Precise engineering of targeted nanoparticles by using ...NPs by macromolecular self-assembly in one...

6
Precise engineering of targeted nanoparticles by using self-assembled biointegrated block copolymers Frank Gu* , Liangfang Zhang*, Benjamin A. Teply* , Nina Mann*, Andrew Wang †‡ , Aleksandar F. Radovic-Moreno* , Robert Langer* †§ , and Omid C. Farokhzad †‡§ *Department of Chemical Engineering, Harvard–MIT Center of Cancer Nanotechnology Excellence, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139; and Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115 Contributed by Robert Langer, December 17, 2007 (sent for review December 3, 2007) There has been progressively heightened interest in the develop- ment of targeted nanoparticles (NPs) for differential delivery and controlled release of drugs. Despite nearly three decades of re- search, approaches to reproducibly formulate targeted NPs with the optimal biophysicochemical properties have remained elusive. A central challenge has been defining the optimal interplay of parameters that confer molecular targeting, immune evasion, and drug release to overcome the physiological barriers in vivo. Here, we report a strategy for narrowly changing the biophysicochemical properties of NPs in a reproducible manner, thereby enabling systematic screening of optimally formulated drug-encapsulated targeted NPs. NPs were formulated by the self-assembly of an amphiphilic triblock copolymer composed of end-to-end linkage of poly(lactic-co-glycolic-acid) (PLGA), polyethyleneglycol (PEG), and the A10 aptamer (Apt), which binds to the prostate-specific mem- brane antigen (PSMA) on the surface of prostate cancer (PCa) cells, enabling, respectively, controlled drug release, ‘‘stealth’’ proper- ties for immune evasion, and cell-specific targeting. Fine-tuning of NP size and drug release kinetics was further accomplished by controlling the copolymer composition. By using distinct ratios of PLGA-b-PEG-b-Apt triblock copolymer with PLGA-b-PEG diblock copolymer lacking the A10 Apt, we developed a series of targeted NPs with increasing Apt densities that inversely affected the amount of PEG exposure on NP surface and identified the narrow range of Apt density when the NPs were maximally targeted and maximally stealth, resulting in most efficient PCa cell uptake in vitro and in vivo. This approach may contribute to further devel- opment of targeted NPs as highly selective and effective thera- peutic modalities. aptamer controlled release chemotherapy combinatorial screening T he development of polymeric nanoparticles (NPs) for tar- geted delivery and controlled drug release may improve the therapeutic index of drugs. Such improvement is particularly important when administering chemotherapeutics that have toxicities that often limit their dose, resulting in suboptimal efficacy (1– 4). The potential of drug targeting was first suggested by the visionary Paul Ehrlich in 1906 and despite significant work over many years (1, 3, 5–8), targeted NPs have yet to make an impact on human health. A central challenge has been defining the optimal interplay of biophysicochemical parameters that confer molecular targeting, immune evasion, and drug release to overcome the physiological barriers in vivo. The challenge is confounded by the fact that the parameters are interrelated and improving one may adversely impact another, and thus the narrow window where each of these are in the optimal range and working in concert has been difficult to achieve. The conven- tional methods of synthesizing targeted NPs involves serial chemical processing of particles during the formulation process, whereby drug-encapsulated NPs are first formed, followed by surface functionalization, which may include the conjugation of biomaterials to render the NPs ‘‘stealth’’ by decreasing their nonspecific immune clearance and biofouling by plasma pro- teins, and the attachments of targeting ligands to direct the cell- or tissue-specific delivery of NPs. In some cases the chemical processing of NPs has been done with functionalized biomate- rials that simultaneously render the NPs both stealth and tar- geted (9–11), or alternatively self-assembled stealth NPs have been conjugated with a targeting ligands to achieve targeted NPs (12, 13). Regardless of the strategies taken, the methods for postsynthesis NP surface modification often require the addition of an excess amount of reactants to drive the chemical reaction (14), thus making it difficult to adjust the properties of NP surface in a meaningfully reproducible manner. Consequently, the postsynthesis particle-processing methods offer limited abil- ity to precisely engineer the NP surface properties, and the targeted NPs produced by such methods may have significant batch-to-batch variations in their biophysicochemical properties (2, 4, 14). The development of prefunctionalized biomaterials that have all of the desired NP components present, and engineering these biomaterials to self-assemble into a targeted NP would eliminate the need of postparticle modification. Furthermore, the use of prefunctionalized biomaterials for the self-assembly of NPs could result in precisely engineered NPs, whereas simpler conjugation and purification procedures are amenable to scale-up with little batch-to-batch variability (15– 17). In addition, the ability to precisely engineer targeted NPs may enable the formulation of distinct NPs that narrowly vary from each other, making it possible to optimize their biophysi- cochemical properties. In this study, we used the A10 2-fluoropyrimidine RNA aptamer (Apt) (18), which binds to the prostate-specific mem- brane antigen (PSMA) on the surface of prostate cancer (PCa) cells as a model hydrophilic targeting molecule; the poly(D,L- lactide-co-glycolide) (PLGA) as a model controlled-release polymer system; and polyethylene glycol (PEG) as a model hydrophilic polymer with antibiofouling properties, to develop a proof-of-concept biointegrated amphiphilic triblock copolymer (TCP) composed of PLGA-b-PEG-b-Apt that forms targeted NPs by macromolecular self-assembly in one single step. Using distinct ratios of PLGA-b-PEG-b-Apt TCP with PLGA-b-PEG diblock copolymer (DCP) lacking the A10 PSMA Apt, we developed NPs with varying Apt surface density and identified the narrow range of Apt density for maximum PCa cell uptake in vitro and in vivo. This narrow range was determined experi- mentally to be the minimum amount of Apt on the NP surface that conferred the maximal targeted cellular uptake. By avoiding the unnecessary masking of PEG on NP surface by excess Apt, Author contributions: F.G., B.A.T., R.L., and O.C.F. designed research; F.G., L.Z., B.A.T., N.M., A.W., and A.F.R.-M. performed research; F.G. contributed new reagents/analytic tools; F.G., L.Z., B.A.T., and O.C.F. analyzed data; and F.G. and O.C.F. wrote the paper. Conflict of interest statement: O.C.F. and R.L. have financial interest in BIND Biosciences. The rest of the authors declare no conflict of interest. § To whom correspondence may be addressed. E-mail: [email protected] or [email protected]. © 2008 by The National Academy of Sciences of the USA 2586 –2591 PNAS February 19, 2008 vol. 105 no. 7 www.pnas.orgcgidoi10.1073pnas.0711714105 Downloaded by guest on September 1, 2020

Transcript of Precise engineering of targeted nanoparticles by using ...NPs by macromolecular self-assembly in one...

Page 1: Precise engineering of targeted nanoparticles by using ...NPs by macromolecular self-assembly in one single step. Using distinct ratios of PLGA-b-PEG-b-Apt TCP with PLGA-b-PEG diblock

Precise engineering of targeted nanoparticles byusing self-assembled biointegrated block copolymersFrank Gu*†, Liangfang Zhang*, Benjamin A. Teply*‡, Nina Mann*, Andrew Wang†‡, Aleksandar F. Radovic-Moreno*†,Robert Langer*†§, and Omid C. Farokhzad†‡§

*Department of Chemical Engineering, †Harvard–MIT Center of Cancer Nanotechnology Excellence, Massachusetts Institute of Technology,77 Massachusetts Avenue, Cambridge, MA 02139; and ‡Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology,Perioperative and Pain Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115

Contributed by Robert Langer, December 17, 2007 (sent for review December 3, 2007)

There has been progressively heightened interest in the develop-ment of targeted nanoparticles (NPs) for differential delivery andcontrolled release of drugs. Despite nearly three decades of re-search, approaches to reproducibly formulate targeted NPs withthe optimal biophysicochemical properties have remained elusive.A central challenge has been defining the optimal interplay ofparameters that confer molecular targeting, immune evasion, anddrug release to overcome the physiological barriers in vivo. Here,we report a strategy for narrowly changing the biophysicochemicalproperties of NPs in a reproducible manner, thereby enablingsystematic screening of optimally formulated drug-encapsulatedtargeted NPs. NPs were formulated by the self-assembly of anamphiphilic triblock copolymer composed of end-to-end linkage ofpoly(lactic-co-glycolic-acid) (PLGA), polyethyleneglycol (PEG), andthe A10 aptamer (Apt), which binds to the prostate-specific mem-brane antigen (PSMA) on the surface of prostate cancer (PCa) cells,enabling, respectively, controlled drug release, ‘‘stealth’’ proper-ties for immune evasion, and cell-specific targeting. Fine-tuning ofNP size and drug release kinetics was further accomplished bycontrolling the copolymer composition. By using distinct ratios ofPLGA-b-PEG-b-Apt triblock copolymer with PLGA-b-PEG diblockcopolymer lacking the A10 Apt, we developed a series of targetedNPs with increasing Apt densities that inversely affected theamount of PEG exposure on NP surface and identified the narrowrange of Apt density when the NPs were maximally targeted andmaximally stealth, resulting in most efficient PCa cell uptake invitro and in vivo. This approach may contribute to further devel-opment of targeted NPs as highly selective and effective thera-peutic modalities.

aptamer � controlled release � chemotherapy � combinatorial screening

The development of polymeric nanoparticles (NPs) for tar-geted delivery and controlled drug release may improve the

therapeutic index of drugs. Such improvement is particularlyimportant when administering chemotherapeutics that havetoxicities that often limit their dose, resulting in suboptimalefficacy (1–4). The potential of drug targeting was first suggestedby the visionary Paul Ehrlich in 1906 and despite significant workover many years (1, 3, 5–8), targeted NPs have yet to make animpact on human health. A central challenge has been definingthe optimal interplay of biophysicochemical parameters thatconfer molecular targeting, immune evasion, and drug release toovercome the physiological barriers in vivo. The challenge isconfounded by the fact that the parameters are interrelated andimproving one may adversely impact another, and thus thenarrow window where each of these are in the optimal range andworking in concert has been difficult to achieve. The conven-tional methods of synthesizing targeted NPs involves serialchemical processing of particles during the formulation process,whereby drug-encapsulated NPs are first formed, followed bysurface functionalization, which may include the conjugation ofbiomaterials to render the NPs ‘‘stealth’’ by decreasing theirnonspecific immune clearance and biofouling by plasma pro-

teins, and the attachments of targeting ligands to direct the cell-or tissue-specific delivery of NPs. In some cases the chemicalprocessing of NPs has been done with functionalized biomate-rials that simultaneously render the NPs both stealth and tar-geted (9–11), or alternatively self-assembled stealth NPs havebeen conjugated with a targeting ligands to achieve targeted NPs(12, 13). Regardless of the strategies taken, the methods forpostsynthesis NP surface modification often require the additionof an excess amount of reactants to drive the chemical reaction(14), thus making it difficult to adjust the properties of NPsurface in a meaningfully reproducible manner. Consequently,the postsynthesis particle-processing methods offer limited abil-ity to precisely engineer the NP surface properties, and thetargeted NPs produced by such methods may have significantbatch-to-batch variations in their biophysicochemical properties(2, 4, 14). The development of prefunctionalized biomaterialsthat have all of the desired NP components present, andengineering these biomaterials to self-assemble into a targetedNP would eliminate the need of postparticle modification.Furthermore, the use of prefunctionalized biomaterials for theself-assembly of NPs could result in precisely engineered NPs,whereas simpler conjugation and purification procedures areamenable to scale-up with little batch-to-batch variability (15–17). In addition, the ability to precisely engineer targeted NPsmay enable the formulation of distinct NPs that narrowly varyfrom each other, making it possible to optimize their biophysi-cochemical properties.

In this study, we used the A10 2�-f luoropyrimidine RNAaptamer (Apt) (18), which binds to the prostate-specific mem-brane antigen (PSMA) on the surface of prostate cancer (PCa)cells as a model hydrophilic targeting molecule; the poly(D,L-lactide-co-glycolide) (PLGA) as a model controlled-releasepolymer system; and polyethylene glycol (PEG) as a modelhydrophilic polymer with antibiofouling properties, to develop aproof-of-concept biointegrated amphiphilic triblock copolymer(TCP) composed of PLGA-b-PEG-b-Apt that forms targetedNPs by macromolecular self-assembly in one single step. Usingdistinct ratios of PLGA-b-PEG-b-Apt TCP with PLGA-b-PEGdiblock copolymer (DCP) lacking the A10 PSMA Apt, wedeveloped NPs with varying Apt surface density and identifiedthe narrow range of Apt density for maximum PCa cell uptakein vitro and in vivo. This narrow range was determined experi-mentally to be the minimum amount of Apt on the NP surfacethat conferred the maximal targeted cellular uptake. By avoidingthe unnecessary masking of PEG on NP surface by excess Apt,

Author contributions: F.G., B.A.T., R.L., and O.C.F. designed research; F.G., L.Z., B.A.T., N.M.,A.W., and A.F.R.-M. performed research; F.G. contributed new reagents/analytic tools; F.G.,L.Z., B.A.T., and O.C.F. analyzed data; and F.G. and O.C.F. wrote the paper.

Conflict of interest statement: O.C.F. and R.L. have financial interest in BIND Biosciences.The rest of the authors declare no conflict of interest.

§To whom correspondence may be addressed. E-mail: [email protected] [email protected].

© 2008 by The National Academy of Sciences of the USA

2586–2591 � PNAS � February 19, 2008 � vol. 105 � no. 7 www.pnas.org�cgi�doi�10.1073�pnas.0711714105

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

1, 2

020

Page 2: Precise engineering of targeted nanoparticles by using ...NPs by macromolecular self-assembly in one single step. Using distinct ratios of PLGA-b-PEG-b-Apt TCP with PLGA-b-PEG diblock

which did not confer additional targeting benefit, we were ableto maximize the exposure of PEG and its antibiofouling prop-erties, resulting in maximally stealth and maximally targetedNPs. The delicate balance between each distinct biophysico-chemical property of NPs needs to be experimentally deter-mined and precisely and reproducibly engineered for ultimate invivo success.

Results and DiscussionsDevelopment of PLGA-b-PEG-b-Apt TCP for Self-Assembly of TargetedNPs. To engineer biomaterials that can self-assemble into atargeted NP, we developed a biointegrated block copolymerconsisting of the following three polymer blocks: PLGA, PEG,and the A10 2�-f luoropyrimidine RNA Apt (18). The PLGApolymer block provides a biodegradable matrix for the encap-sulation and sustained release of therapeutic agents and the PEGpolymer acts as the antibiofouling coating on the NP surface.Both PLGA and PEG have been approved by the Food and DrugAdministration for a number of clinically used products. TheA10 RNA Apt (18) that binds to the extracellular domain of thePSMA (18–22) directs the delivery and uptake of NPs in acell-specific manner. We postulated that a nucleic acid compo-sition of Apt would be uniquely suited for the development toour TCP to be used for NP self-assembly, because it wouldremain stable in organic solvents during polymer synthesis andNP formulation. The PLGA-b-PEG-b-Apt TCP was synthesizedby using a two-step reaction (Fig. 1A): first, the carboxyl-cappedPLGA was conjugated to the amine terminal of heterobifunc-tional PEG (amine-PEG-carboxyl), forming the PLGA-b-PEGDCP; second, the carboxyl end of PEG-b-PLGA DCP wasconjugated to the amine functional group of the A10 PSMA Apt,forming the PLGA-b-PEG-b-Apt TCP. The NMR characteriza-

tion of PLGA, PLGA-PEG DCP, and PLGA-b-PEG-b-Apt TCPis shown in Fig. 1B. By precipitating the PLGA-b-PEG-b-AptTCP in water, the polymer self-assembles to form PSMA-targeted polymeric NPs without postparticle conjugation steps:the hydrophobic PLGA blocks form a core to minimize theirexposure to aqueous surroundings, the hydrophilic PEG blocksform a corona-like shell to stabilize the core, and the hydrophilicApt blocks thrust into aqueous solution on NP surface as thetargeting moieties (Fig. 1C).

Effects of Copolymer Composition on NP Biophysicochemical Proper-ties. We next demonstrated that by combinatorially varying theindividual components of the TCP we can systematically changeof the following NP biophysicochemical properties: (i) NP size,(ii) drug release kinetics, and (iii) differential targeting.

The optimization of the NP size was performed by varying thecomposition of the TCP. A series of targeted PLGA-b-PEG-b-Apt NPs were formulated by using combinations of variousmolecular masses of PLGA and PEG (Fig. 2A). Previous workon PLGA-PEG DCP showed that NP size depended on size ofthe PEG polymer (23, 24). We sought to determine whethervarying the length of the PEG segment in the TCP could controlthe NP-Apt bioconjugate size. We found that, indeed, themolecular mass of PEG but not PLGA was a key factor incontrolling the NP size for the triblock system. By shortening thePEG segment on the TCP from 10,000 Da to 5,000 Da, thediameter of NP reduced from 291 � 5.2 nm to 160 � 3.7 nm(Fig. 2 A).

Using docetaxel (Dtxl) as a model drug, we varied the intrinsicPLGA viscosity to control drug release kinetics. We determinedthat increasing the length of the PLGA segment of the PLGA-b-PEG-b-Apt TCP prolonged the rate of drug release in vitro.

B C

Docetaxel

1

10

1

20

1

40

tios Mixin ra g

O O

O COOH

O

O CH 3

O O N

H

COOH

PLGA-Acid

Amine-PEG-Acid

O O

O

O

O CH 3

O

O O H 2 N

COOH PLGA- PEG-Acid

A-10 Aptamer-Amine

O O N H

O O

O O

O CH 3

O

H 2 N

O

N H

PLGA- PEG- Aptamer

8 7 6 5 4 3 2 1 0

P L G A - P E G

P L G A

P P M

P L G A - P E G - A p t

A

Fig. 1. Development of PSMA-targeted NPs by using PLGA-b-PEG-b-Apt TCP. (A) The PLGA-b-PEG-b-Apt-biointegrated TCP was synthesized in two steps: (i)synthesis of PLGA-b-PEG by conjugating carboxyl-capped PLGA (PLGA-acid) to the amine terminals of heterobifunctional PEG (amine-PEG-acid) and (ii) formationof PLGA-b-PEG-b-Apt by conjugating the carboxyl ends of PLGA-b-PEG-acid to the amine ends of A10 PSMA Apt. (B) 1H NMR characterization of PLGA-b-PEGand PLGA-b-PEG-b-Apt. For the synthesis of PLGA-b-PEG, the yield of PLGA and PEG conjugation was 73–91% and the purified PLGA-b-PEG DCP was used forthe subsequent conjugation to Apt. The presence of Apt on the PLGA-b-PEG-b-Apt TCP was visualized by the peaks between 1.8 and 2.2 ppm. The Apt conjugationefficiency of the PLGA-b-PEG DCP for seven independent reactions was 13–21%. (C) By titration in water, the PLGA-b-PEG-aptamer TCPs self-assemble and formPSMA-targeted NP-Apt bioconjugates. By using distinct ratios of PLGA-b-PEG-b-Apt TCP with PLGA-b-PEG DCP lacking the A10 Apt during NP formulation, theApt surface density can be precisely and reproducibly changed.

Gu et al. PNAS � February 19, 2008 � vol. 105 � no. 7 � 2587

ENG

INEE

RIN

GM

EDIC

AL

SCIE

NCE

S

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

1, 2

020

Page 3: Precise engineering of targeted nanoparticles by using ...NPs by macromolecular self-assembly in one single step. Using distinct ratios of PLGA-b-PEG-b-Apt TCP with PLGA-b-PEG diblock

The length of PEG chain had a minimal effect on the drugrelease properties of NP-Apt bioconjugates. By using PLGAwith an intrinsic viscosity of �0.67 g/dl in hexafluoroisopropanol(�100,000 molecular mass), Dtxl was released at a sustained ratefor 3 days (Fig. 2B).

We next evaluated the bioactivities of the PSMA Apt ex-

pressed on the NP surface. After the TCP self-assembles intotargeted NPs, the Apt on the particle surface can be heatdenatured and slowly cooled to allow proper Apt tertiaryconformation necessary for PSMA binding and differentialtargeting. We aimed to confirm that the Apt on NP surfacemaintain their bioactivity after the chemical steps of triblockformation and Dtxl encapsulation, by quantifying the differentialaccumulation of Dtxl-encapsulated NP-Apt bioconjugateswithin PCa cells that express the PSMA (LNCaP) and controlcells lacking PSMA (PC3). Tritium-labeled NPs were formulatedby mixing PLGA-b-PEG-b-Apt with a trace amount of tritium-labeled PLGA (3H-PLGA). The percentage of NP-Apt biocon-jugates accumulated in LNCaP and PC3 cells was determined byquantifying the 3H detected in the cells. NPs were formulated byusing two types of TCPs: (i) PLGA-b-PEG-b-Apt by using PSMAApt (NP-Apt) and (ii) PLGA-b-PEG-b-MutApt by using ascrambled nonfunctional aptamer (NP-MutApt). LNCaP cellsretained �30% of NPs at 30 min irrespective of the PLGA andPEG composition (Fig. 2C). The NP-MutApt incubated withLNCaP cells and NP-Apt incubated with PC3 cells demonstratedminimal NP accumulation. These results suggested that the Aptexpressed on the Dtxl-encapsulated NP-Apt bioconjugates werebioactive and were capable of differentially guiding the NPs toaccumulate in PSMA� LNCaP cells.

Cell-Specific Endocytosis and Endosomal Colocalization of TargetedNPs. Our data suggested that PLGA0.67-b-PEG3400-b-Apt TCPhad the most desirable combination of small particle size,sustained drug release kinetics, and differential accumulation inLNCaP cells. Next, LNCaP and PC3 cells were used to examinewhether the self-assembled NP-Apt bioconjugates were differ-entially endocytosed by LNCaP cells. NP-Apt bioconjugateswere visualized by encapsulating a green fluorescent dye, (22-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-23,24-bisnor-5-cholen-3�-ol) cholesterol (NBD-cholesterol). By using z-axisscanning fluorescence microscopy and colocalization staining,both early and late endosomal markers were colocalized withNPs in LNCaP cells, confirming a relatively rapid endocytosis ofNP-Apt bioconjugates by LNCaP cells (Fig. 3). In comparison,the NP-Apt bioconjugates seeded onto PC3 cells showed mini-mal cellular uptake consistent with the lack of PSMA expressionin these cells.

Precise Control of Apt Density on NP Surface. Once we confirmedthat NP-Apt bioconjugates resulting from the PLGA0.67-b-PEG3400-b-Apt TCP were effectively endocytosed by PSMA�LNCaP cells, we proceeded to optimize the Apt density on theNP surface. We postulated that the Apt density on the NPsurface can be narrowly controlled by mixing known quantitiesof Apt in the form of PLGA0.67-b-PEG3400-b-Apt TCPs withvarious molar ratios of PLGA0.67-b-PEG3400 DCPs lacking the

0

20

PE

G 2

000

PE

G 3

400

PE

G 5

000

PE

G 2

000

PE

G 3

400

PE

G 5

000

0

20

40

60

PLGA IV (0.67 g/dL)

NP-Apt

LNCaP Cells

% N

P a

ccum

ulat

ion PC3 Cells

PLGA IV (0.19 g/dL)

Oligo-NP PC3

0.20.3

0.40.5

0.60.7

140

160

180

200

220

240

260

280

20004000

60008000

10000

NP

dia

met

er (

nm)

PEG MW

(g/m

ol)PLGA viscosity (g/dL)

A

B

C

Fig. 2. Fine-tuning the nanoparticle physiochemical properties by usingPLGA-b-PEG-b-Apt TCPs. (A) Effect of TCP composition on NP size. Synthesis ofTCP containing different molecular mass segments of PLGA and PEG. TargetedNPs were formulated by TCP self-assembly. The size of each NP formulationwas measured by dynamic light scattering. Each data point represents theaverage of four experiments (n � 4). The standard deviation varied between1 and 6%. The range of nanoparticle size polydispersity index (PDI) wasbetween 0.05 and 0.10 and that the of zeta potential of various nanoparticlesformulations was between �20 and �29. (B) NP drug release properties as afunction of PLGA-b-PEG-b-Apt TCP composition. Dtxl was encapsulated intovarious NPs at a mass loading of 1.5 wt/wt%. Each data point represents theaverage of four experiments (n � 4). (C) Quantification on the percentage ofNP accumulation in LNCaP and PC3 cells. NP-Apt bioconjugates were formu-lated by self-assembly of the PLGA-b-PEG-b-Apt TCP containing differentlengths of PLGA and PEG segments. LNCaP and PC3 cells were incubated with50 �g of 3H-labeled PLGA-b-PEG-b-Apt NPs for 30 min. The amount of NPsendocytosed was determined by 3H radiation counts collected in the cells. Eachdata point represents the average of four experiments (n � 4).

Fig. 3. NP-Apt cellular uptake. The NBD dyes (green) were formulated intoPLGA-PEG-aptamer triblock nanoparticles by nanoprecipitation. LNCaP(PSMA�) and PC3 (PSMA�) cells were incubated with 50 �g of NBD-encapsulated PLGA-PEG-aptamer nanoparticles for 30 min. The early and lateendosomal markers were visualized in red. The cell nuclei were stained byDAPI (blue).

2588 � www.pnas.org�cgi�doi�10.1073�pnas.0711714105 Gu et al.

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

1, 2

020

Page 4: Precise engineering of targeted nanoparticles by using ...NPs by macromolecular self-assembly in one single step. Using distinct ratios of PLGA-b-PEG-b-Apt TCP with PLGA-b-PEG diblock

A10 Apt (Fig. 1C). The Apt densities on the NP surface weredetermined by Apt cleavage assay and nucleic acid quantificationafter hydrolyzing the Apt from the NP surface. Because PLGAis also prone to alkaline hydrolysis, the NPs were constantlymonitored to ensure that they remained stable during the Aptcleavage assay. We determined that the Apt density can beprecisely fine-tuned by controlling the mixing ratios of PLGA-b-PEG-b-Apt TCP to PLGA-b-PEG DCPs lacking the A10 Apt(Fig. 4A). For example, by achieving a molar ratio of 5:95 ofTCP/DCP (NP-Apt 5%) in the formulation process, we canreproducibly achieve an Apt surface density of 5.31 � 0.233 �gof Apt per mg of NP. By changing the triblock/diblock ratio from5:95 (NP-Apt 5%) to 1:99 (NP-Apt 1%), the Apt density on NPsurface is reproducibly and linearly reduced to 0.225 � 0.0691 �gof Apt per mg of NPs. Because the average particle size was 160nm, and the density of PLGA-PEG NP core was estimated at1.27 g/ml (25) we estimated the Apt ligand density for NP-Apt0.5% formulation to be 2.59 � 0.79 Apt per NP. By increasingthe TCP composition to 2%, 5%, and 10%, the Apt liganddensity increased to 21.7 � 4.69, 61.0 � 2.67, and 139.4 � 8.41,Apt per NPs, respectively (Fig. 4A).

Determination of Optimal Apt Density on NP Surface in Vitro and inVivo. We next determined the optimal Apt density on NP surfacein vitro. The optimal ligand density was defined as the minimum

amount of Apt on the NP surface to confer maximal targetedcellular uptake. We believed that by avoiding the unnecessarymasking of PEG on NP surface by excess Apt that did not conferadditional targeting benefit, we could maximize the exposure ofPEG to retain its antibiofouling properties, resulting in maxi-mally stealth and maximally targeted NPs. Our group and othershad shown that PEG can increase the antibiofouling propertiesof NPs in vivo resulting in long circulating NPs for systemicdelivery (24, 26, 27). NPs containing different Apt liganddensities were synthesized by controlling the mixing ratios ofPLGA0.67-b-PEG3400-b-Apt to PLGA0.67-PEG3400. The experi-mental control groups were: nontargeted NPs made ofPLGA0.67-PEG3400 DCPs lacking the A10 Apt (NP), and NPsmade of mutated Apt TCPs (NP-MutApt). NPs were radiola-beled with 3H-PLGA, and the amount of NPs endocytosed byLNCaP and PC3 cells was quantified by measuring the 3H in thecells. All NP-Apt bioconjugates regardless of the Apt surfacedensities consistently demonstrated minimal uptake by the PC3cells that do not express PSMA (Fig. 4B). In contrast, the amountof NPs endocytosed by LNCaP cells that express the PSMA canbe precisely controlled by adjusting the Apt ligand densities. NPscontaining �0.05% of Apt TCPs (NP-Apt 0.05%) had virtuallyno uptake by the LNCaP cells (P � 0.05). There was a significantincrease in NP-Apt bioconjugate uptake by the LNCaP cellswhen the NPs contain �0.1% PLGA-b-PEG-b-Apt TCP (P �0.05). By increasing the PLGA-b-PEG-b-Apt TCP in the for-mulation to 5%, there was a 5-fold increase in the amount of NPuptake by the LNCaP cells compared with the NP-Apt 0.05%formulation. Any further increase in Apt triblock proportion inthe NP resulted in a modest increase in NP uptake (Fig. 4B).Thus, we estimated the optimum ligand density for PSMA-specific endocytosis in vitro was in the range of 10 to 80 nmol ofApt per �mol of NPs.

After determining the optimal Apt density for targeted celluptake in vitro, we correlated this finding to tumor targeting invivo by using a LNCaP s.c. xenograft mouse models of PCa toconfirm whether a narrow window exists for when a PCa-targeted NP is both maximally targeted and maximally stealth.Comparative biodistribution studies were performed by usingsimilar 3H-labeled NP-Apt bioconjugates that were used in thein vitro studies: NP-Apt 10%, NP-Apt 5%, NP-Apt 1%, NP-Apt0%, and NP-MutApt 10% (Fig. 5 A and B). Systemic adminis-tration of targeted NPs showed that increasing the Apt surfacedensity from 0% (no Apt) to 1% significantly increased NPretention in tumors from 0.576 � 0.116% of injected dose per g(IDPG) of tissue to 1.09 � 0.293% of IDPG (P � 0.05). Themaximal tumor targeting was achieved by using the NP-Apt 5%formulation at 1.82 � 0.421% of IDPG (P � 0.05). As the Apttriblock formulation in the NPs reached �10%, there was adecrease in NP accumulation in the tumors. The decrease intumor accumulation for the NP-Apt 10% (0.743 � 0.282% ofIDPG) is possibly caused by diminishing NP stealth properties bythe increased Apt surface density, resulting in its rapid clearanceby the liver. Interestingly, the NP-MutApt 10% control groupaccumulated less in the tumor compared with the NP-Apt 0%control group, consistent with the hypothesis that former par-ticles are less stealth and nontargeted, whereas the NP-Apt 0%particles are maximally stealth and nontargeted NPs, and thuscan reach the tumor by passive targeting alone.

In addition to affecting the particle accumulation in the tumortissues, the ligand surface density was directly proportional to therate of NP accumulation in the liver. Both NP-Apt 10% (68.1 �4.56% of IDPG) and NP-MutApt 10% (73.0 � 3.89% of IDPG)formulations showed significantly higher particle accumulationin the liver compared with NPs with a lower Apt surface density(P � 0.05). By decreasing the TCP to 5% (NP-Apt 5%), 37.7 �3.85% of IDPG was retained in the liver. The NP-Apt 1%formulation had 33.9 � 4.21% of IDPG accumulations in the

0 0.54 1.3 4.8 12.7 35.6 79.8

0% 0.05% 0.1% 0.5% 2% 5% 10%0

20

40

60

80

100

PC3 cells

2 hours

nmol of aptamers per µmol NP

% N

P a

ccum

ulat

ion

Apt

Lig

and

Den

sity

nmol

Apt

per

um

ol N

P

% of aptamer triblock in NP

30 minutes

LNCaP cells

0

20

B

A

0 2 4 6 8 100

2

4

6

8

10

12

14

0

20

40

60

80A

ptam

er m

ass

on N

P s

urfa

ceµg

of A

ptam

er p

er m

g of

NP

% of aptamer triblock in NP composition

Fig. 4. Fine-tuning the Apt ligand density for targeted cell uptake in vitro.(A) Estimation of Apt ligand density on NP surface. The aptamer density canbe precisely controlled in a linear manner by varying the ratio of PLGA0.67-b-PEG3400-b-Apt and PLGA0.67-b-PEG3400. (B) NPs containing 0.05–10% ofPLGA0.67-PEG3400-Apt were synthesized by mixing with different amounts ofPLGA0.67-PEG3400. LNCaP and PC3 cells were incubated with 3H-labeled PLGA-PEG-Apt NPs for 30 min (black) and 2 h (white). Data represent the mean andthe standard error of mean of triplicates per group. Data points for NP-Apt0.1–10% were statistically significant compared with NP-Apt 0% by t test at95% confidence interval. One-way ANOVA with Fisher’s least significantdifference (LSD) post hoc comparisons at 95% confidence interval was used forstatistical comparisons between groups.

Gu et al. PNAS � February 19, 2008 � vol. 105 � no. 7 � 2589

ENG

INEE

RIN

GM

EDIC

AL

SCIE

NCE

S

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

1, 2

020

Page 5: Precise engineering of targeted nanoparticles by using ...NPs by macromolecular self-assembly in one single step. Using distinct ratios of PLGA-b-PEG-b-Apt TCP with PLGA-b-PEG diblock

liver. NPs without Apt ligands (NP) showed that 26.1 � 7.9% ofthe injected NPs was retained in the liver. These data suggest thata narrow window exists where the ligand density of targeted NPsis in the optimal range for maximal targeting and maximal stealthproperties, and this range needs to be determined experimen-tally by using precisely engineered targeted NPs. A similarapproach can be used to optimize other important NP biophysi-cochemical parameters to achieve the combination of parame-ters that result in maximal targeting in vivo.

Summary. We demonstrated that a biointegrated TCP consistingof PLGA, PEG, and the A10 Apt could be used for theself-assembly of targeted NPs for PCa targeting. NPs withdistinct biophysicochemical properties can be generated bymixing PLGA-b-PEG-b-Apt TCPs with a desired amount ofPLGA-b-PEG DCPs lacking the A10 Apt and having the desiredPLGA and PEG characteristics. Increasing the A10 PSMA Aptdensity on NP surface can increase the rate of NP uptake byLNCaP cells in vitro. However, the presence of high Apt surfacedensity also leads to an increase in NP accumulation in the liverand spleen. This is likely due to Apt masking the PEG layer onthe NP surface, and compromising the NP antibiofouling prop-erties in vivo. Thus, in engineering targeted NPs, one mustbalance the tumor-targeting ligand surface density and theantibiofouling surface properties. The use of a PLGA-b-PEG-b-Apt triblock copolymer approach demonstrated the ability tosystematically fine-tune Apt ligand density to maintain tumortargeting while minimizing the amount of NP accumulation inthe liver. The use of this triblock copolymer approach can beused as a technology platform for developing large-scale syn-thesis processes of prefunctionalized targeted NPs while mini-

mizing production times and NP batch-to-batch variations. Thistechnology may have significance in designing delivery vehiclesthat target cancer cells and may contribute to the developmentof the next generation of nano-scale diagnostic and therapeuticmodalities.

MethodsSynthesis of PLGA-b-PEG-b-Apt TCP. The PLGA-b-PEG-b-Apt TCP was synthe-sized by conjugating a modified A10 2�-fluoropyrimidine RNA Apt to the PEGterminal of PEG-PLGA DCP. All materials were purchased from Sigma Aldrich.All reagents were tissue culture grade unless otherwise noted. The RNA Apt(sequence: 5�-NH2-spacer GGG/AGG/ACG/AUG/CGG/AUC/AGC/CAU/GUU/UAC/GUC/ACU/CCU/UGU/CAA/UCC/UCA/UCG/GCiT-3� with 2�-fluoro pyrimidines, a5�-amino group attached by a hexaethyleneglycol spacer and a 3�-inverted Tcap) was custom synthesized by RNA-TEC. Carboxylate-functionalized copol-ymer PLGA-b-PEG was synthesized by the conjugation of COOH-PEG-NH2

(Nektar Therapeutics) to PLGA-COOH (Lactel) in methylene chloride asdescribed by Farokhzad et al. (2). To synthesize PLGA-b-PEG-b-Apt TCP, PLGA-b-PEG DCP was dissolved in methylene chloride and activated with 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide hydrochloride (EDC) and N-hydroxy-succinimide (NHS) in the procedure described earlier. The resulting PLGA-b-PEG-NHS was washed in cold methanol, dried in vacuum, and resolubilized inacetonitrile. The RNA Apt were solubilized in a mixture of formamide/acetonitrile (vol/vol 50:50), and then reacted with PLGA-b-PEG-NHS at roomtemperature for 24 h. The resulting PLGA-b-PEG-b-Apt TCPs were dialyzed incold methanol to remove formamide from the reaction mixture. The resultingPLGA-b-PEG-b-Apt TCP was dried under vacuum and used for NP preparationwithout further treatment. The presence of PLGA and PEG were visualized byusing a 400-MHz 1H NMR (Bruker) at � 1.6, 3.6, 4.8, and 5.2. The protons fromthe Apt on the TCP were visualized at � 1.8–2.2 and 3.78. The Apt conjugationefficiency to the PLGA-b-PEG was determined by degrading the PLGA segmentof the TCP in 0.5 M Tris buffer at pH 9 at 40°C for 3 h, and the mass of Aptrecovered from the TCP was measured by measuring UV absorbance at 260 nm(Varian).

NP Preparation and Characterization: NPs were prepared by using a nanopre-cipitation method. In brief, PLGA-b-PEG-b-Apt (50 mg/ml) and Dtxl (0.5 mg/ml)were dissolved in acetonitrile/formamide (60:40 vol/vol) and together mixeddropwise into water, giving a final NP concentration of 5 mg/ml. The NPs werestirred for 3 h, and the remaining organic solvent was removed by washing theNPs three times by using an Amicon centrifugation filtration membrane witha molecular mass cutoff of 10 kDa. The Apt ligand surface density wasdetermined by hydrolyzing the Apts from the NP surface in 0.01 M Tris bufferat pH 9.5 at 40°C. Apts were separated from the remaining NP supernatant bycentrifugation by 12,000 g for 30 min. After centrifugation, nanoparticledebris formed a pellet, whereas the free aptamers remained suspended in thesupernatant. The mass of Apt recovered in the supernatant was determinedspectrophotometrically. The NP size was measured by quasi-elastic laser lightscattering (QELS) by using a ZetaPALS dynamic light-scattering detector (15mW laser, incident beam � 676 nm, Brookhaven Instruments).

Dtxl Release Study. NPs encapsulating Dtxl were aliquoted into a series ofsemipermeable dialysis, minidialysis tubes (100 �l in volume, molecular masscutoff 10,000 Da, Pierce) and were then placed in a large 40-liter PBS reservoirto mimic the infinite sink condition. At a predetermined time, a fraction of theNPs sampled were collected from the dialysis tubes and were dissolved inacetonitrile. The amount of Dtxl that remained in the NPs at each release timepoint was measured by HPLC in triplicate as described by Cheng et al. (14).

NP Endocytosis in Vitro. For the fluorescent imaging study, LNCaP (PSMA�),PC3 (PSMA�) were seeded at 30,000 cells per well in a eight-well chamber slideand incubated in humidified incubators with 5% CO2 at 37°C overnight. Greenfluorescent dyes, NBD-cholesterol (Invitrogen), were mixed in various formu-lations of PLGA-b-PEG-b-Apt TCP in organic solutions and were coprecipitatedin sterile PBS. NBD-encapsulated NPs were washed and concentrated at 5mg/ml in PBS. LNCaP and PC3 cells were washed with prewarmed PBS andincubated with 50 �g per well of prewarmed NP formulations for 30 min.Endosomal markers were visualized according to manufacturer-recom-mended procedures. In brief, LNCaP and PC3 cells were washed five times toremove the unbound NPs, and fixed with 4% paraformaldehyde for 20 min.Cells were then washed five times, and permeabilized and blocked in PBScontaining 1% BSA and 0.1% Trition X-100, respectively. Early endosomalmarkers, mouse monoclonal EEA-1, and late endosomal markers, mousemonoclonal Mannose 6 Phosphate Receptor (Abcam), were incubated with

Blood heart Kidney Lung Spleen Liver0

20

40

60

80 NP-MutApt 10% NP-Apt 0% NP-Apt 1% NP-Apt 5% NP-Apt 10%

% ID

PG

NP distribution in tumor

0

1

2

NP-Apt10%

NP-Apt5%

NP-Apt1%

NP-Apt0%

NP-MutApt10%

% ID

PG

A

B

Fig. 5. The effect of Apt surface density on NP biodistribution in vivo. Thecomparative biodistribution study of a single systemic administration of NPscontaining different 1% (green), 5% (blue), and 10% (cyan) of NP-Apt wereadministered by retro-orbital injection into LNCaP tumor-bearing mice. Thecontrol groups were NPs without aptamers (NP-Apt 0%) (red), and NPs withscrambled nonfunctional Apt (NP-MutApt 10%) (black). Data represent themean and the standard error of the mean of five mice per group. Data pointsfor NPs-Apt 1% and 5% were statistically significant compared with NP-Apt0% and NP-MutApt 10% by t test at 95% confidence interval. One-wayANOVA with Fisher’s LSD post hoc comparisons at a 95% confidence intervalwere used for statistical comparisons between groups.

2590 � www.pnas.org�cgi�doi�10.1073�pnas.0711714105 Gu et al.

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

1, 2

020

Page 6: Precise engineering of targeted nanoparticles by using ...NPs by macromolecular self-assembly in one single step. Using distinct ratios of PLGA-b-PEG-b-Apt TCP with PLGA-b-PEG diblock

LNCaP and PC3 cells for 1 h at room temperature. These endosomal markerswere visualized by using Cy5 goat anti-mouse antibodies. The cell nuclei werestained with DAPI. Cells were imaged under a DeltaVision fluorescent micro-scope. To quantify the amount of NPs endocytosed by LNCaP and PC3 cells, NPswere radiolabeled with 3H by coprecipitating various PLGA-PEG-Apt formu-lations with 3H-PLGA (custom synthesized by Perkin–Elmer). LNCaP (PSMA�)and PC3 (PSMA�) were seeded in six-well plates and incubated in humidifiedincubators with 5% CO2 at 37°C overnight. On the day of the experiments, 50�g of 3H-abeled NPs were incubated with LNCaP and PC3 cells for 30 min and2 h, respectively. The cells were washed five times to remove the unbound NPsand subsequently lysed in 100–200 �l of lysis buffer (PBS with 0.1% TritonX-100). The NP-cell mixtures were then collected and solubilized in Solvable(Perkin–Elmer), and then mixed with Hionic-Fluor scintillation mixture(Perkin–Elmer). The 3H content in the cells was assayed in a Packard Tri-CarbScintillation Analyzer (Perkin–Elmer).

Tumor Targeting in Vivo. All animal studies were carried out under thesupervision of the Division of Comparative Medicine, Massachusetts Instituteof Technology, and in compliance with the Principles of Laboratory AnimalCare of the National Institutes of Health. Human xenograft prostate cancer

tumors were induced in 8-week-old balb/c nude mice (Charles River Labora-tories). Mice were injected s.c. in the right flank with 4 106 LNCaP cellssuspended in a 1:1 mixture of media and matrigel (BD Biosciences). Tumor-targeting studies were carried out after the mice developed �300-mm3 tu-mors. Mice were randomly divided to minimize tumor size variations betweengroups. Mice were anesthetized by i.p. injection of avertin (200 mg/kg bodyweight), and dosed with NP formulations by retro-orbital injection. NP bio-distributions were traced by encapsulating 3H-PLGA into the NPs beforeadministration. Mice were killed at 24 h after NP injections, and 200 �l ofblood was drawn by cardiac puncture from each mouse. The tumor, heart,lungs, liver, spleen, and kidneys were harvested from each animal as describedby Cheng et al. (14). The 3H content in the tissues was assayed in a PackardTri-Carb Scintillation Analyzer. To determine 100% dose, vials of the formu-lated NPs were counted along with the tissues.

ACKNOWLEDGMENTS. We thank Drs. Philip Kantoff and Neil Bander forhelpful discussions throughout this study. This work was supported by Na-tional Institutes of Health Grants CA119349 and EB003647 and Koch–ProstateCancer Foundation Award in Nanotherapeutics. F.G. was supported by aPostdoctoral Fellowship from the Canadian Natural Sciences and EngineeringResearch Council.

1. Langer R (1998) Drug delivery and targeting. Nature 392:5–10.2. Farokhzad OC, et al. (2006) Targeted nanoparticle-aptamer bioconjugates for cancer

chemotherapy in vivo. Proc Natl Acad Sci USA 103:6315–6320.3. Ferrari M (2005) Cancer nanotechnology: Opportunities and challenges. Nature Rev

5:161–171.4. Gu F, et al. (2007) Targeted nanoparticles for cancer therapy. Nano Today 2:14–21.5. Farokhzad OC, Langer R (2006) Nanomedicine: Developing smarter therapeutic and

diagnostic modalities. Adv Drug Delivery Rev 58:1456–1459.6. Heath TD, Fraley RT, Papahdjopoulos D (1980) Antibody targeting of liposomes: Cell

specificity obtained by conjugation of F (ab�)2 to vesicle surface. Science 210:539–541.7. Leserman LD, Barbet J, Kourilsky F, Weinstein JN (1980) Targeting to cells of fluorescent

liposomes covalently coupled with monoclonal antibody or protein A Nature 288:602–604.

8. Zhang L, et al. (2007) Nanoparticles in medicine: Therapeutic applications and devel-opments. Clin Pharmacol Ther 10.1038/sj.clpt.6100400.

9. Leamon CP, Cooper SR, Hardee GE (2003) Folate-liposome-mediated antisense oli-godeoxynucleotide targeting to cancer cells: evaluation in vitro and in vivo. Biocon-jugate Chem 14:738–747.

10. Popielarski SR, Pun SH, Davis ME (2005) A nanoparticle-based model delivery system toguide the rational design of gene delivery to the liver. 1. Synthesis and characteriza-tion. Bioconjugate Chem 16:1063–1070.

11. Chiu SJ, Liu S, Perrotti D, Marcucci G, Lee RJ (2006) Efficient delivery of a Bcl-2-specificantisense oligodeoxyribonucleotide (G3139) via transferrin receptor-targeted lipo-somes. J Controlled Release 112:199–207.

12. Farokhzad OC, et al. (2004) Nanoparticle-aptamer bioconjugates: A new approach fortargeting prostate cancer cells. Cancer Res 64:7668–7672.

13. Sun B, Ranganathan B, Feng SS (2008) Multifunctional poly(d,l-lactide-co-glycolide)/montmorillonite (PLGA/MMT) nanoparticles decorated by Trastuzumab for targetedchemotherapy of breast cancer. Biomaterials 29:475–486.

14. Cheng J, et al. (2007) Formulation of functionalized PLGA-PEG nanoparticles for in vivotargeted drug delivery. Biomaterials 28:869–876.

15. Kubowicz S, et al. (2005) Multicompartment micelles formed by self-assembly of linearABC triblock copolymers in aqueous medium. Angew Chem 44:5262–5265.

16. Li G, Shi L, Ma R, An Y, Huang N (2006) Formation of complex micelles with double-responsive channels from self-assembly of two diblock copolymers. Angew Chem45:4959–4962.

17. Reynhout IC, Cornelissen JJ, Nolte RJ (2007) Self-assembled architectures from biohy-brid triblock copolymers. J Am Chem Soc 129:2327–2332.

18. Lupold SE, Hicke BJ, Lin Y, Coffey DS (2002) Identification and characterization ofnuclease-stabilized RNA molecules that bind human prostate cancer cells via theprostate-specific membrane antigen. Cancer Res 62:4029–4033.

19. Ellington AD, Szostak JW (1990) Invitro selection of Rna molecules that bind specificligands. Nature 346:818–822.

20. McNamara JO, II, et al. (2006) Cell type-specific delivery of siRNAs with aptamer-siRNAchimeras. Nat Biotechnol 24:1005–1015.

21. Chu TC, et al. (2006) Aptamer:toxin conjugates that specifically target prostate tumorcells. Cancer Res 66:5989–5992.

22. Chu TC, et al. (2006) Labeling tumor cells with fluorescent nanocrystal-aptamerbioconjugates. Biosens Bioelectron 21:1859–1866.

23. Hu Y, Xie J, Tong YW, Wang CH (2007) Effect of PEG conformation and particle size onthe cellular uptake efficiency of nanoparticles with the HepG2 cells. J ControlledRelease 118:7–17.

24. Avgoustakis K, et al. (2003) Effect of copolymer composition on the physicochemicalcharacteristics, in vitro stability, and biodistribution of PLGA-mPEG nanoparticles. IntJ Pharm 259:115–127.

25. Vauthier C, Schmidt C, Couvreur P (1999) Measurement of the density of polymericnanoparticulate drug carriers by isopycnic centrifugation. J Nanoparticle Res 1:411–418.

26. Mosqueira VC, et al. (2001) Biodistribution of long-circulating PEG-grafted nanocap-sules in mice: effects of PEG chain length and density. Pharm Res 18:1411–1419.

27. Gref R, et al. (1994) Biodegradable long-circulating polymeric nanospheres. Science263:1600–1603.

Gu et al. PNAS � February 19, 2008 � vol. 105 � no. 7 � 2591

ENG

INEE

RIN

GM

EDIC

AL

SCIE

NCE

S

Dow

nloa

ded

by g

uest

on

Sep

tem

ber

1, 2

020