You are on page 1of 32

Clin Pharmacokinet (2013) 52:511–542

DOI 10.1007/s40262-013-0062-9

REVIEW ARTICLE

Clinical Pharmacokinetics of Antibacterials in Cerebrospinal


Fluid
Antonello Di Paolo • Giovanni Gori •
Carlo Tascini • Romano Danesi • Mario Del Tacca

Published online: 20 April 2013


Ó Springer International Publishing Switzerland 2013

Abstract In the past 20 years, an increased discrepancy which is greater than 0.8, and for most of these drugs it is
between new available antibacterials and the emergence of near 1. For all drugs that are currently used for the treat-
multidrug-resistant strains has been observed. This condi- ment of CNS infections, the evaluation of pharmacokinetic/
tion concerns physicians involved in the treatment of pharmacodynamic parameters, on the basis of dosing reg-
central nervous system (CNS) infections, for which clinical imens and their time-dependent or concentration-depen-
and microbiological success depends on the rapid dent pattern of bacterial killing, remains an important
achievement of bactericidal concentrations. In order to aspect of clinical investigation and medical practice.
accomplish this aim, the choice of drugs is based on their
disposition toward the cerebrospinal fluid (CSF), which is
influenced by the physicochemical characteristics of anti- 1 Introduction
bacterials. A reduced distribution into CSF has been doc-
umented for beta-lactams, especially cephalosporins and The appropriate treatment of infections of the central ner-
carbapenems, on the basis of their hydrophilic nature. vous system (CNS), namely meningitis, cerebral abscesses,
However, they represent a cornerstone of the majority of and encephalitis, represents a difficult goal of modern
combined therapeutic schemes for their ability to achieve chemotherapy. Antibacterial drugs should rapidly achieve
bactericidal concentrations, especially in the presence of concentrations high enough to exert their killing effect,
inflamed meninges. The good tolerability of beta-lactams which is dependent on their penetration within the CNS.
makes possible high daily dose intensities, which may be However, disposition of antimicrobial agents within the
associated with increased probability of cure. Furthermore, CNS is strongly influenced by several pharmacokinetic
the adoption of continuous infusion seems to be a fruitful factors related to the drug with its own physicochemical
option. Fluoroquinolones, namely moxifloxacin, and anti- properties (i.e., molecular weight, hydrophilic nature,
tuberculosis drugs, together with the agents such as lin- plasma protein binding, etc.) and the pathophysiology of
ezolid, reach the highest CSF/plasma concentration ratio, barriers [both blood–brain and blood–cerebrospinal fluid
(CSF) barriers] surrounding the CNS. Furthermore, the
pharmacokinetic/pharmacodynamic characteristics of che-
A. Di Paolo  R. Danesi
Division of Pharmacology, Department of Clinical and motherapeutic agents (i.e., their time-dependent or con-
Experimental Medicine, University of Pisa, Via Roma 55, 56126 centration-dependent killing pattern) and the bacterial
Pisa, Italy sensitivity to the drugs [i.e., resistant versus sensitive
strains, and minimal inhibitory concentration (MIC)] may
G. Gori  M. Del Tacca (&)
Clinical Pharmacology Centre for Drug Experimentation, Pisa be considered as additional pharmacodynamic factors. As a
University Hospital, Via Roma 67, 56126 Pisa, Italy modern approach to anti-infective chemotherapy, pharma-
e-mail: m.deltacca@med.unipi.it cokinetic data from clinical studies have helped clinicians
to improve drug-dosing regimens, while the same infor-
C. Tascini
Unit of Infectious Diseases, Pisa University Hospital, via mation is allowing the elaboration and adoption of thera-
Paradisa 2, 56124 Pisa, Italy peutic drug monitoring protocols, especially for critically
512 A. Di Paolo et al.

ill patients in the presence of severe or life-threatening 1.1 The Blood Brain and Cerebrospinal Fluid (CSF)
diseases. Barriers
The presence of barriers surrounding the CNS ensures
protection of encephalic and spinal structures against pos- 1.1.1 Morphology of the Blood–Brain Barrier and Blood–
sible damage from chemicals, including drugs. All organ- CSF Barrier (BCSFB)
isms with a well-developed CNS have a blood–brain
barrier (BBB). In the brain and spinal cord of mammals, Although there are several similar features between the
including humans, the BBB is constituted by the endo- BBB and the BCSFB, the cellular basis of these two
thelial cells that form the walls of the capillaries [1] structures as well as their primary functions differ signifi-
(Fig. 1). The combined surface area of these microvessels cantly. The BBB is located in brain capillaries and thus it is
represents the large interface for blood–brain exchange. an endothelial structure with a primary role to protect the
This surface area consists of about 150–200 cm2/g tissue, brain from physiological fluctuations in plasma levels of
giving a total area for exchange in the brain of 12–18 m2 various solutes and from blood-generated substances that
for an adult human [2, 3]. A second interface is formed by could interfere with neurotransmission. However, at the
the epithelial cells of the choroid plexus facing the CSF, same time it acts to provide mechanisms for exchange of
which constitute the blood–CSF barrier (BCSFB). The CSF nutrients, metabolic waste products, signaling molecules,
is secreted across the choroid plexus epithelial cells into the and ions between the blood and the brain. By contrast, the
brain ventricular system [3, 4], while the remainder of the BCSFB consists of a layer of modified cuboidal epithelium
brain extracellular fluid, the interstitial one, is derived, at and the choroid plexus that secretes CSF and this process
least in part, by secretion across the capillary endothelium could be considered as the main function of this epithe-
of the BBB [5, 6]. lium. The differences concerning the principal functions
The third interface is represented by the avascular are derived from variations in morphology and molecular
arachnoid epithelium, underlying the dura mater, and biology [8].
completely enclosing the CNS (Fig. 1). This completes the Brain endothelial cells and choroid plexus epithelial
seal between the extracellular fluids of the CNS and the rest cells are connected at a junctional complex by tight junc-
of the body [7]. tions and adherens junctions (Fig. 2) [7, 9, 10]. Brain
At all three interfaces, the barrier function results from a endothelial cells also express gap junctions, but their
combination of a physical barrier (the tight junctions functional significance is not clear.
between cells reduce the flux through the intercellular cleft All tight junctions and adherens junctions are composed
of paracellular pathway), a transport barrier (specific of transmembrane proteins and cytoplasmic plaque pro-
transport mechanisms mediating solute flux), and a meta- teins. Plaque proteins cluster integral tight junction proteins
bolic barrier (enzymes metabolizing molecules in transit). and form a platform for interaction with scaffolding and
The barrier function is not fixed, but can be modulated both signaling proteins. In addition, a circumferential actin belt
under physiological and pathological conditions [3, 7, 8]. that encircles each endothelial/epithelial cell at the level of
Based on this premise, the two barriers (BBB and BCSFB) tight junctions is important for the formation and the nor-
must be regarded not only as anatomical barriers, but also as mal function of tight junctions [8].
dynamic structures that express multiple transporters, recep-
tors, and enzymes. The two main functions of these barriers 1.1.2 Physiology of BCSFB Barrier
are to inhibit free diffusion between brain fluids and blood, as
well as to provide transport processes for essential nutrients, At the end of the nineteenth century, Paul Ehrlich demon-
ions, metabolic waste products, and drugs. strated for the first time the existence of the blood–brain/
The aim of the present review is to describe the CSF blood–CSF barrier by injecting aniline dye into the blood of
disposition of antibacterial drugs currently employed for preclinical models, observing that all the organs, with the
the treatment of CNS infections, with special attention to exception of the brain, were stained [11, 12]. At the
the pharmacokinetic/pharmacodynamic characteristics of beginning of the twentieth century, Edwin Goldmann, a
these agents, particularly in the clinical management of pupil of Ehrlich, injected trypan blue both intravenously
bacterial meningitis. and subcutaneously in experimental animals. Trypan blue
PubMed database was reviewed up to January 2013 stained both the choroid plexus and the dura mater without
using the following keywords: antibacterials, pharmacoki- entering the CSF. On the contrary, after direct injection of
netics, CSF, pharmacokinetic(s)/pharmacodynamic(s). trypan blue into the CSF, the brain and spinal cord were
Moreover, names of antibacterials (i.e., moxifloxacin, stained, indicating the absence of a tight diffusional barrier
aminoglycosides, etc.) were included within the keywords between the CSF and brain tissues [12, 13]. Large hydro-
and references of retrieved papers were further examined. philic molecules, which cannot penetrate tight junctions and
Pharmacokinetics of Antibacterials in CSF 513

Fig. 1 Sites of the blood–brain barrier (BBB). The BBB includes fluid. Reproduced from Taylor [1] with permission from Springer
tight junctions between: a the endothelial cells of brain capillaries; International Publishing AG (ÓAdis Data Information BV 2002. All
b epithelial cells of the choroid plexus; and c epithelial cells of the rights reserved)
arachnoid membrane. ECF extracellular fluid, CSF cerebrospinal

cell membranes, enter the interstitial space of the brain and 1.1.3 Meningeal Inflammation
the CSF. The surface area of brain capillaries without tight
junctions is approximately 1/5,000 of the total capillary Penetration of antimicrobial agents into the CSF depends
surface area in the brain. The morphological equivalent of on numerous factors, including their serum protein binding,
the blood–CSF barrier consists of the cylindrical epithelium molecular size, lipid solubility, and the degree of local
of the choroid plexus linked by tight junctions [12, 14]. inflammation that could affect BBB permeability. Some
This evidence indicates that the central nervous com- molecules with appropriate lipophilicity, small molecular
partments are bordered by a diffusional barrier consisting weight, and suitable charge could well diffuse from blood
of at least one lipid membrane with the exception of a few into the CNS. However, the overwhelming majority of
leaky regions allowing even large hydrophilic molecules, small molecules (500 daltons), proteins, and peptides do
such as immunoglobulins, to enter the extracellular space not cross the BBB by diffusion. The BBB permeability
of the brain and the CSF. Several compounds with low alterations in the presence of meningeal inflammation
molecular weight are cleaved enzymatically at the blood– could affect CNS and tissue antimicrobial drug concen-
brain/blood–CSF barrier [15]. By contrast, several active trations in a more doubtful manner. Under the conditions of
drug efflux transporters contribute markedly to the function a severe disruption of the blood–brain/blood–CSF barrier,
of the blood–brain/blood–CSF barrier [16, 17]. the physicochemical properties of drugs, which govern
514 A. Di Paolo et al.

Fig. 2 Various transport processes occurring at the blood–brain adherens junctions. Occludin and claudins are the most important
barrier (BBB) and schematic diagram of the endothelial cells that membranous components and are represented with their four trans-
form the BBB and main routes for molecular traffic across the BBB membrane domains and two extracellular loops. In the figure,
are shown. a Water-soluble compounds, including polar drugs, are vascular endothelial cadherin (VE-cadherin), the principal molecule
usually unable to reach astrocytes through tight junction restriction, of endothelial adherens junctions, and the platelet–endothelial cell
while the lipid membranes of the endothelium offer an effective adhesion molecule (PECAM), which mediates homophilic adhesion,
diffusive route for lipid-soluble agents. Ion channels, transport are also shown. Primary linker molecules between adherens junctions
carriers (e.g., for glucose, amino acids, purine bases, nucleosides, and the cytoskeleton are represented by catenine and actin filaments.
choline) or energy-dependent proteins (e.g., P-glycoprotein) and Modified and reprinted by permission from Macmillan Publishers
receptor-mediated transporters (for insulin, transferrin) are also Ltd: [Nat Rev Neurosci.] (Abbott et al. [7], copyright (2010)
represented. b Molecular composition of endothelial tight and

drug entry into the CNS in the absence of meningeal Given that meningitis is characterized by an inflamma-
inflammation, become a secondary aspect [12]. tory process, it could be reasonable to expect that the BBB
Usually, drug concentrations measured in the absence of disruption could significantly jeopardize the pharmacoki-
meningeal inflammation represent the minimum concen- netic/pharmacodynamic features of an antimicrobial agent.
trations that can be encountered early in the course of a Actually, the knowledge of CSF penetration of an antibi-
CNS infection, when the whole BBB morpho-physiologi- otic across inflamed meninges is still unclear. Meningeal
cal structure is still uninjured. In the case of inflamed inflammation may be moderate and even absent in the early
meninges, the BBB could become leaky by the opening of stage of a meningitis when antibiotic penetration into the
intercellular tight junctions [18]. Moreover, the CSF out- CSF is extremely important to inhibit the progress of the
flow resistance increases, leading to a moderate reduction disease. Moreover, immunocompromised patients may
of the CSF production and absorption rates [19]. The have little or no inflammatory response throughout their
activity of P-glycoprotein, a transmembrane transporter entire course of infection [22]. Furthermore, a subject
that determines cellular drug efflux, can also be modulated affected by meningitis usually represents a critically ill
by proinflammatory cytokines, thus reducing the antibac- patient at risk of sepsis. The pathophysiological changes
terial drug efflux from the endothelial cells of the BBB and occurring during sepsis, such as alterations in plasma
from the neuronal cells [20, 21]. All these mechanisms proteins, renal perfusion, capillary permeability, and organ
synergistically lead to a rise of the CSF concentrations of failure could lead to modification of the pharmacokinetic
the drugs, particularly those that do not enter the CSF properties of antimicrobials, because their volume of dis-
readily in the absence of meningeal inflammation [12]. tribution and clearance could be affected [23]. During a
Pharmacokinetics of Antibacterials in CSF 515

Table 1 CSF penetration of


Antimicrobial class/ CSF/plasma AUC ratioa Inflammation/
antimicrobial drugs according to
compound noninflammation ratiob
meningeal status (adapted from Normal/mildly affected Inflamed
[12, 154]) meninges meninges

Penicillins 0.02 0.2 10


Beta-lactamase 0.07 0.1 1.4
inhibitors
Cephalosporins 0.007–0.1 0.15 21–1.5
Carbapenems 0.2 0.3 1.5
Fluoroquinolones 0.3–0.7 0.7–0.9 2.3–1.3
Chloramphenicol 0.6–0.7 0.6–0.7 1
Tetracyclines 0.2 0.2 1
Tigecycline n.a. 0.5 n.c.
Trimethoprim 0.18 0.42–0.51 2.8–2.3
Sulfamethoxazole 0.12 0.24–0.30 2–2.5
Glycopeptides 0.14–0.18 0.30 2.1–1.6
AUC area under the Macrolides n.a. 0.18 n.c.
concentration-time curve, CSF Aminoglycosides 0.2 0.1 n.c.
cerebrospinal fluid, n.a. not Fosfomycin 0.18 n.a. n.c.
available, n.c. not computable
a
Linezolid 0.5–0.9 0.7 n.c.
Estimated CSF penetration
Daptomycin n.a. 0.05 n.c.
ratio based on the arithmetic
mean of available data Colistin 0.03–0.05 0.06–0.68 0.5–0.07
b
Ratio between CSF/plasma Metronidazole n.a. 0.87 n.c.
AUC calculated in the presence/ Rifamycins 0.22 0.3 n.c.
absence of meningeal Antitubercular drugs n.a. 0.86 n.c.
inflammation

septic process, an increased volume of distribution could infection, whereas in the case of brain abscesses anaerobic
decrease peak serum concentrations of aminoglycosides, bacilli may play a role. The epidemiology of meningitis has
beta-lactams, and carbapenems, thus requiring a dose been changed in the recent past because of the introduction
adjustment in order to reach active plasma and/or CSF of routine vaccination of children against Haemophilus
concentrations [24, 25]. influenzae and some strains of Streptococcus pneumoniae.
For a better analysis of the permeability of antibacterials Despite these efforts, S. pneumoniae still represents the
into the CNS with or without meningeal inflammation, a major cause of the most severe diseases at all ages, with an
summary of data is presented in Table 1. increasing incidence of antibiotic-resistant strains, espe-
cially those resistant to penicillin [26, 27]. Neisseria
1.2 Infections of CNS: The Most Common Pathogens meningitidis, Group B Streptococcus, H. influenzae, and
and Drugs of Choice Listeria monocytogenes are other microorganisms respon-
sible for community-acquired infections, sometimes in the
A wide variety of bacterial species is responsible for CNS presence of risk factors (i.e., immunocompromised
infections, including meningitis and abscesses (Table 2), patient). Nosocomial infections are characterized by a
which may render difficult the early adoption of the most different epidemiology, depending on the adoption of
appropriate chemotherapy regimen. For this reason, the required neurosurgical procedures and devices (i.e., ven-
choice of empiric combination treatments against most tricular drainages). In these cases, Gram-negative bacteria,
bacterial infections is the first step toward therapeutic including Escherichia coli, Klebsiella spp., Pseudomonas
success when the identification of bacterial species is not aeruginosa, Enterobacter spp., and Acinetobacter spp.,
yet available. Then, a specific chemotherapy is chosen in may be identified, as well as staphylococci and streptococci
order to increase the probability of an early cure other than S. pneumoniae. Chronic meningitis displays a
attainment. different pattern of causative microorganism, with Myco-
In the case of meningitis, Gram-positive and Gram- bacterium tuberculosis one of the major causes, while
negative bacterial strains may be responsible for the Brucella, Treponema, and Borrelia spp. may represent
516 A. Di Paolo et al.

Table 2 Common bacterial pathogens responsible for CNS infections and current recommended antimicrobial treatment (adapted from [31, 115,
213])
Disease Bacterial pathogens Antimicrobial treatments

Abscess Primary Aerobic and anaerobic bacilli Cefotaxime1/ceftriaxone ? metronidazole


Postsurgery S. aureus, ConS, Enterobacteriaceae, Cefotaxime1/ceftriaxone ? nafcillin/oxacillin/
Haemophilus spp., Fusobacterium spp. vancomycin*,2
Nocardia spp. Trimethoprim-sulfamethoxazole ? ceftriaxone
Acute meningitis
Empiric Preterm–1 month S. agalactiae, E. coli, L. monocytogenes Ampicillin3 ? cefotaxime
therapy 1–3 months As above plus S. pneumoniae, Ampicillin3 ? vancomycin4,5 ? ceftriaxone/cefotaxime
N. menigitidis, H. influenzae
3 months–50 years S. pneumoniae, N. meningitidis, Vancomycin4,5 ? ceftriaxone/cefotaxime
S. agalactiae, H. influenzae, E. coli
Immunocompromised S. pneumoniae, L. monocytogenes, Vancomycin4 ? (ampicillin3/cefotaxime/ceftriaxone or
H. influenzae meropenem)
Postsurgery/infected S. aureus, ConS, Enterobacteriaceae, Vancomycin2,6 ? (cefepime/ceftazidime or meropenem)
shunt Haemophilus spp., Streptococcus spp.,
P. aeruginosa, Clostridium spp.
Infection due to: S. pneumoniae Ceftriaxone/cefotaxime ? vancomycin**
N. meningitidis Ceftriaxone/cefotaxime
L. monocytogenes (Ampicillin ? gentamicin) or trimethoprim-
sulfamethoxazole or meropenem
H. influenzae, Coliforms, P. aeruginosa Ceftriaxone/cefotaxime ? gentamicin
Specific H. influenzae Ampicillin or ceftriaxone/cefotaxime or cefepime or
therapy fluoroquinolones or chloramphenicol
L. monocytogenes (Ampicillin ? gentamicin/trimethoprim-
sulfamethoxazole7) or meropenem or
(linezolid ? rifampicin)
N. meningitidis Penicillin G/ampicillin or cefotaxime/ceftriaxone or
meropenem or moxifloxacin
S. pneumoniae MIC [0.1 Penicillin G or ampicillin or ceftriaxone or cefotaxime
MIC 0.1–1 Ceftriaxone/cefotaxime or cefepime or meropenem
MIC [2 (Vancomycin ? ceftriaxone/cefotaxime) or
fluoroquinolones
MICceftriaxone [1 (Vancomycin ? ceftriaxone/cefotaxime) or
fluoroquinolones
E. coli Cefepime/ceftazidime or aztreonam or meropenem or
ciprofloxacin
P. aeruginosa Cefepime/ceftazidime or aztreonam
Prophylaxis N. meningitidis, H. influenzae Rifampicin8,9/ceftriaxone10/ciprofloxacin11/azithromycin12
Chronic meningitis
Specific M. tuberculosis Isoniazid ? rifampicin13 ? ethambutol ? pyrazinamide
therapy Brucella spp. Gentamicin ? (doxycycline/ trimethoprim-
sulfamethoxazole14)***
T. pallidum Penicillin G
Borrelia spp. Ceftriaxone
Pharmacokinetics of Antibacterials in CSF 517

Table 2 continued
Disease Bacterial pathogens Antimicrobial treatments

Encephalitis# M. pneumonia Azithromycin, doxycycline, fluoroquinolones


B. bacilliformis Chloramphenicol, ciprofloxacin, doxycycline, ampicillin,
or trimethoprim-sulfamethoxazole
B. henselae Doxycycline or azithromycin, with or without rifampicin
L. monocytogenes Ampicillin plus gentamicin; trimethoprim-
sulfamethoxazole
T. whipplei Ceftriaxone, followed by either trimethoprim-
sulfamethoxazole or cefixime
M. tuberculosis 4-Drug antituberculous therapy should be initiated;
dexamethasone should be added in patients with
meningitis
B. burgdorferi Ceftriaxone, cefotaxime, or penicillin G
R. rickettsii Doxycycline; chloramphenicol can be considered an
alternative
C. burnetii Doxycycline plus a fluoroquinolone plus rifampicin
A. phagocytophilum Doxycycline
E. chaffeensis Doxycycline
T. pallidum Penicillin G; ceftriaxone
ConS coagulase-negative staphylococci, MIC minimum inhibitory concentration
* In case of methicillin-resistant S. aureus
** plus dexamethasone
*** age: \8 years/[8 years. Recommended dosages of antimicrobials: penicillin G, 2 g intravenously every 4 h; ampicillin, 2 g intravenously
every 4 h, 50 mg/kg intravenously every 6/12 h
#
No recommended dosages are reported by the authors [213]
1
200 mg/kg per day in 3–4 doses
ceftriaxone, 2 g intravenously every 12 h, 100 mg/kg per day in two doses, 250 mg intramuscularly
2
500–750 mg intravenously every 6 h
3
nafcillin and oxacillin, 2 g intravenously every 4 h; cefotaxime, 2 g intravenously every 6 or 4 h
4
15 mg/kg intravenously every 6 h
5
azythromycin, 500 mg orally
6
or every 12 h
7
5/25 mg/kg orally every 12 h
8
or 600 mg every 24 h for 4 days
9
10 mg/kg intravenously or orally every 24 h (up to 600 mg/day)
10
ceftazidime and cefepime, 2 g intravenously every 8 h
meropenem, 2 g intravenously every 8 h; vancomycin, 1 g intravenously every 6 h
11
moxifloxacin, 400 mg orally every 24 h
trimethoprim/sulfamethoxazole, 15/75 mg/kg per day in 2–4 doses, 5 mg/kg intravenously every 6–8 h
12
gentamicin, 2 mg/kg intravenously loading dose, then 1.7 mg/kg intravenously every 8 h; ciprofloxacin, 400 mg intravenously every 8–12 h,
500 mg orally
13
isoniazid, 10 mg/kg every 24 h (up to 300 mg/day)
ethambutol, 25 mg/kg every 24 h
pyrazinamide, 25 mg/kg every 24 h (up to 2.5 g/day)
metronidazole, 7.5 mg/kg intravenously every 6 h
linezolid, 600 mg intravenously every 12 h
chloramphenicol, 1–1.5 g intravenously every 6 h
14
doxycycline, 100 mg orally every 12 h
rifampicin, 600 mg every 12–24 h, 20 mg/kg orally every 24 h
518 A. Di Paolo et al.

further causes of infection. Among the bacteria responsible During antimicrobial treatment of bacterial meningitis,
for meningitis, L. monocytogenes may also be a cause of an inflammatory response to antibiotics (Jarisch-Herxhei-
encephalitis [28]. mer reaction) can occur, particularly in the case of spiro-
In order to start antibiotic therapy as soon as possible, chetal infections, especially Treponema pallidum, when
the empiric treatment should begin before identification of antibacterial concentrations are sustained [38]. This reac-
the causative microorganism. Drugs used for empiric tion may be the result of both an endotoxin-like bacterial
therapy include beta-lactams (penicillins, third- and fourth- substance release as well as cytokine elevation, but the
generation cephalosporins, carbapenems), glycopeptides exact mechanism behind the Jarisch-Herxheimer reaction
(vancomycin), and other agents [gentamicin, trimethoprim/ is still unknown, so that possible preventative measures are
sulfamethoxazole, fluoroquinolones, linezolid, rifampicin lacking.
(rifampin)]. The same agents should then be administered In the Taiwan prospective observational study on
as a specific therapy for the time required to obtain clinical patients affected by the Jarisch-Herxheimer reaction,
and microbiological response when the causative pathogen multivariate analysis showed that risk factors for its
has been identified. Finally, some drugs like metronidazole, development include high rapid plasma reagin titers, early
doxycycline, and colistin have a very narrow use in the syphilis, and prior penicillin exposure [39]. A randomized,
presence of a limited number of susceptible bacterial controlled trial, performed to prevent Jarisch-Herxheimer
strains, while they may be considered as alternative choices reaction, showed that pretreatment with sheep anti-TNF-
in the case of multi-drug-resistant bacteria [29, 30]. alpha antigen-binding fragment (Fab) suppressed the
The use of adjunctive corticosteroid therapy is another reaction that can occur after penicillin treatment for louse-
aspect that should be evaluated. Adjuvant dexamethasone borne relapsing fever and reduced the associated increase
is recommended with or shortly before the first intravenous in plasma concentrations of interleukin-6 and interleukin-8
dose of antibiotic in both immunocompetent and immu- [40]. In some cases, pharmacological treatment with cor-
nosuppressed adults with pneumococcal meningitis at a ticosteroids showed a significant role in the prevention [41]
dose of 10 mg every 6 h for 4 days and in children at a or in the limitation [42] of the systemic effects caused by
dose of 0.15 mg/kg every 6 h for 4 days for H. influenzae the Jarisch-Herxheimer response.
B and pneumococcal meningitis [31].
The results of a European controlled trial demonstrated 1.3 Key Issues of Antibacterials Within CNS
that adjunctive dexamethasone, given before or with the
first dose of antibiotic therapy, was associated with a slight The study of pharmacokinetics of antibacterial agents
reduction in mortality [relative risk (RR) 0.48, 95 % CI within the CNS represents an important area of research for
0.24–0.96] [32]. However, the last Cochrane meta-analysis, possible translation of results into clinical practice, with the
performed on 4,041 patients affected by bacterial menin- aim to improve treatment efficacy and to reduce the
gitis to evaluate the effect of adjuvant corticosteroid ther- development of mutant strains. For these reasons, the early
apy versus placebo, showed that corticosteroids were achievement of bactericidal concentrations of drugs
associated with lower rates of severe hearing loss (RR 0.67, appears as an urgent requirement in modern antimicrobial
95 % CI 0.51–0.88), any hearing loss (RR 0.76, 95 % CI chemotherapy. However, while reading and discussing
0.64–0.89), and low rates of neurological sequelae (RR results obtained in several studies aimed at evaluating
0.83, 95 % CI 0.69–1.00) with no effect on mortality (RR pharmacokinetic and pharmacodynamic characteristics of
0.74, 95 % CI 0.53–1.05, p = 0.09) [33]. antibacterials, it is likely that comparison of data is
By reducing subarachnoid space inflammation and difficult.
BBB permeability, corticosteroids could decrease CSF
penetration of antibiotics; thus, patients receiving vanco- 1.3.1 Pharmacokinetics of Antibacterials in CSF
mycin for penicillin-resistant pneumococcal meningitis
require close clinical and CSF drug concentration moni- The antibacterial drug distribution into the CSF depends on
toring [31]. Some authors, however, reported that con- several factors that characterize the biological matrix (i.e.,
comitant dexamethasone infusion did not affect ventricular or lumbar CSF, brain abscess) and its features
vancomycin [34] or ceftriaxone [35, 36] CSF concentra- (protein content, pH, presence of leukocytes), and on the
tion in infected patients (Table 3). barriers (inflamed or uninflamed meninges, the use of
Clinical data concerning teicoplanin–steroid interaction corticosteroids) and the pharmacological properties of each
are lacking. When administered in experimental animal drug, along with its physicochemical characteristics,
models with dexamethasone, teicoplanin resulted in lower plasma protein binding, etc. The following discussion
CSF concentrations in the face of the single agent admin- examines some of these aspects, while Sect. 2 discusses the
istration, but not in pharmacological failures [37]. pharmacokinetics of each class of drugs.
Table 3 Penetration of antibacterials in CSF following standard dosage regimens
Drug Daily dose Regimen1 Patients Age2 (years) Way of Inflamed Serum4 Cmax (mg/l)5 tmax (h)5 AUCCSF AUC ratio6 Note Reference
sampling3 meninges (mg  h/l)5

Amoxicillin 50 mg/kg IV, 0.5 21 adults 14–76 L Yes Free 2.53 ± 0.59 &2 6.21° 0.06° AUC4 51a
Potassium 5 mg/kg IV, 0.5 21 adults 14–76 L Yes Total 0.27 ± 0.04 &2 0.84° 0.084° AUC4 51
clavulanate
Cefepime 2g IV, 0.5 7h ? 2i 41–79 V No Free 7.4, 15.8i 1, 2i – 0.08h AUC?g 92b, 96c
adults
Ceftriaxone 2g IV, 0.5, S 7 adults 65–90 V No Total 0.18–1.04 1–16 5.8–41.3 0.006–0.018 AUC? 80d
e
Ceftriaxone 75 mg/kg* IV, 0.5, M 30 0.08–16.5 L, V Yes Total 7.2/2.5 6 – – – 97e
children
Cefotaxime 2g IV, 0.5, S 7 adults 60–90 V No Total 0.14 ± 1.81 0.5–8 3.6–16.3 0.03–0.21 80d
Pharmacokinetics of Antibacterials in CSF

AUC?
Ceftazidime 3g IV, 0.5, S 8 adults 45–65 V No Free – – 0.02–0.08 AUC? 66f
Meropenem 2g IV, 0.5, M 10 adults 48–75 V No Total 0.63 ± 0.50 4.1 ± 2.6 8.55 ±6.73 0.047 ± 0.022 AUC? 53g
Amikacin 7.5 mg/kg IV, 0.5, M 16 0.33–8 L Yes Total 1.65 ± 1.6 3 – – – 102h
children
Moxifloxacin 400 mg OS, –, S 50 adults 66.5 ± 10.6 L No Total 4.07 ± 1.15 4–6 – – – 105i
Moxifloxacin 400 mg OS, –, M 4 adults 30–65* L Yes Total 1.29 4 (3.8–5.5) 18.1 0.82 AUC24 104j
(1.04–1.50) (14.8–19.8) (0.70–0.94)
Moxifloxacin 800 mg OS, –, M 4 adults 30–65* L Yes Total 2.22 4 (3.8–5.5) 28.4 0.71 AUC24 104j
(1.72–2.63) (24.9–28.9) (0.58–0.84)
Ciprofloxacin 500 mg 9 2 OS, –, M 2 adults 17–75 L Yes Total – – 6.97 0.31 AUC24 109k
(4.45, 9.49) (0.31, 0.31)
Ciprofloxacin 750 mg 9 2 OS, –, M 13 adults 17–75 L Yes Total – – 6.95 0.26 AUC24 109k
(2.30, 14.9) (0.11, 0.77)
Gatifloxacin 400 mg OS, –, M 14 adults 16–70 L Yes Total – – 20.0 0.48 AUC24 109k
(16.5, 41.5) (0.47, 0.50)
Levofloxacin 500 mg 9 2 OS, –, M 14 adults 18–68 L Yes Total – – 94.1 0.74 AUC24 109k
(53.1, 208) (0.58, 1.03)
Levofloxacin 500 mg 9 2 IV, 1, M 10 adults 26–74 V No Total 4.06 ± 1.26 2.70 ± 1.32 33.42 ± 10.69 0.71 ± 0.07 AUC12 110l
Colistin 2.61 mg/ IVen, 9 adults 18–73 V – – 6.2–22.1 2.4–5.5 72.5–293.3 – AUC24 55m
24 h– bolus, M
5.22 mg/
12 h
Tigecycline 100 mg IV, 0.5, S 17 adults 43 ± 13 L No Total 0.46 0.12 AUC24 113n
Linezolid 600 mg 9 2 IV, 1, M 9 adults 58.7 ± 17.3* V No Total 10.8 ± 5.7 2.6 ± 0.9 101.6 ± 59.6 0.7 ± 0.1 AUC24 160p
519
Table 3 continued
520

Drug Daily dose Regimen1 Patients Age2 (years) Way of Inflamed Serum4 Cmax (mg/l)5 tmax (h)5 AUCCSF AUC ratio6 Note Reference
sampling3 meninges (mg  h/l)5

Daptomycin 10 mg/kg IV, 1, S 6 adults 26–67 L, V Yes Total – – 1.01–19.35 0.001–0.017, AUC24 164q
0.11*
1
Route of administration (IV, intravenous OS, oral IVen, intraventricular), duration of infusion (h), single (S) or multiple doses (M)
2
range (minimum–maximum) or mean ± standard deviation
3
L lumbar, V ventricular
4
drug concentrations in serum were measured as total or free, unbound fraction
5
pharmacokinetic parameters of drugs within CSF: Cmax, tmax, and AUC, maximum concentration, time to Cmax and area under the concentration-time curve, respectively
6
CSF/serum AUC ratio
a
sampling was performed 48–96 h after starting the therapy while maximum daily dose was 2 g, results are expressed as mean values (°)
b
Monte Carlo simulation
c
Cmax and tmax values in two patients
d
values are expressed as range (minimum–maximum)
e
drug concentration 6 h postdose, at the beginning/end of infectious disease while maximum daily dose was 2 g
f
Monte Carlo simulation, drug concentrations in plasma were corrected for plasma protein binding
g
results are expressed as mean ± standard deviation
h
CSF was obtained 3 h postdosing at putative tmax value, while Cmax values are expressed as mean ± standard deviation
i
results are expressed as mean ± standard deviation
j
abnormal mononuclear cell count (but not glucose and protein concentrations) in CSF was observed, while results are expressed as median value (interquartile range)
k
population/pharmacokinetic analysis on sparse sampling time points, results are expressed as mean value (minimum–maximum)
l
results are expressed as mean ± standard deviation
m
colistimethate sodium was administered and results are expressed as range (minimum–maximum)
n
results are expressed as mean values
p
results are expressed as mean ± standard deviation
q
results are expressed as range (minimum–maximum), while the free fraction of the drug in plasma was considered for calculation (*)
A. Di Paolo et al.
Pharmacokinetics of Antibacterials in CSF 521

the CSF remains questionable, because it may occur in


purulent CSF [50].
Another factor responsible for result variability is the
delay of the time profile of antimicrobial drug concentra-
tions in the CSF with respect to plasma (the hysteresis
phenomenon), a delay which could be smallest when the
CSF is sampled through an EVD (according to the rate of
drug diffusion through barriers), and highest when using a
lumbar puncture. In this manner, when CSF is harvested
through a lumbar puncture, the calculation of CSF/plasma
concentration ratio at the same time point after drug
administration does not completely reflect the real dispo-
sition of the drug within the CNS [51], or at least within the
upper part of CSF spaces (i.e., ventricles). The hydrophilic/
Fig. 3 Plot of concentrations in serum and ventricular cerebrospinal lipophilic properties of drugs may concur to the above-
fluid (CSF) of linezolid 600 mg given intravenously twice daily
infused over 1 h (open circles) [52], intravenous levofloxacin 500 mg
described hysteresis phenomenon also within ventricular
twice a day (filled squares) [110], 2 g intravenous meropenem infused CSF when passage of drugs through the BBB is delayed by
over 30 min (open triangles) [53], 3 g intravenous ceftazidime poor lipid solubility, as in the case of beta-lactams (Fig. 3).
infused over 30 min (filled circles) [212]. Points of each plot For these reasons, the assessment of drug disposition
represent the CSF/plasma concentration ratio at different times after
dosing (i.e., numbers next to symbols represent the time of sampling
into ventricular CSF has the dual advantage of making
expressed in hours after the end of drug administration). For each feasible both a dense sampling scheme [52] and the sub-
drug, the variable ratio is due to the delay of drug entry into CSF (the sequent calculation of area under the concentration–time
hysteresis phenomenon). Furthermore, the high and more rapid curve (AUC) ratios between CSF and plasma. Use of AUC
penetration of antibacterials within the central nervous system
characterizes the drugs (namely linezolid and levofloxacin) whose
ratios is superior to use of the CSF/plasma ratio at a single
curve lies on the left part of the graph. For clarity, dashed lines time point, and can be more definitive for evaluation of
represent the CSF/serum ratio of 1 (on the left) and 0.1 (on the right) drug disposition into CSF. However, the presence of an
EVD is not a common condition, because it depends on the
In the case of bacterial meningitis, CSF should be clinical status and management of patients; therefore,
considered as a suitable matrix to investigate drug dispo- several studies have adopted lumbar puncture to obtain
sition and pharmacokinetics of antibacterial agents, at CSF [53, 54]. Other strategies to improve the pharmaco-
variance with that occurring in the cerebral tissue infection. kinetic analysis of drugs within CSF are external lumbar
In this case, the time profile of CSF concentrations may not drainage [54] and the application of population pharma-
reflect the antibacterial drug diffusion within the infected cokinetic modeling. The latter approach ensures a complete
tissue, because several factors (i.e., extension of the lesion, pharmacokinetic analysis on the basis of a more feasible,
increased interstitial pressure, changes in pH, etc.) may sparse sampling protocol, instead of a dense one [55]. A
influence tissue penetration of the drugs [43]. Furthermore, possible drawback in these studies occurs when drug
the availability of cerebral tissue is restricted to particular administration through the EVD is adopted to obtain CSF
events and clinical cases (i.e., when a surgical curettage drug concentrations that are not achievable after extra-
or drainage is required) and published data are limited ventricular administration. This strategy may influence the
[44–47]. pharmacokinetics of antibacterials, inasmuch that the drain
One of the most important factors, responsible for the is closed for a variable length of time (i.e., from 1 up to
variability among the studies, could be the method of CSF 3 h) after drug administration, hence affecting drug clear-
sample collection. CSF undergoes a concentration process ance. On the other hand, the volume of CSF drained from
as it passes along ventricles and the spinal cord mainly due the EVD before drug administration may play a consider-
to the extracellular fluid of nervous tissue together with able role in interpatient variability of drug pharmacoki-
CSF diffusing from meninges and produced by the choroid netics [56].
plexus [48]. Therefore, an increase in drug concentration From a general point of view, studies that deal with the
should be expected when a lumbar puncture is chosen to distribution of antibacterials from systemic circulation into
collect CSF instead of using an external ventricular drain the CSF should only consider the free fraction of drugs in
(EVD). However, another consequence of this process is a plasma, which is responsible for the pharmacological effect
significant ventricular-lumbar increase in protein and in tissues. However, in many studies the laboratory end-
albumin [49], but whether this condition could have a point is represented by the measurement of total plasma
significant influence on the free fraction of the drug within concentration, whereas, in a limited number of studies,
522 A. Di Paolo et al.

drug concentrations are measured in plasma ultrafiltrate can be easily accessed and investigated, in contrast with CSF
[57]; otherwise, the plasma protein binding percentage is or cerebral tissue. Therefore, a major question to arise is
considered a posteriori [52]. whether antibacterials maintain their plasma pharmacoki-
Several factors may significantly influence the diffusion netic/pharmacodynamic characteristics (i.e., time-dependent
of drugs from plasma into CSF. During bacterial menin- or concentration-dependent killing effect) in other compart-
gitis, the reduction of CSF pH from 7.3 to 7.0 (or lower) ments such as CSF. In addition to this, because the treatment
may increase the diffusion of weak acid antibacterials, of CNS infections requires the attainment of bactericidal
namely penicillins and cephalosporins, from CNS com- concentrations, the minimal bactericidal concentration
partments to blood [58]. In addition, the increased protein (MBC) should be considered rather than the MIC value.
content in purulent CSF may reduce the free portion of Furthermore, in some studies there was no distinction
antibacterials [59, 60]. These two factors are potentially between MIC and MBC, and the maximal difference between
associated with reduced treatment effectiveness. MBC and MIC values was within one dilution [66].
Age plays an important role in the variability of protein For beta-lactams displaying a time-dependent bacterial
content of CSF and its produced volume. In fact, the pro- killing activity, the most predictive pharmacokinetic/phar-
tein content may change from 1,770 and 1,350 mg/l in macodynamic parameter is represented by the time
preterm neonates at 26–28 and 38–40 weeks of age, between two consecutive doses during which the drug
respectively, up to 900 mg/l in healthy-term neonates [61]. concentration is higher than the MIC value of the causative
However, the CSF-to-serum albumin ratio is a direct bacteria (T[MIC). The value of T[MIC should be greater
function of the BBB permeability, while CSF flow is high than 40 % in the case of carbapenems, which are charac-
in newborns and in elderly people and is lowest in children terized by a postantibiotic effect, or 60–70 % for penicil-
at the age of 4 years [62, 63]. Therefore, an increased CSF- lins and cephalosporins. However, preclinical studies
to-serum albumin ratio reflects an augmented disposition suggest that the beta-lactam bactericidal concentration in
into CSF of substances and drugs, whose concentrations the CSF may be predicted by the maximum concentration
are often higher in infants and old people when compared (Cmax)/MIC (or Cmax/MBC) ratio. In fact, ampicillin Cmax
with those measured in children and young adults [12]. By in the CSF must correspond to 10–30 times the MBC value
contrast, no relationship between amikacin disposition and in order to obtain a bacterial killing activity of approxi-
protein content in CSF has been found in neonates [64]. mately 1 log10 CFU/ml per hour [67, 68]. This apparent
Finally, the choice of methods and techniques for the difference of pharmacodynamic behavior has been
measurement of drug concentrations within the matrix explained by the fact that high Cmax values in the presence
could affect the results. In fact, the use of high-perfor- of longer terminal half-lives of drugs in CSF with respect
mance liquid chromatography (HPLC) or agar diffusion to plasma half-lives, are associated with higher minimum
methods could lead to different results, as observed when concentration (Cmin), and hence higher T[MIC values.
the free fraction of ceftriaxone was measured in plasma Moreover, the Cmax/MBC, AUC/MBC, and T[MBC ratios
[65]. At variance with plasma, the measurement of CSF of ceftriaxone could strictly depend on each other, but the
drug concentrations was not affected by the method, sug- highest predictive value was retained by T[MBC [69]. The
gesting that the analytical methodology employed for the same conclusions were drawn by Lodise and colleagues for
prediction and/or calculation of antibacterial penetration meropenem [66].
into CNS should be carefully evaluated [65]. For antibacterial drugs with a concentration-dependent
killing potency, both Cmax and AUC pharmacokinetic
1.3.2 Pharmacodynamics of Antibacterials in CSF parameters may be predictive of bacterial killing. For
example, in experimental meningitis induced by E. coli,
The classification of drugs in terms of concentration- moxifloxacin was effective when the AUC/MBC ratio in
dependent or time-dependent bacterial killing activity CSF was 220–720 for total drug and 160–500 for the free
offers the basis for use of antibacterials at doses and drug [70]. For fluoroquinolones, a Cmax equal to 30–40
schemes that should maximize therapeutic effects. Fur- times MBC has been suggested to obtain the best thera-
thermore, the post-antibiotic effect and/or inhibitory effect peutic benefit. As in the case of antimicrobials with a time-
when concentrations are below the bacterial MIC (the so- dependent activity, all three pharmacokinetic/pharmaco-
called sub-MIC effect) are adjunctive factors that may dynamic parameters were found to be interrelated when
influence the overall antimicrobial effects of drugs. bacterial killing of gatifloxacin was evaluated, even if the
Overall, the definition of the most predictive pharmaco- AUC/MBC ratio had the highest correlation with drug
kinetic/pharmacodynamic parameter in terms of clinical and/ activity [71].
or microbiological efficacy has been based on plasma phar- Several antimicrobial drugs, currently used for the
macokinetics, because plasma represents a compartment that treatment of bacterial meningitis, are characterized by a
Pharmacokinetics of Antibacterials in CSF 523

post-antibiotic effect of variable length when measured in 2 Pharmacokinetic/Pharmacodynamic Profile


plasma, including carbapenems [72] and quinolones [73]. of Antibacterials in the CSF
Whether this characteristic is still present when the drugs
diffuse into CSF is still under investigation. In vitro 2.1 Beta-Lactams
studies have demonstrated that the post-antibiotic effect in
CSF may be higher than that observed in culture medium Drugs belonging to the class of beta-lactams display some
for cefotaxime, gentamicin, and ciprofloxacin [74], while features that could be responsible for their poor penetration
for trovafloxacin similar post-antibiotic effect values were into CSF. Although they are characterized by low molec-
observed in human pooled CSF samples and in culture ular weight, their hydrophilicity, the marked variability in
media against S. pneumoniae, H. influenzae, and plasma protein binding, and substrate affinity for trans-
N. meningitidis [75]. For other drugs, like beta-lactams, membrane transporters [85] may explain their limited
the delay in bacterial regrowth may depend on CSF penetration into CSF, unless meningeal inflammation is
concentrations lower than MIC (sub-MIC effect) rather present. Although the rate of penetration does not change,
than on the post-antibiotic effect [76, 77]. For example, some studies suggest that beta-lactam concentrations into
the inhibitory effect of ampicillin on bacterial growth was CNS may be increased by administering higher doses with
reduced when the drug was removed from the medium or respect to standard regimens, without a corresponding
inactivated, suggesting that the overall ‘‘post-exposure’’ increased risk of severe toxicities. Beta-lactams have a
inhibitory effect was mainly due to sub-MIC concentra- time-dependent activity and the division of the total daily
tions rather than to a post-antibiotic effect [78]. Further- dose in multiple doses or the adoption of extended infu-
more, for the exhaustive analysis of the phenomenon, it sions may give some advantages in terms of both greater
should be considered that several antibacterial drugs dis- T[MIC values and increased susceptibility [86, 87]. Nev-
play longer half-lives in CSF than in plasma (e.g., blood ertheless, how this approach could be transferred to CSF
and CSF half-lives were 0.7–1.4 and 2.1–3.6 h for should be carefully evaluated. In general, T[MIC values of
ampicillin and 0.9–1.7 and 9.3 h for ceftriaxone, respec- 40–70 % of the time interval between two consecutive
tively) [79, 80]. Of note, the antimicrobial activity of doses are considered predictive of treatment efficacy even
fosfomycin was found to be impaired in CSF [81]. if, in some selected cases, the attainment of a T[MIC value
In vitro experiments demonstrated that sustained bacterial equal to 100 % should be reached in order to reduce the
killing activity in human pooled CSF samples was found risk of treatment failure and to prevent the selection of
when drug concentrations of fosfomycin greatly exceeded mutant strains. Overall, beta-lactams are widely used for
MIC values (i.e., at least eightfold MIC, 16 mg/l), the treatment of CNS infections, for empiric or specific
whereas bacterial regrowth was observed 24 h after therapies. These drugs are also employed to prevent CNS
exposure at lower drug concentrations (i.e., from one up infections after trauma and neurosurgical procedures, but
to fourfold MIC). This result was not evident under the the analysis of available randomized controlled trials led to
same experimental conditions in plasma, when bacterial the conclusion that the benefit for the patient is negligible
regrowth occurred only for drug concentrations lower [88].
than the MIC. Therefore, an effective regimen (at least for
S. aureus infections) should be aimed at attaining CSF 2.1.1 Penicillins
antibacterial concentrations higher than eightfold MIC
during the entire dosing interval [81]. 2.1.1.1 Amoxicillin Although the use of amoxicillin in
Finally, CSF is devoid of an effective immune combination with clavulanic acid is not a first choice
response against bacteria [59]; therefore, drug concen- therapeutic option for bacterial meningitis, a CSF/free-
trations should achieve the bactericidal range as early as plasma AUC ratio for amoxicillin, administered at the
possible. However, in the presence of an infectious intravenous dose of 50 mg/kg body weight, was 0.06 in
meningitis, several cytokines and degradation products patients aged 14–76 years with inflamed meninges, and the
from bacteria act as chemoattractant stimuli for leuko- magnitude of the flogistic process seemed to directly and
cyte recruitment [82], with the subsequent CSF pleocy- significantly influence drug disposition when CSF was
tosis and tissue damage. Under these conditions, changes obtained by lumbar puncture [51]. In particular, the time
of pH and high protein content may play a role in profile of CSF levels revealed that drug concentration
reducing the activity of antibacterial drugs. For example, peaked as early as 2 h postdose, and a plateau was evident
preclinical studies demonstrated that bactericidal activity at 3–4 h postinfusion, with concentrations ranging from
of drugs like most macrolides and quinolones was approximately 0.7 up to 3 mg/l (mean 2.25 mg/l), with
reduced at CSF pH values lower than the physiological 68 % of values exceeding 1 mg/l. This level is sufficiently
pH [83, 84]. high to be effective, in combination with clavulanate,
524 A. Di Paolo et al.

against beta-lactamase-producing pathogens [51]. Clavu- same parameters were analyzed using a population phar-
lanic acid disposition into CSF was also investigated. The macokinetic approach and a subsequent Monte Carlo
major findings were that clavulanate reached higher CSF/ simulation [92], it was found that the mean CSF/plasma
plasma AUC ratio (0.08) than amoxicillin and the degree of AUC from time zero to infinity (AUC?) ratio for cefepime
inflammation did not significantly influence drug penetra- was 0.23 and none of the dosing regimens evaluated (2 g
tion. Furthermore, the amoxicillin/clavulanate ratio in CSF intravenous as a 30-min infusion every 8 or 12 h) allowed
ranged between 6.87 and 12.44, suggesting different attainment of T[MIC values of 50 and 100 % in CSF for at
pharmacokinetic behaviors of the two drugs [51]. These least 90 % of patients when a MIC value of 0.5 mg/l was
data suggest that amoxicillin plus clavulanic acid could obtained. However, when researchers took into consider-
have a limited role in treating staphylococcal and strepto- ation both a reduced probability of target achievement
coccal meningitis. (80 %) and MIC values lower than 0.5 mg/l, as in the case
of S. pneumoniae, a cefepime dosing regimen of 2 g
2.1.1.2 Ampicillin Ampicillin is used for empiric and intravenously every 8 h proved to be effective [95], in
specific therapy of meningitis in children and adults. agreement with previous data [96]. In fact, a T[MIC
Ampicillin may be employed for the treatment of bacterial value [90 %, which is predictive of treatment efficacy,
meningitis caused by Group B Streptococci, and other was observed in one of two patients given 2 g cefepime
pathogens (L. monocytogenes, N. meningitidis) [89]. The intravenously every 8 h, whereas six subjects, receiving
usual intravenous daily dose of 150–250 mg/kg should be 1 g cefepime intravenously every 8 h, had no measurable
given at various time intervals of 3–4 h in adult patients, up drug concentrations in CSF [96].
to a total dose of 6–12 g/day. At these dosages, and con- Of interest, Monte Carlo simulation indicated that the
sidering a pharmacokinetic/pharmacodynamic target of best target attainment for cefepime CSF concentrations was
T[MIC C50 %, Monte Carlo simulation demonstrates that associated with a dosing regimen using an intravenous
more than 90 % of N. meningitidis isolates is susceptible to loading dose of 2 g followed by continuous infusion at a
ampicillin at the plasma and CSF breakpoints of 4 and rate of 250 mg/h for a daily total dose of 8 g [92]. On the
0.5 mg/l, respectively [89], even if the clinical breakpoint basis of these results, continuous infusion after a loading
for susceptibility for ampicillin against N. meningitidis is dose could be associated with increased clinical effec-
0.125 mg/l [90, 91]. tiveness, in agreement with the results obtained when
In summary, the use of penicillins to treat meningitis is cefepime was administered by prolonged intravenous
widely recognized in empiric and specific treatments, infusions for infections other than those involving the CNS
regardless of the age of the patients. These drugs are lar- for which a metastatic spreading to the CNS could be
gely prescribed despite their relatively poor permeability of possible [87]. The study by Lodise and colleagues [92] also
the BBB, which may be increased by the presence of a showed that the enrolled patients had a minimal meningeal
flogistic process. inflammation, suggesting that in the case of meningitis the
distribution of cefepime into CSF should be increased, thus
2.1.2 Cephalosporins allowing prompt achievement of bactericidal concentra-
tions within the CNS. Moreover, beta-lactam effectiveness
Cephalosporins are largely used in combination with other could be best predicted by the Cmax/MIC or the ratio
antibacterial drugs for the treatment of CNS infections, between Cmax and MBC rather than the T[MIC parameter,
both in children and adults (Table 2). As already discussed because the Cmax/MBC ratio could be considered a surro-
in the above paragraph, beta-lactams display poor pene- gate marker of T[MIC [92]. It is noteworthy that cefepime
tration into the CNS, with the attainment of low concen- Cmax in CSF ranged from 7.4 up to 15.8 mg/l after the
trations when a minimal meningeal inflammation is present intravenous administration of a 2-g dose [96], and these
(‘‘the worst case scenario’’) [92], and this condition also concentrations were always higher than MICs for most
affects cephalosporin pharmacokinetics. For a general common pathogens [90].
overview regarding properties of cephalosporins and their
passage into CSF through the BBB, see the review by 2.1.2.2 Ceftriaxone Ceftriaxone shows a CSF/serum
Lutsar and Friedland [93]. AUC ratio \0.02 after an intravenous 30-min infusion of
2 g [80]; however, its probability of reaching target con-
2.1.2.1 Cefepime In the case of cefepime, a fourth-gen- centrations may be comparable to that of the fourth-gen-
eration cephalosporin, its pharmacokinetic and pharmaco- eration cephalosporins for low MIC values [95], as those
dynamic profiles in ventricular CSF and plasma were reported for S. pneumoniae [90]. Indeed, ceftriaxone
evaluated in patients treated with the drug at doses of 2 g remains one of the most used drugs for the treatment of
every 12 h by 30-min intravenous infusion [94]. When the meningitis (Table 2). In children given a single dose of
Pharmacokinetics of Antibacterials in CSF 525

ceftriaxone 75 mg/kg intravenously (maximum dose 2 only for MIC values of 0.5 and 0.25 mg/l, respectively,
g/day), the presence of an infection or the early adminis- indicating a moderate bactericidal activity of ceftazidime
tration of the drug ensured higher lumbar CSF drug con- against P. aeruginosa isolates (breakpoint 8 mg/l) among
centrations with respect to a late administration when the other Gram-negative bacteria [66].
infective process, as well as the flogistic meningeal reac- In summary, the new cephalosporins represent the
tion, were decreased [97]. In particular, at 6 h postdose, the backbone of several antimicrobial regimens for the treat-
mean lumbar CSF concentrations were 7.2 and 2.5 mg/l at ment of bacterial meningitis. From our survey of the lit-
the beginning or at the end of the infectious disease, erature, their penetration into CSF is high enough to
respectively. CSF concentrations were higher than MIC achieve bactericidal concentrations against the most com-
values against the most common bacterial isolates observed monly involved pathogens (H. influenzae, N. meningitidis,
in children (B0.03 mg/l) and the EUCAST breakpoints and penicillin-sensitive S. pneumoniae), especially in the
(0.125–0.5 mg/l) [90]. These results were partially con- case of cefepime. However, in some cases, MIC values
firmed in adult patients without inflamed meninges who make bacterial strains less sensitive to these drugs, thus
received a 2-g intravenous dose of ceftriaxone [80]. In requiring an increase in dosage.
particular, ventricular CSF Cmax values were in the range
of 0.18–1.04 mg/l, exceeding MIC values for highly sen- 2.1.3 Carbapenems
sitive bacteria, such as H. influenzae, N. meningitidis, and
penicillin-sensitive S. pneumoniae (0.125–0.5 mg/l). Meropenem, the most documented carbapenem derivative,
When ceftriaxone fails to inhibit bacterial species is selectively used for the treatment of severe CNS infec-
characterized by high MICs, like Enterobacteriaceae (MIC tions caused by Gram-positive and Gram-negative bacteria,
1 mg/l), the dose should be increased up to 4 g/day; including species producing beta-lactamases (extended-
otherwise, a carbapenem must be employed [80]. spectrum beta-lactamase and AmpC-producing strains).
Furthermore, at variance with imipenem, its use for the
2.1.2.3 Cefotaxime Compared with ceftriaxone, cefotax- treatment of severe bacterial meningitis is supported by a
ime showed a greater ventricular CSF distribution in adults minor incidence of seizures, which allows the use of high
(CSF/total-serum AUC? ratio range, 0.03–0.21) for the doses [98]. The administration of intravenous doses of 20
same single 2-g intravenous dose [80]. However, according and 40 mg/kg is associated with meropenem peaks in CSF
to clinical indications, daily doses can be augmented up to of about 0.1–2.8 and 0.3–6.5 mg/l, respectively, approxi-
12 g/day without increasing the risk of CNS toxicity. mately 2–3 h after dosing [99, 100].
Taking into account the efficacy predictive value of the The absence of meningeal inflammation is concomitant
Cmax/MIC ratio, a single 2-g intravenous dose of cefotax- with a reduced penetration of meropenem into CSF, despite
ime was associated with ventricular CSF Cmax values of the negligible plasma protein binding of approximately
0.14–1.81 mg/l. As stated above for ceftriaxone, these 2 % [53]. In adult patients exposed to meropenem for non-
Cmax values were higher than MIC values for the strains CNS infections and provided with an EVD, the adminis-
most commonly involved in meningitis, but ineffective tration of 2 g as a short 30-min infusion led to CSF Cmax
against Enterobacteriaceae. Therefore, the cefotaxime dose mean values of 0.63 ± 0.50 mg/l, with a range varying
should be increased or a carbapenem must be selected in from 0.13 up to 1.60 mg/l. These values were much lower
the presence of less sensitive bacterial strains [80]. than the corresponding mean plasma levels (84.7 ±
23.7 mg/l). Moreover, time to reach Cmax (tmax) values in
2.1.2.4 Ceftazidime Adult patients treated with ceftazi- ventricular CSF were 4.1 ± 2.6 h, suggesting a slow dis-
dime (3 g, 30-min intravenous infusion) for extracerebral tribution of meropenem into the CNS [53]. Interestingly,
infections were enrolled in a pharmacokinetic study aimed AUC? values for CSF were found to be related with those
at evaluating the probability of achieving bactericidal calculated for serum; hence, the increase in dosage could
concentrations within the CSF for the most frequent bac- be associated with an augmented disposition of meropenem
terial strains encountered in clinical practice [66]. Serum from plasma to CSF. Furthermore, the analysis of the most
pharmacokinetic results were calculated by considering a predictive pharmacokinetic/pharmacodynamic parameter
10 % protein binding. The disposition of the drug into for meropenem showed that T[MIC values were C8 h in
ventricular CSF was poor, as witnessed by the low median patients with uninflamed meninges when MIC values of
value of the CSF/free-serum AUC ratio of 0.04 (inter- 0.125–0.5 mg/l were taken into consideration. These
quartile range 0.02–0.08). Furthermore, when a regimen results suggest that, in the presence of uninflamed meninges,
consisting of 3 g of ceftazidime every 8 h as intravenous meropenem could not reach bactericidal concentrations in
infusion (30 min) was applied, the probability of achieving CSF of some patients, and high doses (up to 12 g/day) are
a T[MIC value of 50 and 100 % was higher than 80 % required [53].
526 A. Di Paolo et al.

In agreement with these results, Lodise and colleagues the decreased clinical use of these drugs in the past 10 years.
[66] found that the probability of a T[MIC target attain- Although intrathecal administration can overcome the poor
ment equal to 50 and 100 % was 80 % when MICs were distribution into CSF, the availability of more effective and
B0.25 and B0.125 mg/l, respectively, in patients equipped safer drugs justifies the diminished request of aminoglyco-
with an EVD and in the absence of meningeal inflamma- sides for the treatment of CNS infections.
tion. In agreement with a preclinical model [69], Lodise Antimicrobial therapy with amikacin (7.5 mg/kg intra-
and colleagues suggested that the pharmacodynamic target venously twice a day) in children affected by community-
of meropenem might be represented by a T[MIC value acquired bacterial meningitis led to a mean concentration
ranging from 50 to 100 % [66]. Nevertheless, various in lumbar CSF of 1.65 mg/l at 3 h postdose, which was
reports showed that meropenem was effective in the considered the expected time to peak [102]. The corre-
treatment of infections caused by microorganisms less sponding Cmax values in total serum (obtained 30 min after
susceptible to cephalosporins like Enterobacteriaceae, the end of infusion) was 19.6 mg/l, for a Cmax CSF/total
which are considered resistant if MIC values are C1 mg/l serum ratio of 0.08.
[90]. Furthermore, serum and CSF samples (from lumbar
A more recent pharmacokinetic study of meropenem puncture performed to exclude or to document meningitis)
was performed by Tsumura and colleagues [54] in neuro- from newborns were analyzed for amikacin concentrations
surgical adult patients fitted with an external lumbar drain. by using a population pharmacokinetic approach that
Several CSF and blood samples were collected over 16 h demonstrated significant correlation between total serum
after the first meropenem dose (0.5 g every 8 h infused in and CSF concentrations of amikacin [64] when the drug was
30 min), and drug concentrations were then fitted by using administered on a weight-adjusted basis as a 20-min intra-
a three-compartment model. A Monte Carlo simulation was venous infusion. Median CSF concentration of the drug was
performed considering both a T[MIC target equal to 1.08 mg/l (range 0.34–2.65 mg/l), while Cmax and Cmin
100 % and a probability of target attainment of 95 %; then serum values were 35.7 ± 5.9 and 3.8 ± 2.5 mg/l,
the CSF pharmacodynamic-derived breakpoint was calcu- respectively, with a partition coefficient between CSF and
lated. Although higher concentrations of meropenem total serum of 0.103. These results suggest that after the
should be expected when CSF is harvested by lumbar third dose, the amikacin mean peak concentration in CSF
puncture instead of ventricular drainage, results showed could be 2.5 mg/l, a value that could ensure a Cmax/MIC
that the administration of intravenous meropenem 1.5–2 g ratio of 10 for bacterial MICs of 0.25 mg/l or lower, while
every 8 h was associated with a pharmacodynamic break- other sensitive strains show pharmacodynamic breakpoints
point of 0.25 mg/l against S. pneumoniae, N. meningitidis, B8 mg/l [90]. It is of interest that ototoxicity, given repe-
and H. influenzae [54], a value similar to that adopted by ated doses, showed higher occurrence in preterm newborns,
EUCAST for sensitive bacterial strains of the three species, in whom an inflamed status of meninges is a known factor
in the case of meningitis [90]. of increased risk of hearing loss, because of higher BBB
Moreover, the BBB was not expected to be damaged or permeability [64, 103]. Therefore, the use of amikacin in
inflamed in the above-mentioned study [54], suggesting a bacterial meningitis should be carefully evaluated.
putative increased disposition of meropenem into CSF in
the presence of bacterial meningitis. However, the disrup- 2.3 Fluoroquinolones
tion of barriers due to a surgical procedure requiring EVD
equipment still remains a matter of debate. Finally, changes The administration of fluoroquinolones in one dose or in
in drug administration modalities (i.e., 3-h rather than 0.5-h two refracted doses per day could increase their penetration
infusions) could improve target attainment, as demon- into the CSF, with the achievement of bactericidal con-
strated in previous studies [101]. centrations both in terms of Cmax/MIC and/or AUC/MIC,
In summary, meropenem represents a drug for which the because fluoroquinolones are characterized by a concen-
recommended dosage is 2 g every 8 h as an intravenous tration-dependent bactericidal activity. Moreover, a major
3-hour infusion, with MIC values B0.25 mg/l. Its disposi- advantage of fluoroquinolones is their capability to cross
tion into CSF may increase in the presence of inflamed the BBB for the treatment of CNS infections due to their
meninges. rapid penetration and appearance in CSF, making intra-
thecal administration unnecessary. In fact, when CSF and
2.2 Aminoglycosides plasma concentrations were calculated at the same time
points, pharmacokinetic analysis revealed the achievement
The limited penetration of aminoglycosides in several tis- of CSF/plasma AUC ratio values of 0.4–0.6 or higher,
sues such as the CNS, lung, and bone and their poor toler- suggesting a rapid disposition of the drugs within the CNS
ability should be viewed as the main factors responsible for [104].
Pharmacokinetics of Antibacterials in CSF 527

2.3.1 Moxifloxacin gatifloxacin 400 mg every 24 h) as add-on oral therapy in


association with standard antitubercular chemotherapy.
Among fluoroquinolones, moxifloxacin has demonstrated a Plasma and corresponding CSF samples from lumbar
marked penetration into CSF. A pharmacokinetic analysis puncture were obtained and pharmacokinetic parameters
of moxifloxacin was carried out in adult male patients were compared among the three groups. Oral levofloxacin
given moxifloxacin in a 400-mg oral dose as prophylaxis displayed the highest mean value of the CSF/plasma
for a short urological surgical procedure, and CSF was AUC24 ratio at steady state (median 0.74; range 0.58–1.03),
obtained from lumbar puncture [105]. The highest total whereas the ratio value was lower for gatifloxacin (median
plasma concentrations (6.55 ± 3.61 mg/l) were obtained 0.48; range 0.47–0.50) and ciprofloxacin (median 0.26;
within 2–4 h postdose, whereas in CSF, Cmax (4.07 ± range 0.11–0.77). Interestingly, U-shaped relationships
1.15 mg/l) was observed between 4 and 6 h postdose, thus between drug exposure and clinical outcome were identi-
explaining why the highest CSF/plasma ratio was observed fied, for which the major benefit from chemotherapy was
in the range of 4–6 h postdose. The 400-mg oral dose of associated with intermediate rather than high exposure
moxifloxacin suggests the attainment of Cmax/MIC values levels (i.e., in the case of levofloxacin, the AUC24/MIC
of nearly 10 for Klebsiella spp., Enterobacter spp., and ratio was in the ranges of 112–220 and 14–252 in plasma
Proteus [90]. and CSF, respectively). The study further showed that the
Four patients affected by tubercular meningo-encepha- penetration ratios for levofloxacin and ciprofloxacin were
litis received moxifloxacin at oral doses of 400 and more variable than those for gatifloxacin [109]. Indeed, Pea
800 mg/day in association with other antituberculosis and coworkers [110] reported levofloxacin pharmacoki-
drugs, including rifampicin [104]. The coadministration of netics at steady state in adult patients endowed with an
rifampicin may be able to negatively influence plasma EVD and treated with the drug at the dose of 500 mg twice
concentrations of moxifloxacin, probably by inducing a day administered as a 1-h infusion. On the basis of a
glucuronidation or sulfation processes [106]. However, dense sampling schedule of both blood and ventricular
results showed that the mean value of CSF/total-plasma CSF, the disposition of the drug within CSF was demon-
AUC from 0 to 24 h (AUC24) ratio ranged from 0.82 to strated to be substantial also in the absence of inflamed
0.71 at oral doses of 400 and 800 mg/day, respectively, meninges, with an AUC from 0 to 12 h (AUC12) CSF/total
suggesting a good penetration of the drug within the CNS plasma ratio of 0.71, and a Cmax ratio ranging from 0.32 up
[104]. Because CSF was sampled through an external to 0.79 (i.e., a coefficient of variation of approximately
lumbar drain, tmax in CSF was observed approximately 4 h 30 % for both parameters). However, the CSF AUC12
after drug administration [104]. The free fraction of values ranged from 20.31 up to 48.73 mgh/l, suggesting
moxifloxacin was 40–60 % in plasma and up to 90–95 % that the adopted schedule (intravenous 500 mg every 12 h)
in CSF, while in two out of four enrolled patients the CSF/ could not be effective in some patients when MIC values
plasma AUC24 ratio for ultrafiltrate ranged from 0.85 to are higher than 1 mg/l (i.e., AUC24/MIC values lower than
1.16 and from 1.31 to 1.75 for oral doses of 800 and 100) [110]. Therefore, the authors suggest the administra-
400 mg/day, respectively [104]. Furthermore, the geomet- tion of levofloxacin in combination with a beta-lactam
ric mean values of AUC24/MIC in CSF were 84 and 132 at when the causative microorganism is unknown [110].
doses of 400 and 800 mg, respectively [104], and these In conclusion, fluoroquinolones may be regarded as
values were associated with at least a 2-log10 CFU decrease antimicrobials able to achieve bactericidal concentrations
in experimental models of lung tuberculosis [107]. More in CSF, even if at higher MIC values these drugs should be
recently, Pranger et al. [108] described the pharmacoki- used in combination therapy with other antibacterials.
netics of moxifloxacin (400 mg orally) in a patient affected
by tubercular meningitis. In this case report, a CSF/plasma 2.4 Colistin
AUC24 ratio of approximately 60 % was calculated, with
an AUC24/MIC ratio of 208 in plasma (the unbound frac- Although the use of colistin has been reduced in the past
tion) and 175 in CSF, with the subsequent achievement of mainly because of the occurrence of severe toxicitiy, the
bactericidal concentrations within the CNS. emergence of multidrug resistant strains of P. aeruginosa,
Acinetobacter, and Klebsiella pneumoniae has renewed the
2.3.2 Levofloxacin, Ciprofloxacin, and Gatifloxacin interest for this drug, whose more reliable pharmacoki-
netic/pharmacodynamic parameter seems to be represented
Interesting results have been reported by Thwaites et al. by AUC/MIC of the unbound fraction of the drug [111].
[109], who evaluated the penetration into the CNS and the The introduction of colistimethate sodium, as a more tol-
therapeutic role of three different fluoroquinolones (levo- erable prodrug, has allowed the use of colistin in pediatric
floxacin 500 mg or ciprofloxacin 750 mg every 12 h and patients. Antachopoulos et al. [112] investigated colistin
528 A. Di Paolo et al.

penetration into the CNS of infants and adolescents, who achievement of clinical response in eight out of nine
were affected by Gram-negative infections and fitted with patients in the absence of CNS toxicity [55].
an EVD. Colistin and colistimethate sodium concentrations Recently, physicians have faced the outbreak of severe
were measured both in plasma and CSF before and 30 min infection caused by K. pneumoniae carbapenemase (KPC)-
after intravenous drug administration at doses ranging from producing bacteria, which is a condition characterized by a
60,000 to 225,000 IU/kg per day. The drug did not reach high mortality rate among patients. Both colistin and
effective concentrations in CSF when MIC values for colistimethate sodium have been shown to have a signifi-
Acinetobacter baumannii, P. aeruginosa, and K. pneumo- cant role in combination with rifampicin and tigecycline
niae were C2–4 mg/l, in accordance with EUCAST [116]. Although MIC for colistin may be higher than the
guidelines [90]. In particular, peak and trough values in drug breakpoint, these regimens display a synergic bacte-
CSF ranged from 0.05 to 0.15 mg/l, but they increased up ricidal activity, resulting in clinical cure of patients.
to 0.50 and 0.46 mg/l, respectively, in the presence of Moreover, in order to obtain the highest concentrations as
meningitis. Overall, the CSF/total serum penetration ratio early as possible, the intravenous regimen of colistimethate
at the ventricular level ranged from 0.03 to 0.67, in sodium should use a loading dose of 9–12 9 106 IU fol-
agreement with previous studies [113]. Interestingly, all lowed by maintenance doses of 4.5 9 106 IU every 12 h in
these values are below the MIC values of multidrug- association with other drugs [117].
resistant Gram-negative rods. In summary, colistin and colistimethate sodium repre-
Similar results were obtained by Markantonis and sent drugs of choice when the causative microorganisms
coworkers [114] in adults exposed to colistimethate sodium are multidrug-resistant strains. However, these drugs
(150–225 mg daily intravenous dose) for infections caused should be used in combination with other antibacterials,
by multiresistant Gram-negative bacilli. The CSF samples and, in selected cases, intraventricular administration
were obtained by lumbar puncture, and CSF/total-serum should be adopted to augment the probability of cure
ratio was found in the range of 0.051–0.057 1–3 h after because of modest and variable drug penetration into CSF.
drug administration, while CSF/total-serum AUC ratio at
steady state was 0.051. 2.5 Macrolides
The systemic administration of colistin or colistimethate
sodium was associated with low CSF concentrations, sug- The limited penetration of macrolides into CSF is related to
gesting that in the presence of multidrug-resistant bacteria their high molecular weight and affinity for P-glycoprotein;
the drug could be ineffective [114]. The availability of a consequently, they do not reach sufficient active concen-
more tolerable prodrug, like colistimethate sodium, allows trations in the CSF when meningeal inflammation is lack-
intraventricular administration, thus increasing the proba- ing [118, 119]. Because of a failure of bactericidal activity
bility of achieving bactericidal concentrations in the CSF. in experimental S. pneumoniae meningitis, the role of
A recent investigation by Imberti et al. [55] evaluated macrolides in the treatment of CNS infection is limited to
the pharmacokinetics of colistin in CSF from patients given Legionella pneumophila infections.
intraventricular colistimethate sodium for CNS infections
due to pan-resistant Gram-negative bacteria. Colistimethate 2.5.1 Azithromycin
sodium was administered as an intraventricular bolus
(1–2 min) at doses ranging from 2.61 mg every 24 h up to Azithromycin, a 15-membered ring azolide antibiotic
5.22 mg every 12 h, a dose level close to the schedule of related to macrolide erythromycin, differs in antimicrobial
10 mg/day recommended by Tunkel et al. [115]. The activity and pharmacokinetic properties from the prototype
average steady-state concentrations of active colistin in the macrolide [120] and is superior to erythromycin against
ventricular CSF ranged between 3 and 12.2 mg/l, while the some pathogens, including H. influenzae, Moraxella ca-
mean tmax value was 3.7 h. Minimum CSF concentrations tarrhalis, and L. pneumophila [121, 122]. The antibiotic
ranged from 2.0 up to 9.7 mg/l, and these values were not has excellent pharmacokinetic properties, including wide
observed when intravenous delivery was used in pediatric distribution in body fluids or tissues and high intracellular
patients [112]. Cmin values were higher than MICs of pan- storage [123–125].
resistant bacteria for all evaluated dose regimens, with the Concentrations of azithromycin in the CNS were mea-
exception of the lowest dose (i.e., 2.61 mg intraventricu- sured in patients with brain tumors receiving multiple oral
larly every 24 h). Finally, the dense CSF sampling sche- doses of antibiotic prior to the surgery [120]. Brain tissue
dule showed that the estimated AUC24 ranges were samples were taken from the periphery of the tumor mass,
72.5–248.6 and 149.1–282.9 mgh/l at doses of 2.61 mg specifically from the cerebral cortex and cerebellum. The
every 24 h and 5.22 mg every 12 h, respectively. These mean concentrations of azithromycin in brain tissue at 24,
pharmacokinetic results were associated with the 48, 72, and 96 h after dosing were 2.63 ± 2.58,
Pharmacokinetics of Antibacterials in CSF 529

3.64 ± 3.81, 0.74 ± 0.37, and 0.41 lg/g, respectively, penetration of the macrolide was low, regardless of the
whereas the mean levels in serum at the same times were administration route (oral or intravenous). The highest CSF
0.031 ± 0.044, 0.016 ± 0.011, 0.012 ± 0.005, and levels were found 3–6 h after drug administration; the
0.008 mg/l, respectively. The mean ratios of the concen- maximum index of penetration from the blood into the CSF
tration of the drug in brain tissue to that in serum at 24, 48, was about 10 %. In cases of meningeal inflammation,
72, and 96 h after dosing were 255.76 ± 429.39, erythromycin penetration significantly increases [130].
199.06 ± 188.90, 62.71 ± 5.60, and 51.25, respectively. In conclusion, because of their relatively high molecular
The CSF levels of azithromycin ranged from undetectable weight, protein binding, lipophilic properties, and P-gly-
to 0.015 mg/l at 24 and 48 h after a single 500-mg oral coprotein affinity, the entry of macrolides into the CSF is
dose [120]. limited. Despite great activity against S. pneumonia
in vitro, clarithromycin was only bacteriostatic in experi-
2.5.2 Clarithromycin mental pneumococcal meningitis. At various times fol-
lowing oral treatment with azithromycin, high levels of the
Clarithromycin is the most active macrolide against rapidly macrolide are found in cerebral tissues whereas low con-
growing mycobacteria. Monotherapy with clarithromycin centrations are detected in CSF. Due to the poor CSF
has been successfully employed, although resistance has penetration of macrolides, some authors recommend the
been observed among isolates of M. abscessus from addition of rifampicin or amikacin to macrolide regimens
patients with disseminated disease treated with clarithro- when CNS symptoms are manifested [50].
mycin monotherapy. Resistance has been associated with a
point mutation in the gene encoding pr23SRNA [126]. For 2.6 Tetracyclines and Tigecycline
this reason, the combination of two drugs might be indi-
cated, like the addition of amikacin, which showed sig- Tetracyclines display a time-dependent killing pattern and
nificant additive effect especially for disseminated disease exhibit a considerable postantibiotic effect [131, 132]. This
[127]. is also the case for tigecycline, which is a new member of
Limited information is available concerning the pene- this class [133]; the best pharmacokinetic/pharmacody-
tration of clarithromycin into CSF in humans. In a post- namic index is the AUC24/MIC ratio. In fact, in clinical
traumatic meningitis due to M. abscessus, a patient was trials on patients affected by complicated skin or intra-
treated initially with oral clarithromycin at a dosage of abdominal infections, the AUC24/MIC ratio was chosen as
1,000 mg twice daily and intravenous amikacin plus the pharmacokinetic/pharmacodynamic index for target
intrathecal amikacin. In the presence of meningeal development. Owing to the prolonged postantibiotic effect
inflammation, intraventricular CSF concentrations of clar- and AUC24/MIC ratios of 17.9 and 6.96 for skin and intra-
ithromycin increased to 15–18 % of the simultaneous abdominal infections, respectively, good clinical efficacy
serum levels and to 25–27 % for its metabolite, after 4 and was observed [134, 135]. However, the time above the
9 h, respectively, and they were significantly higher (15- to MIC of free drug level versus effect had a high correlation
17-fold) than the MIC for clarithromycin (0.125 mg/ml). coefficient [133].
Despite CSF levels of clarithromycin and amikacin in
excess of their in vitro MIC for the microorganism, 2.6.1 Doxycycline
the CSF cultures remained continuously positive for
M. abscessus [128]. Among tetracyclines, the most effective treatment of
In experimental pneumococcal meningitis, clarithro- CNS infections is with doxycycline. Clinical studies
mycin lacks bactericidal activity in CSF despite its CSF indicate that doxycycline is slowly absorbed after oral
levels exceeding the MBC of the test strains and a bacte- administration, taking 2–3 h to reach peak concentra-
ricidal in vitro effect in time–kill studies [84]. A possible tions. The elimination half-life is long, ranging from 12
explanation may be related to the fact that the MBC of to 25 h. Average AUCs for oral doses of 200 mg/day
clarithromycin is substantially increased when the pH of range from 40–123 and 61–112 mgh/l for intravenous
the medium is acidified, which is the case for CSF during doses [136].
meningitis [129]. Doxycycline appears to be an effective agent for the
treatment of early syphilis and neurosyphilis [137, 138],
2.5.3 Erythromycin and appears to have both antimicrobial and anti-inflam-
matory activities against Borrelia burgdorferi in Lyme
In neurosurgical patients, erythromycin penetrates into the disease [139, 140].
CSF after oral administration of a 300–500 mg dose or After single oral or intravenous doses of doxycycline
intravenous administration of 200 mg. However, the (100 mg), the concentrations found in the lumbar CSF did
530 A. Di Paolo et al.

not exceed 0.1 mg/l, while daily oral administration for Despite its high efficacy against multidrug-resistant
2–10 days gave a level of 0.1–0.76 mg/l, whose mean pathogens, tigecycline is not usually recommended in
value (0.37 mg/l) corresponded to 14 % of the serum cases of meningeal infections, based on data showing
concentration [141]. modest penetration into the CSF in healthy volunteers
Based on these data, Yim and colleagues [142] inves- [146, 147]. After intravenous administration of a single
tigated doxycycline concentrations in the lumbar CSF from 100-mg dose of tigecycline to subjects with noninflamed
patients affected by neurosyphilis treated with oral doses of meninges [146], mean lumbar CSF and serum AUC24
200 mg twice a day for 21 days. The mean doxycycline values, obtained from a total of 17 cases, were 0.46 and
total serum level was 5.8 mg/l, and the mean CSF level 4.18 mgh/l, respectively, with an AUC24 ratio of 0.11. In
was 1.3 mg/l, which exceeded the standard MIC for the same study, mean and median CSF concentrations of
Treponema pallidum. Penetration into CSF varied from 11 tigecycline, measured between 0.92 and 2.1 h after the
to 56 %, with a mean of 26 % [142]. start of infusion, were 0.015 and 0.014 mg/l (range
In patients treated with oral doxycycline (200 mg once 0.011–0.020 mg/l). Individual ratios of CSF-to-serum
daily) for neuroborreliosis, the median concentration in concentrations during the sampling period ranged from
lumbar CSF was 0.6 mg/l after 4 h, corresponding to 0.016 to 0.095, with a mean of 0.055. In comparison, the
3–36 % (mean 15 %) of the simultaneous concentration respective mean and median CSF concentrations of the six
in total serum [143]. Although patients with a peak samples obtained between 18.4 and 26 h after the start of
doxycycline concentration below the median of 6.1 mg/l infusion were 0.025 and 0.022 mg/l (range 0.021–0.033
in serum had a mean concentration in CSF of 0.62 mg/l mg/l). Individual ratios of CSF-to-serum concentration
compared with 0.97 mg/l for patients with a higher during this sampling period ranged from 0.33 to 0.52,
concentration in serum, no significant correlation with a mean of 0.41 [146].
between the concentrations of doxycycline in serum and In contrast to the moderate penetration ratio observed in
CSF was observed [143]. By contrast, Dotevall et al. subjects with noninflamed meninges, successful treatment
[144] found significantly higher concentrations of CSF has been reported in patients with multidrug-resistant
doxycycline when the antimicrobial was administered as pathogen-induced meningitis, receiving parenteral tigecy-
oral doses of 100 mg twice a day (serum mean concen- cline [148–150]. Moreover, in a case report, Lengerke et al.
trations 4.7 mg/l; CSF mean concentration 0.6 mg/l) or [151] investigated tigecycline brain penetration after
200 mg twice a day (serum mean concentrations 7.5 mg/ 11 days of parenteral administration of 50 mg every 12 h
l; CSF mean concentration 1.1 mg/l), showing a steady in a neutropenic patient affected by Enterococcus faecium-
state of CSF doxycycline levels on day 7. The penetra- induced meningitis, and carrying an EVD. The authors
tion of doxycycline into CSF, compared with serum observed comparable mean total plasma and CSF steady-
concentrations, was lower (15 %) than that reported by state levels in samples collected within 5–9 h after expo-
Yim et al. (26 %) [142], probably due to differences in sure to 50 mg tigecycline (plasma 0.0366 ± 0.0014 mg/l;
the time of sampling [144]. Although it is generally CSF 0.031 ± 0.0045 mg/l) and identical mean trough
agreed that patients with a damaged BBB have increased levels of tigecycline (plasma 0.0254 mg/l; CSF 0.0250 mg/l).
CSF concentrations of antibiotics, the mean concentra- The reported AUC and the CSF/plasma ratio of tigecycline
tion of doxycycline in CSF has been found to be similar concentrations yielded comparable values (0.85) and
in patients with BBB dysfunction (calculated as the CSF/ peak levels assessed in plasma 30 min after infusion
serum albumin ratio) (mean 0.77 mg/l with a CSF/serum (0.232 mg/l) could not be reproduced in CSF analyzed at a
concentration ratio of 0.18) and those without BBB corresponding time point (0.0386 mg/l). These data sug-
dysfunction (mean 0.83 mg/l with a CSF/serum concen- gest a higher penetration of tigecycline into CSF in cases of
tration ratio of 0.13) [144]. inflamed meninges than that reported in patients with
noninflamed meninges, where CSF peak levels reached,
2.6.2 Tigecycline respectively, 52 and 41 % of the corresponding total
plasma levels [146, 148].
Tigecycline is a glycylglycine antimicrobial with bacte- Furthermore, two case reports have been published for
riostatic activity. In in vitro evaluations, tigecycline is patients with cerebral and meningeal infections. The case
active against the most prevalent pathogens including reported by Ray et al. [152] presented a patient affected by
Gram-positive, Gram-negative, aerobic, and anaerobic A. baumannii-induced cerebritis that was treated with an
bacteria isolated from diabetic foot infections with 95 % of intravenous 100-mg loading dose of tigecycline followed
MICs B2 mg/l [145]. It is considered an emerging anti- by 50 mg twice daily. The patient showed CSF concen-
microbial for the treatment of multidrug-resistant strains, trations at the steady state slightly higher (range
especially A. baumannii. 0.035–0.048 mg/l, with a penetration ratio between 0.09
Pharmacokinetics of Antibacterials in CSF 531

and 0.52) than those reported in patients with noninflamed daily by 1-h intravenous infusion) that the antimicrobial
meninges at 1.5 and 24 h after dosing (mean achieved maximum and minimum CSF concentrations of
0.015–0.025 mg/l) [146], suggesting a facilitated tigecy- 10.8 ± 5.7 and 6.1 ± 4.2 mg/l, respectively, with a mean
cline penetration through the compromised BBB. These penetration ratio of 0.66. This roughly corresponds to the
data are similar to those reported for a patient affected by free plasma fraction of the drug and the mean Cmin value
K. pneumoniae meningitis [153], in whom tigecycline was higher than the pharmacodynamic breakpoint of 2 mg/l
penetration was about 0.35 after the parenteral adminis- [90].
tration of 100 mg every 12 h. The authors suggested a 1:1 Interestingly, the adoption of a different schedule of
tigecycline dose:concentration response in CSF, because drug administration in children (i.e., 10 mg/kg intrave-
they found twofold higher CSF tigecycline levels when the nously every 12 h for 3 days instead of every 8 h for
patient was treated with a twofold higher dose. Although 2 days) was associated with a similar linezolid penetration
these concentrations were less than the MIC determined for into ventricular CSF after the last dose [CSF/total plasma
tigecycline, the patient responded clinically and the culture AUC12 ratio 0.98 and CSF/total plasma AUC from 0 to 8 h
of CSF, obtained by EVD, became sterile [153]. (AUC8) ratio 0.95], with a small advantage in terms of
In conclusion, tigecycline is active against many Gram- increased mean Cmin values (1.26 vs 1.94 mg/l, respec-
negative and Gram-positive bacteria, but data about its use tively) [158]. Furthermore, meningeal inflammation did not
in meningitis are limited to some case reports. In these play a role concerning linezolid penetration into CSF. In
cases, tigecycline offers good CSF penetration even if agreement with the above-mentioned studies, a wide range
concentrations achieved at current standard doses could be of values from analyzed cases was observed, in terms of
insufficient to ensure bacterial killing [154]. both Cmax and Cmin range, further supporting the variable
disposition of linezolid into CSF. In a case report on lin-
2.7 Oxazolidinones ezolid and rifampicin interaction, Gebhart et al. [128]
hypothesized a role for ABCB1/P-glycoprotein, for which
Linezolid is the first member of a synthetic class of anti- the drug could represent a substrate, thus explaining in-
microbials, the oxazolidinones, which are effective against terpatient variability. However, results from other studies
Gram-positive pathogens commonly responsible for CNS do not support ABCB1 or organic anion-transporting
infections. Linezolid has gained attention for the treatment polypeptide (OATP) as transporters involved in trans-
of CNS infections because of its effectiveness against membrane passage of linezolid [161].
multiresistant Gram-positive microorganisms such as In summary, CSF linezolid concentrations have been
methicillin-resistant Staphylococcus aureus, methicillin- found similar to those of the free fraction in plasma [162],
resistant coagulase-negative Staphylococcus, and vanco- whereas the presence of inflammation seems not to influ-
mycin-resistant Enterococcus with MIC values ranging ence linezolid penetration into CSF. When strains sensitive
from 0.5 up to 4 mg/l [90, 155]. For these reasons, the drug to linezolid are identified as causative bacteria, then the
is often used to prevent or to treat severe Gram-positive drug represents a possible choice for the treatment of CNS
infections at the CNS level, as may occur after neurosur- infections.
gical interventions. The maximal (i.e., bactericidal) thera-
peutic efficacy corresponds to free AUC/MIC values of 2.8 Daptomycin
80–120 and T[MIC values greater than 80 %, even if the
highest probability to obtain a bactericidal effect is asso- Daptomycin, a lipopeptide antibiotic with significant
ciated with T[MIC values of 100 % or Cmin values 4–5 activity against a wide range of Gram-positive bacteria,
times the MIC value [156]. To date, some studies have offers the possibility to treat infections caused by resistant
investigated drug pharmacokinetics in plasma and CSF, Gram-positive strains that could be less sensitive to lin-
which are characterized by wide interpatient variability. ezolid. Its concentration-dependent activity makes the
Indeed, careful analysis shows that some patients seem not AUC/MIC parameter the best predictor of drug effective-
to receive a therapeutic benefit on the basis of established ness, ranging from 400 (bacteriostatic effect) up to 800
pharmacokinetic/pharmacodynamic cutoff values [52, 57, (bactericidal effect) against methicillin-resistant Staphylo-
157, 158], even in the absence of known factors associated coccus aureus and vancomycin-resistant Enterococcus
with pharmacokinetic variability [159]. strains. However, the Cmax/MIC value should be regarded
Despite the observed clinical response variability, the as an additional parameter when a dense sampling schedule
role of linezolid as a therapeutic option for severe CNS is not feasible, taking into account a lower limit of thera-
infections cannot be doubted. Of note, Myrianthefs and peutic range equal to 100, as suggested by preclinical
colleagues [160] demonstrated on neurosurgical patients studies [163]. Experimental evidence supports the possible
carrying an EVD and treated with linezolid (600 mg twice use of daptomycin for the treatment of severe meningitis
532 A. Di Paolo et al.

caused by fluoroquinolone-resistant and/or penicillin- moderate and severe infections, respectively. On the basis
resistant pneumococcal strains [164]. However, previous of these considerations, some studies have examined the
studies described the relatively low penetration of dapto- advantage of continuous infusions over intermittent brief
mycin into CSF (approximately 6 %), when high peak and infusions. In some cases, the adoption of a continuous
trough concentrations were observed. In fact, in a case infusion may give a benefit [168, 169], while in other cases,
report, intravenous daptomycin (9 mg/kg) reached con- the adoption of an increased dose intensity (at least at the
centrations in total plasma and lumbar CSF of 11.21 and beginning of the treatment) may be associated with an
0.52 mg/l, respectively, with a CSF/plasma ratio of 0.05 in improved efficacy [170]. Whether these results may be
the presence of inflamed meninges [165]. More recently, transferred or extrapolated to CSF should be carefully
the disposition of a single intravenous dose (10 mg/kg) of evaluated.
daptomycin has been investigated in neurosurgical patients
with indwelling external CSF shunts (lumbar or ventricular 2.9.1 Vancomycin
drainages) [166]. The results demonstrated a CSF/plasma
AUC24 ratio of 0.08, which increased up to 0.11 when the In patients who have undergone neurosurgery, disruption
ratio took into account only the free fraction (approxi- of the BBB allows the achievement of therapeutic levels of
mately 0.30) of plasma daptomycin. The reduced disposi- vancomycin in CSF [171]. In adult patients, the intrave-
tion of daptomycin into CSF was probably due to the nous administration of vancomycin (1 g) was associated
variability of meningeal inflammation and it was witnessed with mean CSF concentrations of 6.24 ± 3.46 and
by high tmax values (6 h after administration) and reduced 2.55 ± 1.13 mg/l in ventricles and 4.49 ± 3.14 and
Cmax values (0.12 mg/l), which suggest the difficult 2.43 ± 0.41 mg/l in the spinal cord at 15–30 min (Cmax)
achievement of effective bactericidal concentrations in and 12 h (Cmin) after the end of drug infusion, respectively.
CSF. In view of the low variability of daptomycin phar- The mean Cmax values were higher than MIC values of
macokinetics, further studies are required to assess the role methicillin-resistant S. aureus and methicillin-resistant
of the drug for the treatment of CNS infections, even if coagulase-negative staphylococci strains (2 and 4 mg/l,
bacterial strains are found to be sensitive. respectively) [90, 171].
In summary, penetration of daptomycin into CSF is After administration of a single intravenous dose
limited, even if an increased penetration has been docu- (15–20 mg/kg) of vancomycin before insertion of a CNS
mented in the presence of inflamed meninges. Future shunt in pediatric patients, over 40 % of ventricular CSF
studies should investigate CSF disposition of the drug samples had undetectable drug concentrations [172]. On
when administered at high dosages (i.e., 12 mg/kg). the contrary, the administration of doses of 10–20 mg/kg
every 6–12 h for a documented or suspected shunt infec-
2.9 Glycopeptides tion led to mean CSF concentrations of 2.48 ± 0.52 mg/l,
even if the range of variability in both CSF concentrations
Glycopeptides possess some characteristics that could be (0–6.58 mg/l) and CSF/total plasma AUC ratio
responsible for their low disposition into CSF. In particular, (0.077–0.18) was large enough to assume that in some
hydrophilicity, a molecular mass approaching 1.4 kDa, and patients the schedule was clinically ineffective. These
protein binding ranging from 50 % (vancomycin) up to results suggest poor penetration of vancomycin into CSF,
90 % (teicoplanin) explain why drug disposition into CNS with concentrations equal or less than 10 % of those
should be considered inadequate. On the other hand, the measured in plasma [172]. In summary, the penetration of
administration of excessively high doses may expose vancomycin into CSF depends on both the integrity of the
patients to intolerable toxic effects. However, inflamed BBB and the inflammatory meningeal status, which
meninges enhance the distribution of vancomycin into CSF ensures greater penetration of the drug within CSF when
when the drug is administered in suitable doses. In fact, administered at adequate doses.
vancomycin 500–750 mg intravenously every 6 h may be
employed in association with a cephalosporin for the 2.10 Antituberculosis Drugs
empiric therapy of acute bacterial meningitis [167]
(Table 2). Tuberculous meningitis represents a particular form of
Although the effectiveness of vancomycin and teicopl- meningitis, often characterized by its occurrence in
anin is time dependent, the vancomycin effect is better immunocompromised patients, especially those affected by
predicted by AUC/MIC values higher than 400, whereas HIV infection in developing countries, where it is a com-
for teicoplanin a Cmin value greater than the bacterial MIC mon cause of bacterial meningitis. As for the pulmonary
is considered predictive of bactericidal effect. In particular, form of tuberculosis, several drugs are available for the
a trough value higher than 10 and 20 mg/l is required for treatment of tuberculous meningitis, displaying important
Pharmacokinetics of Antibacterials in CSF 533

advantages in terms of CSF disposition and achievement of 179]. In particular, mean Cmax values, occurring 3–5 h
bactericidal concentrations. postdose, were higher than 30 mg/l at oral doses of 30 mg/kg
per day. Pyrazinamide shows a concentration-dependent
2.10.1 Isoniazid effect, whose 90 % maximal effect of bacterial killing
occurs with an AUC24/MIC ratio of about 209 in the case
Isoniazid, which is still indicated as a first-line therapy for its of pulmonary tuberculosis, while T[MIC is associated
capability to protect other drugs from mycobacterium with the prevention of mutant strains [176, 180]. However,
resistance, has been demonstrated to be an agent with a Monte Carlo simulation demonstrated that pyrazinamide at
concentration-dependent activity with AUC/MIC being the oral doses of 15–30 mg/kg per day led to an AUC0–24/MIC
best pharmacokinetic/pharmacodynamic predictive param- value in epithelial lining fluid of about 209 in 15–53 % of
eter [173], even if monitoring of drug effectiveness has been patients, suggesting that daily doses of 60 mg/kg ([2
performed on the basis of maximal CSF concentrations. In g/day) have greater probability of target attainment [180].
fact, the drug showed its major effectiveness with plasma Whether these results can be transposed to pyrazinamide
Cmax values of 3 mg/l (or better 5 mg/l) [174], while the pharmacokinetics in CSF should be carefully evaluated.
achievement of these Cmax values in CSF is possible when
the drug is administered at oral doses of 5 mg/kg, or better at 2.10.4 Other Antituberculosis Drugs
8–10 mg/kg. Indeed, a study in children affected by tuber-
culous meningitis showed that CSF tmax ranged from 2 to 4 h Among the other drugs used for the treatment of tubercu-
postdose and mean ± SD Cmax values were 4.6 ± 2.4 and lous meningitis, streptomycin may be employed at doses of
11.6 ± 2.7 mg/l following oral doses of 10 and 20 mg/kg, 12–18 mg/kg intramuscularly, and the most appropriate
respectively. Of interest, acetylator genotype significantly pharmacokinetic/pharmacodynamic parameter to predict
influence Cmax values of isoniazid in CSF [175]. the best antibacterial effect is the Cmax/MIC ratio, which
should be equal or greater than 8 [181]. For example,
2.10.2 Rifampicin streptomycin CSF levels ranged between 1.6 and 2.2 mg/l
at 2–6 h after a dose of 13 mg/kg, while at higher dosages,
Rifampicin exerts its bactericidal activity in a concentra- CSF concentrations ranged between 3 and 16 mg/l.
tion-dependent manner [176], and in particular the maxi- Amikacin and kanamycin show similar pharmacokinetic
mal bacterial killing activity is associated with AUC24/MIC behavior for CSF disposition, with Cmax values of 4–8 mg/l
ratio values of approximately 24. Because its plasma pro- and a Cmax/MIC ratio of 10, after administration of doses of
tein binding is about 80 %, it is expected that only 20 % of 15 mg/kg per day intravenously. However, in the presence
the total plasma concentration could be responsible for the of MIC values of 0.5–1 and 2–4 mg/l for amikacin and
bactericidal effect. Of interest, the emergence of drug kanamycin, respectively, it is likely that a bactericidal
resistance may compromise treatment outcome and a free effect could be achieved in a minority of patients [182].
drug Cmax/MIC ratio greater than 175 has been found to A poor penetration into CSF characterizes ethambutol,
prevent the selection of resistant strains of M. tuberculosis for which CSF concentrations ranged from 0 up to
[176]. Further analysis demonstrated that the postantibiotic 1.98 mg/l at 3–4 h after administration of oral standard
effect of more than 5 days was related to the free rifam- doses [183]. In the case of ethionamide, a time-dependently
picin Cmax/MIC ratio. Therefore, the achievement of active drug with a T[MIC value of &4 h, daily oral doses
total and free Cmax concentrations of C9 mg/l (for of 250 mg in adults were associated with CSF concentra-
MIC & 0.05 mg/l) should be effective for bacterial killing tions of 1.0–2.6 mg/l at 3 h after dosing [184]. In children,
and mutant strain prevention, respectively [174]. When a regimen consisting of oral ethionamide at 15 or 20 mg/kg
rifampicin was administered at doses of 600 mg as 3-h led to mean lumbar CSF concentrations of 2.17 or 2.83 mg/l,
intravenous infusion, median Cmax values of 0.73 mg/l respectively, 3–4.5 h after drug administration [185], sug-
were obtained, whereas higher CSF concentrations were gesting that high dosing regimens could offer a better
found in children receiving intravenous dosages up to therapeutic benefit.
20 mg/kg [177]. Finally, cycloserine is characterized by a good CFS
penetration, with mean trough CSF concentrations of
2.10.3 Pyrazinamide 12.8 mg/l after oral doses of 250–500 mg, while Cmax
values of 22.5 and 19.9 mg/l at 9 and 6 h after dosing were
The third antitubercular drug indicated for a classic com- observed in a child and an adult patient, respectively [186].
bination for the treatment of tuberculous meningitis is In conclusion, pyrazinamide and cycloserine display the
pyrazinamide, which displays a good CSF disposition, with highest penetration into CSF with respect to isoniazid,
lumbar CSF/total plasma ratios greater than 0.75 [178, rifampicin, and other antituberculosis agents, especially
534 A. Di Paolo et al.

ethambutol, which is characterized by a poor disposition. delivery strategies have been developed [189]. These
For some drugs, like ethionamide and isoniazid, increased strategies generally fall into one or more of the following
dosage has been suggested to increase the probability of categories: [190] manipulation of drugs; disruption of the
target attainment. BBB; coadministration of antimicrobials with compounds
affecting drug influx/efflux; alternative routes for drug
2.11 Fosfomycin, Trimethoprim, delivery.
and Sulfamethoxazole Manipulation of drugs includes the possibility of mod-
ifying molecules, originally unable to enter the BBB, into
Fosfomycin is used for the treatment of multiresistant more permeable compounds. To date, various models have
Gram-positive and Gram-negative infections. Despite its been attempted, mainly in oncological settings, to increase
hydrophilic nature, its disposition into CSF is more rapid the lipophilicity of a drug by the use of lipophilic analogs
than beta-lactams because of its small molecular weight [191] or a lipophilic precursor or prodrug of a parent
and negligible plasma protein binding. The pharmacoki- compound [192, 193]. Another method is the use of lipo-
netics of the drug have been evaluated in patients carrying somes or lipid-based nanocarriers [194] to surround a
an EVD who received the drug at doses of 8 g intrave- hydrophilic molecule with a sphere of lipids to allow them
nously three times a day [187]. After single and multiple to flow through the BBB, as demonstrated for ciprofloxacin
intravenous doses, the CSF/total plasma AUC8 ratio was [195]. Polymeric micelles self-assembled from cholesterol-
0.23 ± 0.07 and 0.27 ± 0.08, while CSF Cmax values were conjugated polyethylene glycol and anchored with trans-
43 ± 20 and 62 ± 38 mg/l, respectively. Fosfomycin is a activator of transcription peptide have been created with a
time-dependently active agent, and T[MIC values range size of less than 180 nm. Ciprofloxacin was loaded into the
from 98 to 61 % for MIC values of 8 and 32 mg/l, micelles by membrane dialysis and sustained release of the
respectively. Therefore, in the presence of bacterial strains antimicrobial drug was observed both in vitro (human
with lower MIC values (i.e., 2 mg/l), the T[MIC may astrocytes) and in animal models, providing a promising
approach 100 %, suggesting that the drug could be effec- carrier of antibiotics across the BBB in a clinical setting
tive even against ‘‘difficult-to-treat’’ bacteria [29]. [195].
The disposition of trimethoprim and sulfamethoxazole A further invasive strategy to enhance CNS drug entry
into CSF is higher than beta-lactams, also in the presence involves the systemic administration of drugs in conjunc-
of uninflamed meninges because of their lipophilic nature. tion with transient BBB disruption. Entry of drugs into
Trimethoprim and sulfamethoxazole are time-dependently brain tissue and CSF can be facilitated by the artificial
active agents, for which a T[MIC value of 50 % has been temporary disruption of the diffusional barriers among
tentatively established [55], even if no differences were blood, CSF, and brain tissue, by the intra-arterial injection
found between T[MIC and AUC/MIC parameters. Lumbar of hyperosmotic compounds or alkyl glycerols [12].
CSF penetration (calculated as CSF/total serum AUC? However, the exact benefit/risk ratio of these procedures is
ratio) accounts for 0.18 and 0.12 for trimethoprim and still under examination and several risk factors, like the
sulfamethoxazole after oral and 2-h intravenous doses of 5 passage of plasma proteins, altered glucose uptake,
and 25 mg/kg, respectively [188]. Moreover, only 46 % of expression of heat shock proteins, microembolism, or
bacterial isolates from N. meningitidis infections were abnormal neuronal function [165], suggest a cautious
susceptible to trimethoprim and sulfamethoxazole [55]. approach.
In conclusion, trimethoprim and sulfamethoxazole dis- Recently, new and potentially safer biochemical tech-
play a penetration into CSF higher than beta-lactams. For niques have been developed to disrupt the BBB. Ultra-
these reasons, they are currently used for the treatment of sounds can be noninvasively delivered to the brain through
meningitis caused by L. monocytogenes. In the case of the intact skull. When combined with preformed micro-
fosfomycin, its rapid disposition into CSF supports its use bubbles, ultrasounds can safely induce transient, localized,
in the treatment of multiresistant Gram-positive and Gram- and reversible disruption of the BBB, allowing antibacte-
negative infections as well as ‘‘difficult-to-treat’’ bacterial rial drugs to be delivered. Investigations to date have
infections. shown positive response to ultrasound BBB disruption
combined with therapeutic agent delivery in preclinical
models of Alzheimer’s disease and in neuro-oncological
3 Methods to Increase Antibacterial Penetration settings [196–199], and could represent a promising tool
into CNS for release of antibacterials into the brain.
All these categories have been shown to work in proof-
To circumvent the multitude of barriers inhibiting CNS of-principle experiments and some have increased the
penetration of potential therapeutic agents, numerous drug efficacy of drugs that have very little ability to enter the
Pharmacokinetics of Antibacterials in CSF 535

brain [190, 200, 201]. However, two factors have limited aminoglycosides, in addition to intravenous antibiotics, a
the use of these promising approaches: toxicity and threefold increase in mortality was observed when com-
inability to deliver enough drug into the CNS; conse- pared to standard treatment with only intravenous antibi-
quently, these procedures are not yet included in routine otics [207, 208]. However, in a more recent retrospective
clinical use for treating CNS disorders. study [209], the efficacy of treatment with intraventricular
Some authors have reported increased penicillin pene- gentamicin was compared with that of systemic antibiotics
tration into CSF when administered together with proben- in patients with Gram-negative bacillary ventriculitis or
ecid, an inhibitor of the multidrug resistance-associated meningitis after neurosurgery. The intravenous antibacte-
proteins and of the renal tubular excretion of organic acids, rials were meropenem, cefotaxime, ceftazidime, imipenem,
which enhances the CSF concentrations of penicillins and and trimethoprim-sulfamethoxazole. The patients treated
cephalosporins [202, 203]. Although this pharmacological with intraventricular gentamicin had a significantly higher
association is not recommended by actual guidelines, this cure rate and a lower relapse rate than did those treated
procedure represents a theoretical aspect of specific trans- with other intravenous antibiotics with a similar mortality
porter blockade that could be a useful tool in cases unre- rate of 19 %.
sponsive to antimicrobial treatment, secondary to an Because of their relatively low systemic toxicity and high
insufficient drug concentration at a level of target tissue. potential to induce epileptic seizures after intraventricular
Recent experimental research showed nasal-to-CNS application [210], beta-lactam antibiotics are not good can-
drug release as a new and promising drug administration didates for this route of administration. However, whether
route. Data from preclinical models and investigatory intrathecal administration of antibacterials should be used to
human clinical trials have shown efficacy and tolerability treat CNS infections is a topic requiring careful evaluation
of this technique [204], suggesting the feasibility of this [208, 211], even though it represents the only chance to treat
mode of releasing antibacterials, such as cefotaxime, into infections in the presence of multiresistant organisms,
the brain [205]. Indeed cefotaxime levels, attained in the requiring high antimicrobial CSF concentrations.
brain following intravenous and intranasal administrations,
were comparable, suggesting that intranasal administration
of the antibacterials could be a potential noninvasive 4 Conclusions
method of drug transfer that might improve patient com-
pliance [205]. CNS infections, caused by bacteria with reduced sensitivity
An effective but invasive method to obtain high drug to antimicrobial drugs, represent an increasing challenge
concentrations within the CNS is intraventricular injection worldwide. Effective treatments of these infections require
in addition to intravenous administration, as discussed in careful consideration of several factors that may signifi-
Sect. 1.3. This method is most useful with antibiotics with cantly affect the therapeutic activity of antibacterials.
a low penetration rate into the CSF and/or with a high The uneven distribution of anti-infective drugs within
systemic toxicity, which precludes any increase of the daily the various CNS compartments, including CSF, is an
systemic dose (e.g., aminoglycosides, vancomycin). It important issue, even if simple analysis of drug penetration
should be noted that the administration of antibacterials cannot sufficiently predict clinical and microbiological
into the ventricular or lumbar CSF is almost always an off- success. The choice of appropriate laboratory and clinical
label use, and therefore requires a clear indication. For a endpoints (i.e., the measurement of a pharmacokinetic/
large hydrophilic compound such as vancomycin, the pharmacodynamic parameter) may improve the manage-
apparent volume of distribution after intraventricular ment of antibacterial chemotherapy, at both initial acute/
injection was 250 ml [206]. Vancomycin and other mole- empiric and maintenance stages for long-term periods.
cules with high molecular weight and strong hydrophilicity However, some hurdles remain present and will need to be
have a long elimination half-life in CSF, because they do solved before obtaining maximum effectiveness. Low BBB
not readily cross the BBB by diffusion after intracisternal permeability mainly affects hydrophilic drugs, namely
injection. The long half-lives in CSF ensure therapeutic beta-lactams and glycopeptides, for which the presence of
CSF concentrations with once-daily dosing. The high inflamed meninges is associated with an increase in CNS
interpatient variation of CSF elimination half-lives is penetration. However, cephalosporins, namely ceftriaxone,
caused by the heterogeneity of critically ill patients (e.g., ceftazidime, and the more recent cefepime, play an
CSF outflow from an external ventriculostomy, obstruction important role in modern chemotherapeutic regimens for
of ventricles or the subarachnoid space by blood, or drugs the treatment of CNS infections. Interestingly, these drugs
affecting the CSF secretion) [206]. may achieve therapeutic concentrations in CSF, supporting
In treatment of infants with Gram-negative meningitis their use to attain high clinical and microbiological cure
and ventriculitis by routine use of intraventricular rates, often in association with other drugs. At the same
536 A. Di Paolo et al.

time, meropenem still represents an antimicrobial of choice strategies have been proposed. Among them, alternative
in several infections because of the achievement of T[MIC routes of drug administration or BBB disruption appear to
values associated with a bactericidal effect even in the be associated with efficient penetration of antibiotics into
presence of a reduced CSF disposition. For beta-lactams, the CSF. However, the duration of this effect seems to be
alternative regimens (i.e., continuous instead of short limited and complete information is still lacking.
infusions) could give a significant advantage in terms of
cure rates. Furthermore, even these drugs have a time- Acknowledgments No source of funding was used in the prepara-
tion of this review. The authors have no conflict of interest with
dependent bactericidal pattern (for which the T[MIC respect to the manuscript.
parameter should be the best predictor of treatment effec-
tiveness) and clinical pharmacological studies suggest that
their efficacy could be monitored and predicted by calcu- References
lating the Cmax/MIC ratio.
In comparison with beta-lactams, other classes of anti- 1. Taylor EM. The impact of efflux transporters in the brain on the
bacterial drugs display better penetration into CSF, as in development of drugs for CNS disorders. Clin Pharmacokinet.
2002;41(2):81–92.
the case of fluoroquinolones, antitubercular drugs, and 2. Nag S, Begley DJ. Blood–brain barrier, exchange of metabolites
other compounds, such as linezolid, for which the CSF/ and gases. In: Kalimo H (ed) Cerebrovascular diseases. P.a.
plasma ratio is greater than 0.8 or is approaching 1 in the genetics. Basel; ISN Neuropath. Press: 2005. p. 22–29.
case of meningitis. In spite of these relevant characteristics, 3. Abbott NJ, Patabendige AA, Dolman DE, et al. Structure and
function of the blood–brain barrier. Neurobiol Dis. 2010;37(1):
it is well known that some drugs display a significant in- 13–25.
terpatient variability that becomes evident from a survey of 4. Brown PD, Davies SL, Speake T, et al. Molecular mechanisms
literature and that could be responsible for individual of cerebrospinal fluid production. Neuroscience. 2004;129(4):
treatment failures. Among the other drugs, colistin repre- 957–70.
5. Abbott NJ. Evidence for bulk flow of brain interstitial fluid:
sents a useful option for the treatment of multidrug-resis- significance for physiology and pathology. Neurochem Int.
tant strains of P. aeruginosa, Acinetobacter, and 2004;45(4):545–52.
Klebsiella. However, due to its poor penetration into CSF, 6. Dolman D, Drndarski S, Abbott NJ, et al. Induction of aquaporin
it often should be administered intrathecally while a 1 but not aquaporin 4 messenger RNA in rat primary brain
microvessel endothelial cells in culture. J Neurochem. 2005;
loading dose should be considered. 93(4):825–33.
Concerning the drugs under evaluation, the best way to 7. Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial
measure their distribution includes the calculation of the interactions at the blood–brain barrier. Nat Rev Neurosci.
ratio between AUC-time curves in CSF and in serum, 2006;7(1):41–53.
8. Redzic Z. Molecular biology of the blood–brain and the blood–
although a dense sampling schedule is not always feasible. cerebrospinal fluid barriers: similarities and differences. Fluids
Similarly, the CSF/plasma ratio, obtained from single time Barriers CNS. 2011;8(1):3.
point concentrations, is susceptible to misinterpretation 9. Hawkins BT, Davis TP. The blood–brain barrier/neurovascular
when there is a delayed passage of drugs into the liquor unit in health and disease. Pharmacol Rev. 2005;57(2):173–85.
10. de Boer AG, Gaillard PJ. Strategies to improve drug delivery
(hysteresis phenomenon). Indeed, the penetration of anti- across the blood–brain barrier. Clin Pharmacokinet. 2007;
biotics from serum into CSF is a complex pharmacokinetic 46(7):553–76.
process, because the concentrations of antibiotics in CSF 11. Ehrlich P. Über das Sauerstoffbedürfnis des Organismus. Eine
are not always parallel to those in serum. The elimination farbenanalytische Studie,1885, Berlin.
12. Nau R, Sorgel F, Eiffert H. Penetration of drugs through the
of an antibacterial from CSF may be two or three times blood–cerebrospinal fluid/blood–brain barrier for treatment of
slower than from serum, while the CSF undergoes a con- central nervous system infections. Clin Microbiol Rev.
centration process when it passes from ventricles to the 2010;23(4):858–83.
spinal cord. Finally, technical issues (i.e., methods of 13. Goldmann EE, Vitalfärbung am Zentralnervensystem, ed. A.P.i.
Akad1913: Wiss. Phys. Math. Kl.
measurement, EVD vs lumbar puncture for CSF sampling) 14. McComb JG. Recent research into the nature of cerebrospinal
are further points that should be taken into consideration. fluid formation and absorption. J Neurosurg. 1983;59(3):
The attained concentrations of antimicrobial drugs in 369–83.
CSF are influenced not only by the degree of meningeal 15. Ghersi-Egea JF, Leininger-Muller B, Cecchelli R, et al. Blood–
brain interfaces: relevance to cerebral drug metabolism. Toxicol
inflammation but also by the method of administration and Lett. 1995;82–83:645–53.
the dose regimens adopted. Further studies are required to 16. Loscher W, Potschka H. Blood–brain barrier active efflux
elucidate the relationship between the concentrations and transporters: ATP-binding cassette gene family. NeuroRx.
the bacterial killing rates for available and novel 2005;2(1):86–98.
17. Miller DS, Bauer B, Hartz AM. Modulation of P-glycoprotein at
antibiotics. the blood–brain barrier: opportunities to improve central ner-
In order to facilitate the entry and attainment of bacte- vous system pharmacotherapy. Pharmacol Rev. 2008;60(2):
ricidal concentrations of antimicrobials in the CNS, novel 196–209.
Pharmacokinetics of Antibacterials in CSF 537

18. Quagliarello VJ, Wispelwey B, Long WJ Jr, et al. Recombinant adult bacterial meningitis. Int J Antimicrob Agents. 2003;
human interleukin-1 induces meningitis and blood–brain barrier 21(5):5.
injury in the rat. Characterization and comparison with tumor 37. Fernandez A, Cabellos C, Tubau F, et al. Experimental study of
necrosis factor. J Clin Invest. 1991;87(4):1360–6. teicoplanin, alone and in combination, in the therapy of cepha-
19. Scheld WM, Dacey RG, Winn HR, et al. Cerebrospinal fluid losporin-resistant pneumococcal meningitis. J Antimicrob Che-
outflow resistance in rabbits with experimental meningitis. mother. 2005;55(1):78–83.
Alterations with penicillin and methylprednisolone. J Clin 38. Li J, Wang LN. Zheng HY. J Eur Acad Dermatol Venereol:
Invest. 1980;66(2):243–53. Jarisch-Herxheimer reaction among syphilis patients in China;
20. Bauer B, Hartz AM, Miller DS. Tumor necrosis factor alpha and 2012.
endothelin-1 increase P-glycoprotein expression and transport 39. Yang CJ, Lin YH, Lee HC, Ko WC, Liao CH, Wu CH, Hsieh
activity at the blood–brain barrier. Mol Pharmacol. 2007;71(3): CY, Wu PY, Liu WC, Chang YC, Hung CC. Jarisch-Herxheimer
667–75. reaction after penicillin therapy among patients with syphilis in
21. Roberts DJ, Goralski KB. A critical overview of the influence of the era of the HIV infection epidemic: incidence and risk fac-
inflammation and infection on P-glycoprotein expression and tors. Clin Infect Dis. 2010;51(8):9.
activity in the brain. Expert Opin Drug Metab Toxicol. 2008; 40. Fekade D, Knox K, Hussein K, et al. Prevention of Jarisch-
4(10):1245–64. Herxheimer reactions by treatment with antibodies against
22. Dumas N, Seguela JP, Giroud JP. Acute nonspecific inflamma- tumor necrosis factor alpha. N Engl J Med. 1996;335(5):311–5.
tory reaction and modification of resistance to Toxoplasma 41. Gudjonsson HSkog E. The effect of prednisolone on the Jarisch-
gondii. Bull Soc Pathol Exot Filiales. 1984;77(2):190–5. Herxheimer reaction. Acta Derm Venereol. 1968;48(1):15–8.
23. Roberts JA, Lipman J (2009) Pharmacokinetic issues for anti- 42. Butler T, Jones PK, Wallace CK. Borrelia recurrent is infection:
biotics in the critically ill patient. Crit Care Med. 37(3):840–51 single-dose antibiotic regimens and management of the Jarisch-
(quiz 859). Herxheimer reaction. J Infect Dis. 1978;137(5):5.
24. Pea F. Plasma pharmacokinetics of antimicrobial agents in 43. Arlotti M, Grossi P, Pea F, et al. Consensus document on con-
critically ill patients. Curr Clin Pharmacol. 2013;8(1):5–12. troversial issues for the treatment of infections of the central
25. Hayashi Y, Lipman J, Udy AA, et al. Beta-lactam therapeutic nervous system: bacterial brain abscesses. Int J Infect Dis.
drug monitoring in the critically ill: optimising drug exposure in 2010;14(Suppl 4):S79–92.
patients with fluctuating renal function and hypoalbuminaemia. 44. Sjolin J, Eriksson N, Arneborn P, et al. Penetration of cefotax-
Int J Antimicrob Agents. 2013;41(2):162–6. ime and desacetylcefotaxime into brain abscesses in humans.
26. van de Beek D, de Gans J, Spanjaard L, et al. Clinical features Antimicrob Agents Chemother. 1991;35(12):2606–10.
and prognostic factors in adults with bacterial meningitis. 45. Green HT, O’Donoghue MA, Shaw MD, Dowling C. Penetra-
N Engl J Med. 2004;351(18):1849–59. tion of ceftazidime into intracranial abscess. J Antimicrob
27. Brouwer MC, van de Beek D. Bacterial meningitis. Ned Chemother. 1989;24(3):6.
Tijdschr Tandheelkd. 2012;119(5):238–42. 46. Asensi V, Carton JA, Maradona JA, et al. Therapy of brain
28. Eckburg PB, Montoya JG, Vosti KL. Brain abscess due to Lis- abscess with imipenem—a safe therapeutic choice? J Antimic-
teria monocytogenes: five cases and a review of the literature. rob Chemother. 1996;37(1):200–3.
Medicine (Baltimore). 2001;80(4):223–35. 47. Malacarne P, Viaggi B, Di Paolo A, et al. Linezolid cerebro-
29. Giamarellou H. Multidrug-resistant Gram-negative bacteria: spinal fluid concentration in central nervous system infection.
how to treat and for how long. Int J Antimicrob Agents. J Chemother. 2007;19(1):90–3.
2010;36(Suppl 2):S50–4. 48. Battal B, Kocaoglu M, Bulakbasi N, et al. Cerebrospinal fluid
30. Falagas ME, Kastoris AC, Kapaskelis AM, et al. Fosfomycin for flow imaging by using phase-contrast MR technique. Br J
the treatment of multidrug-resistant, including extended-spec- Radiol. 2011;84(1004):8.
trum beta-lactamase producing, Enterobacteriaceae infections: a 49. Sommer JB, Gaul C, Heckmann J, et al. Does lumbar cerebro-
systematic review. Lancet Infect Dis. 2010;10(1):43–50. spinal fluid reflect ventricular cerebrospinal fluid? A prospective
31. Chaudhuri A, Martinez-Martin P, Kennedy PG, et al. EFNS study in patients with external ventricular drainage. Eur Neurol.
guideline on the management of community-acquired bacterial 2002;47(4):224–32.
meningitis: report of an EFNS Task Force on acute bacterial 50. Nau R, Sorgel F, Prange HW. Pharmacokinetic optimisation of
meningitis in older children and adults. Eur J Neurol. the treatment of bacterial central nervous system infections. Clin
2008;15(7):649–59. Pharmacokinet. 1998;35(3):223–46.
32. de Gans J, van de Beek D. European Dexamethasone in 51. Bakken JS, Bruun JN, Gaustad P, et al. Penetration of amoxi-
Adulthood Bacterial Meningitis Study Investigators. Dexa- cillin and potassium clavulanate into the cerebrospinal fluid of
methasone in adults with bacterial meningitis. NEJM. 2002; patients with inflamed meninges. Antimicrob Agents Chemo-
347(20):8. ther. 1986;30(3):481–4.
33. Brouwer MC, McIntyre P, de Gans J, et al. Corticosteroids for 52. Viaggi B, Di Paolo A, Danesi R, et al. Linezolid in the central
acute bacterial meningitis. Cochrane Database Syst Rev. nervous system: comparison between cerebrospinal fluid and
2010;9:CD004405. plasma pharmacokinetics. Scand J Infect Dis. 2011;43(9):721–7.
34. Ricard JD, Wolff M, Lacherade JC, et al. Levels of vancomycin 53. Nau R, Lassek C, Kinzig-Schippers M, et al. Disposition and
in cerebrospinal fluid of adult patients receiving adjunctive elimination of meropenem in cerebrospinal fluid of hydroce-
corticosteroids to treat pneumococcal meningitis: a prospective phalic patients with external ventriculostomy. Antimicrob
multicenter observational study. Clin Infect Dis. 2007; Agents Chemother. 1998;42(8):2012–6.
44(2):250–5. 54. Tsumura R, Ikawa K, Morikawa N, et al. The pharmacokinetics
35. Gaillard JL, Abadie V, Cheron G, et al. Concentrations of cef- and pharmacodynamics of meropenem in the cerebrospinal fluid
triaxone in cerebrospinal fluid of children with meningitis of neurosurgical patients. J Chemother. 2008;20(5):615–21.
receiving dexamethasone therapy. Antimicrob Agents Chemo- 55. Imberti R, Cusato M, Accetta G, et al. Pharmacokinetics of
ther. 1994;38(5):1209–10. colistin in cerebrospinal fluid after intraventricular administra-
36. Buke AC, Karasulu E, Karakartal G. Does dexamethasone affect tion of colistin methanesulfonate. Antimicrob Agents Chemo-
ceftriaxone (corrected) penetration into cerebrospinal fluid in ther. 2012;56(8):4416–21.
538 A. Di Paolo et al.

56. Cook AM, Mieure KD, Owen RD, et al. Intracerebroventricular and broth culture media. Diagn Microbiol Infect Dis. 2000;
administration of drugs. Pharmacotherapy. 2009;29(7):832–45. 36(4):241–7.
57. Adembri C, Fallani S, Cassetta MI, et al. Linezolid pharmaco- 76. Sande MA, Korzeniowski OM, Allegro GM, Brennan RO, Zak
kinetic/pharmacodynamic profile in critically ill septic patients: O, Scheld WM. Intermittent or continuous therapy of experi-
intermittent versus continuous infusion. Int J Antimicrob mental meningitis due to Streptococcus pneumoniae in rabbits:
Agents. 2008;31(2):122–9. preliminary observations on the postantibiotic effect in vivo.
58. Thea D, Barza M. Use of antibacterial agents in infections of the Rev Infect Dis. 1981;3(1):12.
central nervous system. Infect Dis Clin North Am. 1989;3(3):8. 77. Tauber MG, Zak O, Scheld WM, et al. The postantibiotic effect
59. Sinner SW, Tunkel AR. Antimicrobial agents in the treatment of in the treatment of experimental meningitis caused by Strepto-
bacterial meningitis. Infect Dis Clin North Am. 2004;18(3):12. coccus pneumoniae in rabbits. J Infect Dis. 1984;149(4):575–83.
60. Levison ME. Pharmacodynamic of antimicrobial drugs. Infect 78. Meng X, Pei Y, Nightingale CH, et al. Determination of the
Dis Clin North Am. 2004;18(3):15. in vivo post-antibiotic effects of ciprofloxacin and rifampicin.
61. Volpe JJ. Specialized studies in the neurological evaluation. In: J Antimicrob Chemother. 1995;36(6):987–96.
Neurology of the newborn. Philadelphia; W. B. Saunders 79. Wilson HD, Haltalin KC. Ampicillin in Haemophilus influenzae
Company: 2001. p. 134–177. meningitis. Clinicopharmacologic evaluation of intramuscular
62. Blennow K, Fredman P, Wallin A, et al. Protein analysis in vs intravenous administration. Am J Dis Child. 1975;129(2):
cerebrospinal fluid. II. Reference values derived from healthy 208–15.
individuals 18–88 years of age. Eur Neurol. 1993;33(2):129–33. 80. Nau R, Prange HW, Muth P, et al. Passage of cefotaxime and
63. May C, Kaye JA, Atack JR, Schapiro MB, Friedland RP, ceftriaxone into cerebrospinal fluid of patients with uninflamed
Rapoport SI. Cerebrospinal fluid production is reduced in meninges. Antimicrob Agents Chemother. 1993;37(7):1518–24.
healthy aging. Neurology. 1990;40(3 Pt 1):4. 81. Sauermann R, Schwameis R, Fille M, et al. Cerebrospinal fluid
64. Allegaert K, Scheers I, Adams E, et al. Cerebrospinal fluid impairs antimicrobial activity of fosfomycin in vitro. J Anti-
compartmental pharmacokinetics of amikacin in neonates. An- microb Chemother. 2009;64(4):821–3.
timicrob Agents Chemother. 2008;52(6):1934–9. 82. Zwijnenburg PJ, van der Poll T, Roord JJ, et al. Chemotactic
65. Kohlhepp SJ, Gilbert DN, Leggett JE. Influence of assay factors in cerebrospinal fluid during bacterial meningitis. Infect
methodology on the measurement of free serum ceftriaxone Immun. 2006;74(3):1445–51.
concentrations. Antimicrob Agents Chemother. 1998;42(9): 83. Tanaka M, Hoshino K, Hohmura M, et al. Effect of growth
2259–61. conditions on antimicrobial activity of DU-6859a and its bac-
66. Lodise TP, Nau R, Kinzig M, et al. Pharmacodynamics of ce- tericidal activity determined by the killing curve method. J An-
ftazidime and meropenem in cerebrospinal fluid: results of timicrob Chemother. 1996;37(6):1091–102.
population pharmacokinetic modelling and Monte Carlo simu- 84. Schmidt T, Froula JTauber MG. Clarithromycin lacks bacteri-
lation. J Antimicrob Chemother. 2007;60(5):1038–44. cidal activity in cerebrospinal fluid in experimental pneumo-
67. Decazes JM, Ernst JD, Sande MA. Correlation of in vitro time- coccal meningitis. J Antimicrob Chemother. 1993;32(4):627–32.
kill curves and kinetics of bacterial killing in cerebrospinal fluid 85. Kuroda M, Kusuhara H, Endou H, et al. Rapid elimination of
during ceftriaxone therapy of experimental Escherichia coli cefaclor from the cerebrospinal fluid is mediated by a benzyl-
meningitis. Antimicrob Agents Chemother. 1983;24(4):463–7. penicillin-sensitive mechanism distinct from organic anion
68. Tauber MG, Kunz S, Zak O, et al. Influence of antibiotic dose, transporter 3. J Pharmacol Exp Ther. 2005;314(2):855–61.
dosing interval, and duration of therapy on outcome in experi- 86. Tomaselli F, Maier A, Matzi V, et al. Penetration of meropenem
mental pneumococcal meningitis in rabbits. Antimicrob Agents into pneumonic human lung tissue as measured by in vivo micro-
Chemother. 1989;33(4):418–23. dialysis. Antimicrob Agents Chemother. 2004;48(6):2228–32.
69. Lutsar I, Ahmed A, Friedland IR, et al. Pharmacodynamics and 87. Nicasio AM, Ariano RE, Zelenitsky SA, et al. Population
bactericidal activity of ceftriaxone therapy in experimental pharmacokinetics of high-dose, prolonged-infusion cefepime in
cephalosporin-resistant pneumococcal meningitis. Antimicrob adult critically ill patients with ventilator-associated pneumonia.
Agents Chemother. 1997;41(11):2414–7. Antimicrob Agents Chemother. 2009;53(4):1476–81.
70. Rodriguez-Cerrato V, McCoig CC, Michelow IC, et al. Phar- 88. Ratilal BO, Costa J, Sampaio C, Pappamikail L. Antibiotic
macodynamics and bactericidal activity of moxifloxacin in prophylaxis for preventing meningitis in patients with basilar
experimental Escherichia coli meningitis. Antimicrob Agents skull fractures. Cochrane Database Syst Rev 2011;8:CD004884.
Chemother. 2001;45(11):3092–7. 89. Burgess DS, Frei CR, Lewis Ii JS, et al. The contribution of
71. Lutsar I, Friedland IR, Wubbel L, et al. Pharmacodynamics of pharmacokinetic-pharmacodynamic modelling with Monte
gatifloxacin in cerebrospinal fluid in experimental cephalospo- Carlo simulation to the development of susceptibility break-
rin-resistant pneumococcal meningitis. Antimicrob Agents points for Neisseria meningitidis. Clin Microbiol Infect. 2007;
Chemother. 1998;42(10):2650–5. 13(1):33–9.
72. Nadler HL, Pitkin DH, Sheikh W. The postantibiotic effect of 90. (EUCAST) ECoAST. Clinical breakpoints (online). http://
meropenem and imipenem on selected bacteria. J Antimicrob www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Disk_
Chemother. 1989;24(Suppl A):225–31. test_documents/EUCAST_Breakpoint_table_v_3.0.pdf. Acces-
73. Zhanel GG, Noreddin AM. Pharmacokinetics and pharmaco- sed 2013 Jan 18.
dynamics of the new fluoroquinolones: focus on respiratory 91. Cacho C, Brito B, Palacios J, et al. Speciation of nickel by
infections. Curr Opin Pharmacol. 2001;1(5):5. HPLC–UV/MS in pea nodules. Talanta. 2010;83(1):78–83.
74. Zhanel GG, Karlowsky JA, Davidson RJ, et al. Effect of pooled 92. Lodise TP Jr, Rhoney DH, Tam VH, et al. Pharmacodynamic
human cerebrospinal fluid on the postantibiotic effects of cefo- profiling of cefepime in plasma and cerebrospinal fluid of hos-
taxime, ciprofloxacin, and gentamicin against Escherichia coli. pitalized patients with external ventriculostomies. Diagn
Antimicrob Agents Chemother. 1992;36(5):1136–9. Microbiol Infect Dis. 2006;54(3):223–30.
75. Tessier PR, Nightingale CH, Nicolau DP. Postantibiotic effect 93. Lutsar I, Friedland IR. Pharmacokinetics and pharmacodynam-
of trovafloxacin against Streptococcus pneumoniae, Haemophi- ics of cephalosporins in cerebrospinal fluid. Clin Pharmacokinet.
lus influenzae, and Neisseria meningitidis in cerebrospinal fluid 2000;39(5):335–43.
Pharmacokinetics of Antibacterials in CSF 539

94. Rhoney DH, Tam VH, Parker D Jr, et al. Disposition of cefe- 112. Antachopoulos C, Karvanen M, Iosifidis E, et al. Serum and
pime in the central nervous system of patients with external cerebrospinal fluid levels of colistin in pediatric patients. Anti-
ventricular drains. Pharmacotherapy. 2003;23(3):310–4. microb Agents Chemother. 2010;54(9):3985–7.
95. Lodise TP Jr, Nau R, Kinzig M, et al. Comparison of the 113. Jimenez-Mejias ME, Pichardo-Guerrero C, Marquez-Rivas FJ,
probability of target attainment between ceftriaxone and cefe- et al. Cerebrospinal fluid penetration and pharmacokinetic/
pime in the cerebrospinal fluid and serum against Streptococcus pharmacodynamic parameters of intravenously administered
pneumoniae. Diagn Microbiol Infect Dis. 2007;58(4):445–52. colistin in a case of multidrug-resistant Acinetobacter baumannii
96. Jiménez Palacios FJ, Callejon Mochon M, Jiménez Sànchez JC. meningitis. Eur J Clin Microbiol Infect Dis. 2002;21(3):212–4.
Validation of an HPLC method for determination of cefepime (a 114. Markantonis SL, Markou N, Fousteri M, et al. Penetration of
fourth-generation cephalosporin). Determination in human colistin into cerebrospinal fluid. Antimicrob Agents Chemother.
serum, cerebrospinal fluid, and urine. Pharmacokinetic profiles. 2009;53(11):4907–10.
Chromatographica. 2005;62(7/8):7. 115. Tunkel AR, Hartman BJ, Kaplan SL, et al. Practice guidelines
97. Latif R, Dajani AS. Ceftriaxone diffusion into cerebrospinal for the management of bacterial meningitis. Clin Infect Dis.
fluid of children with meningitis. Antimicrob Agents Chemo- 2004;39(9):1267–84.
ther. 1983;23(1):46–8. 116. Sbrana F, Malacarne P, Viaggi B, et al. Carbapenem-sparing
98. Norrby SR. Neurotoxicity of carbapenem antibiotics: conse- antibiotic regimens for infections caused by Klebsiella pneu-
quences for their use in bacterial meningitis. J Antimicrob moniae carbapenemase-producing K. pneumoniae in intensive
Chemother. 2000;45(1):5–7. care unit. Clin Infect Dis. 2013;56(5):697–700.
99. Zhanel GG, Simor AE, Vercaigne L, et al. Imipenem and me- 117. Plachouras D, Karvanen M, Friberg LE, et al. Population
ropenem: comparison of in vitro activity, pharmacokinetics, pharmacokinetic analysis of colistin methanesulfonate and
clinical trials and adverse effects. Can J Infect Dis. 1998;9(4): colistin after intravenous administration in critically ill patients
215–28. with infections caused by gram-negative bacteria. Antimicrob
100. Zhanel GG, Wiebe R, Dilay L, et al. Comparative review of the Agents Chemother. 2009;53(8):3430–6.
carbapenems. Drugs. 2007;67(7):1027–52. 118. Periti P, Mazzei T, Mini E, et al. Clinical pharmacokinetic
101. Li C, Kuti JL, Nightingale CH, et al. Population pharmacoki- properties of the macrolide antibiotics. Effects of age and var-
netic analysis and dosing regimen optimization of meropenem in ious pathophysiological states (Part II). Clin Pharmacokinet.
adult patients. J Clin Pharmacol. 2006;46(10):1171–8. 1989;16(5):261–82.
102. Gaillard JL, Silly C, Le Masne A, et al. Cerebrospinal fluid 119. Periti P, Mazzei T, Mini E, et al. Clinical pharmacokinetic
penetration of amikacin in children with community-acquired properties of the macrolide antibiotics. Effects of age and var-
bacterial meningitis. Antimicrob Agents Chemother. 1995; ious pathophysiological states (Part I). Clin Pharmacokinet.
39(1):253–5. 1989;16(4):193–214.
103. Eisenhut M, Meehan TBatchelor L. Cerebrospinal fluid glucose 120. Jaruratanasirikul S, Hortiwakul R, Tantisarasart T, et al. Dis-
levels and sensorineural hearing loss in bacterial meningitis. tribution of azithromycin into brain tissue, cerebrospinal fluid,
Infection. 2003;31(4):247–50. and aqueous humor of the eye. Antimicrob Agents Chemother.
104. Alffenaar JW, van Altena R, Bokkerink HJ, et al. Pharmacoki- 1996;40(3):825–6.
netics of moxifloxacin in cerebrospinal fluid and plasma in 121. Williams JD. Spectrum of activity of azithromycin. Eur J Clin
patients with tuberculous meningitis. Clin Infect Dis. 2009; Microbiol Infect Dis. 1991;10(10):813–20.
49(7):1080–2. 122. Peters DH, Friedel HA, McTavish D. Azithromycin. A review of
105. Kanellakopoulou K, Pagoulatou A, Stroumpoulis K, et al. its antimicrobial activity, pharmacokinetic properties and clini-
Pharmacokinetics of moxifloxacin in non-inflamed cerebrospi- cal efficacy. Drugs. 1992;44(5):750–99.
nal fluid of humans: implication for a bactericidal effect. J An- 123. Baschiera F, Fornai M, Lazzeri G, et al. Improved tonsillar dispo-
timicrob Chemother. 2008;61(6):1328–31. sition of azithromycin following a 3-day oral treatment with 20 mg/
106. Nijland HM, Ruslami R, Suroto AJ, et al. Rifampicin reduces kg in paediatric patients. Pharmacol Res. 2002;46(1):95–100.
plasma concentrations of moxifloxacin in patients with tuber- 124. Lucchi M, Damle B, Fang A, et al. Pharmacokinetics of azith-
culosis. Clin Infect Dis. 2007;45(8):1001–7. romycin in serum, bronchial washings, alveolar macrophages
107. Shandil RK, Jayaram R, Kaur P, et al. Moxifloxacin, ofloxacin, and lung tissue following a single oral dose of extended or
sparfloxacin, and ciprofloxacin against Mycobacterium tuber- immediate release formulations of azithromycin. J Antimicrob
culosis: evaluation of in vitro and pharmacodynamic indices that Chemother. 2008;61(4):884–91.
best predict in vivo efficacy. Antimicrob Agents Chemother. 125. Di Paolo A, Barbara C, Chella A, Angeletti CA, Del Tacca M.
2007;51(2):576–82. Pharmacokinetics of azithromycin in lung tissue, bronchial
108. Pranger AD, Alffenaar JW, Wessels AM, et al. Determination of washing, and plasma in patients given multiple oral doses of 500
moxifloxacin in human plasma, plasma ultrafiltrate, and cere- and 1000 mg daily. Pharmacol Res. 2002;46(6):6.
brospinal fluid by a rapid and simple liquid chromatography– 126. Wallace RJ Jr, Meier A, Brown BA, et al. Genetic basis for
tandem mass spectrometry method. J Anal Toxicol. 2010;34 clarithromycin resistance among isolates of Mycobacterium
(3):135–41. chelonae and Mycobacterium abscessus. Antimicrob Agents
109. Thwaites GE, Bhavnani SM, Chau TT, et al. Randomized Chemother. 1996;40(7):1676–81.
pharmacokinetic and pharmacodynamic comparison of fluoro- 127. Ingram CW, Tanner DC, Durack DT, et al. Disseminated
quinolones for tuberculous meningitis. Antimicrob Agents infection with rapidly growing mycobacteria. Clin Infect Dis.
Chemother. 2011;55(7):3244–53. 1993;16(4):463–71.
110. Pea F, Pavan F, Nascimben E, et al. Levofloxacin disposition in 128. Maniu CV, Hellinger WC, Chu SY, et al. Failure of treatment
cerebrospinal fluid in patients with external ventriculostomy. for chronic Mycobacterium abscessus meningitis despite ade-
Antimicrob Agents Chemother. 2003;47(10):3104–8. quate clarithromycin levels in cerebrospinal fluid. Clin Infect
111. Dudhani RV, Turnidge JD, Nation RL, et al. fAUC/MIC is the Dis. 2001;33(5):745–8.
most predictive pharmacokinetic/pharmacodynamic index of 129. Fernandes PB, Bailer R, Swanson R, et al. In vitro and in vivo
colistin against Acinetobacter baumannii in murine thigh and lung evaluation of A-56268 (TE-031), a new macrolide. Antimicrob
infection models. J Antimicrob Chemother. 2010;65(9):1984–90. Agents Chemother. 1986;30(6):865–73.
540 A. Di Paolo et al.

130. Imshenetskaia VF. Erythromycin penetration into the cerebro- 149. Jaspan HB, Brothers AW, Campbell AJ, et al. Multidrug-resis-
spinal fluid of patients. Antibiotiki. 1976;21(11):1002–4. tant Enterococcus faecium meningitis in a toddler: character-
131. Petersen PJ, Jones CH, Bradford PA. In vitro antibacterial ization of the organism and successful treatment with
activities of tigecycline and comparative agents by time-kill intraventricular daptomycin and intravenous tigecycline. Pediatr
kinetic studies in fresh Mueller-Hinton broth. Diagn Microbiol Infect Dis J. 2010;29(4):379–81.
Infect Dis. 2007;59(3):347–9. 150. Tutuncu EE, Kuscu F, Gurbuz Y, et al. Tigecycline use in two
132. Noviello S, Ianniello F, Leone S, et al. In vitro activity of cases with multidrug-resistant Acinetobacter baumannii men-
tigecycline: MICs, MBCs, time-kill curves and post-antibiotic ingitis. Int J Infect Dis. 2010;14(Suppl 3):e224–6.
effect. J Chemother. 2008;20(5):577–80. 151. Lengerke C, Haap M, Mayer F, et al. Low tigecycline concen-
133. van Ogtrop ML, Andes D, Stamstad TJ, et al. In vivo pharma- trations in the cerebrospinal fluid of a neutropenic patient with
codynamic activities of two glycylcyclines (GAR-936 and WAY inflamed meninges. Antimicrob Agents Chemother. 2011;55(1):
152,288) against various gram-positive and gram-negative bac- 449–50.
teria. Antimicrob Agents Chemother. 2000;44(4):943–9. 152. Ray L, Levasseur K, Nicolau DP, et al. Cerebral spinal fluid
134. Meagher AK, Passarell JA, Cirincione BB, et al. Exposure- penetration of tigecycline in a patient with Acinetobacter bau-
response analyses of tigecycline efficacy in patients with com- mannii cerebritis. Ann Pharmacother. 2010;44(3):582–6.
plicated skin and skin-structure infections. Antimicrob Agents 153. Dandache P, Nicolau, DP, Sakoulas, G. Tigecycline for the
Chemother. 2007;51(6):1939–45. treatment of multidrug-resistant Klebsiella pneumoniae menin-
135. Passarell JA, Meagher AK, Liolios K, et al. Exposure-response gitis. Infect Dis Clin Pract. 2009;17(Abstract):66.
analyses of tigecycline efficacy in patients with complicated 154. van de Beek D, Brouwer M, Thwaites GE, Tunkel AR.
intra-abdominal infections. Antimicrob Agents Chemother. Advances in treatment of bacterial meningitis. Lancet. 2012;
2008;52(1):204–10. 380(9854):10.
136. Agwuh KN, MacGowan A. Pharmacokinetics and pharmaco- 155. Chien JW, Kucia ML, Salata RA. Use of linezolid, an oxazo-
dynamics of the tetracyclines including glycylcyclines. J Anti- lidinone, in the treatment of multidrug-resistant gram-positive
microb Chemother. 2006;58(2):256–65. bacterial infections. Clin Infect Dis. 2000;30(1):146–51.
137. Kang-Birken SL, Castel UPrichard JG. Oral doxycycline for 156. Andes D, van Ogtrop ML, Peng J, et al. In vivo pharmacody-
treatment of neurosyphilis in two patients infected with human namics of a new oxazolidinone (linezolid). Antimicrob Agents
immunodeficiency virus. Pharmacotherapy. 2010;30(4):119e– Chemother. 2002;46(11):3484–9.
22e. 157. Beer R, Engelhardt KW, Pfausler B, et al. Pharmacokinetics of
138. Psomas KC, Brun M, Causse A, et al. Efficacy of ceftriaxone intravenous linezolid in cerebrospinal fluid and plasma in neu-
and doxycycline in the treatment of early syphilis. Med Mal rointensive care patients with staphylococcal ventriculitis asso-
Infect. 2012;42(1):15–9. ciated with external ventricular drains. Antimicrob Agents
139. Johnson SE, Klein GC, Schmid GP, et al. Susceptibility of the Chemother. 2007;51(1):379–82.
Lyme disease spirochete to seven antimicrobial agents. Yale J 158. Yogev R, Damle B, Levy G, et al. Pharmacokinetics and dis-
Biol Med. 1984;57(4):549–53. tribution of linezolid in cerebrospinal fluid in children and
140. Bernardino AL, Kaushal D, Philipp MT. The antibiotics doxy- adolescents. Pediatr Infect Dis J. 2010;29(9):827–30.
cycline and minocycline inhibit the inflammatory responses to 159. Di Paolo A, Malacarne P, Guidotti E, et al. Pharmacological
the Lyme disease spirochete Borrelia burgdorferi. J Infect Dis. issues of linezolid: an updated critical review. Clin Pharmaco-
2009;199(9):1379–88. kinet. 2010;49(7):439–47.
141. Andersson H, Alestig K. The penetration of doxycycline into 160. Myrianthefs P, Markantonis SL, Vlachos K, et al. Serum and
CSF. Scand J Infect Dis Suppl. 1976;9:17–9. cerebrospinal fluid concentrations of linezolid in neurosur-
142. Yim CW, Flynn NM, Fitzgerald FT. Penetration of oral doxy- gical patients. Antimicrob Agents Chemother. 2006;50(12):
cycline into the cerebrospinal fluid of patients with latent or 3971–6.
neurosyphilis. Antimicrob Agents Chemother. 1985;28(2): 161. Gandelman K, Zhu T, Fahmi OA, et al. Unexpected effect of
347–8. rifampin on the pharmacokinetics of linezolid: in silico and
143. Karlsson M, Hammers S, Nilsson-Ehle I, et al. Concentrations of in vitro approaches to explain its mechanism. J Clin Pharmacol.
doxycycline and penicillin G in sera and cerebrospinal fluid of 2011;51(2):229–36.
patients treated for neuroborreliosis. Antimicrob Agents Che- 162. Tsuji Y, Hiraki Y, Matsumoto K, et al. Pharmacokinetics and
mother. 1996;40(5):1104–7. protein binding of linezolid in cerebrospinal fluid and serum in a
144. Dotevall L, Hagberg L. Penetration of doxycycline into cere- case of post-neurosurgical bacterial meningitis. Scand J Infect
brospinal fluid in patients treated for suspected Lyme neuro- Dis. 2011;43(11–12):982–5.
borreliosis. Antimicrob Agents Chemother. 1989;33(7): 163. Safdar N, Andes D, Craig WA. In vivo pharmacodynamic
1078–80. activity of daptomycin. Antimicrob Agents Chemother.
145. Petersen PJ, Ruzin A, Tuckman M, et al. In vitro activity of 2004;48(1):63–8.
tigecycline against patient isolates collected during phase 3 164. Cottagnoud P, Pfister M, Acosta F, et al. Daptomycin is highly
clinical trials for diabetic foot infections. Diagn Microbiol Infect efficacious against penicillin-resistant and penicillin- and quin-
Dis. 2010;66(4):407–18. olone-resistant pneumococci in experimental meningitis. Anti-
146. Rodvold KA, Gotfried MH, Cwik M, et al. Serum, tissue and microb Agents Chemother. 2004;48(10):3928–33.
body fluid concentrations of tigecycline after a single 100 mg 165. Riser MS, Bland CM, Rudisill CN, et al. Cerebrospinal fluid
dose. J Antimicrob Chemother. 2006;58(6):1221–9. penetration of high-dose daptomycin in suspected Staphylo-
147. Van Wart SA, Cirincione BB, Ludwig EA, et al. Population coccus aureus meningitis. Ann Pharmacother. 2010;44(11):
pharmacokinetics of tigecycline in healthy volunteers. J Clin 1832–5.
Pharmacol. 2007;47(6):727–37. 166. Kullar R, Chin JN, Edwards DJ, et al. Pharmacokinetics of
148. Wadi JA, Al Rub MA. Multidrug resistant Acinetobacter nos- single-dose daptomycin in patients with suspected or confirmed
ocomial meningitis treated successfully with parenteral tigecy- neurological infections. Antimicrob Agents Chemother. 2011;
cline. Ann Saudi Med. 2007;27(6):456–8. 55(7):3505–9.
Pharmacokinetics of Antibacterials in CSF 541

167. Brouwer MC, Tunkel AR, van de Beek D. Epidemiology, 185. Donald PR, Seifart HI. Cerebrospinal fluid concentrations of
diagnosis, and antimicrobial treatment of acute bacterial men- ethionamide in children with tuberculous meningitis. J Pediatr.
ingitis. Clin Microbiol Rev. 2010;23(3):467–92. 1989;115(3):483–6.
168. Jourdan C, Convert J, Peloux A, et al. Adequate intrathecal 186. Baron H, Epstein IG, Mulinos MG, et al. Absorption, distribu-
diffusion of teicoplanin after failure of vancomycin, adminis- tion, and excretion of cycloserine in man. Antibiot Annu.
tered in continuous infusion in three cases of shunt associated 1955;3:136–40.
meningitis. Pathol Biol (Paris). 1996;44(5):389–92. 187. Pfausler B, Spiss H, Dittrich P, et al. Concentrations of fosfo-
169. Cataldo MA, Tacconelli E, Grilli E, et al. Continuous versus mycin in the cerebrospinal fluid of neurointensive care patients
intermittent infusion of vancomycin for the treatment of Gram- with ventriculostomy-associated ventriculitis. J Antimicrob
positive infections: systematic review and meta-analysis. J An- Chemother. 2004;53(5):848–52.
timicrob Chemother. 2012;67(1):17–24. 188. Dudley MN, Levitz RE, Quintiliani R, et al. Pharmacokinetics
170. Pea F, Brollo L, Viale P, et al. Teicoplanin therapeutic drug of trimethoprim and sulfamethoxazole in serum and cerebro-
monitoring in critically ill patients: a retrospective study spinal fluid of adult patients with normal meninges. Antimicrob
emphasizing the importance of a loading dose. J Antimicrob Agents Chemother. 1984;26(6):811–4.
Chemother. 2003;51(4):971–5. 189. Misra A, Ganesh S, Shahiwala A, et al. Drug delivery to the
171. Wang Q, Shi Z, Wang J, et al. Postoperatively administered central nervous system: a review. J Pharm Pharm Sci. 2003;
vancomycin reaches therapeutic concentration in the cerebral 6(2):252–73.
spinal fluid of neurosurgical patients. Surg Neurol. 2008;69(2): 190. Tiwari SB, Amiji MM. A review of nanocarrier-based CNS
126–9 (discussion 129). delivery systems. Curr Drug Deliv. 2006;3(2):219–32.
172. Jorgenson L, Reiter PD, Freeman JE, et al. Vancomycin 191. Witt KA, Gillespie TJ, Huber JD, et al. Peptide drug modifica-
disposition and penetration into ventricular fluid of the tions to enhance bioavailability and blood–brain barrier per-
central nervous system following intravenous therapy in meability. Peptides. 2001;22(12):2329–43.
patients with cerebrospinal devices. Pediatr Neurosurg. 2007; 192. Rautio J, Kumpulainen H, Heimbach T, et al. Prodrugs: design
43(6):449–55. and clinical applications. Nat Rev Drug Discov. 2008;7(3):
173. Budha NR, Lee RB, Hurdle JG, et al. A simple in vitro PK/PD 255–70.
model system to determine time-kill curves of drugs against 193. Lin C, Sunkara G, Cannon JB, et al. Recent advances in pro-
Mycobacteria. Tuberculosis (Edinb). 2009;89(5):378–85. drugs as drug delivery systems. Am J Ther. 2012;19(1):33–43.
174. Peloquin CA, Jaresko GS, Yong CL, et al. Population pharma- 194. Micheli MR, Bova R, Magini A, et al. Lipid-based nanocarriers
cokinetic modeling of isoniazid, rifampin, and pyrazinamide. for CNS-targeted drug delivery. Recent Pat CNS Drug Discov.
Antimicrob Agents Chemother. 1997;41(12):2670–9. 2012;7(1):71–86.
175. Donald PR, Gent WL, Seifart HI, et al. Cerebrospinal fluid 195. Liu L, Venkatraman SS, Yang YY, et al. Polymeric micelles
isoniazid concentrations in children with tuberculous meningitis: anchored with TAT for delivery of antibiotics across the blood–
the influence of dosage and acetylation status. Pediatrics. brain barrier. Biopolymers. 2008;90(5):617–23.
1992;89(2):247–50. 196. Marquet F, Tung YS, Teichert T, et al. Noninvasive, transient
176. Gumbo T, Louie A, Deziel MR, et al. Concentration-dependent and selective blood–brain barrier opening in non-human pri-
Mycobacterium tuberculosis killing and prevention of resistance mates in vivo. PLoS One. 2011;6(7):e22598.
by rifampin. Antimicrob Agents Chemother. 2007;51(11): 197. O’Reilly MA, Hynynen K. Blood–brain barrier: real-time
3781–8. feedback-controlled focused ultrasound disruption by using an
177. Nau R, Prange HW, Menck S, et al. Penetration of rifampicin acoustic emissions-based controller. Radiology. 2012;263(1):
into the cerebrospinal fluid of adults with uninflamed meninges. 96–106.
J Antimicrob Chemother. 1992;29(6):719–24. 198. Liu HL, Yang HW, Hua MY, et al. Enhanced therapeutic agent
178. Ellard GA, Humphries MJ, Gabriel M, et al. Penetration of delivery through magnetic resonance imaging-monitored
pyrazinamide into the cerebrospinal fluid in tuberculous men- focused ultrasound blood–brain barrier disruption for brain
ingitis. Br Med J (Clin Res Ed). 1987;294(6567):284–5. tumor treatment: an overview of the current preclinical status.
179. Phuapradit P, Supmonchai K, Kaojarern S, et al. The blood/ Neurosurg Focus. 2012;32(1):E4.
cerebrospinal fluid partitioning of pyrazinamide: a study during 199. Etame AB, Diaz RJ, Smith CA, et al. Focused ultrasound disruption
the course of treatment of tuberculous meningitis. J Neurol of the blood–brain barrier: a new frontier for therapeutic delivery in
Neurosurg Psychiatry. 1990;53(1):81–2. molecular neurooncology. Neurosurg Focus. 2012;32(1):E3.
180. Gumbo T, Dona CS, Meek C, et al. Pharmacokinetics–phar- 200. Pardridge WM. Blood–brain barrier delivery. Drug Discov
macodynamics of pyrazinamide in a novel in vitro model of Today. 2007;12(1–2):54–61.
tuberculosis for sterilizing effect: a paradigm for faster assess- 201. Kumar P, Wu H, McBride JL, et al. Transvascular delivery of
ment of new antituberculosis drugs. Antimicrob Agents Che- small interfering RNA to the central nervous system. Nature.
mother. 2009;53(8):3197–204. 2007;448(7149):39–43.
181. Ellard GA, Humphries MJ, Allen BW. Cerebrospinal fluid drug 202. Dunlop EM, Al-Egaily SS, Houang ET. Penicillin levels in
concentrations and the treatment of tuberculous meningitis. Am blood and CSF achieved by treatment of syphilis. JAMA.
Rev Respir Dis. 1993;148(3):650–5. 1979;241(23):3.
182. Rastogi N, Labrousse V, Goh KS. In vitro activities of fourteen 203. van der Valk PG, Kraai EJ, van Voorst Vader PC, et al. Peni-
antimicrobial agents against drug susceptible and resistant cillin concentrations in cerebrospinal fluid (CSF) during repos-
clinical isolates of Mycobacterium tuberculosis and comparative itory treatment regimen for syphilis. Genitourin Med. 1988;
intracellular activities against the virulent H37Rv strain in 64(4):223–5.
human macrophages. Curr Microbiol. 1996;33(3):167–75. 204. Landis MS, Boyden T, Pegg S. Nasal-to-CNS drug delivery:
183. Pilheu JA, Maglio F, Cetrangolo R, et al. Concentrations of where are we now and where are we heading? An industrial
ethambutol in the cerebrospinal fluid after oral administration. perspective. Ther Deliv. 2012;3(2):195–208.
Tubercle. 1971;52(2):117–22. 205. Manda P, Hargett JK, Vaka SR, et al. Delivery of cefotaxime to
184. Hughes IE, Smith H. Ethionamide: its passage into the cere- the brain via intranasal administration. Drug Dev Ind Pharm.
brospinal fluid in man. Lancet. 1962;1(7230):616–7. 2011;37(11):1306–10.
542 A. Di Paolo et al.

206. Reesor C, Chow AW, Kureishi A, et al. Kinetics of intraven- 210. De Sarro A, Ammendola D, Zappala M, et al. Relationship
tricular vancomycin in infections of cerebrospinal fluid shunts. between structure and convulsant properties of some beta-lactam
J Infect Dis. 1988;158(5):1142–3. antibiotics following intracerebroventricular microinjection in
207. McCracken GH Jr, Mize SG, Threlkeld N. Intraventricular rats. Antimicrob Agents Chemother. 1995;39(1):232–7.
gentamicin therapy in gram-negative bacillary meningitis of 211. Arnell K, Enblad P, Wester T, et al. Treatment of cerebrospinal
infancy. Report of the Second Neonatal Meningitis Cooperative fluid shunt infections in children using systemic and intraven-
Study Group. Lancet. 1980;1(8172):787–91. tricular antibiotic therapy in combination with externalization of
208. Shah S, Ohlsson A, Shah V. Intraventricular antibiotics for the ventricular catheter: efficacy in 34 consecutively treated
bacterial meningitis in neonates. Cochrane Database Syst Rev. infections. J Neurosurg. 2007;107(3 Suppl):213–9.
2004;4:CD004496. 212. Nau R, Prange HW, Kinzig M, et al. Cerebrospinal fluid ceft-
209. Tangden T, Enblad P, Ullberg M, et al. Neurosurgical gram- azidime kinetics in patients with external ventriculostomies.
negative bacillary ventriculitis and meningitis: a retrospective Antimicrob Agents Chemother. 1996;40(3):763–6.
study evaluating the efficacy of intraventricular gentamicin 213. Tunkel AR, Glaser CA, Bloch KC, et al. The management of
therapy in 31 consecutive cases. Clin Infect Dis. 2011;52(11): encephalitis: clinical practice guidelines by the Infectious Dis-
1310–6. eases Society of America. Clin Infect Dis. 2008;47(3):303–27.

You might also like