651
|
Wang Y, de Rochefort L, Liu T, Kressler B. Magnetic source MRI: a new quantitative imaging of magnetic biomarkers. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2010; 2009:53-6. [PMID: 19965112 DOI: 10.1109/iembs.2009.5335128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A new approach to generating MRI contrast by solving the magnetic field to susceptibility source inverse problem is presented to address the quantification difficulties associated with traditional T1/T2 relaxation and susceptibility weighted T2* methods. The forward problem from source to field is reviewed. Its inverse field to source problem is ill posed. Accurate solutions are found by conditioning the data acquisition or regularizing the solution. Preclinical and clinical applications using this magnetic source MRI are discussed for quantitative mapping magnetic biomarkers such as contrast agents in molecular MRI and iron deposits in diseases.
Collapse
Affiliation(s)
- Yi Wang
- Radiology and Biomedical Engineering, Cornell University, New York, NY 10022, USA.
| | | | | | | |
Collapse
|
652
|
18F-FDG PET/CT-related metabolic parameters and their value in early prediction of chemotherapy response in a VX2 tumor model. Nucl Med Biol 2010; 37:327-33. [DOI: 10.1016/j.nucmedbio.2009.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Revised: 11/15/2009] [Accepted: 12/02/2009] [Indexed: 11/22/2022]
|
653
|
AuNP-DG: Deoxyglucose-Labeled Gold Nanoparticles as X-ray Computed Tomography Contrast Agents for Cancer Imaging. Mol Imaging Biol 2010; 12:463-7. [DOI: 10.1007/s11307-010-0299-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
654
|
Jiang L, Kimura RH, Miao Z, Silverman AP, Ren G, Liu H, Li P, Gambhir SS, Cochran JR, Cheng Z. Evaluation of a (64)Cu-labeled cystine-knot peptide based on agouti-related protein for PET of tumors expressing alphavbeta3 integrin. J Nucl Med 2010; 51:251-258. [PMID: 20124048 DOI: 10.2967/jnumed.109.069831] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Recently, a truncated form of the agouti-related protein (AgRP), a 4-kDa cystine-knot peptide of human origin, was used as a scaffold to engineer mutants that bound to alpha(v)beta(3) integrin with high affinity and specificity. In this study, we evaluated the potential of engineered integrin-binding AgRP peptides for use as cancer imaging agents in living subjects. METHODS Engineered AgRP peptides were prepared by solid-phase peptide synthesis and were folded in vitro and purified by reversed-phase high-performance liquid chromatography. Competition assays were used to measure the relative binding affinities of engineered AgRP peptides for integrin receptors expressed on the surface of U87MG glioblastoma cells. The highest-affinity mutant, AgRP clone 7C, was site-specifically conjugated with 1,4,7,10-tetra-azacyclododecane-N,N',N''N'''-tetraacetic acid (DOTA). The resulting bioconjugate, DOTA-AgRP-7C, was radiolabeled with (64)Cu for biodistribution analysis and small-animal PET studies in mice bearing U87MG tumor xenografts. In addition to serum stability, the in vivo metabolic stability of (64)Cu-DOTA-AgRP-7C was assessed after injection and probe recovery from mouse kidney, liver, tumor, and urine. RESULTS AgRP-7C and DOTA-AgRP-7C bound with high affinity to integrin receptors expressed on U87MG cells (half maximal inhibitory concentration values, 20 +/- 4 and 14 +/- 2 nM, respectively). DOTA-AgRP-7C was labeled with (64)Cu with high radiochemical purity (>99%). In biodistribution and small-animal PET studies, (64)Cu-DOTA-AgRP-7C displayed rapid blood clearance, good tumor uptake and retention (2.70 +/- 0.93 percentage injected dose per gram [%ID/g] and 2.37 +/- 1.04 %ID/g at 2 and 24 h, respectively), and high tumor-to-background tissue ratios. The integrin-binding specificity of (64)Cu-DOTA-AgRP-7C was confirmed in vitro and in vivo by showing that a large molar excess of the unlabeled peptidomimetic c(RGDyK) could block probe binding and tumor uptake. Serum stability and in vivo metabolite assays demonstrated that engineered AgRP peptides are sufficiently stable for in vivo molecular imaging applications. CONCLUSION A radiolabeled version of the engineered AgRP peptide 7C showed promise as a PET agent for tumors that express the alpha(v)beta(3) integrin. Collectively, these results validate AgRP-based cystine-knot peptides for use in vivo as molecular imaging agents and provide support for the general use of AgRP as a scaffold to develop targeting peptides, and hence diagnostics, against other tumor receptors.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China.,Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Cancer Center, Bio-X Program, Stanford University, Stanford, California
| | - Richard H Kimura
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Cancer Center, Bio-X Program, Stanford University, Stanford, California
| | - Zheng Miao
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Cancer Center, Bio-X Program, Stanford University, Stanford, California
| | - Adam P Silverman
- Department of Bioengineering, Cancer Center, Bio-X Program, Stanford University, Stanford, California
| | - Gang Ren
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Cancer Center, Bio-X Program, Stanford University, Stanford, California
| | - Hongguang Liu
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Cancer Center, Bio-X Program, Stanford University, Stanford, California
| | - Peiyong Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Sanjiv Sam Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Cancer Center, Bio-X Program, Stanford University, Stanford, California.,Department of Bioengineering, Cancer Center, Bio-X Program, Stanford University, Stanford, California
| | - Jennifer R Cochran
- Department of Bioengineering, Cancer Center, Bio-X Program, Stanford University, Stanford, California
| | - Zhen Cheng
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Cancer Center, Bio-X Program, Stanford University, Stanford, California
| |
Collapse
|
655
|
Abstract
BACKGROUND Optical imaging (OI) techniques such as bioluminescence and fluorescence imaging have been widely used to track diseases in a non-invasive manner within living subjects. These techniques generally require bioluminescent and fluorescent probes. Here we demonstrate the feasibility of using radioactive probes for in vivo molecular OI. METHODOLOGY/PRINCIPAL FINDINGS By taking the advantages of low energy window of light (1.2-3.1 eV, 400-1000 nm) resulting from radiation, radionuclides that emit charged particles such as beta(+) and beta(-) can be successfully imaged with an OI instrument. In vivo optical images can be obtained for several radioactive probes including 2-deoxy-2-[(18)F]fluoro-D-glucose ([(18)F]FDG), Na(18)F, Na(131)I, (90)YCl(3) and a (90)Y labeled peptide that specifically target tumors. CONCLUSIONS/SIGNIFICANCE These studies demonstrate generalizability of radioactive OI technique. It provides a new molecular imaging strategy and will likely have significant impact on both small animal and clinical imaging.
Collapse
|
656
|
Boyer C, Whittaker MR, Chuah K, Liu J, Davis TP. Modulation of the surface charge on polymer-stabilized gold nanoparticles by the application of an external stimulus. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2010; 26:2721-2730. [PMID: 19894684 DOI: 10.1021/la902746v] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
A new approach to controlling the charge on gold nanoparticle surfaces is described. The method exploits the simultaneous coattachment of both charged and neutral polymers onto gold surfaces. The charged and neutral polymers were synthesized by reversible addition-fragmentation chain transfer (RAFT) polymerization, and the RAFT end-group functionality was used as the anchor for attachment to gold. The approach described is general and can be applied to a wide range of monomers; those exemplified in the paper are poly(2-aminoethyl methacrylamide) (P(AEA)), poly(acrylic acid) (PAA), and poly(N,N-diemthylaminoethyl acrylate) (P(DMAEA)) together with neutral polymers based on poly(oligoethylene oxide) acrylate (P(OEG-A)). The hybrid polymer-stabilized GNPs thus formed were characterized in solution using dynamic light scattering and zeta potential measurements, transmission electron microscopy, UV-visible spectroscopy, X-ray photoelectron spectroscopy, and attenuated total reflection-Fourier-transform IR spectroscopy. The grafting densities of the polymers on GNPs were measured using thermal gravimetric analyses (TGA), as 0.4 chains/nm(2) (for PAA), 0.9 chains/nm(2) (for neutral polymers, such as P(NIPAAm), and 0.6 chain/nm(2) for the positive charged polymers P(AEA) and P(DMAEA). The directed coassembly of two different polymers (one charged and one noncharged) on the gold nanoparticle surfaces provided a mechanism (dependent on molecular weight) for shielding the surface charge imparted by the charged polymer component, allowing for a range of surface charges on the GNPs from -30 to +39 mV. In further work, the surface-charges were modulated by an external stimulus (temperature). The charge-modulation was controlled by the use of thermosensitive neutral polymers coassembled with charged polymers. The thermosensitive polymers exemplified in this paper are P(oligoethylene oxide acrylate-co-diethylene oxide acrylate) (P(OEG-A-co-DEG-A)) and P(N-isopropyl acrylamide) (P(NIPAAm). The temperature of the aqueous phase (from 15 to 70 degrees C) was then adjusted to tune the zeta potentials of the hybrid GNPs from +39 or -30 to approximately 0 mV. Finally, by manipulating the solution pH, reversible aggregation behavior of the hybrid GNPs could be induced.
Collapse
Affiliation(s)
- Cyrille Boyer
- Centre for Advanced Macromolecular Design, School of Chemical Sciences and Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | | | | | | | | |
Collapse
|
657
|
Pu KY, Li K, Liu B. Cationic oligofluorene-substituted polyhedral oligomeric silsesquioxane as light-harvesting unimolecular nanoparticle for fluorescence amplification in cellular imaging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2010; 22:643-6. [PMID: 20217765 DOI: 10.1002/adma.200902409] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Affiliation(s)
- Kan-Yi Pu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, Singapore
| | | | | |
Collapse
|
658
|
Chitosan-based systems for molecular imaging. Adv Drug Deliv Rev 2010; 62:42-58. [PMID: 19861142 DOI: 10.1016/j.addr.2009.09.007] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 09/18/2009] [Accepted: 09/29/2009] [Indexed: 11/24/2022]
Abstract
Molecular imaging enables the non-invasive assessment of biological and biochemical processes in living subjects. Such technologies therefore have the potential to enhance our understanding of disease and drug activity during preclinical and clinical drug development. Molecular imaging allows a repetitive and non-invasive study of the same living subject using identical or alternative biological imaging assays at different time points, thus harnessing the statistical power of longitudinal studies, and reducing the number of animals required and cost. Chitosan is a hydrophilic and non-antigenic biopolymer and has a low toxicity toward mammalian cells. Hence, it has great potential as a biomaterial because of its excellent biocompatibility. Conjugated to additional materials, chitosan composites result in a new class of biomaterials that possess mechanical, physicochemical and functional properties, which have potential for use in advanced biomedical imaging applications. The present review will discuss the strengths, limitations and challenges of molecular imaging as well as applications of chitosan nanoparticles in the field of molecular imaging.
Collapse
|
659
|
Tsotsalas MM, Kopka K, Luppi G, Wagner S, Law MP, Schäfers M, De Cola L. Encapsulating (111)In in nanocontainers for scintigraphic imaging: synthesis, characterization, and in vivo biodistribution. ACS NANO 2010; 4:342-348. [PMID: 20020752 DOI: 10.1021/nn901166u] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
A new strategy for the radiolabeling of porous nanocontainers has been developed, and the first experiments in vivo are reported. Our approach consists of the use of nanometer-sized zeolites whose channels have been filled with the positively charged gamma-emitter (111)In(3+) via simple ion exchange. To avoid leaching of the isotope under physiological conditions, the entrances of the channels have been closed using a specifically designed molecular stopcock. This stopcock has a positively charged group that enters the channels and entraps the loaded radionuclides via electrostatic and steric repulsion. The other side of the stopcock is a bulky triethoxysilane group that can covalently bind to the walls of the zeolite entrances, thereby irreversibly closing the channels. The surface of the zeolites has been functionalized with different chemical groups in order to investigate the different biodistributions depending of the nature of the functionalizations. Preliminary in vivo experiments with Wistar rats have been performed and showed the potential of the approach. This strategy leads to a nanoimaging probe with a very high density of radioisotopes in a confined space, which is highly stable in physiological solution and could allow a large variety of functionalities on its external surface.
Collapse
Affiliation(s)
- Manuel M Tsotsalas
- Physikalisches Institut and NRW Graduate School of Chemistry, Westfalische Wilhelms-Universitat Munster, Mendelstrasse 7, D-48149 Munster, Germany
| | | | | | | | | | | | | |
Collapse
|
660
|
Jiang S, Gnanasammandhan MK, Zhang Y. Optical imaging-guided cancer therapy with fluorescent nanoparticles. J R Soc Interface 2010; 7:3-18. [PMID: 19759055 PMCID: PMC2839386 DOI: 10.1098/rsif.2009.0243] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2009] [Accepted: 08/17/2009] [Indexed: 01/01/2023] Open
Abstract
The diagnosis and treatment of cancer have been greatly improved with the recent developments in nanotechnology. One of the promising nanoscale tools for cancer diagnosis is fluorescent nanoparticles (NPs), such as organic dye-doped NPs, quantum dots and upconversion NPs that enable highly sensitive optical imaging of cancer at cellular and animal level. Furthermore, the emerging development of novel multi-functional NPs, which can be conjugated with several functional molecules simultaneously including targeting moieties, therapeutic agents and imaging probes, provides new potentials for clinical therapies and diagnostics and undoubtedly will play a critical role in cancer therapy. In this article, we review the types and characteristics of fluorescent NPs, in vitro and in vivo imaging of cancer using fluorescent NPs and multi-functional NPs for imaging-guided cancer therapy.
Collapse
Affiliation(s)
- Shan Jiang
- Division of Bioengineering, Faculty of Engineering, National University of Singapore, Singapore 117576, Republic of Singapore
| | - Muthu Kumara Gnanasammandhan
- Division of Bioengineering, Faculty of Engineering, National University of Singapore, Singapore 117576, Republic of Singapore
| | - Yong Zhang
- Division of Bioengineering, Faculty of Engineering, National University of Singapore, Singapore 117576, Republic of Singapore
- Nanoscience and Nanotechnology Initiative, National University of Singapore, Singapore 117576, Republic of Singapore
| |
Collapse
|
661
|
Reumann MK, Weiser MC, Mayer-Kuckuk P. Musculoskeletal molecular imaging: a comprehensive overview. Trends Biotechnol 2010; 28:93-101. [PMID: 20045210 DOI: 10.1016/j.tibtech.2009.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 10/26/2009] [Accepted: 11/12/2009] [Indexed: 01/01/2023]
Abstract
Molecular imaging permits non-invasive visualization and measurement of molecular and cell biology in living subjects, thereby complementing conventional anatomical imaging. Herein, we review the emerging application of molecular imaging for the study of musculoskeletal biology. Utilizing mainly bioluminescence and fluorescence techniques, molecular imaging has enabled in-vivo studies of (i) the activity of osteoblasts, osteoclasts, and hormones, (ii) the mechanisms of pathological cartilage and bone destruction, (iii) skeletal gene and cell therapy with and without biomaterial support, and (iv) the cellular processes in osteolysis and osteomyelitis. In these applications, musculoskeletal molecular imaging demonstrated feasibility for research in a myriad of musculoskeletal conditions ranging from bone fracture and arthritis to skeletal cancer. Importantly, these advances herald great potential for innovative clinical imaging in orthopedics, rheumatology, and oncology.
Collapse
Affiliation(s)
- Marie K Reumann
- Bone Cell Biology and Imaging Laboratory, Caspary Research Building, Rm. 623, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
| | | | | |
Collapse
|
662
|
Verma A, Stellacci F. Effect of surface properties on nanoparticle-cell interactions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2010; 6:12-21. [PMID: 19844908 DOI: 10.1002/smll.200901158] [Citation(s) in RCA: 1796] [Impact Index Per Article: 119.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The interaction of nanomaterials with cells and lipid bilayers is critical in many applications such as phototherapy, imaging, and drug/gene delivery. These applications require a firm control over nanoparticle-cell interactions, which are mainly dictated by surface properties of nanoparticles. This critical Review presents an understanding of how synthetic and natural chemical moieties on the nanoparticle surface (in addition to nanoparticle shape and size) impact their interaction with lipid bilayers and cells. Challenges for undertaking a systematic study to elucidate nanoparticle-cell interactions are also discussed.
Collapse
Affiliation(s)
- Ayush Verma
- Department of Materials Science and Engineering, Massachusetts Institute of Technology (MIT), 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | | |
Collapse
|
663
|
Alford R, Ogawa M, Hassan M, Gandjbakhche AH, Choyke PL, Kobayashi H. Fluorescence lifetime imaging of activatable target specific molecular probes. CONTRAST MEDIA & MOLECULAR IMAGING 2010; 5:1-8. [PMID: 20101762 PMCID: PMC3404610 DOI: 10.1002/cmmi.360] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In vivo optical imaging using fluorescently labeled self-quenched monoclonal antibodies, activated through binding and internalization within target cells, results in excellent target-to-background ratios. We hypothesized that these molecular probes could be utilized to accurately report on cellular internalization with fluorescence lifetime imaging (FLI). Two imaging probes were synthesized, consisting of the antibody trastuzumab (targeting HER2/neu) conjugated to Alexa Fluor750 in ratios of either 1:8 or 1:1. Fluorescence intensity and lifetime of each conjugate were initially determined at endosomal pHs. Since the 1:8 conjugate is self-quenched, the fluorescence lifetime of each probe was also determined after exposure to the known dequencher SDS. In vitro imaging experiments were performed using 3T3/HER2(+) and BALB/3T3 (HER2(-)) cell lines. Changes in fluorescence lifetime correlated with temperature- and time-dependent cellular internalization. In vivo imaging studies in mice with dual flank tumors [3T3/HER2(+) and BALB/3T3 (HER2(-))] detected a minimal difference in FLI. In conclusion, fluorescence lifetime imaging monitors the internalization of target-specific activatable antibody-fluorophore conjugates in vitro. Challenges remain in adapting this methodology to in vivo imaging.
Collapse
Affiliation(s)
- Raphael Alford
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
- Case Western Reserve School of Medicine, Cleveland, Ohio, USA
| | - Mikako Ogawa
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Moinuddin Hassan
- Program in Physical Biology, Laboratory of Integrative and Medical Biophysics, National Institute of Child Health and Human Development; National Institutes of Health, Bethesda, Maryland, USA
| | - Amir H. Gandjbakhche
- Program in Physical Biology, Laboratory of Integrative and Medical Biophysics, National Institute of Child Health and Human Development; National Institutes of Health, Bethesda, Maryland, USA
| | - Peter L. Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
664
|
Cooperative nanomaterial system to sensitize, target, and treat tumors. Proc Natl Acad Sci U S A 2009; 107:981-6. [PMID: 20080556 DOI: 10.1073/pnas.0909565107] [Citation(s) in RCA: 226] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A significant barrier to the clinical translation of systemically administered therapeutic nanoparticles is their tendency to be removed from circulation by the mononuclear phagocyte system. The addition of a targeting ligand that selectively interacts with cancer cells can improve the therapeutic efficacy of nanomaterials, although these systems have met with only limited success. Here, we present a cooperative nanosystem consisting of two discrete nanomaterials. The first component is gold nanorod (NR) "activators" that populate the porous tumor vessels and act as photothermal antennas to specify tumor heating via remote near-infrared laser irradiation. We find that local tumor heating accelerates the recruitment of the second component: a targeted nanoparticle consisting of either magnetic nanoworms (NW) or doxorubicin-loaded liposomes (LP). The targeting species employed in this work is a cyclic nine-amino acid peptide LyP-1 (Cys-Gly-Asn-Lys-Arg-Thr-Arg-Gly-Cys) that binds to the stress-related protein, p32, which we find to be upregulated on the surface of tumor-associated cells upon thermal treatment. Mice containing xenografted MDA-MB-435 tumors that are treated with the combined NR/LyP-1LP therapeutic system display significant reductions in tumor volume compared with individual nanoparticles or untargeted cooperative system.
Collapse
|
665
|
Chiral porphyrazine near-IR optical imaging agent exhibiting preferential tumor accumulation. Proc Natl Acad Sci U S A 2009; 107:1284-8. [PMID: 20080563 DOI: 10.1073/pnas.0912811107] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A chiral porphyrazine (pz), H(2)[pz(trans-A(2)B(2))] (247), has been prepared that exhibits preferential in vivo accumulation in the cells of tumors. Pz 247 exhibits near-infrared (NIR) emission with lambda > 700 nm in the required wavelength range for maximum tissue penetration. When MDA-MB-231 breast tumor cells are treated with 247, the agent shows strong intracellular fluorescence with an emission maximum, 704 nm, which indicates that it localizes within a hydrophobic microenvironment. Pz 247 is shown to associate with the lipophilic core of LDL and undergo cellular entry primarily through receptor-mediated endocytosis accumulating in lysosomes. Preliminary in vivo studies show that 247 exhibits preferential accumulation and retention in the cells of MDA-MB-231 tumors subcutaneously implanted in mice, thereby enabling NIR optical imaging with excellent contrast between tumor and surrounding tissue. The intensity of fluorescence from 247 within the tumor increases over time up to 48 h after injection presumably due to the sequestration of circulating 247/LDL complex by the tumor tissue. As the need for cholesterol, and thus LDL, is elevated in highly proliferative tumor cells over nontumorigenic cells, 247 has potential application for all such tumors.
Collapse
|
666
|
Chung E, Yamashita H, Au P, Tannous BA, Fukumura D, Jain RK. Secreted Gaussia luciferase as a biomarker for monitoring tumor progression and treatment response of systemic metastases. PLoS One 2009; 4:e8316. [PMID: 20016816 PMCID: PMC2789383 DOI: 10.1371/journal.pone.0008316] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Accepted: 11/09/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Currently, only few techniques are available for quantifying systemic metastases in preclinical model. Thus techniques that can sensitively detect metastatic colonization and assess treatment response in real-time are urgently needed. To this end, we engineered tumor cells to express a naturally secreted Gaussia luciferase (Gluc), and investigated its use as a circulating biomarker for monitoring viable metastatic or primary tumor growth and their treatment responses. METHODOLOGY/PRINCIPAL FINDINGS We first developed orthotopic primary and metastatic breast tumors with derivative of MDA-MB-231 cells expressing Gluc. We then correlated tumor burden with Gluc activity in the blood and urine along with bioluminescent imaging (BLI). Second, we utilized blood Gluc assay to monitor treatment response to lapatinib in an experimental model of systemic metastasis. We observed good correlation between the primary tumor volume and Gluc concentration in blood (R(2) = 0.84) and urine (R(2) = 0.55) in the breast tumor model. The correlation deviated as a primary tumor grew due to a reduction in viable tumor fraction. This was also supported by our mathematical models for tumor growth to compare the total and viable tumor burden in our model. In the experimental metastasis model, we found numerous brain metastases as well as systemic metastases including bone and lungs. Importantly, blood Gluc assay revealed early growth of metastatic tumors before BLI could visualize their presence. Using secreted Gluc, we localized systemic metastases by BLI and quantitatively monitored the total viable metastatic tumor burden by blood Gluc assay during the course of treatment with lapatinib, a dual tyrosine kinase inhibitor of EGFR and HER2. CONCLUSION/SIGNIFICANCE We demonstrated secreted Gluc assay accurately reflects the amount of viable cancer cells in primary and metastatic tumors. Blood Gluc activity not only tracks metastatic tumor progression but also serves as a longitudinal biomarker for tumor response to treatments.
Collapse
Affiliation(s)
- Euiheon Chung
- Edwin L. Steele Laboratory, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
667
|
Wang K, Wang K, Li W, Huang T, Li R, Wang D, Shen B, Chen X. Characterizing breast cancer xenograft epidermal growth factor receptor expression by using near-infrared optical imaging. Acta Radiol 2009; 50:1095-103. [PMID: 19922304 DOI: 10.3109/02841850903008800] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) overexpression is associated with several key features of cancer development and growth. Therefore, EGFR is a very promising biological target for tumor diagnosis and anticancer therapy. Characterization of EGFR expression is important for clinicians to select patients for EGFR-targeted therapy and evaluate therapeutic effects. PURPOSE To investigate whether near-infrared (NIR) fluorescent dye Cy5.5-labeled anti-EGFR monoclonal antibody Erbitux can characterize EGFR expression level in MDA-MB-231 and MCF-7 breast cancer xenografts using an in vivo NIR imaging method. MATERIAL AND METHODS A fluorochrome probe was designed by coupling Cy5.5 to Erbitux through acidylation, and the fluorescence property of the Erbitux-Cy5.5 conjugate was characterized by fluorospectroscopy. Flow cytometry and laser confocal microscopy were used to test the EGFR specificity of the antibody probe in vitro. Erbitux-Cy5.5 was also injected intravenously into immune-deficient mice bearing MDA-MB-231 or MCF-7 tumors. Whole-body and region-of-interest fluorescence images were acquired and analyzed. The EGFR expression was also analyzed and confirmed by immunohistochemical assay. RESULTS The maximum excitation/emission wavelength for the Erbitux-Cy5.5 probe was 674/697 nm, similar to that of free Cy5.5 (674/712 nm). Confocal microscopy confirmed receptor-specific uptake in MDA-MB-231 and MCF-7 cells. In flow cytometry probe specificity assay, Erbitux-Cy5.5 showed a 9.32-fold higher affinity for MDA-MB-231 than MCF-7 cells. In vivo NIR imaging also indicated specific uptake in EGFR-positive tumors. Probe uptake rate and maximum intake dose in MDA-MB-231 were significantly higher than those in MCF-7 xenografts (P < 0.001). Immunohistochemical staining confirmed the in vivo imaging results, showing differentiated EGFR expression in MDA-MB-231 (+ + +) and MCF-7 (+) tumor tissues. CONCLUSION Erbitux-Cy5.5 may be used as a specific NIR contrast agent for the noninvasive characterization of EGFR expression level in breast cancer xenografts.
Collapse
Affiliation(s)
- Kezheng Wang
- Department of Medical Imaging and Nuclear Medicine, Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Kai Wang
- Department of Medical Imaging and Nuclear Medicine, Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Weihual Li
- Department of Medical Imaging and Nuclear Medicine, Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Tao Huang
- Department of Medical Imaging and Nuclear Medicine, Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Renfei Li
- Department of Medical Imaging and Nuclear Medicine, Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Dan Wang
- Department of Medical Imaging and Nuclear Medicine, Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Baozhong Shen
- Department of Medical Imaging and Nuclear Medicine, Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiaoyuan Chen
- Department of Radiology, Bio-X & Biophysics, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
668
|
Thurber GM, Figueiredo JL, Weissleder R. Multicolor fluorescent intravital live microscopy (FILM) for surgical tumor resection in a mouse xenograft model. PLoS One 2009; 4:e8053. [PMID: 19956597 PMCID: PMC2779447 DOI: 10.1371/journal.pone.0008053] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 10/30/2009] [Indexed: 11/18/2022] Open
Abstract
Background Complete surgical resection of neoplasia remains one of the most efficient tumor therapies. However, malignant cell clusters are often left behind during surgery due to the inability to visualize and differentiate them against host tissue. Here we establish the feasibility of multicolor fluorescent intravital live microscopy (FILM) where multiple cellular and/or unique tissue compartments are stained simultaneously and imaged in real time. Methodology/Principal Findings Theoretical simulations of imaging probe localization were carried out for three agents with specificity for cancer cells, stromal host response, or vascular perfusion. This transport analysis gave insight into the probe pharmacokinetics and tissue distribution, facilitating the experimental design and allowing predictions to be made about the localization of the probes in other animal models and in the clinic. The imaging probes were administered systemically at optimal time points based on the simulations, and the multicolor FILM images obtained in vivo were then compared to conventional pathological sections. Our data show the feasibility of real time in vivo pathology at cellular resolution and molecular specificity with excellent agreement between intravital and traditional in vitro immunohistochemistry. Conclusions/Significance Multicolor FILM is an accurate method for identifying malignant tissue and cells in vivo. The imaging probes distributed in a manner similar to predictions based on transport principles, and these models can be used to design future probes and experiments. FILM can provide critical real time feedback and should be a useful tool for more effective and complete cancer resection.
Collapse
Affiliation(s)
- Greg M. Thurber
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jose L. Figueiredo
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
669
|
Sista AK, Knebel RJ, Tavri S, Johansson M, DeNardo DG, Boddington SE, Kishore SA, Ansari C, Reinhart V, Coakley FV, Coussens LM, Daldrup-Link HE. Optical imaging of the peri-tumoral inflammatory response in breast cancer. J Transl Med 2009; 7:94. [PMID: 19906309 PMCID: PMC2780997 DOI: 10.1186/1479-5876-7-94] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Accepted: 11/11/2009] [Indexed: 11/10/2022] Open
Abstract
PURPOSE Peri-tumoral inflammation is a common tumor response that plays a central role in tumor invasion and metastasis, and inflammatory cell recruitment is essential to this process. The purpose of this study was to determine whether injected fluorescently-labeled monocytes accumulate within murine breast tumors and are visible with optical imaging. MATERIALS AND METHODS Murine monocytes were labeled with the fluorescent dye DiD and subsequently injected intravenously into 6 transgenic MMTV-PymT tumor-bearing mice and 6 FVB/n control mice without tumors. Optical imaging (OI) was performed before and after cell injection. Ratios of post-injection to pre-injection fluorescent signal intensity of the tumors (MMTV-PymT mice) and mammary tissue (FVB/n controls) were calculated and statistically compared. RESULTS MMTV-PymT breast tumors had an average post/pre signal intensity ratio of 1.8+/- 0.2 (range 1.1-2.7). Control mammary tissue had an average post/pre signal intensity ratio of 1.1 +/- 0.1 (range, 0.4 to 1.4). The p-value for the difference between the ratios was less than 0.05. Confocal fluorescence microscopy confirmed the presence of DiD-labeled cells within the breast tumors. CONCLUSION Murine monocytes accumulate at the site of breast cancer development in this transgenic model, providing evidence that peri-tumoral inflammatory cell recruitment can be evaluated non-invasively using optical imaging.
Collapse
Affiliation(s)
- Akhilesh K Sista
- Department of Radiology and Biomedical Engineering, University of California, San Francisco, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
670
|
Optical molecular imaging of epidermal growth factor receptor expression to improve detection of oral neoplasia. Neoplasia 2009; 11:542-51. [PMID: 19484143 DOI: 10.1593/neo.09188] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 03/08/2009] [Accepted: 03/11/2009] [Indexed: 11/18/2022]
Abstract
BACKGROUND The development of noninvasive molecular imaging approaches has the potential to improve management of cancer. METHODS In this study, we demonstrate the potential of noninvasive topical delivery of an epidermal growth factor-Alexa 647 (EGF-Alexa 647) conjugate to image changes in epidermal growth factor receptor expression associated with oral neoplasia. We report a series of preclinical analyses to evaluate the optical contrast achieved after topical delivery of EGF-Alexa 647 in a variety of model systems, including cells, three-dimensional tissue cultures, and intact human tissue specimens using wide-field and high-resolution fluorescence imaging. Data were collected from 17 different oral cancer patients: eight pairs of normal and abnormal biopsies and nine resected tumors were examined. RESULTS The EGF-dye conjugate can be uniformly delivered throughout the oral epithelium with a penetration depth exceeding 500 microm and incubation time of less than 30 minutes. After EGF-Alexa 647 incubation, the presence of oral neoplasia is associated with a 1.5- to 6.9-fold increase in fluorescence contrast compared with grossly normal mucosa from the same patient with both wide-field and high-resolution fluorescence imaging. CONCLUSIONS Results illustrate the potential of EGF-targeted fluorescent agents for in vivo molecular imaging, a technique that may aid in the diagnosis and characterization of oral neoplasia and allow real-time detection of tumor margins.
Collapse
|
671
|
Ratiometric pH-nanosensors based on rhodamine-doped silica nanoparticles functionalized with a naphthalimide derivative. J Colloid Interface Sci 2009; 339:266-70. [DOI: 10.1016/j.jcis.2009.07.044] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Revised: 07/17/2009] [Accepted: 07/17/2009] [Indexed: 11/18/2022]
|
672
|
Nune SK, Gunda P, Thallapally PK, Lin YY, Forrest ML, Berkland CJ. Nanoparticles for biomedical imaging. Expert Opin Drug Deliv 2009; 6:1175-94. [PMID: 19743894 PMCID: PMC3097035 DOI: 10.1517/17425240903229031] [Citation(s) in RCA: 251] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Synthetic nanoparticles are emerging as versatile tools in biomedical applications, particularly in the area of biomedical imaging. Nanoparticles 1 - 100 nm in diameter have dimensions comparable to biological functional units. Diverse surface chemistries, unique magnetic properties, tunable absorption and emission properties, and recent advances in the synthesis and engineering of various nanoparticles suggest their potential as probes for early detection of diseases such as cancer. Surface functionalization has expanded further the potential of nanoparticles as probes for molecular imaging. OBJECTIVE To summarize emerging research of nanoparticles for biomedical imaging with increased selectivity and reduced nonspecific uptake with increased spatial resolution containing stabilizers conjugated with targeting ligands. METHODS This review summarizes recent technological advances in the synthesis of various nanoparticle probes, and surveys methods to improve the targeting of nanoparticles for their application in biomedical imaging. CONCLUSION Structural design of nanomaterials for biomedical imaging continues to expand and diversify. Synthetic methods have aimed to control the size and surface characteristics of nanoparticles to control distribution, half-life and elimination. Although molecular imaging applications using nanoparticles are advancing into clinical applications, challenges such as storage stability and long-term toxicology should continue to be addressed.
Collapse
Affiliation(s)
- Satish K Nune
- Pacific Northwest National Laboratory, 902 Battelle Boulevard, PO Box 999, MSIN K6-81, Richland, WA 99352, USA.
| | | | | | | | | | | |
Collapse
|
673
|
Koh I, Josephson L. Magnetic nanoparticle sensors. SENSORS 2009; 9:8130-45. [PMID: 22408498 PMCID: PMC3292100 DOI: 10.3390/s91008130] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 09/29/2009] [Accepted: 09/30/2009] [Indexed: 01/08/2023]
Abstract
Many types of biosensors employ magnetic nanoparticles (diameter = 5–300 nm) or magnetic particles (diameter = 300–5,000 nm) which have been surface functionalized to recognize specific molecular targets. Here we cover three types of biosensors that employ different biosensing principles, magnetic materials, and instrumentation. The first type consists of magnetic relaxation switch assay-sensors, which are based on the effects magnetic particles exert on water proton relaxation rates. The second type consists of magnetic particle relaxation sensors, which determine the relaxation of the magnetic moment within the magnetic particle. The third type is magnetoresistive sensors, which detect the presence of magnetic particles on the surface of electronic devices that are sensitive to changes in magnetic fields on their surface. Recent improvements in the design of magnetic nanoparticles (and magnetic particles), together with improvements in instrumentation, suggest that magnetic material-based biosensors may become widely used in the future.
Collapse
Affiliation(s)
- Isaac Koh
- T2 Biosystems, 286 Cardinal Medieros Ave, Cambridge, MA 02141, USA; E-Mail:
| | - Lee Josephson
- Center for Translational Nuclear Medicine, Department of Nuclear Medicine and Molecular Imaging and Center for Molecular Imaging Research, Massachusetts General Hospital/Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1 617-726-6478; Fax: +1 617-723-7212
| |
Collapse
|
674
|
Sun L, Guan YS, Pan WM, Luo ZM, Wei JH, Zhao L, Wu H. Clinical value of 18F-FDG PET/CT in assessing suspicious relapse after rectal cancer resection. World J Gastrointest Oncol 2009; 1:55-61. [PMID: 21160775 PMCID: PMC2999093 DOI: 10.4251/wjgo.v1.i1.55] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Revised: 02/17/2009] [Accepted: 02/24/2009] [Indexed: 02/05/2023] Open
Abstract
AIM: To evaluate the value of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) in the restaging of resected rectal cancer.
METHODS: From January 2007 to Sep 2008, 21 patients who had undergone curative surgery resection for rectal carcinoma with suspicious relapse in conventional imaging or clinical findings were retrospectively enrolled in our study. The patients underwent 28 PET/CT scans (two patients had two scans, one patient had three and one had four scans). Locoregional recurrences and/or distant metastases were confirmed by histological analysis or clinical and imaging follow-up.
RESULTS: Final diagnosis was confirmed by histopathological diagnosis in 12 patients (57.1%) and by clinical and imaging follow-up in nine patients (42.9%). Eight patients had extrapelvic metastases with no evidence of pelvic recurrence. Seven patients had both pelvic recurrence and extrapelvic metastases, and two patients had pelvic recurrence only. 18F-FDG PET/CT was negative in two patients and positive in 19 patients. 18F-FDG PET/CT was true positive in 17 patients and false positive in two. The accuracy of 18F-FDG PET/CT was 90.5%, negative predictive value was 100%, and positive predictive value was 89.5%. Five patients with perirectal recurrence underwent 18F-FDG PET/CT image guided tissue core biopsy. 18F-FDG PET/CT also guided surgical resection of pulmonary metastases in three patients and monitored the response to salvage chemotherapy and/or radiotherapy in four patients.
CONCLUSION: 18F-FDG PET/CT is useful for evaluating suspicious locoregional recurrence and distant metastases in the restaging of rectal cancer after curative resection.
Collapse
Affiliation(s)
- Long Sun
- Long Sun, Wei-Min Pan, Zuo-Ming Luo, Ji-Hong Wei, Long Zhao, Hua Wu, Minnan PET Center and Department of Nuclear Medicine, the First Hospital of Xiamen University, Xiamen 316003, Fujian Province, China
| | | | | | | | | | | | | |
Collapse
|
675
|
Effect of surface charge and agglomerate degree of magnetic iron oxide nanoparticles on KB cellular uptake in vitro. Colloids Surf B Biointerfaces 2009; 73:294-301. [DOI: 10.1016/j.colsurfb.2009.05.031] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 02/27/2009] [Accepted: 05/29/2009] [Indexed: 11/23/2022]
|
676
|
Ma LL, Feldman MD, Tam JM, Paranjape AS, Cheruku KK, Larson TA, Tam JO, Ingram DR, Paramita V, Villard JW, Jenkins JT, Wang T, Clarke GD, Asmis R, Sokolov K, Chandrasekar B, Milner TE, Johnston KP. Small multifunctional nanoclusters (nanoroses) for targeted cellular imaging and therapy. ACS NANO 2009; 3:2686-96. [PMID: 19711944 PMCID: PMC2841963 DOI: 10.1021/nn900440e] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The ability of 20-50 nm nanoparticles to target and modulate the biology of specific types of cells will enable major advancements in cellular imaging and therapy in cancer and atherosclerosis. A key challenge is to load an extremely high degree of targeting, imaging, and therapeutic functionality into small, yet stable particles. Herein we report approximately 30 nm stable uniformly sized near-infrared (NIR) active, superparamagnetic nanoclusters formed by kinetically controlled self-assembly of gold-coated iron oxide nanoparticles. The controlled assembly of nanocomposite particles into clusters with small primary particle spacings produces collective responses of the electrons that shift the absorbance into the NIR region. The nanoclusters of approximately 70 iron oxide primary particles with thin gold coatings display intense NIR (700-850 nm) absorbance with a cross section of approximately 10(-14) m(2). Because of the thin gold shells with an average thickness of only 2 nm, the r(2) spin-spin magnetic relaxivity is 219 mM(-1) s(-1), an order of magnitude larger than observed for typical iron oxide particles with thicker gold shells. Despite only 12% by weight polymeric stabilizer, the particle size and NIR absorbance change very little in deionized water over 8 months. High uptake of the nanoclusters by macrophages is facilitated by the dextran coating, producing intense NIR contrast in dark field and hyperspectral microscopy, both in cell culture and an in vivo rabbit model of atherosclerosis. Small nanoclusters with optical, magnetic, and therapeutic functionality, designed by assembly of nanoparticle building blocks, offer broad opportunities for targeted cellular imaging, therapy, and combined imaging and therapy.
Collapse
Affiliation(s)
- Li Leo Ma
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712
- South Texas Veterans Affairs Hospital System, San Antonio, Texas 78229
| | - Marc D. Feldman
- South Texas Veterans Affairs Hospital System, San Antonio, Texas 78229
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
- Address correspondence to: ,
| | - Jasmine M. Tam
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712
| | - Amit S. Paranjape
- Departments of Biomedical Engineering and Imaging Physics, M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Kiran K. Cheruku
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Timothy A. Larson
- Departments of Biomedical Engineering and Imaging Physics, M.D. Anderson Cancer Center, Houston, Texas 77030
- Center for Nano and Molecular Science and Technology, University of Texas at Austin, Austin, Texas 78712
| | - Justina O. Tam
- Departments of Biomedical Engineering and Imaging Physics, M.D. Anderson Cancer Center, Houston, Texas 77030
- Center for Nano and Molecular Science and Technology, University of Texas at Austin, Austin, Texas 78712
| | - Davis R. Ingram
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712
| | - Vidia Paramita
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712
| | - Joseph W. Villard
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - James T. Jenkins
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Tianyi Wang
- Departments of Biomedical Engineering and Imaging Physics, M.D. Anderson Cancer Center, Houston, Texas 77030
| | - Geoffrey D. Clarke
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Reto Asmis
- Office of the Dean, School of Health Professions, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Konstantin Sokolov
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712
- Departments of Biomedical Engineering and Imaging Physics, M.D. Anderson Cancer Center, Houston, Texas 77030
- Center for Nano and Molecular Science and Technology, University of Texas at Austin, Austin, Texas 78712
| | - Bysani Chandrasekar
- South Texas Veterans Affairs Hospital System, San Antonio, Texas 78229
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Thomas E. Milner
- Departments of Biomedical Engineering and Imaging Physics, M.D. Anderson Cancer Center, Houston, Texas 77030
- Center for Nano and Molecular Science and Technology, University of Texas at Austin, Austin, Texas 78712
| | - Keith P. Johnston
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712
- Center for Nano and Molecular Science and Technology, University of Texas at Austin, Austin, Texas 78712
- Address correspondence to: ,
| |
Collapse
|
677
|
Hong H, Sun J, Cai W. Multimodality imaging of nitric oxide and nitric oxide synthases. Free Radic Biol Med 2009; 47:684-98. [PMID: 19524664 DOI: 10.1016/j.freeradbiomed.2009.06.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 05/28/2009] [Accepted: 06/10/2009] [Indexed: 01/27/2023]
Abstract
Nitric oxide (NO) and NO synthases (NOSs) are crucial factors in many pathophysiological processes such as inflammation, vascular/neurological function, and many types of cancer. Noninvasive imaging of NO or NOS can provide new insights in understanding these diseases and facilitate the development of novel therapeutic strategies. In this review, we will summarize the current state-of-the-art multimodality imaging in detecting NO and NOSs, including optical (fluorescence, chemiluminescence, and bioluminescence), electron paramagnetic resonance (EPR), magnetic resonance (MR), and positron emission tomography (PET). With continued effort over the last several years, these noninvasive imaging techniques can now reveal the biodistribution of NO or NOS in living subjects with high fidelity which will greatly facilitate scientists/clinicians in the development of new drugs and/or patient management. Lastly, we will also discuss future directions/applications of NO/NOS imaging. Successful development of novel NO/NOS imaging agents with optimal in vivo stability and desirable pharmacokinetics for clinical translation will enable the maximum benefit in patient management.
Collapse
Affiliation(s)
- Hao Hong
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705-2275, USA
| | | | | |
Collapse
|
678
|
Hall NC, Zhang J, Povoski SP, Martin EW, Knopp MV. New developments in imaging and functional biomarker technology for the assessment and management of cancer patients. Expert Rev Med Devices 2009; 6:347-51. [PMID: 19572788 DOI: 10.1586/erd.09.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
679
|
Dave SR, Gao X. Monodisperse magnetic nanoparticles for biodetection, imaging, and drug delivery: a versatile and evolving technology. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2009; 1:583-609. [DOI: 10.1002/wnan.51] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
680
|
Veiseh O, Sun C, Fang C, Bhattarai N, Gunn J, Kievit F, Du K, Pullar B, Lee D, Ellenbogen RG, Olson J, Zhang M. Specific targeting of brain tumors with an optical/magnetic resonance imaging nanoprobe across the blood-brain barrier. Cancer Res 2009; 69:6200-7. [PMID: 19638572 DOI: 10.1158/0008-5472.can-09-1157] [Citation(s) in RCA: 247] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nanoparticle-based platforms have drawn considerable attention for their potential effect on oncology and other biomedical fields. However, their in vivo application is challenged by insufficient accumulation and retention within tumors due to limited specificity to the target, and an inability to traverse biological barriers. Here, we present a nanoprobe that shows an ability to cross the blood-brain barrier and specifically target brain tumors in a genetically engineered mouse model, as established through in vivo magnetic resonance and biophotonic imaging, and histologic and biodistribution analyses. The nanoprobe is comprised of an iron oxide nanoparticle coated with biocompatible polyethylene glycol-grafted chitosan copolymer, to which a tumor-targeting agent, chlorotoxin, and a near-IR fluorophore are conjugated. The nanoprobe shows an innocuous toxicity profile and sustained retention in tumors. With the versatile affinity of the targeting ligand and the flexible conjugation chemistry for alternative diagnostic and therapeutic agents, this nanoparticle platform can be potentially used for the diagnosis and treatment of a variety of tumor types.
Collapse
Affiliation(s)
- Omid Veiseh
- Departments of Materials Science and Engineering, University of Washington, Seattle, Washington 99195, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
681
|
Boyer C, Whittaker MR, Luzon M, Davis TP. Design and Synthesis of Dual Thermoresponsive and Antifouling Hybrid Polymer/Gold Nanoparticles. Macromolecules 2009. [DOI: 10.1021/ma9013127] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Cyrille Boyer
- Centre for Advanced Macromolecular Design (CAMD), School of Chemical Sciences and Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Michael R. Whittaker
- Centre for Advanced Macromolecular Design (CAMD), School of Chemical Sciences and Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Mario Luzon
- Instituto de Ciencia y Tecnología de Polímeros, C.SIC; C/Juan de la Cierva 3, 28006 Madrid, Spain
| | - Thomas P. Davis
- Centre for Advanced Macromolecular Design (CAMD), School of Chemical Sciences and Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
682
|
Yang L, Peng XH, Wang YA, Wang X, Cao Z, Ni C, Karna P, Zhang X, Wood WC, Gao X, Nie S, Mao H. Receptor-targeted nanoparticles for in vivo imaging of breast cancer. Clin Cancer Res 2009; 15:4722-32. [PMID: 19584158 DOI: 10.1158/1078-0432.ccr-08-3289] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE Cell-surface receptor-targeted magnetic iron oxide nanoparticles provide molecular magnetic resonance imaging contrast agents for improving specificity of the detection of human cancer. EXPERIMENTAL DESIGN The present study reports the development of a novel targeted iron oxide nanoparticle using a recombinant peptide containing the amino-terminal fragment of urokinase-type plasminogen activator (uPA) conjugated to magnetic iron oxide nanoparticles amino-terminal fragment conjugated-iron oxide (ATF-IO). This nanoparticle targets uPA receptor, which is overexpressed in breast cancer tissues. RESULTS ATF-IO nanoparticles are able to specifically bind to and be internalized by uPA receptor-expressing tumor cells. Systemic delivery of ATF-IO nanoparticles into mice bearing s.c. and i.p. mammary tumors leads to the accumulation of the particles in tumors, generating a strong magnetic resonance imaging contrast detectable by a clinical magnetic resonance imaging scanner at a field strength of 3 tesla. Target specificity of ATF-IO nanoparticles showed by in vivo magnetic resonance imaging is further confirmed by near-IR fluorescence imaging of the mammary tumors using near-IR dye-labeled amino-terminal fragment peptides conjugated to iron oxide nanoparticles. Furthermore, mice administered ATF-IO nanoparticles exhibit lower uptake of the particles in the liver and spleen compared with those receiving nontargeted iron oxide nanoparticles. CONCLUSIONS Our results suggest that uPA receptor-targeted ATF-IO nanoparticles have potential as molecularly targeted, dual modality imaging agents for in vivo imaging of breast cancer.
Collapse
Affiliation(s)
- Lily Yang
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, C-4088, 1365 C Clifton Road NE, Atlanta, GA 30322, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
683
|
Unciti-Broceta A, Rahimi Yusop M, Richardson PR, Walton JG, Bradley M. A fluorescein-derived anthocyanidin-inspired pH sensor. Tetrahedron Lett 2009. [DOI: 10.1016/j.tetlet.2009.03.223] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
684
|
Feasibility study of near-infrared fluorescence tomography using a positron emission tomograph equipped with depth-of-interaction PET detectors. Radiol Phys Technol 2009; 2:189-97. [DOI: 10.1007/s12194-009-0065-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Revised: 06/10/2009] [Accepted: 06/10/2009] [Indexed: 11/30/2022]
|
685
|
Wang H, Chen X. Applications for site-directed molecular imaging agents coupled with drug delivery potential. Expert Opin Drug Deliv 2009; 6:745-68. [DOI: 10.1517/17425240902889751] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
686
|
Webster JM, Zhang R, Gambhir SS, Cheng Z, Syud FA. Engineered Two-Helix Small Proteins for Molecular Recognition. Chembiochem 2009; 10:1293-6. [DOI: 10.1002/cbic.200900062] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
687
|
Abstract
Cancer occurs as a result of misregulation of cell growth, which appears to be a consequence of alteration in the function of oncogenes and tumour suppressor genes. Ionising radiation has been used, since the discovery of X-rays in 1896 by Roentgen, both in cancer research and treatment of the disease. The main purpose of cancer research is to understand the molecular alterations involved in the development and progression of the disease in order to improve diagnosis and develop personalised therapies, by focusing on the features of the tumoral cell and the biological events associated to carcinogenesis. Radioisotopic techniques have been used routinely for in vitro research in the molecular and cellular biology of cancer for more than 20 years and are in the process of being substituted by alternative non-radioactive techniques. However in vivo techniques such as irradiation of cells in culture and/or experimental animal models and radioactive labelling are in development, due in part to advances in molecular imaging technologies. The objective of this review is to analyse in an integrative way the applications of ionising radiation in cancer research and therapy. It had been divided into two parts. The first one will approach the techniques applied to cancer research and the second will summarise how ionising radiation is applied to the treatment of neoplastic disease.
Collapse
Affiliation(s)
- M T Macías
- Servicio de Protección Radiológica, Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.
| |
Collapse
|
688
|
Tamerler C, Sarikaya M. Molecular biomimetics: nanotechnology and bionanotechnology using genetically engineered peptides. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2009; 367:1705-1726. [PMID: 19376767 DOI: 10.1098/rsta.2009.0018] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Nature provides inspiration for designing materials and systems that derive their functions from highly organized structures. Biological hard tissues are hybrid materials having inorganics within a complex organic matrix, the molecular scaffold controlling the inorganic structures. Biocomposites incorporate both biomacromolecules such as proteins, lipids and polysaccharides, and inorganic materials, such as hydroxyapatite, silica, magnetite and calcite. The ordered organization of hierarchical structures in organisms begins via the molecular recognition of inorganics by proteins that control interactions and is followed by the highly efficient self-assembly across scales. Following the molecular biological principle, proteins could also be used in controlling materials formation in practical engineering via self-assembled, hybrid, functional materials structures. In molecular biomimetics, material-specific peptides could be the key in the molecular engineering of biology-inspired materials. With the recent developments of nanoscale engineering in physical sciences and the advances in molecular biology, we now combine genetic tools with synthetic nanoscale constructs to create a novel methodology. We first genetically select and/or design peptides with specific binding to functional solids, tailor their binding and assembly characteristics, develop bifunctional peptide/protein genetic constructs with both material binding and biological activity, and use these as molecular synthesizers, erectors and assemblers. Here, we give an overview of solid-binding peptides as novel molecular agents coupling bio- and nanotechnology.
Collapse
Affiliation(s)
- Candan Tamerler
- Genetically Engineered Materials Science and Engineering Center, University of WashingtonSeattle, WA 98195, USA
| | | |
Collapse
|
689
|
Yang L, Mao H, Cao Z, Wang YA, Peng X, Wang X, Sajja HK, Wang L, Duan H, Ni C, Staley CA, Wood WC, Gao X, Nie S. Molecular imaging of pancreatic cancer in an animal model using targeted multifunctional nanoparticles. Gastroenterology 2009; 136:1514-25.e2. [PMID: 19208341 PMCID: PMC3651919 DOI: 10.1053/j.gastro.2009.01.006] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Revised: 12/09/2008] [Accepted: 01/08/2009] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Identification of a ligand/receptor system that enables functionalized nanoparticles to efficiently target pancreatic cancer holds great promise for the development of novel approaches for the detection and treatment of pancreatic cancer. Urokinase plasminogen activator receptor (uPAR), a cellular receptor that is highly expressed in pancreatic cancer and tumor stromal cells, is an excellent surface molecule for receptor-targeted imaging of pancreatic cancer using multifunctional nanoparticles. METHODS The uPAR-targeted dual-modality molecular imaging nanoparticle probe is designed and prepared by conjugating a near-infrared dye-labeled amino-terminal fragment of the receptor binding domain of urokinase plasminogen activator to the surface of functionalized magnetic iron oxide nanoparticles. RESULTS We have shown that the systemic delivery of uPAR-targeted nanoparticles leads to their selective accumulation within tumors of orthotopically xenografted human pancreatic cancer in nude mice. The uPAR-targeted nanoparticle probe binds to and is subsequently internalized by uPAR-expressing tumor cells and tumor-associated stromal cells, which facilitates the intratumoral distribution of the nanoparticles and increases the amount and retention of the nanoparticles in a tumor mass. Imaging properties of the nanoparticles enable in vivo optical and magnetic resonance imaging of uPAR-elevated pancreatic cancer lesions. CONCLUSIONS Targeting uPAR using biodegradable multifunctional nanoparticles allows for the selective delivery of the nanoparticles into primary and metastatic pancreatic cancer lesions. This novel receptor-targeted nanoparticle is a potential molecular imaging agent for the detection of pancreatic cancer.
Collapse
Affiliation(s)
- Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | - Hui Mao
- Department of Radiology, Emory University School of Medicine, Atlanta, Georgia,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Zehong Cao
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | | | - Xianghong Peng
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Xiaoxia Wang
- Department of Radiology, Emory University School of Medicine, Atlanta, Georgia
| | - Hari K. Sajja
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Liya Wang
- Department of Radiology, Emory University School of Medicine, Atlanta, Georgia
| | - Hongwei Duan
- Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Georgia
| | - Chunchun Ni
- Department of Radiology, Emory University School of Medicine, Atlanta, Georgia
| | - Charles A. Staley
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - William C. Wood
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Xiaohu Gao
- Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Georgia
| | - Shuming Nie
- Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Georgia,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
690
|
Kelloff GJ, Sigman CC, Contag CH. Early Detection of Oral Neoplasia: Watching with New Eyes. Cancer Prev Res (Phila) 2009; 2:405-8. [DOI: 10.1158/1940-6207.capr-09-0064] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
691
|
Braband H, Tooyama Y, Fox T, Alberto R. Syntheses of high-valent fac-[99mTcO3]+ complexes and [3+2] cycloadditions with alkenes in water as a direct labelling strategy. Chemistry 2009; 15:633-8. [PMID: 19035614 DOI: 10.1002/chem.200801757] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Reported herein is a new concept for the labelling of biomolecules with small [(99m)TcO(3)](+) complexes through a [3+2] cycloaddition with alkenes for radiopharmaceutical applications. We developed convenient reactions for the synthesis of small, water stable fac-[TcO(3)(tacn-R)](+) complexes ((99)Tc and (99m)Tc, tacn = 1,4,7-triazacyclononane, R = H, -CH(2)-C(6)H(5), -CH(2)-C(6)H(4)COOH). With alkenes, these high valent [(99m)TcO(3)](+) complexes undergo [3+2] cycloaddition with formation of the corresponding Tc(V)-glycolato complexes. The (99m)Tc(V) and (99m)Tc(VII) complexes are stable at 37 degrees C in water and in the presence of serum proteins. Therefore, new opportunities in technetium chemistry are enabled with a high potential for medicinal and biological applications. In contrast to classical labelling, the presented strategy is ligand and not metal-centred.
Collapse
Affiliation(s)
- Henrik Braband
- Institute of Inorganic Chemistry, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | | | | | | |
Collapse
|
692
|
Beretta L. Comparative analysis of the liver and plasma proteomes as a novel and powerful strategy for hepatocellular carcinoma biomarker discovery. Cancer Lett 2009; 286:134-9. [PMID: 19232462 DOI: 10.1016/j.canlet.2009.01.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 01/06/2009] [Indexed: 10/21/2022]
Abstract
The extraordinary developments made in the past decade in proteomic technologies, in particular in mass spectrometry, have enabled investigators to consider designing studies to search for diagnostic and therapeutic biomarkers by scanning complex proteome samples. We developed a method based on extensive fractionation of intact proteins, to comprehensively and quantitatively profile the liver and plasma proteomes in health and disease. We have applied this method to samples collected from patients with early hepatocellular carcinoma (HCC) and from patients with liver cirrhosis as well as to samples collected from three mouse models of HCC. This method allowed for the identification of proteins that differ in expression levels in liver tissue or in plasma with disease progression from liver fibrosis, cirrhosis or steatohepatitis to HCC. The comparative analysis of the liver and plasma proteomes generated from human and mouse specimens, constitutes a novel and powerful strategy for HCC biomarker discovery.
Collapse
Affiliation(s)
- Laura Beretta
- Molecular Diagnostics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center (M5-A864), 1100 Fairview Avenue North Seattle, WA 98109, USA.
| |
Collapse
|
693
|
Chang C, Marszlowicz G, Waldman Z, Li P, Snook AE, Lin JE, Schulz S, Waldman SA. Guanylyl cyclase C as a biomarker for targeted imaging and therapy of metastatic colorectal cancer. Biomark Med 2009; 3:33-45. [DOI: 10.2217/17520363.3.1.33] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The guanylyl cyclase C (GCC) receptor posseses several well-established properties ideal for use as a biomarker in gastrointestinal malignancies. The GCC receptor is constitutively expressed in the apical membranes of the intestine and its expression is universally preserved in primary colorectal tumors and their metastases. Moreover, receptor binding is retained by GCC’s cognate ligand, the bacterial enterotoxin ST, even after conjugation to functional moieties. Selective tumor, but not gastrointestinal, uptake of ST in mice bearing GCC-expressing colon cancer xenografts demonstrates the potential of exploiting ST–GCC interaction for diagnostic imaging and targeted therapy of metastatic colorectal cancer. We expect this specific targeting provided by ST–GCC interaction to improve diagnosis, staging and management of colorectal cancer metastases, and ultimately prolong patient survival in this disease.
Collapse
Affiliation(s)
- Chang Chang
- School of Biomedical Engineering, Science & Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Glen Marszlowicz
- School of Biomedical Engineering, Science & Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Zac Waldman
- Department of Physics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Peng Li
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam E Snook
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jieru E Lin
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Stephanie Schulz
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Scott A Waldman
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
694
|
Veiseh O, Gunn JW, Kievit FM, Sun C, Fang C, Lee JSH, Zhang M. Inhibition of tumor-cell invasion with chlorotoxin-bound superparamagnetic nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2009; 5:256-64. [PMID: 19089837 PMCID: PMC2692352 DOI: 10.1002/smll.200800646] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Nanoparticles have been investigated as drug delivery vehicles, contrast agents, and multifunctional devices for patient care. Current nanoparticle-based therapeutic strategies for cancer treatment are mainly based on delivery of chemotherapeutic agents to induce apoptosis or DNA/siRNA to regulate oncogene expression. Here, a nanoparticle system that demonstrates an alternative approach to the treatment of cancers through the inhibition of cell invasion, while serving as a magnetic resonance and optical imaging contrast agent, is presented. The nanoparticle comprises an iron oxide nanoparticle core conjugated with an amine-functionalized poly(ethylene glycol) silane and a small peptide, chlorotoxin (CTX), which enables the tumor cell-specific binding of the nanoparticle. It is shown that the nanoparticle exhibits substantially enhanced cellular uptake and an invasion inhibition rate of approximately 98% compared to unbound CTX ( approximately 45%). Significantly, the investigation from flow cytometry analysis, transmission electron microscopy, and fluorescent imaging reveals that the CTX-enabled nanoparticles deactivated the membrane-bound matrix metalloproteinase 2 (MMP-2) and induced increased internalization of lipid rafts that contain surface-expressed MMP-2 and volume-regulating ion channels through receptor-mediated endocytosis, leading to enhanced prohibitory effects. Since upregulation and activity of MMP-2 have been observed in tumors of neuroectodermal origin, and in cancers of the breast, colon, skin, lung, prostate, ovaries, and a host of others, this nanoparticle system can be potentially used for non-invasive diagnosis and treatment of a variety of cancer types.
Collapse
Affiliation(s)
- Omid Veiseh
- Department of Materials Science and Engineering, University of Washington Seattle, WA 98195, USA
| | | | | | | | | | | | | |
Collapse
|
695
|
Lopalco M, Koini EN, Cho JK, Bradley M. Catch and release microwave mediated synthesis of cyanine dyes. Org Biomol Chem 2009; 7:856-9. [PMID: 19225665 DOI: 10.1039/b820719b] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Unsymmetrical functionalised cyanine dyes, covering the whole colour range, were readily synthesised (in 100 mg amounts) by a combination of microwave and solid-phase methodologies.
Collapse
Affiliation(s)
- Maria Lopalco
- School of Chemistry, The University of Edinburgh, West Mains Road, Edinburgh EH93JJ, United Kingdom
| | | | | | | |
Collapse
|
696
|
Ge Y, Zhang Y, He S, Nie F, Teng G, Gu N. Fluorescence Modified Chitosan-Coated Magnetic Nanoparticles for High-Efficient Cellular Imaging. NANOSCALE RESEARCH LETTERS 2009; 4:287-295. [PMID: 20596545 PMCID: PMC2893437 DOI: 10.1007/s11671-008-9239-9] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Accepted: 12/30/2008] [Indexed: 05/21/2023]
Abstract
Labeling of cells with nanoparticles for living detection is of interest to various biomedical applications. In this study, novel fluorescent/magnetic nanoparticles were prepared and used in high-efficient cellular imaging. The nanoparticles coated with the modified chitosan possessed a magnetic oxide core and a covalently attached fluorescent dye. We evaluated the feasibility and efficiency in labeling cancer cells (SMMC-7721) with the nanoparticles. The nanoparticles exhibited a high affinity to cells, which was demonstrated by flow cytometry and magnetic resonance imaging. The results showed that cell-labeling efficiency of the nanoparticles was dependent on the incubation time and nanoparticles' concentration. The minimum detected number of labeled cells was around 10(4) by using a clinical 1.5-T MRI imager. Fluorescence and transmission electron microscopy instruments were used to monitor the localization patterns of the magnetic nanoparticles in cells. These new magneto-fluorescent nanoagents have demonstrated the potential for future medical use.
Collapse
Affiliation(s)
- Yuqing Ge
- Department of Biological Science and Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, State Key Laboratory of Bioelectronics, Nanjing, People’s Republic of China
| | - Yu Zhang
- Department of Biological Science and Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, State Key Laboratory of Bioelectronics, Nanjing, People’s Republic of China
| | - Shiying He
- Department of Biological Science and Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, State Key Laboratory of Bioelectronics, Nanjing, People’s Republic of China
| | - Fang Nie
- Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, 210096, People’s Republic of China
| | - Gaojun Teng
- Department of Radiology, Zhongda Hospital, Southeast University, Nanjing, 210096, People’s Republic of China
| | - Ning Gu
- Department of Biological Science and Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, State Key Laboratory of Bioelectronics, Nanjing, People’s Republic of China
| |
Collapse
|
697
|
Abstract
Our understanding of the intricate inflammation biology underlying atherosclerosis is rapidly progressing. Molecular imaging strategies, harnessing this body of knowledge, have been developed to visualize some key cellular and molecular events in plaque evolution and vulnerability. Here, we discuss recent advances in magnetic resonance and fluorescence imaging of key biomarkers including adhesion molecules, inflammatory cells, and enzyme activity. We discuss strengths and limitations of respective imaging technologies, and comment on the potential of multi-modality imaging approaches.
Collapse
|
698
|
Sokolov K, Tam J, Tam J, Travis K, Larson T, Aaron J, Harrison N, Emelianov S, Johnston K. Cancer imaging and therapy with metal nanoparticles. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2009; 2009:2005-7. [PMID: 19964034 DOI: 10.1109/iembs.2009.5333430] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nanotechnology offers unique opportunities for cancer detection, therapy and the ability to monitor therapeutic interventions. This potential has to be analyzed in context of challenges that need to be overcome in translation of nanoparticles to clinical applications including specific delivery in tissues and clearance from the body. Here, we will present a case study of plasmonic nanoparticles in cancer imaging and therapy.
Collapse
Affiliation(s)
- Konstantin Sokolov
- Department of Imaging Physics, The UT M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
699
|
Park C, Youn H, Kim H, Noh T, Kook YH, Oh ET, Park HJ, Kim C. Cyclodextrin-covered gold nanoparticles for targeted delivery of an anti-cancer drug. ACTA ACUST UNITED AC 2009. [DOI: 10.1039/b816209c] [Citation(s) in RCA: 151] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
700
|
Wang AZ, Bagalkot V, Vasilliou CC, Gu F, Alexis F, Zhang L, Shaikh M, Yuet K, Cima MJ, Langer R, Kantoff PW, Bander NH, Jon S, Farokhzad OC. Superparamagnetic iron oxide nanoparticle-aptamer bioconjugates for combined prostate cancer imaging and therapy. ChemMedChem 2008; 3:1311-5. [PMID: 18613203 DOI: 10.1002/cmdc.200800091] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Andrew Z Wang
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesia, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|