+ All Categories
Home > Documents > Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

Date post: 03-Feb-2017
Category:
Upload: hieu
View: 213 times
Download: 0 times
Share this document with a friend
13
1. Introduction 2. Brain pathophysiology and anti-HIV therapeutic agents 3. Delivering anti-HIV medication to brain bypassing the BBB 4. Absorption of anti-HIV therapeutic agents through BBB 5. Conclusion 6. Expert opinion Review Advances in brain targeting and drug delivery of anti-HIV therapeutic agents Abeer M Al-Ghananeem , Michael Smith, Maria L Coronel & Hieu Tran Sullivan University, College of Pharmacy, Department of Pharmaceutical Sciences, Louisville, KY, USA Introduction: Human immunodeficiency virus (HIV) is a neurotropic virus that enters the central nervous system (CNS) early in the course of infection. Although antiretroviral drugs are able to eliminate the majority of the HIV virus in the bloodstream, however, no specific treatment currently exist for CNS infections related to HIV. This is mainly attributed to the poor penetrabil- ity of antiretroviral therapy across the blood--brain barrier (BBB), and the protective nature of the BBB. Therefore, in order to increase the efficacy of anti-HIV drugs, novel drug delivery methodologies that can exhibit activity in the CNS are most needed and warranted. Areas covered: In this review article, the authors discussed the challenges with delivering drugs to the brain especially under HIV infection pathophysiology status. Also, they discussed the approaches currently being investigated to enhance brain targeting of anti-HIV drugs. A literature search was performed to cover advances in major approaches used to enhance drug delivery to the brain. Expert opinion: If drugs could reach the CNS in sufficient quantity by the methodologies discussed, mainly through intranasal administration and the utilization of nanotechnology, this could generate interest in previously aban- doned therapeutic agents and enable an entirely novel approach to CNS drug delivery. Keywords: AID, BBB, brain targeting, CNS, dementia, HIV, nanotechnology, nasal delivery, prodrug Expert Opin. Drug Deliv. (2013) 10(7):973-985 1. Introduction Human immunodeficiency virus (HIV) appears to be harbored in the brain, as indi- cated by the presence of large quantities of unintegrated viral DNA in the brain of HIV-infected individuals [1]. A wide variety of nervous system disorders occur in patients with HIV [2,3]. The most common and devastating central nervous system (CNS) complication associated with HIV infection is a progressive dementia, which is also referred to as dementia [4,5]. Dementia is a metabolic encephalopathy induced by HIV infection and fueled by immune activation of macrophages and microglia [6]. NeuroAIDS is the neurological complications associated with acquired immuno- deficiency syndrome (AIDS). The exact mechanism of virus entry into the brain is not clearly elucidated; how- ever the resulting infection leads to a number of CNS disorders [7,8]. Currently, no specific treatment exists for neuroAIDS, which is mainly attributed to the poor pen- etrability of antiretroviral therapy (ART) across the blood--brain barrier (BBB). The selective permeability of the BBB is due to the distinct morphology and enzymatic properties of endothelial cells that enable them to form complex tight junctions 10.1517/17425247.2013.781999 © 2013 Informa UK, Ltd. ISSN 1742-5247, e-ISSN 1744-7593 973 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by Gazi Univ. on 08/15/14 For personal use only.
Transcript
Page 1: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

1. Introduction

2. Brain pathophysiology and

anti-HIV therapeutic agents

3. Delivering anti-HIV medication

to brain bypassing the BBB

4. Absorption of

anti-HIV therapeutic agents

through BBB

5. Conclusion

6. Expert opinion

Review

Advances in brain targeting anddrug delivery of anti-HIVtherapeutic agentsAbeer M Al-Ghananeem†, Michael Smith, Maria L Coronel & Hieu TranSullivan University, College of Pharmacy, Department of Pharmaceutical Sciences, Louisville,

KY, USA

Introduction: Human immunodeficiency virus (HIV) is a neurotropic virus that

enters the central nervous system (CNS) early in the course of infection.

Although antiretroviral drugs are able to eliminate the majority of the HIV

virus in the bloodstream, however, no specific treatment currently exist for

CNS infections related to HIV. This is mainly attributed to the poor penetrabil-

ity of antiretroviral therapy across the blood--brain barrier (BBB), and the

protective nature of the BBB. Therefore, in order to increase the efficacy of

anti-HIV drugs, novel drug delivery methodologies that can exhibit activity

in the CNS are most needed and warranted.

Areas covered: In this review article, the authors discussed the challenges with

delivering drugs to the brain especially under HIV infection pathophysiology

status. Also, they discussed the approaches currently being investigated to

enhance brain targeting of anti-HIV drugs. A literature search was performed

to cover advances in major approaches used to enhance drug delivery to

the brain.

Expert opinion: If drugs could reach the CNS in sufficient quantity by the

methodologies discussed, mainly through intranasal administration and the

utilization of nanotechnology, this could generate interest in previously aban-

doned therapeutic agents and enable an entirely novel approach to CNS

drug delivery.

Keywords: AID, BBB, brain targeting, CNS, dementia, HIV, nanotechnology, nasal delivery,

prodrug

Expert Opin. Drug Deliv. (2013) 10(7):973-985

1. Introduction

Human immunodeficiency virus (HIV) appears to be harbored in the brain, as indi-cated by the presence of large quantities of unintegrated viral DNA in the brain ofHIV-infected individuals [1]. A wide variety of nervous system disorders occur inpatients with HIV [2,3]. The most common and devastating central nervous system(CNS) complication associated with HIV infection is a progressive dementia, whichis also referred to as dementia [4,5]. Dementia is a metabolic encephalopathy inducedby HIV infection and fueled by immune activation of macrophages andmicroglia [6].

NeuroAIDS is the neurological complications associated with acquired immuno-deficiency syndrome (AIDS).

The exact mechanism of virus entry into the brain is not clearly elucidated; how-ever the resulting infection leads to a number of CNS disorders [7,8]. Currently, nospecific treatment exists for neuroAIDS, which is mainly attributed to the poor pen-etrability of antiretroviral therapy (ART) across the blood--brain barrier (BBB). Theselective permeability of the BBB is due to the distinct morphology and enzymaticproperties of endothelial cells that enable them to form complex tight junctions

10.1517/17425247.2013.781999 © 2013 Informa UK, Ltd. ISSN 1742-5247, e-ISSN 1744-7593 973All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 2: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

with minimal endocytic activity. This provides a physiologicalbarrier that limits the transport of many blood-borneelements such as macromolecules and circulating leukocytesto the brain [9]. Previous studies report that delivery of ARTto the brain is limited especially due to the physical structureof the BBB, presence of efflux pumps and higher expression ofmetabolizing enzymes, which makes BBB an effective barrieragainst many antiretroviral drugs [10]. Therefore, in order toincrease the efficacy of anti-HIV drugs, novel approaches todeliver antiretroviral drugs to the brain are warranted.Brain drug delivery is a challenge in the treatment of dis-

eases such as neurological disorders. Drugs may be adminis-tered directly into the CNS, or indirectly through oral andintravenous administration. However, transport of drugsfrom systemic circulation into the CNS is restricted by thepresence of the BBB and blood--cerebrospinal fluid barrier(BCSFB), which are featured by tight junctions connectingthe cerebral endothelial and epithelial cells of the choroidplexus, respectively. Most charged, hydrophilic, water-soluble substances, and large therapeutic agents are inhibitedor prevented from entering the brain by the BBB [11,12].Accordingly, to resolve the inhibitory nature of the BBB to

some of the ART drugs, there is a dire need for novel thera-peutics that exhibit antiviral activities in the CNS, attenuateinflammatory pathways and enhance drug delivery intothe brain.This review presented an overview of the brain pathophys-

iology and the current advances in delivery of anti-HIV drugs.

An overview of options in delivering anti-HIV medications tothe brain bypassing the BBB including intranasal delivery andsurgical alternatives were provided. The absorption of anti-HIV medications through BBB was discussed utilizing thenanotechnology, structural manipulation as well as throughaltering BBB passage pathways.

2. Brain pathophysiology and anti-HIVtherapeutic agents

2.1 Pathogenesis of HIV infectionHIV is an obligate intracellular ribonucleic acid (RNA) retro-virus that is highly dependent on host cellular functions tosurvive, replicate and proliferate. This virus is transmittedprimarily through blood and body fluids causing AIDS.The virus interacts with a large number of different cells inthe body and is capable of escaping numerous host immunedefenses against it [13]. Although the exact mechanism of virusentry into the brain is not clearly elucidated, a sequence ofevents occurs to achieve survival. HIV surface gp120 glyco-protein will attach to the CD4 receptor located on the hostcell membrane and the interaction of the gp120 proteinand CD4 complex leads to virus--cell membrane fusion medi-ated by transmembrane gp41 protein [14]. Effective survival ofretroviruses depends on exploiting host cell processes withinthe cell. Viral integrase is responsible for integration of areverse-transcribed viral cDNA into the chromosome of theinfected cell which is a crucial step in the early phases ofthe retroviral life cycle. Retroviral integration causes hostcell chromosome damage. Activation of DNA repair canresult in arrest of cell cycle, which provides time for DNArepair. However, if DNA repair system was left unrestored,apoptosis or cell death can occur [15]. Retroviruses differ intheir preferences for sites for viral DNA integration in thechromosomes of infected cells. HIV integrates preferentiallywithin active transcription units [16]. Proper integration byblocking either integrase catalysis or the function of cellularintegration may potentially offset chromatin alteration pre-venting cell apoptosis.

HIV contains a viral envelope surrounded by projectionsformed from the proteins gp120 and gp41. During an activeinfection, the HIV envelope gp120 glycoprotein is shed bythe virus from HIV-infected immune cells [17]. In the CNS,this generates inflammation and oxidative stress that contrib-utes to the development of the AIDS Dementia Complex(ADC). The correlation of the excitotoxic effects ofgp120 to astroglial cells has been demonstrated through lipidperoxidation and altered glutamine release to brain cell dam-age. Maintaining an active balance activity of glutamine syn-thase (GS), the enzyme that metabolizes glutamate intoglutamine in astroglial cells plays a neuroprotective role [18].

There are further suggestions that neuronal injury observedin the brain of AIDS patients is related to excessive influx ofCa2+ associated with the neurotoxic injurious effect inducedby gp120. This mechanism of toxicity involves the activation

Article highlights

. Dementia is the most significant and devastating centralnervous system (CNS) complication associated withhuman immunodeficiency virus (HIV) infection.

. Currently, no specific treatment exists for CNS infectionsrelated to HIV. This is mainly attributed to the poorpenetrability of antiretroviral therapy (ART) across theblood--brain barrier (BBB), and the protective nature ofthe BBB making the brain tissue HIV sanctuary site.

. Various strategies were implemented for enhanced BBBpenetration, including nanotechnology, structuralmanipulation as well as the use of transport/carrier systems. Other strategies for drug delivery to thebrain involve bypassing the BBB such as intranasaladministration and direct invasive methods.

. Research efforts should focus on enhancing the portionof the nasal dose that reaches the CNS.

. Novel developments emerging in the field of polymerscience and nanotechnology provide an option by whichthe obstacles of limited brain entry can be surmounted.

. A better appreciation of the transporters present at thebrain barriers could prove a valuable milestone inunderstanding the limited brain penetration of anti-HIVdrugs in HIV. This also could aid the development ofnew anti-HIV drugs, new nanotechnology templates anddrug structural manipulations, with enhanced efficacy inthe CNS.

This box summarizes key points contained in the article.

A. M. Al-Ghananeem et al.

974 Expert Opin. Drug Deliv. (2013) 10(7)

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 3: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

of voltage-dependent Ca2+ channels and N-methyl-D-aspartate(NMDA) receptor-operated channels. These suggestionsoffer avenues for future pharmacological intervention withfocus on clinically tolerated calcium channel antagonists andNMDA antagonists as potential trials in humans with AIDSdementia in the near future [19,20]. Further pathogenesis inHIV-infected patients is explained in Table 1 [21-27].

2.2 Dementia and AIDS in HIVThe term dementia is used to describe a set of symptomsresulting from damages and disorders affecting the brain.The associated symptomatology can be due to the specificregions affected by a multitude of diseases known as neurode-generative diseases. The symptoms presented vary fromdecreasing cognitive functions to behavioral, affective, motorand psychiatric disorders. The terms ADC, and HIV-1 associ-ated dementia (HAD), are used to describe these neurologicaland psychiatric symptoms caused by human immunodefi-ciency virus type 1 (HIV-1) infection [22,25]. HIV-1 is themost common cause of dementia in young adults in theUSA [25,26].

2.3 Brain pathophysiological status and its impact

on anti-HIV drugsThe HIV virus commonly affects the basal ganglia, deep whitematter and cerebral cortex in HIV/AIDS dementia. Patholog-ical features include pervasive reactive astrocytosis, myelin pal-lor, activated resident microglia, infiltration by circulatingmonocytic cells, perivascular inflammation, microglial nod-ules, multinucleated giant cells, dendritic simplification andcell death with both astrocytic and neuronal damage [27].

Despite the current therapeutic management with activeART, neuronal cell death persists and has become a constantproblem affecting the CNS of HIV-infected patients.Although anti-HIV therapy has successfully prevented manyof the former end-stage complications of AIDS leading to

increased survival times, the HAD is still prevalent in treatedpatients as well as attenuated forms of CNS opportunisticdisorders. This suggests that there may be other indirectmechanisms of neuronal injury and loss/death occur inHIV/AIDS as a basis for the dementia which can still beexplored [13].

The anatomical--physiological protective mechanism of theBBB and the BCSFB has garnered interest due to the chal-lenges imposed in therapeutics and adverse outcomes ofdrug interactions. Faced with challenges in altered efficacyand enhanced neurotoxicity of drugs, clinicians are posedwith problems in drug delivery and treatment of diseaseslike AIDS dementia [28].

Recent and past clinical therapeutic trials for the treatmentof dementia have focused on some drugs as adjunct therapy tocurrent anti-HIV therapy. Several recent, large-scale trialsconducted through the AIDS Clinical Trials Group(ACTG) focus on three drugs: minocycline, memantine andselegiline as adjunct therapy to anti-HIV drugs (Table 2).Interestingly, data suggested neuroprotective properties ofminocycline that makes it an excellent candidate for limitingHIV replication within the brain, which has remained a diffi-cult anatomical compartment for therapeutic interventionsfor HIV due to the lack of BBB permeability of many suchdrugs [27].

The BBB and BCFSB are not merely passive barriers butalso play a dynamic role in the expression of influx and effluxtransporters and drug-metabolizing enzymes. Studies indicatethat efflux transporters such as P-glycoprotein (P-gp) play animportant role in drug delivery [29]. Furthermore, chemicalinhibition of transporters predicts lesser magnitude ofexpected drug--drug interaction in human BBB but may beclinically significant [29]. Defining the impact of such drug--drug interactions is important for improving efficacy of drugsused in the treatment of CNS diseases while minimizingneurotoxicity.

Table 1. Pathogenesis and neurodegeneration mechanisms in HIV infection.

Pathogenesis in HIV Levels in HIV infection Mechanism of neurodegeneration

Direct effect of HIV infection by proteins Increase HIV-1 gp120 Stimulate nitric oxide synthaseIncrease release of arachidonic acidInhibition of glutamate uptake by astrocytes and neurons

Increase HIV-1 Tat Disruption of the BBBInfiltration of inflammatory cells into the CNSIncreases cytokines and chemokines

HIV-1 Vpr Induce apoptosis of human neuronal-precursor cellsActivation of caspase-8

Indirect consequence of infectionthrough inflammation

Increase chemokineand cytokine

Leukoencephalopathy and neuronal apoptosisGlutamate-associated neurotoxicityTNF-a-associated damage to the BBB increasingentry of HIV proteins and cytokines

Increase neurotoxins Synaptic disruptionImpaired neurogenesis

BBB: Blood--brain barrier; CNS: Central nervous system; gp120: Glycoprotein 120; HIV: Human immunodeficiency; HIV-1: Human immunodeficiency virus type 1;

Tat: Transcriptional transactivator; Vpr: Viral protein R.

Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

Expert Opin. Drug Deliv. (2013) 10(7) 975

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 4: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

3. Delivering anti-HIV medication to brainbypassing the BBB

3.1 Intranasal drug deliveryDelivery of drugs into the brain via intranasal administrationhas gained increased attention in recent years. Intranasal deliv-ery is a non-invasive method that targets drugs to the brainand spinal cord along olfactory and trigeminal neural path-ways, bypassing the BBB and minimizing systemic exposureand side effects. Nasal dosage forms can be self-administeredby the patient and do not need a healthcare provider like par-enteral administration. Furthermore, the nasally applied drugis immediately absorbed through the nasal mucosal mem-branes, largely avoiding hepatic first-pass metabolism. More-over, a part of the absorbed drug is distributed directly tothe CNS as well as via blood flow.In addition to the systemic absorption of therapeutic agents

after intranasal administration, direct delivery of a wide varietyof therapeutic agents to the CNS following intranasal adminis-tration, as well as the therapeutic benefit of intranasal drug deliv-ery has been demonstrated by the authors’ group andothers [30-36]. The direct anatomical connection exists betweenthe nasal cavity and the CNS makes it possible to deliver sometherapeutic agents into the CNS by circumventing the BBB,which provides the basis for the development of therapeuticagents for intranasal administration. Potential mechanisms fornose-to-brain drug transport are thought to involve the trigemi-nal nerve receptors and/or one or a combination of two generalmechanisms namely: axonal transport through the olfactorynerve and absorption across the olfactory sustentacular epithelialcells [36]. Thus, the poor penetration of antiviral agents into theCNS may potentially be overcome by intranasal administration.The nasally administered gp160-HVJ-liposome to normal

mice has shown to be a good immunization tool that induces

necessary Ag-specific immune responses at different stages ofHIV [37]. Furthermore, brain tissue, CSF and olfactory bulbwere shown to have higher concentrations of intranasal didan-osine (2¢,3¢-dideoxyinosine, ddI) following nasal administra-tion when compared with intravenous administration. Theresults suggest that ddI can reach CNS compartments via adirect pathway initiating in the nasal cavity that does notinclude systemic circulation [38].

Intranasal administration of zidovudine (3¢-azido-3¢-deoxy-thymidine, AZT) in rats showed rapid absorption and highCSF concentrations of AZT. Furthermore, nasal delivery ofAZT co-administered with probenecid resulted in high CSFconcentration of AZT [39]. Accordingly, the nasal route wassuggested as a non-oral and non-parenteral dosage form choicefor AIDS treatment to patients with CNS dysfunctions.

The CSF and brain levels of AZT after intranasal adminis-tration of a thermo-reversible gel formulation were greaterthan those attained after intravenous injection. The pharma-cokinetic and brain distribution levels revealed that a polarantiviral compound such as AZT could preferentially transferinto the CSF and brain tissue via an alternative pathway,possibly olfactory route after intranasal administration [40].

Depending on the therapeutic agent physicochemical prop-erties, sometimes nasal administration of therapeutic agentsdoes not result in high levels in CNS. The extent of brain dis-tribution of stavudine after intranasal administration in ratswas comparable or sometimes less than that following sys-temic intravenous administration [41]. It seems that the directtransport pathway from the nasal cavity to brain did not con-tribute significantly to the overall brain exposure of stavudine.

In summary, although intranasal administration has beenused for brain delivery of some therapeutic agents, the effi-ciency of this route in human is still uncertain due to the smallolfactory mucosa area with respect to rodents or dogs. Note

Table 2. Potential effects of adjunct therapy on HIV pathology per studies reported from the ACTG [49].

Drug Drug class Mechanism of action Crosses

BBB

Advantages Disadvantages

Minocycline Anti-inflammatory:second-generationtetracycline

Lowers nitric oxide levelsInhibition of immune cellinfiltration and activationof microgliaInhibits HIV replication in vitro

Yes Inexpensive to produceReadily available inpharmaciesSafe for long-term treatment

Infections related tosustained suppressionof immune system dueto antibiotic effects(i.e., HIV periodontitis)

Memantine Anti-excitotoxicity:open channelNMDAR blocker

Decreases conductance ofcalcium throughuncompetitivebimolecular reactionwith the receptor

Yes Effectively limits abnormalBBB permeability andneurological deficitsConfirms that the neuronaldamage is mediated byNMDAR signaling

Unclear if successful forlong-term useRecent short-term clinicaltrial results aredisappointing

Selegiline Antioxidant:MAOIB

Low-dose trophic effecton neuronsReduce oxygenatedfree radicals

Yes Transdermal administrationof selegiline may avoidtoxicities

Disappointing results in arecent short-term clinicaltrial on transdermal

ACTG: AIDS Clinical Trials Group; BBB: Blood--brain barrier; HIV: Human immunodeficiency; MAOIB: Monoamine oxidase type B inhibitor;

NMDAR: N-methyl-D-aspartate.

A. M. Al-Ghananeem et al.

976 Expert Opin. Drug Deliv. (2013) 10(7)

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 5: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

that the olfactory area constitutes 50% of the nasal cavity inrat, but approximately 3% in human [36]. Thus, the transportmechanism to CNS through this pathway requires furtherclinical investigation to better understand the criteria definingtherapeutic agents’ properties needed for better absorptionand direct delivery to brain.

3.1.1 Surgical optionsInvasive surgical approaches for drug delivery are a means tobypass the BBB that carries risks for the patients withoutassurance of adequate drug distribution. This entails specialdelivery methods that involve direct introduction of the ther-apeutic substances into the brain or into the CSF such as neu-rosurgical procedures, implantation of cells or tissues or theuse of special devices [42-44].

Manipulation of catheters into the CSF compartment (lumbarsubarachnoid space, cisterns and ventricles) during angiographyallows direct injection of contrast media or therapeutic substan-ces into the CNS [45]. This enables a precise placement of electro-des or probes in the brain with minimal invasion achievingsuccessful drug distribution. Direct convection delivery can alsobe used to deliver an infusate and distribute macromolecules ina predictable, homogeneous manner over significant volumes [46].Interstitial delivery of drugs to the brain from biological tissues isa promising strategy to achieve the desired therapeutic agentthrough the implantation of genetically engineered cells thatsecretes the required drug [47-49]. Chimeric peptide technology,wherein a non-transportable drug is conjugated to a BBB trans-port vector holds as much promise for future success [50,51].Intraventricular and intrathecal route uses an Ommaya reser-voir implanted subcutaneously in the scalp and connected tothe ventricles of the brain. Anti-HIV drugs are introduced sub-cutaneously into the implanted reservoir, which subsequentlydelivers the administered drugs to the cerebral ventricles, bymanual compression of the reservoir through the scalp.A smaller dose may be used to offset any potential toxicity.Unfortunately, this delivery method has not been favored dueto its slow rate of drug distribution with the CSF and theincrease in intracranial pressure. Also, it carries high risks forhemorrhage, CSF leaks, infections and neurotoxicity [52-54].

4. Absorption of anti-HIV therapeutic agentsthrough BBB

4.1 Formulation approaches through nanotechnologyDue to their small size range, nanoparticle systems have beenproposed as potential brain targeting drug delivery sys-tems [55,56]. Recent advances in nanotechnology have providedpromising solutions to brain targeting challenge. Orallyadministered stavudine-loaded solid lipid nanoparticles wereeffectively targeted to cells of the reticuloendothelial system(RES) and brain. The solid lipid nanoparticles of stavudineshowed an 11-fold increase in stavudine brain uptake in com-parison with orally administered stavudine solution [57]. Fur-thermore, solid lipid nanoparticles were reported to enhance

the delivery of atazanavir by human brain endothelial cellline [58]. Nanoparticle drug delivery systems have beenincreasingly used to improve the efficiency of antiretroviraldrugs such as saquinavir [59-62]. Recent studies show that thespecificity and efficiency of antiretrovirals delivery can be fur-ther enhanced by using nanotechnology with specific braintargeting, cell penetrating ligands or ATP-binding cassette(ABC)-transporters inhibitors [63].

Several nanotechnology systems ranging from the moreestablished systems, for example, polymeric nanoparticles,solid lipid nanoparticles, liposomes, micelles to the newer sys-tems, for example, dendrimers, nanogels, nanoemulsions andnanosuspensions have been studied for the delivery of CNStherapeutics. Many of these nanomedicines can be effectivelytransported across various in vitro and in vivo BBB modelsby endocytosis and/or transcytosis, and demonstrated earlypreclinical success for the management of CNS conditionssuch as HIV encephalopathy [38,63-65].

Significant enhancement in ddI brain tissue concentrationwas observed when administering the drug nasally inchitosan-loaded nanoparticles in comparison with nasallyadministered ddI solution [38].

These nanotechnology studies suggest considerable oppor-tunities to enhance delivery of therapeutic agents to theCNS for HIV therapy.

4.2 Targeted delivery through structural

manipulationsIn order to overcome the bioavailability issue associated withantiretroviral agents in the CNS, a number of structural mod-ifications to nucleoside reverse transcriptase inhibitors (NRTIs)and protease inhibitors (PIs) have been created in order toimprove BBB penetration. The antiviral AZT is classified as aNRTI which was the first drug approved for the treatment ofAIDS. On drug administration, monitored levels of AZT inthe brain are low relative to the periphery [66]. This phenome-non is explained through a number of mechanisms; AZT hasbeen shown to cross the BBB by a carrier-mediated mechanismand a passive diffusion process [67,68]. Additionally, AZT is sus-ceptible to active efflux from the brain into the blood throughP-gp-mediated efflux [69-71]. AZT has undergone a number ofmodifications with the intent of increasing CNS penetration(Table 3). A high volume of promoieties have been introducedto 5¢-hydroxyl group connected by a labile ester linkage [72-78].Some notable promoieties introduced are retinoic acid whichalone has the capacity to inhibit HIV replication [73]. Unfortu-nately, after subsequent prodrug conversion an additive effectwith AZT was not observed but it contained the highestH9 cellular uptake in that series of prodrugs. In the same study,amino acid ester prodrugs were synthesized with purpose ofenhanced penetration into the CNS possibly through theL-amino acid transporter, the isoleucine prodrug displayedthe highest lipid partition coefficient and not surprisinglydisplayed higher H9 cell uptake relative to AZT. A glycosylphosphotriester prodrug showed improved brain delivery of

Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

Expert Opin. Drug Deliv. (2013) 10(7) 977

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 6: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

AZT, and is metabolized to a monophosphate metabolitebypassing the need for viral kinase activation [74]. Modificationsto the 5,6-olefin bond of AZT yielding a series of 5-halo-6-alk-oxy (or azido) derivatives which led to enhanced lipophilicityand a comparably higher distribution in the brain relative tothe parent compound [79,80]. Unfortunately, increasing lipophi-licity is not the only hurdle to overcome, there are a number ofmetabolic enzymes located at brain endothelial cells which havebeen referred to as an enzymatic or ‘metabolic’ BBB [81]. Aden-osine deaminase (ADA) and purine nucleoside phosphorylase(PNP) are two significant enzymes located in this region andare involved in purine metabolism. One notable prodrug deriv-ative to ddI, 2¢-b-fluoro-2¢,3¢-dideoxyadenosine (F-ddA) is anacid stable analog of 2¢,3¢-didesoxyadenosine (ddA), which isdeaminated by ADA to the active 2¢-b-fluoro-2¢,3¢-dideoxyino-sine (F-ddI) [82,83]. In addition to improve lipophilicity,F-ddA exploits the enzymatic BBB resulting in an eightfoldhigher brain/plasma ratio compared with equivalent F-ddIinfusion [84].In general, prodrug strategies involve the introduction of a

lipophilic moiety connected by a labile linkage in order toenhance diffusion across biological membranes. Chemicalmodifications of antiretroviral agents with the intent ofenhanced uptake via protein transporters at the BBB areanother viable strategy [85-96].A class of guanidine-rich molecular transporters has been

synthesized that possess a high molecular weight, good watersolubility and can penetrate the mouse brain in vivo [85].More specifically, an eight guanidine (G8) molecular trans-porter was shown to be the most effective at penetrating theBBB. Consequently, a covalently linked AZT with sorbitolG8 (Table 3) was synthesized [86]. Tissue distribution studiesin mice indicated the modified AZT possesses an affinitytoward both the brain and liver.Platelet-activating factor (PAF) has been identified as an

HIV-induced neurotoxin [87,88]. One possible route of neuro-toxicity is through a PAF-activated increase in Ca2+ leading toenhanced excitatory neurotransmission through glutamaterelease [89]. These findings prompted the investigation ofPAF antagonists for potential antiviral activity resulting in atrisubstituted piperazine, PMS-601 possessing both anti-HIV-1 effects as well PAF antagonist activities in the micro-molar range in vitro [90]. Additionally, it was shown to havethe capacity to cross the BBB in rat models using the in situbrain perfusion method, further SAR studies were completedresulting in analogs with enhanced dual activities [91,92]. Thesestudies display the potential to both penetrate the viral poolsin the brain as well as attenuate neurotoxicity.Attempts have also been made to utilize the body’s own

nutrient carrier transport through conjugation of antiviralswith amino acids or D-glucose [93-95]. Amino acid conjugatessuch as L-valyl, L-phenylalanyl and L-leucyl esters of indinavirand saquinavir showed an enhanced transport across theCaco-2 cell monolayer, however D-glucose conjugates showeda decrease in absorption [96]. Further investigations of the

valine derivative of indinavir showed no brain distributionand decreased oral bioavailability relative to indinavir [97].

4.3 Altering BBB passage pathwaysABC transporters such as P-gp and multidrug resistance-associated proteins (MRP) are efflux membrane transporterswith a broad capacity of xenobiotic substrates. They are foundin various tissues of the body including the gastrointestinal tract,liver and kidney [98]. P-gp is expressed at high levels in the braincapillaries and is the primary cause for efflux of antiretrovirals outof the CNS [99,100]. Thus, inhibition of P-gp has advantageousclinical implications in the treatment of NeuroAIDS. Com-monly, PIs (a P-gp substrate) are co-administered with a lowdose of ritonavir which functions to block both P-gp as well ascytochrome P450 metabolizing enzymes [101]. In fact, ritonavirdisplays a higher potency of inhibition of P-gp relative tomultidrug resistance reversing agent SDZ PSC833 [102].

Probenecid is a uricosuric agent and a known inhibitor ofthe organic anion acid transporter in the renal tubule. Notonly does it act in the kidney but also has been demonstratedto block transport of organic acids from the CNS to plasmasuch as penicillin, pantothenic acid and leukotrieneA4 [103-105]. Probenecid administered in combination withAZT shows an enhanced distribution in the brain, presum-ably through a reduced rate of drug efflux [106,107]. Combina-tions of ddI and probenecid also displayed an enhancement inCNS uptake [108]. Looking at the efflux rate of both AZT andddI after microinjection into the brain, demonstrated AZTefflux from the brain was inhibited by ddI and probenecid.Additionally, the efflux rate of ddI was inhibited by AZTand probenecid [71]. This provides supporting evidence thatAZT and ddI are transported from the brain to blood acrossthe BBB via a probenecid-sensitive efflux transporter. Amphi-philic block copolymer (Pluronic P85) has been under investi-gation for the inhibition of efflux transporters. P85 consists oftwo chains of hydrophilic polyethylene oxide groups surround-ing a hydrophobic polypropylene oxide moiety. Initially beingused in cancer resistance studies, P85 was also shown to inhibitP-gp-mediated drug efflux in bovine brain microvessel endo-thelial cells providing a possible route to improve CNS drugpenetration [109]. When used in combination with antiretroviraldrugs, P85 increased the CNS biodistribution of the AZT/lam-ivudine/nelfinavir cocktail in a mouse model of HIV encepha-litis (HIVE) [110]. The immunodeficient mice were inoculatedwith HIV-infected human monocyte-derived macrophages(MDM) into the basal ganglia and then treated with P85,ART or P85 and ART. A significant decrease in the HIV-infected MDM in all treatment groups relative to the controlwas observed, it is important to note that P85 alone containedantiretroviral effects [110]. Thus, P85 was hypothesized to havetwo mechanisms of P-gp inhibition: decreasing ATP levels inbovine brain endothelial cell monolayers and can compromisethe lipid bilayer causing a destabilization of P-gp. This copoly-mer also effectively inhibits the interaction between saquinavirand nelfinavir in cellular accumulation studies, Pluronic

A. M. Al-Ghananeem et al.

978 Expert Opin. Drug Deliv. (2013) 10(7)

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 7: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

Table 3. A summary of anti-HIV drugs chemical modifications to improve their CNS bioavailability.

Parent drug Derivatives Therapeutic effect

AZT

O

N3

RO

N

NH

O

O5′-OH Promoieties

Retinoic acidCH3 CH3 CH3

CO2R

CH3CH3

Highest H9 cell uptake in thisseries, however a sixfold increasein cytotoxicity observed relative toAZT. Retinoic acid has been shownto have antiviral activity [66]

Isoleucine

H3C

NH2

OR

OCH3

Isoleucine displayed the highestH9 cell uptake relative to otheramino acid promoieties inthis series [66]

G8 molecular transporter

OR2

R2O

OR2

OR2

O

O

NH25

O

R2 =

O

N5

NH

NH2

HN

HN NH2

NH

O

OR

O

Covalent linkage of a sorbitol-G8transporter displayed enhancedwater solubility and improvedAZT BBB penetration in mousebrain [78]

Glycosylphosphotriester

P ORO

O

O

O

O

HO

OH

OHOHOHOH

Displayed high concentrations ofAZT in vitro via glucosyl dicholphosphate transport, enhancedwater solubility, bypasses viralkinase activation [67]

Modified olefin of AZT

O

N3

HO

N

NH

O

O

RA

RB

RA = Br, Cl, IRB = OMe, OEt, O(CH2)xCH3, N3

Addition to the olefin yielding aclass of 5-halo-6-alkoxy (or azido)AZT lead to enhanced logPand CNS biodistribution,5-bromo/5-iodo analogs mayserve as prodrugs due toregeneration of the olefin [73]

ddI

N

NHN

N

O

OHO

F-ddA

N

O

HN

OH2N

HN O

OR

O

NH

Addition of the 2¢-b-fluoroenhances acid stability, while6-amination yields a prodrug thatundergoes conversion by ADA toF-ddI leading to an eightfoldhigher brain/plasma ratio relativeto F-ddI infusion [75-77]

ADA: Adenosine deaminase; AZT: Zidovudine; BBB: Blood--brain barrier; CNS: Central nervous system; ddI: Didanosine; F-ddI: 2¢-b-fluoro-2¢,3¢-dideoxyinosine;F-ddA: 2¢-b-fluoro-2¢,3¢-dideoxyadenosine; HIV: Human immunodeficiency; P-gp: P-glycoprotein.

Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

Expert Opin. Drug Deliv. (2013) 10(7) 979

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 8: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

F88 and F127 (more hydrophilic copolymers) were also testedbut failed to be active [111].Recently, dimers of abacavir (Table 3) were engineered with a

disulfide linker that was shown to be a potent P-gp inhibi-tor [112]. The prodrug serves a unique dual role of both P-gpinhibition and antiretroviral properties on metabolism. Underthe reducing conditions of the cytosol, the disulfide linker iscleaved ultimately releasing the active monomer. The derivativeAbaS2Me4 displayed the highest activity and a 700-foldincrease in potency over abacavir. Introduction of methyls adja-cent to the carbonyl most likely leads to a reduced metabolismby plasma esterase contributing to the enhanced potency. Thismethod paves the way for varied ART combination in a similardimer which can lead to enhanced CNS penetration, decreaseddosing requirements and a possible lowering of the pill burdenof patients fighting HIV. Targeting P-gp is a viable strategythat shows promise in increasing the brain concentration ofantiretroviral mediations. In fact, the initial thought of in-creasing the drug lipophilicity to enhance drug concentrationsin the brain is possibly counterintuitive. P-gp has beendescribed as a ‘hydrophobic vacuum cleaner’ resulting inincreased removal of lipophilic drugs from the CNS, thusP-gp inhibition combination has clinical significance [112].

5. Conclusion

The restricted entry of anti-HIV drugs to the CNS due to thepresence of BBB hinders the penetration of anti-HIV drugs

into the brain, promoting viral replication, the developmentof drug resistance and ultimately sub-therapeutic concen-trations of drugs reaching the brain, leading to therapeuticfailure. A better appreciation of the transporters present atthe brain barriers could prove a valuable milestone in under-standing the limited brain penetration of anti-HIV drugs inHIV. This also could aid the development of new anti-HIV drugs, new nanotechnology templates and drug structuralmanipulations, with enhanced efficacy in the CNS.

Furthermore, a better understanding of methods thatdeliver anti-HIV drugs to CNS bypassing the BBB is also war-ranted for better control of neurological complications associ-ated with HIV.

6. Expert opinion

The treatment of HIV/AIDS dementia continues to be chal-lenging as we pursue ways to achieve the delivery of optimallevels of drug therapy in the brain for the cure of this debilitat-ing disease. Of further importance is the possibility that theCNS may serve as a sanctuary for the virus after its systemiceradication. The major challenge to CNS drug delivery is theBBB, which limits the access of drugs to the brain substance.Currently approved drugs for the treatment of HIV infectionsmay not attain sufficient drug concentrations in the CNS, andthus may not completely suppress viral replication in the brainover a sustained period of time.

During the last years, considerable efforts were focused onthe field of brain-targeted drug delivery. The ultimate goals

Table 3. A summary of anti-HIV drugs chemical modifications to improve their CNS bioavailability (continued).

Parent drug Derivatives Therapeutic effect

Saquinavir

N

N

N

N

HN

H2N

OH

R = ester/carbamate linked glucoseR = ValR = PheR = Leu

These amino acid prodrugsdisplayed enhanced transportacross Caco-2 monolayer. Furtherstudies of the valine analogdisplayed no brain distributionin vivo. D-Glucose analogsdisplayed a decrease in absorption,additionally ester linkedpromoieties displayed enhancedanti-HIV activity relative to thecarbamate [86-90]

Abacavir

N

NN

N

H2N

OF

HO

Abacavir dimer

N

N

N

N

HN

NH2

O

O

S

N

N

N

N

HN

H2N

O

O

S

Dimeric prodrugs of abacavirdisplay potential for P-gp inhibitionat the BBB and revert to the activemonomer. This presents a platformthat could have application intherapies involved with limitationsof P-gp drug efflux [105]

ADA: Adenosine deaminase; AZT: Zidovudine; BBB: Blood--brain barrier; CNS: Central nervous system; ddI: Didanosine; F-ddI: 2¢-b-fluoro-2¢,3¢-dideoxyinosine;F-ddA: 2¢-b-fluoro-2¢,3¢-dideoxyadenosine; HIV: Human immunodeficiency; P-gp: P-glycoprotein.

A. M. Al-Ghananeem et al.

980 Expert Opin. Drug Deliv. (2013) 10(7)

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 9: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

of those research efforts were mainly to deliver the anti-HIVtherapeutic agents in therapeutically effective concentrationsto the brain. Various strategies were implemented forenhanced BBB penetration, including nanotechnology, struc-tural manipulation as well as the use of transport/carrier sys-tems. Other strategies for drug delivery to the brain involvebypassing the BBB such as intranasal administration anddirect invasive methods. Both strategies have advantages anddisadvantages (Table 4). Although, each strategy may offermany intriguing possibilities for brain delivery and targeting,but only some have reached the phase where they can providesafe and effective human applications.

It is important to consider not only the net delivery of theagent to the CNS, but also the ability of the agent to accessthe relevant target site within the CNS. Site-target and theuse of targeting enhancement factors can be used to quantita-tively assess the effectiveness from a pharmacokinetic perspec-tive of chemical delivery systems. However, from drugdevelopment and regulatory aspects, pharmaceutical compa-nies usually tend toward designing improved formulationsof well-known therapeutic agents that are already in the mar-ket, rather than synthesizing a new chemical entity. Note thatany slight structural manipulation, even simple ester-pro-drugs, will make the therapeutic agent considered as a newchemical entity. The latter would require an extensive regula-tory efforts through new drug application (NDA) necessitat-ing a full safety and efficacy assessment from preclinical andclinical prospective.

A fruitful area of future research will be to determine ifthere are any systems that can target drug delivery to the brain.Novel developments emerging in the field of polymer scienceand nanotechnology provide an option by which the obstaclesof limited brain entry can be surmounted. Thus, the use ofnanotechnology based on Food and Drug Administration(FDA)-approved polymeric carriers would be an attractive

approach to deliver known anti-HIV drugs to the CNS. Theplethora of transporters expressed at the brain barriers all actas selective gatekeepers, and this remains a major obstacle forantiviral therapy. Future directions in nanotechnology as atool to enhance anti-HIV drugs bioavailability in CNS shouldconsider nanocarriers with ligands specific for certain BBBtransports to overcome this obstacle. In the authors’ opinion,this is a milestone enhancement in delivering therapeuticagents including anti-HIV drugs to CNS. Future directionshould focus on two main directions in utilizing nanotechnol-ogy for brain targeting: i) allowing drug trafficking by endocy-tosis (non-specific or receptor-mediated) and ii) blockingdrug efflux transporters. Thus, a better appreciation of BBBtransporters is warranted for successful drug delivery.

Furthermore, applying anti-HIV therapeutic agents encap-sulated into nanoparticles could potentially improve the directCNS delivery via the nasal cavity. Intranasal drug deliveryholds a promising future in brain targeting. However, a chal-lenge of the intranasal administration to target the brain, thatneeds to be in mind during drug development effort, is thatthe quantities of drug administered nasally that have beenshown to be transported directly from nose-to-brain are rela-tively low. Research efforts should focus in enhancing the por-tion of the nasal dose that reaches the CNS. If drugs couldreach the CNS in sufficient quantity by this route, it couldgenerate interest in previously abandoned drug compoundsand enable an entirely novel approach to CNS drug delivery.

Currently, there is no approved nanotechnology-based CNSdrug available; the future for such nanotechnology-baseddelivery system developments is promising. A thorough under-standing of BBB transporter, its location, level of expressionand transport potency is needed to help improve the treatmentof HIV infection in the brain.

Interestingly, there are many nanotechnology anti-HIVplatforms tested but most are still in preclinical research

Table 4. Main advantages and disadvantages of the two drug delivery strategies (absorption through BBB or

bypassing the BBB) used for anti-HIV drug delivery to CNS.

Method Advantage Disadvantage

Bypassing the BBB Bypass hepatic first-pass metabolismReduction of systemic side effectsRapid absorption and onset of action

Devices needed for drug delivery

Surgical options Targeted drug delivery andCNS abundanceControlled and sustained drug delivery

InvasiveSurgical needsHigh risk small site for absorption

Intranasal drug delivery Ease of administrationNon-invasive

Mucociliary clearancePotential nasal irritationCNS drug bioavailability depends on drugabsorption from nasal cavityDose size limitation

Absorption through BBB Ease of administrationPotential technology enhancement withspecific receptors ligands

Systemic exposureLow CNS bioavailabilityOnset of action may be delayedPotential side effects

BBB: Blood--brain barrier; CNS: Central nervous system; HIV: Human immunodeficiency.

Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

Expert Opin. Drug Deliv. (2013) 10(7) 981

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 10: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

with one immunotherapy-based product making it to clinicaltrials. However, the authors expect to see in the futurenanotechnology-based anti-HIV marketed products, as theFDA expressed interest in such technology translated in open-ing multi-nanotechnology research centers. Research done inthis field holds up hope, despite the BBB shielding challenges.Future research should focus on achieving brain delivery ofanti-HIV therapeutic agents in a safe, efficient and yet cost-effective manner.

Acknowledgment

The authors would like to thank Amber Jeffries for heradministrative assistance in this review article.

Declaration of interest

The authors state no conflict of interest and have received nopayment in preparation of this manuscript.

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Pang S, Koyangi Y, Miles S, et al. High

levels of unintegrated HIV-1 DNA in

brain tissue of AIDS dementia patients.

Nature 1990;34:85-9

2. Koppel BS, Wormser GP, Tuchman AJ,

et al. Central nervous system involvement

in patients with acquired immune

deficiency syndrome (AIDS).

Acta Neurol Scand 1985;71:337-53

3. Carne CA; ABC of AIDS. Neurological

manifestations. Br Med J

1987;294:1399-401

4. Anderson BD, Hoesterey BL, Baker DC,

Galinsky RE. Uptake kinetics of

2,3-dideoxyinosine into brain and

cerebrospinal fluid of rats: intravenous

infusion studies. J Pharm Exp Therap

1989;253:113-18

5. Minagar A, Commins D, Alexander JS,

et al. NeuroAIDS: characteristics and

diagnosis of the neurological

complications of Aids. Mol Diagn Ther

2008;12:25-43

6. Gray F, Adle-Biassette H, Chretien F,

et al. Neuropathology and

neurodegeneration in human

immunodeficiency virus infection.

Pathogenesis of HIV-induced lesions of

the brain, correlations with

HIV-associated disorders and

modifications according to treatments.

Clin Neuropathol 2001;20:146-55

. An important reference detailing the

pathogenesis of HIV harbored in

brain tissue.

7. Spencer DC, Price RW. Human

immunodeficiency virus and the central

nervous system. Annu Rev Microbiol

1992;46:655-93

. A high impact report on HIV-related

neurodegenerative disorders.

8. Lipton SA, Gendelman HE. Dementia

associated with the acquired

immunodeficiency syndrome. N Engl

J Med 1995;332:934-40

9. Lesniak MS, Brem H. Targeted therapy

for brain tumors. Nat Rev Drug Discov

2004;3:499-508

10. Nowacek A, Gendelman HE. NanoART,

neuroAIDS and CNS drug delivery.

Nanomed 2009;4:557-74

11. Begley DJ. The blood--brain barrier:

principles for targeting peptides and

drugs to the central nervous system.

J Pharm Pharmacol 1996;48:136-46

12. Frey WH. Bypassing the blood-brain

barrier to deliver therapeutic agents to

the brain and spinal cord.

Drug Deliv Technol 2002;2:46-9

.. An important article detailing

bypassing the BBB technologies to

enhance therapeutic agents

brain bioavailability.

13. Levy JA. Pathogenesis of human

immunodeficiency virus infection.

Microbiol Rev 1993;57(1):183-289

14. Rao KS, Ghorpade A, Labhasetwar V.

Targeting anti-HIV drugs to the CNS.

Expert Opin Drug Deliv

2009;6(8):771-84

15. Skalka AM, Katz RA. Retroviral

DNA integration and the DNA damage

response. Cell Death Differ

2005;12(1):971-8

16. Engelman A. The roles of cellular factors

in retroviral integration. Curr Top

Microbiol Immunol 2003;281:209-38

17. Lipton SA. HIV related neurotoxicity.

Brain Pathol 1991;1(3):193-9

18. Visalli V, Muscoli C, Sacco I, et al.

N-Acetylcysteine prevents HIV gp

120-related damage of human cultured

astrocytes: correlation with glutamine

synthase dysfunction. Neuro Sci

2007;8(1):106

19. Dreyer EB, Kaiser PK, Offermann JT,

et al. HIV-1 coat protein neurotoxicity

prevented by calcium channel

antagonists. Science

1990;248(4953):364-7

20. Lipton SA. AIDS-related dementia and

calcium homeostasis. Ann NY Acad Sci

1994;747:205-24

21. Reger M, Welsh R, Razani J, et al.

A meta-analysis of the neuropsychological

sequelae of HIV infection. J Int

Neuropsychol Soc 2002;8(3):410-24

22. McArthur JC, Sacktor N, Selnes O.

Review human immunodeficiency

virus-associated dementia. Semin Neurol

1999;19(2):129-50

23. Kramer-Hammerle S, Rothenaigner I,

Wolff H, et al. Review cells of the

central nervous system as targets and

reservoirs of the human

immunodeficiency virus. Virus Res

2005;111(2):194-213

24. Song L, Nath A, Geiger JD, et al.

Human immunodeficiency virus

type 1 Tat protein directly activates

neuronal N-methyl-D-aspartate receptors

at an allosteric zinc-sensitive site.

J Neurovirol 2003;9(3):399-403

25. Andras IE, Pu H, Deli MA, et al.

HIV-1 Tat protein alters tight junction

protein expression and distribution in

cultured brain endothelial cells.

J Neurosci Res 2003;74(2):255-65

26. Li W, Galey D, Mattson MP, Nath A.

Molecular and cellular mechanisms of

neuronal cell death in HIV dementia.

Neurotox Res 2005;8:119-34

27. Lindl K, Marks D, Kolson D,

Jordan-Sciutton K. HIV-Associated

neurocognitive disorder: pathogenesis and

therapeutic opportunities.

J Neuroimmune Pharmacol

2010;5(3):294-309

.. An important report on better

understanding of HIV brain

A. M. Al-Ghananeem et al.

982 Expert Opin. Drug Deliv. (2013) 10(7)

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 11: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

pathophysiology and

treatment options.

28. Palmer A. The role of the blood--CNS

barrier in CNS disorders and their

treatment. Neurobiol Dis 2010;37:13-12

29. Eval S, Hsiao P, Unadkat JD. Drug

interactions at the blood brain--barrier:

fact or fantasy? Pharmacol Ther

2009;123(1):80-104

30. Romeo VD, deMeireles J, Sileno AP,

et al. Effects of physicochemical

properties and other factors on systemic

nasal drug delivery. Adv Drug Deliv Rev

1998;29:89-116

31. Chien YW, Chang SF. Intranasal drug

delivery for systemic medications.

Crit Rev Ther Drug Carrier Syst

1987;4(2):67-194

32. Charlton ST, Davis SS, Illum L.

Evaluation of effect of ephedrine on the

transport of drugs from the nasal cavity

to the systemic circulation and the

central nervous system. J Drug Target

2007;15(5):370-7

. An important study on CNS drug

delivery from nasal cavity and factors

that could affect drug

brain bioavailability.

33. Al-Ghananeem AM, Malkawi AH,

Crooks PA. Bioavailability of

D9-tetrahydrocannabinol followingintranasal administration of a

mucoadhesive gel spray delivery system

in conscious rabbits. Drug Dev

Ind Pharm 2011;37(3):329-34

34. Hussain AA. Intranasal drug delivery.

Adv Drug Deliv Rev 1998;29:39-49

35. Al-Ghananeem AM, Traboulsi AA,

Dittert LW, Hussain AA. Targeted brain

delivery of 17b-estradiol via nasally

administered water soluble prodrugs.

AAPS PharmSciTech 2002;3:1

36. Illum L. Is nose-to-brain transport of

drugs in man a reality.

J Pharm Pharmacol 2003;56:3-17

.. An important review article detailing

the mechanistic aspects of

nose-to-brain delivery.

37. Sakaue G, Hiroi T, Nakagawa Y, et al.

HIV mucosal vaccine: nasal

immunization with gp160-encapsulated

hemagglutinating virus of

Japan-Liposome Induces antigen-specific

CTLs and neutralizing antibody

responses. J Immunol 2003;170:495-502

38. Al-Ghananeem AM, Saeed H,

Florence R, et al. Intranasal drug delivery

of didanosine-loaded chitosan

nanoparticles for brain targeting; an

attractive route against infections caused

by AIDS viruses. J Drug Target

2010;18(5):381-8

.. An important study showing enhanced

CNS and CSF uptake of the anti-HIV

drug ddI after intranasal

administration in rat and potential

enhancement effect of nanotechnology

in nasal drug delivery.

39. Seki T, Sato N, Hasegawa T, et al. Nasal

absorption of zidovudine and its

transport to cerebrospinal fluid in rats.

Biol Pharm Bull 1994;17(8):1135-7

40. Ved PM, Kim K. Poly(ethylene oxide/

propylene oxide) copolymer

thermo-reversible gelling system for the

enhancement of intranasal zidovudine

delivery to the brain. Int J Pharm

2011;411(1-2):1-9

41. Yang Z, Huang Y, Gan G, Sawchuk RJ.

Microdialysis evaluation of the brain

distribution of stavudine following

intranasal and intravenous administration

to rats. J Pharm Sci 2005;94(7):1577-88

42. Misral A, Ganesh S, Shahiwala A,

Shrenik P. Drug delivery to the central

nervous system. J Pharm Sci

2003;6(2):252-73

43. Pardridge WM. Recent advances in

blood brain-barrier transport. Annu Rev

Pharmacol Toxicol 1988;28:25-39

44. Taylor EM. The impact of efflux

transporters in the brain on the

development of drugs for CNS disorders.

Clin Pharmacokinet 2002;41:81-92

45. Joshi S, Ergin A, Wang M, et al.

Inconsistent blood brain barrier

disruption by intraarterial mannitol in

rabbits: implications for chemotherapy.

J Neurooncol 2011;104(1):11-19

46. Ding D, Kanaly CW, Bigner DD, et al.

Convection-enhanced delivery of free

gadolinium with the recombinant

immunotoxin MR1-1. J Neurooncol

2010;98(1):1-7

47. Brem H, Langer R. Polymer based drug

delivery to the brain. Sci Med

1996;3(4):1-11

48. Brem H, Gabikian P. Biodegradable

polymer implants to treat brain tumors.

J Control Release 2001;74:63-7

49. Fung LK, Ewend MG, Sills A, et al.

Pharmacokinetics of interstitial delivery

of carmustine,

4-hydroperoxycyclophosphamide and

paclitaxel from a biodegradable polymer

implant in the monkey brain. Cancer Res

1998;58(4):672-84

50. Golden PL, Maccagnan TJ,

Pardridge WM. Human blood--brain

barrier leptin receptor: binding and

endocytosis in isolated human brain

microvessels. J Clin Invest 1997;99:14-18

51. Tamai I, Sai Y, Kobayashi H, et al.

Structure-internalization relationship for

adsorptive-mediated endocytosis of basic

peptides at the blood--brain barrier.

J Pharmacol Exp Ther 1997;280:410-15

52. Harbaugh RE, Saunders RL, Reeder RF.

Use of implantable pumps for central

nervous system drug infusions to treat

neurological disease. Neurosurgery

1988;23(6):693-8

53. Blasberg RG, Patlak C,

Fenstermacher JD. Intrathecal

chemotherapy: brain tissue profiles after

ventriculocisternal perfusion. J Pharmacol

Exp Ther 1975;195:73-83

54. Huang TY, Arita N, Hayakawa T,

Ushio Y. ACNU, MTX and 5-FU

penetration of rat brain tissue and

tumors. J Neurooncol 1999;45:9-17

55. Baratchi S, Kanwar Rupinder K,

Khoshmanesh K, et al. Promises of

nanotechnology for drug delivery to

brain in neurodegenerative diseases.

Curr Nano Sci 2009;5:15-25

56. Kreuter J. Nanoparticulate systems for

brain delivery of drugs. Adv Drug

Deliv Rev 2001;47(1):65-81

57. Dandagi P, Patel P, Gadad A,

Aravapalli AK. RES and brain targeting

stavudine-loaded solid lipid nanoparticles

for AIDS therapy. Asian J Pharm

2012;6(2):116-23

58. Chattopadhyay N, Zastre J, Wong HL,

et al. Solid lipid nanoparticles enhance

the delivery of the HIV protease

inhibitor, atazanavir, by a human brain

endothelial cell line. Pharm Res

2008;25:2262-71

59. Bender AR, Von Briesen H, Kreuter J,

et al. Efficiency of nanoparticles as a

carrier system for antiviral agents in

human immunodeficiency virus-infected

human monocytes/macrophages in vitro.

Antimicrob Agents Chemother

1996;40(6):1467-71

60. Vyas TK, Shah L, Amiji MM.

Nanoparticulate drug carriers for delivery

of HIV/AIDS therapy to viral reservoir

Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

Expert Opin. Drug Deliv. (2013) 10(7) 983

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 12: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

sites. Expert Opin Drug Deliv

2006;3(5):613-28

61. Kinman L, Bui T, Larsen K, et al.

Optimization of lipid-indinavir

complexes for localization in lymphoid

tissues of HIV-infected macaques.

J Acquired Immune Defic Syndr

2006;42(2):155-61

62. Dou H, Morehead J, Destache CJ, et al.

Laboratory investigations for the

morphologic, pharmacokinetic, and

anti-retroviral properties of indinavir

nanoparticles in human

monocyte-derived macrophages. Virology

2007;358(1):148-58

63. Wong HL, Chattopadhyay N, Wu XY,

Bendayan R. Nanotechnology

applications for improved delivery of

antiretroviral drugs to the brain.

Adv Drug Deliv Rev

2010;62(4-5):503-17

. A focused review on the role of

nanotechnology in antiretroviral drugs

brain targeting and its

potential applications.

64. Saiyed ZM, Gandhi NH, Nair MP.

Magnetic nanoformulation of

azidothymidine 5’-triphosphate for

targeted delivery across the blood-brain

barrier. Int J Nanomedicine

2010;5:157-66

65. Saiyed ZM, Gandhi NH, Nair MP. AZT

5’-triphosphate nanoformulation

suppresses human immunodeficiency

virus type 1 replication in peripheral

blood mononuclear cells. J Neurovirol

2009;15(4):343-7

66. Wu D, Clement JG, Pardridge WM.

Low blood--brain barrier permeability to

azidothymidine (AZT), 3TC�, and

thymidine in the rat. Brain Res

1998;791(1--2):313-16

67. Thomas nee Williams SA, Segal MB.

Identification of a saturable uptake

system for deoxyribonucleosides at the

blood-brain and blood-cerebrospinal fluid

barriers. Brain Res 1996;741(1--2):230-9

68. Thomas SA, Segal MB. The passage of

azidodeoxythymidine into and within the

central nervous system: does it follow the

parent compound, thymidine?

J Pharmacol Exp Ther

1997;281(3):1211-18

69. Dykstra KH, Arya A, Arriola DM, et al.

Microdialysis study of zidovudine (AZT)

transport in rat-brain. J Pharmacol

Exp Ther 1993;267(3):1227-36

70. Masereeuw R, Jaehde U,

Langemeijer MWE, et al. In-vitro and

in-vivo transport of zidovudine (AZT)

across the blood-brain-barrier and the

effect of transport inhibitors. Pharm Res

1994;11(2):324-30

71. Takasawa K, Terasaki T, Suzuki H, et al.

In vivo evidence for carrier-mediated

efflux transport of 3’-azido-3’-

deoxythymidine and 2’,3’-dideoxyinosine

across the blood-brain barrier via a

probenecid-sensitive transport system.

J Pharmacol Exp Ther

1997;281(1):369-75

72. Kawaguchi T, Ishikawa K, Seki T, et al.

Ester prodrugs of zidovudine.

J Pharm Sci 1990;79(6):531-3

73. Aggarwal SK, Gogu SR, Rangan SRS,

et al. Synthesis and biological evaluation

of prodrugs of zidovudine. J Med Chem

1990;33(5):1505-10

74. Namane A, Gouyette C, Fillion MP,

et al. Improved brain delivery of AZT

using a glycosyl phosphotriester

prodrug. J Med Chem

1992;35(16):3039-44

75. McGuigan C, Pathirana RN, Balzarini J,

et al. Intracellular delivery of bioactive

AZT nucleotides by aryl phosphate

derivatives of AZT. J Med Chem

1993;36(8):1048-52

76. Hostetler KY, Stuhmiller LM,

Lenting HBM, et al. Synthesis and

antiretroviral activity of phospholipid

analogs of azidothymidine and other

antiviral nucleosides. J Biol Chem

1990;265(11):6112-17

77. Chu CK, Bhadti VS, Doshi KJ, et al.

Brain targeting of anti-HIV nucleosides -

synthesis and in vitro and in vivo studies

of dihydropyridine derivatives of 3’-

azido-2’,3’-dideoxyuridine and 3’-azido-

3’-deoxythymidine. J Med Chem

1990;33(8):2188-92

78. Piantadosi C, Marasco CJ,

Morrisnatschke SL, et al. Synthesis and

evaluation of novel ether lipid nucleoside

conjugates for anti-HIV-1 activity.

J Med Chem 1991;34(4):1408-14

79. Wang L, Morin KW, Kumar R, et al. In

vivo biodistribution, pharmacokinetic

parameters, and brain uptake of

5-halo-6-methoxy (or ethoxy)-5,6-

dihydro-3’-azido-3’-deoxythymidine

diastereomers as potential prodrugs

of 3’-azido-3’-deoxythymidine.

J Med Chem 1996;39(4):826-33

80. Kumar R, Wang LL, Wiebe LI, et al.

Synthesis, in-vitro biological stability,

and anti-HIV activity of 6-halo-6-alkoxy

(or azido)-5,6-dihydro-3’-azido-3’-

deoxythymidine diastereomers as

potential prodrugs to 3’-azido-3’-

deoxythymidine (AZT). J Med Chem

1994;37(25):4297-306

81. Johnson MD, Anderson BD.

Localization of purine metabolizing

enzymes in bovine brain microvessel

endothelial cells: an enzymatic

blood-brain barrier for

dideoxynucleosides? Pharm Res

1996;13(12):1881

82. Masood R, Ahluwalia GS, Cooney DA,

et al. 2’-fluoro-

2’,3’-dideoxyarabinosyladenine - a

metabolically stable analog of the

antiretroviral agent 2’,3’-

dideoxyadenosine. Mol Pharmacol

1990;37(4):590-6

83. Marquez VE, Tseng CKH, Mitsuya H,

et al. Acid-stable 2’-fluoro purine

dideoxynucleosides as active agents

against HIV. J Med Chem

1990;33(3):978-85

84. Johnson MD, Chen J, Anderson BD.

Investigation of the mechanism of

enhancement of central nervous system

delivery of 2’,-beta-fluoro-2’,3’-

dideoxyinosine via a blood-brain barrier

adenosine deaminase-activated prodrug.

Drug Metab Dispos 2002;30(2):191-8

85. Maiti KK, Jeon OY, Lee WS, et al.

Design, synthesis, and

membrane-translocation studies of

inositol-based transporters. Angew Chem

Int Ed Engl 2006;45(18):2907-12

. A novel approach by which AZT is

covalently linked to a

sorbitol-G8 transporter which displays

the ability to cross the BBB in the

mouse brain.

86. Im J, Kim W, Kim KT, et al.

Preparation of a 3’-azido-3’-

deoxythymidine (AZT) derivative, which

is blood-brain barrier permeable.

Chem Commun 2009;31:4669-71

87. Gelbard HA, Nottet H, Swindells S,

et al. Platelet-activating-factor - a

candidate human-immunodeficiency-virus

type 1-induced neurotoxin. J Virol

1994;68(7):4628-35

88. Perry SW, Hamilton JA, Tjoelker LW,

et al. Platelet-activating factor receptor

activation - an initiator step in

A. M. Al-Ghananeem et al.

984 Expert Opin. Drug Deliv. (2013) 10(7)

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.

Page 13: Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

HIV-1 neuropathogenesis. J Biol Chem

1998;273(28):17660-4

89. Bito H, Nakamura M, Honda Z, et al.

Platelet-activating factor (PAF) receptor

in rat brain: PAF mobilizes intracellular

Ca2+ in hippocampal neurons. Neuron

1992;9(2):285-94

90. Martin M, Serradji N,

Dereuddre-Bosquet N, et al. PMS-601, a

new platelet-activating factor receptor

antagonist that inhibits human

immunodeficiency virus replication and

potentiates zidovudine activity in

macrophages.

Antimicrob Agents Chemother

2000;44(11):3150-4

91. Sallem W, Serradji N,

Dereuddre-Bosquet N, et al.

Structure-activity relationships in

platelet-activating factor. Part 14:

synthesis and biological evaluation of

piperazine derivatives with dual anti-PAF

and anti-HIV-1 activity.

Bioorg Med Chem

2006;14(23):7999-8013

92. Serradji N, Martin M, Bensaid O, et al.

Structure-activity relationships in

platelet-activating factor. 12. Synthesis

and biological evaluation of

platelet-activating factor antagonists with

anti-HIV-1 activity. J Med Chem

2004;47(25):6410-19

93. Tamai I, Tsuji A. Transporter-mediated

permeation of drugs across the

blood-brain barrier. J Pharm Sci

2000;89(11):1371-88

94. Farese-Di Giorgio A, Rouquayrol M,

Greiner J, et al. Synthesis and anti-HIV

activity of prodrugs derived from

saquinavir and indinavir.

Antivir Chem Chemother

2000;11(2):97-110

95. Rouquayrol M, Gaucher W, Greiner J,

et al. Synthesis and anti-HIV activity of

glucose-containing prodrugs derived from

saquinavir, indinavir and nelfinavir.

Carbohydr Res 2001;336(3):161-80

96. Rouquayrol M, Gaucher B, Roche D,

et al. Transepithelial transport of

prodrugs of the HIV protease inhibitors

saquinavir, indinavir, and nelfinavir

across Caco-2 Cell monolayers.

Pharm Res 2002;19(11):1704-12

97. Berezovskaya YV, Chudinov MV. Ester

derivatives of nucleoside inhibitors of

reverse transcriptase: 1. Molecular

transport systems for 3’-azido-3’-

deoxythymidine and 2’, 3’-didehydro-3’-

deoxythymidine. Bioorg Khim

2005;31(4):303-19

98. Gottesman MM, Pastan I. Biochemistry

of multidrug-resistance mediated by the

multidrug transporter.

Annu Rev Biochem 1993;62:385-427

99. Polli JW, Jarrett JL, Studenberg SD,

et al. Role of P-glycoprotein on the CNS

disposition of amprenavir (141W94), an

HIV protease inhibitor. Pharm Res

1999;16(8):1206-12

100. Varatharajan L, Thomas SA. The

transport of anti-HIV drugs across

blood-CNS interfaces: summary of

current knowledge and recommendations

for further research (vol 82, pg A99,

2009). Antiviral Res 2009;84(2):203-3

101. Sosnik A, Chiappetta DA,

Carcaboso AM. Drug delivery systems in

HIV pharmacotherapy: what has been

done and the challenges standing ahead.

J Control Release 2009;138(1):2-15

102. Drewe J, Gutmann H, Fricker G, et al.

HIV protease inhibitor ritonavir: a more

potent inhibitor of P-glycoprotein than

the cyclosporine analog SDZ PSC 833.

Biochem Pharmacol

1999;57(10):1147-52

103. Fishman RA. Blood-brain and CSF

barriers to penicillin and related organic

acids. Arch Neurol 1966;15(2):113

104. Spector R, Goetzl EJ.

Leukotriene-c4 transport and metabolism

in the central-nervous-system.

J Neurochem 1986;46(4):1308-12

105. Spector R. Pantothenic-acid transport

and metabolism in the

central-nervous-system. Am J Physiol

1986;250(2):R292-7

106. Hedaya MA, Sawchuk RJ. Effect of

probenecid on the renal and nonrenal

clearances of zidovudine and its

distribution into cerebrospinal fluid in

the rabbit. J Pharm Sci

1989;78(9):716-22

107. Hedaya MA, Elmquist WF, Sawchuk RJ.

Probenecid inhibits the metabolic and

renal clearances of zidovudine (AZT) in

human volunteers. Pharm Res

1990;7(4):411-17

108. Galinsky RE, Flaharty KK,

Hoesterey BL, et al. Probenecid enhances

central-nervous-system uptake of

2’,3’-dideoxyinosine by inhibiting

cerebrospinal-fluid efflux. J Pharmacol

Exp Ther 1991;257(3):972-8

109. Miller DW, Batrakova EV, Waltner TO,

et al. Interactions of Pluronic block

copolymers with brain microvessel

endothelial cells: evidence of two

potential pathways for drug absorption.

Bioconjug Chem 1997;8(5):649-57

110. Spitzenberger TJ, Heilman D,

Diekmann C, et al. Novel delivery

system enhances efficacy of antiretroviral

therapy in animal model for

HIV-1 encephalitis. J Cereb Blood

Flow Metab 2007;27(5):1033-42

111. Shaik N, Pan G, Elmquist WF.

Interactions of Pluronic block

copolymers on P-gp efflux activity:

experience with HIV-1 protease

inhibitors. J Pharm Sci

2008;97(12):5421-33

112. Namanja HA, Emmert D, Davis DA,

et al. Toward eradicating HIV reservoirs

in the brain: inhibiting p-glycoprotein at

the blood-brain barrier with prodrug

abacavir dimers. J Am Chem Soc

2012;134(6):2976-80

.. This study displays a novel approach

through dimerization with a dual

function of P-gp inhibition and

anti-HIV properties.

AffiliationAbeer M Al-Ghananeem† PhD, Michael Smith,

Maria L Coronel & Hieu Tran†Author for correspondence

Sullivan University, College of Pharmacy,

Department of Pharmaceutical Sciences,

2100 Gardiner Lane West Campus,

Louisville, KY 40205, USA

Tel: +1 502 413 8957;

E-mail: [email protected]

Advances in brain targeting and drug delivery of anti-HIV therapeutic agents

Expert Opin. Drug Deliv. (2013) 10(7) 985

Exp

ert O

pin.

Dru

g D

eliv

. Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Gaz

i Uni

v. o

n 08

/15/

14Fo

r pe

rson

al u

se o

nly.


Recommended