Mouse Positron Emission Tomography Study of the ... - ACS Publications

Oct 18, 2016 - of AuNPs by positron emission tomography (PET). AuNPs ... mouse serum for 2 days. ..... bearing mice and evaluated by PET/CT (Figure 5)...
2 downloads 0 Views 13MB Size
Subscriber access provided by CORNELL UNIVERSITY LIBRARY

Article

A Mouse Positron Emission Tomography Study of the Biodistribution of Gold Nanoparticles with Different Surface Coatings Using Embedded Copper-64 Anders F. Frellsen, Anders E. Hansen, Rasmus I. Jølck, Paul Kempen, Gregory W Severin, Palle H. Rasmussen, Andreas Kjær, Andreas T.I. Jensen, and Thomas L. Andresen ACS Nano, Just Accepted Manuscript • DOI: 10.1021/acsnano.6b03144 • Publication Date (Web): 18 Oct 2016 Downloaded from http://pubs.acs.org on October 24, 2016

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Nano is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

A Mouse Positron Emission Tomography Study of the Biodistribution of Gold Nanoparticles with Different Surface Coatings Using Embedded Copper-64 Anders F. Frellsen1,2, Anders E. Hansen2,3, Rasmus I. Jølck2, Paul Kempen2, Gregory W. Severin1, Palle H. Rasmussen1, Andreas Kjær3, Andreas T. I. Jensen1,*, Thomas L. Andresen2,*

1. Technical University of Denmark, The Hevesy Laboratory - Center for Nuclear Technologies, 4000 Roskilde, Denmark 2. Technical University of Denmark, DTU Nanotech, Center for Nanomedicine and Theranostics, 2800 Lyngby, Denmark 3. Rigshospitalet and University of Copenhagen, Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, 2100 Copenhagen, Denmark

*Address correspondence to [email protected], [email protected].

KEYWORDS: Gold nanoparticles, Copper-64, PET, intrinsic radiolabeling, coating materials.

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ABSTRACT By taking advantage of the ability of

64

Page 2 of 47

Cu to bind non-specifically to gold surfaces, we

have developed a methodology to embed this radionuclide inside gold nanoparticles (AuNPs).

64

Cu

enables the in vivo imaging of AuNPs by positron emission tomography (PET). AuNPs have a multitude of uses within health technology and are useful tools for general nanoparticle research. 64CuAuNPs were prepared by incubating AuNP seeds with

64

Cu2+, followed by the entrapment of the

radionuclide by grafting on a second layer of gold. This resulted in radiolabeling efficiencies of 53 ± 6%. The radiolabel showed excellent stability when incubated with EDTA for two days (95% radioactivity retention) and showed no loss of

64

Cu when incubated with 50% mouse serum for two

days. The methodology was chelator-free, removing traditional concerns over chelator instability and altered AuNP properties due to surface modification. Radiolabeled 64Cu-AuNP cores were prepared in biomedically relevant sizes of 20-30 nm and used to investigate the in vivo stability of three different AuNP coatings by PET imaging in a murine xenograft tumor model. We found the longest plasma halflife (T½ about 9 h) and tumor accumulation (3.9 %ID/g) to result from a polyethylene glycol (PEG) coating, while faster elimination from the bloodstream was observed with both a Tween 20-stabilized coating and a zwitterionic coating based on a mixture of sulfonic acids and quaternary amines. In the in vivo model, the

64

Cu was observed to closely follow the AuNPs for each coating, again attributing to

the excellent stability of the radiolabel.

ACS Paragon Plus Environment

Page 3 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

Nanoparticles (NPs) have the ability to accumulate passively in a number of tumor types by the enhanced permeability and retention (EPR) effect.1,2 Gold nanoparticles (AuNPs) have a number of interesting properties for developing health technologies as they have potential as imaging agents,3 are generally biologically inert and show low toxicity,4,5 have the ability to enhance the effect of radiotherapy6,7 and photothermal therapy,8 and show high in vivo stability.9,10 In addition, AuNP surfaces are easily modified, in particular by use of gold-sulfur bonds (Au-S), which are readily formed when AuNPs are mixed with compounds bearing thiol (SH) or disulfide (S-S) functionalities.11 Longcirculating AuNPs can be prepared by coating the surface with hydrophilic polymers, such as polyethylene glycol (PEG), which hinders aggregation and recognition by the reticuloendothelial system.12 Within biomedicine, AuNPs are furthermore useful in photodynamic therapy where drugs coupled to the AuNPs are released due to laser induced thermal heating or excitation.13,14 In addition, AuNPs represent a valuable imaging tool for in vivo studies of nanoparticle pharmacokinetics and biodistribution to obtain general information on nanoparticle behavior in living systems. Accurate knowledge of the in vivo location of AuNPs can be obtained by positron emission tomography (PET), which offers high sensitivity and real-time, non-invasive quantification of wholebody biodistribution.15 PET is based on the detection of photons that arise from the annihilation of an electron (e-) with a positron (e+) emitted by a PET radionuclide. The PET radionuclide

64

Cu is

especially useful when imaging nanoparticles because of its straightforward chemistry and its considerable half-life of 12.7 h, which allows imaging in animals for up to 72 h.16 In addition, the low positron energy (Eave = 278 keV, Emax = 653 keV) of

64

Cu offers good spatial resolution, almost

identical to that of the most commonly used PET isotope 18F (Eave = 250 keV, T½ = 110 min).17 Further,

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 47

64

Cu has possible uses beyond imaging as it is considered a theranostic isotope due to its β-emission,18

and as part of a matched theranostic pair with the therapeutic isotope 67Cu.19 Traditionally,

64

Cu and other copper radioisotopes have been conjugated to NPs through

bifunctional chelators, which complex the radionuclide and are tethered to the NP-surface. 1,4,7,10Tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA),20 triethylenetetramine (TETA),21 and 1,4,7triazacyclononane-triacetic acid (NOTA)22 have been widely employed as chelators for 64Cu, since they allow relatively fast and efficient radiolabeling at room temperature. However, the in vivo stability of such chelators has been questioned,23–25 and since free liver, loss of

64

64

Cu accumulates significantly in tumors and

Cu may seriously impact the interpretation of PET images.26 As molecular imaging of

NPs in drug delivery applications primarily focuses on accurate and quantitative predictions of active substances in organs of interest, such inaccuracies are highly undesirable. Despite newer chelators, such as 2,2'-(1,4,8,11-tetraazabicyclo[6.6.2]hexadecane-4,11-diyl)diacetic acid (CB-TE2A), appearing to offer somewhat increased in vivo stability, these usually require harsh labeling conditions that are incompatible with many NPs.27,28 Furthermore, as the conjugation between the AuNP-surface and chelators is not always covalent or resistant to degradation, there is a risk of disassociation of the entire chelator-metal complex from the AuNPs. In addition, the very presence of macrocyclic chelators on NP surfaces has been implicated in altering their biodistribution.29,30 These factors have fueled a search for reliable, chelator-free alternative radiolabeling strategies and the so-called intrinsic methods are quickly gaining momentum.31,32 Within the last couple of years, a few such methodologies for 64Cu-labeling of NPs have been reported: 1) incorporation of 64Cu into iron oxide NPs using a microwave technique;33 2) formation by co-reduction of a nanoscale 64Cu-containing Cu-Au alloy in a 1:2.9 ratio;34 3) reduction

ACS Paragon Plus Environment

Page 5 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

of

ACS Nano

64

Cu2+ onto the surface of AuNPs using hydrazine;35 4) formation of AuNPs in the presence of

64

CuCl2 by reduction with sodium borohydride.36 In this article, we present methodology for chelator-free preparation of AuNPs embedded with

64

Cu and demonstrate its excellent in vitro and in vivo stability. The AuNPs are prepared exclusively in

aqueous media in biomedically relevant sizes. The method involves embedding of 64Cu in AuNP where the incorporated 64Cu is covered by a controllable layer of gold, thereby embedding it deep within the AuNPs. These characteristics eliminate the risk of disassociation of the radionuclide from the NP. As AuNPs are potentially useful as computerized tomography (CT) contrasts, the radiolabeling methodology presented here also ensures that the CT contrast provided by the AuNPs is not diluted by the addition of significant amounts of lighter atoms, such as Cu. Finally, we have used the developed radiolabeling methodology to investigate the in vivo properties and biodistribution of 64Cu-AuNPs with three different types of surface coating by PET imaging.

RESULTS AND DISCUSSION The radiolabeling methodology described in the present article was based on an observation that 64

Cu2+ adhered non-specifically to citrate-stabilized AuNPs when mixed. Initially, we imagined that

this was due to electrostatic interactions and complexation between citrate molecules and 64Cu2+ on the gold surface. However, we observed that when a piece of solid gold was added to an aqueous solution CuCl2 for 10 min, about 13% of the total

64

with additional water (84% of the associated

64

of

64

Based on this we speculated that the

64

Cu adhered to the gold and could not be washed off

Cu remained on the gold after two hours in water).

Cu2+ adhered or reacted with the gold surface itself. However,

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 47

this non-specific interaction was not stable, as the addition of ethylenediaminetetraacetic acid (EDTA) completely removed the radioactivity from the AuNP surfaces within 10 min. On this basis, we concluded that surface-bound

64

Cu was only loosely bound to the AuNP surface. Therefore, we

investigated possible methods by which to physically entrap the

64

Cu2+ and prevent sequestering by

exogenous Cu-binding agents, with the objective of preparing chelator-free 64Cu-AuNPs with no nonradioactive Cu added and with high in vivo radiolabel stability. Chelator-free

64

Cu labeling of AuNPs. The Turkevich-Frens method was chosen for AuNP

preparation since it allowed quick and simple synthesis of AuNPs in aqueous solution (Scheme 1).37,38 Using this methodology, first generation AuNP seeds (1) and second generation AuNPs grown from seeds (2) were prepared. Aqueous methods were preferred due to compatibility with simplification of the final formulation for in vivo use. Initially, AuNPs (1, 2) and

64

64

Cu2+ and

CuAuNPs (5, 7)

were prepared in refluxing water. The drawback of this approach was a somewhat polydisperse mixture of particle sizes.39 Other aqueous methods, such as sodium acrylate, provide very narrow size distributions, but take several days to carry out, which is incompatible with the 12.7 h half-life of 64Cu. However, we found that when forming the AuNPs at temperatures below boiling, as described by Nguyen et al, 40 the polydispersity index (PDI) was greatly improved. AuNP synthesis at 75 oC in water lowered PDIs from around 0.25 to around 0.10 for first generation synthesis, and from around 0.35 to about 0.15 for second generation, without a significant influence on the radiolabeling efficiency or mean intensity weighted AuNP diameter.

ACS Paragon Plus Environment

Page 7 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

Three main radiolabeling strategies were investigated (Scheme 1): 1) Passive surface labeling (4); 2) direct embedding (5, 6); 3) seeded embedding (7, 8). Passive surface labeling by adhesion was conceived as a way in which

64

Cu bound non-specifically to AuNPs could be trapped by coating the

AuNPs. This would potentially shield weakly bound synthesized and incubated with

64

64

Cu from transchelation. AuNPs (2) were

Cu2+ and subsequently coated with a 1-dodecanethiol/Tween 20

mixture to yield 4. In addition to passive surface labeling, we investigated the embedding of 64Cu inside AuNPs. Wanting to utilize the affinity of

64

Cu towards gold, we formed AuNPs in the presence of

64

Cu2+. We imagined that the 64Cu2+ would adhere to the AuNP during formation and then be trapped

inside the growing AuNP. Following this rationale, direct embedding was achieved by adding 64Cu2+ to the HAuCl4 solution at the beginning of the AuNP synthesis to yield 5 (Scheme 1a). In addition, seeded embedding was achieved by adding

64

Cu2+ to preformed AuNP seeds (1), onto which an

additional layer of gold was grafted to yield 7. In seeded embedding the 64Cu-associated AuNP seeds would act as nucleation sites for further particle growth, controlling the particle size, while simultaneously facilitating the embedding of the 64Cu (Scheme 1b). In order to prevent aggregation of citrate-stabilized AuNPs during purification by filtration, 5 and 7 were coated with a mixture of 1dodecanethiol and Tween 20 to yield 6 and 8 respectively. The radiolabeling efficiencies of 4, 6 and 8 were measured after removing unbound

64

Cu by

incubation with EDTA for 30 min (Figure 1a). It was found that the optimal procedure for 5 and 7 was to incubate with EDTA to remove loosely bound 64Cu, then coat the AuNPs to ensure colloidal stability to yield 6 and 8, whereafter

64

Cu-EDTA was removed by filtration. Control experiments showed that

64

Cu-EDTA did not associate with AuNPs coated with a 1-dodecanethiol/Tween 20 mixture, making

EDTA scavenging a suitable way to remove unincorporated

64

Cu. A radiolabeling efficiency of 26 ±

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 47

11% was obtained for the preparation of 4, and of the two embedding methods, 8 (53 ± 6%) showed a significantly higher radiolabeling efficiency than 6 (23 ± 7%). As the intention of 4 was to trap

64

Cu

under a layer of coating material, the EDTA was added after coating in this particular case. In addition, 64

Cu had been observed to associate with Tween micelles, making it necessary to centrifuge 4 and

remove the micelle-containing supernatant, before a radiolabeling efficiency could be determined. The 64

Cu associated with 6 and 8 was assumed to be embedded within the AuNPs, since EDTA did not

remove it from the citrate-stabilized 5 and 7 before coating. No difference in size (Figure 1b) and no increase in PDI (Figure 1c) were observed between 5 and 7 and their non-radioactive counterparts 1 and 2, respectively. This indicated that embedding of 64Cu2+ did not influence the size and stability of the AuNPs. Additionally, UV-VIS analysis showed similar spectra for 1 and 5 as well as for 2 and 7, with the coated AuNPs 6 and 8 showing slightly higher absorption maxima (Figure 1d & 1e). All UVVIS spectra correlated well with sizes measured by DLS.41 The size and morphology of AuNPs prepared by the methods described (1, 2) was investigated by TEM (Figure 1f & 1g). TEM images are not capable of revealing surface functionalization of the AuNPs, only the size and shape of the solid gold core. These images confirmed the DLS measurements as the diameters measured by TEM were similar (1: 14 ± 1.3 nm, 2: 29 ± 2.4 nm) to the DLS results. In addition, TEM showed that the methodology that was used resulted in AuNPs of quasi-spherical shape and narrow size-distribution (for TEM based size distribution histograms see Figure S1, Supporting Information). Stability of the 64Cu radiolabels against EDTA and serum. The stability of the radiolabels of 4 and 6 was investigated by incubation with EDTA for 2 days. The stability of 7 was investigated by coating it with all the three coatings (8, 9 and 10, Chart 1) that were later to be investigated in vivo.

ACS Paragon Plus Environment

Page 9 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

64

Cu-AuNPs and

64

Cu-EDTA were quantified by separation of the AuNPs from the EDTA fraction

using filter centrifugation (Molecular weight cut off (MWCO) 50 kDa, 7 nm) and by radioactivity-TLC analysis. Both methods gave comparable results. For 4 this revealed a constant leakage of the AuNP leading to increasing amounts of

64

64

Cu from

Cu-EDTA (Figure 2). In contrast, long-term incubation

of 6, 8, 9 and 10 with EDTA showed excellent stability of the 64Cu associated with these AuNPs. For 6, 95 ± 3% (n = 3) of the embedded radioactivity was retained in the 64Cu-AuNP fraction after 22.5 h and 90 ± 3% (n = 3) was retained after 44 h. For the coated versions of 7, 95 ± 0% (n = 3) was retained by 8 (41.5 h), 94 ± 4% (n = 3) was retained by 9 (44.0 h) and 98 ± 2% (n = 3) was retained by 10 (43.8 h) (Figure 2). This demonstrated that

64

Cu embedded inside AuNPs, by both direct and seeded

embedding, provided a remarkably stable radiolabel which was not influenced by the coating. Since seeded embedding (8) had the highest radiolabeling efficiency (53 ± 6%), this method was concluded to be superior. We suggest that this may be due to the 64Cu2+ having longer time to associate with the AuNPs compared to the direct embedding method (6), which resulted in moderate labeling efficiency (23 ± 7%). All further studies were carried out using 64Cu-AuNP prepared by seeded embedding. As an additional measure of radiolabel stability, 8 was incubated with 50% mouse serum in phosphate buffered saline (PBS) for two days at 37 oC. Mouse serum alone as well as 8 incubated with PBS were used as controls. The mixtures were separated by size-exclusion chromatography and fractions were analyzed by liquid scintillation, while protein content and AuNPs were measured by UV-VIS at 280 nm and 533 nm respectively (Figure 3). Identical radioactivity profiles were observed for

64

Cu-AuNPs with or without serum, confirming that no activity had transferred to the serum

proteins. Visually, the highest concentration of AuNPs was found in fraction 9 to 11 (these fractions also contained 87% of the total collected activity). Furthermore, the UV-VIS absorbance at 533 nm,

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 47

corresponding to the surface plasmon resonance peak of the AuNPs, matched the radioactivity in all fractions (See Figure S2, Supporting Information), which confirmed that the activity remained associated with the 64Cu-AuNPs. Further investigations of seeded embedding. As a means to improve embedding and possibly elucidate the embedding mechanism, non-radioactive Cu2+ as a percentage of the total amount of gold was added during seeded embedding. This did not improve the radiolabeling; on the contrary, radiolabeling efficiencies decreased when increasing amounts of nonradioactive CuCl2 were added. This can be overcome by reduction to elemental Cu,35,42 but as no reductants for Cu2+ were added in our system, Cu2+ could be expected to retain its oxidation state. Even when nonradioactive Cu2+ was added in amounts that could not cover the entire AuNP surface, the radiolabeling efficiency still decreased. This indicates that

64

Cu2+ may only associate with specific areas of the AuNP surface and

that these areas can be occupied by extra Cu2+. Alternatively, the increased presence of Cu2+ in the reaction mixture may disturb the embedding process (for an expanded discussion of this topic, see Supporting Information). Increasing the radiolabeling efficiency was attempted by adding the chelators EDTA and DOTA during seeded embedding. However, the Turkevich/Frens method of synthesizing AuNPs is relatively sensitive and the addition of EDTA or DOTA to the reaction mixture did not improve radiolabeling yield, but interfered with the formation of AuNPs, thus increasing the PDI to >0.30. Accordingly, our experiences were in contradiction with previous reports suggesting that addition of EDTA would improve the Turkevich synthesis of AuNPs.43

ACS Paragon Plus Environment

Page 11 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

While we used as small amounts of gold as possible in order to achieve the highest possible specific activity, we investigated the scalability of the seeded embedding reaction. We obtained similar results at 100 mL/15 mg HAuCl4 as at for 10 mL/1.5 mg HAuCl4 scale, indicating that the procedure is scalable. The highest specific activity we obtained was 438 MBq/mg of gold and we have seen no indications of reaching an upper limit of how much activity it is possible to embed. The final gold concentration could be adjusted by concentrating the coated AuNPs on a centrifuge filter and redispersing in the desired volume, as described in the experimental section. Preparation of coated

64

Cu-AuNPs for in vivo evaluation (8, 9 and 10). Utilizing the

developed seeded-embedding methodology we prepared labeled 64Cu-AuNPs for in vivo investigations to study the pharmacokinetics and biodistribution of the AuNPs with three distinct surface coatings, the molecular structures of which are given in Chart 1. Please note that 8 in Chart 1 is identical to 8 in Scheme 1. A single batch of 7 was prepared, which was coated to give 8, 9 and 10 and compared in vivo by PET/CT imaging. This ensured that the observed biodistributions did not originate from secondary factors, such as differences in size, morphology or embedding, due to batch-to-batch variations, i.e. only the coatings are different on 8, 9 and 10. AuNP 8 was prepared by coating 7 with a 1:2 mixture of 1-dodecanethiol and Tween 20 as previously described by Hainfeld.44 Surface coating with a combination of Tween 20 and 1dodecanethiol has been reported to result in biocompatible AuNPs with excellent stability in media of high ionic strength as well as in thiolytic environments, due to the formation of a hydrophobic bilayer protecting the AuNP core.44 Such particles have been employed for in vivo use with a circulation halflife of about 2 h.44 Accordingly, the coating on 8 results in a relatively short in vivo half-life. AuNP 9 was prepared by coating 7 with MeO-PEG5000-SH. PEGylation of AuNPs and other NPs is a common

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 47

technique to prevent opsonization and thereby enhance circulation half-life. MeO-PEG5000-SH was chosen since this polymer size has previously been described as optimal for obtaining intermediate circulation half-life AuNPs (10-20 h).45 Finally, 10 was prepared by coating 7 with a 1:1 mixture of sodium 10-mercaptodecane-sulfonic acid and (10-mercaptodecyl)trimethylammonium bromide. Mixed charge zwitterionic surface modification of AuNPs have showed excellent stability in physiological conditions in vitro and have been reported to be highly biocompatible.46 The phagocytic resistance of the zwitterionic AuNPs was shown to be superior compared to PEGylated AuNPs when evaluated in vitro.46,47 It has previously been reported that zwitterionic AuNPs have a circulation half-life of about 10-30 h.48,49 Accordingly, such AuNPs can be characterized as long circulating. Inclusion of these three distinct surface coating with expected short-, intermediate and long circulating properties enabled an efficient evaluation of the developed chelator-free seeded-embedding methodology. For the in vivo study, 8, 9 and 10 (Chart 1) were incubated with EDTA and purified by filtration to remove unincorporated

64

Cu. Centrifugation was used to remove excess coating materials and to

concentrate the 64Cu-AuNPs. For all three coatings, the pellets containing the 64Cu-AuNPs were easily redispersed in aqueous media. Characterization of the final in vivo formulations is shown in Table 1, with TEM images shown in Figure 4. The TEM images revealed homogenous populations of

64

Cu-

AuNPs in all three cases with mean core diameters ranging from 26.6 to 28.3 (Figure 4A-C), which was comparable to the DLS derived mean intensity-weighted diameter for the uncoated 2nd generation 64

Cu-AuNP (7) of 24.2 ± 7.7 nm. In addition, visualization by negative staining showed the coating

coronas, with the zwitterionic coating (10) having a thickness of 1.3 ± 0.3 nm (n = 50) and the PEG coating (9) a thickness of 5.9 ± 2.1 nm (n = 50). The PEG coating being slightly thicker than the zwitter

ACS Paragon Plus Environment

Page 13 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

coating is expected from the size of the coating molecules. The large diameter of 9 observed by DLS (73 ± 6) is considered to be caused by a larger hydrodynamic diameter due to the PEG hydration. The 1-dodecanethiol/Tween 20 coating (8) was not visible, which is considered to be due to the sample drying process during TEM imaging. All coated

64

Cu-AuNPs exhibited increased PDI and

hydrodynamic diameter as compared to 7, when measured by DLS. This was most pronounced for 9, as a result of the comparatively larger PEG5000 polymer, while 10 was larger than 8, possibly due to a increased hydrated shell retained by the more charged nanoparticle surface (for UV-VIS spectra see Figure S3, Supporting Information). Modest PDI values and the TEM analysis for all three preparations did not show sign of aggregation of the nanoparticles. After coating, all 64Cu-AuNPs were in the range 10-100 nm, where long-circulating, untargeted NPs, can be expected to show passive tumor accumulation and limited initial clearance by liver, spleen and kidneys.23,29,50 Table 1: Characterization of 64Cu-AuNPs used for in vivo investigation. Size, absorption spectra and zeta potentials were measured in PBS. Diameter

PDI

(nm)

Abs. max.

Zeta pot.

Radioactivity

Au

(nm)

(mV)

(MBq/mL)

(mg/mL)

8

25 ± 5

0.18 ± 0.01

533

-7.0 ± 6.9

11.4

0.38

9

73 ± 6

0.29 ± 0.01

532

-22 ± 2.9

6.21

0.27

10

40 ± 9

0.20 ± 0.01

531

-31 ± 1.7

9.66

0.37

In vivo investigation of 8, 9 and 10 (Chart 1). 64Cu-AuNPs were administered intravenously in FaDu tumor-bearing mice and evaluated by PET/CT (Figure 5). Accumulation values are reported as percent injected dose per gram tissue (%ID/g). The injected amounts of Au and 64Cu were 76 µg and 2.28 MBq for 8, 54 µg and 1.24 MBq for 9, and 74 µg and 1.93 MBq for 10, respectively. This corresponds to

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 47

each animal receiving 2.79·1011 NPs for 8, 1.98·1011 NPs for 9, and 2.71·1011 NPs for 10. The amount of Au was slightly lower for 9, which could slightly increase the first-pass clearance due to opsonization.51 However, we did not see any evidence of this in our results as will be seen below. PETderived accumulation in tumors, blood, liver and spleen at 1, 4 and 24 h post-injection (Figure 5a) mirrored the ex vivo biodistribution data obtained by well-counting of organs excised at 24 h (Figure 6). Notable differences were observed between the three coatings. The 1-dodecanethiol/Tween 20 coated

64

Cu-AuNPs (8) showed relatively rapid elimination from the bloodstream, being almost

completely cleared at 4 h (2.3 ± 1.1%ID/g), when compared to long-circulating NPs, such as liposomes, polymeric micelles and other AuNPs, where plasma half-lives of 10-30 h in murine models are observed.23,29,35,52 Hepatic uptake was substantial and rapid (29.9 ± 1.2%ID/g at 4h), while tumor and spleen accumulation were both modest after 24 h (1.29 ± 0.1%ID/g and 2.2 ± 0.2%ID/g, respectively), likely as a result of the limited circulation time. Prolonged circulation is a prerequisite for passive accumulation of NPs in tumors.2 The observed rapid clearance is in accordance with the reported plasma half-life on the order of a few hours.44 In addition, Hainfeld reported low liver contrast immediately following injection,44 which is also in accordance with our observation that hepatic accumulation of 8 is a gradual process with an increase from 1 h to 4 h. It may be speculated that the relatively high critical micelle concentration (CMC) of 99 mg/L and hydrophilic-lipophilic balance (HLB) number of 16.7 of the stabilizing Tween 20,53 could cause it to detach from the

64

Cu-AuNPs

under in vivo sink conditions. This would expose the lipophilic surface to interaction with blood stream components, such as opsonins, causing relatively rapid liver uptake and clearance from blood. As the

ACS Paragon Plus Environment

Page 15 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

only

ACS Nano

64

Cu-AuNPs investigated here, 8 showed accumulation in lymph nodes (for magnification of

lymph node accumulation, see Figure S4, Supporting Information). The PEG-coated

64

Cu-AuNPs (9) showed improved circulation compared to 8 with an

approximate plasma half-life (exponential function estimate based on three blood data points) of 9 h. This corresponds well with reported values for long-circulating NPs in murine models52 and literature data for this type of coating.45 Liver uptake of 9 was limited and relatively constant throughout the imaging period (9.0-9.8%ID/g), while uptake in spleen was low at first but reached a high level of 29.3 ± 2.2%ID/g at 24 h. The tumor uptake of 9 was rising throughout the imaging period and reached a level of 3.89 ± 0.1%ID/g at 24 h. This also corresponds well with reported values for long-circulating NPs such as liposomes.23,52 Accordingly, the PEG-coating (9) appears to be stable in vivo and to confer long-circulating properties to the 64Cu-AuNPs. The sulphonate / quaternary ammonium coated 64Cu-AuNPs (10) exhibited the fastest elimination from the blood stream of the three coatings that were investigated. At the 1 h time point, only 5.1 ± 1.2 %ID/g remained in the blood stream, which was markedly lower than the other two coatings. At later time points, very little radioactivity was present in the blood. 10 appeared to rapidly locate to the liver, with accumulation reaching 41.6 ± 3.3 %ID/g within 1h and thereafter remaining relatively constant throughout the imaging period. Virtually no accumulation in spleen or tumor was observed, suggesting that hepatic sequestration was too rapid for uptake in these organs to take place. These results are in contradiction with the literature where a circulation half-life of 30 h have been reported for a system identical to the one we have tested48 and a half-life about 6-8 h being observed for a similar system using carboxylic acids instead of sulphonates.49 The reason for this discrepancy is currently unknown but further research may clarify the matter.

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 47

In addition to PET/CT and well counting, we measured the gold content in selected organs by digestion in aqua regia followed by ICP-OES measurement. We hypothesized that if the

64

Cu was

stably embedded within the AuNPs, we could expect a correlation between %ID/g of 64Cu and %ID/g of Au. This correlation was found to exist for all three coatings. The stable association of the 64Cu with the AuNPs is further underlined by the relatively different biodistributions of 8, 9 and 10 which were reflected by both 64Cu and Au (Figure 6). Free 64Cu would be expected to primarily accumulate in the liver (20-30%ID/g after 1 h, falling to 10-15%ID/g at 22 h) and tumor (around 5%ID/g after 22 h in FaDu tumors).26 This is fairly similar to long-circulating NPs, which is often a point of critique when using

64

Cu to image these, although free

64

Cu is removed faster from the blood stream than long-

circulating NPs. However, here we demonstrate that the

64

Cu closely follows the Au in three unique

biodistributions of AuNPs, which underlines that the 64Cu is indeed stably embedded with the AuNPs. Final remarks. In the present article, we have described methods to stably embed

64

Cu within

AuNPs. Using this system, we showed that 64Cu-AuNPs coated with PEG thiols (9) exhibited superior long-circulating properties and better passive tumor accumulation than

64

Cu-AuNPs coated with 1-

dodecanethiol/Tween 20 (8) or a zwitterionic sulphonate / quartenary ammonium mixture (10). Since our prepared

64

Cu-AuNPs do not have chelators attached to the surface nor are subjected to reductive

conditions post-formation, they can be freely modified with other compounds, such as targeting ligands, and their biodistribution can be expected to mirror that of non-radiolabeled counterparts. 64CuAuNPs have potential uses as a theranostic tool to predict or monitor the biodistribution of AuNPs administered for other purposes, such as photothermal ablation. In thermal ablation however, a uniform distribution of AuNPs within the tumor tissue may be desirable in order to obtain a homogenous delivery of heat. Since our results indicate relatively large intratumoral variation in accumulation

ACS Paragon Plus Environment

Page 17 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

(Figure 5c) with high local maxima (Figure 5d), the effects of using systemically administered AuNPs for this purpose require detailed investigations. As a biologically inert system, 64Cu-AuNPs will remain intact in vivo and when the 64Cu is stably embedded, the system will accurately report the general in vivo trafficking of NPs, both whole-body and inter-organ, making it a very useful tool in NP research. Another possible use for AuNPs is as CT agents. However, the amount of gold needed to obtain sufficient CT contrast by systemic administration is most likely prohibitive to human use, because large amounts would accumulate in liver and spleen and remain there for extended periods of time.54 A way to circumvent this problem is local administration of the AuNPs e.g. fixed in a gel, in order to obtain a sufficient gold concentration.55

CONCLUSION By adding [64Cu]CuCl2 to AuNPs during their formation, we have developed a chelator-free method for stable embedding of the PET isotope 64Cu inside AuNP cores. Seeded embedding by incubation of 64Cu with gold seeds followed by growth of a further gold layer gave superior radiolabeling yields of 53% compared to 23% for direct radiolabeling of AuNP seeds. The labeling was stable against EDTA and serum challenge as well as in vivo. This method allows for 64Cu-AuNPs to be imaged by PET without interference from unbound 64Cu. In addition, we used the developed 64Cu-AuNPs to investigate the in vivo properties of three distinct coatings for AuNPs in FaDu tumor-bearing mice. In our hands, a PEG5000-coating attached by thiols performed better in terms of long circulation and tumor accumulation than a coating stabilized by Tween 20 or a zwitter-ionic coating based on a 1:1 mixture

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 47

of sulphonates and quaternary ammoniums. Nanoparticle coatings are attracting increasing attention as the role of protein absorption to nanoparticle surfaces forming a protein carona is being recognized as highly important and may drive a range of biological effects.56 Future work should aim to couple studies on nanoparticle biodistribution as a function of surface coatings with quantitative analysis of protein binding and carona formation. It has been found that zwitter-ionic surfaces can lower protein binding and may furthermore effect tumor targeting,57 however, in the present study we observed PEG coatings were superior. Another important aspect is potential antibody responses, IgM and IgG, which will be dependent on the nanoparticle surface coatings and may prevent multiple injections of the same particle type.

MATERIALS AND METHODS Further experimental details are available in Supporting Information, including materials, characterization of AuNPs and synthesis of coating materials. Synthesis of first generation AuNP seeds (1). The method was adapted from literature procedures.37,40 A solution of HAuCl4·4H2O (14.8 mg, 35.9 µmol) in Milli-Q water (100 mL) was prepared. The solution was heated to 75 °C under vigorous stirring and aq. trisodium citrate (8.00 mL, 288 µmol, pH 7.0) was added. The mixture was stirred for 60 min at 75 °C. The resulting dark red dispersion was heated to reflux for 30 min followed by cooling to room temperature, furnishing citrate stabilized first generation AuNPs (0.066 mg/mL) (1).

ACS Paragon Plus Environment

Page 19 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

Synthesis of second generation AuNPs (2), followed by C12H25-SH/Tween 20-coating (3). The method was adapted from literature procedures.37,40 AuNP seeds (1) (20 mL, 0.14 mg/mL, pH 6.0) were mixed with Milli-Q water (80 mL). To the dispersion was added aq. trisodium citrate (2.0 mL, 72 nmol, pH 7.0). The reaction mixture was heated to 75 °C under vigorous stirring. Then aq. HAuCl4·4H2O (15.0 mL, 18.5 mg, 45.0 µmol) was added dropwise to the warm dispersion. The reaction mixture was then heated to reflux for 30 min, and subsequently cooled to room temperature, furnishing a dispersion of citrate-stabilized second generation AuNPs (0.087 mg/mL) (2). Coating was achieved by quick addition of a mixture of 1% (v/v) 1-dodecanethiol in ethanol (1.00 mL, 41.7 nmol) and 1% (v/v) Tween 20 in ethanol (2.00 mL, 17.8 nmol) under vigorous stirring. Stirring was continued for 10 min furnishing the coated second generation AuNPs (3). Preparation of 64Cu-AuNPs by passive surface labeling with C12H25-SH/Tween 20 coating (4). To a vial containing dried [64Cu]CuCl2 was added aqueous NH4OAc buffer (100 µL, 0.1 M, pH 5.5), Milli-Q water (700 µL) and a dispersion of citrate-stabilized second generation AuNPs (2) (200 µL, 0.087 mg/mL). The reaction mixture was magnetically stirred for 10 min at room temperature and 1% (v/v) 1-dodecanethiol solution in ethanol (24 µL, 1.00 nmol) and 1% (v/v) Tween 20 solution in ethanol (48 µL, 0.428 nmol) were mixed and added quickly under vigorous stirring. Stirring was continued for 10 min, after which the reaction mixture was mixed with aq. tripotassium EDTA (50 µL, 100 mM, 5 µmol, pH 7.5) and incubated for 30 min, before measuring the labeling yield and stability. Preparation of

64

Cu-AuNPs by direct embedding (5), followed by C12H25-SH/Tween 20

coating (6). A solution of HAuCl4·4H2O (1.5 mg, 3.6 µmol) and [64Cu]CuCl2 in Milli-Q water (10 mL) was prepared. The solution was heated to 75 °C under vigorous stirring. To the warm solution was added aq. trisodium citrate (800 µL, 28.8 µmol, pH 7.0). The yellow solution changed color through

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 47

faint pink to a red dispersion. After stirring for 40 min at 75 °C the dispersion was heated to reflux for 20 min, followed by cooling to room temperature. The resulting 64Cu-AuNPs (5) were mixed with aq. tripotassium EDTA (100 µL, 10.0 µmol, pH 7.5) and incubated for 30 min. Then a mixture of 1% (v/v) 1-dodecanethiol in ethanol (100 µL, 4.17 nmol) and 1% (v/v) Tween 20 in ethanol (200 µL, 1.78 nmol) were added under vigorous stirring, furnishing C12-SH/Tween 20 coated first generation

64

Cu-AuNPs

(6). Stirring was continued for 10 min before measuring the labeling yield and stability. Preparation of

64

Cu-AuNPs by seeded embedding (7), followed by C12H25-SH /Tween 20

coating (8). AuNP seeds (1) (20.0 mL, 0.14 mg/mL, pH 6.0)) were mixed with Milli-Q water (80 mL) and aq. trisodium citrate (2.0 mL, 72 µmol, pH 7.0) followed by addition of [64Cu]CuCl2 to the resulting mixture. The mixture was incubated for 20-30 min during heating to 75 °C under vigorous stirring. This was followed by dropwise addition of HAuCl4·4H2O (18.5 mg, 44.9 µmol) in Milli-Q water (15 mL). During the addition the color changed from red to pink. The dispersion was heated to reflux for 20 min causing a color change to purple and then dark red, followed by cooling to room temperature. The resulting 64Cu-AuNPs (7) were then incubated with aq. tripotassium EDTA (100 µL, 100 mM, 10.0 µmol, pH 7.5) for 30 min. Then 1% (v/v) 1-dodecanethiol in ethanol (1.00 mL, 41.7 nmol) and 1% (v/v) Tween 20 in ethanol (2.00 mL, 17.8 nmol) were mixed and added quickly under vigorous stirring, furnishing C12-SH/Tween 20 coated second generation 64Cu-AuNPs (8). Stirring was continued for 10 min before measuring the labeling yield and stability. Test of radiolabeling stability against EDTA. After preparation, 64Cu-AuNPs were mixed with aq. tripotassium EDTA (100 µL, 100 mM, 10.0 µmol, pH 7.5) and incubated for 30 min to sequester any unincorporated 64Cu. Separation was obtained by centrifugation (15 min, 4500 G) in Amicon Ultra centrifuge filters (MWCO 50 kDa, 7 nm). The radioactivities (A) in the filtrate (small molecules incl.

ACS Paragon Plus Environment

Page 21 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

64

Cu-EDTA) and the filtrand (64Cu-AuNPs) were compared to provide the radiolabeling efficiency

(rad.eff. = Afiltrand / (Afiltrate + Afiltrand)). The radiolabeled AuNPs were then re-dispersed in aq. tripotassium EDTA (10 mL, 10 mM, pH 7.5). For approximately 48 h the stability was monitored by spinning down the AuNPs through centrifuge filters and measuring the activities in filtrand and filtrate, to show any disassociated 64Cu as 64Cu-EDTA. After each centrifugation the AuNPs were re-dispersed in aq. tripotassium EDTA (10 mL, 10 mM, pH 7.5). In addition to centrifuge filtration, the labeling stability was determined by analyzing the mixture on radio-TLC using 5% (w/v) NH4OAc in H2OMeOH (1:1). Radioactivity at the origin was reported as associated with the 64Cu-AuNPs, while 64CuEDTA represented unbound 64Cu. Test of radiolabeling stability in serum. The method was adapted from Seo et al.29 Freshly prepared 64Cu-AuNPs (8) (1.8 mg) dispersed in phosphate buffered saline (PBS) (0.5 mL, pH 7.4) were mixed with mouse serum (0.5 mL) and incubated for two days at 37 C. Then an aliquot of the mixture (100 µL) was applied to a size-exclusion chromatography column, packed with Sephacryl-300HR, and eluted with PBS-buffer – 40 fractions (~1.5 mL/each) were collected. The UV absorption at 280 nm of each fraction was measured. From each fraction was taken a sample (100 µL) that was mixed with liquid scintillation (LSC) cocktail (10 mL) and counted on a liquid scintillator. Two control experiments were carried out in the same way as described above: 1) Mouse serum (0.5 mL) was mixed with PBS (0.5 mL), 2) 64Cu-AuNPs (8) (1.8 mg) dissolved in phosphate buffered saline (PBS) (1.0 mL, pH 7.4). Preparation of coated

64

Cu-AuNPs for in vivo evaluation (8, 9 and 10). Citrate-stabilized

64

Cu-AuNPs (7) were prepared as described above, and then split into three portions prior to coating.

Three different coatings were used: 8) A 2:1 mixture of 1-dodecanethiol and Tween 20, 9) MeO-

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

PEG5000-SH,

10)

A

1:1

mixture

of

sodium

10-mercaptodecane-sulfonic

Page 22 of 47

acid

and

(10-

mercaptodecyl)trimethylammonium bromide. The materials for coating 10 were synthesized as described in Supporting Information (for 1H NMR spectra see Figure S4 and S5). Formulation 8 was obtained as previously described by premixing 1% (v/v) ethanol solutions of 1-dodecanethiol (205 µL, 8.55 nmol) and Tween 20 (410 µL, 3.65 nmol) then mixing with the 64Cu-AuNPs (8.2 mL, c(Au) 0.075 mg/mL). Formulation 9 was obtained by stirring the 64Cu-AuNPs (8.2 mL, c(Au) 0.075 mg/mL) with aq. MeO-PEG5000-SH (85 µL, 85 nmol), which corresponds to eight PEG molecules pr. nm2 of gold surface. According to Wuelfing et al. the “footprint” of one PEG-S-ligand is 0.35 nm², while the “footprint” of one 1-dodecanethiol ligand is 0.21 nm².58 This gives a moderate excess of 1.7 times surface saturation for 8 and 2.8 times the surface saturation for 9. Formulation 10 was prepared by mixing the

64

Cu-AuNPs (8.2 mL, c(Au) 0.075 mg/mL) with a 1:1 aqueous solution (82 µL) of the

sulphonate (50 mM) and quaternary ammonium lipids (50 mM), which corresponds to 495 thiols pr. nm² or 100 times surface saturation of thiols. The large excess of thiols used for 10 was due to observed instability, when using less material. After coating, the 64Cu-AuNP centrifuge filters (MWCO 50 kDa, 7 nm) were used to remove

64

Cu-EDTA. Excess coating material was removed by

centrifugation (2x35 min at 17000G) each time removing the supernatant and re-dispersing the AuNPpellet in Milli-Q water. Finally the

64

Cu-AuNP dispersions were concentrated 6.5 times and re-

dispersed in physiological phosphate buffer solution (NaCl (8.0 g), KCl (0.20 g), Na2HPO4·12H2O (2.9 g), KH2PO4 (0.2 g) in 1000 mL Milli-Q water (pH 7.4) to give a final concentration of gold of approximately 0.5 mg/mL. Animal Model. Female Naval Medical Research Institute (NMRI) nude mice, five weeks old (Taconic Europe, Lille Skensved, Denmark) were acclimatized for one week and had free access to

ACS Paragon Plus Environment

Page 23 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

water and chow. Human pharyngeal squamous cell carcinoma (FaDu) cells, purchased from American Type Culture Collection (ATCC), were cultured in Minimum Essential Medium (MEM) with Earle's salts and sodium bicarbonate supplemented with 2 mM L-glutamine (both from Sigma-Aldrich), 1 mM sodium pyruvate, 1% MEM non-essential amino acids solution (100 ×), 10% fetal calf serum, 100 units/mL penicillin and 100 µg/mL streptomycin (all from Invitrogen) at 37 °C in 5% CO2. Mice were sc. inoculated with 5 x 106 FaDu cells suspended in 100 µL (1:1 cell culture medium and BD™ Matrigel™ (VWR, Søborg, Denmark)) on both flanks. Tumors were allowed to grow for 18 days reaching sizes of 393 ± 221 mm3. Mice were randomized on to three groups of five mice to receive 8, 9 or 10. All animal experiments were approved by the Danish Animal Welfare Council, Ministry of Justice. PET/CT imaging procedures. PET/CT imaging was performed 1, 4 and 24 h after injection of 8, 9 or 10. Mice were anesthetized by inhalation anesthesia (~3.5% sevoflurane in 40% O2 and medial grade air) and 64Cu-AuNPs were injected into a tail vein. Mice had 2.76 mg/kg Au (8), 1.98 mg/kg Au (9), 2.72 mg/kg Au (10) injected, equaling a mean (±SD) [64Cu] activity of 16.7 ± 0.46 MBq (8), 9.1 ± 0.60 MBq (9) and 14.0 ± 0.41 MBq (10).

64

Cu-AuNPs were allowed to distribute for 1 h before

commencing a 5-min PET scan (termed 1-hour scan) with the mouse under anesthesia and positioned on a scanning bed. After completion the scanning bed was moved into the CT field-of-view for CTscanning. After the scanning procedure mice were allowed to cage rest until performing an identical PET/CT scan at 4 h (termed 4-hour scan, 5 min PET acquisition) and again at 24 h (termed 24-hour scan, 15 min PET acquisition) after injection of 64Cu-AuNPs. All imaging procedures were performed using inhalation anesthesia as described above. PET/CT imaging was performed on a dedicated Inveon® small animal PET/CT system with CT based PET image attenuation (Siemens Medical

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 47

Systems, PA, USA). PET scans were reconstructed using a maximum a posteriori (MAP) reconstruction algorithm (0.815 x 0.815 x 0.796 mm, resolution; 1.2 mm). Emission data were corrected for dead time and decay and attenuation correction was performed based on the corresponding CT scan. Image analysis was performed using commercially available Inveon® software (Siemens Medical Systems, PA, USA). 3D Regions of interest (ROIs) were manually constructed and decay corrected data (%injected dose pr. gram tissue (%ID/g)) reported. Ex vivo autoradiography and [64Cu] gamma counting. After completion of the 24-hour PET/CT scans, mice were terminally perfused under inhalation anesthesia (~5% sevoflurane in 40% O2 and medial grade air) with >40 ml of isotonic saline solution. Tumors were removed by dissection and one half of two tumors from each 64Cu-AuNP group was snap frozen in dry-ice cooled isopentane and mounted in freezing media (Tissue Tek® Sakura Finetek, USA). Tumors were subsequently cryosectioned in multiple 8 µm sections and thaw mounted on microscopy plus slides (Superfrost Ultra Plus® Thermo Scientific, Germany). Semi-quantitative autoradiography images of microregional 64CuAuNP distribution were acquired by exposing tumor sections to a phosphor imaging screen for 90 min and developed using a phosphor imaging system (Cyclone Plus Phosphor Imager, Perkin Elmer, Waltham, MA., USA) to form semi-quantitative photo-stimulated luminescence images . Tumor sections were subsequently stored in PBS until further analysis. The 64Cu levels in tumors and tissues of interests were determined by gamma counting the excised tissues. The following tissues were analyzed; Blood, tumor, liver, spleen, kidney, lung, muscle, heart, brain, intestine. Tissues were placed in pre-weighed well-counting tubes and well-counted for 120 seconds using a [64Cu] calibrated automatic gamma counter, (Wizard2TM3, Perkin Elmer, Waltham, MA., USA). Tumors and tissues were stored at -80 °C until further analysis.

ACS Paragon Plus Environment

Page 25 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

Ex vivo ICP-MS measurements of gold in tissue samples. Tissue samples from tumor, liver, spleen and muscle from each mouse were analyzed. Tissue samples of approximately 100 mg were cut and weighed in 15 mL plastic tubes. To each tube was added: HNO3 (500 µL, 16 M), H2O2 (300 µL, 10 M) and HCl (50 µL, 11 M). The samples were incubated at 65 °C for 12 h to dissolve the tissue. To the digested samples were added H2O (10 mL) pr. tube. The water contained 0.5 ppb iridium as an internal reference. Dilutions of the digested tissue samples were prepared. For tumor and muscle a 10x dilution was prepared. For liver and spleen a 100x dilution was prepared. Diluting was done with a 2% (w/v) HCl solution containing 0.5 ppb iridium as an internal standard. A standard curve for gold was prepared, and each sample was measure by ICP-MS. The gold concentration in each kind of tissue was calculated as an average %ID/g for each of the three groups of mice injected with 8, 9 or 10 respectively.

Conflict of interest: The authors declare no competing financial interests. Acknowledgments: This research was funded by the Danish Council for Strategic Research, the European Research Council Excellence (ERC) programme and the Lundbeck foundation. We would like to thank Søren Bredmose Simonsen (DTU Energy) for TEM measurements and Dennis Ringkjøbing Elema (DTU Nutech) for supplying 64CuCl2. Supporting information available: Figures S1, S2, S3, S4 and S5 included, along with more experimental details. This material is available free of charge via the Internet at http://pubs.acs.org.

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 26 of 47

REFERENCES (1)

Davis, M. E.; Chen, Z. G.; Shin, D. M. Nanoparticle Therapeutics: An Emerging Treatment Modality for Cancer. Nat. Rev. Drug Discov. 2008, 7, 771–782.

(2)

Matsumura, Y.; Maeda, H. A New Concept for Macromolecular Therapeutics in Cancer Chemotherapy: Mechanism of Tumoritropic Accumulation of Proteins and the Antitumor Agent Smancs. Cancer Res. 1986, 46, 6387–6392.

(3)

Kim, D.; Park, S.; Lee, J. H.; Jeong, Y. Y.; Jon, S. Antibiofouling Polymer-Coated Gold Nanoparticles as a Contrast Agent for In Vivo X-Ray Computed Tomography Imaging. J. Am. Chem. Soc. 2007, 129, 7661–7665.

(4)

Connor, E. E.; Mwamuka, J.; Gole, A.; Murphy, C. J.; Wyatt, M. D. Gold Nanoparticles Are Taken up by Human Cells but Do Not Cause Acute Cytotoxicity. Small 2005, 1, 325–327.

(5)

Goodman, C. M.; McCusker, C. D.; Yilmaz, T.; Rotello, V. M. Toxicity of Gold Nanoparticles Functionalized with Cationic and Anionic Side Chains. Bioconjug. Chem. 2004, 15, 897–900.

(6)

Lee, J.; Chatterjee, D. K.; Lee, M. H.; Krishnan, S. Gold Nanoparticles in Breast Cancer Treatment: Promise and Potential Pitfalls. Cancer Lett. 2014, 347, 46–53.

(7)

Hainfeld, J. F.; Slatkin, D. N.; Smilowitz, H. M. The Use of Gold Nanoparticles to Enhance Radiotherapy in Mice. Phys. Med. Biol. 2004, 49, N309–N315.

(8)

Kennedy, L. C.; Bickford, L. R.; Lewinski, N. a; Coughlin, A. J.; Hu, Y.; Day, E. S.; West, J. L.;

ACS Paragon Plus Environment

Page 27 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

Drezek, R. a. A New Era for Cancer Treatment: Gold-Nanoparticle-Mediated Thermal Therapies. Small 2011, 7, 169–183. (9)

Alkilany, A. M.; Lohse, S. E.; Murphy, C. J. The Gold Standard: Gold Nanoparticle Libraries to Understand the Nano-Bio Interface. Acc. Chem. Res. 2013, 46, 650–661.

(10)

Dykman, L.; Khlebtsov, N. Gold Nanoparticles in Biomedical Applications: Recent Advances and Perspectives. Chem. Soc. Rev. 2012, 41, 2256–2282.

(11)

Yeh, Y.-C.; Creran, B.; Rotello, V. M. Gold Nanoparticles: Preparation, Properties, and Applications in Bionanotechnology. Nanoscale 2012, 4, 1871–1880.

(12)

Klibanov, a L.; Maruyama, K.; Torchilin, V. P.; Huang, L. Amphipathic Polyethyleneglycols Effectively Prolong the Circulation Time of Liposomes. FEBS Lett. 1990, 268, 235–237.

(13)

Sershen, S.; Westcott, S. Temperature-sensitive Polymer-Nanoshell Composites for Photothermally Modulated Drug Delivery. J. Biomed. Mater. Res. 2000, 51, 293–298.

(14)

Tuchina, E. S.; Tuchin, V. V; Khlebtsov, B. N.; Khlebtsov, N. G. Phototoxic Effect of Conjugates of Plasmon-Resonance Nanoparticles with Indocyanine Green Dye on Staphylococcus Aureus Induced by IR Laser Radiation. Quantum Electron. 2011, 41, 354–359.

(15)

Willmann, J. K.; van Bruggen, N.; Dinkelborg, L. M.; Gambhir, S. S. Molecular Imaging in Drug Development. Nat. Rev. Drug Discov. 2008, 7, 591–607.

(16)

Cai, W.; Wu, Y.; Chen, K.; Cao, Q.; Tice, D. a; Chen, X. In Vitro and In Vivo Characterization of 64Cu-Labeled Abegrin, a Humanized Monoclonal Antibody against Integrin Alpha v Beta 3.

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 28 of 47

Cancer Res. 2006, 66, 9673–9681. (17)

Williams, H. A.; Robinson, S.; Julyan, P.; Zweit, J.; Hastings, D. A Comparison of PET Imaging Characteristics of Various Copper Radioisotopes. Eur. J. Nucl. Med. Mol. Imaging 2005, 32, 1473–1480.

(18)

Yoshii, Y.; Furukawa, T.; Kiyono, Y.; Watanabe, R.; Mori, T.; Yoshii, H.; Asai, T.; Okazawa, H.; Welch, M. J.; Fujibayashi, Y. Internal Radiotherapy with Copper-64-Diacetyl-Bis (N4Methylthiosemicarbazone) Reduces CD133+ Highly Tumorigenic Cells and Metastatic Ability of Mouse Colon Carcinoma. Nucl. Med. Biol. 2011, 38, 151–157.

(19)

Muller, C.; Bunka, M.; Haller, S.; Koster, U.; Groehn, V.; Bernhardt, P.; van der Meulen, N.; Turler, A.; Schibli, R. Promising Prospects for 44Sc-/47Sc-Based Theragnostics: Application of 47Sc for Radionuclide Tumor Therapy in Mice. J. Nucl. Med. 2014, 55, 1658–1664.

(20)

Shokeen, M.; Anderson, C. J. Molecular Imaging of Cancer with Copper-64 Radiopharmaceuticals and Positron Emission Tomography (PET). Acc. Chem. Res. 2009, 42, 832–841.

(21)

Jones-Wilson, T. M.; Deal, K. a; Anderson, C. J.; McCarthy, D. W.; Kovacs, Z.; Motekaitis, R. J.; Sherry, a D.; Martell, a E.; Welch, M. J. The In Vivo Behavior of Copper-64-Labeled Azamacrocyclic Complexes. Nucl. Med. Biol. 1998, 25, 523–530.

(22)

Zhang, Y.; Hong, H.; Engle, J. W.; Bean, J.; Yang, Y.; Leigh, B. R.; Barnhart, T. E.; Cai, W. Positron Emission Tomography Imaging of CD105 Expression with a 64Cu-Labeled Monoclonal Antibody: NOTA is Superior to DOTA. PLoS One 2011, 6, e28005.

ACS Paragon Plus Environment

Page 29 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(23)

ACS Nano

Jensen, A. I.; Binderup, T.; Kumar EK, P.; Kjær, A.; Rasmussen, P. H.; Andresen, T. L. Positron Emission Tomography Based Analysis of Long-Circulating Cross-Linked Triblock Polymeric Micelles in a U87MG Mouse Xenograft Model and Comparison of DOTA and CB-TE2A as Chelators of Copper-64. Biomacromolecules 2014, 15, 1625–1633.

(24)

Bartholomä, M. D. Recent Developments in the Design of Bifunctional Chelators for MetalBased Radiopharmaceuticals Used in Positron Emission Tomography. Inorganica Chim. Acta 2012, 389, 36–51.

(25)

Anderson, C. J.; Ferdani, R. Copper-64 Radiopharmaceuticals for PET Imaging of Cancer: Advances in Preclinical and Clinical Research. Cancer Biother. Radiopharm. 2009, 24, 379– 393.

(26)

Jørgensen, J. T.; Persson, M.; Madsen, J.; Kjær, A. High Tumor Uptake of (64)Cu: Implications for Molecular Imaging of Tumor Characteristics with Copper-Based PET Tracers. Nucl. Med. Biol. 2013, 40, 345–350.

(27)

Boswell, C. A.; Sun, X.; Niu, W.; Weisman, G. R.; Wong, E. H.; Rheingold, A. L.; Anderson, C. J. Comparative In Vivo Stability of Copper-64-Labeled Cross-Bridged and Conventional Tetraazamacrocyclic Complexes. J. Med. Chem. 2004, 47, 1465–1474.

(28)

Sprague, J. E.; Kitaura, H.; Zou, W.; Ye, Y.; Achilefu, S.; Weilbaecher, K. N.; Teitelbaum, S. L.; Anderson, C. J. Noninvasive Imaging of Osteoclasts in Parathyroid Hormone-Induced Osteolysis Using a 64Cu-Labeled RGD Peptide. J. Nucl. Med. 2007, 48, 311–318.

(29)

Seo, J. W.; Mahakian, L. M.; Kheirolomoom, A.; Zhang, H.; Meares, C. F.; Ferdani, R.;

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 30 of 47

Anderson, C. J.; Ferrara, K. W. Liposomal Cu-64 Labeling Method Using Bifunctional Chelators: Poly(ethylene Glycol) Spacer and Chelator Effects. Bioconjug. Chem. 2010, 21, 1206–1215. (30)

Fonge, H.; Huang, H.; Scollard, D.; Reilly, R. M.; Allen, C. Influence of Formulation Variables on the Biodistribution of Multifunctional Block Copolymer Micelles. J. Control. Release 2012, 157, 366–374.

(31)

Goel, S.; Chen, F.; Ehlerding, E. B.; Cai, W. Intrinsically Radiolabeled Nanoparticles: An Emerging Paradigm. Small 2014, 10, 3825–3830.

(32)

Sun, X.; Cai, W.; Chen, X. Positron Emission Tomography Imaging Using Radiolabeled Inorganic Nanomaterials. Acc. Chem. Res. 2015, 48, 286–294.

(33)

Wong, R. M.; Gilbert, D. A.; Liu, K.; Louie, A. Y. Rapid Size-Controlled Synthesis of Oxide Nanoparticles. ACS Nano 2012, 6, 3461–3467.

(34)

Zhao, Y.; Sultan, D.; Detering, L.; Cho, S.; Sun, G.; Pierce, R.; Wooley, K. L.; Liu, Y. Copper64-Alloyed Gold Nanoparticles for Cancer Imaging: Improved Radiolabel Stability and Diagnostic Accuracy. Angew. Chem. Int. Ed. Engl. 2014, 53, 156–159.

(35)

Sun, X.; Huang, X.; Yan, X.; Wang, Y.; Guo, J.; Jacobson, O.; Liu, D.; Szajek, L. P.; Zhu, W.; Niu, G.; Kiesewetter, D. O.; Shouheng, S.; Chen, X. Chelator-Free (64)Cu-Integrated Gold Nanomaterials for Positron Emission Tomography Imaging Guided Photothermal Cancer Therapy. ACS Nano 2014, 8438–8446.

ACS Paragon Plus Environment

Page 31 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(36)

ACS Nano

Zhao, Y.; Sultan, D.; Detering, L.; Luehmann, H. Facile Synthesis, Pharmacokinetic and Systemic Clearance Evaluation, and Positron Emission Tomography Cancer Imaging of 64Cu– Au Alloy Nanoclusters. Nanoscale 2014, 6, 13501–13509.

(37)

Turkevich, J.; Stevenson, P. C.; Hillier, J. A Study of the Nucleation and Growth Processes in the Synthesis of Colloidal Gold. Discuss. Faraday Soc. 1951, 11, 55.

(38)

Frens, G. Controlled Nucleation for the Regulation of the Particle Size in Monodisperse Gold Suspensions. Nat. Phys. Sci., 1973, 241, 20–22.

(39)

Zhao, P.; Li, N.; Astruc, D. State of the Art in Gold Nanoparticle Synthesis. Coord. Chem. Rev. 2013, 257, 638–665.

(40)

Nguyen, D. T.; Kim, D.-J.; So, M. G.; Kim, K.-S. Experimental Measurements of Gold Nanoparticle Nucleation and Growth by Citrate Reduction of HAuCl4. Adv. Powder Technol. 2010, 21, 111–118.

(41)

Bastús, N. G.; Comenge, J.; Puntes, V. Kinetically Controlled Seeded Growth Synthesis of Citrate-Stabilized Gold Nanoparticles of up to 200 Nm: Size Focusing versus Ostwald Ripening. Langmuir 2011, 27, 11098–11105.

(42)

Alvarez-Paneque, A. F.; Rodríguez-González, B.; Pastoriza-Santos, I.; Liz-Marzán, L. M. Shape-Templated Growth of Au@Cu Nanoparticles. J. Phys. Chem. C, 2013, 117, 2474–2479.

(43)

Schulz, F.; Homolka, T.; Bastu, N. G.; Puntes, V.; Weller, H.; Vossmeyer, T. Little Adjustments Significantly Improve the Turkevich Synthesis of Gold Nanoparticles. Langmuir 2014, 30,

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 32 of 47

10779–10784. (44)

Hainfeld, J. F. Functional Associative Coatings for Nanoparticles. US 20080089836 A1, 2008.

(45)

Zhang, G.; Yang, Z.; Lu, W.; Zhang, R.; Huang, Q.; Tian, M.; Li, L.; Liang, D.; Li, C. Influence of Anchoring Ligands and Particle Size on the Colloidal Stability and In Vivo Biodistribution of Polyethylene Glycol-Coated Gold Nanoparticles in Tumor-Xenografted Mice. Biomaterials 2009, 30, 1928–1936.

(46)

Liu, X.; Huang, H.; Jin, Q.; Ji, J. Mixed Charged Zwitterionic Self-Assembled Monolayers as a Facile Way to Stabilize Large Gold Nanoparticles. Langmuir 2011, 27, 5242–5251.

(47)

Liu, X.; Jin, Q.; Ji, Y.; Ji, J. Minimizing Nonspecific Phagocytic Uptake of Biocompatible Gold Nanoparticles with Mixed Charged Zwitterionic Surface Modification. J. Mater. Chem. 2012, 22, 1916.

(48)

Liu, X.; Li, H.; Chen, Y.; Jin, Q.; Ren, K.; Ji, J. Mixed-Charge Nanoparticles for Long Circulation, Low Reticuloendothelial System Clearance, and High Tumor Accumulation. Adv. Healthc. Mater. 2014, 1439–1447.

(49)

Liu, X.; Chen, Y.; Li, H.; Huang, N.; Jin, Q.; Ren, K.; Ji, J. Enhanced Retention and Cellular Uptake of Nanoparticles in Tumors by Controlling Their Aggregation Behavior. ACS Nano 2013, 7, 6244–6257.

(50)

Maeda, H.; Wu, J.; Sawa, T.; Matsumura, Y.; Hori, K. Tumor Vascular Permeability and The EPR Effect in Macromolecular Therapeutics: A Review. J. Control. Release 2000, 65, 271–284.

ACS Paragon Plus Environment

Page 33 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(51)

ACS Nano

Laverman, P.; Brouwers, A. H.; Dams, E. T.; Oyen, W. J.; Storm, G.; van Rooijen, N.; Corstens, F. H.; Boerman, O. C. Preclinical and Clinical Evidence for Disappearance of LongCirculating Characteristics of Polyethylene Glycol Liposomes at Low Lipid Dose. J. Pharmacol. Exp. Ther. 2000, 293, 996–1001.

(52)

Petersen, A. L.; Binderup, T.; Jølck, R. I.; Rasmussen, P.; Henriksen, J. R.; Pfeifer, A. K.; Kjær, A.; Andresen, T. L. Positron Emission Tomography Evaluation of Somatostatin Receptor Targeted 64Cu-TATE-Liposomes in a Human Neuroendocrine Carcinoma Mouse Model. J. Control. Release 2012, 160, 254–263.

(53)

Kim, C.; Hsieh, Y. Lo. Wetting and Absorbency of Nonionic Surfactant Solutions on Cotton Fabrics. Colloids Surfaces A Physicochem. Eng. Asp. 2001, 187-188, 385–397.

(54)

Sadauskas, E.; Danscher, G.; Stoltenberg, M.; Vogel, U.; Larsen, A.; Wallin, H. Protracted Elimination of Gold Nanoparticles from Mouse Liver. Nanomedicine 2009, 5, 162–169.

(55)

Jølck, R. I.; Binderup, T.; Hansen, A. E.; Scherman, J. B.; Munch af Rosenschold, P.; Kjær, A.; Andresen, T. L. Injectable Colloidal Gold in A Sucrose Acetate Isobutyrate Gelating Matrix with Potential Use in Radiation Therapy. Adv. Healthc. Mater. 2014, 3, 1680–1687.

(56) Monopoli, M. P.; Åberg, C.; Salvati, A.; Dawson, K. A. Biomolecular Coronas Provide The Biological Identity of Nanosized Materials. Nature Nanotech. 2012, 7, 779–786. (57)

Mizuhara, T.; Saha, K.; Moyano, D. F.; Kim, C. S.; Yan, B.; Kim, Y.; Rotello V. M. Acylsulfonamide-Functionalized Zwitterionic Gold Nanoparticles for Enhanced Cellular Uptake at Tumor pH. Angew. Chem. Int. Ed. 2015, 54, 6567-6570.

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(58)

Page 34 of 47

Wuelfing, W. P.; Gross, S. M.; Miles, D. T.; Murray, R. W. Nanometer Gold Clusters Protected by Surface-Bound Monolayers of Thiolated Poly(ethylene Glycol) Polymer Electrolyte. J. Am. Chem. Soc. 1998, 120, 12696–12697.

ACS Paragon Plus Environment

Page 35 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

Figure, scheme and chart legends: Scheme 1: Preparation and coating of AuNPs and 64Cu-AuNPs. Depicted is non-radioactive AuNP synthesis (1, 2, 3). The top horizontal lane depicts direct embedding (5, 6), the middle lane depicts seeded embedding (7, 8) and the bottom lane depicts passive surface labeling (4). a) Sodium citrate, 65 °C to reflux. b) Sodium citrate, dropwise addition of HAuCl4 at 65 °C, then reflux. c) 1-Dodecanethiol and Tween 20. Radiolabeling of 4, 5 and 7 was achieved by adding [64Cu]CuCl2 directly to the reaction mixture. Figure 1: Characterization of AuNPs and

64

Cu-AuNPs. Passive surface labeling shown in red,

direct embedding shown in blue, seeded embedding shown in green. (A) Average radiolabeling yields of the three strategies given as the amount of 64Cu remaining associated with the AuNP fraction after incubation with EDTA for 30 min. Passive surface labeling (4) (n = 3), direct embedding (6) (n = 6) and seeded embedding (8) (n = 5). (B) Mean sizes of AuNPs as determined by DLS. Uncoated

64

Cu-

AuNPs (5, 7) did not show significant change in size as compared to non-radiolabeled AuNPs prepared by an identical method (1, 2), (n = 5). Coated 64Cu-AuNPs (6, 8) showed an increase in size. (C) PDIs of AuNPs as determined by DLS. Embedding of 64Cu did not show a significant increase in PDI, (n = 5). (D & E) Absorption spectra of AuNPs. The characteristic absorption peak for AuNPs is observed at 521 nm for 1, 523 nm for 5 and 532 nm for 6. This redshift corresponds to an increase in the size of the AuNPs, correlating with DLS data. A similar pattern was observed for 2 (527 nm), 7 (526 nm) and 8 (531 nm). (F & G): Representative TEM images of first generation AuNPs (1), with sizes of 14 ± 1.3 nm (G) and second generation AuNPs (2) with sizes of 29 ± 2.4 nm (F). Size distributions are based on counts of n = 335 (1) and n = 443 (2) AuNPs. See Supporting Information (Figure S1) for size distribution histograms.

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 36 of 47

Figure 2: Stability of the radiolabels over two days against EDTA (10 mM, pH 7.5). The associated with

64

Cu

64

Cu-AuNPs prepared by direct embedding (6) and seeded embedding (8, 9 and 10)

showed excellent stability against EDTA with 90% ± 3% (6), 95% ± 0% (8), 94% ± 4% (9) and 98% ± 2% (10) remaining associated with the

64

Cu-AuNPs after two days.

64

surface labeling appeared less stable and showed substantial leak of

Cu-AuNPs prepared by passive 64

Cu. Numbers are reported as

mean ± SD (n = 3). Chart 1: Molecular structures of gold coatings investigated in vivo by PET/CT. 8: 1-Dodecanethiol / Tween 20, 9: MeO-PEG5000SH, 10: Sulphonate / quaternary ammonium. Figure 3: Serum stability study of

64

Cu-AuNPs (8) incubated with PBS/mouse serum (1:1) for two

days at 37 °C, and then separated by size-exclusion chromatography. The left y-axis shows percent of total applied radioactivity in fractions of the

64

Cu-AuNPs and 50% serum mixture (purple) and the

64

Cu-AuNPs alone (red), while the right y-axis shows the absorbance at 280 nm for serum alone. The

eluted radioactivity for the mixture and the 64Cu-AuNPs alone were virtually identical, indicating that no transfer of 64Cu to serum proteins had taken place. Figure 4. TEM characterization of coated

64

Cu-AuNPs used in vivo (8, 9, 10). (A-C) Representative

low magnification TEM micrographs with high resolution inset, A: 8, B: 9, C: 10. Average diameters of the 64Cu-AuNP cores were found to be 26.6 ± 3.3 nm (n = 327) (8), 28.3 ± 3.5 nm (n = 440) (9) and 26.9 ± 2.5 nm (n = 267) (10). (D-F) Representative negative stained TEM images of coated

64

Cu-

AuNPs showing a corona of organic material surrounding the Au Core, D: 8, E: 9, F: 10. No Corona was seen for 8 whereas coronas were observed for both 9 and 10 measuring 5.9 ± 2.1 (n = 50) and 1.3 ±

ACS Paragon Plus Environment

Page 37 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Nano

0.3 nm (n = 50) in thickness, respectively. Scale bars equal 100 nm for A-C (10 nm for inserts) and 50 nm for D-F. Figure 5:

64

Cu-AuNP biodistribution by PET/CT.

64

Cu-AuNPs (8, 9, 10) were injected

intravenously in FaDu tumor-bearing mice and region-of-interest (ROI) analysis was constructed on PET/CT images acquired 1h, 4h and 24h after injection. (A) Mean tissue accumulation given as %ID/g ± SD. n = 5 in all cases, except tumors where n = 10. For 10 at 4h and 24h n = 4, and tumors, n = 8. (B) Representative PET/CT images of 8 (top), 9 (middle) and 10 (bottom). Organs are indicated as: (H) heart and the large intracardiac blood volume, (L) liver, (T) tumors, were they can be appreciated. Yellow arrows illustrate the observed accumulation in lymph nodes for 8 at the 24h scan (for magnification of lymph node accumulation, see Figure S4, Supporting Information). Color bar indicates activity level of 64Cu; for the 1 h and 4 h scans the scale is 0 – 6.5 %ID/g and 0 – 16 %ID/g for the 24 h scans. (C) Left column: Representative axial PET/CT images at 24 h after intravenous injection of 8, 9 and 10. Tumors are indicated by white arrows (scale bar 0 – 16 %ID/g). Right column: Autoradiography of 8, 9 and 10 in 8 µm tumor cryosections. The autoradiography illustrates the heterogeneous intratumoral distribution of the NPs. Scale bars represent semi-quantiative accumulation level, white line 2 mm long. (D) Max tumor accumulation given as %ID/g ± SD (left three bars for each coating) and percent injected dose per tumor given as %ID/tumor ± SD (right three bars). n = 10 in all cases, except 10 (4h, 24h) where n = 8. The max tumor accumulation data mirror the heterogeneous distribution of the

64

Cu-AuNPs in the tumors when compared to mean tumor

accumulation. Figure 6: Ex vivo analysis of

64

Cu-AuNP biodistribution. Tissue accumulation (%ID/g) based on

radioactivity was determined by well-counting of excised organs. In addition, tumor, liver, spleen and

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

muscle were analyzed for Au content by ICP-OES. We saw good correlation between

Page 38 of 47

64

Cu and Au

accumulation in each organ, underlining the in vivo stability of the radiolabel. The Au and corresponding

64

Cu accumulation data were subjected to a two-tailed, unpaired t-test, with a

hypothesized mean difference of 0. Significant difference is shown as (-) p > 0.25, (*) p < 0.25, (**) p < 0.05, (***) p < 0.01. Number given as mean ± SD, n = 5, except tumors where n = 10.

ACS Paragon Plus Environment

Page 39 of 47

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Paragon Plus Environment

Page 40 of 47

Page 41 of 47

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Paragon Plus Environment

Page 42 of 47

Page 43 of 47

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Paragon Plus Environment

Page 44 of 47

Page 45 of 47

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Paragon Plus Environment

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Paragon Plus Environment

Page 46 of 47

Page 47 of 47

ACS Nano

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Paragon Plus Environment