You are on page 1of 6

ARTICLES

PUBLISHED ONLINE: 15 FEBRUARY 2016 | DOI: 10.1038/NNANO.2015.330

Protein adsorption is required for stealth effect of


poly(ethylene glycol)- and poly(phosphoester)-
coated nanocarriers
Susanne Schöttler1,2, Greta Becker1, Svenja Winzen1, Tobias Steinbach1, Kristin Mohr1,
Katharina Landfester1, Volker Mailänder1,2*† and Frederik R. Wurm1*†

The current gold standard to reduce non-specific cellular uptake of drug delivery vehicles is by covalent attachment of
poly(ethylene glycol) (PEG). It is thought that PEG can reduce protein adsorption and thereby confer a stealth effect.
Here, we show that polystyrene nanocarriers that have been modified with PEG or poly(ethyl ethylene phosphate) (PEEP)
and exposed to plasma proteins exhibit a low cellular uptake, whereas those not exposed to plasma proteins show high
non-specific uptake. Mass spectrometric analysis revealed that exposed nanocarriers formed a protein corona that
contains an abundance of clusterin proteins (also known as apolipoprotein J). When the polymer-modified nanocarriers
were incubated with clusterin, non-specific cellular uptake could be reduced. Our results show that in addition to reducing
protein adsorption, PEG, and now PEEPs, can affect the composition of the protein corona that forms around nanocarriers,
and the presence of distinct proteins is necessary to prevent non-specific cellular uptake.

dsorption of proteins from physiological fluids to nano-

A
been proposed as PEG alternatives include hydroxyethyl starch
carriers leads to the formation of a protein shell1,2. This (HES), polysialic acid and dextrin13. However, another very interest-
rapidly forming protein corona has previously been shown ing polymer class—the poly(phosphoester)s (PPEs)—has never
to be responsible for the biological fate of nanocarriers3–5. Poly- been investigated with respect to stealth behaviour. In recent
(ethylene glycol) (PEG) is widely used to suppress any non-specific years, we have been studying PPEs with respect to novel synthetic
protein adsorption, and PEGylated drugs and (nano)carriers show protocols and biomedical applications22,23. The chemical structure
longer blood half-lives and less non-specific cellular uptake com- of PPEs is highly modular, they are degradable, and their degra-
pared to unmodified drugs6,7. This is a prerequisite for specific dation products and time can be adjusted by means of precise
targeting8,9. For surfaces, PEGylation is also the typical approach chemistry24,25. Several studies have dealt with the preparation of
to reduce protein adsorption10,11. This feature, usually referred to PPE-based drug carriers, but none has investigated the protein
as the ‘stealth’ effect, is generally explained by the high level of interactions of the hydrophilic PPEs with blood proteins26,27.
hydration of the hydrophilic polyether backbone, which also pre- Three distinct findings are reported herein. First, the polymer-
vents protein adsorption on typically hydrophobic polymer surfaces modified nanocarriers (with PEG and PPE) exhibit decreased
by means of steric repulsion12,13. When the stealth properties of PEG protein adsorption after incubation in human plasma compared
are discussed it is often neglected that even if the overall protein to unmodified particles. Second, and more importantly, mass spec-
adsorption is reduced, it cannot be fully suppressed; a certain trometric analysis of the protein corona generated on the particles’
amount of serum proteins, for example, is always detected on the surface after plasma incubation revealed a similar pattern of proteins
drug carriers, thus altering the surface properties14,15. on PEGylated and ‘PPEylated’ surfaces. Clusterin—also termed apo-
PEG is a non-biodegradable polyether and its accumulation in the lipoprotein J (ApoJ)—was identified as a major component on both
body must be prevented. This is especially important when drug–PEG surfaces. Third, interestingly, a high non-specific cellular uptake for
conjugates are administered for chronic diseases over a period of both polymer-modified nanocarriers was found without previous
several years. In such cases, an accumulation of PEG in the body is plasma incubation, indicating the requirement for distinct proteins
likely, and can cause unwanted side effects13. Indeed, the development to prevent non-specific cellular uptake. This non-specific cellular
of PEG antibodies16,17 and severe hypersensitivity reactions have been uptake could eventually be reduced by non-covalent ‘pre-loading’
reported18. Antibody formation can lead to an accelerated blood clear- with clusterin onto the nanocarriers, demonstrating a strong effect
ance following repeated systemic administration19. Furthermore, PEG of reduced cell internalization. Unravelling the effects of protein
has been shown to trigger complement activation, which can lead to type and stealth polymer structure will produce new efficient drug
anaphylactic reactions in sensitive individuals20. These factors have delivery devices.
given rise to a search for alternatives to PEG, and other biocompa-
tible polymers have been recognized to improve the in vivo proper- PEEP as novel stealth polymers
ties of pharmaceuticals. For example, zwitterionic molecules such as The nanocarriers (Fig. 1) used in this study were monodisperse
polybetaines or polysaccharides can also generate hydrophilic shells polystyrene nanoparticles with diameters of ∼100 nm, which were
when coupled to nanoparticles21. Degradable polymers that have prepared by free radical terpolymerization of three monomers

1
Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany. 2 Johannes Gutenberg University Mainz, University Medical Center,
Department of Dermatology, Langenbeckstr. 1, 55131 Mainz, Germany. †These authors jointly supervised this work. * e-mail: mailaender@mpip-mainz.mpg.de;
wurm@mpip-mainz.mpg.de

372 NATURE NANOTECHNOLOGY | VOL 11 | APRIL 2016 | www.nature.com/naturenanotechnology

© 2016 Macmillan Publishers Limited. All rights reserved


NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2015.330 ARTICLES
a b H2N NH2 c
PS-NH2
H2N NH2
x y z
O O
O O O O
NO R
O
NH3CI
R = PEG R = PEEP

N O H
H
O N
F
B N
O
O N OP O
O nO 100 nm
F nO
m m

Ø (nm) 106 117 119 117 122

n − 44 110 49 92

m − 3,500 2,900 3,200 2,400

ζ (mV ) 46 15 8 −9 −10

PS-NH2 PS-PEG44 PS-PEG110 PS-PEEP49 PS-PEEP92

Figure 1 | Characteristics of nanocarriers. Amine-functionalized polystyrene nanoparticles were modified with PEG or PEEP. a, The terpolymer of styrene,
2-aminoethyl methacrylate hydrochloride and BODIPY-methacrylate, used as the carrier material for the nanoparticles investigated here. b, PEG and PEEP
attachment to the amino groups of polystyrene nanoparticles. c, Scanning electron microscopy image of PS-PEEP49. d, Schematic representation of
nanocarriers (average diameter Ø determined by dynamic light scattering (DLS), number of repeating units n, number of polymer chains per nanoparticle m,
and zeta potential ζ).

via miniemulsion28. Styrene was copolymerized with 2 wt% that is, grafted polystyrene, proved that no unreacted polymer was
2-aminoethyl methacrylate hydrochloride to introduce reactive in the mixture (compare Supplementary Fig. 19).
groups for subsequent polymer attachment and in the presence of To investigate the stealth effect of the polymer-modified nano-
a fluorescently active boron-dipyrromethene (BODIPY)-methacrylate carriers, the amount of proteins adsorbed after plasma incubation
to label the nanocarriers for further biological studies (see was analysed (Fig. 2a). The nanocarriers were incubated with
Supplementary Section ‘Synthesis’ for details). The nanoparticles human blood plasma for 1 h and separated from unbound proteins
were characterized by standard procedures (Fig. 1: light scattering, by centrifugation and several washing steps. The adsorbed proteins
zeta potential, number of polymer chains per particle, Mn of each were removed from the surface by treatment with urea and quanti-
chain = 190,000 g mol–1). N-hydroxysuccinimide-functionalized fied with a colorimetric protein assay. The adsorption of plasma
PEG (Mn = 2,000 g mol–1, degree of polymerization (DPn) = 44 proteins was significantly reduced by surface functionalization
and Mn = 5,000 g mol–1, DPn = 110) and the water-soluble PPE- with PEG and PEEP. Decreases of 79% (PS-PEG44), 66%
derivative PEEP (poly(ethyl ethylene phosphate), Mn = 7,600 g mol–1, (PS-PEG110), 73% (PS-PEEP49) and 70% (PS-PEEP92) were
DPn = 49 and Mn = 14,100 g mol–1, DPn = 92) were coupled to the observed compared to the adsorption of proteins onto PS-NH2
amino-functionalized nanocarriers. Attachment of the PEG and nanocarriers (Fig. 2a). The reduction in protein adsorption to a
PEEP chains to the nanocarriers was demonstrated by the reduction value similar to that for PEG is the first indication of a stealth
in the zeta potential (PEGylated particles, ∼+10 mV; PPEylated effect provoked by PPEs. The adsorbed protein patterns were
particles, ∼–10 mV). The number of attached PEG or PEEP chains further visualized using SDS–polyacrylamide gel electrophoresis
on each particle could be determined accurately via 1H NMR spectro- (PAGE; Supplementary Fig. 28). At first glance, a similar protein
scopy, and was found, typically, to be in the range 2,000–4,000, pattern can be observed for all stealth nanocarriers, with enrichment
corresponding to a degree of functionalization of the amine groups of a 38 kDa protein, but several differences between PEGylated and
of 11–16% (for the list of results see Supplementary Tables 7 and 8). PPEylated surfaces are also detected. The molecular weight differ-
After 15% grafting of the surface amino groups, an area of ∼10 nm2 ences of PEG and PEEP seem not to play a crucial role. The
was available for each polymer chain. Kuga29 has reported the hydro- amino-functionalized control nanocarrier is mainly covered by
dynamic radii of different PEGs to be 1.3 nm (PEG Mn = 2,000) and albumin, the most abundant protein in human blood plasma (the
2.7 nm (PEG Mn = 6,000), resulting in surface areas of up to 23 nm² band at ∼62 kDa). Again, the overall stronger protein adsorption
for these polymers. Assuming a PEG chain to have the same confor- to PS-NH2 is clearly illustrated. In addition to the covalently
mation and size when on the surface of a particle as is found in sol- PEG- and PPEylated nanocarriers, Lutensol AT50 (a PEG-based sur-
ution, the calculations show that the maximum number of polymer factant) was self-assembled on amino-functional and neutral PS
chains is almost reached for PEG110. Gel permeation chromato- nanoparticles as controls. With different concentrations of surfactant,
graphy (GPC) analysis of the dissolved modified nanocarriers, similar protein patterns were identified by SDS–PAGE or mass

NATURE NANOTECHNOLOGY | VOL 11 | APRIL 2016 | www.nature.com/naturenanotechnology 373

© 2016 Macmillan Publishers Limited. All rights reserved


ARTICLES NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2015.330

a c −Plasma +Plasma
5

4
PS-NH2

Proteins (mg m−2)

PS-PEEP49

PS-PEEP92
3

PS-PEG110
PS-PEG44
PS-NH2
2

1 PS-PEG44

b 1,250
Plasma −Plasma
Relative fluorescence intensity

1,000 PS-PEG110

750

500

250 PS-PEEP49

0
-P 44

-P 44
0

0
-P 49

PS 92

-P 49
92
-P 2

-P 2
PS H

PS H
-P 11

-P 11
PS EG

PS EG
P

P
PS EEP

PS EEP
PS EG

PS EG
-N

-N
EE

EE
PS

PS-PEEP92

Figure 2 | Cellular uptake of PEEP and PEG nanocarriers. Functionalization of nanoparticles with PEEP and PEG inhibits uptake into macrophages only in the
presence of proteins. a, Quantification of human plasma proteins adsorbed to the nanocarriers’ surface (mg protein m–²). Values are expressed as mean ± s.d.
of biological triplicates. b, Flow cytometry analysis of RAW264.7 cells incubated with different nanocarriers for 2 h. The nanocarriers were incubated in human
plasma or in water (–plasma) for 1 h at 37 °C and separated from residual proteins by centrifugation before being added to cells. Values are expressed as
mean ± s.d. of biological triplicates. The experiment was further replicated three times in the laboratory. c, Laser scanning microscopy images of RAW264.7
cells incubated with PS-NH2 , PS-PEEP49, PS-PEEP92, PS-PEG44 and PS-PEG110 for 1 h in 100% human plasma or DMEM without plasma. The cell membrane
is stained with CellMask Orange (red) and nanocarriers are shown in green. Scale bars, 10 μm.

spectrometry (Supplementary Figs 29 and 30). The lower protein proteins alone that is responsible for the inhibition of cellular inter-
binding of the stealth nanocarriers compared with the unmodified nalization by the stealth polymers. It is, instead, a secondary effect;
control was also confirmed by isothermal titration calorimetry that is, the specific protein patterns of PEG- or PEEP-functionalized
(Supplementary Fig. 31). nanocarriers consist of ‘don’t-eat-me’ proteins. Laser scanning
microscopy (Fig. 2c) also shows this effect clearly: strong inter-
Proteins are necessary for the stealth effect nalization of the unmodified nanocarriers (PS-NH2) and both
The advantage of stealth nanocarriers is their ability to remain in the PEEP-modified nanocarriers (PEEP-NP) into RAW264.7 cells was
blood circulation for extended periods of time by evading clearance observed (even after only 1 h) when the cells were cultured in
by the immune system30. Macrophages are important components culture medium without plasma proteins. In contrast, macrophages
of the immune defence system and play a major role in clearing cultured with plasma did not internalize PEGylated or PPEylated
foreign molecules from the blood. Only by eluding these phagocytic nanocarriers, but the unmodified nanoparticles were taken up.
cells can nanocarriers reach their destination within the body and Even after 24 h incubation, no significant uptake of the stealth nano-
fulfil their task. Therefore, the internalization of the described nano- carriers pre-coated with plasma proteins was observed, in contrast
carriers into a murine macrophage-like cell line, namely RAW264.7, to the control nanocarrier (Supplementary Fig. 32). Similar results
was studied. Flow cytometry analysis revealed that uptake of the were also obtained for the cervical cancer cell line HeLa cultured
‘naked’ nanocarriers (PS-NH2) is inhibited by the attachment of in medium with or without serum (Supplementary Fig. 33a,c). A
PEEP to the same degree as by PEG when the nanocarriers are high uptake of PEGylated and PPEylated nanocarriers without the
previously incubated with plasma (Fig. 2b). presence of proteins, and no uptake after incubation with 10%
Most intriguingly, a high uptake of all nanocarriers in protein- fetal bovine serum (FBS), was observed by flow cytometry analysis
free medium was observed. As these nanocarriers carry no adsorbed and live cell imaging. Furthermore, human immature dendritic
proteins, this strongly indicates that it is not the reduced amount of cells, also professional phagocytes in the human body, did not

374 NATURE NANOTECHNOLOGY | VOL 11 | APRIL 2016 | www.nature.com/naturenanotechnology

© 2016 Macmillan Publishers Limited. All rights reserved


NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2015.330 ARTICLES
a PS-NH2 PS-PEG44 PS-PEG110 functions shows that the control nanocarrier is mainly covered by
albumin (as visualized by SDS–PAGE) and the proteins involved
in coagulation, and lipoproteins bind preferentially to the polymer-
modified nanocarriers. Interestingly, for PEEP-modified nanocarriers,
lipoproteins constitute up to 85% of the protein corona.
Figure 3b depicts the most abundant proteins identified in the
so-called ‘hard’ corona of the nanocarriers (Supplementary
Acute phase
PS-PEEP49 PS-PEEP92 Table 9). Albumin, fibrinogen and vitronectin are the most promi-
Coagulation
Complement system
nent proteins on the control nanocarrier (PS-NH2 , >80%), but these
Immunoglobulins proteins are strongly depleted on PEGylated and PPEylated nano-
Lipoproteins carriers. In these latter nanocarriers, clusterin—an apolipoprotein—
Other plasma components is the major protein in the hard corona, and can be assigned to the
Tissue leakage
38 kDa band in the SDS–PAGE results (Supplementary Fig. 28). It
makes up 77% of the protein corona on the PEEP-functionalized
PS-PEEP49

PS-PEEP92
nanocarriers, with all other proteins contributing less than 2%. This
PS-PEG110
PS-PEG44
PS-NH2

b significant enrichment of one single protein is remarkable. In previous


publications, apolipoproteins have been found to be enriched3,31,32,
Apolipoprotein A-I 80 some literature has described that hydrophobic materials adsorb
Apolipoprotein A-IV apolipoproteins even more33, and hydrophilic nanomaterials have
Apolipoprotein C-III been reported to adsorb mainly albumin, fibrinogen and IgG as
Apolipoprotein D more abundant proteins. As apolipoproteins have lipid-binding
Clusterin
domains, their attraction to hydrophobic surfaces is readily concei-
Percentage of total protein corona

Complement C1q subcomponent 60


subunit B vable. Interestingly, PEG is supposed to be a rather hydrophilic
Complement C1q subcomponent material. However, different apolipoproteins have different
subunit C propensities to adsorb: Walkey and Chan found that ApoA4,
Fibrinogen alpha chain ApoB, ApoC3 and ApoE adsorb preferentially to hydrophobic
Fibrinogen beta chain 40
quantum dots, while ApoA1, ApoA2 and ApoC1 adsorb preferen-
Fibrinogen gamma chain tially to more hydrophilic NH2-modified quantum dots33.
Fibronectin
Ig gamma-1 chain C region
Apolipoproteins therefore appear to be a class of proteins of interest
Ig gamma-2 chain C region for different nanocarriers, and will need to be further explored for
20
Ig kappa chain C region their different functions.
Ig mu chain C region Clusterin has been reported to adsorb to the surface of several
Serum albumin nanocarriers with varying affinities. Relatively high amounts of clus-
Serum amyloid P-component terin were detected on solid lipid nanoparticles, with an enhanced
Vitronectin 0 quantity with increasing incubation time34. Furthermore, a signifi-
cantly enhanced affinity was detected for silica nanoparticles31.
Figure 3 | Proteomic analysis of protein corona on the surface of On polystyrene nanoparticles with distinct surface characteristics,
nanocarriers. The protein corona of PEG- and PEEP-functionalized clusterin has been identified as one of the most abundant proteins32,
nanoparticles mainly consists of the protein clusterin. a, Classification of and an increasing amount of clusterin has been reported for nano-
protein corona components identified by quantitative LC-MS. The average particles with increasing hydrophobicity35,36. To our knowledge,
amount of each protein (in fmol) was calculated from three biological and such a high abundance of clusterin has not been described pre-
two technical replicates. Identified proteins were grouped according to their viously for other nanocarriers. On PEGylated nanocarriers, apolipo-
function in biological processes, and the amounts of proteins were summed. protein A1 (as well as clusterin) is also found (∼11%, corresponding
b, Heat map of the most abundant proteins in the protein corona of PS-NH2 , to the band at ∼28 kDa in the SDS–PAGE results).
PS-PEG44 , PS-PEG110 , PS-PEEP49 and PS-PEEP92 determined by proteomic These differences in the composition of the protein corona for
mass spectrometry. Values were calculated from the molar masses of each two different hydrophilic polymers is an additional handle for the
protein identified by LC-MS. Only those proteins that constitute at least 1% future design of novel drug carriers or diagnostic devices, if the mol-
of the protein corona on one of the nanocarriers are shown. ecular structure of the stealth polymer can be tailored in a way to
control not only the amount but also the type of plasma protein,
show any significant internalization of the four stealth nanocarriers for example by copolymerization.
when incubated with serum (Supplementary Fig. 33b). All nanocar- The significant enrichment of clusterin on PEG-functionalized
riers were investigated with respect to their toxicity. The PEGylated nanocarriers (and even more on PEEP-functionalized nanocarriers)
and PPEylated nanocarriers did not promote the cell death of HeLa suggests its important function in stealth properties. In blood, the
cells, but the control nanocarrier (PS-NH2) showed minor toxicity native glycoprotein clusterin is assembled with other apolipopro-
(Supplementary Fig. 34). teins, such as ApoA1, into high-density-lipoprotein (HDL) par-
From these results, fundamental conclusions can be drawn. It is ticles. The clusterin/ApoJ gene encodes for a different protein
not only the reduction in protein adsorption that explains the stealth isoform: a highly conserved disulphide-linked heterodimeric
effect of both PEG- and PEEP-functionalized nanocarriers; more secreted glycoprotein of ∼75–80 kDa (sCLU)37, as well as cyto-
importantly, the type of protein must be highly important. This plasmic (cCLU) and a nuclear (nCLU) isoform38. The secreted
renders the stealth effect a secondary effect caused by selective form of clusterin in plasma has a concentration of 40–60 μg ml–1
protein adsorption. and is thus not among the most abundant of proteins, but it is
still a protein with a relevant concentration. sCLU was first
Analysis of protein corona reveals high clusterin content described in testis fluid39 and, as well as in blood plasma, sCLU is
With liquid chromatography–mass spectrometry (LC-MS) analysis, also found in other body fluids including milk, urine and cerebro-
a total of 171 proteins were identified in the corona of all five nano- spinal or lacrimal fluid37,40. Various physiological functions have
carriers (Fig. 3). Grouping the identified proteins according to their been proposed for clusterin: complement attack prevention,

NATURE NANOTECHNOLOGY | VOL 11 | APRIL 2016 | www.nature.com/naturenanotechnology 375

© 2016 Macmillan Publishers Limited. All rights reserved


ARTICLES NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2015.330

necessity for a mixture of dysopsonins on stealth nanocarriers. Due


to the washing of the nanocarriers before the proteomic analysis, pro-
Water teins in the soft corona are not identified but may also contribute to
the stealth effect. Although some first steps have been taken45,
methods to investigate the soft corona in its complexity are not yet
as elaborate as those carried out for the hard corona. These comp-
HSA lementary methods are urgently needed to add to the knowledge
PS-PEEP92
about the protein corona. While dysopsonization has also been
800
achieved by the coupling of self-peptides derived from cell surface
700 proteins46, this is the first report on a polymeric surface resulting in
a specific plasma adsorption.
Relative fluorescence intensity

600 Although, opsonization of nanocarriers with plasma proteins is


Plasma
500 or thought to be the major cause for clearance from the bloodstream,
clusterin we have shown that stealth nanocarriers functionalized with either
400 PEG or PEEP were internalized by macrophages when no proteins
were present, and so opsonization cannot have occurred. Walkey
300
and colleagues have suggested that PEG does not eliminate serum
200 protein adsorption in total, but selectively suppresses adsorption
of specific proteins, resulting in a minimized macrophage
100 uptake47. We want to go even further: we postulate that it is not
0
only the selective reduction of opsonizing proteins such as immuno-
globulins and complement factors, but in fact the selective enrich-
rin

A
a
2O

HS
te

ment of a single type of protein, depending on the polymer


H

as

us

+
+

Pl

Cl
+

structure, that triggers the (secondary) stealth effect.


+

Apolipoproteins have been recognized previously as an impor-


Figure 4 | Clusterin reduces non-specific cellular uptake. Clusterin tant class of proteins governing the in vitro and in vivo fate of nano-
functions as a dysopsonin and assists the ‘stealth effect’. Cellular uptake of carriers1,4,48. ApoE, in particular, has been identified as an
PS-PEEP92 into RAW264.7 cells after incubation in water, human plasma, interesting molecule, because it enhances the passage of nanocarriers
clusterin (66.6 μg ml–1) or human serum albumin (HSA, 66.6 μg ml–1). through the blood–brain barrier49, while ApoA1 and ApoB-100
The nanocarriers were incubated in each medium for 1 h at 37 °C before also promote transport into the central nervous system50. Other
being added to the cells maintained in DMEM. After 1 h, the cells were apolipoproteins, such as ApoA4 and ApoC3, have been described
analysed by flow cytometry. Values are expressed as mean ± s.d. of to function as dysopsonins32. Clearly, these proteins could also con-
biological triplicates. The experiment was further replicated three times tribute to the stealth effect and may even substitute for clusterin on
in the laboratory. other stealth nanocarriers. Nevertheless, we found clusterin to
be such a prominent protein in the proteome of the nanoparticle
promotion of cell aggregation, regulation of apoptosis and lipid corona that it appears to be the most important one.
transport, but all are mainly based on binding partners41. The In summary, PEGylated and PPEylated (PEEP-modified) model
secreted form of clusterin has been described to prevent protein nanocarriers demonstrate a distinct reduction and modification of
aggregation and precipitation by binding to hydrophobic regions protein adsorption after incubation with human plasma when com-
of misfolding-prone proteins42. It is therefore believed to be the pared with non-modified nanocarriers. The uptake of the
first secreted mammalian chaperone to be identified. One could PEGylated and PPEylated nanocarriers by different cell lines,
speculate that PEG and PEEP appear to clusterin to be such including a macrophage-like cell line, was completely inhibited.
proteins, although it remains unclear how they are structurally We identified clusterin as the major protein in the corona of both
related. Alternatively, other proteins that can be detected under polymer-modified nanocarriers, with its highest enrichment on
these conditions in our proteomic approach could have been mis- PPEylated surfaces. Furthermore, pre-loading of the PPEylated
folded during binding to the nanoparticles’ surfaces, and sub- nanocarriers with clusterin reduced macrophage uptake (while
sequently attracted clusterin. However, binding to polymeric albumin pre-loading increased the uptake), providing evidence for
surfaces of this protein has not been studied to date. a dysopsonizing function of clusterin concerning internalization
into macrophages. When comparing these results with other
Clusterin reduces non-specific cellular uptake attempts to pursue a ‘stealth effect’, for example, by Discher and
To further analyse the effect of clusterin, PPEylated nanocarriers were colleagues46, who used peptides of hCD47 to prohibit clearance by
incubated with native clusterin at its physiological concentration43, phagocytes, our work can be seen to report a facile way to enrich
and uptake into RAW264.7 cells was studied. Figure 4 confirms the ‘stealth effect’ of nanoparticles, just by mixing with
that incubation with clusterin reduces the uptake of the nanocarriers human plasma.
into macrophages by 75.4% (in the absence of plasma or clusterin a These results lead to fundamental insights that have to be con-
high cellular uptake is detected). To prove that this effect cannot be sidered for future nanocarrier design: (1) the stealth effect is not
provoked by any plasma protein, human serum albumin (HSA) a polymer effect alone, but a secondary effect; (2) degradable
was used as control. Interestingly, HSA incubation increased the poly(phosphoester)s are potential PEG alternatives; (3) the chemical
internalization by 34%, although albumin is often described as a dys- structure of the stealth polymer can trigger the amount and type of
opsonin44. Although pure clusterin does not have the same effect as plasma protein adsorbed and thus the biological fate of the
whole plasma, which decreases the uptake by as much as 99.5%, it nanocarrier; (4) clusterin is an interesting candidate for further
shows that clusterin functions as a strong dysopsonin for macro- investigations into the stealth effect.
phages and supports the idea of its predominant participation in
the stealth effect of PEG- and PPEylated nanocarriers. The lower effi- Methods
cacy of clusterin alone in hindering nanoparticle uptake compared to All experimental details, materials, and methods can be found in the
the complex protein corona formed in whole plasma indicates the Supplementary Information.

376 NATURE NANOTECHNOLOGY | VOL 11 | APRIL 2016 | www.nature.com/naturenanotechnology

© 2016 Macmillan Publishers Limited. All rights reserved


NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2015.330 ARTICLES
Received 9 March 2015; accepted 10 December 2015; 29. Kuga, S. Pore-size distribution analysis of gel substances by size exclusion
published online 15 February 2016 chromatography. J. Chromatogr. A 206, 449–461 (1981).
30. Kang, B. et al. Tailoring the stealth properties of biocompatible polysaccharide
nanocontainers. Biomaterials 49, 125–134 (2015).
References 31. Tenzer, S. et al. Nanoparticle size is a critical physicochemical determinant of the
1. Tenzer, S. et al. Rapid formation of plasma protein corona critically affects human blood plasma corona: a comprehensive quantitative proteomic analysis.
nanoparticle pathophysiology. Nature Nanotech. 8, 772–781 (2013). ACS Nano 5, 7155–7167 (2011).
2. Cedervall, T. et al. Understanding the nanoparticle–protein corona using 32. Ritz, S. et al. Protein corona of nanoparticles: distinct proteins regulate the
methods to quantify exchange rates and affinities of proteins for nanoparticles. cellular uptake. Biomacromolecules 16, 1311–1321 (2015).
Proc. Natl Acad. Sci. USA 104, 2050–2055 (2007). 33. Walkey, C. D. & Chan, W. C. Understanding and controlling the interaction of
3. Monopoli, M. P., Aberg, C., Salvati, A. & Dawson, K. A. Biomolecular coronas nanomaterials with proteins in a physiological environment. Chem. Soc. Rev. 41,
provide the biological identity of nanosized materials. Nature Nanotech. 7, 2780–2799 (2012).
779–786 (2012). 34. Goppert, T. M. & Muller, R. H. Adsorption kinetics of plasma proteins on solid
4. Aggarwal, P. et al. Nanoparticle interaction with plasma proteins as it relates to lipid nanoparticles for drug targeting. Int. J. Pharm. 302, 172–186 (2005).
particle biodistribution, biocompatibility and therapeutic efficacy. Adv. Drug 35. Blunk, T. et al. Colloidal carriers for intravenous drug targeting: plasma
Deliv. Rev. 61, 428–437 (2009). protein adsorption patterns on surface-modified latex particles evaluated by
5. Sacchetti, C. et al. Surface polyethylene glycol conformation influences the two-dimensional polyacrylamide gel electrophoresis. Electrophoresis 14,
protein corona of polyethylene glycol-modified single-walled carbon 1382–1387 (1993).
nanotubes: potential implications on biological performance. ACS Nano 7, 36. Gessner, A. et al. Nanoparticles with decreasing surface hydrophobicities:
1974–1989 (2013). influence on plasma protein adsorption. Int. J. Pharm. 196, 245–249 (2000).
6. Veronese, F. M. & Pasut, G. PEGylation, successful approach to drug delivery. 37. Vanhooren, V. et al. Protein modification and maintenance systems as
Drug Discov. Today 10, 1451–1458 (2005). biomarkers of ageing. Mech. Ageing Dev. 151, 71–84 (2015).
7. Alconcel, S. N. S., Baas, A. S. & Maynard, H. D. FDA-approved poly(ethylene 38. Koltai, T. Clusterin: a key player in cancer chemoresistance and its inhibition.
glycol)–protein conjugate drugs. Polym. Chem. 2, 1442–1448 (2011). Oncol. Targets Ther. 7, 447–456 (2014).
8. Baier, G. et al. Suppressing unspecific cell uptake for targeted delivery using 39. Blaschuk, O., Burdzy, K. & Fritz, I. B. Purification and characterization of a cell-
hydroxyethyl starch nanocapsules. Biomacromolecules 13, 2704–2715 (2012). aggregating factor (clusterin), the major glycoprotein in ram rete testis fluid.
9. Landfester, K. & Mailander, V. Nanocapsules with specific targeting and release J. Biol. Chem. 258, 7714–7720 (1983).
properties using miniemulsion polymerization. Expert Opin. Drug Deliv. 10, 40. Nilselid, A. M. et al. Clusterin in cerebrospinal fluid: analysis of carbohydrates
593–609 (2013). and quantification of native and glycosylated forms. Neurochem. Int. 48,
10. Worz, A. et al. Protein-resistant polymer surfaces. J. Mater. Chem. 22, 718–728 (2006).
19547–19561 (2012). 41. Silajdzic, E., Minthon, L., Bjorkqvist, M. & Hansson, O. No diagnostic value of
11. Otsuka, H., Nagasaki, Y. & Kataoka, K. PEGylated nanoparticles for biological plasma clusterin in Alzheimer’s disease. PLoS ONE 7, e50237 (2012).
and pharmaceutical applications. Adv. Drug Deliv. Rev. 64, 246–255 (2012). 42. Poon, S. et al. Clusterin is an ATP-independent chaperone with very broad
12. Pino, P. d. et al. Protein corona formation around nanoparticles—from the past substrate specificity that stabilizes stressed proteins in a folding-competent state.
to the future. Mater. Horiz. 1, 301–313 (2014). Biochemistry 39, 15953–15960 (2000).
13. Pelegri-O’Day, E. M., Lin, E. W. & Maynard, H. D. Therapeutic protein–polymer 43. Jenne, D. E. & Tschopp, J. Clusterin: the intriguing guises of a widely expressed
conjugates: advancing beyond PEGylation. J. Am. Chem. Soc. 136, glycoprotein. Trends Biochem. Sci. 17, 154–159 (1992).
14323–14332 (2014). 44. Thiele, L. et al. Competitive adsorption of serum proteins at microparticles
14. Gref, R. et al. ‘Stealth’ corona–core nanoparticles surface modified by affects phagocytosis by dendritic cells. Biomaterials 24, 1409–1418 (2003).
polyethylene glycol (PEG): influences of the corona (PEG chain length and 45. Winzen, S. et al. Complementary analysis of the hard and soft protein corona:
surface density) and of the core composition on phagocytic uptake and plasma sample preparation critically effects corona composition. Nanoscale 7,
protein adsorption. Colloids Surf. B 18, 301–313 (2000). 2992–3001 (2015).
15. Kim, H. R. et al. Analysis of plasma protein adsorption onto PEGylated 46. Rodriguez, P. L. et al. Minimal ‘self’ peptides that inhibit phagocytic clearance
nanoparticles by complementary methods: 2-DE, CE and protein lab-on-chip and enhance delivery of nanoparticles. Science 339, 971–975 (2013).
system. Electrophoresis 28, 2252–2261 (2007). 47. Walkey, C. D. et al. Nanoparticle size and surface chemistry determine
16. Ishida, T. et al. Injection of PEGylated liposomes in rats elicits PEG-specific IgM, serum protein adsorption and macrophage uptake. J. Am. Chem. Soc. 134,
which is responsible for rapid elimination of a second dose of PEGylated 2139–2147 (2012).
liposomes. J. Control Rel. 112, 15–25 (2006). 48. Barran-Berdon, A. L. et al. Time evolution of nanoparticle–protein corona
17. Ishida, T. & Kiwada, H. Anti-polyethyleneglycol antibody response to PEGylated in human plasma: relevance for targeted drug delivery. Langmuir 29,
substances. Biol. Pharm. Bull. 36, 889–891 (2013). 6485–6494 (2013).
18. Shah, S., Prematta, T., Adkinson, N. F. & Ishmael, F. T. Hypersensitivity to 49. Zensi, A. et al. Albumin nanoparticles targeted with Apo E enter the CNS by
polyethylene glycols. J. Clin. Pharmacol. 53, 352–355 (2013). transcytosis and are delivered to neurones. J. Control. Rel. 137, 78–86 (2009).
19. Bertrand, N. & Leroux, J. C. The journey of a drug-carrier in the body: an 50. Kreuter, J. et al. Covalent attachment of apolipoprotein A-I and apolipoprotein
anatomo-physiological perspective. J. Control. Rel. 161, 152–163 (2012). B-100 to albumin nanoparticles enables drug transport into the brain. J. Control.
20. Hamad, I., Hunter, A. C., Szebeni, J. & Moghimi, S. M. Poly(ethylene glycol)s Rel. 118, 54–58 (2007).
generate complement activation products in human serum through increased
alternative pathway turnover and a MASP-2-dependent process. Mol. Immunol. Acknowledgements
46, 225–232 (2008). T.S. and F.R.W. acknowledge support from the Max Planck Graduate Center (MPGC). G.B.
21. Amoozgar, Z. & Yeo, Y. Recent advances in stealth coating of nanoparticle and T.S. acknowledge support from the Graduate School of Excellence ‘MAINZ’ (Materials
drug delivery systems. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 4, Science in Mainz). This work was supported by the DFG/SFB1066 (‘Nanodimensionale
219–233 (2012). polymere Therapeutika für die Tumortherapie’, Q1 and Q2). F.R.W. thanks the DFG WU
22. Steinbach, T., Ritz, S. & Wurm, F. R. Water-soluble poly(phosphonate)s via 750/6-1 for funding.
living ring-opening polymerization. ACS Macro Lett. 3, 244–248 (2014).
23. Marsico, F. et al. Hyperbranched unsaturated polyphosphates as a protective Author contributions
matrix for long-term photon upconversion in air. J. Am. Chem. Soc. 136, F.R.W., V.M. and K.L. supervised the project. F.R.W., V.M., S.S. and G.B. conceived and
11057–11064 (2014). designed the experiments. G.B. and T.S. synthesized the polymers and nanoparticles. S.S.
24. Troev, K. D. Polyphosphoesters: Chemistry and Application (Elsevier, 2012). performed the cell experiments and proteomic analysis. S.W. and K.M. performed the
25. Steinbach, T. & Wurm, F. R. Poly(phosphoester)s: a new platform for degradable isothermal titration calorimetry measurements. F.R.W. and S.S. wrote the manuscript. All
polymers. Angew. Chem. Int. Ed. 54, 6098–6108 (2015). authors discussed the results and commented on the manuscript.
26. Zhang, S. Y. et al. Poly(ethylene oxide)-block-polyphosphester-based paclitaxel
conjugates as a platform for ultra-high paclitaxel-loaded multifunctional Additional information
nanoparticles. Chem. Sci. 4, 2122–2126 (2013). Supplementary information is available in the online version of the paper. Reprints and
27. Wang, Y. C. et al. Recent progress in polyphosphoesters: from controlled permissions information is available online at www.nature.com/reprints. Correspondence and
synthesis to biomedical applications. Macromol. Biosci. 9, 1154–1164 (2009). requests for materials should be addressed to V.M. and F.R.W.
28. Holzapfel, V. et al. Preparation of fluorescent carboxyl and amino functionalized
polystyrene particles by miniemulsion polymerization as markers for cells. Competing financial interests
Macromol. Chem. Phys. 206, 2440–2449 (2005). The authors declare no competing financial interests.

NATURE NANOTECHNOLOGY | VOL 11 | APRIL 2016 | www.nature.com/naturenanotechnology 377

© 2016 Macmillan Publishers Limited. All rights reserved

You might also like