FormalPara Key Points

The penetration of drugs into the central nervous system is very limited.

Transport proteins expressed at barriers between the central nervous system and the systemic circulation are gate keepers for drugs.

Knowledge on protein expression levels and localization of transporters is lagging behind knowledge on messenger RNA expression of transporters.

Novel imaging methodologies are rapidly progressing and hold the promise to visualize individual transporter function in vivo.

1 Introduction

All organs in the mammalian body are connected via the blood circulatory system, which provides both the supply of vital nutrients and disposes waste products. In most instances, drugs also reach their target via the circulatory system regardless of the route of application. Organs are separated from the circulatory system by barriers, which may be leaky such as in the liver or very tight such as in the brain. The brain is separated from the blood by the blood–brain barrier (BBB) and from the cerebrospinal fluid (CSF) by the choroid plexus (CP) [1, 2]. In addition, the brain also communicates with body extracellular fluids via the arachnoid epithelium [3], but its pathway only has a minor contribution to the exchange and is not the subject of this overview. To cross barriers, which ultimately are always plasma membranes, transport systems are needed [4, 5]. Moreover, a tissue-specific expression of transporters allows the body to accumulate substances, such as drugs in an organ-specific manner [1, 4]. The tightness of the BBB severely limits the access of drugs to the brain and presents a major challenge in the development of drugs with targets in the central nervous system (CNS) [68]. Therefore, the aim of this review is to summarize the current knowledge on the expression of drug transporters in the BBB and in the CP. As the retina is also part of the CNS, we also address transporters in the blood–retina barrier (BRB). Furthermore, we highlight the role of the transporters encountered by drugs and other substances once they have crossed the barriers surrounding the tissues of the CNS.

2 Drug Transporters

Solutes, like drugs need transporters to enter or exit cells. Generally, transporters mediating the cellular uptake of drugs belong to the superfamily of solute carriers (SLC). The efflux of drugs (or their metabolites) frequently occurs against a concentration gradient and is often mediated by members of the adenosine triphosphate (ATP)-binding cassette (ABC) transporters. Numerous reviews have been published on both superfamilies of transporters. The SLC superfamily represents currently 52 families and 395 genes for individual transporters and has been covered recently in a special issue [9]. Human ABC transporter genes number to 48 members and are divided into seven families [10], but not all of them act as transporters [11]. It is beyond the scope and space of this review to describe the individual drug transporter families. This overview focuses on members of the SLCO and SLC22A gene families, which are well known to mediate, in addition to endogenous substrates, the transport of drugs. Among the ABC protein families, multidrug resistance protein 1 (MDR1) (ABCB1), ABCG2 (also called breast cancer resistance protein or BCRP, ABCG2), and members of the ABCC family are known to be important drug and drug metabolite transporters and are therefore covered here.

Human drug transporters being members of the SLC superfamily and expressed in cerebral blood–tissue barriers are listed together with rodent transporters in Tables 1, 2, 3, 4 and 5. Rodent species are included as they are used as preclinical species in drug development and because they allow in vivo experiments not possible in humans for investigating the role of transporters in drug transport in the CNS. In these tables, a selection of references (we apologize for omissions) was made and data on transport systems obtained from microperfusion experiments as well as from work with microcapillary endothelial cell lines are not included. Microperfusion experiments are most valuable for the elucidation of the in vivo situation for drug access to brain tissue but face the difficulty that many drug transporters have an overlapping substrate specificity [1214]. Brain capillary endothelial cell lines and very likely also other established cell lines display altered transporter expression levels in comparison to their in vivo counterparts [15, 16]. It should be realized that there are often conflicting data in the literature. Good examples are the members of the ABCC family, about which conflicting data on the expression in the BBB currently exist [17]. This may relate to the fact that for animals, within a species, different strains show different transporter expression. For example, in mice, mouse multidrug resistance-associated protein 2 (MRP2) could be detected in the BBB of C57BL/6, Swiss, and SvJ mice, but not FVB mice, while the liver and kidney showed positive staining in all strains [18]. Similarly, the expression levels of the mouse monocarboxylate transporter MCT1 in the BBB of C57BL/6J mice were significantly lower than in ddY or FVB mice, while the expression of mBCRP was significantly higher in C57BL/6J mice compared with ddY or FVB mice when analyzed by quantitative targeted proteomics [19]. In human studies, tissue procurement and storage prior to analysis as well as sampling biases will considerably contribute to variable data sets. For protein expression, preference to data obtained from proteomic approaches where available was given over data obtained by western blotting. Transporter expression in blood–neural tissue barriers has additionally been covered in many overviews [17, 2031].

Table 1 Expression of SLC transporters at the blood–brain barrier
Table 2 Expression of ABC transporters at the blood–brain barrier
Table 3 Expression of SLC transporters in choroid plexus
Table 4 Expression of ABC transporters in choroid plexus
Table 5 Transporter expression in blood–retina barriers

3 Blood–Brain Barrier

To provide a stable environment for the CNS, the BBB needs to be able to tightly control the access of substances to the brain. To this end, the endothelial cells lining the walls of the brain capillaries form together with tight junctions an impervious barrier [2]. Brain access of substances (e.g., nutrients such as d-glucose) is consequently controlled by transport proteins specifically expressed in the luminal and/or abluminal membrane of brain capillary endothelial cells [2]. Nutrients are transported into the brain by influx systems such as amino acids by members of the SLC1A family [32]. Many of these transporters are equilibrative, i.e., they cannot work against concentration gradients. Extrusion of substances from the brain occurs at the luminal membrane and is mediated by ABC transporters such as MDR1 [33]. ABC transporters use energy provided from ATP hydrolysis and can therefore establish steep concentration gradients. While SLC transporters expressed in plasma membranes are often uptake transporters, some members act as exchangers of solutes and consequently may mediate efflux of a substrate in exchange for uptake of another substance [9]. Consequently, the direction of solute transport by such transporters has to be determined experimentally, ideally in situ in the organ of interest.

Drug transporters being members of the SLC superfamily and expressed in the BBB are listed in Table 1. Specifically, the protein expression of several SLC superfamily members involved in drug transport (four organic anion transporting polypeptides (OATPs) (SLCO), two organic cation transporters (OCTs) (SLC22A), one organic cation transporter novel type (OCTN) (SLC22A), one concentrative nucleoside transporter (CNT) (SLC28A), and two equilibrative nucleoside transporters (ENTs) (SLC29A) has been reported for the human BBB (Table 1). SLC family members are either facilitating uptake transporters or secondary active transporters capable of working against concentration gradients [9]. Consequently, drug transporters expressed in the luminal membrane of the BBB are potential entry sites for drugs or toxins into the BBB. In the case of non-polar expression (i.e., in the luminal and in the abluminal membrane of the BBB), these transporters may allow their substrates to cross the endothelial cells of the BBB and enter into the brain. The number of substrates including drugs for SLC family members known currently is overwhelming and listing them is beyond the scope of this overview. Lists of substrates can be found in the following (as well as many additional) reviews: for OATPs [13, 34, 35]; for OATs [3638]; for OCTs [37, 39]; for CNTs [40, 41]; for ENTs [41, 42], and for MATEs [43, 44].

Several examples demonstrate indirectly and directly the pharmacologic and toxicologic role of SLC transporters in the BBB of humans. Drugs used for the treatment of pain often need to enter the CNS [45]. Triptans are drugs used to treat migraine. It was recently demonstrated that several triptans are substrates of OATP1A2 expressed in the BBB (Table 1) [46]. Hence, it is reasonable that hydrophilic triptans may use OATP1A2 to cross the BBB. The relative transport rate of OATP1A2-mediated transport decreases from triptans with tertiary amines to triptans with primary amines in heterologous expression systems [46]. While the transport of drugs across the BBB is considered to be beneficial this is not the case for toxins. This is exemplified by an incidence in Brazil, where 126 patients of a hemodialysis unit experienced a microcystin intoxication and 60 patients subsequently died [47]. The patients developed acute neurotoxicity and subacute hepatotoxicity. Expressing OATP1A2 in Xenopus laevis oocytes demonstrated that this transporter mediates uptake of microcystin [48]. Moreover, OATP1A2 expression was required for microcystin to exert its toxic effects on oocytes. Recently, it was reported that OATP1A2 is expressed in neurons in the human brain [49]. This finding adds an additional piece to the mechanistic understanding of microcystin toxicity: microcystin inhibits protein phosphatases at nanomolar concentrations [50]. Hence, the expression of OATP1A2 in neurons may allow microcystein, once it has crossed the BBB, to enter into neurons followed by impairment of neuronal functions. Looking at an endogenous compound, thyroid hormones are instrumental for the development of the brain and in adult life for metabolic adaptation [51]. OATP1C1, which is expressed at the BBB (Table 1) [52] is a high-affinity thyroid hormone transporter [53] and consequently allows the entry of thyroid hormones into brain. These examples clearly demonstrate that expression of transport proteins in the BBB in addition to endogenous substances allows the entry of xenobiotics into the brain. Hence, understanding the molecular properties of transporters working in the BBB will contribute to a better understanding of the penetration of drugs across the BBB to reach pharmacodynamics targets in the brain. Therefore, the relevance of the BBB as a selective guard of the brain is not only recognized by physiologists and pharmacologists but has also initiated large efforts for developing tools to study the impact of the BBB early in drug development [8, 54].

ABC transporters are mostly cellular efflux transporters and either act as cellular defense systems for substances or export them from the cytoplasm [9]. They are often located in strategic organ boundaries including the BBB, where they are most important for controlling access to body sanctuaries [55]. The importance of ABC transporters is further emphasized by the observation that more than 20 (out of 48) human ABC transporters are important in various acquired and inherited human diseases [56]. Drug transporters being members of the ABC superfamily and expressed in the human BBB are listed in Table 2. The protein expression of several ABC transporter superfamily members involved in drug transport (one MDR) (ABCB), four MRPs (ABCC), and one ABCG (ABCG) has been reported for the human BBB (Table 2). With respect to drug transport, members of the ABCB, ABCC, and ABCG family are capable of transporting numerous drugs. A list of substrates including drugs for transporting ABC family members can be found in the following reviews [27, 5767].

The brain protective role of ABC transporters at the BBB is best illustrated with the clinical studies aimed at inhibiting MDR1 in the drug treatment of cancer. For example, in a phase I trial, co-administration of etoposide and cyclosporine lead to more severe nausea in some patients receiving both drugs [68]. In another phase I study where etoposide and the second-generation MDR1 inhibitor PSC 833 were combined to treat cancer patients, severe ataxia was observed as dose-limiting toxicity of PSC833 [69]. In this case, the MDR1 inhibitor allowed etoposide to cross the BBB inducing neurotoxicity. The same toxicity was later observed in a phase III trial [70]. Similarly, a high-dose, oral tamoxifen phase I trial in combination with verapamil revealed dose-limiting neurologic side effects [71]. Taken together, these few examples in humans demonstrate the importance of luminal ABC transporters in the BBB as gate keepers preventing or lowering the exposure of the brain to potentially neurotoxic agents. In principle, given the access of a substrate into the BBB, abluminal ABC transporters should enhance the exposure of the brain to their substrates. To the best of our knowledge we found no such examples in the literature.

4 Choroid Plexus

The CP is located in the lateral, third and fourth brain ventricles and produces the CSF. It is a highly vascularized organ containing in the stroma loose connective tissue and the fenestrated endothelium. A tight monolayer of CP epithelial cells connected by tight junctions near the apical surface forms the blood–CSF barrier (BCSFB) [7274]. In addition to its central role in the production of CSF, it also removes organic anions as well as drugs and drug metabolites from the CSF, making the CP an important detoxifying system for the CSF [72, 73].

The protein expression of several SLC superfamily members involved in drug transport has to date been reported for CP. In humans, two OATPs and two OATs are identified in the CP (Table 3) and one member of the ABCB and two members of the ABCC family have been demonstrated at the protein level (Table 4). Inferring from rodent tissues, the two MRPs likely are expressed in the basolateral membrane of human CP epithelial cells and consequently mediate export of substances form the CSF back into the blood after their uptake across the apical membrane. The individual role of these transporters in drug transport cannot be directly assessed in humans despite the fact that, for example, analgetics and anticancer drugs are administered to patients via an intrathecal route. Clearance of such drugs from the CSF is obvious, but in addition to the CP, the villi of the arachnoids may also be involved in elimination of substances from the CSF. Drug transporter expression in arachnoid villi is still a largely uncharted area. In addition, the hydrodynamics of the CSF, which may contribute to drug elimination from this body compartment is rather controversial [74, 75].

5 Blood–Retina Barrier

The retina is an organ rich in neurons. The retina is exposed on the anterior side to the vitreous humor and at the posterior side to the choroid. In the retina, there exists two BRBs, namely the inner BRB formed by the retinal capillary endothelial cells and the outer BRB formed by the retinal pigmented epithelial cells [21, 76]. These two barriers prevent uncontrolled entry of blood constituents into the eye. Consequently, either one or both of these barriers needs to be overcome by drugs, which are systemically administered for the treatment of retinal diseases.

The protein expression of several SLC superfamily members involved in drug transport (three OATPs, one OAT, and one member of the ABCB and ABCC families each) has to date been reported for the human BRB (Table 5). Direct information on the role of transporters in drug permeation through the BRB in humans is missing, but it should be noted that systemically administered antibiotics reach the vitreous humor, e.g., ciprofloxacin [77] or daptomycin [78]. Ciprofloxacin is known to interact with OATPs [79]. Additionally, prostaglandins are used as first-line treatment for glaucoma [80] and are substrates of OATPs [81, 82]. While these examples do not prove that transport systems are involved in the ocular disposition of drugs, they are nevertheless strongly indicative, as in particular daptomycin is rather membrane impermeable.

6 Animal Models for Investigating the Role of Drug Transporters in the Central Nervous System

Animal models are a potent means to investigate the role of transporters in the CNS. Such models yield most valuable information on the function of drug transporters at blood–tissue barriers in the CNS as well as on their physiologic role in the CNS. For example, more than 50 years ago it was demonstrated in a goat model that phenolsulfonphthalein (also called phenol red), which at physiologic pH is a dianionic compound, and the anionic angiographic contrast agent diodrast are actively transported out from the CSF into blood [83]. The understanding and consequent appreciation of the role of transporters in the BBB changed with the seminal work by Schinkel and coworkers who demonstrated that in mice with an inactivated Mdr1a gene, the tissue concentration of ivermectin in brain was increased 87-fold in comparison to controls and 22.4-fold for vinblastine [84]. In addition, the same team found no negative effect on the physiology of mice when both Mdr1 genes were inactivated indicating that in this species mMDR1 (Table 2) plays no vital role [85]. In contrast, mice with a disrupted Abcg2 gene gave conflicting results on the role of mABCG2 in the BBB (Table 2) [86]. However, if studies were performed in mice, which in addition to Abcg2 had also disrupted Mdr1 genes, it became clear that for some drugs Abcg2 contributes to preventing drugs from crossing the BBB. This example nicely illustrates the complexity of in vivo studies with drugs sharing multiple transporters. Furthermore, the role of mOATPs (Table 1) in penetrating the BBB became evident in mice with a knockout of Slco1a4 [87], as well as with the Slco1a/1b locus, as in such animals statins showed a considerably lower entry into the brain [88].

Genetically modified mice can also be used to study the efflux of drug metabolites produced in the brain. Oseltamivir is an ethylester prodrug for RO 64-0802. The latter is an inhibitor of neuraminidase and as such is used in the prophylaxis and treatment of influenza virus infections [89]. This drug is associated with adverse psychiatric effects [90]. Oseltamivir is activated by carboxylesterase 1 [89], which is among other organs also expressed in the brain [91]. Studies with Abcb1 knockout mice showed that mMDR1 isoforms limit the brain’s access to oseltamivir across the BBB [92]. Microinjection of RO 64-0802 into the brain of mice deficient either for Abcc4 or SLC22a8 demonstrated that both mMRP4 (Table 2) and mOAT3 (Table 1) are involved in the elimination of RO 64-0802 from the brain across the BBB [93].

The opposite localization with respect to the lumen of blood vessels in the BBB (Table 4) and in the CP (Table 5) of MDR1 and ABCG2 leads to differential effects on the brain entry of drugs across the BBB and into the CSF. Mice with an Abcb1 or Abcg2 knockout show an increased accumulation of topotecan in the brain parenchyma, while its penetration into the CSF is reduced [94]. In double-knockout animals, these effects were additive for both barriers. In mice with an inactivated Scl22a8 gene, accumulation of fluorescein into the isolated CP was greatly reduced compared with wild-type animals [95]. Hence, knock-out mice provide a most valuable tool to investigate the impact of transporters not only in the BBB but also in the CP [96].

OATP1C1 was identified as a high-affinity thyroxine transporter [53], which is expressed at the BBB (Table 1) and in the CP (Table 5). Mice having an inactivated Oatp1c1 gene showed a significantly reduced brain content of T4 and T3 with no change in the plasma concentration of these two thyroid hormones [97], clearly demonstrating the important role of this transporter for thyroid hormone supply to the brain. In the same knock-out mice strain, uptake of sulforhodamine 101 into astrocytes of the hippocampus is severely impaired [98].

7 Outlook

Ample evidence accumulated in recent years indicates the importance of transporters expressed in the BBB, CP, and BRB for mediating the passage of drugs as well as nutrients and metabolites. Great progress has also been made, in particular in model animals, in the identification and quantification of transport proteins in these barriers. However, the depth of knowledge varies considerably between the different barriers as the availability of the CP and even more so of the BRB is very limited, in particular from humans. Hence, alternate tools such as good antibodies are urgently needed for defining the transporter inventory in these barriers. Importantly, antibodies have a major advantage in that they are keys to define the subcellular expression of transporters in barriers. As movement of substances across barriers into and out from the CNS is often unidirectional and in some instances may occur against concentration gradients, exact knowledge on the subcellular expression of transporters together with an understanding of their transport mechanism is key to understanding the contribution of individual transporters to the passage or blockage of compounds across these barriers. Progress in this area may come from a systems biology approach generating antibodies against a larger number of human proteins, such as the Human Protein Atlas Project [99]. In addition, advances in the field of targeted proteomics should certainly help to increase the knowledge needed for developing pharmacokinetic models for the uptake of drugs into the brain and for the export of drug metabolites from the brain [16]. In addition, for developing novel kinetic models for brain uptake and export, the contribution of drug metabolism, e.g., in the BBB, has to be taken into account [23].

Imaging methods and in particular positron emission tomography (PET) have made rapid and large progress in recent years such that PET has become a feasible tool for studying the function of transporters in vivo [100]. First, studies with healthy subjects [101] and with patients with epilepsy [102, 103] have clearly provided data demonstrating that the imaging in vivo function of transporters not only in animal models but also in humans will soon become a very valuable tool for understanding drug transport across barriers shielding the brain. It is important to note that the transporter function in vivo cannot only be monitored by PET, but also by single-photon emission-coupled tomography [104]. The development of novel imaging probes should in the future certainly help the development of novel drugs with targets in the CNS, as their passage through the barriers can be followed in vivo. This methodology, together with stringent quality control of the label [105], will certainly also help to address issues of transporter-based drug–drug interactions at the BBB, where clearly more information for clinical practice is needed [20, 106, 107].