Abstract
In 1900, Ernst Overton found that the entry of anilin dyes through the cell membranes of living cells depended on the lipophilicity of the dyes. The brain is surrounded by barriers consisting of lipid layers that possess several inward and outward active transport systems. In the absence of meningeal inflammation, the cerebrospinal fluid (CSF) penetration of anti-infectives in humans estimated by the ratio of the area under the concentration-time curve (AUC) in CSF (AUCCSF) to that in serum (AUCCSF/AUCS) correlated positively with the lipid-water partition coefficient at pH 7.0 (log D) (Spearman's rank correlation coefficient rS = 0.40; P = 0.01) and negatively with the molecular mass (MM) (rS = −0.33; P = 0.04). The ratio of AUCCSF to the AUC of the fraction in serum that was not bound (AUCCSF/AUCS,free) strongly correlated with log D (rS = 0.67; P < 0.0001). In the presence of meningeal inflammation, AUCCSF/AUCS also correlated positively with log D (rS = 0.46; P = 0.002) and negatively with the MM (rS = −0.37; P = 0.01). The correlation of AUCCSF/AUCS,free with log D (rS = 0.66; P < 0.0001) was as strong as in the absence of meningeal inflammation. Despite these clear correlations, Overton's rule was able to explain only part of the differences in CSF penetration of the individual compounds. The site of CSF withdrawal (lumbar versus ventricular CSF), age of the patients, underlying diseases, active transport, and alterations in the pharmacokinetics by comedications also appeared to strongly influence the CSF penetration of the drugs studied.
INTRODUCTION
The entry of many endogenous and exogenous compounds into the central nervous system (CNS) is restricted by the blood-brain and blood-cerebrospinal fluid (CSF) barriers, which ensure proper neuronal function. Paul Ehrlich was the first to demonstrate the existence of these barriers by injecting dyes into the circulation systems of laboratory animals at the end of the 19th century: he noticed that all organs with the exception of the brain were stained (26). Twenty-five years later, Ehrlich's student Edwin Goldmann injected trypan blue systemically or into the CNS. After systemic injection, trypan blue stained the choroid plexus and the dura mater, but not the brain and CSF. In contrast, after direct injection of trypan blue into the CSF, the brain and spinal cord were stained (32, 33). These and subsequent morphological studies gave rise to the idea that the CNS was surrounded by a lipid layer comparable to a second cell membrane.
In 1900, Ernst Overton discovered that the entry of dyes into plant cells depends on the lipophilicity of the dyes (100). Later, this rule was refined by the acknowledgment of the importance of molecular size for the penetration through lipid membranes and was applied to the penetration of drugs through the blood-brain or blood-CSF barrier (for example, see references 12, 50, 96, and 99). Recently, the validity of Overton's rule has been questioned by membrane physiologists. In particular, violations of Overton's rule for small molecules, including carboxylic acids and gases were reported (for example, see reference 35). With some exceptions, however, Overton's rule is still considered a valid explanation of how molecules pass through lipid membranes (82).
The pharmacokinetic interactions between blood and the different compartments of the CNS are complex (Fig. 1). Unlike other deep compartments, the steady-state concentrations of many drugs, including most anti-infectives, do not reach CSF concentrations equal to the respective serum levels or concentrations of unbound drug in serum. For some drugs, this is caused by strong outward transport systems. However, this phenomenon also occurs in the absence of active transport, where it is caused by CSF bulk flow (approximately 20 to 30 ml/h under physiological conditions). For this reason, in the absence of active transport, the intercompartmental outward clearance CLout is greater than the inward clearance CLin by approximately 20 to 30 ml/h (86, 95). Active outward transport and CSF bulk flow often represent obstacles for the achievement of intracranial concentrations of anti-infectives effective for the treatment of CNS infections.
Fig 1.
Pharmacokinetic relationships between blood, brain, and cerebrospinal fluid (CSF) illustrated on a normal cranial computer tomogram (CCT). (Left) The different spaces and their average volumes are listed. The volumes of these spaces vary greatly and depend on age, skull volume, genetic factors, and underlying diseases. Tight diffusional barriers containing active inward and outward transport systems separate blood and brain tissue (blood-brain barrier) as well as blood and CSF (blood-CSF barrier). Conversely, no tight barrier separates CSF from the extracellular fluid of the brain. The arachnoid granulations, one-way valves for the return of CSF to the blood, even allow the passage of bacteria and red and white blood cells. The CSF space and the interstitial space of the central nervous system are not homogeneous compartments, e.g., at equal concentration-versus-time curves in serum (plasma), the pharmacokinetics of a drug in ventricular and lumbar CSF can strongly differ. (Right) The sites of CSF production and drainage are listed. CSF flow has a strong circadian rhythm. It also depends on underlying diseases, blood and intracranial pressure, and drugs influencing the activity of the choroid plexus or the permeability of the blood-brain barrier. In addition to physiological drainage sites, an external ventricular and lumbar catheter are also shown: in many studies analyzed, CSF was drawn from indwelling catheters. Regions where the CSF flow is often interrupted in clinical routine are also depicted. Blockade of the CSF flow can alter the volume of distribution of the CSF space. The average volumes and fluxes were taken from Davon et al. (20). Interindividual variation is great. The host-dependent variables influencing CSF kinetics of an anti-infective make it very difficult to predict the course of the concentration-time curves in the CSF of patients. The CCT was kindly provided by J. Gossner, Department of Radiology, Evangelisches Krankenhaus Göttingen-Weende, Germany, reproduced with permission.
The present study reanalyzes previously published pharmacokinetic data with the aim of assessing the influence of the drugs' physicochemical properties on the entry of anti-infectives into the CSF.
MATERIALS AND METHODS
Previously generated and compiled data (95) were analyzed. Relevant publications were identified by a PubMed search using the following algorithm: name of the antibiotic and location (cerebrospinal or brain) and the concentration or concentrations and human. We also contacted the manufacturers for information on the penetration of the respective drugs into the cerebrospinal fluid. Previously we outlined the importance of the ratios of the areas under the concentration-time curves in CSF and serum or plasma (AUCCSF/AUCS or AUCCSF/AUCplasma) for the description of drug entry into the CSF (96, 97). Only pharmacokinetic studies that either reported AUCCSF/AUCS ratios or that permitted calculation of these ratios or that clearly reported CSF-to-serum concentration ratios at steady state (CCSFss/CSss) were analyzed. For subsequent calculations, we used the means or (in the absence of Gaussian distribution) medians of AUCCSF/AUCS (CCSFss/CSss) ratios determined for individual patients. When several investigations were identified for an individual compound, the data from all studies meeting the criteria outlined above were used independently. No pharmacokinetic CSF data after intraventricular or intraspinal injection of antibiotics alone or in combination with systemic administration were included in this analysis.
First, we analyzed data derived from patients with uninflamed meninges (31 compounds; 40 independent sets of data derived from different studies). Then, data reporting the entry of antibiotics into the CSF in patients during meningitis in patients were studied (27 compounds; 47 independent sets of data).
We established a correlation matrix between the following parameters: AUCCSF/AUSS, ratio of the areas under the concentration-time curves in CSF and of the fraction that was not bound in serum (AUCCSF/AUCS,free), common or decadic logarithm of the octanol-water partition coefficient of the un-ionized drug (log P), decadic logarithm of the octanol-water partition coefficient at pH 7.0 (log D), molecular mass (MM), fraction unbound to serum proteins (FF), and the ratios of log P/√MM and log D/√MM. Moreover, the polar surface area (PSA) and molecular volume were correlated with AUCCSF/AUCS and AUCCSF/AUCS,free. To ensure a homogeneous set of data, we used log P, log D, PSA, and molecular volume calculated with Advanced Chemistry Development (ACD/Labs, Toronto, Canada) software (V8.14 and V11.02) published online by SciFinder 2007, unless indicated otherwise. Data on binding to serum (plasma) proteins were taken from the references indicated in Tables 1 and 2. Since the relation between individual variables was clearly nonlinear in many cases even after transformation, Spearman's nonparametric correlation coefficient rS was used (112).
Table 1.
CSF penetration versus physicochemical properties of anti-infectives in patients with uninflamed meninges
| Drug | Molecular mass (g/mol) | Partition coefficienta |
Protein binding (%) | Protein binding reference | AUCCSF/AUCS | AUCCSF/AUCS,free | Pharmacokinetic reference(s) | |
|---|---|---|---|---|---|---|---|---|
| Log PO/W | Log DO/W | |||||||
| Penicillins | ||||||||
| Cloxacillin | 436 | 2.53 ± 0.34b | −0.87c | 93 | 68 | 0.0087 | 0.12 | 114 |
| Piperacillin | 518 | 1.88 ± 0.37b | −2.69c | 25 | 118 | 0.034 | 0.045 | 87 |
| β-Lactamase inhibitors | ||||||||
| Clavulanate | 199 | −1.98 ± 0.78b | −3.47c | 26 | 67 | 0.037 | 0.05 | F. Sörgel et al., unpublished data |
| Tazobactam | 300 | −1.70 ± 0.84b | −3.10c | 21.5 | 118 | 0.106 | 0.135 | 87 |
| Cephalosporins | ||||||||
| Cefotaxime | 455 | 1.20 ± 0.89b | −2.47b | 25.5 | 71 | 0.12 | 0.16 | 93 |
| Ceftriaxone | 555 | −0.25 ± 1.12b | −4.43c | 94.5 | 73 | 0.007 | 0.13 | 93 |
| Ceftazidime | 547 | −1.21d | −2.07 (135) | 14.5 | 72 | 0.057 | 0.067 | 90 |
| Carbapenem meropenem | 383 | −3.13 ± 0.75b | −3.74c | 2 | 7 | 0.047, 0.21, 0.25 | 0.048, 0.21, 0.26 | 61, 89 |
| Aminoglycoside netilmicin | 476 | −1.90 ± 0.84b | −7.02c | 0 | 16 | 0.24 | 0.24 | 94 |
| Fluoroquinolones | ||||||||
| Ciprofloxacin | 331 | 1.31 ± 0.79b | −0.33c | 30 | 52 | 0.24, 0.43 | 0.34, 0.61 | 91, 133 |
| Ofloxacin | 361 | 1.61 ± 0.81b | −0.20c | 32.5 | 52 | 0.65 | 0.96 | 88 |
| Levofloxacin | 361 | 1.61 ± 0.81b | −0.20c | 32.5 | 132 | 0.71 | 1.05 | 103 |
| Moxifloxacin | 401 | 1.90 ± 0.82b | −0.63b | 37.5 | 132 | 0.46 | 0.74 | 43 |
| Fosfomycin | 138 | −2.98 ± 0.60b | −4.77c | 0 | 111 | 0.092, 0.128 | 0.092, 0.128 | 45 |
| Chloramphenicol | 323 | 1.02 ± 0.37b | 1.10c | 60 | 75 | 0.67 | 1.68 | 31 |
| Colistin | 1,155 | −1.28d | 55 | 28 | 0.051 | 0.11 | 59 | |
| Rifamycin rifampin | 823 | 1.09 ± 0.74b | −0.38c | 87.5 | 78 | 0.22 | 1.76 | 92 |
| Folate antagonists | ||||||||
| Trimethoprim | 290 | 0.79 ± 0.38b | 0.27c | 44 | 77 | 0.18 | 0.32 | 24 |
| Sulfamethoxazole | 253 | 0.89 ± 0.42b | −0.22c | 70 | 64 | 0.12 | 0.4 | 24 |
| Glycopeptide vancomycin | 1,449 | −1.44 ± 1.02b | −4.67c | 40 | 47 | 0.14, 0.18 | 0.30, 0.23 | 1 |
| Antiherpesvirus nucleoside analogues | ||||||||
| Aciclovir | 225 | −1.76 ± 0.49b | −1.48c | 21 | 63 | 0.31 | 0.39 | 56, 57 |
| Foscarnet | 126 | −1.63d | −6.78c | 15 | 53 | 0.27, 0.43 | 0.32, 0.51 | 108, 116 |
| Antiretroviral drugs | ||||||||
| Abacavir | 286 | 0.72 ± 0.82b | 0.58b | 48 | 11 | 0.35 | 0.67 | 62 |
| Zidovudine | 267 | 1.15 (31a) | −0.58 (127a) | 27 | 11 | 0.75 | 1.03 | 110 |
| Indinavir | 614 | 2.88 ± 0.86b | 2.86b | 60 | 11 | 0.06, 0.147 | 0.15, 0.37 | 36, 49 |
| Lopinavir | 629 | 6.26 ± 0.84b | 6.26b | 98 | 11 | 0.285 | 14.3 | 13 |
| Antiparasitic drugs | ||||||||
| Albendazole | 265 | 3.07 ± 0.83b | 2.99c | 70 | 21 | 0.37, 0.38, 0.43 | 1.23, 1.27, 1.43 | 42, 123 |
| Praziquantel | 312 | 2.44 ± 0.69b | 2.66c | 80 | 21 | 0.24 | 1.2 | 42 |
| Sulfadiazine | 250 | −0.12 ± 0.26b | −0.68c | 44 | 69 | 0.27, 0.34 | 0.54 | 2 |
| Tetroxoprim | 335 | 0.46 ± 0.43b | −0.03c | 15 | 135 | 0.23, 0.39 | 0.36 | 2 |
The logarithm of the octanol-water partition coefficient of the drug (log PO/W) and the logarithm of the octanol-water partition coefficient (log DO/W) are shown. The drug was tested at pH 7 unless specified otherwise.
Calculated using Advanced Chemistry Development (ACD/Labs) software V8.14 (1994 to 2011 ACD/Labs) published online by SciFinder 2007.
Calculated using Advanced Chemistry Development (ACD/Labs) software V11.02 (1994 to 2011 ACD/Labs) published online by SciFinder 2007.
Data from the DrugBank database (http://drugbank.ca/).
Table 2.
CSF penetration versus physicochemical properties of anti-infectives in patients with inflamed meninges
| Drug | Molecular mass (g/mol) | Partition coefficienta | Protein binding (%) | Protein binding reference | AUCCSF/AUCS | AUCCSF/AUCS,free | Pharmacokinetics reference(s) | |
|---|---|---|---|---|---|---|---|---|
| Log PO/W | Log DO/W | |||||||
| Penicillins | ||||||||
| Amoxicillin | 365 | 0.61 ± 0.33b | −2.08c | 18.5 | 76 | 0.058 | 0.071 | 8 |
| Piperacillin | 518 | 1.88 ± 0.37b | −2.69c | 25 | 118 | 0.32 | 0.43 | 22 |
| β-Lactamase inhibitor clavulanate | 199 | −1.98 ± 0.78b | −3.47c | 26 | 67 | 0.084 | 0.11 | 8 |
| Cephalosporins | ||||||||
| Ceftriaxone | 555 | −0.25 ± 1.12b | −4.43c | 94.5 | 73 | 0.17, 0.041 | 3.09, 0.75 | 60 |
| Cefepime | 481 | −0.36d | −1.07e | 20 | 70 | 0.103 | 0.13 | 113 |
| Cefpirome | 515 | −1.70 (82a) | −1.5e | 10 | 83 | 0.145, 0.31 | 0.16, 0.34 | 30 |
| Carbapenems | ||||||||
| Imipenem | 299 | −2.78 ± 0.76b | −5.28b | 17 | 66 | 0.14 | 0.17 | 134 |
| Cilastatin | 358 | 2.42 ± 0.55b | −2.77c | 40 | 66 | 0.1 | 0.17 | 134 |
| Meropenem | 383 | −3.13 ± 0.75b | −3.74c | 2 | 7 | 0.39 | 0.4 | 15 |
| Fluoroquinolones | ||||||||
| Ciprofloxacin | 331 | 1.31 ± 0.79b | −0.33c | 30 | 52 | 0.92 | 1.31 | 133 |
| Moxifloxacin | 401 | 1.90 ± 0.82b | −0.63b | 37.5 | 132 | 0.94, 0.74, 0.82, 0.71 | 1.50, 1.18, 1.31, 1.14 | 3, 4 |
| Macrolide | ||||||||
| clarithromycin | 748 | 3.16 ± 0.78b | 1.71c | 69.5 | 19 | 0.18 | 0.59 | 58 |
| Fosfomycin | 138 | −2.98 ± 0.60b | −4.77c | 0 | 111 | 0.23, 0.27 | 0.23, 0.27 | 106 |
| Chloramphenicol | 323 | 1.02 ± 0.37b | 1.10c | 60 | 75 | 0.60, 0.65, 0.67 | 1.50, 1.63, 1.68 | 31, 136 |
| Colistin | 1,155 | −1.28d | 55 | 28 | 0.051, 0.16 | 0.11, 0.36 | 41, 59 | |
| Oxazolidinone linezolid | 337 | 0.43 ± 0.73b | 0.41b | 31 | 117 | 0.8, 1.0 | 1.16, 1.45 | 9 |
| Nitroimidazole metronidazole | 171 | −0.01 ± 0.30b | −0.14c | 20 | 65 | 0.87 | 1.09 | 130 |
| Rifamycin rifampin | 823 | 1.09 ± 0.74b | −0.38c | 87.5 | 78 | 0.05 | 0.4 | 27 |
| Folate antagonists | ||||||||
| Trimethoprim | 290 | 0.79 ± 0.38b | 0.27c | 44 | 77 | 0.47 | 0.84 | 51 |
| Sulfamethoxazole | 253 | 0.89 ± 0.42b | −0.22c | 70 | 64 | 0.3 | 1 | 51 |
| Glycopeptide vancomycin | 1,449 | −1.44 ± 1.02b | −4.67c | 40 | 47 | 0.07, 0.30, 0.29, 0.48 | 0.12, 0.50, 0.48, 0.80 | 1, 105, 109 |
| Antituberculosis drugs | ||||||||
| Isoniazid | 137 | −0.89 ± 0.24b | −0.77c | 0 | 39 | 0.78, 0.86, 1.17 | 0.78, 0.86, 1.17 | 23, 27, 115 |
| Streptomycin | 582 | −2.53 ± 0.85b | −5.46c | 46.2 | 39 | 0.12 | 0.22 | 27 |
| Foscarnet | 126 | −1.63d | −6.78c | 15 | 53 | 0.23, 0.66, 0.27 | 0.27, 0.78, 0.32 | 38, 108 |
| Antifungal drug fluconazole | 306 | 0.50 ± 0.89b | 0.45c | 12 | 10 | 0.74, 0.86, 0.89 | 0.84, 0.98, 1.01 | 80, 126 |
| Antiparasitic drugs | ||||||||
| Albendazole | 265 | 3.07 ± 0.83b | 2.99c | 70 | 21 | 0.38, 0.43 | 1.27, 1.43 | 42, 123 |
| Praziquantel | 312 | 2.44 ± 0.69b | 2.66c | 80 | 21 | 0.24 | 1.2 | 42 |
The logarithm of the octanol-water partition coefficient of the drug at pH 7 (log PO/W) and the logarithm of the octanol-water partition coefficient at pH 7.0 (log DO/W) are shown. The drug was tested at pH 7 unless specified otherwise.
Calculated using Advanced Chemistry Development (ACD/Labs) software V8.14 (1994 to 2011 ACD/Labs) published online by SciFinder 2007.
Calculated using Advanced Chemistry Development (ACD/Labs) software V11.02 (1994 to 2011 ACD/Labs) published online by SciFinder 2007.
Data from the DrugBank database (http://drugbank.ca/).
Calculated for pH 7.4. Data were provided by LookChem (www.lookchem.com).
RESULTS
In the absence of meningeal inflammation (n = 40), the CSF penetration of anti-infectives in humans as estimated by AUCCSF/AUCS (CCSFss/CSss) correlated positively with the lipid-water partition coefficient at pH 7.0 (D or log D) (Spearman's rank correlation coefficient rS = 0.40; P = 0.01) and negatively with the molecular mass (MM) (rS = −0.33; P = 0.04). The correlation of AUCCSF/AUCS with log D/√MM was not stronger (rS = 0.40; P = 0.01) than the correlation with log D. AUCCSF/AUCS did not correlate with the fraction unbound in serum (rS = −0.06; P = 0.70). The ratio of AUCCSF to the unbound fraction of AUCS (AUCCSF/AUCS,free) strongly correlated with log D (rS = 0.67; P < 0.0001) and log D/√MM (rS = 0.69; P < 0.0001) (Fig. 2). The correlation of AUCCSF/AUCS and AUCCSF/AUCS,free with log P was substantially weaker than with log D (0.31 versus 0.40 and 0.56 versus 0.67).
Fig 2.

Entry of anti-infectives into the CSF in the absence (A) and presence (B) of meningeal inflammation depends on the octanol-water partition coefficient at pH 7.0. The abscissa shows the log of the octanol-water partition coefficient at pH 7 (log D). The ordinate shows the ratio of the concentration-time curves in cerebrospinal fluid and serum (AUCCSF/AUCS). Log D correlated with AUCCSF/AUCS (Spearman's rank correlation coefficient rS was 0.40 in the absence of meningeal inflammation [A], and rS was 0.46 in the presence of meningeal inflammation [B]; P = 0.01 and P =0.002, respectively). The correlation would have been even stronger if the highly hydrophilic foscarnet (log D of −6.78), which readily enters the CSF because of its small molecular size, and if lipophilic compounds (log D of >1.5 corresponding to a partition coefficient of >30), which were often highly bound to serum proteins, had been excluded from the analysis (for patients with uninflamed meninges, rS = 0.52, P = 0.003, and n = 31; for patients with meningeal inflammation, rS = 0.62, P < 0.0001, and n = 38).
In the presence of meningeal inflammation (n = 47), AUCCSF/AUCS also correlated positively with log D (rS = 0.46; P = 0.002) and negatively with MM (rS = −0.37; P = 0.01). Moreover, AUCCSF/AUCS correlated with the fraction unbound in serum (rS = 0.33; P = 0.025). The correlation of AUCCSF/AUCS,free with log D (rS = 0.66; P < 0.0001) was as strong as in the absence of meningeal inflammation. The use of log D/√MM instead of log D did not increase the correlation with AUCCSF/AUCS,free (rS = 0.66 for both; P < 0.0001). Again, the correlation of AUCCSF/AUCS and AUCCSF/AUCS,free with log P was substantially weaker than with log D (0.28 versus 0.46 and 0.60 versus 0.66).
The AUCCSF/AUCS,free of a few highly protein-bound compounds both with uninflamed (lopinavir) and inflamed meninges (one of two reports on ceftriaxone) strongly exceeded 1.0.
In both patients with uninflamed meninges and patients with meningitis, the polar surface area (PSA) correlated with AUCCSF/AUCS (rS = −0.51, P = 0.001, and rS = −0.64, P < 0.0001) to a higher degree than log D correlated with AUCCSF/AUCS, but PSA correlated less strongly with AUCCSF/AUCS,free (rS = −0.38, P = 0.02, n = 38, and rS = −0.36, P = 0.02, n = 42) than log D correlated with AUCCSF/AUCS,free. The molecular volume did not significantly correlate with either AUCCSF/AUCS or AUCCSF/AUCS,free.
DISCUSSION
We found a strong correlation between the octanol-water partition coefficient at pH 7.0 (log D) and AUCCSF/AUCS both in the absence and presence of meningeal inflammation. The assumption of a Gaussian distribution of the data and use of Pearson correlation with or without logarithmic transformation of the scales instead of Spearman's rank correlation coefficient did not make the correlation between AUCCSF/AUCS and D stronger. In accordance with Overton's rule in its original form, which focuses on lipophilicity and neglects molecular mass and binding to serum protein, D (log D) had a great explanatory value for the behavior of anti-infective drugs at the human blood-CSF barrier. The correlation between log D and AUCCSF/AUCS (Fig. 2A and B) would have been even stronger if the highly hydrophilic foscarnet (log D −6.78), which readily enters the CSF because of its small molecular size, and if the lipophilic compounds (log D of >1.5 corresponding to a partition coefficient of >30), which are usually highly bound to serum proteins (128), had been excluded from the analysis (for patients with uninflamed meninges, rS = 0.52, P = 0.003, and n = 31; for patients with meningeal inflammation, rS = 0.62, P < 0.0001, and n = 38). The partition coefficient of the un-ionized drug (log P) explains less well the intercompound variations of drug entry into the CSF than does the partition coefficient at a fixed pH (log D), in this case pH 7. Since most of the substances discussed have acidic or basic properties and therefore are ionizable, this result is not astonishing.
In vitro and in murine animal models, penetration through the blood-brain barrier was proportional to the product of the lipid-water partition coefficient and diffusion coefficient. Because the diffusion coefficient in aqueous solvents inversely correlates with the molecular size, it approximately correlates with 1/√MM (50). In the present study, the correlation between the MM and drug entry into the CSF was statistically significant, but comparatively low. Use of the molecular volume instead of the MM did not lead to a stronger correlation with CSF penetration. Similarly, use of log D/√MM instead of log D did not substantially increase the correlation with AUCCSF/AUCS or AUCCSF/AUCS,free both with uninflamed and inflamed meninges. The probable reason for the comparatively low influence of MM is the small range of molecular size studied (126 to 1,449 Da). For large hydrophilic molecules, e.g., proteins, molecular size is a strong determinant of drug entry into the CSF (29). Both with uninflamed and inflamed meninges, the correlation of AUCCSF/AUCS,free with log D was substantially stronger than the correlation of AUCCSF/AUCS with log D, underlining the importance of drug binding to serum (plasma) proteins.
The polar surface area (PSA) correlated to a higher degree with AUCCSF/AUCS than log D did, but PSA correlated less strongly with AUCCSF/AUCS,free than log D did. For this reason, its use did not represent a substantial advantage compared to log D under these conditions. The molecular volume did not correlate significantly either with AUCCSF/AUCS or with AUCCSF/AUCS,free in the absence or presence of meningeal inflammation.
Reasons for failure to establish nomograms.
Our data do not allow us to establish nomograms to predict CSF penetration on the grounds of physicochemical drug properties and binding to serum proteins (e.g., Fig. 2A and B). There are several reasons for this failure, involving the great variability in CSF pharmacokinetics.
(i) Variation in serum pharmacokinetics.
Particularly in critically ill patients, the pharmacokinetics of anti-infectives is very heterogeneous (104). A meta-analysis of 57 studies of 6 different β-lactam antibiotics showed a more than 2-fold variation both in the volume of distribution (V) and in total body clearance (CL) (34).
(ii) Influence of age of the subjects investigated on the permeability of the blood-CSF barrier and the volumes of the intracranial compartments.
Many studies included patients of different ages, including newborns. Physiologically, the CSF-to-serum albumin ratio as a combined measure of the state of the blood-CSF barrier and the CSF flow, which both affect the AUCCSF/AUCS of a drug administered into the circulation system, depends on the subject's age. In newborns, the CSF-to-serum albumin ratio is high. It then decreases to its minimum at approximately 4 years and then slowly increases with age (138). As a consequence of the age-related physiological involution of the brain, the ratio of CSF space/brain tissue increases with age (137).
(iii) Different underlying diseases in patients without meningeal inflammation.
The studies reporting CSF entry in the absence of meningeal inflammation did not investigate healthy persons in most cases, but patients with different degrees of abnormal blood-CSF barrier or CSF flow. In many studies, patients with external CSF drains were included. The additional clearance by external bulk flow in patients treated with either ventricular or lumbar CSF drainage is an important determinant of CSF pharmacokinetics of anti-infectives after intravenous (i.v.) administration. The clinical decision, whether the drain is open, and the amount of CSF drained or whether the drain is closed during the pharmacokinetic study can strongly influence the CSF kinetics of the drug studied. However, data on the flow through an external CSF drain are usually not reported in pharmacokinetic studies. Moreover, a blockade of the CSF circulation at the level of the aqueduct or of the basal cisterns decreases the volume of distribution of the CSF space, thereby influencing the pharmacokinetics of a drug in the CNS.
(iv) Intensity of meningeal inflammation.
In patients with meningitis, capillary permeability is increased, and transport across the epithelium of the choroid plexus may be affected (107). The degree of meningeal inflammation in the patients studied was highly variable. In experimental animals, dexamethasone, which is recommended as adjunct therapy for community-acquired bacterial meningitis in industrialized countries, causes a reduction in the CSF concentrations of vancomycin by approximately 30%, resulting in a delay in CSF sterilization not observed in non-dexamethasone-treated animals (102). In children with bacterial meningitis receiving adjunct dexamethasone treatment, the CSF concentrations of cefuroxime were reduced on an average by one dilution step (i.e., approximately 50%) (48).
(v) Lumbar versus ventricular CSF.
Some studies investigated lumbar CSF, others used ventricular CSF, and several studies either investigated CSF from both sites or did not clearly state from which site CSF was obtained. Physiologically, the CSF-to-serum albumin ratio is approximately 2 times lower in the ventricular CSF than in the lumbar CSF (124, 131).
(vi) Binding to serum proteins.
It is generally believed that only the free fraction in serum is available for penetration into the compartments of the central nervous system (98). For some highly protein-bound drugs (e.g., ceftriaxone) (122), the percentage of protein binding in serum depends on the total serum concentration. Binding to serum proteins is also influenced by the patient's protein concentration and composition in blood and on the comedication (79, 121, 127). Moreover, the notion that only the free serum fraction equilibrates with the drug fraction unbound in CSF has been questioned: the amount of some highly protein-bound drugs available for transport into the CNS may be not restricted to the free (dialyzable) fraction but may include the much larger globulin-bound fraction (101). Because of the low protein concentrations in normal CSF, binding of anti-infectives to CSF proteins has usually been neglected (60). In the case of drugs with a high affinity to serum proteins, however, this is a simplification. In children with Haemophilus influenzae meningitis, 19% ± 6% of the total CSF ceftriaxone concentration was bound to protein (40). Drug binding to CSF proteins is probably one explanation for the AUCCSF/AUCS,free ratios far above 1 encountered with some compounds (lopinavir in patients with uninflamed meninges, ceftriaxone in patients with inflamed meninges) in the present study.
(vii) Possible limitation of the validity of Overton's rule to molecules with a molecular mass below 500 Da.
In experimental animals, molecules with a molecular mass above 500 Da crossed the blood-brain barrier less readily than expected by Overton's rule (50). Equivalent data for the blood-CSF barrier in humans are not available.
(viii) Active transport.
In recent decades, many anti-infectives have been identified as ligands of active transport systems that remove toxic compounds from the compartments of the central nervous system (81, 120). The influence of these systems on the drug concentrations in the intracranial compartments varies from strong to absent. Since these transport systems in most cases bind to several drugs (for a review, see reference 55), interactions between antibiotics and the comedication can occur. P-glycoprotein (P-gp) has a broad spectrum of ligands favoring lipophilic compounds. This pump can remove substrates of approximately 300 to 4,000 Da from the compartments of the central nervous system (81). It is difficult to predict whether a drug is a strong ligand of P-gp (5). Polymorphisms of the ABCB1 gene, which codes for P-gp, can influence the kinetics of several drugs (14). The organic anion transporter 3 (OAT3) and peptide transporter 2 (PEPT2) remove several penicillins and cephalosporins from the CNS (46, 84). OAT3 is an active outward transport system for weak organic acids. Cephalothin, a strong ligand of OAT3, appears to be unsuitable for treatment of bacterial meningitis, because it is rapidly removed from CSF (120). Among other compounds, OAT3 is involved in the uptake of histamine H2 receptor antagonists used in intensive care medicine for the prophylaxis and therapy of gastric ulcers (cimetidine, ranitidine, and famotidine). It has been shown that drug-drug interactions cause an increase in the CSF concentrations of H2 receptor antagonists without affecting their plasma concentrations (85). OAT3 decreases CSF concentrations of ligands particularly in the absence of meningeal inflammation and is inhibited by probenecid. In experimental animals with uninflamed meninges, inhibition of OAT3 by probenecid increased the CSF concentrations of penicillin G after intracisternal injection by a factor of 1.5 to 3 (129) and the CSF-to-serum concentration ratio during continuous i.v. infusion by 2 to 3 times (18). This effect tended to be weaker in patients with meningitis (18). In 4 patients with late uncomplicated syphilis receiving 600,000 IU penicillin G procaine intramuscularly, CSF penicillin concentrations were ≤0.015 μg/ml. Coadministration of probenecid 500 mg every 6 h orally caused an increase in the CSF penicillin concentration to 0.016 to 0.29 μg/ml (median of 0.033 μg/ml) (25).
The influence of the transport system of weak organic acids on the exchange of many β-lactam antibiotics at the blood-CSF barrier appears to be moderate (for example, see reference 119). In children on the 10th day of treatment for bacterial meningitis (i.e., after the acute phase), coadministration of probenecid increased the AUC from 0 to 4 h (AUC0–4) of amoxicillin in plasma by approximately 40% and in CSF by approximately 75%. The average AUCCSF,0–4/AUCS,0–4 of amoxicillin rose from 0.04 without probenecid coadministration to 0.05 with probenecid (estimated from Tables 2 and 3 in reference 17). Ceftriaxone, cefotaxime, and meropenem, belonging to the standard therapy of community- and hospital-acquired bacterial meningitis, have low affinity to OAT3 and PEPT2 (95).
In rats, the antivirals aciclovir and zidovudine had a high affinity to OAT3: probenecid increased the CSF-to-unbound plasma concentration ratio from 17 to 29% (zidovudine) and from 8 to 17% (aciclovir), whereas the concentration ratio of cytarabine (AraC) was unchanged (44). In laboratory rabbits, coadministration of probenecid increased AUCCSF/AUCplasma of zidovudine from 0.15 ± 0.02 to 0.57 ± 0.22 (37).
Detailed reviews on the affinity of anti-infectives and other drugs for the different transport systems located at the blood-CSF and blood-brain barriers have been published recently (54, 81, 95, 120).
(ix) pH gradient.
In patients with uninflamed meninges, there is a small pH gradient between blood (pH approximately 7.4) and CSF (pH approximately 7.3) (20, 125). The pH gradient between blood and CNS is greater in patients with meningitis, where a brain interstitial pH as low as 6.9 has been encountered (6). The un-ionized more-lipophilic fraction of acids therefore is higher in the compartments of the central nervous system than in blood. For this reason, weak acids diffuse more readily from CSF to blood than from blood to CSF, and this effect is greater in patients with inflamed meninges than in patients with uninflamed meninges (125).
(x) Publication bias.
In the present analysis, several studies reporting AUC ratios for vancomycin from 0.07 to as high as 0.48 were included (Tables 1 and 2). Conversely, after 1 g of vancomycin was administered i.v., we were unable to detect measurable vancomycin concentrations in CSF (detection limit of 0.05 μg/ml) (R. Nau and J. Bircher, unpublished observation). Since negative results are often not published, the CSF penetration of some compounds, in particular vancomycin, colistin, and netilmicin, may have been overestimated by the analysis of published data.
In conclusion, the entry of an anti-infective into the compartments of the central nervous system is determined by the variable properties of the drug, host, or both. The most important host-independent physicochemical drug property is lipophilicity at pH 7. In other words, Overton's rule is still valid. At the molecular sizes studied, molecular mass accounted for less of the interdrug variation than did lipophilicity at a neutral pH. Active transport and drug binding to serum proteins both depend on the drug's properties, on comedications which either influence drug protein binding or the activity of transport systems, on polymorphisms of genes encoding the transport proteins, and on the protein content in the circulation system and CSF. Host variables independent of the drug's physicochemical properties are the subject's age, an underlying disease affecting the state of the barriers and CSF flow, and the site of CSF withdrawal. Physicochemical drug properties help to predict which anti-infective will readily enter the CNS, and from data generated in animals under uniform experimental conditions, nomograms for the estimation of penetration into the compartments of the central nervous system have been constructed. Because of the great heterogeneity of published human data, predicting exact CSF concentrations based on physicochemical drug properties and pharmacokinetic data in the circulation system, however, in clinical practice is very difficult.
ACKNOWLEDGMENTS
We thank Christine Crozier for carefully going over the language of the manuscript.
H.E. and R.N. were supported by grants from the European Commission (CAREPNEUMO) and the Else Kröner-Fresenius-Stiftung. M.D. received a grant from the Robert-Bosch-Stiftung.
Footnotes
Published ahead of print 21 November 2011
REFERENCES
- 1. Albanese J, et al. 2000. Cerebrospinal fluid penetration and pharmacokinetics of vancomycin administered by continuous infusion to mechanically ventilated patients in an intensive care unit. Antimicrob. Agents Chemother. 44: 1356–1358 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Albert F, Bishop-Freudling GB, Vergin H. 1984. Diffusion of tetroxoprim and sulfadiazine in the cerebrospinal fluid of neurosurgery patients. Fortschr. Med. 102: 1064–1066 (In German) [PubMed] [Google Scholar]
- 3. Alffenaar JW, et al. 2008. Plasma and cerebrospinal fluid pharmacokinetics of moxifloxacin in a patient with tuberculous meningitis. Antimicrob. Agents Chemother. 52: 2293–2295 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Alffenaar JW, et al. 2009. Pharmacokinetics of moxifloxacin in cerebrospinal fluid and plasma in patients with tuberculous meningitis. Clin. Infect. Dis. 49: 1080–1082 [DOI] [PubMed] [Google Scholar]
- 5. Aller SG, et al. 2009. Structure of P-glycoprotein reveals a molecular basis for poly-specific drug binding. Science 323: 1718–1722 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Andersen NE, Gyring J, Hansen AJ, Laursen H, Siesjo BK. 1989. Brain acidosis in experimental pneumococcal meningitis. J. Cereb. Blood Flow Metab. 9: 381–387 [DOI] [PubMed] [Google Scholar]
- 7. AstraZeneca 2007. Product monograph “Merrem.” AstraZeneca, Mississauga, Ontario, Canada [Google Scholar]
- 8. Bakken JS, Bruun JN, Gaustad P, Tasker TC. 1986. Penetration of amoxicillin and potassium clavulanate into the cerebrospinal fluid of patients with inflamed meninges. Antimicrob. Agents Chemother. 30: 481–484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Beer R, et al. 2007. Pharmacokinetics of intravenous linezolid in cerebrospinal fluid and plasma in neurointensive care patients with staphylococcal ventriculitis associated with external ventricular drains. Antimicrob. Agents Chemother. 51: 379–382 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Bellmann R. 2007. Clinical pharmacokinetics of systemically administered antimycotics. Curr. Clin. Pharmacol. 2: 37–58 [DOI] [PubMed] [Google Scholar]
- 11. Boffito M, et al. 2003. Protein binding in antiretroviral therapies. AIDS Res. Hum. Retroviruses 19: 825–835 [DOI] [PubMed] [Google Scholar]
- 12. Brodie BB, Kurz H, Schanker LS. 1960. The importance of dissociation constant and lipid-solubility in influencing the passage of drugs into the cerebrospinal fluid. J. Pharmacol. Exp. Ther. 130: 20–25 [PubMed] [Google Scholar]
- 13. Capparelli EV, et al. 2005. Lopinavir concentrations in cerebrospinal fluid exceed the 50% inhibitory concentration for HIV. AIDS 19: 949–952 [DOI] [PubMed] [Google Scholar]
- 14. Choong E, Dobrinas M, Carrupt PA, Eap CB. 2010. The permeability P-glycoprotein: a focus on enantioselectivity and brain distribution. Expert Opin. Drug Metab. Toxicol. 6: 953–965 [DOI] [PubMed] [Google Scholar]
- 15. Chou YW, Yang YH, Chen JH, Kuo CC, Chen SH. 2007. Quantification of meropenem in plasma and cerebrospinal fluid by micellar electrokinetic capillary chromatography and application in bacterial meningitis patients. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 856: 294–301 [DOI] [PubMed] [Google Scholar]
- 16. Contrepois A, et al. 1985. Renal disposition of gentamicin, dibekacin, tobramycin, netilmicin, and amikacin in humans. Antimicrob. Agents Chemother. 27: 520–524 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Craft JC, Feldman WE, Nelson JD. 1979. Clinicopharmacological evaluation of amoxicillin and probenecid against bacterial meningitis. Antimicrob. Agents Chemother. 16: 346–352 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Dacey RG, Sande MA. 1974. Effect of probenecid on cerebrospinal fluid concentrations of penicillin and cephalosporin derivatives. Antimicrob. Agents Chemother. 6: 437–441 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Davey PG. 1991. The pharmacokinetics of clarithromycin and its 14-OH metabolite. J. Hosp. Infect. 19(Suppl. A): 29–37 [DOI] [PubMed] [Google Scholar]
- 20. Davson H, Welch K, Segal MB. 1987. Physiology and pathophysiology of the cerebrospinal fluid. Churchill Livingstone, London, United Kingdom [Google Scholar]
- 21. Dayan AD. 2003. Albendazole, mebendazole and praziquantel. Review of non-clinical toxicity and pharmacokinetics. Acta Trop. 86: 141–159 [DOI] [PubMed] [Google Scholar]
- 22. Dickinson GM, Droller DG, Greenman RL, Hoffman TA. 1981. Clinical evaluation of piperacillin with observations on penetrability into cerebrospinal fluid. Antimicrob. Agents Chemother. 20: 481–486 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Donald PR, Gent WL, Seifart HI, Lamprecht JH, Parkin DP. 1992. Cerebrospinal fluid isoniazid concentrations in children with tuberculous meningitis: the influence of dosage and acetylation status. Pediatrics 89: 247–250 [PubMed] [Google Scholar]
- 24. Dudley MN, Levitz RE, Quintiliani R, Hickingbotham JM, Nightingale CH. 1984. Pharmacokinetics of trimethoprim and sulfamethoxazole in serum and cerebrospinal fluid of adult patients with normal meninges. Antimicrob. Agents Chemother. 26: 811–814 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Dunlop EM, Al-Egaily SS, Houang ET. 1979. Penicillin levels in blood and CSF achieved by treatment of syphilis. JAMA 241: 2538–2540 [PubMed] [Google Scholar]
- 26. Ehrlich P. 1885. Über das Sauerstoffbedürfnis des Organismus. Eine farbenanalytische Studie. Hirschwald-Verlag, Berlin, Germany [Google Scholar]
- 27. Ellard GA, Humphries MJ, Allen BW. 1993. Cerebrospinal fluid drug concentrations and the treatment of tuberculous meningitis. Am. Rev. Respir. Dis. 148: 650–655 [DOI] [PubMed] [Google Scholar]
- 28. Falagas ME, Kasiakou SK. 2005. Colistin: the revival of polymyxins for the management of multidrug-resistant gram-negative bacterial infections. Clin. Infect. Dis. 40: 1333–1341 [DOI] [PubMed] [Google Scholar]
- 29. Felgenhauer K. 1974. Protein size and cerebrospinal fluid composition. Klin. Wochenschr. 52: 1158–1164 [DOI] [PubMed] [Google Scholar]
- 30. Friedland IR, Sultan E, Lehr KH, Lenfant B. 1998. Concentrations of cefpirome in cerebrospinal fluid of children with bacterial meningitis after a single intravenous dose. Antimicrob. Agents Chemother. 42: 199–201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Friedman CA, Lovejoy FC, Smith AL. 1979. Chloramphenicol disposition in infants and children. J. Pediatr. 95: 1071–1077 [DOI] [PubMed] [Google Scholar]
- 31a. GlaxoSmithKline 2008. Trizivir. GlaxoSmithKline Shire Canada, Missauga, Ontario, Canada [Google Scholar]
- 32. Goldmann EE. 1909. Die äußere und innere Sekretion des gesunden und kranken Organismus im Lichte der “vitalen Färbung.” Beitr. Klin. Chirurg. 64: 192–265 [Google Scholar]
- 33. Goldmann EE. 1913. Vitalfärbung am Zentralnervensystem. Abh. Preuß. Akad. Wiss. Phys.-Math. Klasse. 1: 1–60 [Google Scholar]
- 34. Goncalves-Pereira J, Povoa P. 2011. Antibiotics in critically ill patients - a systematic review of the pharmacokinetics of beta-lactams. Crit. Care 15: R206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Grime JM, Edwards MA, Rudd NC, Unwin PR. 2008. Quantitative visualization of passive transport across bilayer lipid membranes. Proc. Natl. Acad. Sci. U. S. A. 105: 14277–14282 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Haas DW, et al. 2000. Steady-state pharmacokinetics of indinavir in cerebrospinal fluid and plasma among adults with human immunodeficiency virus type 1 infection. Clin. Pharmacol. Ther. 68: 367–374 [DOI] [PubMed] [Google Scholar]
- 37. Hedaya MA, Sawchuk RJ. 1989. Effect of probenecid on the renal and nonrenal clearances of zidovudine and its distribution into cerebrospinal fluid in the rabbit. J. Pharm. Sci. 78: 716–722 [DOI] [PubMed] [Google Scholar]
- 38. Hengge UR, Brockmeyer NH, Malessa R, Ravens U, Goos M. 1993. Foscarnet penetrates the blood-brain barrier: rationale for therapy of cytomegalovirus encephalitis. Antimicrob. Agents Chemother. 37: 1010–1014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Holdiness MR. 1984. Clinical pharmacokinetics of the antituberculosis drugs. Clin. Pharmacokinet. 9: 511–544 [DOI] [PubMed] [Google Scholar]
- 40. Hoshino T, Ishiwada N, Kohno Y. 2010. Free concentration and protein-binding ratio of ceftriaxone in cerebrospinal fluid in paediatric patients with purulent meningitis caused by Haemophilus influenzae type b. Int. J. Antimicrob. Agents 35: 512–513 [DOI] [PubMed] [Google Scholar]
- 41. Jimenez-Mejias ME, et al. 2002. Cerebrospinal fluid penetration and pharmacokinetic/pharmacodynamic parameters of intravenously administered colistin in a case of multidrug-resistant Acinetobacter baumannii meningitis. Eur. J. Clin. Microbiol. Infect. Dis. 21: 212–214 [DOI] [PubMed] [Google Scholar]
- 42. Jung H, Hurtado M, Sanchez M, Medina MT, Sotelo J. 1990. Plasma and CSF levels of albendazole and praziquantel in patients with neurocysticercosis. Clin. Neuropharmacol. 13: 559–564 [DOI] [PubMed] [Google Scholar]
- 43. Kanellakopoulou K, et al. 2008. Pharmacokinetics of moxifloxacin in non-inflamed cerebrospinal fluid of humans: implication for a bactericidal effect. J. Antimicrob. Chemother. 61: 1328–1331 [DOI] [PubMed] [Google Scholar]
- 44. Kawaguchi T, et al. 2000. Roles of hydrophobicity, protein binding and the probenecid-sensitive transport system in the cerebrospinal fluid delivery of nucleoside analogues with anti-viral activity. Biol. Pharm. Bull. 23: 979–983 [DOI] [PubMed] [Google Scholar]
- 45. Kuhnen E, Pfeifer G, Frenkel C. 1987. Penetration of fosfomycin into cerebrospinal fluid across non-inflamed and inflamed meninges. Infection 15: 422–424 [DOI] [PubMed] [Google Scholar]
- 46. Kuroda M, Kusuhara H, Endou H, Sugiyama Y. 2005. Rapid elimination of cefaclor from the cerebrospinal fluid is mediated by a benzylpenicillin-sensitive mechanism distinct from organic anion transporter 3. J. Pharmacol. Exp. Ther. 314: 855–861 [DOI] [PubMed] [Google Scholar]
- 47. Larsson AJ, Walker KJ, Raddatz JK, Rotschafer JC. 1996. The concentration-independent effect of monoexponential and biexponential decay in vancomycin concentrations on the killing of Staphylococcus aureus under aerobic and anaerobic conditions. J. Antimicrob. Chemother. 38: 589–597 [DOI] [PubMed] [Google Scholar]
- 48. Lebel MH, et al. 1988. Dexamethasone therapy for bacterial meningitis. Results of two double-blind, placebo-controlled trials. N. Engl. J. Med. 319: 964–971 [DOI] [PubMed] [Google Scholar]
- 49. Letendre SL, Capparelli EV, Ellis RJ, McCutchan JA. 2000. Indinavir population pharmacokinetics in plasma and cerebrospinal fluid. The HIV Neurobehavioral Research Center Group. Antimicrob. Agents Chemother. 44: 2173–2175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Levin VA. 1980. Relationship of octanol/water partition coefficient and molecular weight to rat brain capillary permeability. J. Med. Chem. 23:682–684 [DOI] [PubMed] [Google Scholar]
- 51. Levitz RE, Dudley MN, Quintiliani R, Mullany LD, Nightingale CH. 1984. Cerebrospinal fluid penetration of trimethoprim-sulphamethoxazole in two patients with gram-negative bacillary meningitis. J. Antimicrob. Chemother. 13:400–401 [DOI] [PubMed] [Google Scholar]
- 52. Lode H, et al. 1990. Quinolone pharmacokinetics and metabolism. J. Antimicrob. Chemother. 26(Suppl. B):41–49 [DOI] [PubMed] [Google Scholar]
- 53. Lopez-Cortes LF, et al. 2000. Intravitreal, retinal, and central nervous system foscarnet concentrations after rapid intravenous administration to rabbits. Antimicrob. Agents Chemother. 44:756–759 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Loscher W, Potschka H. 2005. Blood-brain barrier active efflux transporters: ATP-binding cassette gene family. NeuroRx 2:86–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Loscher W, Potschka H. 2005. Role of drug efflux transporters in the brain for drug disposition and treatment of brain diseases. Prog. Neurobiol. 76:22–76 [DOI] [PubMed] [Google Scholar]
- 56. Lycke J, Andersen O, Svennerholm B, Appelgren L, Dahlof C. 1989. Acyclovir concentrations in serum and cerebrospinal fluid at steady state. J. Antimicrob. Chemother. 24:947–954 [DOI] [PubMed] [Google Scholar]
- 57. Lycke J, Malmestrom C, Stahle L. 2003. Acyclovir levels in serum and cerebrospinal fluid after oral administration of valacyclovir. Antimicrob. Agents Chemother. 47:2438–2441 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Maniu CV, Hellinger WC, Chu SY, Palmer R, Alvarez-Elcoro S. 2001. Failure of treatment for chronic Mycobacterium abscessus meningitis despite adequate clarithromycin levels in cerebrospinal fluid. Clin. Infect. Dis. 33:745–748 [DOI] [PubMed] [Google Scholar]
- 59. Markantonis SL, et al. 2009. Penetration of colistin into cerebrospinal fluid. Antimicrob. Agents Chemother. 53:4907–4910 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Martin E, Koup JR, Paravicini U, Stoeckel K. 1984. Pharmacokinetics of ceftriaxone in neonates and infants with meningitis. J. Pediatr. 105:475–481 [DOI] [PubMed] [Google Scholar]
- 61. Matsuda T, et al. 2009. LC method for the determination of meropenem in cerebrospinal fluid: application to therapeutic drug monitoring. Chromatographia 69:1031–1034 [Google Scholar]
- 62. McDowell JA, et al. 1999. Pharmacokinetics of [14C]abacavir, a human immunodeficiency virus type 1 (HIV-1) reverse transcriptase inhibitor, administered in a single oral dose to HIV-1-infected adults: a mass balance study. Antimicrob. Agents Chemother. 43:2855–2861 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. McEvoy GK. 2002. Anti-infective agents: antivirals: acyclovir, p 527–537 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 64. McEvoy GK. 2002. Anti-infective agents: miscellaneous anti-infectives: co-trimoxazole, p 852–861 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 65. McEvoy GK. 2002. Anti-infective agents: miscellaneous anti-infectives: metronidazole, p 864–875 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 66. McEvoy GK. 2002. Anti-infective agents: miscellaneous β-lactam antibiotics: imipenem and cilastin, p 257–267 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 67. McEvoy GK. 2002. Anti-infective agents: penicillins: amoxicillin and clavulanate, p 388–394 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 68. McEvoy GK. 2002. Anti-infective agents: penicillins: cloxacillin, p 360–362 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 69. McEvoy GK. 2002. Anti-infective agents: sulfonamides: sulfadiazine, p 826–827 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 70. McEvoy GK. 2002. Anti-infective agents: cephalosporines: cefepime, p 154–157 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 71. McEvoy GK. 2002. Anti-infective agents: cephalosporines: cefotaxime, p 1170–1180 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 72. McEvoy GK. 2002. Anti-infective agents: cephalosporines: ceftazidime, p 188–198 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 73. McEvoy GK. 2002. Anti-infective agents: cephalosporines: ceftriaxone, p 207–224 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 74. McEvoy GK. 2002. Anti-infective agents: cephalosporines: cephalosporines general statement, p 129–141 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 75. McEvoy GK. 2002. Anti-infective agents: chloramphenicol: chloramphenicol, p 129–141 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 76. McEvoy GK. 2002. Anti-infective agents: penicillins: aminopenicillins general statement, p 369–384 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 77. McEvoy GK. 2002. Anti-infective agents: urinary anti-infectives: trimethoprim, p 841–844 In McEvoy GK. (ed), AHFS drug information. American Society of Health-System Pharmacists, Bethesda, MD [Google Scholar]
- 78. McEvoy GK. 2002. Anti-infective agents: antituberculosis agents: rifampin, p 513–524 In McEvoy GK. (ed), AHFS drug information, Bethesda, MD [Google Scholar]
- 79. McNamara PJ, Trueb V, Stoeckel K. 1990. Ceftriaxone binding to human serum albumin. Indirect displacement by probenecid and diazepam. Biochem. Pharmacol. 40: 1247–1253 [DOI] [PubMed] [Google Scholar]
- 80. Menichetti F, et al. 1996. High-dose fluconazole therapy for cryptococcal meningitis in patients with AIDS. Clin. Infect. Dis. 22: 838–840 [DOI] [PubMed] [Google Scholar]
- 81. Miller DS, Bauer B, Hartz AM. 2008. Modulation of P-glycoprotein at the blood-brain barrier: opportunities to improve central nervous system pharmacotherapy. Pharmacol. Rev. 60: 196–209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Missner A, Pohl P. 2009. 110 years of the Meyer-Overton rule: predicting membrane permeability of gases and other small compounds. ChemPhysChem 10: 1405–1414 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82a. Mrestani Y, El-Mokdad N, Rüttinger HH, Neubert R. 1998. Characterization of partitioning behavior of cephalosporins using microemulsion and micellar electrokinetic chromatography. Electrophoresis 19: 2895–2899 [DOI] [PubMed] [Google Scholar]
- 83. Muller M, et al. 1997. Relationship between serum and free interstitial concentrations of cefodizime and cefpirome in muscle and subcutaneous adipose tissue of healthy volunteers measured by microdialysis. J. Clin. Pharmacol. 37: 1108–1113 [DOI] [PubMed] [Google Scholar]
- 84. Nagata Y, Kusuhara H, Endou H, Sugiyama Y. 2002. Expression and functional characterization of rat organic anion transporter 3 (rOat3) in the choroid plexus. Mol. Pharmacol. 61: 982–988 [DOI] [PubMed] [Google Scholar]
- 85. Nagata Y, Kusuhara H, Hirono S, Endou H, Sugiyama Y. 2004. Carrier-mediated uptake of H2-receptor antagonists by the rat choroid plexus: involvement of rat organic anion transporter 3. Drug Metab. Dispos. 32: 1040–1047 [PubMed] [Google Scholar]
- 86. Nau R, Emrich D, Prange HW. 1993. Inverse correlation between disappearance of intrathecally injected 111In-DTPA from CSF with CSF protein content and CSF-to-serum albumin ratio. J. Neurol.Sci. 115: 102–104 [DOI] [PubMed] [Google Scholar]
- 87. Nau R, et al. 1997. Kinetics of piperacillin and tazobactam in ventricular cerebrospinal fluid of hydrocephalic patients. Antimicrob. Agents Chemother. 41: 987–991 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Nau R, et al. 1994. Kinetics of ofloxacin and its metabolites in cerebrospinal fluid after a single intravenous infusion of 400 milligrams of ofloxacin. Antimicrob. Agents Chemother. 38: 1849–1853 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Nau R, et al. 1998. Disposition and elimination of meropenem in cerebrospinal fluid of hydrocephalic patients with external ventriculostomy. Antimicrob. Agents Chemother. 42: 2012–2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Nau R, et al. 1996. Cerebrospinal fluid ceftazidime kinetics in patients with external ventriculostomies. Antimicrob. Agents Chemother. 40: 763–766 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Nau R, et al. 1990. Penetration of ciprofloxacin into the cerebrospinal fluid of patients with uninflamed meninges. J. Antimicrob. Chemother. 25: 965–973 [DOI] [PubMed] [Google Scholar]
- 92. Nau R, et al. 1992. Penetration of rifampicin into the cerebrospinal fluid of adults with uninflamed meninges. J. Antimicrob. Chemother. 29: 719–724 [DOI] [PubMed] [Google Scholar]
- 93. Nau R, et al. 1993. Passage of cefotaxime and ceftriaxone into cerebrospinal fluid of patients with uninflamed meninges. Antimicrob. Agents Chemother. 37: 1518–1524 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Nau R, et al. 1993. Netilmicin cerebrospinal fluid concentrations after an intravenous infusion of 400 mg in patients without meningeal inflammation. J. Antimicrob. Chemother. 32: 893–896 [DOI] [PubMed] [Google Scholar]
- 95. Nau R, Sorgel F, Eiffert H. 2010. Penetration of drugs through the blood-cerebrospinal fluid/blood-brain barrier for treatment of central nervous system infections. Clin. Microbiol. Rev. 23: 858–883 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Nau R, Sorgel F, Prange HW. 1994. Lipophilicity at pH 7.4 and molecular size govern the entry of the free serum fraction of drugs into the cerebrospinal fluid in humans with uninflamed meninges. J. Neurol. Sci. 122: 61–65 [DOI] [PubMed] [Google Scholar]
- 97. Nau R, Zysk G, Thiel A, Prange HW. 1993. Pharmacokinetic quantification of the exchange of drugs between blood and cerebrospinal fluid in man. Eur. J. Clin. Pharmacol. 45: 469–475 [DOI] [PubMed] [Google Scholar]
- 98. Norrby SR. 1985. Role of cephalosporins in the treatment of bacterial meningitis in adults. Overview with special emphasis on ceftazidime. Am. J. Med. 79: 56–61 [DOI] [PubMed] [Google Scholar]
- 99. Orbach E, Finkelstein A. 1980. The nonelectrolyte permeability of planar lipid bilayer membranes. J. Gen. Physiol. 75: 427–436 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Overton E. 1900. Studien über die Aufnahme der Anilinfarben durch die lebende Zelle. Jahrb. Wiss. Bot. 34: 669–701 [Google Scholar]
- 101. Pardridge WM, Sakiyama R, Fierer G. 1983. Transport of propranolol and lidocaine through the rat blood-brain barrier. Primary role of globulin-bound drug. J. Clin. Invest. 71: 900–908 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Paris MM, et al. 1994. Effect of dexamethasone on therapy of experimental penicillin- and cephalosporin-resistant pneumococcal meningitis. Antimicrob. Agents Chemother. 38: 1320–1324 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Pea F, et al. 2005. Levofloxacin disposition over time in aqueous humor of patients undergoing cataract surgery. Antimicrob. Agents Chemother. 49: 2554–2557 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Pea F, Viale P, Furlanut M. 2005. Antimicrobial therapy in critically ill patients: a review of pathophysiological conditions responsible for altered disposition and pharmacokinetic variability. Clin. Pharmacokinet. 44: 1009–1034 [DOI] [PubMed] [Google Scholar]
- 105. Pfausler B, et al. 2003. Treatment of staphylococcal ventriculitis associated with external cerebrospinal fluid drains: a prospective randomized trial of intravenous compared with intraventricular vancomycin therapy. J. Neurosurg. 98: 1040–1044 [DOI] [PubMed] [Google Scholar]
- 106. Pfausler B, et al. 2004. Concentrations of fosfomycin in the cerebrospinal fluid of neurointensive care patients with ventriculostomy-associated ventriculitis. J. Antimicrob. Chemother. 53: 848–852 [DOI] [PubMed] [Google Scholar]
- 107. Quagliarello VJ, Long WJ, Scheld WM. 1986. Morphologic alterations of the blood-brain barrier with experimental meningitis in the rat. Temporal sequence and role of encapsulation. J. Clin. Invest. 77: 1084–1095 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Raffi F, Taburet AM, Ghaleh B, Huart A, Singlas E. 1993. Penetration of foscarnet into cerebrospinal fluid of AIDS patients. Antimicrob. Agents Chemother. 37: 1777–1780 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Ricard JD, et al. 2007. Levels of vancomycin in cerebrospinal fluid of adult patients receiving adjunctive corticosteroids to treat pneumococcal meningitis: a prospective multicenter observational study. Clin. Infect. Dis. 44: 250–255 [DOI] [PubMed] [Google Scholar]
- 110. Rolinski B, Bogner JR, Sadri I, Wintergerst U, Goebel FD. 1997. Absorption and elimination kinetics of zidovudine in the cerebrospinal fluid in HIV-1-infected patients. J. Acquir. Immune Defic. Syndr. Hum. Retrovirol. 15: 192–197 [DOI] [PubMed] [Google Scholar]
- 111. Roussos N, Karageorgopoulos DE, Samonis G, Falagas ME. 2009. Clinical significance of the pharmacokinetic and pharmacodynamic characteristics of fosfomycin for the treatment of patients with systemic infections. Int. J. Antimicrob. Agents 34: 506–515 [DOI] [PubMed] [Google Scholar]
- 112. Sachs L. 1986. A guide to statistical methods and to the pertinent literature. Springer-Verlag, Berlin, Germany [Google Scholar]
- 113. Saez-Llorens X, et al. 1995. Prospective randomized comparison of cefepime and cefotaxime for treatment of bacterial meningitis in infants and children. Antimicrob. Agents Chemother. 39: 937–940 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Schievink HI, Mattie H, Thomeer RT, Van Strijen E. 1993. The passage of cloxacillin into cerebrospinal fluid in the absence of meningitis. Br. J. Clin. Pharmacol. 36: 57–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Shin SG, et al. 1990. Kinetics of isoniazid transfer into cerebrospinal fluid in patients with tuberculous meningitis. J. Korean Med. Sci. 5: 39–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Sjovall J, Bergdahl S, Movin G, Ogenstad S, Saarimaki M. 1989. Pharmacokinetics of foscarnet and distribution to cerebrospinal fluid after intravenous infusion in patients with human immunodeficiency virus infection. Antimicrob. Agents Chemother. 33: 1023–1031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Slatter JG, et al. 2001. Pharmacokinetics, metabolism, and excretion of linezolid following an oral dose of [(14)C]linezolid to healthy human subjects. Drug Metab. Dispos. 29: 1136–1145 [PubMed] [Google Scholar]
- 118. Sorgel F, Kinzig M. 1993. The chemistry, pharmacokinetics and tissue distribution of piperacillin/tazobactam. J. Antimicrob. Chemother. 31 (Suppl. A): 39–60 [DOI] [PubMed] [Google Scholar]
- 119. Spector R. 1986. Ceftriaxone pharmacokinetics in the central nervous system. J. Pharmacol. Exp. Ther. 236: 380–383 [PubMed] [Google Scholar]
- 120. Spector R. 2010. Nature and consequences of mammalian brain and CSF efflux transporters: four decades of progress. J. Neurochem. 112: 13–23 [DOI] [PubMed] [Google Scholar]
- 121. Stoeckel K, Koup JR. 1984. Pharmacokinetics of ceftriaxone in patients with renal and liver insufficiency and correlations with a physiologic nonlinear protein binding model. Am. J. Med. 77: 26–32 [PubMed] [Google Scholar]
- 122. Stoeckel K, McNamara PJ, Brandt R, Plozza-Nottebrock H, Ziegler WH. 1981. Effects of concentration-dependent plasma protein binding on ceftriaxone kinetics. Clin. Pharmacol. Ther. 29: 650–657 [DOI] [PubMed] [Google Scholar]
- 123. Takayanagui OM, Bonato PS, Dreossi SA, Lanchote VL. 2002. Enantioselective distribution of albendazole metabolites in cerebrospinal fluid of patients with neurocysticercosis. Br. J. Clin. Pharmacol. 54: 125–130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Tarnaris A, et al. 2011. Rostrocaudal dynamics of CSF biomarkers. Neurochem. Res. 36: 528–532 [DOI] [PubMed] [Google Scholar]
- 125. Thea D, Barza M. 1989. Use of antibacterial agents in infections of the central nervous system. Infect. Dis. Clin. North Am. 3: 553–570 [PubMed] [Google Scholar]
- 126. Tucker RM, et al. 1988. Pharmacokinetics of fluconazole in cerebrospinal fluid and serum in human coccidioidal meningitis. Antimicrob. Agents Chemother. 32: 369–373 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Ulldemolins M, Roberts JA, Rello J, Paterson DL, Lipman J. 2011. The effects of hypoalbuminaemia on optimizing antibacterial dosing in critically ill patients. Clin. Pharmacokinet. 50: 99–110 [DOI] [PubMed] [Google Scholar]
- 127a. Valko K, Nunhuck S, Bevan C, Abraham MH, Reynolds DP. 2003. Fast gradient HPLC method to determine compounds binding to human serum albumin. Relationships with octanol/water and immobilized artificial membrane lipophilicity. J. Pharm. Sci. 92: 2236–2248 [DOI] [PubMed] [Google Scholar]
- 128. van de Waterbeemd H, Smith DA, Jones BC. 2001. Lipophilicity in PK design: methyl, ethyl, futile. J. Comput. Aided Mol. Des. 15: 273–286 [DOI] [PubMed] [Google Scholar]
- 129. Vladic A, et al. 2000. Homeostatic role of the active transport in elimination of [3H]benzylpenicillin out of the cerebrospinal fluid system. Life Sci. 67: 2375–2385 [DOI] [PubMed] [Google Scholar]
- 130. Warner JF, Perkins RL, Cordero L. 1979. Metronidazole therapy of anaerobic bacteremia, meningitis, and brain abscess. Arch. Intern. Med. 139: 167–169 [PubMed] [Google Scholar]
- 131. Weisner B, Bernhardt W. 1978. Protein fractions of lumbar, cisternal, and ventricular cerebrospinal fluid. Separate areas of reference. J. Neurol. Sci. 37: 205–214 [DOI] [PubMed] [Google Scholar]
- 132. Wise R. 2003. Maximizing efficacy and reducing the emergence of resistance. J. Antimicrob. Chemother. 51 (Suppl. 1): 37–42 [DOI] [PubMed] [Google Scholar]
- 133. Wolff M, et al. 1987. Penetration of ciprofloxacin into cerebrospinal fluid of patients with bacterial meningitis. Antimicrob. Agents Chemother. 31: 899–902 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Wong VK, et al. 1991. Imipenem/cilastatin treatment of bacterial meningitis in children. Pediatr. Infect. Dis. J. 10: 122–125 [DOI] [PubMed] [Google Scholar]
- 135. Yamazaki K, Kanaoka M. 2004. Computational prediction of the plasma protein-binding percent of diverse pharmaceutical compounds. J. Pharm. Sci. 93: 1480–1494 [DOI] [PubMed] [Google Scholar]
- 136. Yogev R, Kolling WM, Williams T. 1981. Pharmacokinetic comparison of intravenous and oral chloramphenicol in patients with Haemophilus influenzae meningitis. Pediatrics 67: 656–660 [PubMed] [Google Scholar]
- 137. Zatz LM, Jernigan TL, Ahumada AJ., Jr 1982. Changes on computed cranial tomography with aging: intracranial fluid volume. AJNR Am. J. Neuroradiol. 3: 1–11 [PMC free article] [PubMed] [Google Scholar]
- 138. Zettl UK, Lehmitz R, Mix E. 2005. Klinische Liquordiagnostik, vol 2 Walter de Gruyter, Berlin, Germany [Google Scholar]

