Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect
ACS Publications. Most Trusted. Most Cited. Most Read
In Vivo Tumor Vasculature Targeted PET/NIRF Imaging with TRC105(Fab)-Conjugated, Dual-Labeled Mesoporous Silica Nanoparticles
My Activity
CONTENT TYPES

Figure 1Loading Img
  • Open Access
Article

In Vivo Tumor Vasculature Targeted PET/NIRF Imaging with TRC105(Fab)-Conjugated, Dual-Labeled Mesoporous Silica Nanoparticles
Click to copy article linkArticle link copied!

View Author Information
Department of Radiology, University of Wisconsin—Madison, Madison, Wisconsin 53792, United States
Materials Science Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
§ Department of Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
TRACON Pharmaceuticals, Inc., San Diego, California 92122, United States
University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53792, United States
*E-mail: [email protected]. Address: Departments of Radiology and Medical Physics, University of Wisconsin—Madison, Room 7137, 1111 Highland Avenue, Madison, Wisconsin 53705−2275, United States.
Open PDFSupporting Information (1)

Molecular Pharmaceutics

Cite this: Mol. Pharmaceutics 2014, 11, 11, 4007–4014
Click to copy citationCitation copied!
https://doi.org/10.1021/mp500306k
Published June 17, 2014

Copyright © 2014 American Chemical Society. This publication is licensed under these Terms of Use.

Abstract

Click to copy section linkSection link copied!

Multifunctional mesoporous silica nanoparticles (MSN) with well-integrated multimodality imaging properties have generated increasing research interest in the past decade. However, limited progress has been made in developing MSN-based multimodality imaging agents to image tumors. We describe the successful conjugation of, copper-64 (64Cu, t1/2 = 12.7 h), 800CW (a near-infrared fluorescence [NIRF] dye), and TRC105 (a human/murine chimeric IgG1 monoclonal antibody) to the surface of MSN via well-developed surface engineering procedures, resulting in a dual-labeled MSN for in vivo targeted positron emission tomography (PET) imaging/NIRF imaging of the tumor vasculature. Pharmacokinetics and tumor targeting efficacy/specificity in 4T1 murine breast tumor-bearing mice were thoroughly investigated through various in vitro, in vivo, and ex vivo experiments. Dual-labeled MSN is an attractive candidate for future cancer theranostics.

Copyright © 2014 American Chemical Society

SPECIAL ISSUE

This article is part of the Positron Emission Tomography: State of the Art special issue.

Introduction

Click to copy section linkSection link copied!

Vasculature targeting of tumor angiogenesis can be done based on biomarkers on the surface of tumor endothelial cells and does not require nanoparticle extravasation. (1) Vasculature targeted ligands, including vascular endothelial growth factor (VEGF) and arginine–glycine–aspartic acid (RGD) peptides, have been conjugated to the surface of different functional nanoparticles, including but not limited to, quantum dots (QDs), (2) single-walled carbon nanotubes (SWNTs), (3) and nanographene oxide (GO), (4, 5) for enhanced tumor targeted imaging and drug delivery.
TRC105 (a human/murine chimeric IgG1 monoclonal antibody) is a promising vasculature targeting moiety, (6) which binds to both human and murine CD105 (endoglin), a vascular-specific marker for tumor angiogenesis. (7, 8) Using TRC105 antibody and its fragments (i.e., TRC105(Fab) or F(ab′)2), we reported the first positron emission tomography (PET) imaging of CD105 expression in cancer, (9, 10) demonstrating the potential of CD105 for cancer-targeted imaging and therapy.
Fluorescence imaging, especially near-infrared fluorescence (NIRF) imaging, is economical and highly sensitive in certain scenarios. However, it is not quantitative in nature. (11) PET imaging, in contrast, has superb tissue penetration of signal, high sensitivity, and quantitative evaluation of the in vivo biodistribution. (12) The combination of PET with NIRF may provide more complementary information than either single modality alone and has attracted increasing interests recently. (13-20) For example, to mitigate the qualitative nature of QDs optical imaging, copper-64 (64Cu, t1/2 = 12.7 h) labeled QDs have been developed for in vivo PET/NIRF targeted dual-modality imaging using RGD peptides as the targeting ligands. (13)
Mesoporous silica nanoparticle (MSN) possesses many attractive properties and have been intensively investigated as a novel and biocompatible drug delivery system. (21-24) Although MSN itself emits no light, many strategies have been developed for engineering fluorescent MSN that can be tracked by optical imaging. For example, fluorescent dyes with varied excitation/emission combinations have been chemically linked to the surface or loaded into the matrix of MSN for studying the dynamic distribution of MSN in vivo. (25, 26) Moreover, optical nanocrystals, including QDs (27) and upconversion nanoparticles, (28) have also been encapsulated into MSN for imaging deeper tissues. Efforts have focused on the design of various fluorescent MSNs for in vivo cancer imaging, but little progress has been made with respect to in vivo tumor targeted imaging of the functionalized MSN.
Herein, using TRC105 (Fab) vascular targeting moiety as the targeting ligand, we report the first example of 64Cu (a PET tracer) and 800CW (a NIRF dye) labeled MSN for PET/NIRF dual-modality imaging of the tumor vasculature. Pharmacokinetics and tumor targeting efficacy/specificity in 4T1 murine breast tumor-bearing mice were thoroughly investigated through various in vitro, in vivo, and ex vivo experiments. Our dual-labeled MSN could become a promising candidate for future image-guided drug delivery and targeted cancer therapy.

Experimental Section

Click to copy section linkSection link copied!

Materials

TRC105 was provided by TRACON Pharmaceuticals Inc. (San Diego, CA). PD-10 columns were purchased from GE Healthcare (Piscataway, NJ). IR Dye 800CW-NHS (NHS denotes N-hydroxysuccinimide) ester was acquired from LI-COR Biosciences Co. (Lincoln, NE). SCM-PEG5k-Mal was obtained from Creative PEGworks. NOTA-SCN (i.e., 2-S-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid) was acquired from Macrocyclics, Inc. (Dallas, TX). NHS-fluorescein, Chelex 100 resin (50–100 mesh), tetraethyl orthosilicate (TEOS), ammonia (NH3·H2O), triethylamine (TEA), 3-aminopropyl)triethoxysilane (APS), dimethyl sulfoxide, cetyltrimethylammonium chloride (CTAC, 25 wt %), and Kaiser test kit were purchased from Sigma-Aldrich (St. Louis, MO). Traut’s reagent (2-Iminothiolane·HCl) and Ellman’s reagent (5,5′-dithiobis(2-nitrobenzoic acid) or DTNB) were purchased from Fisher Scientific. Water and all buffers were of Millipore grade and pretreated with Chelex 100 resin to ensure that the aqueous solution was free of heavy metals. All chemicals were used as received without further purification.

Generation of TRC105(Fab)

Procedures for the synthesis of TRC105(Fab) were the same as we reported previously. (29) Typically, TRC105 (2 mg/mL) was digested out in a reaction buffer (20 mM sodium phosphate monobasic, 10 mM disodium ethylenediaminetetraacetic acid [EDTA], and 80 mM cysteine hydrochloride) for 4 h at 37 °C, with immobilized papain/TRC105 at a weight ratio of 1:40. Afterward, the reaction mixture was centrifuged at 5000g for 1 min to remove the immobilized papain. The supernatant was purified by size exclusion column chromatography on a Sephadex G-75 column to yield TRC105(Fab), using phosphate-buffered saline (PBS) as the mobile phase.
For the thiolation of TRC105(Fab), in 700 μL (1.5 mg/mL) of TRC105(Fab) was added 50 μL (2 mg/mL) of Traut’s reagent and adjusted the pH to 8.0 using 0.1 M Na2CO3. The mixture was kept shaking for 2 h at room temperature. As-synthesized TRC105(Fab)-SH was purified using PD-10 columns with PBS as the mobile phase. Fraction from 3.0 to 4.0 mL was collected and concentrated using a 10 k filter at 5000 rpm for 15 min. The final concentration of TRC105(Fab)-SH was found to be about 1.7 mg/mL (∼300 μL).

Synthesis of MSN

Procedures for the synthesis of ∼80 nm sized MSN were the same as we reported previously. (30) In a typical synthesis, CTAC (2 g) and TEA (20 mg) were dissolved in 20 mL of high Q water and stirred at room temperature for 1 h. Afterward, 1.0 mL of TEOS was added rapidly and the resulting mixture was stirred for 1 h at 95 °C in a water bath. The mixture was then cooled down, collected by centrifugation, and washed with water and ethanol to remove residual reactants. Subsequently, the product was extracted for 24 h with a 1 wt% solution of NaCl in methanol at room temperature to remove the template CTAC. This process was carried out for at least three times to ensure complete removal of CTAC.
For amino group modification, as-synthesized MSN was first dispersed in 20 mL of absolute ethanol, followed by addition of 1 mL of APS. The system was sealed and kept at 86–90 °C in a water bath for 48 h. Afterward, the mixture was centrifuged and washed with ethanol for several times to remove the residual APS. The MSN-NH2 could be well-dispersed in water, and the concentration of −NH2 groups (nmol/mL) was measured using a Kaiser test kit.

Synthesis of NOTA-MSN-800CW-PEG-TRC105(Fab)

To conjugated MSN with 800CW, 2 nmol of 800CW-NHS ester was mixed with MSN-NH2 (with ∼100 nmol of −NH2 groups) and reacted for 2 h at room temperature (pH 8.5–9.0) to form MSN-800CW-NH2. Then, NOTA-SCN (∼53 nmol) in dimethyl sulfoxide was allowed to react with MSN-800CW-NH2 at pH 8.5 to obtain NOTA-MSN-800CW-NH2. Afterward, 2 mg (400 nmol) of SCM-PEG5k-Mal was added and reacted for another 2 h, resulting in NOTA-MSN-800CW-PEG-Mal. NOTA-MSN-800CW-PEG-TRC105(Fab) could be obtained by reacting TRC105(Fab)-SH with NOTA-MSN-800CW-PEG-Mal at room temperature for overnight. The final sample was kept at 4 °C before 64Cu labeling. Note, both “NOTA” and “PEG” were omitted from the acronyms of the final nanoconjugates for clarity considerations in the following sections.

Flow Cytometry

Cells were first harvested and suspended in cold PBS with 2% bovine serum albumin at a concentration of 5 × 106 cells/mL and then incubated with fluorescein conjugated MSN-800CW-TRC105(Fab) (targeted group) or fluorescein conjugated MSN-800CW (non-targeted group) for 30 min at room temperature. The cells were washed three times with cold PBS and centrifuged for 5 min. Afterward, the cells were washed and analyzed using a BD FACSCalibur four-color analysis cytometer, which is equipped with 488 and 633 nm lasers (Becton-Dickinson, San Jose, CA) and FlowJo analysis software (Tree Star, Ashland, OR). A “blocking” experiment was also performed in cells incubated with the same amount of fluorescein conjugated MSN-800CW-TRC105(Fab), where 500 μg/mL of unconjugated TRC105 was added to evaluate the CD105 specificity of fluorescein conjugated MSN-800CW-TRC105(Fab).

Radiolabeling with 64Cu

64CuCl2 (74–148 MBq) was diluted in 300 μL of 0.1 M sodium acetate buffer (pH 6.5) and added to MSN-800CW-TRC105(Fab) or MSN-800CW. Note, all MSN nanoconjugates were already conjugated with NOTA. The reaction was allowed to proceed at 37 °C for 30 min with constant stirring. 64Cu-MSN-800CW-TRC105(Fab) and 64Cu-800CW-MSN were purified using PD-10 columns with PBS as the mobile phase. The radioactivity fractions (typically between 3.5 and 4.5 mL) were collected for further in vivo imaging experiments. After 6 mL of PBS, the unreacted 64Cu started to elute from the column.

4T1 Murine Breast Cancer Model

All animal studies were conducted under a protocol approved by the University of Wisconsin Institutional Animal Care and Use Committee. To generate the 4T1 tumor model, 4–5 week old female BALB/c mice were purchased from Harlan (Indianapolis, IN, USA), and tumors were established by subcutaneously injecting 2 × 106 cells, suspended in 100 μL of 1:1 mixture of RPMI 1640 and Matrigel (BD Biosciences, Franklin Lakes, NJ, USA), into the front flank of mice. The tumor sizes were monitored every other day, and the animals were subjected to in vivo experiments when the tumor diameter reached 5–8 mm.

PET/NIRF Imaging and Biodistribution Studies

PET scans at various time points postinjection (pi) using a microPET/microCT Inveon rodent model scanner (Siemens Medical Solutions USA, Inc.), image reconstruction, and region-of-interest (ROI) analysis of the PET data were performed similarly to that described previously. (31) Quantitative PET data were presented as percentage injected dose per gram of tissue (%ID/g). Tumor-bearing mice were each injected with 5–10 MBq of 64Cu-MSN-800CW-TRC105(Fab) or 64Cu-MSN-800CW via tail vein before serial PET scans. Another group of three 4T1 tumor-bearing mice were each injected with 1 mg of unlabeled TRC105 at 1 h before 64Cu-MSN-800CW-TRC105(Fab) administration to evaluate the CD105 specificity of 64Cu-MSN-800CW-TRC105(Fab) in vivo (i.e., blocking experiment).
After the last PET scans at 48 h pi, biodistribution studies were carried out to confirm that the %ID/g values based on PET imaging truly represented the radioactivity distribution in tumor-bearing mice. Mice were euthanized, and blood, 4T1 tumor, and major organs/tissues were collected and wet-weighed. The radioactivity in the tissue was measured using a gamma-counter and presented as %ID/g (mean ± SD).
For in vivo NIRF imaging, each 4T1 tumor-bearing mouse was injected with MSN-800CW-TRC105(Fab) (targeted group) with the amount of 800CW estimated to be ∼400 pmol. The mouse was then imaged using an IVIS spectrum in vivo imaging system (Ex = 745 nm, Em = 800 nm) at 4 h pi. For non-targeted and blocking groups, mice were injected with MSN-800CW and MSN-800CW-TRC105(Fab) (together with 1 mg blocking dose of free TRC105), respectively, with equal amount of 800CW dyes.

Results and Discussion

Click to copy section linkSection link copied!

Synthesis and Characterizations of Thiolated TRC105(Fab)

Thiolated TRC105(Fab), i.e., TRC105(Fab)-SH, was generated following our previous reported procedures with an addition thiolation step (Figure 1a). (29) After papain digestion of full TRC105 antibody, a Sephadex G 75 column was used to separate TRC105(Fab) from other components in the reaction mixture. Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) was then used to confirm the successful generation of TRC105(Fab), where the disappearance of the TRC105 band (∼148 kDa, lane 2 in Figure 1b) and the appearance of pure TRC105(Fab) band (∼47.5 kDa, lane 3 in Figure 1b) were shown. Traut’s reagent was later used for the thiolation of TRC105(Fab), forming TRC105(Fab)-SH. The presence of −SH groups was further confirmed by using Ellman’s reagent. Taken together, these results indicate the complete digestion of TRC105 after papain treatment to yield high purity TRC105(Fab) and the successful thiolation of TRC105(Fab) to form TRC105(Fab)-SH for further bioconjugations.

Figure 1

Figure 1. Synthesis and characterization of TRC105(Fab). (a) Schematic illustration showing the generation of TRC105(Fab) and its thiolation. (b) SDS-PAGE of molecular weight markers (lane 1), intact TRC105 antibody (lane 2), and TRC105(Fab) after purification on the Sephadex G-75 column (lane 3).

Synthesis and Characterizations of 64Cu-MSN-800CW-TRC105(Fab)

Multiple steps of surface engineering were required for the synthesis 64Cu-MSN-800CW-TRC105(Fab), as shown in Scheme 1. As-synthesized uniform MSN (1) was first functionalized with amino groups (−NH2) with (3-aminopropyl)triethoxysilane (APS), to form MSN-NH2 (2), leaving amino groups on the surface for further conjugation. Successful −NH2 modification was confirmed by ninhydrin testing (Supportiong Information Figure S1). The desired amount of NIRF dyes (800CW-NHS ester) and NOTA-SCN (a 64Cu chelator) and were then reacted with MSN-NH2 at pH 8.5–9 to produce NOTA-MSN-800CW-NH2 (3). Afterward, the heterobifunctional succinimidyl carboxy methyl ester-poly ethylene glycol(5 kDa)-maleimide (SCM-PEG5k-Mal) was used to generate NOTA-MSN-800CW-PEG-Mal (4), which was then reacted with TRC105(Fab)-SH to yield NOTA-MSN-800CW-PEG-TRC105(Fab) (5). Lastly, the NOTA-MSN-800CW-PEG-TRC105(Fab) was labeled with 64Cu to form the 64Cu-NOTA-MSN-800CW-PEG-TRC105(Fab) nanoconjugate (6), abbreviated as 64Cu-MSN-800CW-TRC105(Fab). Because all the MSN nanoconjugates will contain the same NOTA and PEG chains (5 kDa), both “NOTA” and “PEG” were omitted from the acronyms of the final nanoconjugates for clarity.

Scheme 1

Scheme 1. Schematic Illustration of the Synthesis of 64Cu-MSN-800CW-TRC105(Fab)a

Scheme aUniform MSN nanoparticle (1) was first modified with −NH2 groups with APS to form MSN-NH2 (2). As-synthesized MSN-NH2 was then subjected to 800CW and NOTA conjugation to yield NOTA-MSN-800CW-NH2 (3). Afterwards, PEGylation step was introduced to render the stability of NOTA-MSN-800CW-PEG-Mal (4) in PBS, at the same time adding maleimide groups. NOTA-MSN-800CW-PEG-TRC105(Fab) (5) could be obtained by reacting TRC105(Fab)-SH with 4 at room temperature. 64Cu-labeling was performed in the last step to generate 64Cu-NOTA-MSN-800CW-PEG-TRC105(Fab) (6), short for 64Cu-MSN-800CW-TRC105(Fab).

Figure 2a shows the representative transmission electron microscopy (TEM) image of ∼80 nm sized uniform MSN, which was synthesized following as described. (30) No obvious changes in the morphology of MSN were observed after multistep surface modifications, as evidenced by TEM image of MSN-800CW-TRC105(Fab) (Figure 2b). Successful conjugation of 800CW to MSN was confirmed by the NIRF imaging of MSN-800CW-TRC105(Fab) (inset in Figure 2b) using the IVIS spectrum in vivo imaging system (Ex = 745 nm, Em = 800 nm). Dynamic light scattering (DLS) and ζ potential measurements indicated the final Z-average size and surface charge of MSN-800CW-TRC105(Fab) to be 175.3 ± 9.7 nm and −3.25 ± 0.8 mV (pH 7.4, PBS), respectively.

Figure 2

Figure 2. Synthesis and characterization of MSN and MSN-800CW-TRC105(Fab). TEM images of (a) pure MSN, (b) MSN-800CW-TRC105(Fab). Insets in (a) and (b) show the schemes of MSN and MSN-800CW-TRC105(Fab). An optical image of MSN-800CW-TRC105(Fab) acquired from IVIS spectrum in vivo imaging system (Ex = 745 nm, Em = 800 nm) is also shown in the upper right portion of (b).

In Vitro CD105 Targeting of MSN-800CW-TRC105(Fab)

Previously, we have demonstrated that fluorescein conjugated TRC105(Fab) binds with high avidity and specificity to human umbilical vein endothelial cells (HUVECs), (29) indicating that papain digestion does not affect antigen recognition. To assess CD105 targeting efficiency of MSN nanoconjugates, a flow cytometry study using HUVECs was also performed after NHS-fluorescein was conjugated to the surface of nanoparticles. Flow cytometry results (Figure 3) indicated that incubation with fluorescein conjugated MSN-800CW-TRC105(Fab) significantly enhanced the mean fluorescence intensity of HUVECs, compared to fluorescein conjugated MSN-800CW. The specificity of in vitro CD105 targeting with MSN-800CW-TRC105(Fab) was confirmed in the blocking study, where only background level uptake of MSN-800CW-TRC105(Fab) was observed after blocking HUVECs with a large dose of free TRC105 (500 μg/mL).

Figure 3

Figure 3. Flow cytometry analysis of MSN nanoconjugates in HUVECs (CD105 positive) after 30 min incubation and subsequent washing. Targeted group: fluorescein conjugated MSN-800CW-TRC105(Fab). Non-targeted group: fluorescein conjugated MSN-800CW. Blocking group: fluorescein conjugated MSN-800CW-TRC105(Fab) with a blocking dose of TRC105 (500 μg/mL).

In Vivo PET/NIRF Dual Modal CD105-Targeted Imaging of 64Cu-MSN-800CW-TRC105(Fab)

For in vivo tumor targeted PET imaging, both MSN-800CW-TRC105(Fab) and MSN-800CW were labeled with 64Cu for in vivo imaging and biodistribution studies. As-synthesized 64Cu-MSN-800CW-TRC105(Fab) (targeted group) and 64Cu-MSN-800CW (non-targeted group) were purified using PD-10 columns with PBS as the mobile phase. The radioactivity fractions (typically elute between 3.0 and 4.0 mL) were collected for in vivo experiments. Unreacted 64Cu eluted from the column by the 6 mL fractions.
In vivo tumor targeted imaging was carried out in 4T1 murine breast tumor-bearing mice, which express a high level of CD105 on the tumor neovasculature. (7, 8) Each mouse was intravenously (iv) injected with 5–10 MBq of 64Cu-MSN-800CW-TRC105(Fab) or MSN-800CW-TRC105(Fab) (had ∼400 pmol of dyes) for PET/NIRF imaging to show in vivo biodistribution pattern as well as tumor accumulation (Figures 4, 5 and 6). Non-targeted and blocking groups were also included to assess the targeting specificity of dual-labeled MSN nanoconjugates. Quantitative data obtained from ROI analysis of these PET images are also shown in Supporting Information Table S1–S3.

Figure 4

Figure 4. Serial coronal PET images of 4T1 tumor-bearing mice at different time points postinjection of (a) 64Cu-MSN-800CW-TRC105(Fab), (b) 64Cu-MSN-800CW, and (c) 64Cu-MSN-800CW-TRC105(Fab) with a blocking dose of TRC105 (1 mg/mouse). Tumors are indicated by yellow arrowheads.

Figure 5

Figure 5. In vivo NIRF imaging of 4T1 tumor-bearing mice at 4 h postinjection. (a) Targeted group, 64Cu-MSN-800CW-TRC105(Fab); (b) non-targeted group, 64Cu-MSN-800CW; (c) blocking group, 64Cu-MSN-800CW-TRC105(Fab) with a blocking dose of TRC105 (1 mg/mouse). Tumors are indicated by yellow arrowheads. Each mouse was iv injected with MSN nanoconjugates with equal amounts of 800CW dyes (∼400 pmol). All images were acquired by using IVIS spectrum in vivo imaging system (Ex = 745 nm, Em = 800 nm).

Figure 6

Figure 6. Quantitative analysis of the PET data. (a) Time–activity curve of the liver, 4T1 tumor, blood, and muscle upon iv injection of 64Cu-MSN-800CW-TRC105(Fab) (targeted group, n = 3). (b) Time–activity curve of tumor-to-muscle (T/M), tumor-to-blood (T/B), and tumor-to-liver (T/L) ratios from the same targeted group (n = 3). (c) Comparison of 4T1 tumor uptake among the three groups. The differences between 4T1 tumor uptake of 64Cu-MSN-800CW-TRC105(Fab) and the two control groups were statistically significant (**P < 0.01) in all cases. (d) Biodistribution of three groups in 4T1 tumor-bearing mice at the end of the PET scans at 48 h pi (n = 3).

The accumulation of 64Cu-MSN-800CW-TRC105(Fab) in the 4T1 tumor was clearly demonstrated in PET imaging and was found to be 5.4 ± 0.2%ID/g at 4 h pi, as shown in Figures 4a and 6a and Supporting Information Table S1 (n = 3). In contrast, the 4T1 tumor uptake of 64Cu-MSN-800CW was found to be ∼2%ID/g at all of the time points examined (n = 3; Figure 4b, Supporting Information Figure S2a and Table S2), indicating that TRC105(Fab) conjugation was required for enhanced tumor accumulation of 64Cu-MSN-800CW-TRC105(Fab) in vivo. To further confirm the CD105 targeting specificity of 64Cu-MSN-800CW-TRC105(Fab), blocking studies were performed. The administration of a blocking dose (1 mg/mouse) of free TRC105 at 1 h before 64Cu-MSN-800CW-TRC105(Fab) injection significantly reduced tumor uptake to 2.3 ± 0.2%ID/g at 4 h pi (n = 3, Figure 4c, Supporting Information Figure S2b and Table S3), demonstrating the CD105 specific targeting of 64Cu-MSN-800CW-TRC105(Fab) in vivo. Figure 6c further summarizes the comparison of 4T1 tumor uptake of three groups at different time points. 64Cu-MSN-800CW-TRC105(Fab) showed the highest tumor uptake throughout the study period.
In vivo NIRF imaging was used to further confirm the enhanced accumulation of MSN-800CW-TRC105(Fab). Different MSN nanoconjugates which contained equal amounts of dyes (∼400 pmol) were injected intravenously into 4T1 tumor-bearing mice. In vivo NIRF images that were generated at 4 h pi (when the nanoparticles showed the highest tumor accumulation based on the PET imaging, shown in Figure 4a) were acquired and are shown in Figure 5. A significantly stronger signal from the targeted group was observed compared to the non-targeted and blocking groups, indicating the successful detection by NIRF imaging of MSN-800CW-TRC105(Fab) nanoconjugates. Notably, the liver and spleen were the major organs for the accumulation of MSN-800CW-TRC105(Fab). However, due to the difference in scattering behaviors and depth, only 800CW emission signalsfrom xenograft 4T1 tumor on the surface of mouse skin were detected by the IVIS spectrum in vivo imaging system. Taken together, these data demonstrated the CD105 specificity of dual-modality PET/NIRF targeted imaging using 64Cu-MSN-800CW-TRC105(Fab) in vivo.
The conjugate 64Cu-MSN-800CW-TRC105(Fab) also improved tumor-to-muscle (T/M) biodistribution ratios. As shown in Figure 6b, T/M value of the targeted group was estimated to be 5.2 ± 0.2 at 0.5 h pi and increased up to 7.3 ± 1.1 at 4 h pi, which was significantly higher than that of non-targeted and blocking groups (Supporting Information Figure S2c,d and Table S4–S6). Similar to other TRC105-conjugated nanoparticles, (4, 5, 30) besides tumor accumulation, most of the 64Cu-MSN-800CW-TRC105(Fab) nanoconjugates were cleared by the reticuloendothelial system (RES) with liver uptake found to be 23.2 ± 3.5%ID/g at 0.5 h pi, decreasing gradually to 9.2 ± 0.6%ID/g by 48 h pi (n = 3; Figures 4a and 6a, and Supporting Information Table S1), which was validated by the ex vivo biodistribution study (Figure 6d). Similar trends were also observed in the non-targeted and blocking groups (Supporting Information Figure S2a,b and Table S2–S3).

Conclusion

Click to copy section linkSection link copied!

In conclusion, we report the in vivo tumor vasculature targeted PET/NIRF dual-modality imaging using well-functionalized 64Cu-MSN-800CW-TRC105(Fab) nanoconjugates. Pharmacokinetics and tumor targeting efficacy/specificity in 4T1 murine breast tumor-bearing mice were thoroughly investigated through various in vitro, in vivo, and ex vivo experiments. Vascular targeting led to ∼2-fold enhancement of tumor accumulation over passive targeting alone. Other targeting ligands could also be conjugated to MSN for further improvement of tumor accumulation in vivo. Given the presence of tunable pore size and larger surface area that could accommodate therapeutic agents, as-designed 64Cu-MSN-800CW-TRC105(Fab) could be employed as both imaging and therapeutic agents.

Supporting Information

Click to copy section linkSection link copied!

Ninhydrin testing, tables of PET region-of-interest quantifications, quantitative analysis of PET imaging data from non-targeted and blocking groups. This material is available free of charge via the Internet at http://pubs.acs.org.

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

Click to copy section linkSection link copied!

  • Corresponding Author
    • Weibo Cai - Department of Radiology, University of Wisconsin—Madison, Madison, Wisconsin 53792, United StatesMaterials Science Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United StatesDepartment of Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53705, United StatesUniversity of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53792, United States Email: [email protected]
  • Authors
    • Feng Chen - Department of Radiology, University of Wisconsin—Madison, Madison, Wisconsin 53792, United States
    • Tapas R. Nayak - Department of Radiology, University of Wisconsin—Madison, Madison, Wisconsin 53792, United States
    • Shreya Goel - Materials Science Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
    • Hector F. Valdovinos - Department of Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
    • Hao Hong - Department of Radiology, University of Wisconsin—Madison, Madison, Wisconsin 53792, United StatesMaterials Science Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United StatesDepartment of Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53705, United StatesTRACON Pharmaceuticals, Inc., San Diego, California 92122, United StatesUniversity of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53792, United States
    • Charles P. Theuer - Department of Radiology, University of Wisconsin—Madison, Madison, Wisconsin 53792, United StatesTRACON Pharmaceuticals, Inc., San Diego, California 92122, United States
    • Todd E. Barnhart - Department of Medical Physics, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
  • Notes
    The authors declare the following competing financial interest(s): Charles P. Theuer is an employee of TRACON. The other authors declare no competing financial interest..

Acknowledgment

Click to copy section linkSection link copied!

This work is supported, in part, by the University of Wisconsin—Madison, the National Institutes of Health (NIBIB/NCI 1R01CA169365, P30CA014520), the Department of Defense (W81XWH-11-1-0644), and the American Cancer Society (125246-RSG-13-099-01-CCE).

References

Click to copy section linkSection link copied!

This article references 31 other publications.

  1. 1
    Chen, F.; Cai, W. Tumor Vasculature Targeting: A Generally Applicable Approach for Functionalized Nanomaterials Small 2014, 10, 1887 1893
  2. 2
    Cai, W.; Shin, D. W.; Chen, K.; Gheysens, O.; Cao, Q.; Wang, S. X.; Gambhir, S. S.; Chen, X. Peptide-Labeled Near-Infrared Quantum Dots for Imaging Tumor Vasculature in Living Subjects Nano Lett. 2006, 6, 669 676
  3. 3
    Liu, Z.; Cai, W.; He, L.; Nakayama, N.; Chen, K.; Sun, X.; Chen, X.; Dai, H. In Vivo Biodistribution and Highly Efficient Tumour Targeting of Carbon Nanotubes in Mice Nature Nanotechnol. 2007, 2, 47 52
  4. 4
    Hong, H.; Yang, K.; Zhang, Y.; Engle, J. W.; Feng, L.; Yang, Y.; Nayak, T. R.; Goel, S.; Bean, J.; Theuer, C. P.; Barnhart, T. E.; Liu, Z.; Cai, W. In Vivo Targeting and Imaging of Tumor Vasculature with Radiolabeled, Antibody-Conjugated Nanographene ACS Nano 2012, 6, 2361 2370
  5. 5
    Shi, S.; Yang, K.; Hong, H.; Valdovinos, H. F.; Nayak, T. R.; Zhang, Y.; Theuer, C. P.; Barnhart, T. E.; Liu, Z.; Cai, W. Tumor Vasculature Targeting and Imaging in Living Mice with Reduced Graphene Oxide Biomaterials 2013, 34, 3002 3009
  6. 6
    Rosen, L. S.; Hurwitz, H. I.; Wong, M. K.; Goldman, J.; Mendelson, D. S.; Figg, W. D.; Spencer, S.; Adams, B. J.; Alvarez, D.; Seon, B. K.; Theuer, C. P.; Leigh, B. R.; Gordon, M. S. A Phase I First-in-Human Study of TRC105 (Anti-Endoglin Antibody) in Patients with Advanced Cancer Clin. Cancer Res. 2012, 18, 4820 9
  7. 7
    Seon, B. K.; Haba, A.; Matsuno, F.; Takahashi, N.; Tsujie, M.; She, X.; Harada, N.; Uneda, S.; Tsujie, T.; Toi, H.; Tsai, H.; Haruta, Y. Endoglin-Targeted Cancer Therapy Curr. Drug Delivery 2011, 8, 135 143
  8. 8
    Fonsatti, E.; Nicolay, H. J.; Altomonte, M.; Covre, A.; Maio, M. Targeting Cancer Vasculature Via Endoglin/CD105: A Novel Antibody-Based Diagnostic and Therapeutic Strategy in Solid Tumours Cardiovasc. Res. 2010, 86, 12 19
  9. 9
    Hong, H.; Yang, Y.; Zhang, Y.; Engle, J. W.; Barnhart, T. E.; Nickles, R. J.; Leigh, B. R.; Cai, W. Positron Emission Tomography Imaging of CD105 Expression During Tumor Angiogenesis Eur. J. Nucl. Med. Mol. Imaging 2011, 38, 1335 1343
  10. 10
    Hong, H.; Zhang, Y.; Orbay, H.; Valdovinos, H. F.; Nayak, T. R.; Bean, J.; Theuer, C. P.; Barnhart, T. E.; Cai, W. Positron Emission Tomography Imaging of Tumor Angiogenesis with a (61/64)Cu-Labeled F(ab′)(2) Antibody Fragment Mol. Pharm. 2013, 10, 709 716
  11. 11
    James, M. L.; Gambhir, S. S. A Molecular Imaging Primer: Modalities, Imaging Agents, and Applications Physiol. Rev. 2012, 92, 897 965
  12. 12
    Gambhir, S. S. Molecular Imaging of Cancer with Positron Emission Tomography Nature Rev. Cancer 2002, 2, 683 693
  13. 13
    Cai, W.; Chen, K.; Li, Z. B.; Gambhir, S. S.; Chen, X. Dual-Function Probe for PET and Near-Infrared Fluorescence Imaging of Tumor Vasculature J. Nucl. Med. 2007, 48, 1862 1870
  14. 14
    Chen, K.; Li, Z. B.; Wang, H.; Cai, W.; Chen, X. Dual-Modality Optical and Positron Emission Tomography Imaging of Vascular Endothelial Growth Factor Receptor on Tumor Vasculature Using Quantum Dots Eur. J. Nucl. Med. Mol. Imaging 2008, 35, 2235 2244
  15. 15
    Lee, J.; Lee, T. S.; Ryu, J.; Hong, S.; Kang, M.; Im, K.; Kang, J. H.; Lim, S. M.; Park, S.; Song, R. RGD Peptide-Conjugated Multimodal NaDdF4:Yb3+/Er3+ Nanophosphors for Upconversion Luminescence, MR, and PET Imaging of Tumor Angiogenesis J. Nucl. Med. 2013, 54, 96 103
  16. 16
    Sun, Y.; Yu, M.; Liang, S.; Zhang, Y.; Li, C.; Mou, T.; Yang, W.; Zhang, X.; Li, B.; Huang, C.; Li, F. Fluorine-18 Labeled Rare-Earth Nanoparticles for Positron Emission Tomography (PET) Imaging of Sentinel Lymph Node Biomaterials 2011, 32, 2999 3007
  17. 17
    Xie, J.; Chen, K.; Huang, J.; Lee, S.; Wang, J.; Gao, J.; Li, X.; Chen, X. PET/NIRF/MRI Triple Functional Iron Oxide Nanoparticles Biomaterials 2010, 31, 3016 3022
  18. 18
    Zhou, J.; Yu, M.; Sun, Y.; Zhang, X.; Zhu, X.; Wu, Z.; Wu, D.; Li, F. Fluorine-18-Labeled Gd3+/Yb3+/Er3+ Co-Doped NaYF4 Nanophosphors for Multimodality PET/MR/UCL Imaging Biomaterials 2011, 32, 1148 1156
  19. 19
    Huang, X.; Zhang, F.; Lee, S.; Swierczewska, M.; Kiesewetter, D. O.; Lang, L.; Zhang, G.; Zhu, L.; Gao, H.; Choi, H. S.; Niu, G.; Chen, X. Long-Term Multimodal Imaging of Tumor Draining Sentinel Lymph Nodes Using Mesoporous Silica-Based Nanoprobes Biomaterials 2012, 33, 4370 4378
  20. 20
    Zhang, Y.; Hong, H.; Engle, J. W.; Yang, Y.; Barnhart, T. E.; Cai, W. Positron Emission Tomography and Near-Infrared Fluorescence Imaging of Vascular Endothelial Growth Factor with Dual-Labeled Bevacizumab Am. J. Nucl. Med. Mol. Imaging 2012, 2, 1 13
  21. 21
    Vallet-Regi, M.; Rámila, A.; del Real, R. P.; Pérez-Pariente, J. A New Property of Mcm-41: Drug Delivery System Chem. Mater. 2000, 13, 308 311
  22. 22
    Lu, J.; Liong, M.; Li, Z.; Zink, J. I.; Tamanoi, F. Biocompatibility, Biodistribution, and Drug-Delivery Efficiency of Mesoporous Silica Nanoparticles for Cancer Therapy in Animals Small 2010, 6, 1794 1805
  23. 23
    Meng, H.; Xue, M.; Xia, T.; Ji, Z.; Tarn, D. Y.; Zink, J. I.; Nel, A. E. Use of Size and a Copolymer Design Feature to Improve the Biodistribution and the Enhanced Permeability and Retention Effect of Doxorubicin-Loaded Mesoporous Silica Nanoparticles in a Murine Xenograft Tumor Model ACS Nano 2011, 5, 4131 4144
  24. 24
    Pan, L.; He, Q.; Liu, J.; Chen, Y.; Ma, M.; Zhang, L.; Shi, J. Nuclear-Targeted Drug Delivery of TAT Peptide-Conjugated Monodisperse Mesoporous Silica Nanoparticles J. Am. Chem. Soc. 2012, 134, 5722 5725
  25. 25
    Wang, K.; He, X.; Yang, X.; Shi, H. Functionalized Silica Nanoparticles: A Platform for Fluorescence Imaging at the Cell and Small Animal Levels Acc. Chem. Res. 2013, 46, 1367 1376
  26. 26
    Lee, C. H.; Cheng, S. H.; Wang, Y. J.; Chen, Y. C.; Chen, N. T.; Souris, J.; Chen, C. T.; Mou, C. Y.; Yang, C. S.; Lo, L. W. Near-Infrared Mesoporous Silica Nanoparticles for Optical Imaging: Characterization and in Vivo Biodistribution Adv. Funct. Mater. 2009, 19, 215 222
  27. 27
    Pan, J.; Wan, D.; Gong, J. Pegylated Liposome Coated QDs/Mesoporous Silica Core–Shell Nanoparticles for Molecular Imaging Chem. Commun. 2011, 47, 3442 3444
  28. 28
    Liu, J.; Bu, W.; Zhang, S.; Chen, F.; Xing, H.; Pan, L.; Zhou, L.; Peng, W.; Shi, J. Controlled Synthesis of Uniform and Monodisperse Upconversion Core/Mesoporous Silica Shell Nanocomposites for Bimodal Imaging Chemistry 2012, 18, 2335 2341
  29. 29
    Zhang, Y.; Hong, H.; Orbay, H.; Valdovinos, H. F.; Nayak, T. R.; Theuer, C. P.; Barnhart, T. E.; Cai, W. Pet Imaging of CD105/Endoglin Expression with a (6)(1)/(6)(4)Cu-Labeled Fab Antibody Fragment Eur. J. Nucl. Med. Mol. Imaging 2013, 40, 759 767
  30. 30
    Chen, F.; Hong, H.; Zhang, Y.; Valdovinos, H. F.; Shi, S.; Kwon, G. S.; Theuer, C. P.; Barnhart, T. E.; Cai, W. In Vivo Tumor Targeting and Image-Guided Drug Delivery with Antibody-Conjugated, Radiolabeled Mesoporous Silica Nanoparticles ACS Nano 2013, 7, 9027 9039
  31. 31
    Orbay, H.; Zhang, Y.; Valdovinos, H. F.; Song, G.; Hernandez, R.; Theuer, C. P.; Hacker, T. A.; Nickles, R. J.; Cai, W. Positron Emission Tomography Imaging of CD105 Expression in a Rat Myocardial Infarction Model with (64)Cu-Nota-TRC105 Am. J. Nucl. Med. Mol. Imaging 2013, 4, 1 9

Cited By

Click to copy section linkSection link copied!

This article is cited by 82 publications.

  1. Julen Ariztia, Kathleen Solmont, Nadia Pellegrini Moïse, Simon Specklin, Marie Pierre Heck, Sandrine Lamandé-Langle, Bertrand Kuhnast. PET/Fluorescence Imaging: An Overview of the Chemical Strategies to Build Dual Imaging Tools. Bioconjugate Chemistry 2022, 33 (1) , 24-52. https://doi.org/10.1021/acs.bioconjchem.1c00503
  2. Mukaddes Izci, Christy Maksoudian, Bella B. Manshian, Stefaan J. Soenen. The Use of Alternative Strategies for Enhanced Nanoparticle Delivery to Solid Tumors. Chemical Reviews 2021, 121 (3) , 1746-1803. https://doi.org/10.1021/acs.chemrev.0c00779
  3. Yi-Hsien Cheng, Chunla He, Jim E. Riviere, Nancy A. Monteiro-Riviere, Zhoumeng Lin. Meta-Analysis of Nanoparticle Delivery to Tumors Using a Physiologically Based Pharmacokinetic Modeling and Simulation Approach. ACS Nano 2020, 14 (3) , 3075-3095. https://doi.org/10.1021/acsnano.9b08142
  4. Yin-Jia Cheng, Si-Yong Qin, Yi-Han Ma, Xiao-Sui Chen, Ai-Qing Zhang, Xian-Zheng Zhang. Super-pH-Sensitive Mesoporous Silica Nanoparticle-Based Drug Delivery System for Effective Combination Cancer Therapy. ACS Biomaterials Science & Engineering 2019, 5 (4) , 1878-1886. https://doi.org/10.1021/acsbiomaterials.9b00099
  5. Dalong Ni, Dawei Jiang, Emily B. Ehlerding, Peng Huang, Weibo Cai. Radiolabeling Silica-Based Nanoparticles via Coordination Chemistry: Basic Principles, Strategies, and Applications. Accounts of Chemical Research 2018, 51 (3) , 778-788. https://doi.org/10.1021/acs.accounts.7b00635
  6. Michaela Beck, Tamoghna Mandal, Christian Buske, and Mika Lindén . Serum Protein Adsorption Enhances Active Leukemia Stem Cell Targeting of Mesoporous Silica Nanoparticles. ACS Applied Materials & Interfaces 2017, 9 (22) , 18566-18574. https://doi.org/10.1021/acsami.7b04742
  7. Bastian Ruehle, Daniel L. Clemens, Bai-Yu Lee, Marcus A. Horwitz, and Jeffrey I. Zink . A Pathogen-Specific Cargo Delivery Platform Based on Mesoporous Silica Nanoparticles. Journal of the American Chemical Society 2017, 139 (19) , 6663-6668. https://doi.org/10.1021/jacs.7b01278
  8. Gaixia Xu, Shuwen Zeng, Butian Zhang, Mark T. Swihart, Ken-Tye Yong, and Paras N. Prasad . New Generation Cadmium-Free Quantum Dots for Biophotonics and Nanomedicine. Chemical Reviews 2016, 116 (19) , 12234-12327. https://doi.org/10.1021/acs.chemrev.6b00290
  9. Anyanee Kamkaew, Feng Chen, Yonghua Zhan, Rebecca L. Majewski, and Weibo Cai . Scintillating Nanoparticles as Energy Mediators for Enhanced Photodynamic Therapy. ACS Nano 2016, 10 (4) , 3918-3935. https://doi.org/10.1021/acsnano.6b01401
  10. Christopher G. England, Reinier Hernandez, Savo Bou Zein Eddine, and Weibo Cai . Molecular Imaging of Pancreatic Cancer with Antibodies. Molecular Pharmaceutics 2016, 13 (1) , 8-24. https://doi.org/10.1021/acs.molpharmaceut.5b00626
  11. Alyssa B. Chinen, Chenxia M. Guan, Jennifer R. Ferrer, Stacey N. Barnaby, Timothy J. Merkel, and Chad A. Mirkin . Nanoparticle Probes for the Detection of Cancer Biomarkers, Cells, and Tissues by Fluorescence. Chemical Reviews 2015, 115 (19) , 10530-10574. https://doi.org/10.1021/acs.chemrev.5b00321
  12. Shreya Goel, Feng Chen, Hao Hong, Hector F. Valdovinos, Reinier Hernandez, Sixiang Shi, Todd E. Barnhart, and Weibo Cai . VEGF121-Conjugated Mesoporous Silica Nanoparticle: A Tumor Targeted Drug Delivery System. ACS Applied Materials & Interfaces 2014, 6 (23) , 21677-21685. https://doi.org/10.1021/am506849p
  13. Moumita Sil, Dipro Mukherjee, Arunava Goswami, Moupriya Nag, Dibyajit Lahiri, Debasmita Bhattacharya. Antibiofilm activity of mesoporous silica nanoparticles against the biofilm associated infections. Naunyn-Schmiedeberg's Archives of Pharmacology 2024, 397 (6) , 3617-3633. https://doi.org/10.1007/s00210-023-02872-0
  14. Santosh Sarnaik, Dhananjay Bhatane, Sharon Rose Pamshong, Amit Alexander. Cutting-edge advancements in anticancer drug delivery and scope for theranostics using biocompatible multifunctional mesoporous silica nanoparticles. Journal of Drug Delivery Science and Technology 2024, 94 , 105504. https://doi.org/10.1016/j.jddst.2024.105504
  15. Bingqing Lu, Jiaqi Wang, A. Jan Hendriks, Tom M. Nolte. Clearance of nanoparticles from blood: effects of hydrodynamic size and surface coatings. Environmental Science: Nano 2024, 11 (1) , 406-417. https://doi.org/10.1039/D3EN00812F
  16. Elif Tugce SARCAN. NANOPARTICLES FOR DUAL IMAGING: PET AND FLUORESCENCE IMAGING. Ankara Universitesi Eczacilik Fakultesi Dergisi 2024, 48 (2) , 3-3. https://doi.org/10.33483/jfpau.1323924
  17. Jessica C. Hsu, Zhongmin Tang, Olga E. Eremina, Alexandros Marios Sofias, Twan Lammers, Jonathan F. Lovell, Cristina Zavaleta, Weibo Cai, David P. Cormode. Nanomaterial-based contrast agents. Nature Reviews Methods Primers 2023, 3 (1) https://doi.org/10.1038/s43586-023-00211-4
  18. Nahid S Awad, Najla M Salkho, Waad H Abuwatfa, Vinod Paul, Nour M AlSawaftah, Ghaleb A Husseini. Tumor vasculature vs tumor cell targeting: Understanding the latest trends in using functional nanoparticles for cancer treatment. OpenNano 2023, 11 , 100136. https://doi.org/10.1016/j.onano.2023.100136
  19. Mokshada Kumar, Priyanka Kulkarni, Shufang Liu, Nagendra Chemuturi, Dhaval K. Shah. Nanoparticle biodistribution coefficients: A quantitative approach for understanding the tissue distribution of nanoparticles. Advanced Drug Delivery Reviews 2023, 194 , 114708. https://doi.org/10.1016/j.addr.2023.114708
  20. Neetu Singh, Sixiang Shi, Shreya Goel. Ultrasmall silica nanoparticles in translational biomedical research: Overview and outlook. Advanced Drug Delivery Reviews 2023, 192 , 114638. https://doi.org/10.1016/j.addr.2022.114638
  21. Fatemeh Ahmadi, Arezoo Sodagar-Taleghani, Pedram Ebrahimnejad, Seyyed Pouya Hadipour Moghaddam, Farzam Ebrahimnejad, Kofi Asare-Addo, Ali Nokhodchi. A review on the latest developments of mesoporous silica nanoparticles as a promising platform for diagnosis and treatment of cancer. International Journal of Pharmaceutics 2022, 625 , 122099. https://doi.org/10.1016/j.ijpharm.2022.122099
  22. Vincent Mittelheisser, Pierre Coliat, Eric Moeglin, Lilian Goepp, Jacky G. Goetz, Loic J. Charbonnière, Xavier Pivot, Alexandre Detappe. Optimal Physicochemical Properties of Antibody–Nanoparticle Conjugates for Improved Tumor Targeting. Advanced Materials 2022, 34 (24) https://doi.org/10.1002/adma.202110305
  23. Marcin Woźniak, Agata Płoska, Anna Siekierzycka, Lawrence W. Dobrucki, Leszek Kalinowski, Iwona T. Dobrucki. Molecular Imaging and Nanotechnology—Emerging Tools in Diagnostics and Therapy. International Journal of Molecular Sciences 2022, 23 (5) , 2658. https://doi.org/10.3390/ijms23052658
  24. Ardalan Chaichi, Elnaz Sheikh, Supratik Mukhopadhyay, Manas R. Gartia. Trends in Nanotechnology Development in Medical Applications. 2022, 913-919. https://doi.org/10.1016/B978-0-12-820352-1.00112-7
  25. Arezoo Sodagar Taleghani, Ali Taghvaie Nakhjiri, Mohammad Javad Khakzad, Seyed Mahdi Rezayat, Pedram Ebrahimnejad, Amir Heydarinasab, Azim Akbarzadeh, Azam Marjani. Mesoporous silica nanoparticles as a versatile nanocarrier for cancer treatment: A review. Journal of Molecular Liquids 2021, 328 , 115417. https://doi.org/10.1016/j.molliq.2021.115417
  26. Teresa González Muñoz, Ana Teresa Amaral, Pilar Puerto-Camacho, Héctor Peinado, Enrique de Álava. Endoglin in the Spotlight to Treat Cancer. International Journal of Molecular Sciences 2021, 22 (6) , 3186. https://doi.org/10.3390/ijms22063186
  27. Mohsen Ghaferi, Maedeh Koohi Moftakhari Esfahani, Aun Raza, Sitah Al Harthi, Hasan Ebrahimi Shahmabadi, Seyed Ebrahim Alavi. Mesoporous silica nanoparticles: synthesis methods and their therapeutic use-recent advances. Journal of Drug Targeting 2021, 29 (2) , 131-154. https://doi.org/10.1080/1061186X.2020.1812614
  28. Ziyuan Li, Yingwen Mu, Cheng Peng, Martin F. Lavin, Hua Shao, Zhongjun Du. Understanding the mechanisms of silica nanoparticles for nanomedicine. WIREs Nanomedicine and Nanobiotechnology 2021, 13 (1) https://doi.org/10.1002/wnan.1658
  29. Gabriela Capriotti, Michela Varani, Chiara Lauri, Gabriele Franchi, Patrizia Pizzichini, Alberto Signore. Copper-64 labeled nanoparticles for positron emission tomography imaging: a review of the recent literature. The Quarterly Journal of Nuclear Medicine and Molecular Imaging 2020, 64 (4) https://doi.org/10.23736/S1824-4785.20.03315-4
  30. Sepanta Hosseinpour, Laurence J. Walsh, Chun Xu. Biomedical application of mesoporous silica nanoparticles as delivery systems: a biological safety perspective. Journal of Materials Chemistry B 2020, 8 (43) , 9863-9876. https://doi.org/10.1039/D0TB01868F
  31. Zhuoran Ma, Feifei Wang, Yeteng Zhong, Felix Salazar, Jiachen Li, Mingxi Zhang, Fuqiang Ren, Anna M. Wu, Hongjie Dai. Cross‐Link‐Functionalized Nanoparticles for Rapid Excretion in Nanotheranostic Applications. Angewandte Chemie International Edition 2020, 59 (46) , 20552-20560. https://doi.org/10.1002/anie.202008083
  32. Zhuoran Ma, Feifei Wang, Yeteng Zhong, Felix Salazar, Jiachen Li, Mingxi Zhang, Fuqiang Ren, Anna M. Wu, Hongjie Dai. Cross‐Link‐Functionalized Nanoparticles for Rapid Excretion in Nanotheranostic Applications. Angewandte Chemie 2020, 132 (46) , 20733-20741. https://doi.org/10.1002/ange.202008083
  33. Sang Bong Lee, Ho Won Lee, Bobby Aditya Darmawan, In-Kyu Lee, Sung Jin Cho, Jungwook Chin, Sang Kyoon Kim, Jong-Oh Park, Kil Soo Kim, Sang-Woo Lee, Jaetae Lee, Yong Hyun Jeon. NIR dye-loaded mesoporous silica nanoparticles for a multifunctional theranostic platform: Visualization of tumor and ischemic lesions, and performance of photothermal therapy. Journal of Industrial and Engineering Chemistry 2020, 88 , 99-105. https://doi.org/10.1016/j.jiec.2020.03.027
  34. Lisa Haddick, Wei Zhang, Sören Reinhard, Karin Möller, Hanna Engelke, Ernst Wagner, Thomas Bein. Particle-Size-Dependent Delivery of Antitumoral miRNA Using Targeted Mesoporous Silica Nanoparticles. Pharmaceutics 2020, 12 (6) , 505. https://doi.org/10.3390/pharmaceutics12060505
  35. Sugata Barui, Valentina Cauda. Multimodal Decorations of Mesoporous Silica Nanoparticles for Improved Cancer Therapy. Pharmaceutics 2020, 12 (6) , 527. https://doi.org/10.3390/pharmaceutics12060527
  36. Aswathy Ravindran Girija, Vivekanandan Palaninathan. Multifunctional Mesoporous Silica Nanoparticles for Biomedical Applications. 2020, 213-235. https://doi.org/10.1007/978-981-15-6255-6_9
  37. Marco A. Downing, Piyush K. Jain. Mesoporous silica nanoparticles: synthesis, properties, and biomedical applications. 2020, 267-281. https://doi.org/10.1016/B978-0-12-816662-8.00016-3
  38. Md Aquib, Muhammad A. Farooq, Parikshit Banerjee, Fahad Akhtar, Mensura S. Filli, Kofi O. Boakye‐Yiadom, Samuel Kesse, Faisal Raza, Mily B. J. Maviah, Rukhshona Mavlyanova, Bo Wang. Targeted and stimuli–responsive mesoporous silica nanoparticles for drug delivery and theranostic use. Journal of Biomedical Materials Research Part A 2019, 107 (12) , 2643-2666. https://doi.org/10.1002/jbm.a.36770
  39. Seong H. Song, Han G. Kang, Young B. Han, Ho-Young Lee, Dae H. Jeong, Soo M. Kim, Seong J. Hong. Characterization and validation of multimodal annihilation-gamma/near-infrared/visible laparoscopic system. Journal of Biomedical Optics 2019, 24 (09) , 1. https://doi.org/10.1117/1.JBO.24.9.096008
  40. Dalong Ni, Emily B. Ehlerding, Weibo Cai. Multimodale Kontrastmittel für die kombinierte Positronenemissionstomographie. Angewandte Chemie 2019, 131 (9) , 2592-2602. https://doi.org/10.1002/ange.201806853
  41. Dalong Ni, Emily B. Ehlerding, Weibo Cai. Multimodality Imaging Agents with PET as the Fundamental Pillar. Angewandte Chemie International Edition 2019, 58 (9) , 2570-2579. https://doi.org/10.1002/anie.201806853
  42. Bong Geun Cha, Jaeyun Kim. Functional mesoporous silica nanoparticles for bio‐imaging applications. WIREs Nanomedicine and Nanobiotechnology 2019, 11 (1) https://doi.org/10.1002/wnan.1515
  43. Feng Chen, Kai Ma, Brian Madajewski, Li Zhuang, Li Zhang, Keith Rickert, Marcello Marelli, Barney Yoo, Melik Z. Turker, Michael Overholtzer, Thomas P. Quinn, Mithat Gonen, Pat Zanzonico, Anthony Tuesca, Michael A. Bowen, Larry Norton, J. Anand Subramony, Ulrich Wiesner, Michelle S. Bradbury. Ultrasmall targeted nanoparticles with engineered antibody fragments for imaging detection of HER2-overexpressing breast cancer. Nature Communications 2018, 9 (1) https://doi.org/10.1038/s41467-018-06271-5
  44. Terisse A. Brocato, Eric N. Coker, Paul N. Durfee, Yu-Shen Lin, Jason Townson, Edward F. Wyckoff, Vittorio Cristini, C. Jeffrey Brinker, Zhihui Wang. Understanding the Connection between Nanoparticle Uptake and Cancer Treatment Efficacy using Mathematical Modeling. Scientific Reports 2018, 8 (1) https://doi.org/10.1038/s41598-018-25878-8
  45. Feng Chen, Shreya Goel, Sixiang Shi, Todd E. Barnhart, Xiaoli Lan, Weibo Cai. General synthesis of silica-based yolk/shell hybrid nanomaterials and in vivo tumor vasculature targeting. Nano Research 2018, 11 (9) , 4890-4904. https://doi.org/10.1007/s12274-018-2078-9
  46. Michela Varani, Filippo Galli, Sveva Auletta, Alberto Signore. Radiolabelled nanoparticles for cancer diagnosis. Clinical and Translational Imaging 2018, 6 (4) , 271-292. https://doi.org/10.1007/s40336-018-0283-x
  47. Antti Rahikkala, Sarah A. P. Pereira, Patrícia Figueiredo, Marieta L. C. Passos, André R. T. S. Araújo, M. Lúcia M. F. S. Saraiva, Hélder A. Santos. Mesoporous Silica Nanoparticles for Targeted and Stimuli‐Responsive Delivery of Chemotherapeutics: A Review. Advanced Biosystems 2018, 2 (7) https://doi.org/10.1002/adbi.201800020
  48. Xiaoke Zheng, Shuli Zeng, Jing Hu, Lan Wu, Xiandeng Hou. Applications of silica-based nanoparticles for multimodal bioimaging. Applied Spectroscopy Reviews 2018, 53 (5) , 377-394. https://doi.org/10.1080/05704928.2017.1355312
  49. Shuang-Lin Deng, Yun-Qian Li, Gang Zhao. Imaging Gliomas with Nanoparticle-Labeled Stem Cells. Chinese Medical Journal 2018, 131 (6) , 721-730. https://doi.org/10.4103/0366-6999.226900
  50. Jonas G. Croissant, Yevhen Fatieiev, Abdulaziz Almalik, Niveen M. Khashab. Mesoporous Silica and Organosilica Nanoparticles: Physical Chemistry, Biosafety, Delivery Strategies, and Biomedical Applications. Advanced Healthcare Materials 2018, 7 (4) https://doi.org/10.1002/adhm.201700831
  51. Feng Chen. Size-, Shape- and Charge-Dependent Pharmacokinetics of Radiolabeled Nanoparticles. 2018, 313-329. https://doi.org/10.1007/978-3-319-67720-0_17
  52. Feby Wijaya Pratiwi, Chiung Wen Kuo, Si-Han Wu, Yi-Ping Chen, Chung Yuan Mou, Peilin Chen. The Bioimaging Applications of Mesoporous Silica Nanoparticles. 2018, 123-153. https://doi.org/10.1016/bs.enz.2018.07.006
  53. Tao Wang, Guorong Jia, Chao Cheng, Qiuhu Wang, Xiao Li, Yanyan Liu, Chaofan He, Luguang Chen, Gaofeng Sun, Changjing Zuo. Active targeted dual-modal CT/MR imaging of VX2 tumors using PEGylated BaGdF 5 nanoparticles conjugated with RGD. New Journal of Chemistry 2018, 42 (14) , 11565-11572. https://doi.org/10.1039/C8NJ01527A
  54. Elisabetta Avitabile, Davide Bedognetti, Gianni Ciofani, Alberto Bianco, Lucia Gemma Delogu. How can nanotechnology help the fight against breast cancer?. Nanoscale 2018, 10 (25) , 11719-11731. https://doi.org/10.1039/C8NR02796J
  55. Nai-Tzu Chen, Jeffrey S. Souris, Shih-Hsun Cheng, Chia-Hui Chu, Yu-Chao Wang, Vani Konda, Urszula Dougherty, Marc Bissonnette, Chung-Yuan Mou, Chin-Tu Chen, Leu-Wei Lo. Lectin-functionalized mesoporous silica nanoparticles for endoscopic detection of premalignant colonic lesions. Nanomedicine: Nanotechnology, Biology and Medicine 2017, 13 (6) , 1941-1952. https://doi.org/10.1016/j.nano.2017.03.014
  56. Anna Watermann, Juergen Brieger. Mesoporous Silica Nanoparticles as Drug Delivery Vehicles in Cancer. Nanomaterials 2017, 7 (7) , 189. https://doi.org/10.3390/nano7070189
  57. Musafar Sikkandhar, Anu Nedumaran, Roopa Ravichandar, Satnam Singh, Induja Santhakumar, Zheng Goh, Sachin Mishra, Govindaraju Archunan, Balázs Gulyás, Parasuraman Padmanabhan. Theranostic Probes for Targeting Tumor Microenvironment: An Overview. International Journal of Molecular Sciences 2017, 18 (5) , 1036. https://doi.org/10.3390/ijms18051036
  58. Shreya Goel, Christopher G. England, Feng Chen, Weibo Cai. Positron emission tomography and nanotechnology: A dynamic duo for cancer theranostics. Advanced Drug Delivery Reviews 2017, 113 , 157-176. https://doi.org/10.1016/j.addr.2016.08.001
  59. , , , Juan L. Vivero-Escoto, Laura Moore Jeffords, Didier Dréau, Merlis Alvarez-Berrios, Pinku Mukherjee. Mucin1 antibody-conjugated dye-doped mesoporous silica nanoparticles for breast cancer detection in vivo. 2017, 100780B. https://doi.org/10.1117/12.2252369
  60. Marie-Caline Z. Abadjian, Jaeyeon Choi, Carolyn J. Anderson. Nanoparticles for PET Imaging of Tumors and Cancer Metastasis. 2017, 229-255. https://doi.org/10.1007/978-3-319-42169-8_11
  61. Daniel A. Richards, Antoine Maruani, Vijay Chudasama. Antibody fragments as nanoparticle targeting ligands: a step in the right direction. Chemical Science 2017, 8 (1) , 63-77. https://doi.org/10.1039/C6SC02403C
  62. Larissa Kramer, Gordon Winter, Benjamin Baur, Andrea J. Kuntz, Thomas Kull, Christoph Solbach, Ambros J. Beer, Mika Lindén. Quantitative and correlative biodistribution analysis of 89 Zr-labeled mesoporous silica nanoparticles intravenously injected into tumor-bearing mice. Nanoscale 2017, 9 (27) , 9743-9753. https://doi.org/10.1039/C7NR02050C
  63. Moumita Roy Chowdhury, Canan Schumann, Dipita Bhakta-Guha, Gunjan Guha. Cancer nanotheranostics: Strategies, promises and impediments. Biomedicine & Pharmacotherapy 2016, 84 , 291-304. https://doi.org/10.1016/j.biopha.2016.09.035
  64. Saeideh Same, Ayuob Aghanejad, Sattar Akbari Nakhjavani, Jaleh Barar, Yadollah Omidi. Radiolabeled theranostics: magnetic and gold nanoparticles. Bioimpacts 2016, 6 (3) , 169-181. https://doi.org/10.15171/bi.2016.23
  65. Jinyu Li, Dandan Liu, Guoxin Tan, Zhinan Zhao, Xinggang Yang, Weisan Pan. A comparative study on the efficiency of chitosan-N-acetylcysteine, chitosan oligosaccharides or carboxymethyl chitosan surface modified nanostructured lipid carrier for ophthalmic delivery of curcumin. Carbohydrate Polymers 2016, 146 , 435-444. https://doi.org/10.1016/j.carbpol.2016.03.079
  66. Weifei Lu, Hao Hong, Weibo Cai. Radio-nanomaterials for biomedical applications: state of the art. European Journal of Nanomedicine 2016, 8 (3) , 151-170. https://doi.org/10.1515/ejnm-2016-0011
  67. Pablo Hervella, Elisa Parra, David Needham. Encapsulation and retention of chelated-copper inside hydrophobic nanoparticles: Liquid cored nanoparticles show better retention than a solid core formulation. European Journal of Pharmaceutics and Biopharmaceutics 2016, 102 , 64-76. https://doi.org/10.1016/j.ejpb.2016.02.015
  68. Yannan Yang, Chengzhong Yu. Advances in silica based nanoparticles for targeted cancer therapy. Nanomedicine: Nanotechnology, Biology and Medicine 2016, 12 (2) , 317-332. https://doi.org/10.1016/j.nano.2015.10.018
  69. Jessica Maria Rosenholm, Jixi Zhang, Mika Linden, Cecilia Sahlgren. Mesoporous Silica Nanoparticles in Tissue Engineering – a Perspective. Nanomedicine 2016, 11 (4) , 391-402. https://doi.org/10.2217/nnm.15.212
  70. Huiyu Liu, Linlin Li, Shunhao Wang, Qi Yang. Multifunctional Mesoporous/Hollow Silica for Cancer Nanotheranostics. 2016, 307-354. https://doi.org/10.1007/978-981-10-0063-8_9
  71. Shanka Walia, Amitabha Acharya. Theragnosis: Nanoparticles as a Tool for Simultaneous Therapy and Diagnosis. 2016, 127-152. https://doi.org/10.1007/978-981-10-0818-4_6
  72. Truffi Marta, Sorrentino Luca, Mazzucchelli Serena, Fiandra Luisa, Corsi Fabio. What Is the Role of Nanotechnology in Diagnosis and Treatment of Metastatic Breast Cancer? Promising Scenarios for the Near Future. Journal of Nanomaterials 2016, 2016 , 1-16. https://doi.org/10.1155/2016/5436458
  73. Derek Reichel, Piotr Rychahou, Younsoo Bae. Polymer Nanoassemblies With solvato- and halo-fluorochromism for Drug Release Monitoring and Metastasis Imaging. Therapeutic Delivery 2015, 6 (10) , 1221-1237. https://doi.org/10.4155/tde.15.59
  74. Diego Stéfani T. Martinez, Amauri J. Paula, Leandro C. Fonseca, Luis Augusto V. Luna, Camila P. Silveira, Nelson Durán, Oswaldo L. Alves. Monitoring the Hemolytic Effect of Mesoporous Silica Nanoparticles after Human Blood Protein Corona Formation. European Journal of Inorganic Chemistry 2015, 2015 (27) , 4595-4602. https://doi.org/10.1002/ejic.201500573
  75. Sixiang Shi, Hao Hong, Hakan Orbay, Stephen A. Graves, Yunan Yang, Jakob D. Ohman, Bai Liu, Robert J. Nickles, Hing C. Wong, Weibo Cai. ImmunoPET of tissue factor expression in triple-negative breast cancer with a radiolabeled antibody Fab fragment. European Journal of Nuclear Medicine and Molecular Imaging 2015, 42 (8) , 1295-1303. https://doi.org/10.1007/s00259-015-3038-1
  76. Sixiang Shi, Hakan Orbay, Yunan Yang, Stephen A. Graves, Tapas R. Nayak, Hao Hong, Reinier Hernandez, Haiming Luo, Shreya Goel, Charles P. Theuer, Robert J. Nickles, Weibo Cai. PET Imaging of Abdominal Aortic Aneurysm with 64 Cu-Labeled Anti-CD105 Antibody Fab Fragment. Journal of Nuclear Medicine 2015, 56 (6) , 927-932. https://doi.org/10.2967/jnumed.114.153098
  77. Metin Sitti, Hakan Ceylan, Wenqi Hu, Joshua Giltinan, Mehmet Turan, Sehyuk Yim, Eric Diller. Biomedical Applications of Untethered Mobile Milli/Microrobots. Proceedings of the IEEE 2015, 103 (2) , 205-224. https://doi.org/10.1109/JPROC.2014.2385105
  78. Claudia Caltagirone, Alexandre Bettoschi, Alessandra Garau, Riccardo Montis. Silica-based nanoparticles: a versatile tool for the development of efficient imaging agents. Chemical Society Reviews 2015, 44 (14) , 4645-4671. https://doi.org/10.1039/C4CS00270A
  79. C. P. Silveira, L. M. Apolinário, W. J. Fávaro, A. J. Paula, N. Durán. Hybrid biomaterial based on porous silica nanoparticles and Pluronic F-127 for sustained release of sildenafil: in vivo study on prostate cancer. RSC Advances 2015, 5 (99) , 81348-81355. https://doi.org/10.1039/C5RA15006J
  80. Ranjith Kumar Kankala, Yaswanth Kuthati, Chen-Lun Liu, Chung-Yuan Mou, Chia-Hung Lee. Killing cancer cells by delivering a nanoreactor for inhibition of catalase and catalytically enhancing intracellular levels of ROS. RSC Advances 2015, 5 (105) , 86072-86081. https://doi.org/10.1039/C5RA16023E
  81. Gonzalo Villaverde, Alejandro Baeza, Gustavo J. Melen, Arantzazu Alfranca, Manuel Ramirez, Maria Vallet-Regí. A new targeting agent for the selective drug delivery of nanocarriers for treating neuroblastoma. Journal of Materials Chemistry B 2015, 3 (24) , 4831-4842. https://doi.org/10.1039/C5TB00287G
  82. Christopher T. Turner, Steven J. P. McInnes, Nicolas H. Voelcker, Allison J. Cowin, . Therapeutic Potential of Inorganic Nanoparticles for the Delivery of Monoclonal Antibodies. Journal of Nanomaterials 2015, 2015 (1) https://doi.org/10.1155/2015/309602
Open PDF

Molecular Pharmaceutics

Cite this: Mol. Pharmaceutics 2014, 11, 11, 4007–4014
Click to copy citationCitation copied!
https://doi.org/10.1021/mp500306k
Published June 17, 2014

Copyright © 2014 American Chemical Society. This publication is licensed under these Terms of Use.

Article Views

2574

Altmetric

-

Citations

Learn about these metrics

Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.

  • Abstract

    Figure 1

    Figure 1. Synthesis and characterization of TRC105(Fab). (a) Schematic illustration showing the generation of TRC105(Fab) and its thiolation. (b) SDS-PAGE of molecular weight markers (lane 1), intact TRC105 antibody (lane 2), and TRC105(Fab) after purification on the Sephadex G-75 column (lane 3).

    Scheme 1

    Scheme 1. Schematic Illustration of the Synthesis of 64Cu-MSN-800CW-TRC105(Fab)a

    Scheme aUniform MSN nanoparticle (1) was first modified with −NH2 groups with APS to form MSN-NH2 (2). As-synthesized MSN-NH2 was then subjected to 800CW and NOTA conjugation to yield NOTA-MSN-800CW-NH2 (3). Afterwards, PEGylation step was introduced to render the stability of NOTA-MSN-800CW-PEG-Mal (4) in PBS, at the same time adding maleimide groups. NOTA-MSN-800CW-PEG-TRC105(Fab) (5) could be obtained by reacting TRC105(Fab)-SH with 4 at room temperature. 64Cu-labeling was performed in the last step to generate 64Cu-NOTA-MSN-800CW-PEG-TRC105(Fab) (6), short for 64Cu-MSN-800CW-TRC105(Fab).

    Figure 2

    Figure 2. Synthesis and characterization of MSN and MSN-800CW-TRC105(Fab). TEM images of (a) pure MSN, (b) MSN-800CW-TRC105(Fab). Insets in (a) and (b) show the schemes of MSN and MSN-800CW-TRC105(Fab). An optical image of MSN-800CW-TRC105(Fab) acquired from IVIS spectrum in vivo imaging system (Ex = 745 nm, Em = 800 nm) is also shown in the upper right portion of (b).

    Figure 3

    Figure 3. Flow cytometry analysis of MSN nanoconjugates in HUVECs (CD105 positive) after 30 min incubation and subsequent washing. Targeted group: fluorescein conjugated MSN-800CW-TRC105(Fab). Non-targeted group: fluorescein conjugated MSN-800CW. Blocking group: fluorescein conjugated MSN-800CW-TRC105(Fab) with a blocking dose of TRC105 (500 μg/mL).

    Figure 4

    Figure 4. Serial coronal PET images of 4T1 tumor-bearing mice at different time points postinjection of (a) 64Cu-MSN-800CW-TRC105(Fab), (b) 64Cu-MSN-800CW, and (c) 64Cu-MSN-800CW-TRC105(Fab) with a blocking dose of TRC105 (1 mg/mouse). Tumors are indicated by yellow arrowheads.

    Figure 5

    Figure 5. In vivo NIRF imaging of 4T1 tumor-bearing mice at 4 h postinjection. (a) Targeted group, 64Cu-MSN-800CW-TRC105(Fab); (b) non-targeted group, 64Cu-MSN-800CW; (c) blocking group, 64Cu-MSN-800CW-TRC105(Fab) with a blocking dose of TRC105 (1 mg/mouse). Tumors are indicated by yellow arrowheads. Each mouse was iv injected with MSN nanoconjugates with equal amounts of 800CW dyes (∼400 pmol). All images were acquired by using IVIS spectrum in vivo imaging system (Ex = 745 nm, Em = 800 nm).

    Figure 6

    Figure 6. Quantitative analysis of the PET data. (a) Time–activity curve of the liver, 4T1 tumor, blood, and muscle upon iv injection of 64Cu-MSN-800CW-TRC105(Fab) (targeted group, n = 3). (b) Time–activity curve of tumor-to-muscle (T/M), tumor-to-blood (T/B), and tumor-to-liver (T/L) ratios from the same targeted group (n = 3). (c) Comparison of 4T1 tumor uptake among the three groups. The differences between 4T1 tumor uptake of 64Cu-MSN-800CW-TRC105(Fab) and the two control groups were statistically significant (**P < 0.01) in all cases. (d) Biodistribution of three groups in 4T1 tumor-bearing mice at the end of the PET scans at 48 h pi (n = 3).

  • References


    This article references 31 other publications.

    1. 1
      Chen, F.; Cai, W. Tumor Vasculature Targeting: A Generally Applicable Approach for Functionalized Nanomaterials Small 2014, 10, 1887 1893
    2. 2
      Cai, W.; Shin, D. W.; Chen, K.; Gheysens, O.; Cao, Q.; Wang, S. X.; Gambhir, S. S.; Chen, X. Peptide-Labeled Near-Infrared Quantum Dots for Imaging Tumor Vasculature in Living Subjects Nano Lett. 2006, 6, 669 676
    3. 3
      Liu, Z.; Cai, W.; He, L.; Nakayama, N.; Chen, K.; Sun, X.; Chen, X.; Dai, H. In Vivo Biodistribution and Highly Efficient Tumour Targeting of Carbon Nanotubes in Mice Nature Nanotechnol. 2007, 2, 47 52
    4. 4
      Hong, H.; Yang, K.; Zhang, Y.; Engle, J. W.; Feng, L.; Yang, Y.; Nayak, T. R.; Goel, S.; Bean, J.; Theuer, C. P.; Barnhart, T. E.; Liu, Z.; Cai, W. In Vivo Targeting and Imaging of Tumor Vasculature with Radiolabeled, Antibody-Conjugated Nanographene ACS Nano 2012, 6, 2361 2370
    5. 5
      Shi, S.; Yang, K.; Hong, H.; Valdovinos, H. F.; Nayak, T. R.; Zhang, Y.; Theuer, C. P.; Barnhart, T. E.; Liu, Z.; Cai, W. Tumor Vasculature Targeting and Imaging in Living Mice with Reduced Graphene Oxide Biomaterials 2013, 34, 3002 3009
    6. 6
      Rosen, L. S.; Hurwitz, H. I.; Wong, M. K.; Goldman, J.; Mendelson, D. S.; Figg, W. D.; Spencer, S.; Adams, B. J.; Alvarez, D.; Seon, B. K.; Theuer, C. P.; Leigh, B. R.; Gordon, M. S. A Phase I First-in-Human Study of TRC105 (Anti-Endoglin Antibody) in Patients with Advanced Cancer Clin. Cancer Res. 2012, 18, 4820 9
    7. 7
      Seon, B. K.; Haba, A.; Matsuno, F.; Takahashi, N.; Tsujie, M.; She, X.; Harada, N.; Uneda, S.; Tsujie, T.; Toi, H.; Tsai, H.; Haruta, Y. Endoglin-Targeted Cancer Therapy Curr. Drug Delivery 2011, 8, 135 143
    8. 8
      Fonsatti, E.; Nicolay, H. J.; Altomonte, M.; Covre, A.; Maio, M. Targeting Cancer Vasculature Via Endoglin/CD105: A Novel Antibody-Based Diagnostic and Therapeutic Strategy in Solid Tumours Cardiovasc. Res. 2010, 86, 12 19
    9. 9
      Hong, H.; Yang, Y.; Zhang, Y.; Engle, J. W.; Barnhart, T. E.; Nickles, R. J.; Leigh, B. R.; Cai, W. Positron Emission Tomography Imaging of CD105 Expression During Tumor Angiogenesis Eur. J. Nucl. Med. Mol. Imaging 2011, 38, 1335 1343
    10. 10
      Hong, H.; Zhang, Y.; Orbay, H.; Valdovinos, H. F.; Nayak, T. R.; Bean, J.; Theuer, C. P.; Barnhart, T. E.; Cai, W. Positron Emission Tomography Imaging of Tumor Angiogenesis with a (61/64)Cu-Labeled F(ab′)(2) Antibody Fragment Mol. Pharm. 2013, 10, 709 716
    11. 11
      James, M. L.; Gambhir, S. S. A Molecular Imaging Primer: Modalities, Imaging Agents, and Applications Physiol. Rev. 2012, 92, 897 965
    12. 12
      Gambhir, S. S. Molecular Imaging of Cancer with Positron Emission Tomography Nature Rev. Cancer 2002, 2, 683 693
    13. 13
      Cai, W.; Chen, K.; Li, Z. B.; Gambhir, S. S.; Chen, X. Dual-Function Probe for PET and Near-Infrared Fluorescence Imaging of Tumor Vasculature J. Nucl. Med. 2007, 48, 1862 1870
    14. 14
      Chen, K.; Li, Z. B.; Wang, H.; Cai, W.; Chen, X. Dual-Modality Optical and Positron Emission Tomography Imaging of Vascular Endothelial Growth Factor Receptor on Tumor Vasculature Using Quantum Dots Eur. J. Nucl. Med. Mol. Imaging 2008, 35, 2235 2244
    15. 15
      Lee, J.; Lee, T. S.; Ryu, J.; Hong, S.; Kang, M.; Im, K.; Kang, J. H.; Lim, S. M.; Park, S.; Song, R. RGD Peptide-Conjugated Multimodal NaDdF4:Yb3+/Er3+ Nanophosphors for Upconversion Luminescence, MR, and PET Imaging of Tumor Angiogenesis J. Nucl. Med. 2013, 54, 96 103
    16. 16
      Sun, Y.; Yu, M.; Liang, S.; Zhang, Y.; Li, C.; Mou, T.; Yang, W.; Zhang, X.; Li, B.; Huang, C.; Li, F. Fluorine-18 Labeled Rare-Earth Nanoparticles for Positron Emission Tomography (PET) Imaging of Sentinel Lymph Node Biomaterials 2011, 32, 2999 3007
    17. 17
      Xie, J.; Chen, K.; Huang, J.; Lee, S.; Wang, J.; Gao, J.; Li, X.; Chen, X. PET/NIRF/MRI Triple Functional Iron Oxide Nanoparticles Biomaterials 2010, 31, 3016 3022
    18. 18
      Zhou, J.; Yu, M.; Sun, Y.; Zhang, X.; Zhu, X.; Wu, Z.; Wu, D.; Li, F. Fluorine-18-Labeled Gd3+/Yb3+/Er3+ Co-Doped NaYF4 Nanophosphors for Multimodality PET/MR/UCL Imaging Biomaterials 2011, 32, 1148 1156
    19. 19
      Huang, X.; Zhang, F.; Lee, S.; Swierczewska, M.; Kiesewetter, D. O.; Lang, L.; Zhang, G.; Zhu, L.; Gao, H.; Choi, H. S.; Niu, G.; Chen, X. Long-Term Multimodal Imaging of Tumor Draining Sentinel Lymph Nodes Using Mesoporous Silica-Based Nanoprobes Biomaterials 2012, 33, 4370 4378
    20. 20
      Zhang, Y.; Hong, H.; Engle, J. W.; Yang, Y.; Barnhart, T. E.; Cai, W. Positron Emission Tomography and Near-Infrared Fluorescence Imaging of Vascular Endothelial Growth Factor with Dual-Labeled Bevacizumab Am. J. Nucl. Med. Mol. Imaging 2012, 2, 1 13
    21. 21
      Vallet-Regi, M.; Rámila, A.; del Real, R. P.; Pérez-Pariente, J. A New Property of Mcm-41: Drug Delivery System Chem. Mater. 2000, 13, 308 311
    22. 22
      Lu, J.; Liong, M.; Li, Z.; Zink, J. I.; Tamanoi, F. Biocompatibility, Biodistribution, and Drug-Delivery Efficiency of Mesoporous Silica Nanoparticles for Cancer Therapy in Animals Small 2010, 6, 1794 1805
    23. 23
      Meng, H.; Xue, M.; Xia, T.; Ji, Z.; Tarn, D. Y.; Zink, J. I.; Nel, A. E. Use of Size and a Copolymer Design Feature to Improve the Biodistribution and the Enhanced Permeability and Retention Effect of Doxorubicin-Loaded Mesoporous Silica Nanoparticles in a Murine Xenograft Tumor Model ACS Nano 2011, 5, 4131 4144
    24. 24
      Pan, L.; He, Q.; Liu, J.; Chen, Y.; Ma, M.; Zhang, L.; Shi, J. Nuclear-Targeted Drug Delivery of TAT Peptide-Conjugated Monodisperse Mesoporous Silica Nanoparticles J. Am. Chem. Soc. 2012, 134, 5722 5725
    25. 25
      Wang, K.; He, X.; Yang, X.; Shi, H. Functionalized Silica Nanoparticles: A Platform for Fluorescence Imaging at the Cell and Small Animal Levels Acc. Chem. Res. 2013, 46, 1367 1376
    26. 26
      Lee, C. H.; Cheng, S. H.; Wang, Y. J.; Chen, Y. C.; Chen, N. T.; Souris, J.; Chen, C. T.; Mou, C. Y.; Yang, C. S.; Lo, L. W. Near-Infrared Mesoporous Silica Nanoparticles for Optical Imaging: Characterization and in Vivo Biodistribution Adv. Funct. Mater. 2009, 19, 215 222
    27. 27
      Pan, J.; Wan, D.; Gong, J. Pegylated Liposome Coated QDs/Mesoporous Silica Core–Shell Nanoparticles for Molecular Imaging Chem. Commun. 2011, 47, 3442 3444
    28. 28
      Liu, J.; Bu, W.; Zhang, S.; Chen, F.; Xing, H.; Pan, L.; Zhou, L.; Peng, W.; Shi, J. Controlled Synthesis of Uniform and Monodisperse Upconversion Core/Mesoporous Silica Shell Nanocomposites for Bimodal Imaging Chemistry 2012, 18, 2335 2341
    29. 29
      Zhang, Y.; Hong, H.; Orbay, H.; Valdovinos, H. F.; Nayak, T. R.; Theuer, C. P.; Barnhart, T. E.; Cai, W. Pet Imaging of CD105/Endoglin Expression with a (6)(1)/(6)(4)Cu-Labeled Fab Antibody Fragment Eur. J. Nucl. Med. Mol. Imaging 2013, 40, 759 767
    30. 30
      Chen, F.; Hong, H.; Zhang, Y.; Valdovinos, H. F.; Shi, S.; Kwon, G. S.; Theuer, C. P.; Barnhart, T. E.; Cai, W. In Vivo Tumor Targeting and Image-Guided Drug Delivery with Antibody-Conjugated, Radiolabeled Mesoporous Silica Nanoparticles ACS Nano 2013, 7, 9027 9039
    31. 31
      Orbay, H.; Zhang, Y.; Valdovinos, H. F.; Song, G.; Hernandez, R.; Theuer, C. P.; Hacker, T. A.; Nickles, R. J.; Cai, W. Positron Emission Tomography Imaging of CD105 Expression in a Rat Myocardial Infarction Model with (64)Cu-Nota-TRC105 Am. J. Nucl. Med. Mol. Imaging 2013, 4, 1 9
  • Supporting Information

    Supporting Information


    Ninhydrin testing, tables of PET region-of-interest quantifications, quantitative analysis of PET imaging data from non-targeted and blocking groups. This material is available free of charge via the Internet at http://pubs.acs.org.


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.