacterial activity of cefpodoxime and cefixime in in332 vitro kinetic models. Int. J. Antimicrob. Agents 25:120-129.333 11. MacGowan, A.P., R. Reynolds, A.R. Noel, and K.E. Bowker. 2009. Bacterial strain-334 to-strain variation in pharmacodynamic index magnitude, a hitherto unconsidered factor335 in establishing antibiotic clinical breakpoints. Antimicrob. Agents Chemother. 53:5181-336 5184.337 12. McGrath, B., K.C. Lamp, and M.J. Rybak. 1993. Pharmacodynamic effects of338 extended dosing intervals of imipenem alone and in combination with amikacin against339 Pseudomonas aeruginosa in an in vitro model. Antimicrob. Agents Chemother. 37:1931-340 1937.341 13. Meagher, A.K., A. Forrest, A. Dalhoff, H. Stass, and J.J. Schentag. 2004. Novel342 pharmacokinetic-pharmacodynamic model for prediction of outcomes with an extended-343 release formulation of ciprofloxacin. Antimicrob. Agents Chemother. 48:2061-2068.344 14. Miller, J.R., S. Dunham, I. Mochalkin, C. Banotai, M. Bowman, S. Buist, B. Dunkle,345 D. Hanna, J.H. Harwood, M.D. Huband, A. Karnovsky, M. Kuhn, C. Limberakis,346 J.Y. Liu, S. Mehrens, T.W. Mueller, L. Narasimhan, A. Ogden, J. Ohren, V.J.V.N.347 Prasad, J.A. Shelly, L. Skerlos, M. Sulavik, H.V. Thomas, S. VanderRoest, L. Wang,348
  • 17. 17 Z. Wang, A. Whitton, T. Zhu, and C.K. Stover. 2009. A class of selective349 antibacterials derived from a protein kinase inhibitor pharmacophore. Proc.Nat. Acad.350 Sci. USA 106:1737-42.351 15. Mochalkin, I., J.R. Miller, L. Narasimhan, V. Thanabal, P. Erdman, P.B. Cox,352 V.J.V.N. Prasad, S. Lightle, M.D. Huband, and C.K. Stover. 2009. Discovery of353 antibacterial biotin carboxylase inhibitors by virtual screening and fragment-based354 approaches. ACS Chem. Bio. 4:473-83.355 16. Mouton, J.W., and A.A. Vinks. 2005. Pharmacokinetic/pharmacodynamic modeling of356 antibacterials in vitro and in vivo using bacterial growth and kill kinetics. Clin.357 Pharmacokinet. 44:201-210.358 17. National Committee for Clinical Laboratory Standards. 1999. Methods for359 determining bactericidal activity of antimicrobial agents. Approved Guideline M26-A.360 National Committee for Clinical Laboratory Standards, Wayne, PA.361 18. Schaper, K.J., S. Schubert, and A. Dalhoff. 2005. Kinetics and quantification of362 antibacterial effects of beta-lactams, macrolides, and quinolones against gram-positive363 and gram-negative RTI pathogens. Infection. 33(Suppl 2): 3-14.364 19. Simeoni, M., P. Magni, C. Cammia, G. De Nicolao, V. Croci, E. Pesenti, M.365 Germani, I. Poggesi, and M. Rocchetti. 2004. Predictive pharmacokinetic-366 pharmacodynamic modeling of tumor growth kinetics in xenograft models after367 administration of anticancer agents. Cancer Res. 64:1094-1101.368 20. Tam, V.H., A.N. Schilling, K. Poole, and M. Nikolaou. 2007. Mathematical modeling369 response of Pseudomonas aeruginosa to meropenem. J. Antimicrob. Chemother.370 60:1302-1309.371
  • 18. 18 21. van de Waterbeemd, H. 2009. Improving compound quality through in vitro and in372 silico physicochemical profiling. Chem. Biodivers. 6:1760-6.373 22. Yano, Y., T. Oguma, H. Nagata, and S. Sasaki. 1998. Application of logistic growth374 model to pharmacodynamic analysis of in vitro bactericidal kinetics. J. Pharm. Sci.375 87:1177-1183.376 23. Zinner, S. H., M. Dudley, and J. Blaser. 1986. In vitro models for the study of377 combination antibiotic therapy in neutropenic patients. Am. J. Med. 80:156-160.378
  • 19. 19 379 380 Figure 1. Schematic representation of the mathematical PK/PD model. The total bacteria381 density (CFUtotal) is the combination of non-replicating (CFU1), replicating (CFU2), and damaged382 cells (CFU3). Bacterial replication eventually results in bacterial counts reaching a stationary383 phase, which is determined by the total amount of bacteria in the system (Nmax). The replication384 rate (krep) is modified as bacterial counts increase [krep×((Nmax−CFUtotal)/Nmax)] and, ultimately,385 achieve a stationary phase. The maximum drug effect (kkill), drug concentration associated with386 50% of the maximum effect (EC50), drug concentration in the media (Cm), and Hill coefficient (γ)387 determine the magnitude of the drug effect [kkill×Cm γ /( Cm γ + EC50 γ )]. Rate constants associated388 with a growth lag (kglag), death lag (kdlag), and flushing of bacteria out of the in vitro system389 (kflush) are also included in the model. See the Materials and Methods section for equations and a390 more complete description of the model.391 392 kelim CmPK: krep kglag kdlag Drug Effect Drug Effect kflush kflush kflush PD: CFU1 CFU2 CFU3
  • 20. 20 393 0 2 4 6 8 10 0 6 12 18 24 BacteriaDensity(logCFU/mL) Time (h) 0 0.5x 1x 2x 4x 8x 16x x MIC 394 0 2 4 6 8 10 0 6 12 18 24 BacteriaDensity(logCFU/mL) Time (h) 0 5 8 11 26 46 130 260 530 1100 AUC/MIC 395 Figure 2. PK/PD modeling of PD-0162819 against H. influenzae 3113 in A) SCTK and B)396 chemostat. Symbols represent observed data. Lines represent model predictions.397 398 A B
  • 21. 21 399 -6 -4 -2 0 2 4 6 0 200 400 600 800 1000 1200 BacteriaDensityChangeOver24h AUC/MIC r2 = 0.95 -6 -4 -2 0 2 4 6 0 50 100 150 200 Cmax/MIC r2 = 0.76 -6 -4 -2 0 2 4 6 0 20 40 60 80 100 T>MIC (%) r2 = 0.88 400 401 Figure 3. Efficacy driver analysis of PD-0162819 against H. influenzae 3113 in the chemostat system. Closed circles represent402 observed data, and solid lines represent model-predicted values.403
  • 22. 22 Table 1. PK/PD modeling results of the time-course effects of PD-0162819 against H.404 influenzae 3113 in SCTK and chemostat systems.405 406 Parameter Units Static Concentration Time Kill Dynamic Concentration Time Kill krep h-1 2.04 ± 0.14a (1.8 – 2.3)b 1.80 ± 0.10 (1.6 – 2.0) Nmax CFU/mL 108.80 ± 0.15 (108.6 – 109.0 ) 109.28± 0.04 (109.2 – 109.3 ) kkill h-1 2.53 ± 0.14 (2.3 – 2.8) 2.02 ± 0.11 (1.8 – 2.2) EC50 µg/mL 0.0456 ± 0.0043 (0.039 – 0.053) 0.340 ± 0.041 (0.27 – 0.41) γ  1.80 ± 0.15 (1.6 – 2.1) 1.88 ± 0.42 (1.2 – 2.6) kglag h-1  0.171 ± 0.065 (0.07 – 0.28) kdlag h-1 1.91 ± 0.20 (1.6 – 2.2) 3.06 ± 1.10 (1.2 – 4.9) r2  0.91 0.98 a NONMEM estimate ± standard error.407 b 90% confidence intervals in parentheses.408