CNS Drug Design: Balancing Physicochemical Properties For .

2y ago
7 Views
2 Downloads
1.57 MB
25 Pages
Last View : 12d ago
Last Download : 3m ago
Upload by : Karl Gosselin
Transcription

Perspectivepubs.acs.org/jmcCNS Drug Design: Balancing Physicochemical Properties for OptimalBrain ExposureZoran Rankovic*Eli Lilly and Company, 893 South Delaware Street, Indianapolis, Indiana 46285, United StatesS Supporting Information*ABSTRACT: The human brain is a uniquely complex organ, which has evolved a sophisticatedprotection system to prevent injury from external insults and toxins. Designing molecules that canovercome this protection system and achieve optimal concentration at the desired therapeutictarget in the brain is a specific and major challenge for medicinal chemists working in CNS drugdiscovery. Analogous to the now widely accepted rule of 5 in the design of oral drugs, thephysicochemical properties required for optimal brain exposure have been extensively studied in anattempt to similarly define the attributes of successful CNS drugs and drug candidates. This bodyof work is systematically reviewed here, with a particular emphasis on the interplay between themost critical physicochemical and pharmacokinetic parameters of CNS drugs as well as their impacton medicinal chemistry strategies toward molecules with optimal brain exposure. A summary ofmodern CNS pharmacokinetic concepts and methods is also provided.1. INTRODUCTIONTraditionally, the development of medicines for central nervoussystem (CNS) disorders has been an intense and profitableresearch activity for the pharmaceutical industry. Despite therecent divestment of research in some areas, such as psychiatricdisorders (i.e., depression and schizophrenia),1 this trend is likelyto continue as a result of the acute medical needs and marketopportunities created by an increasing human life span. It isestimated that, in the absence of disease-modifying treatments,age-related neurodegenerative conditions such as Alzheimer’sdisease and Parkinson’s disease will be the major drivers of thepredicted increase in US health care expenditures from thecurrent 15% of gross domestic product (GDP) to 29% ofGDP in 2040.2 Additionally, the brain is a common site of metastases for some of the most prevalent cancers, such as lung andbreast cancers,3 and is the second leading cause of death in theUnited States among males younger than 40 years and femalesyounger than 20 years.4 Patients who present with metastaticbrain cancer have a poor prognosis with a median survival of only2.5 months,5 which is at least partially a consequence of poorCNS exposure for currently available drugs that were developedto treat systemic disease. Consequently, CNS exposure isincreasingly seen as an important requirement in the development of new cancer treatments.6However, the pressures of medical needs and the incentivesof market opportunities are countered by the challenges andcomplexities of CNS drug discovery. In addition to the difficulties of CNS target validation and clinical translation,designing therapeutic agents that are able to reach and effectivelymodulate targets in the brain makes CNS research one of themost challenging endeavors in drug discovery. This is largely dueto a highly sophisticated protection system that the brain hasevolved to preserve its physiological environment and shielditself from external insults and toxins. An important component 2014 American Chemical Societyof this system is the blood brain barrier (BBB), which lies at theinterface between the blood capillaries of the brain and braintissue.7 The BBB has a very complex multicellular organizationcomprising brain endothelial cells that line the blood vessels andform the brain capillary endothelium as well as surrounding cells,including pericytes, astrocytes, and neurons. The BBB provides afully autonomous milieu for the cells within the CNS, enablingselective access of required nutrients and hormones whileremoving waste and diminishing exposure to potentially harmfulxenobiotics.7 The blood cerebrospinal fluid barrier (BCSFB)is a similar barrier between the blood capillaries and thecerebrospinal fluid (CSF), which fills the ventricles and bathesthe external surface of the brain to provide buoyancy andmechanical protection within the skull.8 In brain tissue, theCSF is exchanged with the brain’s interstitial fluid (ISF), whicheliminates metabolic waste from the CNS and regulates thechemical environment of the brain (Figure 1).Since the density of the capillaries within the brainparenchyma is so high that virtually every neuron is suppliedby its own capillary (the combined capillary length in the humanbrain is 400 miles), it is estimated that the BBB surface area( 120 square feet) is approximately 5000 times larger than theBCSFB surface area. Consequently, the BBB is thought to havea much greater role than the BCSFB in CNS drug delivery tothe brain.9,10 Active transport and passive diffusion throughendothelial cells are the two principal mechanisms by whichmolecules can enter the brain. In contrast to the endothelial cellsof capillaries elsewhere in the body, diffusion through theintercellular space between cells (paracellular) is effectivelyprecluded by the presence of the tight junctions thatcharacterize the BBB and BCSFB. For example, the capillaryReceived: October 5, 2014Published: December 11, 20142584dx.doi.org/10.1021/jm501535r J. Med. Chem. 2015, 58, 2584 2608

Journal of Medicinal ChemistryPerspectiveFigure 1. Principal CNS PK parameters and concepts. (a) Schematic representation of bound and unbound drug concentration equilibrium acrossBBB and brain main compartments: interstitial fluid (ISF), intracellular fluid (ICF), and cerebrospinal fluid (CSF); (b) Kp,uu values significantlylower or higher than 1 indicate active transport across the BBB.is particularly challenging for CNS targets. The existence of theBBB renders classical PK parameters, such as oral bioavailabilityand plasma concentration, insufficient for assessing drug exposureand time courses in the brain. For a meaningful assessment ofCNS drug candidates, additional data and more sophisticatedPK methods are required.17 212.1. Free Drug Hypothesis. The most direct informationon drug exposure at the site of action is provided by receptoroccupancy (RO) studies. In the clinical setting, brain RO ismeasured using radiolabeled tracer ligands whose distributioncan be determined by noninvasive imaging methods, such assingle photon emission computed tomography (SPECT) andpositron emission tomography (PET).22 In preclinical studies,the tracer distribution is established mainly by scintillation spectroscopy of dissected brain tissue. These studies are characterizedby the need for radiolabeled ligands, long turnaround times, andrelatively low success rates (particularly for agonists). Therefore,due to technical complexities as well as time and costs involved,RO studies tend to be performed mainly for the most advancedcompounds. Consequently, the more easily obtained drug totalbrain concentration (Cb) and whole brain-to-plasma ratio (Kp Cb/Cp; also referred to as the B/P ratio) have historically beenused as the main decision-making drivers in CNS drug discovery.However, recent evidence suggests that relying solely on theseparameters is insufficient for adequate understanding of theCNS exposure needed to establish robust PK/PD correlations.23This insufficiency arises from the fact that only a fraction of totaldrug concentration in the plasma (Cp) and tissues, such as brain(Cb), is unbound from the tissue proteins or lipids and is freeto diffuse across biological barriers and tissues to reach theintended therapeutic target (Cu,p Cp f u,p and Cu,b Cb f u,b,respectively), as depicted in Figure 1a. Because the unboundplasma and tissue fractions tend to be different, the unboundbrain-to-plasma ratio (Kp,uu) is often considerably different fromthe total brain-to-plasma ratio (Kp).The potential for misleading conclusions derived from thereliance on total brain and plasma concentrations is wellexemplified by comparative in vivo PK/PD studies of morphine,1, and one of its active metabolites, morphine-6-glucuronide 2(M6G).24 Both compounds are thought to exert their centrallymediated analgesic affect through the activation of mu-opioidreceptors (MOR) expressed in CNS.endothelial cell junctions in the brain are sufficiently tight torestrict the movement of ions such as Na and Cl and consequently display much higher transendothelial electrical resistancethan peripheral capillaries ( 1000 versus 20 ohm/cm2).11 Thisrestrictiveness is thought to protect the brain from fluctuations inionic composition that may occur, e.g., during a meal or physicalactivity.The majority of CNS drugs are small molecules that crossthe BBB via the transcellular passive diffusion route.12Designing molecules that can achieve optimal concentrationat the desired therapeutic target in the brain is a unique andmajor challenge for medicinal chemists working in CNSdrug discovery.13 15 Tremendous progress has been made inrecent years in terms of enabling the development of robustpharmacokinetic-pharmacodynamic (PK/PD) relationships forcentrally acting agents as well as in understanding how theserelationships are influenced by molecular physicochemicalproperties. The objective of this Perspective is to review thisprogress and highlight successful medicinal chemistry strategiestoward molecules that can achieve optimal brain exposure viatranscellular passive diffusion. Alternative brain delivery systemsutilizing BBB active transport are beyond the scope of thisPerspective, and the reader is referred to an excellent review ofthis topic.16At this point, it should also be noted that the term “optimal”is used loosely in this context, since there is no single value ordesirable concentration range, but what is considered to beoptimal exposure depends on a multitude of variables, includingdrug potency, mode of action (e.g., agonist vs antagonist),binding kinetics (e.g., fast vs slow dissociation constant orreversible vs irreversible binders), pharmacokinetic properties,and therapeutic index. One could define the optimal brainexposure as a free drug concentration at the site of the intendedtarget expressed in the brain that produces maximum desiredand minimum undesired effects (best therapeutic window) overthe time course deemed to be suitable for the intended dosingregimen (e.g., once-a-day).2. IMPORTANT CNS PHARMACOKINETIC CONCEPTSAND PARAMETERSAchieving a good understanding of the PK and PD relationshipsin preclinical disease models, which is a critical prerequisite forsuccessful drug discovery programs across all therapeutic areas,2585dx.doi.org/10.1021/jm501535r J. Med. Chem. 2015, 58, 2584 2608

Journal of Medicinal ChemistryPerspectiveused in combination with the drug in vitro potency data toestimate the brain exposure or dose required for in vivo efficacy.However, it is important to note that this assumption is notvalid for compounds thatTable 1. Morphine and Morphine-6-glucuronide ParadoxExplained by Considering Drug Cu,b Data27,28 Display a low rate of passive permeability across theBBB/BCSFB (require a long time to reach equilibriumacross membranes). Are actively transported in or out of the brain.compd12MORaAUCbbKi (nM)(μM/min)226318642KpISF AUCcIn these cases, the disconnect between a compound’s freeplasma and brain concentrations (Cu,b Cu,p) is translatedinto Kp,uu values, which can be either above or below 1. Kp,uuvalues below 1 indicate compounds that are subject to activeefflux and/or have low passive permeability across the BBB(Figure 1b).31 This profile is suitable for peripherally activedrugs, such as (S)-cetirizine (Kp,uu 0.22), a second-generationantihistamine that was intentionally designed to minimize brainexposure to avoid sedation and other CNS side effects generallyassociated with first-generation antihistamines.32 However,pursuing compounds with low Kp,uu values for CNS targets isless desirable, due not only to the higher peripheral exposureneeded to achieve efficacious brain levels and the consequentlyincreased risk of side effects but also to the difficulty withestimating low human Cu,b, which results in low confidencehuman dose predictions.23 Kp,uu values greater than 1, asexemplified by the first-generation antihistamine diphenhydramine (rat Kp,uu 5.5), indicate an active uptake processmediated by influx transporters.33 Active transport may offer analternative approach for achieving optimal brain exposure formolecules whose properties are nonconducive to passive BBBpenetration, as exemplified by gabapentin or L-DOPA that aretransported via L-type amino acid transporter-1 (LAT1).34However, intentionally targeting specific transport proteins ortranscytosis mechanisms for CNS drug delivery remainschallenging, and, although this approach has attracted interestacross the pharmaceutical industry and academia, research inthis area is still in its infancy.16 A successful small molecule CNSdrug candidate typically complies with the free drug hypothesisand displays a ratio of the unbound drug in the brain to theunbound drug in the plasma (Kp,uu value) close to 1.19,35 Witha Kp,uu in mouse of 0.98, the antidepressant venlafaxine is onesuch example.36To simplify the process and increase the throughput, Kp,uuvalues are often estimated from single-time-point measurements. This is generally an acceptable approximation; however,one should be aware that such data can be misleading if theselected time point was before the system could reach steadystate. In this context, more reliable Kp,uu values are generatedusing the area under the concentration time curves (AUCs)for total and unbound concentrations obtained from timecourse experiments (Kp,uu AUCu,b/AUCu,p). Again, AUCsand steady-state concentrations can be used interchangeablyin this equation as they measure the same property.36 Whenmaking important decisions on the basis of Kp,uu data, it isworth remembering that these values include experimentalerrors from four separate in vivo and in vitro measurements(total plasma concentrations, total brain concentrations, PPB,and brain tissue binding). For comparing and prioritizing keycompounds, one might like to consider generating multipleKp,uu data points (e.g., n 2) to increase confidence in the Kp,uuvalues.2.2. Experimental Methods Commonly Used ToMeasure or Estimate Brain Exposure. Microdialysis is theKp,uu(μM/min)0.740.05793360.510.56a 3[ H]-Naloxone displacement binding assay in MOR-expressing cellmembranes.25 bTotal brain AUC concentration in rat, 10 mg/kg (s.c.).cMeasured by in vivo transcortical microdialysis.28Although the two compounds exhibit similar activities in vitro(Table 1),25 their similar in vivo efficacy26 was unexpectedconsidering the poor total brain/plasma ratio (Kp 0.05) andmuch lower rate of BBB permeation, expressed as permeabilitysurface area product (PS 0.11 μL/min/g), for 2 comparedwith those of morphine (Kp 0.54; PS 3.5 μL/min/g).In fact, based on the classic concepts of CNS exposure, 2should have no in vivo efficacy.27 To explain this phenomenon,several hypotheses, such as glucuronide 2 activation of a newand not yet discovered receptor subtype, have been proposed.24However, this apparent paradox could also be explained byconsidering the unbound (free) rather than total drugconcentrations in the brain.28 Actually, whereas the 2 totalbrain AUC concentration is 4 5-fold lower compared with thatof morphine, the drug ISF AUC (unbound) is approximately4-fold higher than morphine (Table 1), which may explain thesimilar in vivo efficacy observed for these two compounds.27These general observations have been confirmed by multipleCNS drug studies reported over the past decade: ISF is therelevant brain compartment for drug pharmacodynamics effect,and ISF drug concentration is not reflected by the total brainconcetration.15,23,27It is important to note that the total B/P ratio (Kp) can bemisleading, even when comparing structurally similar compounds. For example, a Kp value of 0.04 suggests significantlylower brain penetration for (R)-cetirizine compared with thatof its enantiomer, (S)-cetirizine, which has a Kp value of 0.22.29However, the unbound drugs in the brain (Cu,b) and plasma(Cu,p) show that (S)- and (R)-cetirizine have very similar, if notidentical, effective compound levels in the brain, with Kp,uu(Cu,b/Cu,p) values of 0.17 and 0.14, respectively. The observedKp enantioselectivity has been rationalized by the lower plasmaprotein binding (PPB) of (S)-cetirizine (PPB 50%) comparedwith that of (R)-cetirizine (PBB 85%).29The importance of free drug concentrations is embeddedin the central tenet of in vivo pharmacology, the free drughypothesis, which states that: (1) the free (unbound) drugconcentration (Cu) at the site of action is responsible forpharmacological activity in vivo and (2) at steady state and inthe absence of active transport, the free drug concentration isthe same on both sides of any biomembrane (e.g., the BBB).30Thus, at steady state, one could equate the drug unbound brainconcentration (Cu,b) with its experimentally more accessibleunbound plasma concentration (Cu,p). The Cu,p could be then2586dx.doi.org/10.1021/jm501535r J. Med. Chem. 2015, 58, 2584 2608

Journal of Medicinal ChemistryPerspectiveon the equilibrium dialysis of compounds between brainhomogenate and buffer, which is used to calculate the unboundconcentration in the brain (Cu,b f u,b Cb).42 An alternative tothe in vitro f u,b determination method preserves the cellularstructure of the brain tissue using brain slices.43 In contrast tothe brain homogenate method, this approach can capturepotential differences between ISF and intracellular fluid (ICF)drug concentrations, as observed for gabapentin.35 Since thisphenomenon is thought to be relatively rare, generallyassociated with compounds that are actively transported intotissue, the more elaborate slice method is considered mainlywhen other techniques fail to explain the PK/PD relationship.It is important to note that due to a very poor correlation thefree fraction in plasma ( f u,p) is not a suitable surrogate for f u,b.44This lack of correlation is mostly a consequence of the verydifferent lipid and protein contents of the two compartments,with plasma having twice as much protein and the brain having20-fold more lipids.28 However, the free drug hypothesis infersthat at steady state, irrespective of differences in f u,p and f u,tissue,the unbound drug concentration in the tissue of interest ismirrored by Cu,p, i.e., Cu,plasma Cu,tissue. For example, followingeither single or multiple doses of the antifungal drugfluconazole, similar free drug concentrations are found in arange of body fluids, including plasma, CSF, saliva, breast milk,vaginal secretions, sputum, and prostatic and seminal vesiclefluid.45Species dependence of plasma protein binding is a welldocumented phenomenon. Summerfield and colleagues atGlaxoSmithKline reported marked variations on comparinghuman f u,p values with rat (R2 0.49) and pig (R2 0.65) for aset of 21 CNS drugs and PET tracers.46 This is hardly surprisingconsidering the evolutionary divergence in the structure ofblood proteins and overall blood content. In contrast, however,Summerfield et al. found good interspecies correlations for f u,bvalues among rat, pig, and human (R2 0.9; n 21).46 Similarlyhigh degrees of f u,b correlation were reported by Di et al. atPfizer for a set of 47 drugs in six different species and in twodifferent strains of rat.47 This evidence supports the commonlyaccepted practice of using rodent f u,b values in human PKprediction models. It was suggested that interspecies similaritiesin f u,b reflect the conserved brain general morphology acrossspecies and mainly nonspecific nature of drug binding to brainlipids, since the brain-lipid content is higher than that in plasmaand protein binding within brain tissue is thought to beinsignificant.48 Therefore, if species differences in brain exposureof passively diffused drugs are observed, then they are likely tobe driven by differences in plasma protein binding where thehomology varies significantly from one species to another.Plasma protein and brain tissue binding are clearly veryimportant parameters for understanding drug PK, PD, and safetyproperties. However, theoretical analyses and experimentalobservations suggest that these parameters should not be theprimary focus of optimization efforts in drug discovery.30,49Indeed, high tissue binding is generally not considered to bea liability, as many successful drugs exhibit low f u, e.g., 24% of alldugs launched between 2003 and 2013 have PPB 99%,whereas 15 out of 32 the most prescribed CNS drugs have f u,b 5%.49 It has also been pointed out that lowering brain tissuebinding does not necessarily lead to higher C u,b andconsequently, instead of focusing on f u,p and f u,b, discoveryteams should place a stronger emphasis on minimizing intrinsicclearance and BBB efflux transport to achieve high Cu,p andCu,b.49 It is worth noting that improved Cu,p and Cu,b levels,method of choice for the direct measurement of unbound drugconcentrations (Cu,b) within the brain ISF.28 This powerfultechnique enables investigations into the extent as well as therate of drug delivery to the brain, providing critical parametersfor PK/PD modeling. However, similar to RO studies, technicalchallenges and resource requirements limit its use in drugdiscovery to more advanced drug candidates.CSF sampling is a less demanding and therefore more widelyemployed technique for assessing brain exposure during earlydrug discovery. The CSF is separated from the ISF compartment by only a single layer of ependyma, a cell layer that doesnot have tight junctions. Due to the very low protein levelsin CSF, approximately 0.2 mg/mL compared to 70 mg/mL inplasma, drug CSF concentrations (CCSF) can be approximatedby Cu,b. Indeed, microdialysis studies of brain distribution, aswell as PK/PD studies for a range of preclinically and clinicallyused compounds, suggest that the drug CSF level is generallya good approximation of the brain ISF concentration.37 Acomparative study of brain exposure in rats for 39 structurallydiverse drugs demonstrated a 3-fold agreement between drugswith respect to Kp,uu (ISF) and Kp,uu (CSF), within a similarrank order.38 However, one should remember that the CSFapproximation fails for compounds that are subject to activeefflux or influx (Kp,uu 1 or 1), presumably due to thelower active transport capacity of the blood cerebrospinal fluidbarrier compared with that of the blood brain barrier.39For example, while the CCSF significantly overpredicts the Cb,uof highly effluxed drugs, such as loperamide, it underpredictsthe Cb,u of actively influxed drugs, such as oxycodone.38CSF sampling has a high value in CNS translational studies,being the only generally applicable method of obtaining information on free drug concentrations in the human brain.40 Studiescomparing rat and human CSF data showed that the humanKp,uu (CSF) was, on average, approximately 3-fold higher thanthe rat Kp,uu (CSF).38 Some of the proposed explanations forthe observed species difference include the disease state of thepatients (e.g., altered BBB function), active transport, kinetic biasdue to timing of CSF sampling, and the different sampling site(cisterna magna in the rat vs lumbar puncture in the patients).38In any case, the 3-fold error is generally considered to be oflittle pharmacokinetic or pharmacodynamic consequence for theprediction of drug concentrations in human CSF on the basisof rat CSF concentrations. However, prediction of rat unboundbrain exposure based on rat CSF concentration includes anadditional 3-fold error, which results in a cumulative 9-fold errormargin in prediction of human Cu,b solely on the basis of rat CSFexposure.41 This may have significant consequences if a drug hasa narrow therapeutic window. In order to enhance the predictivepower of modeling human Cu,b and related PK/PD relationshipson the basis of preclinical data, more complex physiologicallybased PK (PBPK) algorithms have been developed in recentyears, which also incorporate mechanistically relevant in vitrodata and integrate drug-dependent physiological and biologicalparameters, as they vary between species, subjects, age, or diseasestate.41,70A more screening-based approach commonly used in earlydrug discovery due to much higher throughput is the brainhomogenate-based estimation of unbound drug concentrationsin the brain (Cu,b). This method relies on a combination of thein vitro measured fraction unbound in the brain ( f u,b) and therelatively straightforward in vivo determined brain totalconcentration (Cb). The f u,b data are generated in a highthroughput fashion using a simple and elegant method based2587dx.doi.org/10.1021/jm501535r J. Med. Chem. 2015, 58, 2584 2608

Journal of Medicinal ChemistryPerspectivesignificantly lower incidence of P-gp-mediated efflux than nonCNS drugs, suggesting the greater importance of P-gp efflux forCNS therapeutics.57 This is probably because, in contrast to theintestine, drug plasma concentrations at the BBB can rarely reachP-gp saturation levels.In humans, P-gp is encoded by a single gene (MDR1),whereas in rodents, there are two genes, mdr1a and mdr1b.They are both expressed in brain; however, only mdr1a is foundat the BBB, whereas mdr1b is expressed in brain parenchyma.58The significant species-dependent substrate recognitiondivergence is a relatively rare occurrence,54,59 although compounds that exhibit difference between rodent and humanefflux ratios have been observed.60 These observations areconsistent with a relatively high primary sequence homologybetween human and rodent P-gp transporters, also shared withother species, e.g., there is 85 93% sequence homology amonghuman, mouse, rat, dog, and rhesus monkey.59The continuous research focus on P-gp is a reflection of itsapparent role as the main BBB gatekeeper with the broadestsubstrate specificity. Its molecular recognition promiscuity isbetter understood since the recent publication of the mouseP-gp apo crystal structure at 3.8 Å resolution, along with twoco-crystal structures with the enantiomers of a P-gp inhibitor,which revealed an internal cavity of 6000 Å3 with a very largehydrophobic substrate binding site and two ATP-bindingdomains.61 The crystal structures support a “hydrophobicvacuum cleaner”54 model whereby the substrate partitionedfrom the outside of the cell into the membrane bilayer entersthe P-gp binding pocket through an open portal at the level ofthe inner leaflet of the lipid bilayer. With the substrate in place,ATP binds to the two nucleotide binding domains, triggering alarge conformational change that presents the substrate to theextracellular space while occluding the cytosolic side, therebyproviding unidirectional transport to the outside. ATPhydrolysis and dissociation returns the protein to its inwardfacing conformation, ready for the next transport cycle. Theproposed transport mechanism is also consistent with theobservation that increasing passive permeability is oftenaccompanied by a lower likelihood of P-gp efflux.62 It ispossible that compounds that rapidly diffuse across themembrane could overwhelm the P-gp efflux process and, tosome extent, escape into the cytosol. Therefore, increasing passivepermeability can be seen as a potential strategy for addressing therisk of efflux by P-gp.63Two decades after the discovery of P-gp, another member ofthe ATP-binding cassette transporter superfamily implicated incancer multidrug resistance was identified, first in the multidrugresistant human breast cancer cell line (hence the name breastcancer resistance protein, or BCRP)64 and then found widelyexpressed in tissues including gastrointestinal tract, liver, andkidney, as well as the luminal membrane of the brain capillaries.65Studies using BCRP and the dual P-gp/BCRP knockout mousepoint toward BCRP and P-gp having synergistic and potentiallycompensatory roles in brain exclusion of cancer drugs such asdasatanib66 and sunitinib.67 On the basis of the currently availablestructure activity relationship, an amino-heteroraomatic motif issuggested to be an important substrate recognition element forBCRP.53 However, the relative contribution of BCRP to overalldrug BBB efflux capacity, and its broader impact on brainexposure of therapeutic agents outside oncology, is stillunclear.68,69 Other well-characterized members of the ABCtransporter superfamily, such as multidrug resistant protein 1(MRP1), MRP4, and MRP5, are also found to be expressed inespecially when achieved by reducing lipophilicity, are oftenaccompanied by increased unbound concentrations (e.g., Tables 3and 4). Ultimately, assuming all other properties being equal, onemay still prefer drugs with higher f u levels, which may translateinto lower daily dose requirements.2.3. BBB Permeability and Active Transport. A fineinterplay between passive membrane permeability and activetransport processes at the BBB level, together with plasma/braintissue binding, is widely recognized as being the primarydeterminant of drug disposition within the CNS. However,despite decades of experience in culturing brain endothelial cells,there is still no satisfactory in vitro model of the BBB availableto date. Culturing primary brain endothelial cells is too difficultfor routine applications, whereas immortalized brain endothelialcell lines lack the required junction tightness. Still, usefulhigh-throughput permeability assays have been developed usingalternative cell lines, such as Caco2 (heterogeneous humanepithelial colorectal adenocarcinoma cells), porcine kidney(LLC-PK1), and the industry favorite MDCK (Madin Darbycanine kidney) cell lines.50 Although these cells lines are neitherendothelial cells nor do they originate from the brain, thetightness of the monolayer results in permeability values (Papp)that correlate well with in vivo permeation. Due to its significantly lower cost and higher throughput, a parallel artificialmembrane permeability assay (PAMPA)51 that lack

CNS Drug Design: Balancing Physicochemical Properties for Optimal Brain Exposure Zoran Rankovic* Eli Lilly and Company, 893 South Delaware Street, Indianapolis, Indiana 46285, United States * S Supporting Info

Related Documents:

Effective date: Apr 30, 2020 . IEC/EN 60335-1, IEC/EN 60335-2-2 CNS 3765, CNS 3765-2 100128 電熨斗 IEC/EN 60335-1, IEC/EN 60335-2-3 CNS 3765, CNS 3765-3 100129 旋轉式脫水機 IEC/EN 60335-1, IEC/EN 60335-2-4 CNS 3765-4, CNS 2926 sub-clause 3.3, 3.4 100130 洗碗機 IEC/EN 60335-1

8. Load Balancing Lync Note: It's highly recommended that you have a working Lync environment first before implementing the load balancer. Load Balancing Methods Supported Microsoft Lync supports two types of load balancing solutions: Domain Name System (DNS) load balancing and Hardware Load Balancing (HLB). DNS Load Balancing

CNS Cable to Lake Riverside X Technical Services/CNS 300,000 Budget Completed 2010 Replacement bucket trucks X Technical Services/CNS 175,000 Budget . Thomasville X Technical Services/CNS 1,000,000 Grant & Budget Completed 2009 Automated Meter Reading System X Technical Services/CNS 3,000,000 Budget

to propose/decide on an European CNS rationalisation roadmap Advise SESAR to: Deploy ICAO standardised technology/services in the short term to reduce the congestion and make the CNS infrastructure improving the network performance. Provide a clear path for the CNS technology evolution (cf. Roadmap). Study the Perf.

principles, applications, strengths and limitationsof a variety of modal-ities commonly used to investigate the physicochemical characteristics of nanomaterials (Table 1). 2. Overview of physicochemical characteristics Typically, engineered materials with dimensions in the nanometer scale areintermediatesbetween isolated small molecules andbulk ma-

Balancing involves redistributing the mass which may be carried out by addition or removal of mass from various machine members Balancing of rotating masses can be of 1. Balancing of a single rotating mass by a single mass rotating in the same plane. 2. Balancing of a single rotating mass by two masses rotating in different planes. 3.

Internal Load Balancing IP: 10.10.10.10, Port: 80 Web Tier Internal Tier Internal Load Balancing IP: 10.20.1.1, Port: 80 asia-east-1a User in Singapore Database Tier Database Tier Database Tier External Load Balancing Global: HTTP(S) LB, SSL Proxy Regional: Network TCP/UDP LB Internal Load Balancing ILB Use Case 2: Multi-tier apps

This paper aims to extend this range and introduces a novel engineering application of Origami: Folded Textured Sheets. Existing applications of Origami in engineering can broadly be catego-rized into three areas. Firstly, many deployable structures take inspiration from, or are directly derived from, Origami folding. Examples are diverse and range from wrapping solar sails [Guest and .