ArticlePDF AvailableLiterature Review

Biological barriers, and the influence of protein binding on the passage of drugs across them

Authors:

Abstract and Figures

Drug-protein binding plays a key role in determining the pharmacokinetics of a drug. The distribution and protein binding ability of a drug changes over a lifetime, and are important considerations during pregnancy and lactation. Although proteins are a significant fraction in plasma composition, they also exist beyond the bloodstream and bind with drugs in the skin, tissues or organs. Protein binding influences the bioavailability and distribution of active compounds, and is a limiting factor in the passage of drugs across biological membranes and barriers: drugs are often unable to cross membranes mainly due to the high molecular mass of the drug-protein complex, thus resulting in the accumulation of the active compounds and a significant reduction of their pharmacological activity. This review describes the consequences of drug-protein binding on drug transport across physiological barriers, whose role is to allow the passage of essential substances—such as nutrients or oxygen, but not of xenobiotics. The placental barrier regulates passage of xenobiotics into a fetus and protects the unborn organism. The blood–brain barrier is the most important barrier in the entire organism and the skin separates the human body from the environment.
Content may be subject to copyright.
Vol.:(0123456789)
1 3
Molecular Biology Reports (2020) 47:3221–3231
https://doi.org/10.1007/s11033-020-05361-2
MINI REVIEW ARTICLE
Biological barriers, andtheinuence ofprotein binding
onthepassage ofdrugs acrossthem
KarolinaWanat1
Received: 28 November 2019 / Accepted: 27 February 2020 / Published online: 5 March 2020
© The Author(s) 2020
Abstract
Drug-protein binding plays a key role in determining the pharmacokinetics of a drug. The distribution and protein binding
ability of a drug changes over a lifetime, and are important considerations during pregnancy and lactation. Although proteins
are a significant fraction in plasma composition, they also exist beyond the bloodstream and bind with drugs in the skin, tis-
sues or organs. Protein binding influences the bioavailability and distribution of active compounds, and is a limiting factor
in the passage of drugs across biological membranes and barriers: drugs are often unable to cross membranes mainly due
to the high molecular mass of the drug-protein complex, thus resulting in the accumulation of the active compounds and a
significant reduction of their pharmacological activity. This review describes the consequences of drug-protein binding on
drug transport across physiological barriers, whose role is to allow the passage of essential substances—such as nutrients
or oxygen, but not of xenobiotics. The placental barrier regulates passage of xenobiotics into a fetus and protects the unborn
organism. The blood–brain barrier is the most important barrier in the entire organism and the skin separates the human
body from the environment.
Keywords Breast milk, Drug-protein binding, Skin barrier,
Protein binding, The blood–brain barrier, The placental
barrier.
Drug‑protein binding
Following absorption from the gastrointestinal system
or direct infusion into bloodstream, a drug can bind with
plasma proteins. The main proteins responsible for the bind-
ing in plasma are human serum albumin (HSA) and alpha-
1-acid glycoprotein (AAG) [13]. Their concentrations and
functions are listed in Table1 [46]. While the protein-drug
complex is relatively stable, the connection between mol-
ecules is reversible: molecules can join and separate, and the
equilibrium state is reached a few hours after the administra-
tion of a medicine [3].
The structure and properties of the drug determine the
extent of both: plasma protein binding (PPB) and protein
binding (PB) in the sense of the general process, because
these concepts should be distinguished. Lipophilicity
(described as logP) and acid–base properties have a signifi-
cant correlation with binding [7]. Hydrophobic and acidic
drugs (e.g. warfarin, ketoprofen, ibuprofen, diazepam) bind
preferably to HSA, while AAG connects with the basic
ones (e.g. bupivacaine, clindamycine) [69] which should
be taken into account while setting the therapy. Binding
can also increase the solubility of compounds, especially
hydrophobic ones, which would otherwise not be distributed
in the aqueous environment of plasma [10]. A connection
with the plasma proteins protects compounds from oxida-
tion, lowers their toxicity and increases their half-life; drugs
highly bound to the plasma proteins often reveal low first
pass-metabolism [1012]. Volume of distribution depends
from PB as well and is decreased for drugs highly bound in
plasma or increased for those which bind in tissues [1315].
In addition drugs with higher affinity to a binding site on a
plasma protein can replace one with lower affinity and such
competition can lead to an uncontrolled rise in the concen-
tration of the free, unbound fraction of a drug [16]. This
can have serious consequences for narrow therapeutic index
(NTID) drugs, where the difference between therapeutic and
toxic doses is minimal (e.g. cardenolides, carbamazepine,
phenytoin or warfarin [17, 18]) and any changes in the con-
centration of unbound, active form may be poorly tolerated
* Karolina Wanat
karolina.wanat@umed.lodz.pl
1 Department ofAnalytical Chemistry, Medical University
ofLodz, Muszyńskiego 1, 90-151, Lodz, Poland
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
3222 Molecular Biology Reports (2020) 47:3221–3231
1 3
by the organism. A sudden increase in the unbound fraction
of the drug may provide a toxic effect [19]. This can lead to
clinical consequences such as high risk of bleeding (warfa-
rin) [19] or cardiac arrest (cardenolides) [20].
Level of protein binding depends on the properties of a
drug but also on the surrounding environment for example,
the temperature or pH. The latter can change the ionization
state of the chemical compound [10, 21, 22]. The degree of
plasma protein binding is governed by two variables, these
being the unbound fraction of the drug in plasma (fu,p) and
the percentage of plasma protein binding (PPB%), as given
below in Eqs.1 and 2 [3]:
An important consideration, often omitted in the litera-
ture, is that of drug-protein binding occurring outside the
bloodstream. Compounds can bind with macromolecules
in skin, breast milk, tissues and organs including the pla-
centa [13, 23, 24], where they become ‘stuck’ and are thus
prevented from reaching site of pharmacological action
[23, 25]. These drugs may later pass into the plasma but
in an uncontrolled way, which disturbs the dosage and the
intended result of pharmacotherapy.
Transfer acrossbiological membranes
andbarriers
The cell membrane is a semipermeable phospholipid bilayer,
which separates cell organelles and cytoplasm from the envi-
ronment. The ability of a molecule to cross the membrane
depends on various factors including molecular weight, lipo-
philicity, ionisation state, the concentration on both sides
of the barrier and protein binding [26, 27]. Low-molecular,
lipid, unionised and unbound to plasma proteins molecules are
reckoned as good penetrators through membranes, although
(1)
f
u,p =
unbound drug concentration in plasma
total dose
(2)
PPB%
=
bound drug concentration in plasma
total dose
×
100%
extreme lipophilicity can cause accumulation in lipid environ-
ment [28, 29]. The mechanism of passive transport includes:
simple diffusion (the undisturbed movement of small, lipo-
philic and unionized molecules across membrane) and facili-
tated diffusion, where specialized membrane proteins transport
particles across barriers [30]. Active transport acts against the
concentration gradient and as such requires energy, which is
typically obtained by the hydrolysis of adenosine triphosphate
(ATP). One such family of membrane proteins which actively
transport drugs and other molecules across membranes is that
of the ATP-binding cassette transporters (ABC transporters).
They also contribute significantly to the passage of drugs
through the blood–brain barrier or placenta [31]. Crossing bio-
logical barriers is a far more difficult matter. Their structure is
more complex and there are additional mechanisms involved
which prevent the passage of xenobiotics. Transfer across each
barrier is explained in detail in the appropriate sections of this
review. The most important, and the most difficult to pass, is
the blood–brain barrier (BBB), which separates crucial organs
from the environment.
Binding with HSA and AAG macromolecules affects the
pharmacokinetic properties of pharmacologically-active
compounds by decreasing their bioavailability and slow-
ing their passage across biological membranes and barriers
[3234]; proteins themselves hardly penetrate through the
cell membranes [3537]. On the contrary new approaches
in target therapy also reveal that drug binding to the protein
carrier improves the effectiveness of several pharmacothera-
pies [38], e.g. a simple but effective mechanism was used
in anti-tumour pharmacotherapy. Drug-protein conjugates
penetrate into tumour circulation easily, through fenestrated
capillaries, and stay trapped inside [39]. Albumin is also
used as a protein carrier in commonly used drugs such as
levemir, methotrexate, doxorubicin or paclitaxel [40, 41].
The blood–brain barrier
The blood–brain barrier (BBB) protects the central nerv-
ous system (CNS), which controls the whole body.
Blood vessels, which are part of the BBB, are lined with
Table 1 Physicochemical properties of HSA and AAG
Plasma protein Protein family Concentration in plasma Function
Human serum albumin,
65kDa, 585 amino acids
Albumins 3.5–5g/dL Transport of compounds across the blood-
stream (mainly hydrophobic and acidic ones),
maintenance of the blood oncotic pressure,
antioxidant, anticoagulant and immunomodu-
lating properties
Alpha-1-acid glycoprotein,
44kDa, 183 amino acids
Globulins, acute phase
proteins
Depends from physiological condi-
tion
Transport of compounds across the bloodstream
(mainly ones with basic properties), AAG is
produced during the inflammatory state
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
3223Molecular Biology Reports (2020) 47:3221–3231
1 3
tightly-connected endothelial cells. These unique connec-
tions between the endothelial cells are called tight junctions
(TJs) and adherence junctions (AJs). The BBB is also com-
posed of a basement membrane, glial cells, pericytes and
surrounding neurons [42]. The close cell connections, viz.
TJs and AJs prevent the passage of molecules through the
intercellular space: transport can only take place through the
intracellular route, i.e. within the cells [43]. Further defence
is provided by unique metabolic activity of the barrier, with
enzymes such as γ-glutamyl transpeptidase (γ-GTP) or alka-
line phosphatase (AP) enabling chemical decomposition of
compounds which can cross the BBB from the bloodstream
[42, 44]. The CNS is also protected by the diversity of its
routes of xenobiotic transport mechanisms [42, 43, 45]. Of
the drug efflux transporters, i.e. those of ABC transport-
ers family P-glycoprotein (P-gp), breast cancer resistance
protein (BCRP) and multidrug resistance protein (MRP)
demonstrate the highest activity in the BBB [46]. These
transporters are responsible for drug distribution into the
CNS and they can remove compounds which cross the bar-
rier. Such efflux transporters have various substrates, includ-
ing anti-cancer drugs, such as doxorubicin or methotrexate,
antiepileptics, such as phenytoin and carbamazepine, and
antidepressants, such as venlafaxine and paroxetine. While
some drugs are not intended to act on the CNS, many others
have to penetrate the brain to reach the main site of their
activity and achieve successful therapy [46]. Drug transport
across the blood–brain barrier has been widely described by
Pardridge etal., with a series of articles providing a clear
review of various aspects of barrier structure, the transport
of drugs across it and the development of drugs for use in
the CNS [4752]. New approaches to delivering CNS drugs
are also mentioned in other recent articles [53, 54].
The blood–brain barrier protects the CNS from harm-
ful substances but its main role is to provide nutrients and
oxygen, essential for the brain structures [42]. Oxygen mol-
ecules and drugs with low molecular weight and lipophilic
properties can easily cross the BBB by simple diffusion
[55]. Nutrition such as glucose, crucial for proper CNS
function, or amino acids are carried by specific transport-
ers (e.g. GLUT1 glucose transporter); macromolecules with
high molecular mass, such as insulin, are transported in the
process of endocytosis [43, 56]. Drugs can pass through
the BBB by transmembrane diffusion, especially those that
are lightweight or with high lipophilicity, or are carried by
transporters, as in the case of glucose [55]. Two parameters
(Eqs.3 and 4) describe the amount of a drug that is passed
into the CNS: log BB and log PS [57, 58]. Log BB repre-
sents the ratio between drug concentration in the CNS and
plasma, while log PS indicates the permeability of certain
surface; while the former is easier to obtain and more intui-
tive to understand, the latter is currently receiving more
research attention [57, 58]:
A number of studies have examined protein binding with
drugs and their ability to cross the BBB [55, 5961]. Albu-
min, like other proteins, does not readily pass through the
barrier, and its drug-macromolecule complex, cannot cross.
Based on this assumption, it appears that drugs which bind
more readily to proteins are less able to pass into the CNS
(‘free drug theory’ [34, 62]). This may be true for most
drugs, but there are some exceptions to the rule. Several
drugs which cross the BBB without difficulty, such as ben-
zodiazepines, steroids and a few hormones, demonstrate
higher concentrations in the CNS than their unbound plasma
fraction would indicate [6366]. Similar observations were
made by Videbæk etal. (1999) (Table2) [67]. De Lange and
Danhof [68] collected several papers which describe highly
bound drugs (oxicams [69], imipramine and desimirpamine
[70], isradipine, darodipine [71]) which also penetrate the
BBB in surprisingly high extent (Table2). There are several
explanations of this phenomenon. Pardridge etal. claimed
that the conformation of the protein changes while interact-
ing with capillary walls and a drug molecule is freed from
a complex [64, 65, 72], Tanaka and Mizojiri ended up with
similar conclusion [66]. Another idea was protein-mediated
transport in which binding with protein (especially AAG)
enhances the BBB penetration [62]. Several authors claimed
that more permeable structure of capillary endothelium in
some regions may be the reason of the increased extraction
of a complex into the CNS [67, 68]. There is no doubt that
protein binding has a significant role for penetrating BBB; it
can either decrease the passage or affect it in the other way
with mechanisms still to be discovered. Mentioned studies
reveal that invivo analyses seem to be more applicable in
that case. The unique environment in the CNS or interac-
tions between proteins and brain capillaries apparently have
a high impact on the matter, therefore there is a substantial
difference between invitro and invivo results.
The placental barrier
The placenta is a unique connection between mother and
fetus. It is formed during the sixth week of pregnancy and
exists until the time of birth. Its main function is to deliver
nutrients and oxygen to the foetus and to remove waste and
metabolites. Throughout the pregnancy, the placenta also
adopts other roles: from the tenth week, it also produces
(3)
log BB
=
drug concentration in CNS
drug concentration in plasma
(4)
log PS
=observed permeability across BBB
cm
s
surface area of brain capillary endothelium
cm2
g
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
3224 Molecular Biology Reports (2020) 47:3221–3231
1 3
hormones such as chorionic gonadotropin (CG), human pla-
cental lactogen (HPL), relaxin, progesterone, testosterone,
oestrogens etc. and it manifests metabolic activity [73, 74].
Inside the placenta the blood vessels from a mother and a
child are tangled together but the blood itself does not mix;
despite this, sufficient exchange of substances is maintained
between the organisms [73]. Due to its high permeability,
the placenta acts more as a filter than an actual barrier [75].
Bacterial cells are retained within the placenta as are macro-
molecules, such as insulin or heparin, and immunoglobulins,
except IgG. Most drugs pass through the placental barrier,
including barbiturates, antibiotics, sulphonamides and alco-
hol [75]. Small molecules cross the barrier by simple diffu-
sion, while drugs also cross by facilitated diffusion or active
transport [75] or by endocytosis [73, 7678]. It is assumed
that the penetration of drugs through the placenta is limited
mainly by protein binding rather than lipophilicity [79]. A
significant role in the active transport of drugs is played by
the ATP-binding cassette transporters, with the main ones
being glycoprotein P and breast cancer resistance protein.
They can either transport drug molecules to the fetal side
or return them into maternal circulation; of these, the latter
function is assumed to be more important, and plays a sig-
nificant role in forming the placental barrier [77].
During pregnancy, it is difficult to avoid pharmacother-
apy, and drug usage has increased in recent years. Drugs
are administered in the treatment of chronic diseases such
as epilepsy, diabetes, hypertension or they are prescribed
temporally to treat infections such as the common cold.
Additionally pregnant women often take over-the-counter
drugs and dietary supplements, without medical advice [80,
81]. The amount of a drug which crosses the placental bar-
rier is dependent on various factors: its physicochemical
properties, pharmacokinetics, the concentration gradient on
both sides of the barrier, the differences in pH in between
maternal and foetal plasma and the levels of protein binding
in both organisms [80, 81]. Protein binding is considered the
important property in determining drug transport through
the placenta, influencing both the speed and the extent of
this process [16, 74].
The distribution of a drug between mother and child is
limited also by the concentration of main plasma proteins.
These change continually over the course of pregnancy:
while the concentration of foetal albumin (alpha-fetopro-
tein, AFP) is lower than the maternal HSA level during the
initial stages of pregnancy, it can be up to 20% higher than
maternal HSA at childbirth [16]. The amount of foetal alpha-
1-acid glycoprotein also increases with the development of
the foetus; however, it never exceeds adult levels, remain-
ing about 30–40% lower [16]. There are also differences in
affinities to protein binding sites, with AFP attracting fewer
molecules than HSA in adult plasma [82]. Protein binding
can also occur in both maternal and foetal tissues; drug mol-
ecules can also form a repository in the placenta, from which
it can be released in uncontrolled way into the maternal or
foetal plasma [16].
Table 2 Influence of protein binding on drug penetration into the CNS
* Calculated from unbound fraction data available in the reference paper
** Not available
*** Bovine serum albumin, used as a replacement for HSA
Drug Pharmacological activity Plasma protein binding* CNS penetration Reference
Isoxicam Non-steroidal anti-inflammatory drugs 96,5% in human serum Increased (in the presence of HSA and
AAG)
[69]
Meloxicam 99,7% in human serum Increased (in the presence of AAG)
Imipramine Antidepressants − 52% to HSA − 67% to AAG Higher than predicted from the unbound
fraction (for both proteins)
[70]
Desimipramine − 61% to AAG Higher than predicted from the unbound
fraction (for both proteins)
Isradipine Calcium channel antagonists − 91% to HSA − 92% to AAG Higher than predicted from the unbound
fraction (for both proteins)
[71]
Darodipine − 86% to HSA − 96% to AAG Higher than predicted from the unbound
fraction (for both proteins)
Propranolol Beta blocker NA** Low, compatible with the prediction (in
the presence of BSA***) Higher than
predicted from the unbound fraction (in
the presence of aag)
[62]
Flumazenil GABA receptor antagonists − 39% to HSA Higher than predicted from the unbound
fraction (HSA)
[67]
Iomazenil − 58% to HSA Higher than predicted from the unbound
fraction (HSA)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
3225Molecular Biology Reports (2020) 47:3221–3231
1 3
In vitro experiments with propofol using a human pla-
centa model by He etal. [79, 83] found propofol clearance to
correlate with the concentration of fetal albumin. It appears
that the potential to cross into the placenta is significantly
dependent on binding with alpha-fetoprotein: an increase
of alpha-fetoprotein concentration results in greater drug
penetration. It was also found that infiltration across the
placental barrier diminishes as the concentration of mater-
nal HSA rises. Elsewhere, [84] it was found that HSA has
a great influence on citalopram and fluoxetine placental
transport, with its presence in the perfusion solute increased
the degree of penetration; this effect was correlated with
the affinity of the drugs to HSA: the passage of fluoxetine
(PPB% = 94%) was significantly lower than that of citalo-
pram (PPB% = 50%).
The placenta is considered a very weak barrier against
xenobiotics and most of the administered drugs can easily
cross it. Plasma protein binding appears to affect this process
because it significantly limits placental transit, but alpha-
fetoprotein concentration increase which can enhance the
passage is also an important matter. The accumulation of
drugs in the placenta is still underestimated and it needs to
be studied in detail to get a clearer picture of the processes
that can affect fetal safety during pharmacotherapy.
Skin barrier
The skin is the largest human organ, and one which separates
the internal environment from the surroundings and protects
it from various pathogens. As a barrier, the skin also pre-
vents the penetration of many chemical compounds. This
poses a challenge for the design of dermatological prepara-
tions, which are quite common in modern pharmacotherapy,
mainly due to their easy and convenient application and lack
of side effects typical for the oral administration. Dermato-
logical application can also enhance the systemic activity
of a drug [85]. It has previously been assumed that most of
the administered drug particles are absorbed into the skin
circulation, thus allowing them to pass into the bloodstream,
and that the process was regulated by the skin structure and
condition, the structure of the drug and the type of pharma-
ceutical formulation [85, 86]. However, later studies suggest
that the most important factors determining skin penetration
are the structure and properties of the drug [85]. The per-
meability of the skin varies across its surface in response to
changes in its structure, for example, variation in the num-
bers of follicles or the thickness of the stratum corneum [87].
Externally administered drugs can bind with the proteins
within the skin layers, which can be desirable if only local
action is intended: the drug will accumulate at its site of
activity and will not cause any adverse systemic effects.
However, in the case of transdermal drugs such skin pro-
tein binding will disturb their flow into the circulation, slow
the passage through the skin and reduce the overall amount
of active molecules in the system. Previously, it was found
that highly protein binding drugs achieved lower concentra-
tion in plasma and the time of skin penetration was longer
[88]. A 2008 study [89] examining the different pharmaco-
dynamics of tacrolimus and pimecrolimus with regard to
their ability to penetrate the skin found that pimecrolimus is
more likely to bind non-specifically with various skin pro-
tein than tacrolimus, thus yielding a lower systemic con-
centration (Table3). Similarly, benzocaine has also been
found to accumulate in the skin through non-specific binding
(Table3) [90].
These results suggest that protein binding in the skin
should be carefully studied in case of dermatological for-
mulations, especially for highly protein binding drugs. Albu-
min is present in the skin [91] so the correlation between
binding to HSA in plasma and skin could be a useful tool
in pharmaceutical design. Nowadays, only the process of
skin sensitisation is widely examined, which is supposed to
be the result of non-covalent, reversible binding of various
compound with skin proteins, including albumin [92, 93].
Drug penetration intobreast milk
During lactation, similarly to pregnancy, it can be difficult
to avoid the use of any medicines. Many women abandon
breastfeeding when they take drugs, but often unnecessarily.
The penetration of most xenobiotics into milk is quite low
and only a fraction is typically ingested by an infant [94,
95]. The amount of a drug in breast milk is estimated using
the M/P ratio for a particular drug (Eq.5): this represents
the ratio between concentration of the drug in milk and in
maternal plasma:
Table 3 Influence of protein binding on skin penetration
Drug Pharmacological activity Protein binding in skin Skin permeability Reference
Pimecrolimus Calcineurin inhibitors High, non-specific binding to human skin proteins Lower penetration than in
the case of tacrolimus
[89]
Tacrolimus Low, non-specific binding to human skin proteins
Benzocaine Local anaesthetic Accumulation of significant amount of benzocaine in skin Low penetration [90]
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
3226 Molecular Biology Reports (2020) 47:3221–3231
1 3
This parameter should be calculated for each drug indi-
vidually and can be obtained from clinical studies, observa-
tions of single, medical cases or derived mathematically,
using chemometric methods [96101]. Drugs with M/P
value lower than 1 are considered as safe for breast-feeding
child.
Breast milk is regarded as the best nourishment for a new-
born infant, and for first six months of life, it can be its only
food. Milk is produced in the mammary glands by special-
ised cells called lactocytes [102]. It is composed of a mixture
of water and carbohydrates, proteins, lipids, vitamins and
various other nutrients [103], with the composition changing
over the course of lactation [104, 105]. During the first stage
of lactation, breast milk, colostrum, is composed mainly of
structural proteins and proteins which support the immune
system e.g. lactoferrin or immunoglobulins [106, 107]. This
is later replaced by transitional milk, which has higher levels
of carbohydrates and lipids and it is more nutritious than
colostrum. Mature milk is produced around the third week
after birth, and it consists of around 7% carbohydrates, 4%
lipids and less than 1% proteins [102, 106, 108]. This is
later replaced by transitional milk, which has higher levels
of carbohydrates and lipids and it is more nutritious than
colostrum. Mature milk is produced around the third week
after birth, and it consists of around 7% carbohydrates, 4%
lipids and less than 1% proteins [109].
Drugs mostly penetrate into breast milk by simple dif-
fusion along a concentration gradient. This process is also
limited by various factors connected with the compound
structure: molecular weight, lipophilicity, protein binding
or pKa [94, 110]. The pKa of a drug plays an important role
on its accumulation in milk: the mean pH of breast milk
ranges from 7.1–7.2 while that of plasma is around 7.4 [102,
105]. Weak bases become ionized in breast milk, trapping
them inside the mammary gland and preventing their return
to maternal plasma [111]. In addition, drugs with high lipo-
philicity can also accumulate in the lipid phase of breast
milk, and while protein binding can prevent the passage of
molecules into milk, drugs also bind with the breast milk
proteins themselves [112]. The composition of the protein
phase consists of alpha-S1, alpha-S2, beta- and kappa-
caseins, alpha-lactoalbumin, beta-lactoglobulin, plasma
albumin and lactoferrin, as well as immunoglobulins A, M,
G and lysozyme and alpha-1-acid glycoprotein [104, 113].
However, drug binding is typically weaker in breast milk
than in plasma [111].
Drug transfer into breast milk is still a difficult subject
for invivo study. Although clinical studies have been per-
formed, they are usually based on very small groups of sub-
jects or describe individual cases. Short-term use of drugs,
(5)
M/P = drug concentration in milk
drug concentration in plasma
during infection for example, seems to be less problematic
than in the case of long-term pharmacotherapy. Women
suffering from chronic conditions such as multiple sclero-
sis, epilepsy or psychiatric disorders, or those undergoing
anticancer therapy, often want to maintain breast-feeding.
A review by Constantinescu etal. [114] examined the usage
of various immunosuppressive drugs, including azathio-
prine, belatacept, corticosteroids, cyclosporine A, everoli-
mus, sirolimus and tacrolimus, during lactation. A study of
methylprednisolone levels in the breast milk of two lactating
women, one of them after renal transplantation and the other
with multiple sclerosis [115, 116] found that methylpredni-
solone passes poorly into milk, which could be related with
its high PPB%, estimated to be around 79% (Table4) [116].
A 2013 study of antiepileptic drugs by Davanzo etal.
[117] reviewed a body of pharmacokinetic and clinical data,
including relevant infant dose (RID), and toxicity guidelines
taken from LactMed [118] and Hale [119]. Older-generation
drugs such as carbamazepine, phenobarbital, phenytoin and
valproic acid were found to be relatively safe, even pheno-
barbital, which weakly binds with plasma proteins in mater-
nal plasma (20–45% [120]). The overall conclusion was that
neither pharmacokinetic or literature toxicity parameters are
good predictors of the drug penetration into breast milk. The
penetration of cisplatin across the placenta [121] and into
breast milk (Table4) [121, 122] was also studied. The drug
was found to demonstrate poor penetration into milk as its
platinum ion binds strongly with plasma proteins [120, 122].
However, cisplatin is contraindicated during lactation, prob-
ably due to the fact that that it accumulates during repeated
dosage.
Postpartum depression or anxiety also requires a long-
term treatment. SSRIs (selective serotonin reuptake inhibi-
tors) are believed to be the safest drugs for lactating women
because their high PPB% values, among other factors, pre-
vent them from crossing readily into milk (Table4) [123].
One exception is paroxetine, as it has been linked with an
increased risk of heart dysfunction [124]. A detailed reviews
about CNS drugs usage during lactation by Eberhard-Gran
etal. and by Weissman etal. [125, 126]. They provide data
regarding drug secretion into breast milk and recommenda-
tions for use. The latter study also points out the negative
correlation between PB and M/P values [125]. Further infor-
mation about the use of antidepressants is also given in a
review by Lanza di Scalea and Wisner [127].
In most of the cases described, where the excretion of the
drug into milk is very low, one of the main reasons men-
tioned is high plasma protein binding. This may indicate that
milk penetration may be the most PPB dependent of all the
barriers described in this review. Another issue to consider
is milk protein binding, which further reduces the amount of
medicine an infant ingests. Experts claim that breastfeeding
should not be interrupted during pharmacotherapy unless it
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
3227Molecular Biology Reports (2020) 47:3221–3231
1 3
is necessary, which seems to be a reasonable solution. How-
ever, there should be sufficient evidence that the medicine is
safe for breastfed infants or wouldn’t interrupt the lactation.
Summary
Drug-protein binding has a significant influence on the phar-
macokinetic properties of most compounds. It can limit the
bioavailability of active compounds by controlling their
passage through biological membranes; however, binding
to plasma proteins allows hydrophobic drugs to be trans-
ported in the aqueous environment of the human organism.
The drug-protein complex is less likely to cross the placental
barrier or to enter breast milk, which decreases the nega-
tive effect of medicines on breastfeeding infants; however,
some drugs can accumulate in placental tissues or in milk
by binding with proteins in these regions, and upon their
later release, enter the foetus or infant in uncontrolled way.
Passage through the blood–brain barrier is more complicated
by mechanisms which protect the central nervous system,
such as active efflux and the use of strong protein binding
mechanisms. Additional unknown mechanisms that lead to
the penetration of several protein-bounded drugs make this
matter even more complex. Skin penetration is an impor-
tant issue for transdermal drugs because they have a strong
impact on their bioavailability and protein-binding interacts
with this process.
Protein binding is relatively simple to study invitro, but
its effect on crossing biological barriers in a living organ-
ism could be difficult to grasp with these methods. This is
probably due to the complexity of the entire barrier-crossing
process and additional side-effects that simply cannot be
obtained in the laboratory.
Acknowledgements This work was supported by an internal grant of
the Medical University of Lodz no. 502–34-106.
Compliance with ethical standards
Conflict of interest The author declares no conflict of interest, finan-
cial or otherwise.
Open Access This article is licensed under a Creative Commons Attri-
bution 4.0 International License, which permits use, sharing, adapta-
tion, distribution and reproduction in any medium or format, as long
as you give appropriate credit to the original author(s) and the source,
provide a link to the Creative Commons licence, and indicate if changes
were made. The images or other third party material in this article are
included in the article’s Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in
the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a
copy of this licence, visit http://creat iveco mmons .org/licen ses/by/4.0/.
References
1. Sun H, Zhao H (2016) Physiologic drug distribution and pro-
tein binding. in: applied biopharmaceutics & pharmacokinet-
ics. McGraw-Hill Education, 259
2. Kratz F, Elsadek B (2012) Clinical impact of serum proteins
on drug delivery. J Control Release 161:429–445. https ://doi.
org/10.1016/j.jconr el.2011.11.028
3. Schmidt S, Gonzalez D, Derendorf H (2010) Significance of
protein binding in pharmacokinetics and pharmacodynamics.
J Pharm Sci 99:1107–1122. https ://doi.org/10.1002/jps.21916
4. Spinella R, Sawhney R, Jalan R (2016) Albumin in chronic
liver disease: structure, functions and therapeutic implica-
tions. Hepatol Int 10:124–132. https ://doi.org/10.1007/s1207
2-015-9665-6
5. Caraceni P, Domenicali M, Tovoli A etal (2013) Clinical indica-
tions for the albumin use: still a controversial issue. Eur J Intern
Med 24:721–728. https ://doi.org/10.1016/j.ejim.2013.05.015
Table 4 Penetration into breast milk of highly protein bound drugs
* According to the reference paper
** According to DrugBank database [120]
Drug Pharmacological activity Plasma
protein
binding
Penetration into breast milk Reference
Methylprednisolone Corticosteroid 75–82%* Poor penetration, average infant exposure was 0,188mg/kg/day with
maternal dose of 1000mg/day
[116]
Cisplatin Chemotherapeutic agent 90%* Cisplatin was undetectable in breast milk, maternal dose was 70mg
weekly (repeated in 4 cycles)
[122]
Citalopram Selective serotonin reup-
take inhibitors (SSRI)
80%** Poor penetration, citalopram average milk level was 157 mcg/l with
average maternal dose of 29mg/day
[125]
Fluoxetine SSRI 94%** Poor penetration, fluoxetine average milk level was 76 mcg/l with
average maternal dose of 28mg/day
[125]
Sertraline SSRI 98–99%** Poor penetration, sertraline average milk level was 45 mcg/l with aver-
age maternal dose of 83mg/day
[125]
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
3228 Molecular Biology Reports (2020) 47:3221–3231
1 3
6. Israili ZH, Dayton PG, Frederick J, Di C (2001) Human alpha-
1-glycoprotein and its interactions with drugs. Drug Metab Rev
33:161–235. https ://doi.org/10.1081/DMR-10010 4402
7. Ghuman J, Zunszain PA, Petitpas I etal (2005) Structural basis
of the drug-binding specificity of human serum albumin. J Mol
Biol 353:38–52. https ://doi.org/10.1016/j.jmb.2005.07.075
8. Yamasaki K, Chuang VTG, Maruyama T, Otagiri M (2013)
Albumin-drug interaction and its clinical implication. Bio-
chim Biophys Acta - Gen Subj 1830:5435–5443. https ://doi.
org/10.1016/j.bbage n.2013.05.005
9. Otagiri M (2005) A molecular functional study on the interac-
tions of drugs with plasma proteins. Drug Metab Pharmacoki-
net 20:309–323. https ://doi.org/10.2133/dmpk.20.309
10. Chechłacz M, Korytowska N (2017) Związki wiążące się z
białkami osocza u ludzi: Znaczenie w terapii oraz metody
oznaczania wolnej frakcji. Biul Wydz Farm WUM 6:50–59
11. Svennebring A (2016) The connection between plasma protein
binding and acute toxicity as determined by the LD50 value.
Drug Dev Res 77:3–11. https ://doi.org/10.1002/ddr.21286
12. Zhang F, Xue J, Shao J, Jia L (2012) Compilation of 222 drugs’
plasma protein binding data and guidance for study designs.
Drug Discov Today 17:475–485. https ://doi.org/10.1016/j.
drudi s.2011.12.018
13. Heuberger J, Schmidt S, Derendorf H (2013) When is protein
binding important? J Pharm Sci 102:3458–3467. https ://doi.
org/10.1002/jps.23559
14. Hong L, Rosenbaum S (2014) Developmental pharmacoki-
netics in pediatric populations. J Pediatr Pharmacol Ther
19:262–276
15. Korzekwa K, Nagar S (2017) Drug distribution part 2. predict-
ing volume of distribution from plasma protein binding and
membrane partitioning. Pharm Res 34:544–551. https ://doi.
org/10.1007/s1109 5-016-2086-y
16. Polin RA, Abman SH, Rowitch D, Benitz WE (2017) Develop-
mental pharmacology and pharmacokinetics. In: fetal and neo-
natal physiology. Elsevier B.V., pp 187–249
17. Greenberg RG, Melloni C, Wu H etal (2016) Therapeutic index
estimation of antiepileptic drugs: a systematic literature review
approach. Clin Neuropharmacol 39:232. https ://doi.org/10.1097/
WNF.00000 00000 00017 2
18. Tamargo J, Le Heuzey JY, Mabo P (2015) Narrow therapeu-
tic index drugs: a clinical pharmacological consideration to
flecainide. Eur J Clin Pharmacol 71:549–567. https ://doi.
org/10.1007/s0022 8-015-1832-0
19. Mullokandov E, Ahn J, Szalkiewicz A (2014) Protein binding
drug-drug interaction between warfarin and tizoxanide in human
plasma. Austin J Pharmacol Ther
20. El-Mallakh RS, Brar KS, Yeruva RR (2019) Cardiac glycosides
in human physiology and disease: update for entomologists.
Insects 10:102
21. Cohen L (2004) Plasma protein-binding methods in drug dis-
covery. in: optimization in drug discovery: Invitro methods.
Humana Press, 111–222
22. Howard M, Hill J, Galluppi G, McLean M (2010) Plasma protein
binding in drug discovery and development. Comb Chem High
Throughput Screen 13:170–187. https ://doi.org/10.2174/13862
07107 90596 745
23. Deb PK, Al-Attraqchi O, Prasad MR, Tekade RK (2018) Pro-
tein and tissue binding: implication on pharmacokinetic param-
eters. In: advances in pharmaceutical product development and
research. Academic Press, 371–399
24. Faed EM (1981) Protein binding of drugs in plasma, interstitial
fluid and tissues: effect on pharmacokinetics. Eur J Clin Phar-
macol 21:77–81
25. Sun H, Zhao H (2012) Physiologic drug distribution and pro-
tein binding. In: applied biopharmaceutics & pharmacokinetics.
McGraw-Hill Education, 259–308
26. Peck T, Hill S, Williams M (2008) Drug passage across the cell
membrane. In: pharmacology for anaesthesia and intensive care,
Third Edition. Cambridge University Press, 1–10
27. Benet LZ, Hosey CM, Ursu O, Oprea TI (2016) BDDCS, the
Rule of 5 and drugability. Adv Drug Deliv Rev 101:89–98. https
://doi.org/10.1016/j.addr.2016.05.007
28. Kok-Yong S, Lawrence L (2015) Drug distribution and drug
elimination. In: basic pharmacokinetic concepts and some clini-
cal applications
29. Waring MJ (2010) Lipophilicity in drug discovery. Expert Opin
Drug Discov 5:235–248. https ://doi.org/10.1517/17460 44100
36050 98
30. Tymoczko JL, Berg JM, Stryer L (2009) Biochemia.
Wydawnictwo Naukowe PWN, Warszawa
31. Bamburowicz-Klimkowska M, Bogucka U, Szutowski M (2011)
Funkcje transporterów typu ABC. Biul Wydz Farm WUM
3:34–40
32. Zhivkova ZD (2017) Quantitative structure—pharmacokinetics
relationships for plasma protein binding of basic drugs. J Pharm
Pharm Sci 20: 349–359. https ://doi.org/10.18433 /J3363 3
33. Di L, Kerns EH (2016) Invivo environments affect drug expo-
sure. In: Drug-Like Properties. pp 15–28
34. Bohnert T, Gan LS (2013) Plasma protein binding: from dis-
covery to development. J Pharm Sci 102:2953–2994. https ://doi.
org/10.1002/jps.23614
35. Yang NJ, Hinner MJ (2015) Getting across the cell mem-
brane: an overview for small molecules, peptides, and
proteins. Methods Mol Biol 1266:29–53. https ://doi.
org/10.1007/978-1-4939-2272-7_3
36. Zhang R, Qin X, Kong F et al (2019) Improving cellular
uptake of therapeutic entities through interaction with compo-
nents of cell membrane. Drug Deliv 26:328–342. https ://doi.
org/10.1080/10717 544.2019.15827 30
37. Pellegatti M, Pagliarusco S, Solazzo L, Colato D (2011) Plasma
protein binding and blood-free concentrations: which studies are
needed to develop a drug? Expert Opin Drug Metab Toxicol
7:1009–1020. https ://doi.org/10.1517/17425 255.2011.58633 6
38. Vhora I, Patil S, Bhatt P, Misra A (2015) Protein- and Peptide-
drug conjugates: an emerging drug delivery technology. In:
advances in protein chemistry and structural biology. 98:1–44
39. Maeda H, Wu J, Sawa T etal (2000) Tumor vascular permeability
and the EPR effect in macromolecular therapeutics: a review.
J Control Release 65:271–284. https ://doi.org/10.1016/S0168
-3659(99)00248 -5
40. Larsen MT, Kuhlmann M, Hvam ML, Howard KA (2016) Albu-
min-based drug delivery: harnessing nature to cure disease. Mol
Cell Ther 4:3. https ://doi.org/10.1186/s4059 1-016-0048-8
41. Feng J, Zhao C, Wang L etal (2018) Development of a novel
albumin-based and maleimidopropionic acid-conjugated pep-
tide with prolonged half-life and increased invivo anti-tumor
efficacy. Theranostics 8:2094–2106. https ://doi.org/10.7150/
thno.22069
42. Persidsky Y, Ramirez SH, Haorah J, Kanmogne GD (2006)
Blood-brain barrier: structural components and function under
physiologic and pathologic conditions. J Neuroimmune Pharma-
col 1:223–236. https ://doi.org/10.1007/s1148 1-006-9025-3
43. Brzezińska K, Ziaja M (2012) Struktura i funkcje bariery krew-
mózg: the structure and role of blood-brain barrier. Postępy Biol
Komórki 39:84–99
44. Meyer J, Mischeck U, Veyhl M etal (1990) Blood-brain bar-
rier characteristic enzymatic properties in cultured brain cap-
illary endothelial cells. Brain Res 514:305–309. https ://doi.
org/10.1016/0006-8993(90)91425 -G
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
3229Molecular Biology Reports (2020) 47:3221–3231
1 3
45. Taylor EM (2005) Efflux transporters and the blood-brain barrier.
Nova Science Publishers, New York
46. Löscher W, Potschka H (2005) Blood-brain barrier active efflux
transporters: ATP-binding cassette gene family. NeuroRx 2:86–
98. https ://doi.org/10.1602/neuro rx.2.1.86
47. Pardridge WM (2002) CNS Drug design based on principles of
blood-brain barrier transport. J Neurochem. https ://doi.org/10.1
046/j.1471-4159.1998.70051 781.x
48. Pardridge WM (2005) The blood-brain barrier: Bottleneck in
brain drug development. NeuroRx. https ://doi.org/10.1602/neuro
rx.2.1.3
49. Pardridge WM (2011) Drug transport in brain via the cerebro-
spinal fluid. Fluids Barriers CNS 8:7
50. Pardridge WM (2012) Drug transport across the blood-brain bar-
rier. J Cereb Blood Flow Metab 32:1959–1972
51. Pardridge WM (2015) Targeted delivery of protein and gene
medicines through the blood-brain barrier. Clin Pharmacol Ther
97:347–361
52. Pardridge WM (2019) Blood-brain barrier and delivery of protein
and gene therapeutics to brain. Front Aging Neurosci 11:373.
https ://doi.org/10.3389/fnagi .2019.00373
53. Dong X (2018) Current strategies for brain drug delivery. Thera-
nostics 8:1481–1493. https ://doi.org/10.7150/thno.21254
54. Bors LA, Erdö F (2019) Overcoming the blood-brain barrier:
challenges and tricks for CNS drug delivery. Sci Pharm 87:6.
https ://doi.org/10.3390/sciph arm87 01000 6
55. Banks WA (2009) Characteristics of compounds that cross
the blood-brain barrier. BMC Neurol 9:S3. https ://doi.
org/10.1186/1471-2377-9-S1-S3
56. Ballabh P, Braun A, Nedergaard M (2004) The blood–brain
barrier: an overview. Neurobiol Dis 16:1–13. https ://doi.
org/10.1016/j.nbd.2003.12.016
57. Pardridge WM (2004) Log(BB), PS products and in silico models
of drug brain penetration. Drug Discov Today 9:392–393
58. Misra A, Ganesh S, Shahiwala A, Shah SP (2003) Drug deliv-
ery to the central nervous system: a review. J Pharm Pharm Sci
6:252–273
59. Kakee A, Terasaki T, Sugiyama Y (1996) Brain efflux index as
a novel method of analyzing efflux transport at the blood-brain
barrier. J Pharmacol Exp Ther 277:1550–1559
60. Kalvass JC, Maurer TS (2002) Influence of nonspecific brain
and plasma binding on CNS exposure: implications for rational
drug discovery. Biopharm Drug Dispos 23:327–338. https ://doi.
org/10.1002/bdd.325
61. Kalvass JC, Maurer TS, Pollack GM (2007) Use of plasma and
brain unbound fractions to assess the extent of brain distribu-
tion of 34 drugs: comparison of unbound concentration ratios to
invivo P-glycoprotein efflux ratios. Drug Metab Dispos 35:660–
666. https ://doi.org/10.1124/dmd.106.01229 4
62. Pardridge WM, Sakiyama R, Fierer G (1983) Transport of pro-
pranolol and lidocaine through the rat blood-brain barrier: pri-
mary role of globulin-bound drug. J Clin Invest 71:900–908.
https ://doi.org/10.1172/JCI11 0844
63. Jones DR, Hall SD, Jackson EK etal (1988) Brain uptake of ben-
zodiazepines: effects of lipophilicity and plasma protein binding.
J Pharmacol Exp Ther 245:816–822
64. Kobiler D, Lustig S, Shapira S (2001) The role of plasma protein
binding in drug delivery to brain. In: blood-brain barrier: drug
delivery and brain pathology. Kluwer Academic/Plenum Publish-
ers, 311–321
65. Pardridge WM, Fierer G (1990) Transport of tryptophan
into brain from the circulating, albumin-bound pool in
rats and in rabbits. J Neurochem 54:971–976. https ://doi.
org/10.1111/j.1471-4159.1990.tb023 45.x
66. Tanaka H, Mizojiri K (1999) Drug-protein binding and blood-
brain barrier permeability. J Pharmacol Exp Ther 288:912–918
67. Videbæk C, Ott P, Paulson OB, Knudsen GM (1999) Blood-brain
barrier transport and protein binding of flumazenil and ioma-
zenil in the rat: implications for neuroreceptor studies. J Cereb
Blood Flow Metab 19:948–955. https ://doi.org/10.1097/00004
647-19990 9000-00002
68. de Lange ECM, Danhof M (2002) Considerations in the use of
cerebrospinal fluid pharmacokinetics to predict brain target con-
centrations in the clinical setting: implications of the barriers
between blood and brain. Clin Pharmacokinet 41:691–703. https
://doi.org/10.2165/00003 088-20024 1100-00001
69. Jolliet P, Simon N, Brée F etal (1997) Blood-to-brain transfer of
various oxicams: effects of plasma binding on their brain deliv-
ery. Pharm Res 14:650–656. https ://doi.org/10.1023/A:10121
65414 610
70. Lin TH, Sawada Y, Sugiyama Y etal (1987) Effects of albu-
min and α1-acid glycoprotein on the transport of imipramine
and desipramine through the blood-brain barrier in rats. Chem
Pharm Bull 35:294–301. https ://doi.org/10.1248/cpb.35.294
71. Urien S, Pinquier JL, Paquette B etal (1987) Effect of the bind-
ing of isradipine and darodipine to different plasma proteins on
their transfer through the rat blood-brain barrier: drug binding
to lipoproteins does not limit the transfer of drug. J Pharmacol
Exp Ther 242:349–353
72. Pardridge WM (1988) Recent advances in blood-brain barrier
transport. Annu Rev Pharmacol Toxicol 28:25–39. https ://doi.
org/10.1146/annur ev.pharm tox.28.1.25
73. Van Der Aa EM, Peereboom-Stegeman JH, Noordhoek J etal
(1998) Mechanisms of drug transfer across the human placenta.
Pharm World Sci 20:139–148. https ://doi.org/10.1023/A:10086
56928 861
74. Griffiths SK, Campbell JP (2015) Placental structure, function
and drug transfer. Contin Educ Anaesthesia, Crit Care Pain
15:84–89. https ://doi.org/10.1093/bjace accp/mku01 3
75. Traczyk WZ (2013) Fizjologia człowieka w zarysie. PZWL,
Warszawa
76. Jamroziak K, Kowalczyk M, Robak T (2002) Białko oporności
raka piersi ABCG2 (BCRP/MXR/ABCP) nowy transporter
z nadrodziny ABC związany z opornością wielolekową. Acta
Haematol Pol 33:403–416
77. Iqbal M, Audette MC, Petropoulos S etal (2012) Placental drug
transporters and their role in fetal protection. Placenta 33:137–
142. https ://doi.org/10.1016/j.place nta.2012.01.008
78. Rubinchik-Stern M, Eyal S (2012) Drug interactions at the
human placenta: What is the evidence? Front Pharmacol 3:126.
https ://doi.org/10.3389/fphar .2012.00126
79. He YL, Tsujimoto S, Tanimoto M etal (2000) Effects of protein
binding on the placental transfer of propofol in the human dually
perfused cotyledon invitro. Br J Anaesth 85:281–286. https ://
doi.org/10.1093/bja/85.2.281
80. Evseenko D, Paxton JW, Keelan JA (2006) Active transport
across the human placenta: impact on drug efficacy and tox-
icity. Expert Opin Drug Metab Toxicol 2:51–69. https ://doi.
org/10.1517/17425 255.2.1.51
81. Gedeon C, Koren G (2006) Designing pregnancy centered medi-
cations: drugs which do not cross the human placenta. Placenta
27:861–868. https ://doi.org/10.1016/j.place nta.2005.09.001
82. Unadkat J, Dahlin A, Vijay S (2005) Placental drug transport-
ers. Curr Drug Metab 5:125–131. https ://doi.org/10.2174/13892
00043 48917 1
83. He YL, Seno H, Sasaki K, Tashiro C (2002) The influences of
maternal albumin concentrations on the placental transfer of
propofol in human dually perfused cotyledon invitro. Anesth
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
3230 Molecular Biology Reports (2020) 47:3221–3231
1 3
Analg 94:1312–1314. https ://doi.org/10.1097/00000 539-20020
5000-00048
84. Heikkinen T, Ekblad U, Laine K (2002) Transplacental trans-
fer of citalopram, fluoxetine and their primary demethylated
metabolites in isolated perfused human placenta. BJOG An Int J
Obstet Gynaecol 109:1003–1008. https ://doi.org/10.1016/S1470
-0328(02)01467 -2
85. Cross SE, Roberts MS (1999) Targeting local tissues by trans-
dermal application: understanding drug physicochemical prop-
erties that best exploit protein binding and blood flow effects.
Drug Dev Res 46:309–315. https ://doi.org/10.1002/(SICI)1098-
2299(19990 3/04)46:3/4%3c309 :AID-DDR17 %3e3.0.CO;2-H
86. Benfeldt E, Serup J, Menné T (1999) Effect of barrier pertur-
bation on cutaneous salicylic acid penetration in human skin:
invivo pharmacokinetics using microdialysis and non-invasive
quantification of barrier function. Br J Dermatol 140:739–748.
https ://doi.org/10.1046/j.1365-2133.1999.02859 .x
87. Nielsen JB, Benfeldt E, Holmgaard R (2016) Penetration through
the skin barrier. Curr Probl Dermatology 49:103–111. https ://doi.
org/10.1159/00044 1549
88. Hwang S, Bayne W (1984) Influence of protein binding on the
accumulation and depletion of drug from the skin. J Pharm Sci
73:710–712
89. Weiss HM, Fresneau M, Moenius T etal (2008) Binding of pime-
crolimus and tacrolimus to skin and plasma proteins: implica-
tions for systemic exposure after topical application. Drug Metab
Dispos 36:1812–1818. https ://doi.org/10.1124/dmd.108.02191 5
90. Dalvi UG, Zatz JL (1982) Effect of skin binding on percutaneous
transport of benzocaine from aqueous suspensions and solutions.
J Pharm Sci 71:824–826. https ://doi.org/10.1002/jps.26007 10728
91. Divkovic M, Pease CK, Gerberick GF, Basketter DA (2005)
Hapten-protein binding: From theory to practical application
in the invitro prediction of skin sensitization. Contact Dermat
53:189–200. https ://doi.org/10.1111/j.0105-1873.2005.00683 .x
92. Aleksic M, Thain E, Gutsell SJ etal (2007) The role of non-cova-
lent protein binding in skin sensitisation potency of chemicals.
Cutan Ocul Toxicol 26:161–169. https ://doi.org/10.1080/15569
52070 12122 82
93. Settivari RS, Rowlands JC, Wilson DM, etal (2017) Application
of evolving computational and biological platforms for chemical
safety assessment. In: A Comprehensive Guide to Toxicology in
Nonclinical Drug Development. 843–867
94. Hotham N, Hotham E (2015) Drugs in breastfeeding. Aust Prescr
38:156–159. https ://doi.org/10.18773 /austp rescr .2015.056
95. Lauterbach R (2011) Stosowanie leków a karmienie piersią. Pedi-
atr po Dyplomie 15:77–85
96. Atkinson HC, Begg EJ (1988) Relationship between human
milk lipid-ultrafiltrate and octanol-water partition coefficients. J
Pharm Sci 77:796–798. https ://doi.org/10.1002/jps.26007 70916
97. Atkinson H, Begg E (1988) Prediction of drug concentrations
in human skim milk from plasma protein binding and acid-base
characteristics. Br J Clin Pharmacol 25:495–503. https ://doi.
org/10.1111/j.1365-2125.1988.tb033 34.x
98. Atkinson HC, Begg EJ, Darlow BA (1988) Drugs in human
milk: clinical pharmacokinetic considerations. Clin Pharma-
cokinet 14:217–240. https ://doi.org/10.2165/00003 088-19881
4040-00003
99. Atkinson UC, Begg EJ (1990) Prediction of drug distribution into
human milk from physicochemical characteristics. Clin Phar-
macokinet 18:151–167. https ://doi.org/10.2165/00003 088-19901
8020-00005
100. Agatonovic-Kustrin S, Tucker IG, Zecevic M, Zivanovic LJ
(2000) Prediction of drug transfer into human milk from theo-
retically derived descriptors. Anal Chim Acta 418:181–195. https
://doi.org/10.1016/S0003 -2670(00)00963 -6
101. Abraham MH, Gil-Lostes J, Fatemi M (2009) Prediction of milk/
plasma concentration ratios of drugs and environmental pollut-
ants. Eur J Med Chem 44:2452–2458
102. Florea M (2014) Laktacja i karmienie piersią: Przegląd
piśmiennictwa. Perinatol Neonatol i Ginekol 7:165–170
103. Newton E (2004) Breastmilk: the Gold Standard. Clin Obstet
Gynecol 47:632–642
104. Lis J, Orczyk-Pawiłowicz M, Ka̧tnik-Prastowska I (2013)
Białka mleka ludzkiego zaangażowane w procesy immuno-
logiczne. Postepy Hig Med Dosw 67:529–547. https ://doi.
org/10.5604/17322 693.10516 48
105. Fleishaker JC, Desai N, McNamara PJ (1987) Factors affecting
the milk to plasma drug concentration ratio in lactating women:
physical interactions with protein and fat. J Pharm Sci 76:189–
193. https ://doi.org/10.1002/jps.26007 60302
106. Kowalska D, Gruczyńska E, Bryś J (2015) Mleko matki-pierwsza
żywność w życiu człowieka. Probl Hig Epidemiol 96:387–398
107. Rak K, Bronkowska M (2014) Immunologiczne znaczenie siary
Hygeia Public Heal 49:249–254
108. Jenness R (1979) The composition of human milk. Semin Peri-
natol 3:225–239. https ://doi.org/10.1007/BF027 48197
109. Riodan, Jan; Wambach K (2009) Breastfeeding and human Lacta-
tion. Jones & Bartlett Learning; 4th edition
110. Spencer JP, Gonzalez LS, Barnhart DJ (2001) Medications in the
breast-feeding mother. Am Fam Physician 64:119
111. Breitzka RL, Sandritter TL, Hatzopoulos FK (1997) Principles of
drug transfer into breast milk and drug disposition in the nursing
infant. J Hum Lact 13:155–158. https ://doi.org/10.1177/08903
34497 01300 219
112. Gentile S, Rossi A, Bellantuono C (2007) SSRIs during breast-
feeding: Spotlight on milk-to-plasma ratio. Arch Womens Ment
Health 10:39–51
113. Ballard O, Morrow AL (2013) Human milk composition. nutri-
ents and bioactive factors. Pediatr Clin North Am 60:46–74. https
://doi.org/10.1016/j.pcl.2012.10.002
114. Constantinescu S, Pai A, Coscia LA etal (2014) Breast-feed-
ing after transplantation. Best Pract Res Clin Obstet Gynaecol
28:1163–1173. https ://doi.org/10.1016/j.bpobg yn.2014.09.001
115. Coulam C, Moyer T, Jiang N, Zincke H (1982) Breast-feeding
after renal transplantation Transpl Proc 14:605–609
116. Cooper SD, Felkins K, Baker TE, Hale TW (2015) Transfer of
methylprednisolone into breast milk in a mother with multiple
sclerosis. J Hum Lact 31:237–239. https ://doi.org/10.1177/08903
34415 57097 0
117. Davanzo R, Dal Bo S, Bua J et al (2013) Antiepileptic
drugs and breastfeeding. Ital J Pediatr 39:50. https ://doi.
org/10.1186/1824-7288-39-50
118. Drugs and lactation database (LactMed). https ://toxne t.nlm.nih.
gov/pda/lactm ed.htm. Accessed 11 Nov 2019
119. Hale TW (2012) Medications and mother’s milk, 15th ed. Phar-
masoft Medical Publishing
120. Drugbank. https ://www.drugb ank.ca/drugs /DB011 74. Accessed
20 Nov 2019
121. Hale TW (2012) Medications and mother’s milk, 15th ed.
Pharmasoft Medical Publishing. https ://doi.org/10.1515/
JPM.2011.015
122. Hays KE, Ryu RJ, Swisher EM etal (2013) Duration of cisplatin
excretion in breast milk. J Hum Lact 29:469–472. https ://doi.
org/10.1177/08903 34413 47967 1
123. Williams AS (2007) Antidepressants in pregnancy and breast-
feeding. Aust, Prescr
124. Norris MM (2013) Use of antidepressants during pregnancy and
lactation. Ment Heal Clin. https ://doi.org/10.9740/mhc.n1635 20
125. Weissman AM, Levy BT, Hartz AJ etal (2004) Pooled analysis
of antidepressant levels in lactating mothers, breast milk, and
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
3231Molecular Biology Reports (2020) 47:3221–3231
1 3
nursing infants. Am J Psychiatry 161:1066–1078. https ://doi.
org/10.1176/appi.ajp.161.6.1066
126. Eberhard-Gran M, Eskild A, Opjordsmoen S (2006) Use of psy-
chotropic medications in treating mood disorders during lacta-
tion: Practical recommendations. CNS Drugs 20:187–198
127. Lanza Di Scalea T, Wisner KL (2009) Antidepressant medication
use during breastfeeding. Clin Obstet Gynecol 52:483. https ://
doi.org/10.1097/GRF.0b013 e3181 b52bd 6
Publisher’s Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... 21 characteristics that increase transfer to breast milk include high lipophilicity, low molecular weight, and low protein binding. 50 Furthermore, drugs that are weak bases become ionized in breast milk, trapping them in the mammary glands and preventing the drug from returning to plasma. 50 Plasma protein binding is a key factor when determining the extent of drug transfer into breast milk; drugs with high plasma protein binding tend to have poor distribution in breast milk, which reduces infant drug exposure and results in a lower RID. ...
... 50 Furthermore, drugs that are weak bases become ionized in breast milk, trapping them in the mammary glands and preventing the drug from returning to plasma. 50 Plasma protein binding is a key factor when determining the extent of drug transfer into breast milk; drugs with high plasma protein binding tend to have poor distribution in breast milk, which reduces infant drug exposure and results in a lower RID. 50 In this study, following once-daily administration of zuranolone 30 mg for 5 days, zuranolone was highly protein bound in plasma, with an overall unbound fraction of ≤0.49% in all participants. ...
... 50 Plasma protein binding is a key factor when determining the extent of drug transfer into breast milk; drugs with high plasma protein binding tend to have poor distribution in breast milk, which reduces infant drug exposure and results in a lower RID. 50 In this study, following once-daily administration of zuranolone 30 mg for 5 days, zuranolone was highly protein bound in plasma, with an overall unbound fraction of ≤0.49% in all participants. Additionally, the temporal profile of zuranolone concentration was comparable between plasma and breast milk. ...
Article
Purpose/Background Zuranolone is a positive allosteric modulator of both synaptic and extrasynaptic γ-aminobutyric acid type A receptors and a neuroactive steroid approved as an oral, once-daily, 14-day treatment course for adults with postpartum depression in the United States. This study assessed zuranolone transfer into breast milk. Methods/Procedures Healthy, nonpregnant, lactating adult female participants received once-daily 30 mg zuranolone from day (D)1 through D5 in this phase 1 open-label study. The relative infant dose (RID; weight-adjusted proportion of the maternal dose in breast milk over 24 hours) for 30 mg zuranolone was assessed at D5. An RID for 50 mg zuranolone was estimated using a simulation approach across a range of infant ages and weights. Findings/Results Of 15 enrolled participants (mean age, 30.1 years), 14 completed the study. The mean RID for 30 mg zuranolone at D5 was 0.357%; the mean steady-state milk volume over D3 to D5 decreased from baseline by 8.3%. Overall unbound zuranolone in plasma was low (≤0.49%). Plasma concentrations peaked at D5 before decreasing in a biexponential manner. There was strong concordance between the temporal evolution of zuranolone concentrations in plasma and breast milk. The estimated mean RID for 50 mg zuranolone based on a milk intake of 200 mL/kg per day was 0.984%. All treatment-emergent adverse events reported by participants were mild, the most common being dizziness (n = 3). Implications/Conclusions Zuranolone transfer into the breast milk of healthy, nonpregnant, lactating adult female participants was low; the estimated RID for 50 mg zuranolone was <1%, well below the <10% threshold generally considered compatible with breastfeeding.
... It is important to note that concerns about the baby's safety arise when the RID is greater than 10% [41]. The transfer of drugs into breastmilk is influenced by protein binding, lipid solubility, and ionization [42]. Heparin and Insulin were believed to be too large to cross biological membranes, but recent evidence shows otherwise [43,44]. ...
Preprint
Full-text available
Background The use of cosmetic Botox (Botulinum Neurotoxin Type A, BoNT/A ) has become increasingly prevalent. Particularly after pregnancy, postpartum depression represents one major factor motivating women to use Botox even during the lactation and breastfeeding period. Currently, there is limited understanding of the impact of Botox on lactation and the potential of its active component passing into breastmilk and affecting the infant. Methods Breastmilk samples were acquired from five women aged between 28 - 45 through a clinic in Suzhou, Jiangsu, P.R. China. Three sample sets ranged from 1 hour to 1 year after facial Botox treatments (64 U), whereas the remaining two sample sets were from women who never received Botox treatment. BoNT/A concentrations in samples were detected using standard Enzyme-Linked Immunosorbent Assay (ELISA), unreduced and reduced Western Blotting, confocal micro-Raman Spectroscopy, and Mass Spectrometry(LC-MS). Findings From ELISA, breastmilk BoNT/A concentrations peaked at 33.4 pg/mL 4 days after Botox injection. BoNT/A concentrations were highest overall in the first week and around two months after injection. While non-reducing polyacrylamide gel electrophoresis (PAGE) showed a protein band of 150 kDa peaking at 48 hours, reduced SDS-PAGE detected a 100 kDa protein first peaking at 72 hours, then re-emerging after 7 days, respectively, and in line with previous observations by others. Interestingly, micro-Raman spectroscopy indicated additional Raman peaks at 6 hours and 48 hours that were not present in other breastmilk samples which were evaluated in this study. However, no clear indication of BoNT/A was detected in Mass Spectrometry (LC-MS). Interpretation The amount of BoNT/A in breastmilk peaks around 48 hours, and at 2 months after facial injection. Even over a year after injection, BoNT/A can be detected. However, all quantities of BoNT/A detected in this study are highly likely to be safe for infants. Additionally, our study suggests that alternative methods, besides ELISA, may be utilized for the rapid detection of low concentrations of BoNT/A in body fluid samples. Funding Duke Kunshan University Start-Up funds, Duke Kunshan University Undergraduate Studies Signature Work Research Grant, Synear and Wang-Cai Biochemistry grants, and Kunshan Municipal Government research funding. Graphical Abstract Highlights BoNT/A was detected in the breastmilk of 3 women after facial Botox injections of 64 U. Toxin levels peaked in the first week and at around 2 months after injection. All detected levels (up to 33.40 pg/mL) were significantly below the lethal dose for newborns.
... The extent of distribution is influenced by physicochemical properties of the drug and biological factors. Egress from the intravascular space involves passive diffusion, carrier-mediated, or active transport processes [20]. Volume of distribution describes the relationship between the amount of drug administered and the measured concentration. ...
Article
The widespread use of drugs for unapproved purposes remains common in children, primarily attributable to practical, ethical, and financial constraints associated with pediatric drug research. Pharmacometrics, the scientific discipline that involves the application of mathematical models to understand and quantify drug effects, holds promise in advancing pediatric pharmacotherapy by expediting drug development, extending applications, and personalizing dosing. In this review, we delineate the principles of pharmacometrics, and explore its clinical applications and prospects. The fundamental aspect of any pharmacometric analysis lies in the selection of appropriate methods for quantifying pharmacokinetics and pharmacodynamics. Population pharmacokinetic modeling is a data-driven method (‘top-down’ approach) to approximate population-level pharmacokinetic parameters, while identifying factors contributing to inter-individual variability. Model-informed precision dosing is increasingly used to leverage population pharmacokinetic models and patient data, to formulate individualized dosing recommendations. Physiologically based pharmacokinetic models integrate physicochemical drug properties with biological parameters (‘bottom-up approach’), and is particularly valuable in situations with limited clinical data, such as early drug development, assessing drug–drug interactions, or adapting dosing for patients with specific comorbidities. The effective implementation of these complex models hinges on strong collaboration between clinicians and pharmacometricians, given the pivotal role of data availability. Promising advancements aimed at improving data availability encompass innovative techniques such as opportunistic sampling, minimally invasive sampling approaches, microdialysis, and in vitro investigations. Additionally, ongoing research efforts to enhance measurement instruments for evaluating pharmacodynamics responses, including biomarkers and clinical scoring systems, are expected to significantly bolster our capacity to understand drug effects in children.
... For the compounds ANO61 and AF299, we observed low effectivity-likely due to high plasma bindingupon agonist-induced PRP aggregation. Plasma protein binding are known to affect the bioavailability and distribution of bioactive compounds, which effect partly determines their pharmacokinetics 57 . For instance, clinically used tyrosine kinase inhibitors are known to bind to plasma factors, which reduces the drug efficiency and limits diffusion through the cell membrane 58,59 . ...
Article
Full-text available
Distinct platelet activation patterns are elicited by the tyrosine kinase-linked collagen receptor glycoprotein VI (GPVI) and the G-protein coupled protease-activated receptors (PAR1/4) for thrombin. This is reflected in the different platelet Ca²⁺ responses induced by the GPVI agonist collagen-related peptide (CRP) and the PAR1/4 agonist thrombin. Using a 96 well-plate assay with human Calcium-6-loaded platelets and a panel of 22 pharmacological inhibitors, we assessed the cytosolic Ca²⁺ signaling domains of these receptors and developed an automated Ca²⁺ curve algorithm. The algorithm was used to evaluate an ultra-high throughput (UHT) based screening of 16,635 chemically diverse small molecules with orally active physicochemical properties for effects on platelets stimulated with CRP or thrombin. Stringent agonist-specific selection criteria resulted in the identification of 151 drug-like molecules, of which three hit compounds were further characterized. The dibenzyl formamide derivative ANO61 selectively modulated thrombin-induced Ca²⁺ responses, whereas the aromatic sulfonyl imidazole AF299 and the phenothiazine ethopropazine affected CRP-induced responses. Platelet functional assays confirmed selectivity of these hits. Ethopropazine retained its inhibitory potential in the presence of plasma, and suppressed collagen-dependent thrombus buildup at arterial shear rate. In conclusion, targeting of platelet Ca²⁺ signaling dynamics in a screening campaign has the potential of identifying novel platelet-inhibiting molecules.
... In adult female rats, pregnant and non-pregnant, the free fraction of drug was much higher (40-50%) than when measured in vitro (see Tables 9 and 10). The functional significance of the free fraction of a drug is that it is the free fraction of drug that is exchangeable with cells, tissues and organs (Wanat 2020;Summerfield et al. 2022). These results suggest that in vivo the exchangeable fraction of a drug may be much higher than is apparent from in vitro measurements. ...
Article
Full-text available
Cannabidiol is a major component of cannabis but without known psychoactive properties. A wide range of properties have been attributed to it, such as anti-inflammatory, analgesic, anti-cancer, anti-seizure and anxiolytic. However, being a fairly new compound in its purified form, little is known about cannabidiol brain entry, especially during development. Sprague Dawley rats at four developmental ages: embryonic day E19, postnatal day P4 and P12 and non-pregnant adult females were administered intraperitoneal cannabidiol at 10 mg/kg with [³H] labelled cannabidiol. To investigate the extent of placental transfer, the drug was injected intravenously into E19 pregnant dams. Levels of [³H]-cannabidiol in blood plasma, cerebrospinal fluid and brain were estimated by liquid scintillation counting. Plasma protein binding of cannabidiol was identified by polyacrylamide gel electrophoresis and its bound and unbound fractions measured by ultrafiltration. Using available RNA-sequencing datasets of E19 rat brain, choroid plexus and placenta, as well as P5 and adult brain and choroid plexus, expression of 13 main cannabidiol receptors was analysed. Results showed that cannabidiol rapidly entered both the developing and adult brains. Entry into CSF was more limited. Its transfer across the placenta was substantially restricted as only about 50% of maternal blood plasma cannabidiol concentration was detected in fetal plasma. Albumin was the main, but not exclusive, cannabidiol binding protein at all ages. Several transcripts for cannabidiol receptors were expressed in age- and tissue-specific manner indicating that cannabidiol may have different functional effects in the fetal compared to adult brain.
Article
Tuberculosis (TB), mainly caused by Mycobacterium tuberculosis, remains the leading cause of death from a bacterium in the world. The global prevalence of clinically relevant infections with opportunistically pathogenic non‐tuberculous mycobacteria (NTM) have also been on the rise. Pharmacological treatment of both TB and NTM infections usually requires prolonged regimens of drug combinations, and is often challenging because of developed or inherent resistance to common antibiotic drugs. Medicinal chemistry efforts are thus needed to improve treatment options and therapeutic outcomes. Nα‐aroyl‐N‐aryl‐phenylalanine amides (AAPs) have been identified as potent antimycobacterial agents that target the RNA polymerase with a low probability of cross resistance to rifamycins, the clinically most important class of antibiotics known to inhibit the bacterial RNA polymerase. In this review, we describe recent developments in the field of AAPs, including synthesis, structural characterization, in vitro microbiological profiling, structure‐activity relationships, physicochemical properties, pharmacokinetics and early cytotoxicity assessment.
Article
Full-text available
Wiele substancji endogennych oraz egzogennych wykazuje zdolność do wiązania się z białkami osocza, głównie z ludzką albuminą surowicy oraz α-1-kwaśną glikoproteiną. Substancje te w krwioobiegu występują zarówno w formie wolnej jak i związanej z białkami. Białko posiada na swojej powierzchni miejsca wiązania charakterystyczne dla danego związku, różniące się wielkością, kształtem oraz powinowactwem. Wiązanie z białkami następuje w wyniku oddziaływań hydrofobowych, van der Waalsa oraz elektrostatycznych. Stopień związania z białkami osocza zależy od m.in. obecności stanu zapalnego, występowania chorób nerek i wątroby oraz wieku. Jedynie forma wolna związku jest aktywna biologicznie oraz jest zdolna do pokonywania barier biologicznych. W związku z tym duże znaczenie mają metody pomiaru stężenia frakcji niezwiązanej z białkami w osoczu. Najczęściej stosowana jest dializa równowagowa, uznawana za metodę referencyjną. Inne metody to m.in. ultrafiltracja, ultrawirowanie, mikrodializa, mikroekstrakcja, wysokosprawna analiza czołowa oraz ekstrakcja w punkcie zmętnienia.
Article
Full-text available
Różne rodzaje transporterów obecnych w organizmie wpływają na losy leków w ustroju poprzez udział w procesach ich absorpcji, dystrybucji i eliminacji. Białka te między innymi biorą udział w dwukierunkowym transporcie substancji egzo- i endogennych przez ściany jelita cienkiego, przewodów żółciowych i bariery krew-mózg. W polifarmakoterapii transportery typu ABC obecne w ścianie jelita mogą determinować biodostępność, szybkość i kierunek transportu oraz być przyczyną występowania interakcji pomiędzy przyjmowanymi lekami. Glikoproteina P (Pgp) produkt ekspresji genu MDR1 należy do najbardziej znaczących transporterów typu ABC pod tym względem. Jej dystrybucja tkankowa i narządowa posiada bardzo istotny wpływ na wchłanianie ksenobiotyków, a interakcje leków z tym białkiem mogą prowadzić do zmian biodostępności leków stosowanych jednocześnie. Omówiono poszczególne podrodziny transporterów typu ABC ze szczególnym uwzględnieniem ich funkcji.
Article
Full-text available
Alzheimer’s disease (AD) and treatment of the brain in aging require the development of new biologic drugs, such as recombinant proteins or gene therapies. Biologics are large molecule therapeutics that do not cross the blood-brain barrier (BBB). BBB drug delivery is the limiting factor in the future development of new therapeutics for the brain. The delivery of recombinant protein or gene medicines to the brain is a binary process: either the brain drug developer re-engineers the biologic with BBB drug delivery technology, or goes forward with brain drug development in the absence of a BBB delivery platform. The presence of BBB delivery technology allows for engineering the therapeutic to enable entry into the brain across the BBB from blood. Brain drug development may still take place in the absence of BBB delivery technology, but with a reliance on approaches that have rarely led to FDA approval, e.g., CSF injection, stem cells, small molecules, and others. CSF injection of drug is the most widely practiced approach to brain delivery that bypasses the BBB. However, drug injection into the CSF results in limited drug penetration to the brain parenchyma, owing to the rapid export of CSF from the brain to blood. A CSF injection of a drug is equivalent to a slow intravenous (IV) infusion of the pharmaceutical. Given the profound effect the existence of the BBB has on brain drug development, future drug or gene development for the brain will be accelerated by future advances in BBB delivery technology in parallel with new drug discovery.
Article
Full-text available
Cardiac glycosides, cardenolides and bufadienolides, are elaborated by several plant or animal species to prevent grazing or predation. Entomologists have characterized several insect species that have evolved the ability to sequester these glycosides in their tissues to reduce their palatability and, thus, reduce predation. Cardiac glycosides are known to interact with the sodium- and potassium-activated adenosine triphosphatase, or sodium pump, through a specific receptor-binding site. Over the last couple of decades, and since entomologic studies, it has become clear that mammals synthesize endogenous cardenolides that closely resemble or are identical to compounds of plant origin and those sequestered by insects. The most important of these are ouabain-like compounds. These compounds are essential for the regulation of normal ionic physiology in mammals. Importantly, at physiologic picomolar or nanomolar concentrations, endogenous ouabain, a cardenolide, stimulates the sodium pump, activates second messengers, and may even function as a growth factor. This is in contrast to the pharmacologic or toxic micromolar or milimolar concentrations achieved after consumption of exogenous cardenolides (by consuming medications, plants, or insects), which inhibit the pump and result in either a desired medical outcome, or the toxic consequence of sodium pump inhibition.
Article
Full-text available
Efficient cellular delivery of biologically active molecules is one of the key factors that affect the discovery and development of novel drugs. The plasma membrane is the first barrier that prevents direct translocation of chemic entities, and thus obstructs their efficient intracellular delivery. Generally, hydrophilic small molecule drugs are poor permeability that reduce bioavailability and thus limit the clinic application. The cellular uptake of macromolecules and drug carriers is very inefficient without external assistance. Therefore, it is desirable to develop potent delivery systems for achieving effective intracellular delivery of chemic entities. Apart from of the types of delivery strategies, the composition of the cell membrane is critical for delivery efficiency due to the fact that cellular uptake is affected by the interaction between the chemical entity and the plasma membrane. In this review, we aimed to develop a profound understanding of the interactions between delivery systems and components of the plasma membrane. For the purpose, we attempt to present a broad overview of what delivery systems can be used to enhance the intracellular delivery of poorly permeable chemic entities, and how various delivery strategies are applied according to the components of plasma membrane.
Article
Full-text available
Treatment of certain central nervous system disorders, including different types of cerebral malignancies, is limited by traditional oral or systemic administrations of therapeutic drugs due to possible serious side effects and/or lack of the brain penetration and, therefore, the efficacy of the drugs is diminished. During the last decade, several new technologies were developed to overcome barrier properties of cerebral capillaries. This review gives a short overview of the structural elements and anatomical features of the blood–brain barrier. The various in vitro (static and dynamic), in vivo (microdialysis), and in situ (brain perfusion) blood–brain barrier models are also presented. The drug formulations and administration options to deliver molecules effectively to the central nervous system (CNS) are presented. Nanocarriers, nanoparticles (lipid, polymeric, magnetic, gold, and carbon based nanoparticles, dendrimers, etc.), viral and peptid vectors and shuttles, sonoporation and microbubbles are briefly shown. The modulation of receptors and efflux transporters in the cell membrane can also be an effective approach to enhance brain exposure to therapeutic compounds. Intranasal administration is a noninvasive delivery route to bypass the blood–brain barrier, while direct brain administration is an invasive mode to target the brain region with therapeutic drug concentrations locally. Nowadays, both technological and mechanistic tools are available to assist in overcoming the blood–brain barrier. With these techniques more effective and even safer drugs can be developed for the treatment of devastating brain disorders.
Article
Full-text available
Angiogenesis plays a critical role in tumor aggressiveness, and a lot of anti-angiogenic agents have been used in clinical therapy. The therapeutic efficacy of peptides are generally restricted by the short in vivo life-time, thus, we were interested in developing a novel albumin-based and maleimidopropionic acid-conjugated peptide to prolong the half-life and improve the anti-tumor effect. Methods: We developed a peptide F56 with a maleimidopropionic acid (MPA) at the C-terminal (denoted as F56-CM), which allows immediate and irreversible conjugation with serum albumin. Biological property and anti-tumor activity of F56-CM were evaluated in vitro and in vivo. Results: We showed that F56-CM reduced migration and tube formation of endothelial cells in vitro and inhibited the generation of subintestinal vessels (SIV) in zebrafish embryos in vivo. F56-CM inhibited vascular endothelial growth factor (VEGF) induced phosphorylation of VEGFR1 and activation of the PI3K-AKT axis. Furthermore, F56-CM rapidly conjugated with albumin upon intravenous injection and extended the biological half-life of F56 from 0.4249 h to 6.967 h in rats. Compared with F56, F56-CM exhibited stronger anti-tumor activity on both BGC-823 gastric cancer and HT-29 colon cancer xenografts in nude mice, and the statistical difference was remarkable. More significantly, the efficacy of F56-CM inhibiting lung metastasis of BGC-823 cells was also better than that of F56. The inhibition rates were 62.1% and 78.9% for F56 and F56-CM respectively when administrated every day, and 43.8% and 63.1% when administrated every four days at equal dose. Conclusions: Taken together, our results demonstrated that F56-CM has considerable potential for cancer therapy.
Article
Full-text available
The blood-brain barrier (BBB) has been a great hurdle for brain drug delivery. The BBB in healthy brain is a diffusion barrier essential for protecting normal brain function by impeding most compounds from transiting from the blood to the brain; only small molecules can cross the BBB. Under certain pathological conditions of diseases such as stroke, diabetes, seizures, multiple sclerosis, Parkinson's disease and Alzheimer disease, the BBB is disrupted. The objective of this review is to provide a broad overview on current strategies for brain drug delivery and related subjects from the past five years. It is hoped that this review could inspire readers to discover possible approaches to deliver drugs into the brain. After an initial overview of the BBB structure and function in both healthy and pathological conditions, this review re-visits, according to recent publications, some questions that are controversial, such as whether nanoparticles by themselves could cross the BBB and whether drugs are specifically transferred to the brain by actively targeted nanoparticles. Current non-nanoparticle strategies are also reviewed, such as delivery of drugs through the permeable BBB under pathological conditions and using non-invasive techniques to enhance brain drug uptake. Finally, one particular area that is often neglected in brain drug delivery is the influence of aging on the BBB, which is captured in this review based on the limited studies in the literature.
Chapter
The third edition of this market leading book has been thoroughly updated and expanded, with additional contributions from experts in the field, to include all new drugs available to the anaesthetist and intensive care specialist. Basic pharmacological principles, vital to understanding how individual drugs actually have their effects, are dealt with methodically and with many highly annotated diagrams and tables. With hospital infections becoming increasingly prevalent, the important section on antibiotics has been further expanded. With the third edition, this well established title continues to provide its readers with the most concise yet comprehensive coverage of all aspects of pharmacology. An ideal aid to study and practice for junior and trainee anaesthetists, critical care nurses and all physicians and healthcare professionals working in theatre, accident and emergency departments or intensive care units.
Book
The vasculature of the central nervous system (eNS) is characterized by the existence of the blood-brain barrier (BBB), which can be regarded as both an anatomical and physiological phenomenon. The BBB is formed by a complex cellular system of endothelial cells, astroglia, pericytes, perivascular macrophages and a basal membrane, although the anatomic substrate of the BBB is the interendothelial tight junctions that form a continuous sealing. The BBB serves as an exquisitely controlled, functional gate to the eNS. It not only protects the brain from agents in the blood that could impair neurological function, but also controls the influx and efflux of numerous substances to maintain proper homeostasis and provide the brain with necessary nutrients. The structural and functional integrity of the BBB was shown to be dramatically altered during various diseases of the eNS, including neoplasia, ischemia, trauma, hypertension, inflammation and epilepsy. Recent years research has partially elucidated the mechanisms underlying the development of some of these brain disorders as well as the pathways used by different pathogens, like bacteria and viruses, to initiate eNS infections. The development of in vitro models of the BBB had instrumental role in the understanding of the involvement of the BBB in the pathogenesis of several eNS diseases. The intimate, functional association between the function of the brain and the activity of the BBB makes the later a target for pharmacological modulation that will expand the therapeutic possibilities for a range of neurological diseases.