1601
|
Ricciardelli C, Oehler MK. Diverse molecular pathways in ovarian cancer and their clinical significance. Maturitas 2009; 62:270-5. [DOI: 10.1016/j.maturitas.2009.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2008] [Accepted: 01/01/2009] [Indexed: 12/27/2022]
|
1602
|
Al-Ejeh F, Darby JM, Tsopelas C, Smyth D, Manavis J, Brown MP. APOMAB, a La-specific monoclonal antibody, detects the apoptotic tumor response to life-prolonging and DNA-damaging chemotherapy. PLoS One 2009; 4:e4558. [PMID: 19247492 PMCID: PMC2645692 DOI: 10.1371/journal.pone.0004558] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 01/16/2009] [Indexed: 12/16/2022] Open
Abstract
Background Antineoplastic therapy may impair the survival of malignant cells to produce cell death. Consequently, direct measurement of tumor cell death in vivo is a highly desirable component of therapy response monitoring. We have previously shown that APOMAB® representing the DAB4 clone of a La/SSB-specific murine monoclonal autoantibody is a malignant cell-death ligand, which accumulates preferentially in tumors in an antigen-specific and dose-dependent manner after DNA-damaging chemotherapy. Here, we aim to image tumor uptake of APOMAB® (DAB4) and to define its biological correlates. Methodology/Principal Findings Brisk tumor cell apoptosis is induced in the syngeneic EL4 lymphoma model after treatment of tumor-bearing mice with DNA-damaging cyclophosphamide/etoposide chemotherapy. Tumor and normal organ accumulation of Indium 111 (111In)-labeled La-specific DAB4 mAb as whole IgG or IgG fragments was quantified by whole-body static imaging and organ assay in tumor-bearing mice. Immunohistochemical measurements of tumor caspase-3 activation and PARP-1 cleavage, which are indicators of early and late apoptosis, respectively, were correlated with tumor accumulation of DAB4. Increased tumor accumulation of DAB4 was associated directly with both the extent of chemotherapy-induced tumor cell death and DAB4 binding per dead tumor cell. Tumor DAB4 accumulation correlated with cumulative caspase-3 activation and PARP-1 cleavage as tumor biomarkers of apoptosis and was directly related to the extended median survival time of tumor-bearing mice. Conclusions/Significance Radiolabeled La-specific monoclonal antibody, DAB4, detected dead tumor cells after chemotherapy, rather than chemosensitive normal tissues of gut and bone marrow. DAB4 identified late apoptotic tumor cells in vivo. Hence, radiolabeled DAB4 may usefully image responses to human carcinoma therapy because DAB4 would capture the protracted cell death of carcinoma. We believe that the ability of radiolabeled DAB4 to rapidly assess the apoptotic tumor response and, consequently, to potentially predict extended survival justifies its future clinical development as a radioimmunoscintigraphic agent. This article is part I of a two-part series providing proof-of-concept for the the diagnostic and therapeutic use of a La-specific monoclonal antibody, the DAB4 clone of which is represented by the registered trademark, APOMAB®.
Collapse
Affiliation(s)
- Fares Al-Ejeh
- Experimental Therapeutics Laboratory, Hanson Institute, Adelaide, South Australia, Australia
| | - Jocelyn M. Darby
- Experimental Therapeutics Laboratory, Hanson Institute, Adelaide, South Australia, Australia
| | - Chris Tsopelas
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Douglas Smyth
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Jim Manavis
- Centre for Neurological Disease, Hanson Institute, Adelaide, South Australia, Australia
| | - Michael P. Brown
- Experimental Therapeutics Laboratory, Hanson Institute, Adelaide, South Australia, Australia
- Department of Medical Oncology, Royal Adelaide Hospital Cancer Centre and School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- * E-mail:
| |
Collapse
|
1603
|
Srinivasan S, Carpenter C, Pogue BW, Paulsen KD. Image-guided near infrared spectroscopy using boundary element method: phantom validation. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2009; 7171:717103. [PMID: 20445830 PMCID: PMC2863336 DOI: 10.1117/12.808938] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Image-guided near infrared spectroscopy (IG-NIRS) can provide high-resolution vascular, metabolic and molecular characterization of localized tissue volumes in-vivo. The approach for IG-NIRS uses hybrid systems where the spatial anatomical structure of tissue obtained from standard imaging modalities (such as MRI) is combined with tissue information from diffuse optical imaging spectroscopy. There is need to optimize these hybrid systems for large-scale clinical trials anticipated in the near future in order to evaluate the feasibility of this technology across a larger population. However, existing computational methods such as the finite element method mesh arbitrary image volumes, which inhibit automation, especially with large numbers of datasets. Circumventing this issue, a boundary element method (BEM) for IG-NIRS systems in 3-D is presented here using only surface rendering and discretization. The process of surface creation and meshing is faster, more reliable, and is easily generated automatically as compared to full volume meshing. The proposed method has been implemented here for multi-spectral non-invasive characterization of tissue. In phantom experiments, 3-D spectral BEM-based spectroscopy recovered the oxygen dissociation curve with mean error of 6.6% and tracked variation in total hemoglobin linearly.
Collapse
Affiliation(s)
- Subhadra Srinivasan
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH-03755
| | - Colin Carpenter
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH-03755
| | - Brian W. Pogue
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH-03755
| | - Keith D. Paulsen
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH-03755
| |
Collapse
|
1604
|
Khemtong C, Kessinger CW, Ren J, Bey EA, Yang SG, Guthi JS, Boothman DA, Sherry AD, Gao J. In vivo off-resonance saturation magnetic resonance imaging of alphavbeta3-targeted superparamagnetic nanoparticles. Cancer Res 2009; 69:1651-8. [PMID: 19190328 PMCID: PMC2821739 DOI: 10.1158/0008-5472.can-08-3231] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Magnetic resonance imaging is a powerful clinical imaging technique that allows for noninvasive tomographic visualization of anatomic structures with high spatial resolution and soft tissue contrast. However, its application in molecular imaging of cancer has been limited by the lack of sensitivity and detection accuracy in depicting the biochemical expression of these diseases. Here, we combine an ultrasensitive design of superparamagnetic polymeric micelles (SPPM) and an off-resonance saturation (ORS) method to enhance the imaging efficacy of tumor biomarkers in vivo. SPPM nanoparticles encoded with cyclic(RGDfK) were able to target the alpha(v)beta(3)-expressing microvasculature in A549 non-small cell lung tumor xenografts in mice. ORS greatly improved tumor detection accuracy over the conventional T(2)*-weighted method by its ability to turn "ON" the contrast of SPPM. This combination of ORS imaging with a tumor vasculature-targeted, ultrasensitive SPPM design offers new opportunities in molecular imaging of cancer.
Collapse
Affiliation(s)
- Chalermchai Khemtong
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chase W. Kessinger
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jimin Ren
- The Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Erik A. Bey
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Su-Geun Yang
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jagadeesh Setti Guthi
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - David A. Boothman
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - A. Dean Sherry
- The Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jinming Gao
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
1605
|
Kee HL, Diers JR, Ptaszek M, Muthiah C, Fan D, Lindsey JS, Bocian DF, Holten D. Chlorin-bacteriochlorin energy-transfer dyads as prototypes for near-infrared molecular imaging probes: controlling charge-transfer and fluorescence properties in polar media. Photochem Photobiol 2009; 85:909-20. [PMID: 19222800 DOI: 10.1111/j.1751-1097.2008.00532.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The photophysical properties of two energy-transfer dyads that are potential candidates for near-infrared (NIR) imaging probes are investigated as a function of solvent polarity. The dyads (FbC-FbB and ZnC-FbB) contain either a free base (Fb) or zinc (Zn) chlorin (C) as the energy donor and a free base bacteriochlorin (B) as the energy acceptor. The dyads were studied in toluene, chlorobenzene, 1,2-dichlorobenzene, acetone, acetonitrile and dimethylsulfoxide (DMSO). In both dyads, energy transfer from the chlorin to bacteriochlorin occurs with a rate constant of approximately (5-10 ps)(-1) and a yield of >99% in nonpolar and polar media. In toluene, the fluorescence yields (Phif=0.19) and singlet excited-state lifetimes (tau approximately 5.5 ns) are comparable to those of the benchmark bacteriochlorin. The fluorescence yield and excited-state lifetime decrease as the solvent polarity increases, with quenching by intramolecular electron (or hole) transfer being greater for FbC-FbB than for ZnC-FbB in a given solvent. For example, the Phif and tau values for FbC-FbB in acetone are 0.055 and 1.5 ns and in DMSO are 0.019 and 0.28 ns, whereas those for ZnC-FbB in acetone are 0.12 and 4.5 ns and in DMSO are 0.072 and 2.4 ns. The difference in fluorescence properties of the two dyads in a given polar solvent is due to the relative energies of the lowest energy charge-transfer states, as assessed by ground-state redox potentials and supported by molecular-orbital energies derived from density functional theory calculations. Controlling the extent of excited-state quenching in polar media will allow the favorable photophysical properties of the chlorin-bacteriochlorin dyads to be exploited in vivo. These properties include very large Stokes shifts (85 nm for FbC-FbB, 110 nm for ZnC-FbB) between the red-region absorption of the chlorin and the NIR fluorescence of the bacteriochlorin (lambdaf=760 nm), long bacteriochlorin excited-state lifetime (approximately 5.5 ns), and narrow (<or=20 nm) absorption and fluorescence bands. The latter will facilitate selective excitation/detection and multiprobe applications using both intensity- and lifetime-imaging techniques.
Collapse
Affiliation(s)
- Hooi Ling Kee
- Department of Chemistry, Washington University, St. Louis, MO, USA
| | | | | | | | | | | | | | | |
Collapse
|
1606
|
Chang YC, Ye JY, Thomas TP, Cao Z, Kotlyar A, Tkaczyk ER, Baker JR, Norris TB. Two-photon in vivo flow cytometry using a fiber probe. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2009; 7173:71730I1-71730I10. [PMID: 27182102 DOI: 10.1117/12.808436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We have demonstrated the use of a double-clad fiber probe to conduct two-photon excited flow cytometry in vitro and in vivo. We conducted two-channel detection to measure fluorescence at two distinct wavelengths simultaneously. Because the scattering and absorption problems from whole blood were circumvented by the fiber probe, the detected signal strength from the cells were found to be similar in PBS and in whole blood. We achieved the same detection efficiency of the membrane-binding lipophilic dye DiD labeled cells in PBS and in whole blood. High detection efficiency of green fluorescent protein (GFP)-expressing cells in whole blood was demonstrated. DiD-labeled untransfected and GFP-transfected cells were injected into live mice and the circulation dynamics of the externally injected cells were monitored. The detection efficiency of GFP-expressing cells in vivo was consistent with that observed in whole blood.
Collapse
Affiliation(s)
- Yu-Chung Chang
- Center for Ultrafast Optical Science, University of Michigan, Ann Arbor, MI 48109
| | - Jing Yong Ye
- Center for Ultrafast Optical Science, University of Michigan, Ann Arbor, MI 48109; Michigan Nanotechnology Institute for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Thommey P Thomas
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Zhengyi Cao
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Alina Kotlyar
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Eric R Tkaczyk
- Center for Ultrafast Optical Science, University of Michigan, Ann Arbor, MI 48109; Michigan Nanotechnology Institute for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - James R Baker
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Theodore B Norris
- Center for Ultrafast Optical Science, University of Michigan, Ann Arbor, MI 48109; Michigan Nanotechnology Institute for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
1607
|
Kelly KA, Shaw SY, Nahrendorf M, Kristoff K, Aikawa E, Schreiber SL, Clemons PA, Weissleder R. Unbiased discovery of in vivo imaging probes through in vitro profiling of nanoparticle libraries. Integr Biol (Camb) 2009; 1:311-7. [PMID: 20023731 DOI: 10.1039/b821775k] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In vivo imaging reveals how proteins and cells function as part of complex regulatory networks in intact organisms, and thereby contributes to a systems-level understanding of biological processes. However, the development of novel in vivo imaging probes remains challenging. Most probes are directed against a limited number of pre-specified protein targets; cell-based screens for imaging probes have shown promise, but raise concerns over whether in vitro surrogate cell models recapitulate in vivo phenotypes. Here, we rapidly profile the in vitro binding of nanoparticle imaging probes in multiple samples of defined target vs. background cell types, using primary cell isolates. This approach selects for nanoparticles that show desired targeting effects across all tested members of a class of cells, and decreases the likelihood that an idiosyncratic cell line will unduly skew screening results. To adjust for multiple hypothesis testing, we use permutation methods to identify nanoparticles that best differentiate between the target and background cell classes. (This approach is conceptually analogous to one used for high-dimensionality datasets of genome-wide gene expression, e.g. to identify gene expression signatures that discriminate subclasses of cancer.) We apply this approach to the identification of nanoparticle imaging probes that bind endothelial cells, and validate our in vitro findings in human arterial samples, and by in vivo intravital microscopy in mice. Overall, this work presents a generalizable approach to the unbiased discovery of in vivo imaging probes, and may guide the further development of novel endothelial imaging probes.
Collapse
Affiliation(s)
- Kimberly A Kelly
- Center for Molecular Imaging, Massachusetts General Hospital, Harvard Medical School, 149 13th St., Rm 5420, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | |
Collapse
|
1608
|
Eliceiri KW, Contag CH. Integrated studies of biology: multiplexed imaging assays from molecules to man and back. Curr Opin Biotechnol 2009; 20:1-3. [DOI: 10.1016/j.copbio.2009.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
1609
|
Schellenberger E, Rudloff F, Warmuth C, Taupitz M, Hamm B, Schnorr J. Protease-specific nanosensors for magnetic resonance imaging. Bioconjug Chem 2009; 19:2440-5. [PMID: 19007261 DOI: 10.1021/bc800330k] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Imaging of enzyme activity is a central goal of molecular imaging. With the introduction of fluorescent smart probes, optical imaging has become the modality of choice for experimental in vivo detection of enzyme activity. Here, we present a novel high-relaxivity nanosensor that is suitable for in vivo imaging of protease activity by magnetic resonance imaging. Upon specific protease cleavage, the nanoparticles rapidly switch from a stable low-relaxivity stealth state to become adhesive, aggregating high-relaxivity particles. To demonstrate the principle, we chose a cleavage motif of matrix metalloproteinase 9 (MMP-9), an enzyme important in inflammation, atherosclerosis, tumor progression, and many other diseases with alterations of the extracellular matrix. On the basis of clinically tested very small iron oxide particles (VSOP), the MMP-9-activatable protease-specific iron oxide particles (PSOP) have a hydrodynamic diameter of only 25 nm. PSOP are rapidly activated, resulting in aggregation and increased T2*-relaxivity.
Collapse
Affiliation(s)
- Eyk Schellenberger
- Department of Radiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
1610
|
Abstract
PURPOSE OF REVIEW Tumors recruit various immune cells with seemingly contrasting functions. Yet, the precise role of these cells in situ remains vastly unknown. This review presents a new discovery effort that employs intravital imaging to study immune players directly in tissues. RECENT FINDINGS Cytotoxic T lymphocytes (CTLs) that recognize cognate antigenic peptide can infiltrate tumors from the periphery to the center, and physically engage and eliminate antigen-presenting tumor cells. Nevertheless, the reported kinetics for tumor cell killing by CTLs in vivo is surprisingly low as it takes several hours for one CTL to eliminate one tumor cell. Also, T regulatory (Treg) cells can create a suppressive milieu that restricts the release of CTL cytotoxic granules, which protects tumor cells from being killed. CTLs may be further subverted during lengthy interactions with tumor-associated macrophages (TAMs). Finally, TAMs can directly facilitate tumor invasion by recruiting tumor cells nearby vessels and promoting their intravasation. SUMMARY Intravital imaging has started to uncover tumor-related immune events as they unfold in vivo. The technology should be exploited in the coming years to dissect further the tumor microenvironment and to define therapeutics that augment antitumor immunity.
Collapse
Affiliation(s)
- Mikael J Pittet
- Center for Systems Biology and Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA.
| |
Collapse
|
1611
|
Kobayashi H, Kosaka N, Ogawa M, Morgan NY, Smith PD, Murray CB, Ye X, Collins J, Kumar GA, Bell H, Choyke PL. In vivo multiple color lymphatic imaging using upconverting nanocrystals. ACTA ACUST UNITED AC 2009. [DOI: 10.1039/b910512c] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
1612
|
Stefania R, Tei L, Barge A, Geninatti Crich S, Szabo I, Cabella C, Cravotto G, Aime S. Tuning Glutamine Binding Modes in Gd-DOTA-Based Probes for an Improved MRI Visualization of Tumor Cells. Chemistry 2009; 15:76-85. [DOI: 10.1002/chem.200801567] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
1613
|
Fang C, Zhang M. Multifunctional Magnetic Nanoparticles for Medical Imaging Applications. JOURNAL OF MATERIALS CHEMISTRY 2009; 19:6258-6266. [PMID: 20593005 PMCID: PMC2893338 DOI: 10.1039/b902182e] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Magnetic nanoparticles (MNPs) have attracted enormous research attention due to their unique magnetic properties that enable the detection by the non-invasive medical imaging modality-magnetic resonance imaging (MRI). By incorporating advanced features, such as specific targeting, multimodality, therapeutic delivery, the detectability and applicability of MNPs have been dramatically expanded. A delicate design on structure, composition and surface chemistry is essential to achieving desired properties in MNP systems, such as high imaging contrast and chemical stability, non-fouling surface, target specificity and/or multimodality. This article presents the design fundamentals on the development of MNP systems, from discussion of material selection for nanoparticle cores and coatings, strategies for chemical synthesis and surface modification and their merits and limitations, to conjugation of special biomolecules for intended functions, and reviews the recent advances in the field.
Collapse
Affiliation(s)
- Chen Fang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
1614
|
Tavri S, Jha P, Meier R, Henning TD, Müller T, Hostetter D, Knopp C, Johansson M, Reinhart V, Boddington S, Sista A, Wels WS, Daldrup-Link HE. Optical Imaging of Cellular Immunotherapy against Prostate Cancer. Mol Imaging 2009. [DOI: 10.2310/7290.2009.00002] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
1615
|
Kosaka N, Ogawa M, Longmire MR, Choyke PL, Kobayashi H. Multi-targeted multi-color in vivo optical imaging in a model of disseminated peritoneal ovarian cancer. JOURNAL OF BIOMEDICAL OPTICS 2009; 14:014023. [PMID: 19256711 PMCID: PMC2788995 DOI: 10.1117/1.3083449] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Commonly used in flow cytometry, multiplexed optical probes can diagnose multiple types of cell surface marker, potentially leading to improved diagnosis accuracy in vivo. Herein, we demonstrate the targeting of two different tumor markers in models of disseminated ovarian cancer. Two ovarian cancer cell lines (SKOV3 and SHIN3) were employed; both overexpress D-galactose receptor (D-galR), but only SKOV3 overexpresses HER2/neu. Additionally, fusion tumors composed of SKOV3 and SHIN3/RFP were evaluated. Both galactosyl serum albumin-rhodamine green (GSA-RhodG), which binds D-galR, and trastuzumab-Alexa680, which binds HER2/neu, were administered to tumor-bearing mice for in vivo fluorescence imaging and in situ fluorescence microscopy. In vivo fluorescence imaging depicted 64 of 69 SKOV3 tumors (94.2%) based on their dual spectra corresponding to both RhodG and Alexa680, while all 71 SHIN3 tumors (100%) were detected based on their single spectrum corresponding only to RhodG. All 59 SHIN3 and 36 SKOV3 tumors were correctly diagnosed with in situ microscopy. Additionally, in the mixed tumor model, all tumors could be depicted using the RhodG spectrum, but only SKOV3 components also showed the Alexa680 spectrum. In conclusion, multitargeted multicolor optical imaging enabled specific in vivo diagnosis of tumors expressing distinct patterns of receptors, leading to improved diagnostic accuracy.
Collapse
Affiliation(s)
- Nobuyuki Kosaka
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Dr., Bethesda, MD 20892-1088, USA
| | - Mikako Ogawa
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Dr., Bethesda, MD 20892-1088, USA
| | - Michelle R. Longmire
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Dr., Bethesda, MD 20892-1088, USA
| | - Peter L. Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Dr., Bethesda, MD 20892-1088, USA
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Dr., Bethesda, MD 20892-1088, USA
| |
Collapse
|
1616
|
Controlled Growth of Nanoparticle Clusters through Competitive Stabilizer Desorption. Angew Chem Int Ed Engl 2008; 48:175-8. [DOI: 10.1002/anie.200803895] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
1617
|
Stolarczyk J, Ghosh S, Brougham D. Controlled Growth of Nanoparticle Clusters through Competitive Stabilizer Desorption. Angew Chem Int Ed Engl 2008. [DOI: 10.1002/ange.200803895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
1618
|
Van Beers BE, Vilgrain V. Biomarkers in abdominal imaging. ACTA ACUST UNITED AC 2008; 34:663-7. [DOI: 10.1007/s00261-008-9480-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Accepted: 10/31/2008] [Indexed: 12/22/2022]
|
1619
|
|
1620
|
Cortez-Retamozo V, Swirski FK, Waterman P, Yuan H, Figueiredo JL, Newton AP, Upadhyay R, Vinegoni C, Kohler R, Blois J, Smith A, Nahrendorf M, Josephson L, Weissleder R, Pittet MJ. Real-time assessment of inflammation and treatment response in a mouse model of allergic airway inflammation. J Clin Invest 2008; 118:4058-66. [PMID: 19033674 PMCID: PMC2579705 DOI: 10.1172/jci36335] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Accepted: 09/24/2008] [Indexed: 01/18/2023] Open
Abstract
Eosinophils are multifunctional leukocytes that degrade and remodel tissue extracellular matrix through production of proteolytic enzymes, release of proinflammatory factors to initiate and propagate inflammatory responses, and direct activation of mucus secretion and smooth muscle cell constriction. Thus, eosinophils are central effector cells during allergic airway inflammation and an important clinical therapeutic target. Here we describe the use of an injectable MMP-targeted optical sensor that specifically and quantitatively resolves eosinophil activity in the lungs of mice with experimental allergic airway inflammation. Through the use of real-time molecular imaging methods, we report the visualization of eosinophil responses in vivo and at different scales. Eosinophil responses were seen at single-cell resolution in conducting airways using near-infrared fluorescence fiberoptic bronchoscopy, in lung parenchyma using intravital microscopy, and in the whole body using fluorescence-mediated molecular tomography. Using these real-time imaging methods, we confirmed the immunosuppressive effects of the glucocorticoid drug dexamethasone in the mouse model of allergic airway inflammation and identified a viridin-derived prodrug that potently inhibited the accumulation and enzyme activity of eosinophils in the lungs. The combination of sensitive enzyme-targeted sensors with noninvasive molecular imaging approaches permitted evaluation of airway inflammation severity and was used as a model to rapidly screen for new drug effects. Both fluorescence-mediated tomography and fiberoptic bronchoscopy techniques have the potential to be translated into the clinic.
Collapse
Affiliation(s)
- Virna Cortez-Retamozo
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Filip K. Swirski
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Waterman
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Hushan Yuan
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jose Luiz Figueiredo
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Andita P. Newton
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rabi Upadhyay
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Claudio Vinegoni
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rainer Kohler
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph Blois
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Adam Smith
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Matthias Nahrendorf
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lee Josephson
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ralph Weissleder
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mikael J. Pittet
- Center for Systems Biology and
Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
1621
|
Lucignani G. Hi-tech systems for in-vivo image-guided preclinical radiobiology. Eur J Nucl Med Mol Imaging 2008; 35:2334-8. [DOI: 10.1007/s00259-008-0964-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
1622
|
Ratiometric multimodal chemosensors based on cubic silsesquioxanes for monitoring solvent polarity. Bioorg Med Chem 2008; 16:10029-33. [DOI: 10.1016/j.bmc.2008.10.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 10/04/2008] [Accepted: 10/07/2008] [Indexed: 11/17/2022]
|
1623
|
Zhou YF, Gulaka P, Zhou J, Xiao M, Xu D, Hsieh JT, Kodibagkar VD, Sun X. Preparation and Evaluation of a Radioisotope-Incorporated Iron Oxide Core/Au Shell Nanoplatform for Dual Modality Imaging. J Biomed Nanotechnol 2008. [DOI: 10.1166/jbn.2008.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
1624
|
|
1625
|
Pierce MC, Javier DJ, Richards-Kortum R. Optical contrast agents and imaging systems for detection and diagnosis of cancer. Int J Cancer 2008; 123:1979-90. [PMID: 18712733 PMCID: PMC2902964 DOI: 10.1002/ijc.23858] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Molecular imaging has rapidly emerged as a discipline with the potential to impact fundamental biomedical research and clinical practice. Within this field, optical imaging offers several unique capabilities, based on the ability of cells and tissues to effect quantifiable changes in the properties of visible and near-infrared light. Beyond endogenous optical properties, the development of molecularly targeted contrast agents enables disease-specific morphologic and biochemical processes to be labeled with unique optical signatures. Optical imaging systems can then provide real-time visualization of pathophysiology at spatial scales from the subcellular to whole organ levels. In this article, we review fundamental techniques and recent developments in optical molecular imaging, emphasizing laboratory and clinical systems that aim to visualize the microscopic and macroscopic hallmarks of cancer.
Collapse
Affiliation(s)
- Mark C Pierce
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | | |
Collapse
|
1626
|
Abstract
Preceded by three decades of intense basic research on tumour angiogenesis, we are assisting to the translation of anti-antiangiogenic therapies as medical oncologists are increasingly using pioneering anti-angiogenic drugs in combination with standard treatments. While basic knowledge in the field of angiogenesis is reaching maturity and our level of understanding of the complex process of vessel development and growth in health and disease has been enriched at the molecular and cellular levels, the translation of this knowledge to the clinic is still in its infancy. Identifying the most suitable drugs, and the optimal dosage and schedule, as well as monitoring patients' responses to anti-angiogenic therapy, remain challenging issues that currently limit the benefit of this new therapeutic approach in cancer. This review will focus on a comprehensive description of the experimental assays in which angiogenesis research has been founded and how the different assays complement and provide relevant information for the task of characterising the angiogenic properties of diverse tumours, giving us a variety of tools to follow up tumour angiogenesis in research models. Following up tumour angiogenesis in patients and their response to antiangiogenic therapy is a more challenging task that will benefit in the near future from the use of non-invasive imaging methods as well as molecular and cellular biomarkers of angiogenesis suitable for clinical oncology. As both the design of the anti-angiogenic therapies and monitoring of the response are improved in the coming years to properly tailor them to the angiogenic profile of every patient, we hope to achieve increasing response and benefit of including antiangiogenic drugs as standard in cancer therapy.
Collapse
|
1627
|
Anglim PP, Alonzo TA, Laird-Offringa IA. DNA methylation-based biomarkers for early detection of non-small cell lung cancer: an update. Mol Cancer 2008; 7:81. [PMID: 18947422 PMCID: PMC2585582 DOI: 10.1186/1476-4598-7-81] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Accepted: 10/23/2008] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is the number one cancer killer in the United States. This disease is clinically divided into two sub-types, small cell lung cancer, (10–15% of lung cancer cases), and non-small cell lung cancer (NSCLC; 85–90% of cases). Early detection of NSCLC, which is the more common and less aggressive of the two sub-types, has the highest potential for saving lives. As yet, no routine screening method that enables early detection exists, and this is a key factor in the high mortality rate of this disease. Imaging and cytology-based screening strategies have been employed for early detection, and while some are sensitive, none have been demonstrated to reduce lung cancer mortality. However, mortality might be reduced by developing specific molecular markers that can complement imaging techniques. DNA methylation has emerged as a highly promising biomarker and is being actively studied in multiple cancers. The analysis of DNA methylation-based biomarkers is rapidly advancing, and a large number of potential biomarkers have been identified. Here we present a detailed review of the literature, focusing on DNA methylation-based markers developed using primary NSCLC tissue. Viable markers for clinical diagnosis must be detectable in 'remote media' such as blood, sputum, bronchoalveolar lavage, or even exhaled breath condensate. We discuss progress on their detection in such media and the sensitivity and specificity of the molecular marker panels identified to date. Lastly, we look to future advancements that will be made possible with the interrogation of the epigenome.
Collapse
Affiliation(s)
- Paul P Anglim
- Departments of Surgery and of Biochemistry and Molecular Biology, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089-9176, USA.
| | | | | |
Collapse
|
1628
|
Kaijzel EL, Snoeks TJA, Buijs JT, van der Pluijm G, Löwik CWGM. Multimodal imaging and treatment of bone metastasis. Clin Exp Metastasis 2008; 26:371-9. [PMID: 18941911 DOI: 10.1007/s10585-008-9217-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Accepted: 09/29/2008] [Indexed: 02/06/2023]
Abstract
The role of molecular imaging in pre-clinical research is continuously evolving. Particularly in small animal models in biomedical research, optical imaging technologies are frequently used to visualize normal as well as aberrant cellular processes at a molecular-genetic or cellular level of function. Also in cancer metastasis research, whole body bioluminescent and fluorescent imaging techniques have become indispensable tools that allow non-invasive and real-time imaging of gene expression, tumor progression and metastasis, and response to therapeutic intervention. In this paper, we discuss the use of optical imaging strategies--either alone or in combination with CT--to study intrabone tumor growth, tumor progression and to monitor efficacy of therapeutic agents in metastatic bone disease.
Collapse
Affiliation(s)
- Eric L Kaijzel
- Department of Endocrinology, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
1629
|
|
1630
|
de Ryk J, Namati E, Thiesse J, McLennan G. New imaging approaches for understanding lung cancer response to treatment. Clin Pharmacol Ther 2008; 84:517-22. [PMID: 18769370 PMCID: PMC4367487 DOI: 10.1038/clpt.2008.160] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The survival rate for lung cancer patients has barely improved over the past 30 years. New evaluation benchmarks for cancer response are needed to test therapy agents in a cost-effective and timely manner. From recent work, it is evident that primary lung cancers are very complex structures containing not only cancerous cells but also fibrotic and inflammatory cells and necrotic tissue. A greater understanding of the three-dimensional structure of primary lung cancer is emerging, allowing for the first time an appreciation of how this biomass is represented in medical imaging data. It is only through a greater understanding of the lung cancer biomass that we can define rational and early-response measures, including specific cellular responses such as cancer cell death or growth inhibition. In doing so, we can define response metrics that will shorten new drug discovery times and reduce costs, allowing for the evaluation of many more agents with therapeutic potential.
Collapse
Affiliation(s)
- J de Ryk
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - E Namati
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - J Thiesse
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - G McLennan
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
1631
|
Debbage P, Jaschke W. Molecular imaging with nanoparticles: giant roles for dwarf actors. Histochem Cell Biol 2008; 130:845-75. [PMID: 18825403 DOI: 10.1007/s00418-008-0511-y] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2008] [Indexed: 10/25/2022]
Abstract
Molecular imaging, first developed to localise antigens in light microscopy, now encompasses all imaging modalities including those used in clinical care: optical imaging, nuclear medical imaging, ultrasound imaging, CT, MRI, and photoacoustic imaging. Molecular imaging always requires accumulation of contrast agent in the target site, often achieved most efficiently by steering nanoparticles containing contrast agent into the target. This entails accessing target molecules hidden behind tissue barriers, necessitating the use of targeting groups. For imaging modalities with low sensitivity, nanoparticles bearing multiple contrast groups provide signal amplification. The same nanoparticles can in principle deliver both contrast medium and drug, allowing monitoring of biodistribution and therapeutic activity simultaneously (theranostics). Nanoparticles with multiple bioadhesive sites for target recognition and binding will be larger than 20 nm diameter. They share functionalities with many subcellular organelles (ribosomes, proteasomes, ion channels, and transport vesicles) and are of similar sizes. The materials used to synthesise nanoparticles include natural proteins and polymers, artificial polymers, dendrimers, fullerenes and other carbon-based structures, lipid-water micelles, viral capsids, metals, metal oxides, and ceramics. Signal generators incorporated into nanoparticles include iron oxide, gadolinium, fluorine, iodine, bismuth, radionuclides, quantum dots, and metal nanoclusters. Diagnostic imaging applications, now appearing, include sentinal node localisation and stem cell tracking.
Collapse
Affiliation(s)
- Paul Debbage
- Department of Anatomy, Division of Histology and Embryology, Medical University Innsbruck, Muellerstrasse 59, 6020, Innsbruck, Austria.
| | | |
Collapse
|
1632
|
Debouck C. Integrating genomics across drug discovery and development. Toxicol Lett 2008; 186:9-12. [PMID: 18930125 DOI: 10.1016/j.toxlet.2008.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Accepted: 09/17/2008] [Indexed: 12/22/2022]
Abstract
The sequencing of the human genome was an exceptional achievement, but it was not an end in itself as it set the foundation for building new knowledge in biology and medicine. The laborious, multifaceted science of drug discovery and development also draws tremendous benefits from mining the human genome and exploiting the large palette of genomic technologies. This article discusses how diverse genomic tools have been used to date and how they will continue to be utilized in the future to impact drug discovery and development. Integrating genomics across drug discovery and development will undoubtedly help to shorten timelines, increase success rates at all stages and ultimately bring the right drugs to the right patients at the right times.
Collapse
|
1633
|
Kee HL, Nothdurft R, Muthiah C, Diers JR, Fan D, Ptaszek M, Bocian DF, Lindsey JS, Culver JP, Holten D. Examination of Chlorin-Bacteriochlorin Energy-transfer Dyads as Prototypes for Near-infrared Molecular Imaging Probes†. Photochem Photobiol 2008; 84:1061-72. [DOI: 10.1111/j.1751-1097.2008.00409.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
1634
|
Currid CA, Gallagher WM. Selected highlights of the Third Annual Biomarkers Congress: from discovery to validation in the drug development arena. EXPERT OPINION ON MEDICAL DIAGNOSTICS 2008; 2:1091-1094. [PMID: 23495928 DOI: 10.1517/17530059.2.9.1091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
This report summarises selected presentations that focused on aspects of biomarker discovery and validation, imaging in preclinical and clinical development and biomarkers in clinical drug development, as discussed during the Third Annual Biomarkers Congress held in Manchester, UK on 14 - 15 May 2008. More than 140 delegates attended this event to discuss the application of biomarkers to all facets of the drug development process. The conference consisted of five intensive streams discussing current and future directions in: i) biomarker discovery and validation; ii) molecular diagnostics, data integration, data analysis, modelling and bioinformatics; iii) biomarkers in clinical drug development; iv) biomarker discovery and validation: therapeutic areas; and v) imaging in preclinical and clinical development and safety biomarkers. This review will focus on selected presentations from the first, third and fifth streams.
Collapse
Affiliation(s)
- Caroline A Currid
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland +353 1 7166743 ; +353 1 2837211 ;
| | | |
Collapse
|
1635
|
Mempel TR, Bauer CA. Intravital imaging of CD8+ T cell function in cancer. Clin Exp Metastasis 2008; 26:311-27. [PMID: 18665448 DOI: 10.1007/s10585-008-9196-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Accepted: 07/14/2008] [Indexed: 12/11/2022]
Abstract
Recent technological advances in photonics are making intravital microscopy (IVM) an increasingly powerful approach for the mechanistic exploration of biological processes in the physiological context of complex native tissue environments. Direct, dynamic and multiparametric visualization of immune cell behavior in living animals at cellular and subcellular resolution has already proved its utility in auditing basic immunological concepts established through conventional approaches and has also generated new hypotheses that can conversely be complemented and refined by traditional experimental methods. The insight that outgrowing tumors must not necessarily have evaded recognition by the adaptive immune system, but can escape rejection by actively inducing a state of immunological tolerance calls for a detailed investigation of the cellular and molecular mechanisms by which the anti-cancer response is subverted. Along with molecular imaging techniques that provide dynamic information at the population level, IVM can be expected to make a critical contribution to this effort by allowing the observation of immune cell behavior in vivo at single cell-resolution. We review here how IVM-based investigation can help to clarify the role of cytotoxic T lymphocytes (CTL) in the immune response against cancer and identify the ways by which their function might be impaired through tolerogenic mechanisms.
Collapse
Affiliation(s)
- Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| | | |
Collapse
|
1636
|
Rankin SC. The role of positron emission tomography in staging of non-small cell lung cancer. Target Oncol 2008. [DOI: 10.1007/s11523-008-0085-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
1637
|
Griffiths GL. The imaging probe development center and the production of molecular imaging probes. CURRENT CHEMICAL GENOMICS 2008; 1:65-9. [PMID: 20161829 PMCID: PMC2774657 DOI: 10.2174/1875397300801010065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Revised: 05/17/2008] [Accepted: 05/21/2008] [Indexed: 11/22/2022]
Abstract
The Imaging Probe Development Center (IPDC), part of the NIH Roadmap for Medical Research Initiative (http://nihroadmap.nih.gov/) recently became fully operational at its newly refurbished laboratories in Rockville, MD. The IPDC (http://nihroadmap.nih.gov/molecularlibraries/ipdc/) is dedicated to the production of known and novel molecular imaging probes, with its services currently being used by the NIH intramural community, although in the future it is intended that the extramural community will also benefit from the IPDC's resources. The Center has been set up with the belief that molecular imaging, and the probe chemistry that underpins it, will constitute key technologies going forward. As part of the larger molecular libraries and imaging initiative, it is planned that the IPDC will work closely with scientists from the molecular libraries effort. Probes produced at the IPDC include optical, radionuclide and magnetic resonance agents and may encompass any type of contrast agent. As IPDC is a trans-NIH resource it can serve each of the 27 Institutes and Centers that comprise NIH so its influence can be expected to impact widely different subjects and disease conditions spanning biological research. IPDC is expected to play a key part in interdisciplinary collaborative imaging projects and to support translational R&D from basic research through clinical development, for all of the imaging modalities. Examples of probes already prepared or under preparation are outlined to illustrate the breadth of the chemistries undertaken together with a reference outline of the diverse biological applications for which the various probes are intended.
Collapse
Affiliation(s)
- Gary L Griffiths
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
1638
|
Tanaka K, Kitamura N, Naka K, Chujo Y. Multi-modal 19F NMR probe using perfluorinated cubic silsesquioxane-coated silica nanoparticles for monitoring enzymatic activity. Chem Commun (Camb) 2008:6176-8. [DOI: 10.1039/b815022b] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
1639
|
Abstract
Cancer remains an important and growing health problem. Researchers have made great progress in defining genetic and molecular alterations that contribute to cancer formation and progression. Molecular imaging can identify appropriate patients for targeted cancer therapy and may detect early biochemical changes in tumors during therapy, some of which may have important prognostic implications. Progress in this field continues largely due to a union between molecular genetics and advanced imaging technology. This review details uses of molecular-genetic imaging in the context of tumor-associated viruses. Under certain conditions, and particularly during pharmacologic stimulation, gammaherpesviruses will express genes that enable imaging and therapy in vivo. The techniques discussed are readily translatable to the clinic.
Collapse
Affiliation(s)
- De-Xue Fu
- Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD, 21231, USA
| | - Catherine A. Foss
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, 21231, USA
| | - Sridhar Nimmagadda
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, 21231, USA
| | - Richard F. Ambinder
- Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD, 21231, USA
| | - Martin G. Pomper
- Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD, 21231, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, 21231, USA
| |
Collapse
|
1640
|
Edelhauser G, Funovics M. Breast Cancer Treatment in the Era of Molecular Imaging. Breast Care (Basel) 2008; 3:409-414. [PMID: 21048912 DOI: 10.1159/000181160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Molecular imaging employs molecularly targeted probes to visualize and often quantify distinct disease-specific markers and pathways. Modalities like intravital confocal or multiphoton microscopy, near-infrared fluorescence combined with endoscopy, surface reflectance imaging, or fluorescence-mediated tomography, and radionuclide imaging with positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are increasingly used for small animal high-throughput screening, drug development and testing, and monitoring gene therapy experiments. In the clinical treatment of breast cancer, PET and SPECT as well as magnetic resonance-based molecular imaging are already established for the staging of distant disease and intrathoracic nodal status, for patient selection regarding receptor-directed treatments, and to gain early information about treatment efficacy. In the near future, reporter gene imaging during gene therapy and further spatial and qualitative characterization of the disease can become clinically possible with radionuclide and optical methods. Ultimately, it may be expected that every level of breast cancer treatment will be affected by molecular imaging, including screening.
Collapse
Affiliation(s)
- Gundula Edelhauser
- Workgroup for Experimental Radiology and Preclinical Imaging, Cardiovascular and Interventional Radiology, Department of Radiology, Medical University of Vienna, Austria
| | | |
Collapse
|