+ All Categories
Home > Documents > Factors Affecting the Clear ance and Biodistribution of ...

Factors Affecting the Clear ance and Biodistribution of ...

Date post: 14-Apr-2022
Category:
Upload: others
View: 2 times
Download: 0 times
Share this document with a friend
12
Factors Affecting the Clearance and Biodistribution of Polymeric Nanoparticles The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters. Citation Alexis, Frank et al. “Factors Affecting the Clearance and Biodistribution of Polymeric Nanoparticles.” Molecular Pharmaceutics 5.4 (2008): 505–515. © 2008 American Chemical Society As Published http://dx.doi.org/10.1021/mp800051m Publisher American Chemical Society (ACS) Version Final published version Citable link http://hdl.handle.net/1721.1/75024 Terms of Use Article is made available in accordance with the publisher's policy and may be subject to US copyright law. Please refer to the publisher's site for terms of use.
Transcript
Page 1: Factors Affecting the Clear ance and Biodistribution of ...

Factors Affecting the Clearance andBiodistribution of Polymeric Nanoparticles

The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters.

Citation Alexis, Frank et al. “Factors Affecting the Clearance andBiodistribution of Polymeric Nanoparticles.” MolecularPharmaceutics 5.4 (2008): 505–515. © 2008 American ChemicalSociety

As Published http://dx.doi.org/10.1021/mp800051m

Publisher American Chemical Society (ACS)

Version Final published version

Citable link http://hdl.handle.net/1721.1/75024

Terms of Use Article is made available in accordance with the publisher'spolicy and may be subject to US copyright law. Please refer to thepublisher's site for terms of use.

Page 2: Factors Affecting the Clear ance and Biodistribution of ...

Factors Affecting the Clearance and Biodistribution ofPolymeric Nanoparticles

Frank Alexis,*,†,‡,§ Eric Pridgen,‡,| Linda K. Molnar,⊥ andOmid C. Farokhzad*,†,‡

Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham andWomen’s Hospital, HarVard Medical School, Boston, Massachusetts 02115, MIT-HarVardCenter for Cancer Nanotechnology Excellence, HarVard-MIT DiVision of Health Sciences

and Technology, Department of Chemical Engineering, Massachusetts Institute ofTechnology, Cambridge, Massachusetts 02139, and National Cancer Institute, National

Institutes of Health, Bethesda, Maryland 20892

Received May 12, 2008; Revised Manuscript Received June 14, 2008; Accepted June 19, 2008

Abstract: Nanoparticle (NP) drug delivery systems (5-250 nm) have the potential to improvecurrent disease therapies because of their ability to overcome multiple biological barriers andreleasing a therapeutic load in the optimal dosage range. Rapid clearance of circulatingnanoparticles during systemic delivery is a critical issue for these systems and has made itnecessary to understand the factors affecting particle biodistribution and blood circulation half-life. In this review, we discuss the factors which can influence nanoparticle blood residencetime and organ specific accumulation. These factors include interactions with biological barriersand tunable nanoparticle parameters, such as composition, size, core properties, surfacemodifications (pegylation and surface charge), and finally, targeting ligand functionalization. Allthese factors have been shown to substantially affect the biodistribution and blood circulationhalf-life of circulating nanoparticles by reducing the level of nonspecific uptake, delayingopsonization, and increasing the extent of tissue specific accumulation.

Keywords: Biodistribution; circulation half-life; polymeric nanoparticles

IntroductionNanoparticles have drawn increasing interest from every

branch of medicine1–5 for their ability to deliver drugs inthe optimum dosage range, often resulting in increasedtherapeutic efficacy of the drug, weakened side effects,6,7

and improved patient compliance. Today, there are severalexamples of nontargeted NPs currently used in clinicalpractice (Doxil8–11 and Daunoxome12) and in clinical de-velopment (Cyclosert).13 Early success of these lipid-basedvesicular drug delivery nanoparticles has led to the investiga-tion and development of many different compositions ofpolymeric nanoparticles, including polymeric micelles, den-drimers, drug conjugates, and polypeptide- and polysaccha-ride-based nanoparticles. Among these, Genexol-PM [meth-oxy-PEG-poly(D,L-lactide)Taxol] is the first polymeric micellarnanoparticle in phase II clinical trials in the United States.14,15

Generally, clinical success correlates well with pharmaco-logical and toxicological parameters. Blood circulationresidence, maximal tolerated dose (MTD), and selectivityare the most important factors for achieving a high thera-peutical index and corresponding clinical success. Polymericnanoparticles are defined by their morphology and polymercomposition in the core and corona (Figure 1). The thera-peutic load is typically conjugated to the surface of the

* To whom correspondence should be addressed. O.C.F.: Brighamand Women’s Hospital, 75 Francis St., CWN-L1, Boston, MA02115; telephone, (617) 732-6093; fax, (617) 730-2801;e-mail, [email protected]. F.A.: Brigham andWomen’s Hospital, 75 Francis St., MRB-505, Boston, MA02115; e-mail, [email protected].

† Brigham and Women’s Hospital.‡ MIT-Harvard Center for Cancer Nanotechnology Excellence,

Massachusetts Institute of Technology.§ Harvard-MIT Division of Health Sciences and Technology,

Massachusetts Institute of Technology.| Department of Chemical Engineering, Massachusetts Institute

of Technology.⊥ National Institutes of Health.

reviews

10.1021/mp800051m CCC: $40.75 2008 American Chemical Society VOL. 5, NO. 4, 505–515 MOLECULAR PHARMACEUTICS 505Published on Web 08/04/2008

Page 3: Factors Affecting the Clear ance and Biodistribution of ...

nanoparticle, or encapsulated and protected inside the core.The delivery systems can be designed to provide eithercontrolled release or a triggered release of the therapeuticmolecule.16,17 The nanoparticle surface can then be func-tionalized by various methods to form the corona. Surfacefunctionalization can be utilized to increase residence timein the blood, reduce nonspecific distribution, and, in somecases, target tissues or specific cell surface antigens with atargeting ligand (peptide, aptamer, antibody/antibody frag-ment, small molecule). For instance, it is well establishedthat hydrophilic polymers, most notably poly(ethylene glycol)

(PEG), can be grafted, conjugated, or absorbed to the surfaceof nanoparticles to form the corona, which provides stericstabilization and confers “stealth” properties such as preven-tion of protein absorption.18,19 Surface functionalization canaddress the major limiting factor for long-circulating nano-particle systems, which is protein absorption. Proteinsadsorbed on the surface of the nanoparticle promote op-sonization, leading to aggregation and rapid clearance fromthe bloodstream.20–23 The resultant rapid clearance is dueto phagocytosis by the mononuclear phagocyte system (MPS)in the liver and splenic filtration. Typically, the majority ofopsonized particles are cleared by a receptor-mediatedmechanism in fewer than a few minutes due to the highconcentration of phagocytic cells in the liver and spleen, orthey are excreted.21 Thus, over the past 20 years, numerousapproaches to improving nanoparticle blood residence andaccumulation in specific tissues for the treatment of diseasehave been developed. In this review, we will discuss theeffects of physiological tissue defects (high permeability) andpolymeric nanoparticle physicochemical properties on theirbiodistribution and clearance. Specifically, polymeric com-position, nanoparticle size, pegylation, surface charge, andtargeting functionality will be discussed.

Overcoming Biological Barriers: Effect ofPhysiological Defects

Numerous biological barriers exist to protect the humanbody from invasion by foreign particles. These barriers

(1) Zhang, L.; et al. Nanoparticles in Medicine: Therapeutic Ap-plications and Developments. Clin. Pharmacol. Ther. 2008, 83,761–769.

(2) Moghimi, S. M.; Hunter, A. C. Poloxamers and poloxamines innanoparticle engineering and experimental medicine. Trends inbiotechnology 2000, 18, 412–420.

(3) Farokhzad, O. C.; Langer, R. Nanomedicine: Developing smartertherapeutic and diagnostic modalities. AdV. Drug DeliVery ReV.2006, 58, 1456–1459.

(4) Shaffer, C. Nanomedicine transforms drug delivery. DrugDiscoVery Today 2005, 10, 1581–1582.

(5) Alexis, F.; et al. New frontiers in nanotechnology for cancertreatment. Urol. Oncol. 2008, 26, 74–85.

(6) Safra, T.; et al. Pegylated liposomal doxorubicin (doxil): Reducedclinical cardiotoxicity in patients reaching or exceeding cumula-tive doses of 500 mg/m2. Ann. Oncol. 2000, 11, 1029–1033.

(7) Fassas, A.; Buffels, R.; Kaloyannidis, P.; Anagnostopoulos, A.Safety of high-dose liposomal daunorubicin (daunoxome) forrefractory or relapsed acute myeloblastic leukaemia. Br. Jo.Haematol. 2003, 122, 161–163.

(8) James, N. D.; et al. Liposomal doxorubicin (Doxil): An effectivenew treatment for Kaposi’s sarcoma in AIDS. Clin. Oncol. 1994,6, 294–296.

(9) Doxil receives FDA market clearance. AIDS Patient Care andSTDs, 1996; Vol. 10, p 135.

(10) Muggia, F. M. Doxil in breast cancer. J. Clin. Oncol. 1998, 16,811–812.

(11) Food and Drug Administration. FDA approves DaunoXome asfirst-line therapy for Kaposi’s sarcoma. Journal of the Interna-tional Association of Physicians in AIDS Care 1996, 2, 50–51.

(12) DaunoXome approved. AIDS Patient Care and STDs, 1996; Vol.10, p 263.

(13) Schluep, T.; et al. Preclinical efficacy of the camptothecin-polymer conjugate IT-101 in multiple cancer models. Clin.Cancer Res. 2006, 12, 1606–1614.

(14) Kim, D. W.; et al. Multicenter phase II trial of Genexol-PM, anovel Cremophor-free, polymeric micelle formulation of pacli-taxel, with cisplatin in patients with advanced non-small-cell lungcancer. Ann. Oncol. 2007, 18, 2009–2014.

(15) Lee, K. S.; et al. Multicenter phase II trial of Genexol-PM, aCremophor-free, polymeric micelle formulation of paclitaxel, inpatients with metastatic breast cancer. Breast Cancer Res. Treat.2008, 108, 241–250.

(16) Pridgen, E. M.; Langer, R.; Farokhzad, O. C. Biodegradable,polymeric nanoparticle delivery systems for cancer therapy.Nanomedicine (London, U.K.) 2007, 2, 669–680.

(17) Moghimi, S. M. Recent developments in polymeric nanoparticleengineering and their applications in experimental and clinicaloncology. Anti-Cancer Agents Med. Chem. 2006, 6, 553–561.

(18) Avgoustakis, K. Pegylated poly(lactide) and poly(lactide-co-glycolide) nanoparticles: Preparation, properties and possibleapplications in drug delivery. Curr. Drug DeliVery 2004, 1, 321–333.

(19) Otsuka, H.; Nagasaki, Y.; Kataoka, K. PEGylated nanoparticlesfor biological and pharmaceutical applications. AdV. DrugDeliVery ReV. 2003, 55, 403–419.

(20) Romberg, B.; Hennink, W. E.; Storm, G. Sheddable coatingsfor long-circulating nanoparticles. Pharm. Res. 2008, 25, 55–71.

Figure 1. Nanoparticle platforms for drug delivery.Polymeric nanoparticle platforms are characterizedby their physicochemical structures, including poly-merosome, solid polymeric nanoparticle, nanoshell,dendrimer, polymeric micelle, and polymer-drugconjugates.

reviews Alexis et al.

506 MOLECULAR PHARMACEUTICS VOL. 5, NO. 4

Page 4: Factors Affecting the Clear ance and Biodistribution of ...

include cellular and humoral arms of the immune system aswell as mucosal barriers among others. These barriers mustbe overcome in order for nanoparticles to reach their target(Figure 2). Due to their unique size, and amenability tosurface functionalization to incorporate the desired charac-teristics, nanoparticles are particularly well suited to over-coming these barriers. This is especially true in the case ofabnormal neovascularization. Blood vessels are responsiblefor delivering molecules, nutrients, and oxygen to organsthroughout the body. Endothelia composing the blood vesselshave been classified as continuous, fenestrated, or discon-tinuous, depending on the morphological features of theendothelium. The continuous endothelium morphology ap-pears in arteries, vessels,24,25 and the lungs.26 In contrast,fenestrated endothelium27 appears in glands,28 digestivemucosa, and kidney. Fenestrae have an octagonal symmetrywith radial fibrils interweaving in a central point forming

pores of approximately 60 nm. Discontinuous endotheliumis a characteristic of the liver (fenestrea of 50-100 nm)29

and bone marrow. Endothelial cells from the blood vesselsare able to respond to the physiological environment,resulting in angiogenic activity. Angiogenesis30,31 is well-characterized for cancers32 as well as ocular and inflamma-tory diseases,33 with antiangiogenesis compounds commonlyused for therapy.34,35 Angiogenesis during tumor growthresults in defective hypervasculature and a deficient lym-phatic drainage system, which has given rise to the conceptof passive targeting of nanoparticles to tumors through the“enhanced permeability and retention” (EPR) effect.36,37 TheEPR is a unique feature which allows macromolecules ordrug delivery nanoparticles (cutoff size of >400 nm) topreferentially accumulate and diffuse in tumor tissues.38

Long-circulating drug delivery nanoparticles are able toextravasate into tumor tissues, accumulate, and release thetherapeutic drug locally in the extracellular area. Similarly,abnormal neovascularization or angiogenesis as well asenhanced vascular permeability are major causes of manyocular disorders, including age-related macular degeneration(AMD), retinopathy of prematurity (ROP), ischemic retinalvein occlusions, and diabetic retinopathy (DR), causingirreversible vision loss. In ocular disorders, the angiogenicprocess appears to be due to a stimulus response for retinalneovascularization. The stimulus can be tissue hypoxia,inflammatory cell infiltration, increased local concentrationof cytokines (VEGF, PDGF, FGF5 TNF, IGF etc.). The result

(21) Moghimi, S. M.; Hunter, A. C.; Murray, J. C. Long-circulatingand target-specific nanoparticles: Theory to practice. Pharmacol.ReV. 2001, 53, 283–318.

(22) Vittaz, M.; et al. Effect of PEO surface density on long-circulating PLA-PEO nanoparticles which are very low comple-ment activators. Biomaterials 1996, 17, 1575–1581.

(23) Owens, D. E., III; Peppas, N. A. Opsonization, biodistribution,and pharmacokinetics of polymeric nanoparticles. Int. J. Pharm.2006, 307, 93–102.

(24) Predescu, D.; Palade, G. E. Plasmalemmal vesicles represent thelarge pore system of continuous microvascular endothelium.Am. J. Physiol. 1993, 265, H725–H733.

(25) Simionescu, M.; Simionescu, N.; Palade, G. E. Morphometricdata on the endothelium of blood capillaries. J. Cell Biol. 1974,60, 128–152.

(26) Brigham, K. L. Estimations of permeability properties ofpulmonary capillaries (continuous endothelium). Physiologist1980, 23, 44–46.

(27) Ludatscher, R. M.; Stehbens, W. E. Vesicles of fenestrated andnon-fenestrated endothelium. Z. Zellforsch. Mikrosk. Anat. 1969,97, 169–177.

(28) Ryan, U. S.; Ryan, J. W.; Smith, D. S.; Winkler, H. Fenestratedendothelium of the adrenal gland: Freeze-fracture studies. TissueCell 1975, 7, 181–190.

(29) Braet, F.; et al. Contribution of high-resolution correlativeimaging techniques in the study of the liver sieve in three-dimensions. Microsc. Res. Tech. 2007, 70, 230–242.

(30) Eisenstein, R. Angiogenesis in arteries: review. Pharmacol. Ther.1991, 49, 1–19.

(31) Folkman, J. What is the role of endothelial cells in angiogenesis.Laboratory InVestigation: A Journal of Technical Methods andPathology 1984, 51, 601–604.

(32) Folkman, J. Tumor angiogenesis: Therapeutic implications.N. Engl. J. Med. 1971, 285, 1182–1186.

(33) Folkman, J. Angiogenesis in cancer, vascular, rheumatoid andother disease. Nat. Med. 1995, 1, 27–31.

(34) Folkman, J.; Ingber, D. Inhibition of angiogenesis. Semin. CancerBiol. 1992, 3, 89–96.

(35) Kerbel, R.; Folkman, J. Clinical translation of angiogenesisinhibitors. Nat. ReV. 2002, 2, 727–739.

(36) Maeda, H. The enhanced permeability and retention (EPR) effectin tumor vasculature: The key role of tumor-selective macro-molecular drug targeting. AdV. Enzyme Regul. 2001, 41, 189–207.

(37) Greish, K. Enhanced permeability and retention of macromo-lecular drugs in solid tumors: A royal gate for targeted anticancernanomedicines. J. Drug Targeting 2007, 15, 457–464.

(38) Hobbs, S. K.; et al. Regulation of transport pathways in tumorvessels: Role of tumor type and microenvironment. Proc. Natl.Acad. Sci. U.S.A. 1998, 95, 4607–4612.

Figure 2. Biodistribution and clearance of polymericnanoparticles. Tissue defects, stealth properties,targeting, and the size of the nanoparticles are majorfactors affecting the biodistribution and clearance ofpolymeric nanoparticles.

Factors Affecting the Pharmacokinetics of Polymeric Nanoparticles reviews

VOL. 5, NO. 4 MOLECULAR PHARMACEUTICS 507

Page 5: Factors Affecting the Clear ance and Biodistribution of ...

is the formation of new vessels, which disrupt the organi-zational structure of the neural retina or break through theinner limiting membranes into the vitreous.39 Many otherdisorders are also characterized by angiogenesis or vascu-lature defects such as obesity,40 asthma,41 diabetes,42,43 andmultiple sclerosis.44,45 The development of new imagingsystems and medical knowledge about physiological defectsis leading to novel therapeutic approaches using nanoparticledrug delivery systems. It is now well accepted that nano-particles are suitable for crossing biological barriers throughtissue diffusion, extravasation, and escape from hepaticfiltration.

Nanoparticle PropertiesComposition. Historically, the administration of thera-

peutic agents has been limited by multiple factors, primaryamong these being low solubility, stability and rapid clear-ance. The result is a short circulation half-life and lowefficacy, making frequent administration necessary. Ad-ditionally, there can be significant side effects in non-diseasedtissues that adsorb therapeutic agent. These issues have ledto the development of various targeting strategies aimed atincreasing therapeutic index, including monoclonal antibodiesand immunoconjugates. Some of the strategies are currentlyused in clinical practice and others are in clinical develop-ment.46 In addition to these strategies, it has been shownthat polymer-drug conjugates can substantially improve theblood residence time and weaken side effects. It is anticipatedthat engineered multifunctional nanoparticles can addressissues with targeting as well as carry a more substantial drugpayload. Many polymers have been investigated, includingHPMA [N-(2-hydroxypropyl)methacrylamide], dextran, andpolyglutamate. HPMA and polyglutamate drug conjugatesrepresent ∼35% of all the polymeric drug delivery systemsin clinical development. The clinical success of AP5346,PK1, PK2, Xyotax, and CT-2106 seems to be due to theirlong circulation half-life, passive targeting ability, and, mostimportantly, lower toxicity allowing higher dosages (see

Table 1). Thus far, the main impact of polymer conjugateshas been to improve the pharmacokinetic parameters of drugsalready in clinical use. These polymeric nanocarriers mustbe non-toxic, non-immunogenic, and carry sufficient amountof drug and release the drug at the optimal dose. The use ofpoly(ethylene glycol) (PEG) for nanoparticle surface func-tionalization has had led to very favorable results due to itsintrinsic physicochemical properties but has had limitedimpact as a drug conjugate carrier due to low drug loadings.PEG polymers have low toxicity and no immunogenicity andare approved by the Food and Drug Administration (FDA)for clinical use. PEG-drug conjugates or nanoparticlesfunctionalized with PEG chains have been described as long-circulating drug delivery systems with potential applicationsfor systemic drug administration.21,47 Poly(ethylene glycol)or poly(ethylene oxide) refers to an oligomer or polymer ofethylene oxide in linear or branched structures.48 PEG is ahydrophilic polymer that can be adsorbed or covalentlyattached to the surface of nanoparticles. Hrkach et al. havedemonstrated the formation of PLA-PEG nanoparticles ina core-corona structure with a solid core and anchored PEGchains on the surface. Their results showed for the first timethat PEG chains, covalently attached to a particle surface,could exhibit flexibility similar to that of free PEG polymerdissolved in water. Furthermore, PEG has been shown tosubstantially reduce nonspecific interactions with proteinsthrough its hydrophilicity and steric repulsion effects, reduc-ing opsonization and complement activation.49–55 The chainlength, shape, and density of PEG on the particle surfacehave been shown to be the main parameters affectingnanoparticle surface hydrophilicity and phagocytosis. Themechanism involved in phagocytosis of opsonized nanopar-ticles is receptor-mediated by interaction of specific proteinsabsorbed on the surface of the nanoparticles with phagocytes.

(39) Folkman, J. Fundamental concepts of the angiogenic process.Curr. Mol. Med. 2003, 3, 643–651.

(40) Lijnen, H. R. Angiogenesis and obesity. CardioVasc. Res. 2008,78, 286–293.

(41) Chetta, A.; Zanini, A.; Torre, O.; Olivieri, D. Vascular remodel-ling and angiogenesis in asthma: Morphological aspects andpharmacological modulation. Inflammation Allergy Drug Targets2007, 6, 41–45.

(42) Norrby, K.; Jakobsson, A.; Simonsen, M.; Sorbo, J. Increasedangiogenesis in diabetes. Experientia 1990, 46, 856–860.

(43) Martin, A.; Komada, M. R.; Sane, D. C. Abnormal angiogenesisin diabetes mellitus. Med. Res. ReV. 2003, 23, 117–145.

(44) Kirk, S.; Frank, J. A.; Karlik, S. Angiogenesis in multiplesclerosis: Is it good, bad or an epiphenomenon. J. Neurol. Sci.2004, 217, 125–130.

(45) Siegel, R. C. Angiogenesis in progressive systemic sclerosis.N. Engl. J. Med. 1972, 286, 217.

(46) Wu, A. M.; Senter, P. D. Arming antibodies: Prospects andchallenges for immunoconjugates. Nat. Biotechnol. 2005, 23,1137–1146.

(47) Gref, R.; et al. Biodegradable long-circulating polymeric nano-spheres. Science 1994, 263, 1600–1603.

(48) Ben-Shabat, S.; Kumar, N.; Domb, A. J. PEG-PLA blockcopolymer as potential drug carrier: Preparation and character-ization. Macromol. Biosci. 2006, 6, 1019–1025.

(49) Bazile, D.; et al. Stealth Me.PEG-PLA nanoparticles avoid uptakeby the mononuclear phagocytes system. J. Pharm. Sci. 1995,84, 493–498.

(50) Peracchia, M. T.; et al. Stealth PEGylated polycyanoacrylatenanoparticles for intravenous administration and splenic targeting.J. Controlled Release 1999, 60, 121–128.

(51) Peracchia, M. T.; et al. Visualization of in vitro protein-rejectingproperties of PEGylated stealth polycyanoacrylate nanoparticles.Biomaterials 1999, 20, 1269–1275.

(52) Li, Y.; et al. PEGylated PLGA nanoparticles as protein carriers:Synthesis, preparation and biodistribution in rats. J. ControlledRelease 2001, 71, 203–211.

(53) Gref, R.; et al. ‘Stealth’ corona-core nanoparticles surfacemodified by polyethylene glycol (PEG): Influences of the corona(PEG chain length and surface density) and of the corecomposition on phagocytic uptake and plasma protein adsorption.Colloids Surf. 2000, 18, 301–313.

(54) Scott, M. D.; Murad, K. L. Cellular camouflage: Fooling theimmune system with polymers. Curr. Pharm. Des. 1998, 4, 423–438.

reviews Alexis et al.

508 MOLECULAR PHARMACEUTICS VOL. 5, NO. 4

Page 6: Factors Affecting the Clear ance and Biodistribution of ...

Tab

le1.

Clin

ical

Sta

tus

ofP

olym

eric

Dru

gD

eliv

ery

Nan

opar

ticle

sun

der

Dev

elop

men

t

com

posi

tion

(tra

dena

me)

stat

uspa

rtic

lesi

ze(n

m)

circ

ulat

ion

half-

life

rela

tive

tissu

eac

cum

ulat

ion

ofth

edr

ugm

axim

umto

lera

ted

dose

(MT

D)

effic

acy

met

hoxy

-PE

G-p

oly

(D,L

-lact

ide)

Tax

ol(G

enex

ol-P

M)

Pha

seII

30-

60nm

pacl

itaxe

l,12

h(h

uman

)61

Gen

exol

-PM

vsT

axol

:2×

mor

ein

liver

,sp

leen

,he

art,

and

tum

or(m

ice)

62

390

mg/

m2

adm

inis

tere

din

trav

enou

sly

for

3h

ever

y3

wee

ks(h

uman

)14

75%

ofm

etas

tatic

brea

stca

ncer

patie

ntsh

owed

2ye

ars

over

alls

urvi

val1

5

HP

MA

-DA

CH

pala

tinat

e(P

roLi

ndac

,pr

evio

usly

AP

5346

)

Pha

seII

6-15

nmD

AC

H-p

latin

um,

70h

(hum

an)6

3N

Aa

640

mg/

m2

(initi

alcy

cle)

;re

peat

edcy

cles

ofth

erap

yw

ere

not

asse

ssed

(hum

an)6

4

NA

a

PE

G-a

rgin

ine

deam

inas

e(H

epac

id,

prev

ious

lyA

DI-

SS

PE

G20

000

MW

)

Pha

seI/I

IN

Aa

argi

nine

deam

inas

e,7

days

(hum

an)6

5N

Aa

640

units

/m2

once

aw

eek

(MT

D>

max

imum

feas

ible

dose

byin

trav

enou

s(iv

)ad

min

istr

atio

n)6

6

doub

lem

edia

nsu

rviv

altim

eof

patie

nts

with

met

asta

ticm

elan

oma;

47%

resp

onse

rate

inH

CC

patie

nts

(n)

19)6

6

PE

G-c

ampt

othe

cin

(Pro

thec

an)

Pha

seI/I

IN

Aa

cam

ptot

heci

n,40

h(h

uman

)67

cam

ptot

heci

n%

ID/g

oftis

sue

24h

post

inje

ctio

n:3.

7%in

tum

or,

4.41

%in

bloo

d,2.

32%

inliv

er(m

ice)

68

7000

mg/

m2

adm

inis

tere

din

1h

ivin

fusi

ons

ever

y3

wee

ks(h

uman

)67

NA

a

Plu

roni

c-do

xoru

bici

n(S

P10

49C

)P

hase

II∼2

5nm

doxo

rubi

cin,

3h

(hum

an)6

9en

hanc

edA

UC

intu

mor

(50.

8vs

30.1

)an

dbr

ain

(9.2

vs5.

6)co

mpa

red

with

free

doxo

rubi

cin

(mic

e)7

0

70m

g/m

2ad

min

iste

red

intr

aven

ousl

yev

ery

3w

eeks

for

am

axim

umof

six

cycl

es(h

uman

)69

thre

epa

tient

sov

er21

show

edre

spon

ses

totr

eatm

ent6

9

poly

cycl

odex

trin

cam

ptot

heci

n(I

T-1

01)

Pha

seI

∼40

nmca

mpt

othe

cin,

38h

(mic

e)7

1ca

mpt

othe

cin

%ID

/gof

tissu

e:tu

mor

)1.

3%;

liver

)1.

9%(2

4h)

(mic

e)1

3,7

1

NA

apr

elim

inar

yda

tais

repo

rted

tosh

owst

able

dise

ase

rate

inpa

tient

sw

ithso

lidtu

mor

s13

poly

glut

amat

eca

mpt

othe

cin

(CT

-210

6)P

hase

I/II

NA

aca

mpt

othe

cin,

44-

63h

(hum

an)7

2N

Aa

25m

g/m

2ad

min

iste

red

byiv

infu

sion

wee

kly

ever

y3

of4

wee

ks(h

uman

)72

less

toxi

city

than

free

drug

72

poly

glut

amat

epa

clita

xel

(Xyo

tax)

Pha

seIII

NA

apa

clita

xel,

100

h(h

uman

)73

pacl

itaxe

l%ID

/gof

tissu

e:tu

mor

)2.

2%;

sple

en)

16%

;liv

er)

8%(m

ice)

74

for

aniv

adm

inis

trat

ion,

233

mg/

m2

for

patie

nts

ona

two-

dose

wee

kly

sche

dule

and

177

mg/

m2

ona

thre

e-do

sew

eekl

ysc

hedu

le(h

uman

)73

resp

onse

rate

of10

%fo

r99

patie

nts

and

am

edia

ntim

eto

dise

ase

prog

ress

ion

of2

mon

ths7

5

dext

ran-

doxo

rubi

cin

(AD

-70)

Pha

seI

NA

ado

xoru

bici

n,3-

12h

(hum

an)7

6N

Aa

20m

g/m

2(h

uman

)76

disc

ontin

ued

due

tose

vere

hepa

toto

xici

tylim

iting

the

dose

at20

mg/

m2

76

dext

ran-

cam

ptot

heci

n(D

E-3

10)

Pha

seI/I

IN

Aa

cam

ptot

heci

n,30

0-40

0h

(hum

an)7

7N

Aa

9m

g/m

2gi

ven

once

ever

y4

wee

ks(h

uman

)77

none

wm

ajor

toxi

city

com

pare

dto

drug

besi

dehe

pato

toxi

city

repo

rted

asre

vers

ible

77

HP

MA

-pac

litax

el(P

NU

1669

45)

Pha

seI

NA

apa

clita

xel,

3-12

h(h

uman

)78

NA

a19

6m

g/m

2(h

uman

)78

disc

ontin

ued

due

tose

vere

neur

otox

icity

78

HP

MA

-dox

orub

icin

(PK

1)P

hase

IIN

Aa

doxo

rubi

cin,

93h

(hum

an)7

9N

Aa

320

mg/

m2

(hum

an)7

9he

patic

toxi

city

atdo

ses

>12

0m

g/m

2;

two

part

ial

and

two

min

orre

spon

ses

over

36pa

tient

s79

PE

G-a

spar

ticac

id-d

oxor

ubic

in(N

K91

1)P

hase

I30

-50

nmdo

xoru

bici

n,1.

6-4.

7h

(hum

an)8

067

mg/

m2,

plas

ma

clea

ranc

e40

0-fo

ldhi

gher

than

Dox

il(h

uman

)80

nose

vere

toxi

city

;P

hase

IIcl

inic

altr

ialf

orpa

ncre

atic

canc

er8

0

HP

MA

-dox

orub

icin

with

gala

ctos

amin

e(P

K2)

Pha

seI

∼8.4

nmbi

phas

iccl

eara

nce

with

half-

lives

of2.

9an

d26

.7h

whe

n12

0m

g/m

2

adm

inis

tere

dby

24h

infu

sion

(hum

an)8

1

enha

nced

accu

mul

atio

nin

liver

,he

pato

ma,

and

met

asta

tiche

pato

ma

com

pare

dw

ithno

ntar

gete

dH

PM

A-d

oxor

ubic

in(h

uman

)81

160

mg/

m2

with

adm

inis

trat

ion

byiv

infu

sion

over

1h

ever

y3

wee

ks(h

uman

)81

of18

patie

nts,

thre

ere

spon

ded

totr

eatm

ent,

with

two

inpa

rtia

lrem

issi

onfo

r>

26an

d>

47m

onth

s81

aN

otap

plic

able

.

Factors Affecting the Pharmacokinetics of Polymeric Nanoparticles reviews

VOL. 5, NO. 4 MOLECULAR PHARMACEUTICS 509

Page 7: Factors Affecting the Clear ance and Biodistribution of ...

Gref et al. were the first to report the advantages ofpegylation on PLGA-PEG nanoparticles, resulting in asubstantial increase in blood residence time. Apolipoproteins(A-IV and E) were found to be present on the surface ofpure PLA nanoparticles in contrast to the PLA-PEGnanoparticles using gel electrophoresis analysis.53 Althoughthe assay did not provide a precise quantitative analysis, itwas clearly shown that protein adsorption, particularlyapolipoprotein J and complement protein C3, was substan-tially reduced on the surface of the PLA-PEG nanoparticlescompared with pure PLA formulations. These results suggestthat these specific apolipoproteins might also play a role inthe process of opsonization of the particles. Gref et al.53 havesystematically studied the effect of PEG chain length inpreventing protein adsorption on the surface of the nano-particles. The results showed that an optimal molecular mass(Mw) range exists (between 2 and 5 kDa) in order to reduceplasma protein adsorption. The amount of protein absorbedon PLA-PEG 5 kDa was substantially reduced (∼80%)compared to the amount of nonpegylated PLA nanoparticles.PEG content as low as 0.5 wt % on the surface of thenanoparticles was able to significantly reduce the totalamount of protein when compared to the nonpegylated PLAnanoparticles. The most significant reduction of proteinabsorption was found for pegylated particles (5 wt %). Theeffect of PEG on the surface of the nanoparticle in preventingprotein absorption correlated with the polymorphonuclearleukocyte (PMN) and human monocyte (THP-1) uptake.Interestingly, a threshold of 1-2 nm space between the PEGchains was estimated for minimal protein absorption. Sincethen, many research groups have investigated the interactionof polymeric nanoparticles with serum opsonins, character-izing both uptake mechanisms and kinetics.53,56–58 In onestudy, the relationship between the protein adsorption kineticsof the nanoparticles and their uptake in the liver wasinvestigated.56 The results showed that the rate of hepaticuptake of polystyrene nanoparticles (50 nm) was significantlyhigher when nanoparticles were incubated with serum priorto transfusion in the rat liver. It was suggested that theincreased rate of hepatic uptake was mainly mediated byopsonization. In vitro uptake studies showed that theincreased amount of complement protein C3 and immuno-globulin G (IgG) adsorbed on nanoparticles was directlyreflected in the increased rate of uptake of nanoparticles by

Kupffer cells. These results indicate that the amount ofopsonins associated with the surface of the nanoparticlesincreased over time and triggered substantial hepatic ag-glomeration. Similarly, C. Fang et al.59 have recently shownthe effect of the molecular mass of PEG for passive targetingof stealth poly(cyanoacrylate-co-n-hexadecyl) cyanoacrylate(PHDCA) nanoparticles. As in previous studies, PEG surfacemodification of nanoparticles was able to dramatically reduceprotein absorption as detected using quantitative assays (BCAassay). The amount of protein adsorbed was directly de-pendent on the molecular mass of the PEG. Medium-sizedpegylated nanoparticles (100-200 nm) showed 10-40%protein absorption, and PEG 10 kDa was found to be themost efficient size of PEG as compared to PEG 2 kDa andPEG 5 kDa in preventing protein absorption. The resultssuggest that a dense PEG shielding over a negatively chargedsurface is important in preventing protein absorption. Thetrend in shielding properties was confirmed by the analysisof nanoparticle uptake by murine macrophages and bloodclearance kinetics. Interestingly, the level of tumor necrosisfactor-R (TNF-R) delivered by nanoparticles into tumor-bearing mice was clearly correlated with the shieldingdensity. Another recent example confirmed the advantagesof surface pegylation by utilizing thiolated gelatin nanopar-ticles (∼300 nm) to improve passive tumor targeting in anorthotopic human breast adenocarcinoma xenograft mousemodel.60 Despite the fact that these particles were twice thesize of those in the previous study, the results showed a lowerrate of uptake of the PEG-modified nanoparticles by the liver,indicative of the stealth properties of pegylated nanoparticles.In general, pegylated nanoparticles were found to have longercirculation time and higher levels of tumor accumulation thannonpegylated nanoparticles. In summary, much has beenlearned about PEG molecular mass and PEG density onnanoparticles which has led to reduced plasma proteinadsorption, opsonization, and nonspecific uptake. In turn, thishas resulted in increased nanoparticle circulation half-life andimproved therapeutic efficacy of drugs delivered usingpegylated nanocarriers.

Effect of Size. On the basis of physiological parameterssuch as hepatic filtration, tissue extravasation, tissue diffu-sion, and kidney excretion, it is clear that, along with surfacecomposition, particle size is a key factor in the biodistributionof long-circulating nanoparticles and achieving therapeuticefficacy (Figure 2). In one study, in vivo biodistributionresults of polystyrene nanoparticles with consistent composi-(55) Francis, G. E.; Delgado, C.; Fisher, D.; Malik, F.; Agrawal, A. K.

Polyethylene glycol modification: Relevance of improved meth-odology to tumour targeting. J. Drug Targeting 1996, 3, 321–340.

(56) Nagayama, S.; Ogawara, K.; Fukuoka, Y.; Higaki, K.; Kimura,T. Time-dependent changes in opsonin amount associated onnanoparticles alter their hepatic uptake characteristics. Int.J. Pharm. 2007, 342, 215–221.

(57) Panagi, Z.; et al. Effect of dose on the biodistribution andpharmacokinetics of PLGA and PLGA-mPEG nanoparticles. Int.J. Pharm. 2001, 221, 143–152.

(58) Beletsi, A.; Panagi, Z.; Avgoustakis, K. Biodistribution propertiesof nanoparticles based on mixtures of PLGA with PLGA-PEGdiblock copolymers. Int. J. Pharm. 2005, 298, 233–241.

(59) Fang, C.; et al. In vivo tumor targeting of tumor necrosis factor-R-loaded stealth nanoparticles: Effect of MePEG molecularweight and particle size. Eur. J. Pharm. Sci. 2006, 27, 27–36.

(60) Kommareddy, S.; Amiji, M. Biodistribution and pharmacokineticanalysis of long-circulating thiolated gelatin nanoparticles fol-lowing systemic administration in breast cancer-bearing mice.J. Pharm. Sci. 2007, 96, 397–407.

(81) Rijcken, C. J.; Snel, C. J.; Schiffelers, R. M.; van Nostrum, C. F.;Hennink, W. E. Hydrolysable core-crosslinked thermosensitivepolymeric micelles: Synthesis, characterisation and in vivostudies. Biomaterials 2007, 28, 5581–5593.

reviews Alexis et al.

510 MOLECULAR PHARMACEUTICS VOL. 5, NO. 4

Page 8: Factors Affecting the Clear ance and Biodistribution of ...

tion and varying particle sizes of 50 and 500 nm showedhigher levels of agglomeration of the larger nanoparticles inthe liver.56 It was suggested that the mechanism of hepaticuptake was mediated by surface absorption of proteinsleading to opsonization. However, the effect of temperature(37 °C vs 4 °C) on hepatic elimination showed unexpectedlyfaster uptake of the 50 nm polystyrene nanoparticles at thelower temperature. Similarly, the size of the nanoparticle wasshown to have a substantial effect on the protein absorption.Small (<100 nm), medium (100-200 nm), and large (>200nm) pegylated PHDCA nanoparticles incubated with serumprotein for 2 h showed a significant correlation betweenparticle size and protein absorption.59Protein absorption onsmall nanoparticles (80 nm) was quantified (6%) andcompared to the same nanoparticle formulation with a larger

size (171 and 243 nm, 23 and 34%, respectively). The effectof protein absorption on different sized nanoparticles wasalso confirmed with the analysis of nanoparticle uptake bymurine macrophages and blood clearance kinetics. Bloodclearance of the smaller nanoparticles was twice as slow aswith the larger nanoparticle formulations. More importantly,the amount of drug encapsulated (TNF-R) in nanoparticlesthat accumulated in the tumor within 24 h was twice that ofthe larger nanoparticle formulation. The results suggest thatit might be due to a higher surface PEG density (brushlike)on the surface of smaller nanoparticles. Rijcken et al.81 haveshown that size and polydispersity can substantially affectthe biodistribution of micelles. Cross-linked micelles incu-bated in PBS (pH 7.4) at 37 °C for more than 3 days did notshow substantial increases in size and polydispersity incontrast to non-cross-linked micelles (PD ∼ 0.5 after 10 h).Stable micelles showed a long circulation half-life of ∼8 hdue to a low rate of hepatic uptake (liver ∼ 10% of theinjected dose; spleen ∼ 2% of the injected dose). Similar toresults with small liposomes (<100 nm), micelles were foundto accumulate in the skin.

In summary, it has been consistently shown that pegylatednanoparticles smaller than 100 nm have reduced plasmaprotein adsorption on their surface and also have reducedhepatic filtration. Further, these small pegylated nanoparticleshave a long blood residence time and a high rate ofextravasation into permeable tissues, demonstrating theimportance of tunable particle size and surface compositionfor achieving effective, targeted delivery.

(61) Kim, T. Y.; et al. Phase I and pharmacokinetic study of Genexol-PM, a cremophor-free, polymeric micelle-formulated paclitaxel,in patients with advanced malignancies. Clin. Cancer Res. 2004,10, 3708–3716.

(62) Kim, S. C.; et al. In vivo evaluation of polymeric micellarpaclitaxel formulation: Toxicity and efficacy. J. ControlledRelease 2001, 72, 191–202.

(63) Campone, M.; et al. Phase I and pharmacokinetic trial of AP5346,a DACH-platinum-polymer conjugate, administered weekly forthree out of every 4 weeks to advanced solid tumor patients.Cancer Chemother. Pharmacol. 2007, 60, 523–533.

(64) Rice, J. R.; Gerberich, J. L.; Nowotnik, D. P.; Howell, S. B.Preclinical efficacy and pharmacokinetics of AP5346, a noveldiaminocyclohexane-platinum tumor-targeting drug deliverysystem. Clin. Cancer Res. 2006, 12, 2248–2254.

(65) Ascierto, P. A.; et al. Pegylated arginine deiminase treatment ofpatients with metastatic melanoma: Results from phase I and IIstudies. J. Clin. Oncol. 2005, 23, 7660–7668.

(66) Ensor, C. M.; Holtsberg, F. W.; Bomalaski, J. S.; Clark, M. A.Pegylated arginine deiminase (ADI-SS PEG20,000 mw) inhibitshuman melanomas and hepatocellular carcinomas in vitro andin vivo. Cancer Res. 2002, 62, 5443–5450.

(67) Posey, J. A., III; et al. Phase 1 study of weekly polyethyleneglycol-camptothecin in patients with advanced solid tumors andlymphomas. Clin. Cancer Res. 2005, 11, 7866–7871.

(68) Conover, C. D.; Greenwald, R. B.; Pendri, A.; Gilbert, C. W.;Shum, K. L. Camptothecin delivery systems: Enhanced efficacyand tumor accumulation of camptothecin following its conjuga-tion to polyethylene glycol via a glycine linker. CancerChemother. Pharmacol. 1998, 42, 407–414.

(69) Danson, S.; et al. Phase I dose escalation and pharmacokineticstudy of pluronic polymer-bound doxorubicin (SP1049C) inpatients with advanced cancer. Br. J. Cancer 2004, 90, 2085–2091.

(70) Schluep, T.; Cheng, J.; Khin, K. T.; Davis, M. E. Pharmacoki-netics and biodistribution of the camptothecin-polymer conjugateIT-101 in rats and tumor-bearing mice. Cancer Chemother.Pharmacol. 2006, 57, 654–662.

(71) Homsi, J.; et al. Phase I trial of poly-L-glutamate camptothecin(CT-2106) administered weekly in patients with advanced solidmalignancies. Clin. Cancer Res. 2007, 13, 5855–5861.

(72) Boddy, A. V.; et al. A phase I and pharmacokinetic study ofpaclitaxel poliglumex (XYOTAX), investigating both 3-weeklyand 2-weekly schedules. Clin. Cancer Res. 2005, 11, 7834–7840.

(73) Li, C.; et al. Biodistribution of paclitaxel and poly(L-glutamicacid)-paclitaxel conjugate in mice with ovarian OCa-1 tumor.Cancer Chemother. Pharmacol. 2000, 46, 416–422.

(74) Sabbatini, P.; et al. Phase II study of CT-2103 in patients withrecurrent epithelial ovarian, fallopian tube, or primary peritonealcarcinoma. J. Clin. Oncol. 2004, 22, 4523–4531.

(75) Danhauser-Riedl, S.; et al. Phase I clinical and pharmacokinetictrial of dextran conjugated doxorubicin (AD-70, DOX-OXD).InVest. New Drugs 1993, 11, 187–195.

(76) Soepenberg, O.; et al. Phase I and pharmacokinetic study of DE-310 in patients with advanced solid tumors. Clin. Cancer Res.2005, 11, 703–711.

(77) Meerum Terwogt, J. M.; et al. Phase I clinical and pharmaco-kinetic study of PNU166945, a novel water-soluble polymer-conjugated prodrug of paclitaxel. Anti-Cancer Drugs 2001, 12,315–323.

(78) Vasey, P. A.; et al. Phase I clinical and pharmacokinetic studyof PK1 [N-(2-hydroxypropyl)methacrylamide copolymer doxo-rubicin]: First member of a new class of chemotherapeuticagents-drug-polymer conjugates. Cancer Research CampaignPhase I/II Committee. Clin. Cancer Res. 1999, 5, 83–94.

(79) Matsumura, Y.; et al. Phase I clinical trial and pharmacokineticevaluation of NK911, a micelle-encapsulated doxorubicin. Br. J.Cancer 2004, 91, 1775–1781.

(80) Hopewel, J. W.; Duncan, R.; Wilding, D.; Chakrabarti, K.Preclinical evaluation of the cardiotoxicity of PK2: A novelHPMA copolymer-doxorubicin-galactosamine conjugate antitu-mour agent. Hum. Exp. Toxicol. 2001, 20, 461–470.

(82) Shenoy, D.; Little, S.; Langer, R.; Amiji, M. Poly(ethyleneoxide)-modified poly(-amino ester) nanoparticles as a pH-sensitive system for tumor-targeted delivery of hydrophobicdrugs: Part 2. In vivo distribution and tumor localization studies.Pharm. Res. 2005, 22, 2107–2114.

Factors Affecting the Pharmacokinetics of Polymeric Nanoparticles reviews

VOL. 5, NO. 4 MOLECULAR PHARMACEUTICS 511

Page 9: Factors Affecting the Clear ance and Biodistribution of ...

Effect of the Core. Shenoy et al.82 investigated thebiodistribution of stealth poly(-amino ester) nanoparticles(PbAE) and poly(caprolactone) (PCL)-based nanoparticleswith a similar size range of 100-200 nm. In addition, bothnanoparticle formulations had a very high positive surfacecharge of approximately +30 mV. The results clearly showeda significantly higher level of accumulation in the heartand lung tissues for the PbAE nanoparticles. This wascorrelated with a lower level of accumulation in the liver ascompared to PCL nanoparticles. It was suggested that ahigher degree of aggregation of PbAE nanoparticles in thepresence of serum proteins affected the biodistributionleading, to such high levels of accumulation in the lung.Recently, greater degrees of nanoparticle “flexibility” werehypothesized to improve the binding ability of particles onthe cell surface.83 Shell cross-linked nanoparticles (SCKs)containing partially hydrochlorinated poly(isoprene) coreswere shown to undergo temperature-dependent deforma-tion.84 SCK nanoparticles with similar physiochemicalproperties (size, ca. 20 nm; , ca. -25 mV) possessing alow glass transition temperature (Tg) with a fluidlike poly-(methyl acrylate) (PMA) core or a high Tg with a glassypoly(styrene) (PS) core were synthesized to evaluate theeffects of the rigidity of the polymeric core on the in vivobiodistribution.84 The results showed that high-Tg poly(sty-rene) core nanoparticles exhibited a significantly higher bloodresidence time compared to the low-Tg poly(methyl acrylate)nanoparticles. The low-Tg core is expected to provide greaterflexibility and an increased number of surface interactionsof the nanoparticles with the tissues and biological environ-ment. However, it was not clear if the relative rigidity orother physicochemical properties of the polymers (hydro-phobicity) affected the blood residence time. The resultssuggested that the composition of the core has an importanteffect on the blood residence time. In this case, PEG surfacemodifications did not have a noticeable effect on kidneyaccumulation and clearance most probably due to the smallsize of the nanoparticle formulations. These results areconsistent with previous work investigating the effect of thecore composition on the nature of absorbed proteins using aseries of block copolymers with the same PEG thickness anddensity.85 Copolymers of poly(ethylene glycol) (PEG) andpolyesters with increased hydrophobicity (PLGA, PLA, andPCL) were synthesized.85 Evidence of C3 cleavage was usedas a semiquantitative way to characterize the level ofcomplement activation by naked and pegylated nanocapsules

(NCs) formed with different core copolymers. The resultsshowed that PLA NCs prepared from biodegradable poly-esters act as strong activators of complement. Since PCLshowed a lower level of complement activation, the reasonfor the stronger complement response did not seem tocorrelate with the hydrophobicity of the copolymer. However,pegylated NCs had significantly reduced levels of comple-ment activation which were shown to depend on PEG chainlength and density. In another study, polymeric nanoparticlescoated with polysorbate were shown to be able to accumulatein the brain tissues.86 The brain blood vessels are character-ized by tight junctions between endothelial cells possessinga TEER (trans-endothelial electrical resistance) of ap-proximately 1500-2000 Ω/cm2 which prevents paracellulartransport of molecules and represents a biological barrier fordelivery of the drug to the brain. However, poly(butylcyanoacrylate) nanoparticles coated with polysorbate 80 wereable to deliver significant amounts of doxorubicin to thebrain. Multiple formulations were studied, and polysorbate80-coated nanoparticles exhibited a high doxorubicin con-centration (6 µg/g) in brain tissues as compared to noncoatednanoparticles (0.1 µg/g). It was found that apolipoprotein E(apoE) binds substantially to polysorbate 80 surfactants, andthe results suggest that apoE is involved in the mediatedtransport of the polysorbate 80-coated nanoparticles to thebrain. Further explanation of the high concentration ofdoxorubicin drug delivered to the brain can be derived fromthe fact that polysorbate can act as an anchor for apoE proteinbinding and interact with LDL receptors expressed in thebrain blood vessels. Polysorbate 80 also has been shown tohave Pgp inhibition properties which could provide anadditional rationale for the delivery of the doxorubicin withnanoparticles having a polysorbate 80 coating.87 Recently,Shaw and Weissleder (in press in Proceedings of the NationalAcademy of Sciences of the United States of America) havefound interesting results regarding the core composition andrelative toxicities of nanoparticles utilizing an in vitro, high-throughput, multidimensional analysis across varying con-centrations utilizing multiple cell types and multiple assaysof cellular physiology. Their generalizable, systematic ap-proach indicates that biological effects result from thecombined effects of many aspects of nanoparticle composi-tion, including the core composition which was found to havea somewhat surprising effect on biological properties. Futurework developing such structure-activity relationships mayshed even more light on the complex interplay betweennanoparticle properties and subsequent biological effects. Insummary, it has been found that physicochemical propertiesof the core are critical parameters of the nanoparticleformulations. Precise modifications of these properties areable to dramatically affect the extent of interactions withblood and biodistribution of nanoparticles.

(83) Takeoka, S.; et al. Rolling properties of rGPIbR-conjugatedphospholipid vesicles with different membrane flexibilities onvWf surface under flow conditions. Biochem. Biophys. Res.Commun. 2002, 296, 765–770.

(84) Sun, X.; et al. An assessment of the effects of shell cross-linkednanoparticle size, core composition, and surface PEGylation onin vivo biodistribution. Biomacromolecules 2005, 6, 2541–2554.

(85) Mosqueira, V. C.; et al. Relationship between complementactivation, cellular uptake and surface physicochemical aspectsof novel PEG-modified nanocapsules. Biomaterials 2001, 22,2967–2979.

(86) Gulyaev, A. E.; et al. Significant transport of doxorubicin intothe brain with polysorbate 80-coated nanoparticles. Pharm. Res.1999, 16, 1564–1569.

(87) Kreuter, J. Nanoparticulate systems for brain delivery of drugs.AdV. Drug DeliVery ReV. 2001, 47, 65–81.

reviews Alexis et al.

512 MOLECULAR PHARMACEUTICS VOL. 5, NO. 4

Page 10: Factors Affecting the Clear ance and Biodistribution of ...

Effect of Surface Functionality and Charge. It has beenestablished that the physicochemical characteristics of apolymeric nanoparticle such as surface charge and functionalgroups can affect its uptake by the cells of the phagocyticsystem. It was previously shown that polystyrene micropar-ticles with a primary amine at the surface underwentsignificantly more phagocytosis as compared to micropar-ticles having sulfate, hydroxyl, and carboxyl groups. There-fore, it is well accepted that positively charged nanoparticleshave a higher rate of cell uptake compared to neutral ornegatively charged formulations. Nanoparticles carrying apositively charged surface are also expected to have a highnonspecific internalization rate and short blood circulationhalf-life. Nanoshells having a negative surface charge haveshown a marked reduction in the rate of uptake. The potential for nanoshells with BSA absorbed on the surfaceof the nanoparticle was characterized by a shift to a morenegative value. However, the BSA absorption did notpromote a higher rate of cell uptake. Interestingly, when thebiodistribution of Tyr- and Tyr-Glu-PEG/PDLLA micelleswas investigated in mice,88 both the Tyr- (neutral) and Tyr-Glu (negatively charged) PEG/PDLLA micelles exhibitedno remarkable difference in their blood clearance kinetics.The pharmacokinetic parameters suggest that all the micelleformulations are distributed mainly into the extracellularspace volume of the spleen and liver. However, the anionicTyr-Glu-PEG/PDLLA micelles showed a lower distribution(10 times lower) into the liver and spleen 4 h post-injection.Remarkably, a lower level of accumulation of Tyr-Glu-PEG/PDLLA micelles into the liver and spleen is believed to bedue to synergic steric and electrostatic repulsion whichdecrease the rate of cellular uptake. In addition, urinaryexcretion was the major excretion route, confirming a lowrate of hepatic uptake of the micelle formulation. Recently,thiolated gel nanoparticles (∼250 nm; ca. -5 mV)60 havehad a higher level of tumor accumulation than nonthiolatednanoparticles, and it was correlated with a very short plasmahalf-life of ∼3 h, indicating a possible preferential uptakeor accumulation in the tumor tissues. In addition, the thiolatednanoparticles were found to have a higher rate of uptakeinto the spleen. The reasons for such an impact of thethiolated functional group on the surface of the nanoparticlesare not very clear, but it could be due to aggregation ofnanoparticles through disulfide bond formation or reactionwith thiolated molecules circulating in the blood. In sum-mary, it is established that neutral or negatively chargedsurface nanoparticles have a reduced plasma protein adsorp-tion and low rate of nonspecific cellular uptake. Thus, surfacefunctionality is another critical parameter in controlling thedevelopment of long-circulating nanoparticles.

Effect of Active Targeting. Active targeting of nanopar-ticles involves the conjugation of targeting ligands to thesurface of nanoparticles. These ligands can include antibod-

ies, engineered antibody fragments, proteins, peptides, smallmolecules, and aptamers. The active targeting mechanismtakes advantage of highly specific interactions between thetargeting ligand and certain tissues or cells within the bodyto promote the accumulation of nanoparticles.3,89–92 In thecase of weak binding ligands, low affinity can be offset byincreased avidity through the surface functionalization ofmultiple molecules or multivalent designs and has beenshown to be a valid approach. There are several examplesof FDA-approved antibodies in clinical practice today,93,94

including Rituxan (target, CD20-positive B-cells for thetreatment of non-Hodgkin’s lymphoma and rheumatoidarthritis), Herceptin (target, HER-2-overexpressing breastcancer cells), Erbitux [target, epidermal growth factorreceptor (EGFR) for the treatment of colorectal cancer],Iressa (target, EGFR for the treatment of non-small cell lungcancer and metastatic breast cancer), and Avastin [target,vascular epidermal growth factor (VEGF) for the treatmentof metastatic colorectal, non-small lung, and breast cancers].While the progress with monoclocal antibodies has beenencouraging, they have not been shown to be curative. Thishas led to the development of immunoconjugates with theintent of utilizing the targeting specificity of the antibody todeliver a relatively potent drug. The development of immu-noconjugates has been slow due to various challenges buthas resulted in the FDA approval of three immunoconju-gates46 such as Mylotarg (conjugate of calicheamicin target-ing CD33 for the treatment of acute myeloid leukemia),Zevalin (yttrium-90 radio-immunotherapy conjugates target-ing CD20-positive B-cells for the treatment of non-Hodgkin’slymphoma), and tositumomab (131I radio-immunotherapyconjugates targeting CD20-positive B-cells for the treatmentof non-Hodgkin’s lymphoma). Currently, drug deliverycarriers are being functionalized with proteins, includingantibodies or antibody fragments and various other targetingligands, with the goal of both delivering a high therapeuticdose and delivering this high therapeutic dose to specifictissues or cells. Conjugation approaches for controlling theamount of targeting proteins on the surface of the nanopar-ticles have been developed to increase specificity and bindingaffinity. Research using proteins for targeting applicationshas led to a better understanding of the effect of stabilityand size of the ligand for successful targeting and clinical

(88) Yamamoto, Y.; Nagasaki, Y.; Kato, Y.; Sugiyama, Y.; Kataoka,K. Long-circulating poly(ethylene glycol)-poly(D,L-lactide) blockcopolymer micelles with modulated surface charge. J. ControlledRelease 2001, 77, 27–38.

(89) Freeman, A. I.; Mayhew, E. Targeted drug delivery. Cancer 1986,58, 573–583.

(90) Emerich, D. F.; Thanos, C. G. Targeted nanoparticle-based drugdelivery and diagnosis. J. Drug Targeting 2007, 15, 163–183.

(91) van Vlerken, L. E.; Vyas, T. K.; Amiji, M. M. Poly(ethyleneglycol)-modified nanocarriers for tumor-targeted and intracellulardelivery. Pharm. Res. 2007, 24, 1405–1414.

(92) Peppas, N. A. Intelligent therapeutics: Biomimetic systems andnanotechnology in drug delivery. AdV. Drug DeliVery ReV. 2004,56, 1529–1531.

(93) Schrama, D.; Reisfeld, R. A.; Becker, J. C. Antibody targeteddrugs as cancer therapeutics. Nat. ReV. Drug DiscoVery 2006,5, 147–159.

(94) Weiner, L. M.; Adams, G. P. New approaches to antibodytherapy. Oncogene 2000, 19, 6144–6151.

Factors Affecting the Pharmacokinetics of Polymeric Nanoparticles reviews

VOL. 5, NO. 4 MOLECULAR PHARMACEUTICS 513

Page 11: Factors Affecting the Clear ance and Biodistribution of ...

development.95,96 Antibody molecules are very large andcomplex and can be expensive to manufacture relative tosmall-molecule drugs. In addition, antibodies have a hydro-dynamic size of ∼20 nm that can increase the size of theNPs. Recently, Davis et al.97 have demonstrated the thera-peutic impact of differential distribution between intracellularand extracellular tumor domains. Cyclodextran-based nano-particles containing transferrin targeting ligand (NP size of∼70 nm) showed enhanced intracellular accumulation in ahuman tumor xenograft mouse model. Interestingly, themodel of tumor accumulation and uptake fit the experimentaldata, suggesting that tumor retention time and internalizationinto tumor cells are the critical factors affecting accumulationof nanoparticles in targeted tissues. The same group98

reported the first toxicology study of a targeted, polymericnanoparticle platform in non-human primates as reportedabove. This type of study is crucial for answering thenumerous critical questions about the toxicological responseto polymeric nanoparticles circulating in the blood. Theresults clearly show that their intravenously injected poly-meric nanoparticles were safely administered to non-humanprimates even after multiple administrations over 3 weeks.These promising results led to the submission of aninvestigational new drug application.

While targeting with proteins has been shown to beadvantageous in some cases, for various reasons, it may bedesirable to achieve targeting with relatively small targetingligands that are potentially easier to manufacture. Thediscovery of new peptide targeting domains99 provides theadvantage of being able to utilize small synthetic moleculesfor active targeting. One such example is peptides whichare relatively stable compared to antibodies and are also lesslikely to be immunogenic. Similarly, small molecules canbe very attractive for use as targeting ligands, and smallmolecules such as folic acid or sugar molecules have beenextensively used. For example, cell surface membrane lectinshave been shown to be overexpressed on the surface ofnumerous cancer cells and are able to specifically internalizesugar molecules (lactose, galactose, and mannose).5 Simi-larly, nucleic acid aptamers are able to fold into unique

structures capable of binding to specific targets with highaffinity and specificity.100 Favorable characteristics of aptam-ers have resulted in their rapid progress into clinicalapplications. Our group is interested in developing this classof molecules for targeted delivery of controlled drug-releasing polymer vehicles.101 We described the first proof-of-concept drug delivery polymeric nanoparticle utilizingaptamers as targeting ligands in vitro102 and subsequentlyshowed the efficacy of similarly designed nanoparticlesagainst prostate cancer tumors in vivo.103 In vivo resultsusing a human prostate cancer tumor xenograft mouse modelshowed a significant therapeutic efficacy of aptamer-targetednanoparticles loaded with docetaxel compared to nontargetedformulations. Targeted nanoparticles substantially reducedthe size of the tumor after a single intratumoral injection,and all of the treated mice survived more than 3 months incontrast to other controls. More recently, we reported a novelstrategy for facile synthesis of targeted nanoparticle formula-tions.104 We have shown that targeted nanoparticles specif-ically accumulated in the prostate tumor xenograft mousemodel (∼1.8%ID/g of tissue) compared to nontargetednanoparticles (0.5%ID/g of tissue). In summary, it has nowbeen shown both in vitro and in vivo that targeted nanopar-ticles are able to agglomerate in specific tissues to improvethe therapeutic efficacy, and there is a growing body ofliterature in this area. Clearly, active targeting provides apowerful approach to increasing the drug delivery load at aspecific site for treatment.

ConclusionThe exact role of each of the proteins adsorbed on the

surface of the nanoparticles in the clearance and biodistri-bution is still not clear besides their role in opsonization andenhanced hepatic uptake. It is generally assumed that therapid uptake of injected nanoparticles is triggered by recep-tor-mediated mechanisms of absorbed proteins from theblood (opsonins) onto their surface and complement activa-tion. However, it is not clear if a specific type, a combinationof proteins, or even the protein conformation is the mostimportant factor for a high rate of phagocytotic uptake. Theefficacy of the PEG “brush” in altering the biodistributionof nanoparticles has been clearly demonstrated, and in vivostudies showed a drastic increase in blood circulation time

(95) Clauss, M. A.; Jain, R. K. Interstitial transport of rabbit and sheepantibodies in normal and neoplastic tissues. Cancer Res. 1990,50, 3487–3492.

(96) Graff, C. P.; Wittrup, K. D. Theoretical analysis of antibodytargeting of tumor spheroids: Importance of dosage for penetra-tion, and affinity for retention. Cancer Res. 2003, 63, 1288–1296.

(97) Bartlett, D. W.; Su, H.; Hildebrandt, I. J.; Weber, W. A.; Davis,M. E. Impact of tumor-specific targeting on the biodistributionand efficacy of siRNA nanoparticles measured by multimodalityin vivo imaging. Proc. Natl. Acad. Sci. U.S.A. 2007, 104, 15549–15554.

(98) Heidel, J. D.; et al. Administration in non-human primates ofescalating intravenous doses of targeted nanoparticles containingribonucleotide reductase subunit M2 siRNA. Proc. Natl. Acad.Sci. U.S.A. 2007, 104, 5715–5721.

(99) Scott, J. K.; Smith, G. P. Searching for peptide ligands with anepitope library. Science 1990, 249, 386–390.

(100) Gopinath, S. C. Methods developed for SELEX. Anal. Bioanal.Chem. 2007, 387, 171–182.

(101) Cheng, J.; et al. Formulation of functionalized PLGA-PEGnanoparticles for in vivo targeted drug delivery. Biomaterials2007, 28, 869–876.

(102) Farokhzad, O. C.; et al. Nanoparticle-aptamer bioconjugates: Anew approach for targeting prostate cancer cells. Cancer Res.2004, 64, 7668–7672.

(103) Farokhzad, O. C.; et al. Targeted nanoparticle-aptamer biocon-jugates for cancer chemotherapy in vivo. Proc. Natl. Acad. Sci.U.S.A. 2006, 103, 6315–6320.

(104) Gu, F.; et al. Precise engineering of targeted nanoparticles byusing self-assembled biointegrated block copolymers. Proc. Natl.Acad. Sci. U.S.A. 2008, 105, 2586–2591.

reviews Alexis et al.

514 MOLECULAR PHARMACEUTICS VOL. 5, NO. 4

Page 12: Factors Affecting the Clear ance and Biodistribution of ...

with an increase in PEG surface density. It is also clear thatrelatively small physicochemical differences have significantbiological implications in the fate of the biodistribution ofnanoparticles. Pegylated nanoparticles between 10 and 100nm in size are able to remain in the systemic circulation forhours and extravasate or diffuse into the diseased tissues bya passive targeting mechanism. Recently, targeted nanopar-ticles funtionalized with ligands that have high affinity andspecificity have been shown to efficiently accumulate inspecific tissues and dramatically increase the therapeuticefficacy of long-circulating nanoparticle drug delivery sys-tems. Natural and synthetic polymers are now in preclinical

and clinical phases for drug delivery. More importantly,targeted and nontargeted polymeric nanoparticles are nowin the preclinical and clinical phases and confirm the greatpromise of the past 20 years of research and lessons learnedfrom the failure of some clinical studies to increase thetherapeutic index of drugs approved for clinical use.

Acknowledgment. This work was supported byNational Institutes of Health Grants CA119349 andEB003647 and a Koch-Prostate Cancer FoundationAward in Nanotherapeutics.

MP800051M

Factors Affecting the Pharmacokinetics of Polymeric Nanoparticles reviews

VOL. 5, NO. 4 MOLECULAR PHARMACEUTICS 515


Recommended