201
|
Mouchess ML, Sohara Y, Nelson MD, DeCLerck YA, Moats RA. Multimodal imaging analysis of tumor progression and bone resorption in a murine cancer model. J Comput Assist Tomogr 2006; 30:525-34. [PMID: 16778634 DOI: 10.1097/00004728-200605000-00030] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This study evaluates the use of multimodal imaging to qualitatively and quantitatively measure tumor progression and bone resorption in a xenotransplanted tumor model of human neuroblastoma. METHODS Human neuroblastoma cells expressing a luciferase reporter gene were injected into the femur of nu/nu mice. Tumor progression with and without zoledronic acid treatment was monitored using radiographs, D-luciferin-induced luminescence, micro-computer tomography (CT) and micro-magnetic resonance imaging (MRI). RESULTS We observed a gradual increase in D-luciferin-based bioluminescence concomitant with detectable osteolytic lesions. Tumor growth was inhibited (P=0.003-0.07) with zoledronic acid treatment. Micro-CT analysis in vivo provided a method to quantify bone loss, and its prevention by zoledronic acid. High-resolution MRI images allowed the observation of tumor cells within the bone marrow cavity, as well as distant metastasis. CONCLUSION Multimodal imaging allows to measure tumor growth and bone resorption simultaneously in vivo and also proved useful in the detection distant metastasis.
Collapse
Affiliation(s)
- Maria L Mouchess
- Division of Hematology Oncology, Department of Pediatrics, University of Southern California and the Saban Research Institute of Childrens Hospital Los Angeles, Los Angeles, California 90027, USA
| | | | | | | | | |
Collapse
|
202
|
Al-Gubory KH, Houdebine LM. In vivo imaging of green fluorescent protein-expressing cells in transgenic animals using fibred confocal fluorescence microscopy. Eur J Cell Biol 2006; 85:837-45. [PMID: 16781011 DOI: 10.1016/j.ejcb.2006.03.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2005] [Revised: 03/14/2006] [Accepted: 03/17/2006] [Indexed: 11/20/2022] Open
Abstract
Animal imaging requires the use of reliable long-term fluorescence methods and technology. The application of confocal imaging to in vivo monitoring of transgene expression within internal organs and tissues has been limited by the accessibility to these sites. We aimed to test the feasibility of fibred confocal fluorescence microscopy (FCFM) to image in situ green fluorescent protein (GFP) in cells of living animals. We used transgenic rabbits expressing the enhanced GFP (eGFP) gene. Detailed tissue architecture and cell morphology were visualised and identified in situ by FCFM. Imaging of vasculature by using FCFM revealed a single blood vessel or vasculature network. We also used non-transgenic female rabbits mated with transgenic males to visualise eGFP expression in extra-foetal membranes and the placenta. Expression of the eGFP gene was confirmed by FCFM. This new imaging technology offers specific characteristics: a way to gain access to organs and tissues in vivo, sensitive detection of fluorescent signals, and cellular observations with rapid acquisition at near real time. It allows an accurate visualisation of tissue anatomical structure and cell morphology. FCFM is a promising technology to study biological processes in the natural physiological environment of living animals.
Collapse
Affiliation(s)
- Kaïs H Al-Gubory
- Institut National de la Recherche Agronomique, Département de Physiologie Animale, Unité de Biologie du Développement et de la Reproduction, F-78352 Jouy-en-Josas Cedex, France.
| | | |
Collapse
|
203
|
Phadke PA, Mercer RR, Harms JF, Jia Y, Frost AR, Jewell JL, Bussard KM, Nelson S, Moore C, Kappes JC, Gay CV, Mastro AM, Welch DR. Kinetics of metastatic breast cancer cell trafficking in bone. Clin Cancer Res 2006; 12:1431-40. [PMID: 16533765 PMCID: PMC1523260 DOI: 10.1158/1078-0432.ccr-05-1806] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE In vivo studies have focused on the latter stages of the bone metastatic process (osteolysis), whereas little is known about earlier events, e.g., arrival, localization, and initial colonization. Defining these initial steps may potentially identify the critical points susceptible to therapeutic intervention. EXPERIMENTAL DESIGN MDA-MB-435 human breast cancer cells engineered with green fluorescent protein were injected into the cardiac left ventricle of athymic mice. Femurs were analyzed by fluorescence microscopy, immunohistochemistry, real-time PCR, flow cytometry, and histomorphometry at times ranging from 1 hour to 6 weeks. RESULTS Single cells were found in distal metaphyses at 1 hour postinjection and remained as single cells up to 72 hours. Diaphyseal arrest occurred rarely and few cells remained there after 24 hours. At 1 week, numerous foci (2-10 cells) were observed, mostly adjacent to osteoblast-like cells. By 2 weeks, fewer but larger foci (> or =50 cells) were seen. Most bones had a single large mass at 4 weeks (originating from a colony or coalescing foci) which extended into the diaphysis by 4 to 6 weeks. Little change (<20%) in osteoblast or osteoclast numbers was observed at 2 weeks, but at 4 to 6 weeks, osteoblasts were dramatically reduced (8% of control), whereas osteoclasts were reduced modestly (to approximately 60% of control). CONCLUSIONS Early arrest in metaphysis and minimal retention in diaphysis highlight the importance of the local milieu in determining metastatic potential. These results extend the Seed and Soil hypothesis by demonstrating both intertissue and intratissue differences governing metastatic location. Ours is the first in vivo evidence that tumor cells influence not only osteoclasts, as widely believed, but also eliminate functional osteoblasts, thereby restructuring the bone microenvironment to favor osteolysis. The data may also explain why patients receiving bisphosphonates fail to heal bone despite inhibiting resorption, implying that concurrent strategies that restore osteoblast function are needed to effectively treat osteolytic bone metastases.
Collapse
Affiliation(s)
| | - Robyn R. Mercer
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | | | | | - Andra R. Frost
- Departments of Pathology
- Comprehensive Cancer Center
- NFCR-Center for Metastasis Research, University of Alabama at Birmingham, Birmingham, AL and
| | - Jennifer L. Jewell
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Karen M. Bussard
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Shakira Nelson
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | | | | | - Carol V. Gay
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Andrea M. Mastro
- NFCR-Center for Metastasis Research, University of Alabama at Birmingham, Birmingham, AL and
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Danny R. Welch
- Departments of Pathology
- Comprehensive Cancer Center
- Center for Metabolic Bone Disease
- NFCR-Center for Metastasis Research, University of Alabama at Birmingham, Birmingham, AL and
- Requests for reprints: Danny R. Welch, Ph.D. Department of Pathology, University of Alabama at Birmingham; 1670 University Blvd.; Volker Hall G-019A; Birmingham, AL 35294-0019; Phone: +1-205-934-2961 Fax: +1-205-975-1126; ; or, Andrea M. Mastro, Ph.D., Department of Biochemistry and Molecular Biology, 231 South Frear, Pennsylvania State University, University Park, PA 16802; Phone: +1-814-863-0152; Fax: +1-814-863-7024;
| |
Collapse
|
204
|
Luo Y, Chen X, O'donnell MA. Use of prostate specific antigen to measure bladder tumor growth in a mouse orthotopic model. J Urol 2006; 172:2414-20. [PMID: 15538283 DOI: 10.1097/01.ju.0000143860.50878.b1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PURPOSE Animal orthotopic tumor models are commonly used in bladder cancer studies. However, to our knowledge there is currently no accurate method to quantify tumor growth inside the bladder in living animals. We used prostate specific antigen (PSA) as a marker to cope with this limitation. MATERIALS AND METHODS Infectious but replication incompetent retroviral particles carrying PSA coding sequence were constructed and infected into MB49 cells, a mouse bladder transitional cell carcinoma line of C57BL/6 origin. Syngeneic mice were intravesically implanted with the novel MB49-PSA transfectants. Tumor burden was evaluated by enzyme-linked immunosorbent assay measurement for PSA in urine and bladder tissues. RESULTS The MB49-PSA line actively secreted PSA in culture as well as in urine (18 to 2,062 pg/ml) depending on tumor mass. Immunofluorescence staining confirmed PSA expression in MB49-PSA derived orthotopic tumors. Urinary PSA production paralleled tumor growth and was detectable prior to the development of a palpable tumor. Although urinary PSA did not tightly correlate with tumor mass, all bladders (total of 16 tested) weighing 34 mg or greater (18 to 21 mg for age and sex matched normal bladders) showed 18 pg/ml or greater urinary PSA. In contrast, bladder tissue PSA correlated more with tumor mass in general and it was measurable even before the detection of urinary PSA. This MB49-PSA orthotopic tumor model also demonstrated its usefulness for evaluating the antibladder cancer agents gemcitabine and mitomycin. CONCLUSIONS This novel MB49-PSA line may serve as a useful tool for bladder cancer study because its growth inside the bladder can be noninvasively measured in living animals even during early stages of tumor growth.
Collapse
Affiliation(s)
- Yi Luo
- University of Iowa Department of Urology, Iowa City, Iowa 52242, USA.
| | | | | |
Collapse
|
205
|
Yamauchi K, Yang M, Jiang P, Xu M, Yamamoto N, Tsuchiya H, Tomita K, Moossa AR, Bouvet M, Hoffman RM. Development of real-time subcellular dynamic multicolor imaging of cancer-cell trafficking in live mice with a variable-magnification whole-mouse imaging system. Cancer Res 2006; 66:4208-14. [PMID: 16618743 DOI: 10.1158/0008-5472.can-05-3927] [Citation(s) in RCA: 196] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
With the use of dual-color fluorescent cells and a highly sensitive whole-mouse imaging system with both macro-optics and micro-optics, we report here the development of subcellular real-time imaging of cancer cell trafficking in live mice. To observe cytoplasmic and nuclear dynamics in the living mouse, tumor cells were labeled in the nucleus with green fluorescent protein and with red fluorescent protein in the cytoplasm. Dual-color cancer cells were injected by a vascular route in an abdominal skin flap in nude mice. The mice were imaged with an Olympus OV100 whole-mouse imaging system with a sensitive CCD camera and five objective lenses, parcentered and parfocal, enabling imaging from macrocellular to subcellular. We observed the nuclear and cytoplasmic behavior of cancer cells in real time in blood vessels as they moved by various means or adhered to the vessel surface in the abdominal skin flap. During extravasation, real-time dual-color imaging showed that cytoplasmic processes of the cancer cells exited the vessels first, with nuclei following along the cytoplasmic projections. Both cytoplasm and nuclei underwent deformation during extravasation. Different cancer cell lines seemed to strongly vary in their ability to extravasate. With the dual-color cancer cells and the highly sensitive whole-mouse imaging system described here, the subcellular dynamics of cancer metastasis can now be observed in live mice in real time. This imaging technology will enable further understanding of the critical steps of metastasis and provide visible targets for antimetastasis drug development.
Collapse
MESH Headings
- Animals
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/metabolism
- Carcinoma, Lewis Lung/pathology
- Cell Line, Tumor
- Cell Nucleus/metabolism
- Cytoplasm/metabolism
- Diagnostic Imaging/instrumentation
- Diagnostic Imaging/methods
- Fibrosarcoma/blood supply
- Fibrosarcoma/metabolism
- Fibrosarcoma/pathology
- Green Fluorescent Proteins/analysis
- Humans
- Luminescent Proteins/analysis
- Mammary Neoplasms, Experimental/blood supply
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Nude
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Neoplastic Cells, Circulating/metabolism
- Neoplastic Cells, Circulating/pathology
- Subcellular Fractions/chemistry
- Subcellular Fractions/metabolism
- Red Fluorescent Protein
Collapse
|
206
|
Winkelmann CT, Figueroa SD, Rold TL, Volkert WA, Hoffman TJ. Microimaging Characterization of a B16-F10 Melanoma Metastasis Mouse Model. Mol Imaging 2006. [DOI: 10.2310/7290.2006.00011] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
| | - Said Daibes Figueroa
- University of Missouri-Columbia
- Harry S. Truman Memorial Veterans Hospital-Columbia
| | | | | | - Timothy J. Hoffman
- University of Missouri-Columbia
- Harry S. Truman Memorial Veterans Hospital-Columbia
| |
Collapse
|
207
|
Abstract
Metastasis of melanoma to the central nervous system (CNS) remains one of the major barriers to successful treatment of this disease. Available treatment modalities are of limited clinical efficacy. This problem is compounded by the presence of the blood-brain barrier (BBB), an important consideration in the development of new therapeutic agents. Only in animal models can the dual properties of experimental tumours and the BBB be explored in one system. A variety of rodent models have been developed, utilizing both murine and human melanoma cell lines. These models have highlighted the complex biology of cerebral metastasis, involving apparent disease progression through the selection of subclones at each stage, eventually leading to disease in the brain. As demonstrated in a number of animal studies, different subpopulations of metastatic melanoma cells are likely to be responsible for parenchymal and leptomeningeal CNS disease. In addition, these animal systems have been used to demonstrate the potential efficacy of new chemotherapeutic drugs, radiation treatments and immunotherapeutic approaches for the treatment of melanoma brain metastasis. Key biological questions remain to be answered. In particular, the molecular and cellular mechanisms responsible for establishing cerebral melanoma must be clearly delineated. Several molecules, including vascular endothelial growth factor (VEGF) and integrins, appear to play important, but not definitive, roles. Other, as yet undefined, molecules appear to be critical. The identification of these factors in experimental models, with confirmatory studies in humans, will expand our understanding of cerebral melanoma and provide valuable new therapeutic targets for intervention in this difficult clinical problem.
Collapse
Affiliation(s)
- Lee D Cranmer
- Section of Hematology and Oncology, The Arizona Cancer Center, University of Arizona/University Medical Center, Tucson, Arizona 85724, USA.
| | | | | | | |
Collapse
|
208
|
Abstract
AbstractMolecular-genetic imaging in living organisms has become a new field with the exceptional growth over the past 5 years. Modern imaging is based on three technologies: nuclear, magnetic resonance and optical imaging. Most current molecular-genetic imaging strategies are “indirect,” coupling a “reporter gene” with a complimentary “reporter probe.” The reporter transgene usually encodes for an enzyme, receptor or transporter that selectively interacts with a radiolabeled probe and results in accumulation of radioactivity in the transduced cell. In addition, reporter systems based on the expression of fluorescence or bioluminescence proteins are becoming more widely applied in small animal imaging. This review begins with a description of Positron Emission Tomography (PET)-based imaging genes and their complimentary radiolabeled probes that we think will be the first to enter clinical trials. Then we describe other imaging genes, mostly for optical imaging, which have been developed by investigators working with a variety of disease models in mice. Such optical reporters are unlikely to enter the clinic, at least not in the near-term. Reporter gene constructs can be driven by constitutive promoter elements and used to monitor gene therapy vectors and the efficacy of gene targeting and transduction, as well as to monitor adoptive cell-based therapies. Inducible promoters can be used as “sensors” to monitor endogenous cell processes, including specific intracellular molecular-genetic events and the activity of signaling pathways, by regulating the magnitude of reporter gene expression.
Collapse
|
209
|
Yokoyama H, Nakanishi H, Kodera Y, Ikehara Y, Ohashi N, Ito Y, Koike M, Fujiwara M, Tatematsu M, Nakao A. Biological significance of isolated tumor cells and micrometastasis in lymph nodes evaluated using a green fluorescent protein-tagged human gastric cancer cell line. Clin Cancer Res 2006; 12:361-8. [PMID: 16428473 DOI: 10.1158/1078-0432.ccr-05-1963] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE The biological significance of isolated tumor cells and micrometastasis in lymph node defined by the International Union against Cancer remains essentially unknown because of the lack of appropriate animal models. In the present study, we developed a lymph node micrometastasis model featuring a human gastric cancer cell line tagged with green fluorescent protein gene (GCIY-EGFP), which allows visualization of even isolated tumor cells in the development of metastasis without histologic procedure. Using this model, we investigated the effect of surgery and chemotherapy on the growth of early-phase metastasis formation in the lymph nodes. EXPERIMENTAL DESIGN The time course of spontaneous inguinal lymph node metastasis after s.c. inoculation of GCIY-EGFP cells into nude mice was examined with fluorescence dissecting microscopy. Then, the effects of surgical removal of the primary tumor with or without anti-asialo GM1 treatment or postoperative chemotherapy on the growth of isolated tumor cells and micrometastasis in the lymph nodes were examined. RESULTS GCIY-EGFP cells were found to metastasize spontaneously to the inguinal lymph nodes to form isolated tumor cells, micrometastasis, and, finally, develop macroscopic metastasis at 1 to 2, 3 to 5, and 5 weeks postinjection, respectively. When the primary tumors were removed within 2 weeks of inoculation, isolated tumor cells, but not micrometastasis, in the lymph nodes regressed by 4 weeks after surgery in all the mice examined (five of five). This spontaneous regression of isolated tumor cells was completely reversed by anti-asialo GM1 treatment, which could deplete natural killer cells effectively in nude mice. Chemotherapy following resection of the primary tumor at an early stage partially eliminated the remaining micrometastasis in the lymph nodes. CONCLUSIONS These results suggest that isolated tumor cells in the regional lymph nodes regressed by removal of the primary tumor mainly via natural killer cell-mediated antitumor activity and that micrometastasis in the lymph nodes could be effectively eliminated by the postoperative chemotherapy.
Collapse
Affiliation(s)
- Hiroyuki Yokoyama
- Department of Surgery II, Nagoya University School of Medicine and Division of Oncological Pathology, Aichi Cancer Center Research Institute, Kanokoden, Chukisa-ku, 464-8681 Nagoya, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Pilch J, Brown DM, Komatsu M, Järvinen TAH, Yang M, Peters D, Hoffman RM, Ruoslahti E. Peptides selected for binding to clotted plasma accumulate in tumor stroma and wounds. Proc Natl Acad Sci U S A 2006; 103:2800-4. [PMID: 16476999 PMCID: PMC1413849 DOI: 10.1073/pnas.0511219103] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Screening of a phage library for peptides that bind to clotted plasma in the presence of liquid plasma yielded two cyclic decapeptides, CGLIIQKNEC (CLT1) and CNAGESSKNC (CLT2). When injected intravenously into mice bearing various types of tumors, fluorescein-conjugated CLT peptides accumulated in a fibrillar meshwork in the extracellular compartment of the tumors, but were not detectable in other tissues of the tumor-bearing mice. The tumor homing of both peptides was strongly reduced after coinjection with unlabeled CLT2, indicating that the two peptides recognize the same binding site. The CLT peptide fluorescence colocalized with staining for fibrin(ogen) present in the extravascular compartment of tumors, but not in other tissues. The CLT peptides did not home to tumors grown in fibrinogen-null mice or in mice that lack plasma fibronectin. The CLT peptides also accumulated at the sites of injury in arteries, skeletal muscle, and skin. We conclude that the CLT peptides recognize fibrin-fibronectin complexes formed by clotting of plasma proteins that have leaked into the extravascular space in tumors and other lesions. These peptides may be useful in targeting diagnostic and therapeutic materials into tumors and injured tissues.
Collapse
Affiliation(s)
- Jan Pilch
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Darren M. Brown
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Masanobu Komatsu
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Tero A. H. Järvinen
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Meng Yang
- AntiCancer, Inc., 7917 Ostrow Street, San Diego, CA 92111; and
| | - David Peters
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Robert M. Hoffman
- AntiCancer, Inc., 7917 Ostrow Street, San Diego, CA 92111; and
- Department of Surgery, University of California, 200 West Arbor Drive, San Diego, CA 92103-8220
| | - Erkki Ruoslahti
- *Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037
| |
Collapse
|
211
|
Doyle TC, Nawotka KA, Kawahara CB, Francis KP, Contag PR. Visualizing fungal infections in living mice using bioluminescent pathogenic Candida albicans strains transformed with the firefly luciferase gene. Microb Pathog 2006; 40:82-90. [PMID: 16426810 DOI: 10.1016/j.micpath.2005.11.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Revised: 11/03/2005] [Accepted: 11/07/2005] [Indexed: 11/23/2022]
Abstract
Animal studies with Candida albicans have provided models for understanding fungal virulence and antifungal drug development. To non-invasively monitor long-term Candida murine infections, clinical isolates were stably transformed with a codon-optimized luciferase gene to constitutively express luciferase. Chronic systemic infections were established in mice with engineered strains, and bioluminescent signals were apparent from kidneys by non-invasive imaging using charged-coupled device cameras. These infections were established in immune-competent mice, and bioluminescence was detectable in animals that showed no physiological consequence of infection, as well as those visually succumbing to the disease. Similarly, bioluminescence was measured from the vaginal tissue of mice infected vaginally. Fungal loads determined by plating vaginal lavages showed a similar pattern to the bioluminescent signals measured, and fungal infection could be detected in animals for over 30 days post infection by both modalities. The effect of the antifungal drug miconazole was tested in this model, and clearance in animals was apparent by both direct imaging and fungal load determination. The use of bioluminescence to monitor these and other models of Candida infections will greatly speed up the analysis of drug development studies, both in ease of visualizing infections and decreasing numbers of animals required to run such studies.
Collapse
Affiliation(s)
- Timothy C Doyle
- Xenogen Corporation, 860 Atlantic Avenue, Alameda, CA 94501, USA.
| | | | | | | | | |
Collapse
|
212
|
Yu RMK, Lin CC, Chan PK, Chow ESH, Murphy MB, Chan BP, Müller F, Strähle U, Cheng SH. Four-dimensional imaging and quantification of gene expression in early developing zebrafish (Danio rerio) embryos. Toxicol Sci 2006; 90:529-38. [PMID: 16434501 DOI: 10.1093/toxsci/kfj115] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Four-dimensional (4D) imaging is a powerful tool for studying three-dimensional (3D) changes in an organism through time. Different imaging systems for obtaining 3D data from in vivo specimens have been developed but usually involved large and expensive machines. We successfully used a simple inverted compound microscope and a commercially available program to study and quantify in vivo changes in sonic hedgehog (shh) expression during early development in a green fluorescence protein (GFP) transgenic zebrafish (Danio rerio) line. We applied the 4D system to study the effect of 100 microM cadmium exposure on shh expression. In control zebrafish embryos, shh:GFP expression was detected at about 9 h post-fertilization (hpf) and increased steadily in the next 7 h, peaking at about 17 hpf and decreasing in the following 4 h. In the same time period, different shh expression volumes were observed in cadmium-treated and control embryos. Embryos affected by cadmium-exposure demonstrated a down-regulation in shh expression. The number of GFP-expressing cells measured by flow cytometry decreased, and expression of neurogenin-1, a downstream target of the shh signaling pathway, was down-regulated, providing additional supporting data on the effects of cadmium on shh. In summary, we demonstrated the setup of a 4D imaging system and its application to the quantification of gene expression.
Collapse
Affiliation(s)
- Richard M K Yu
- Department of Biology and Chemistry, City University of Hong Kong, Hong Kong.
| | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Condeelis J, Singer RH, Segall JE. The great escape: when cancer cells hijack the genes for chemotaxis and motility. Annu Rev Cell Dev Biol 2006; 21:695-718. [PMID: 16212512 DOI: 10.1146/annurev.cellbio.21.122303.120306] [Citation(s) in RCA: 270] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The combined use of the new technologies of multiphoton-based intravital imaging, the chemotaxis-mediated collection of invasive cells, and high sensitivity expression profiling has allowed the correlation of the behavior of invasive tumor cells in vivo with their gene expression patterns. New insights have resulted including a gene expression signature for invasive cells and the tumor microenvironment invasion model. This model proposes that tumor invasion and metastasis can be studied as a problem resembling normal morphogenesis. We discuss how these new insights may lead to a better understanding of the molecular basis of the invasive behavior of tumor cells in vivo, which may result in new strategies for the diagnosis and treatment of metastasis.
Collapse
Affiliation(s)
- John Condeelis
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York 10461-1975, USA.
| | | | | |
Collapse
|
214
|
Tsuji K, Yamauchi K, Yang M, Jiang P, Bouvet M, Endo H, Kanai Y, Yamashita K, Moossa AR, Hoffman RM. Dual-color imaging of nuclear-cytoplasmic dynamics, viability, and proliferation of cancer cells in the portal vein area. Cancer Res 2006; 66:303-6. [PMID: 16397243 DOI: 10.1158/0008-5472.can-05-2958] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We used dual-color in vivo cellular imaging to visualize trafficking, nuclear-cytoplasmic dynamics, and the viability of cancer cells after their injection into the portal vein of mice. For these studies, we used dual-color fluorescent cancer cells that express green fluorescent protein (GFP) linked to histone H2B in the nucleus and retroviral red fluorescent protein (RFP) in the cytoplasm. Human HCT-116-GFP-RFP colon cancer and mouse mammary tumor (MMT) cells were HCT-116-GFP-RFP in the portal vein of nude mice. The cells were observed intravitally in the liver at the single-cell level using the Olympus OV100 whole-mouse imaging system. Most HCT-116-GFP-RFP cells remained in sinusoids near peripheral portal veins. Only a small fraction of the cancer cells invaded the lobular area. Extensive clasmocytosis (destruction of the cytoplasm) of the HCT-116-GFP-RFP cells occurred within 6 hours. The number of apoptotic cells rapidly increased within the portal vein within 12 hours of injection. Apoptosis was readily visualized in the dual-color cells by their altered nuclear morphology. The data suggest rapid death of HCT-116-GFP-RFP cells in the portal vein. In contrast, dual-color MMT-GFP-RFP cells injected into the portal vein mostly survived in the liver of nude mice 24 hours after injection. Many surviving MMT-GFP-RFP cells showed invasive figures with cytoplasmic protrusions. The cells grew aggressively and formed colonies in the liver. However, when the host mice were pretreated with cyclophosphamide, the HCT-116-GFP-RFP cells also survived and formed colonies in the liver after portal vein injection. These results suggest that a cyclophosphamide-sensitive host cellular system attacked the HCT-116-GFP-RFP cells but could not effectively kill the MMT-GFP-RFP cells.
Collapse
MESH Headings
- Animals
- Cell Growth Processes/physiology
- Cell Nucleus/metabolism
- Cell Survival/physiology
- Colonic Neoplasms/drug therapy
- Colonic Neoplasms/metabolism
- Colonic Neoplasms/pathology
- Cyclophosphamide/pharmacology
- Cytoplasm/metabolism
- Green Fluorescent Proteins/metabolism
- HCT116 Cells
- Humans
- Image Processing, Computer-Assisted
- Liver Neoplasms, Experimental/blood
- Liver Neoplasms, Experimental/secondary
- Luminescent Proteins/metabolism
- Male
- Mammary Neoplasms, Experimental/blood
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Nude
- NIH 3T3 Cells
- Neoplastic Cells, Circulating/drug effects
- Neoplastic Cells, Circulating/metabolism
- Neoplastic Cells, Circulating/pathology
- Portal Vein
- Red Fluorescent Protein
Collapse
|
215
|
Lun X, Yang W, Alain T, Shi ZQ, Muzik H, Barrett JW, McFadden G, Bell J, Hamilton MG, Senger DL, Forsyth PA. Myxoma virus is a novel oncolytic virus with significant antitumor activity against experimental human gliomas. Cancer Res 2005; 65:9982-9990. [PMID: 16267023 PMCID: PMC4373463 DOI: 10.1158/0008-5472.can-05-1201] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Myxoma virus, a poxvirus previously considered rabbit specific, can replicate productively in a variety of human tumor cells in culture. The purpose of this study was to determine if there was efficacy or toxicities of this oncolytic virus against experimental models of human malignant gliomas in vitro, in vivo, and ex vivo in malignant glioma specimens. In vitro, the majority of glioma cell lines tested (7 of 8, 87.5%) were fully permissive for myxoma virus replication and killed by infection. In vivo, intracerebral (i.c.) myxoma virus inoculation was well tolerated and produced only minimal focal inflammatory changes at the site of viral inoculation. U87 and U251 orthotopic xenograft models were used to assess myxoma virus efficacy in vivo. A single intratumoral injection of myxoma virus dramatically prolonged median survival compared with treatment with UV-inactivated myxoma virus. Median survival was not reached in myxoma virus-treated groups versus 47.3 days (U87; P = 0.0002) and 50.7 days (U251; P = 0.0027) in UV-inactivated myxoma virus-treated groups. Most myxoma virus-treated animals (12 of 13, 92%) were alive and apparently "cured" when the experiment was finished (>130 days). Interestingly, we found a selective and long-lived myxoma virus infection in gliomas in vivo. This is the first demonstration of the oncolytic activity of myxoma virus in vivo. The nonpathogenic nature of myxoma virus outside of the rabbit host, its capacity to be genetically modified, its ability to produce a long-lived infection in human tumor cells, and the lack of preexisting antibodies in the human population suggest that myxoma virus may be an attractive oncolytic agent against human malignant glioma.
Collapse
Affiliation(s)
- Xueqing Lun
- Departments of Oncology, Clinical Neurosciences, and Biochemistry and Molecular Biology, University of Calgary, and Tom Baker Cancer Centre, Calgary, Alberta, Canada
- Clark-Smith Integrative Brain Tumour Research Center, Calgary, Alberta, Canada
| | - Wenqing Yang
- Departments of Oncology, Clinical Neurosciences, and Biochemistry and Molecular Biology, University of Calgary, and Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Tommy Alain
- Departments of Oncology, Clinical Neurosciences, and Biochemistry and Molecular Biology, University of Calgary, and Tom Baker Cancer Centre, Calgary, Alberta, Canada
- Clark-Smith Integrative Brain Tumour Research Center, Calgary, Alberta, Canada
| | - Zhong-Qiao Shi
- Departments of Oncology, Clinical Neurosciences, and Biochemistry and Molecular Biology, University of Calgary, and Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Huong Muzik
- Departments of Oncology, Clinical Neurosciences, and Biochemistry and Molecular Biology, University of Calgary, and Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - John W. Barrett
- BioTherapeutics Research Group, Robarts Institute and Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Grant McFadden
- BioTherapeutics Research Group, Robarts Institute and Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - John Bell
- Ottawa Regional Cancer Centre, Ottawa, Ontario, Canada
| | - Mark G. Hamilton
- Departments of Oncology, Clinical Neurosciences, and Biochemistry and Molecular Biology, University of Calgary, and Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Donna L. Senger
- Departments of Oncology, Clinical Neurosciences, and Biochemistry and Molecular Biology, University of Calgary, and Tom Baker Cancer Centre, Calgary, Alberta, Canada
- Clark-Smith Integrative Brain Tumour Research Center, Calgary, Alberta, Canada
| | - Peter A. Forsyth
- Departments of Oncology, Clinical Neurosciences, and Biochemistry and Molecular Biology, University of Calgary, and Tom Baker Cancer Centre, Calgary, Alberta, Canada
- Clark-Smith Integrative Brain Tumour Research Center, Calgary, Alberta, Canada
| |
Collapse
|
216
|
Zacharakis G, Kambara H, Shih H, Ripoll J, Grimm J, Saeki Y, Weissleder R, Ntziachristos V. Volumetric tomography of fluorescent proteins through small animals in vivo. Proc Natl Acad Sci U S A 2005; 102:18252-7. [PMID: 16344470 PMCID: PMC1317905 DOI: 10.1073/pnas.0504628102] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Volumetric detection and accurate quantification of fluorescent proteins in entire animals would greatly enhance our ability to monitor biological processes in vivo. Here we present a quantitative tomographic technique for visualization of superficial and deep-seated (>2-3 mm) fluorescent protein activity in vivo. We demonstrate noninvasive imaging of lung tumor progression in a murine model, as well as imaging of gene delivery using a herpes virus vector. This technology can significantly improve imaging capacity over the current state of the art and should find wide in vivo imaging applications in drug discovery, immunology, and cancer research.
Collapse
Affiliation(s)
- Giannis Zacharakis
- Laboratory for Bio-optics and Molecular Imaging, Center for Molecular Imaging Research, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | |
Collapse
|
217
|
Roffler SR, Wang HE, Yu HM, Chang WD, Cheng CM, Lu YL, Chen BM, Cheng TL. A membrane antibody receptor for noninvasive imaging of gene expression. Gene Ther 2005; 13:412-20. [PMID: 16267569 DOI: 10.1038/sj.gt.3302671] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Monitoring gene expression is important to optimize gene therapy protocols and ensure that the proper tissue distribution is achieved in clinical practice. We developed a noninvasive imaging system based on the expression of artificial antibody receptors to trap hapten-labeled imaging probes. Functional membrane-bound anti-dansyl antibodies (DNS receptor) were stably expressed on melanoma cells in vitro and in vivo. A bivalent (DNS)2-diethylenetriaminepentaacetic 111Indium probe specifically bound to cells that expressed DNS receptors but not control scFv receptors. Importantly, the 111In probe preferentially localized to DNS receptors but not control receptors on tumors in mice as assessed by gamma camera imaging. By 48 h after intravenous injection, the uptake of the probe in tumors expressing DNS receptors was 72 times greater than the amount of probe in the blood. This targeting strategy may allow noninvasive assessment of the location, extent and persistence of gene expression in living animals and in the clinic.
Collapse
Affiliation(s)
- S R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
218
|
Bouvet M, Spernyak J, Katz MH, Mazurchuk RV, Takimoto S, Bernacki R, Rustum YM, Moossa AR, Hoffman RM. High correlation of whole-body red fluorescent protein imaging and magnetic resonance imaging on an orthotopic model of pancreatic cancer. Cancer Res 2005; 65:9829-33. [PMID: 16267005 DOI: 10.1158/0008-5472.can-05-1548] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have developed genetically fluorescent orthotopic models of human pancreatic cancer. In these models, noninvasive fluorescent protein imaging (FPI) of internal primary tumors and metastatic deposits has been carried out. Whole-body tumor images are easily and inexpensively obtained using FPI, permitting both detection and quantification of tumor load. In this study, we simultaneously compared single mice with a highly fluorescent, red fluorescent protein-expressing orthotopic pancreatic cancer xenografts with both FPI and high-resolution magnetic resonance imaging (MRI). Images were acquired at multiple time points after tumor implantation in the pancreas. Indwelling pancreatic primary tumors and metastatic foci were detected by both FPI and MRI. Moreover, a strong correlation existed between images taken with these two technologies. FPI permitted rapid, high-throughput imaging without the need for either anesthesia or contrast agents. Both FPI and MRI enabled accurate imaging of tumor growth and metastasis, although MRI enabled tissue structure to be visualized as well. FPI has high resolution and is exceedingly rapid with instant image capture. We suggest a complimentary role for these two imaging modalities.
Collapse
Affiliation(s)
- Michael Bouvet
- Department of Surgery, University of California at San Diego, CA 92111, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
219
|
Abstract
Naturally fluorescent proteins have revolutionized biology by enabling what was formerly invisible to be seen clearly. These proteins have allowed us to visualize, in real time, important aspects of cancer in living animals, including tumour cell mobility, invasion, metastasis and angiogenesis. These multicoloured proteins have allowed the colour-coding of cancer cells growing in vivo and enabled the distinction of host from tumour with single-cell resolution. Visualization of many aspects of cancer initiation and progression in vivo should be possible with fluorescent proteins.
Collapse
Affiliation(s)
- Robert M Hoffman
- AntiCancer Inc. and Department of Surgery, University of California, San Diego, 7917 Ostrow Street, San Diego, California 92111, USA.
| |
Collapse
|
220
|
Graham KC, Wirtzfeld LA, MacKenzie LT, Postenka CO, Groom AC, MacDonald IC, Fenster A, Lacefield JC, Chambers AF. Three-dimensional high-frequency ultrasound imaging for longitudinal evaluation of liver metastases in preclinical models. Cancer Res 2005; 65:5231-7. [PMID: 15958568 DOI: 10.1158/0008-5472.can-05-0440] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Liver metastasis is a clinically significant contributor to the mortality associated with melanoma, colon, and breast cancer. Preclinical mouse models are essential to the study of liver metastasis, yet their utility has been limited by the inability to study this dynamic process in a noninvasive and longitudinal manner. This study shows that three-dimensional high-frequency ultrasound can be used to noninvasively track the growth of liver metastases and evaluate potential chemotherapeutics in experimental liver metastasis models. Liver metastases produced by mesenteric vein injection of B16F1 (murine melanoma), PAP2 (murine H-ras-transformed fibroblast), HT-29 (human colon carcinoma), and MDA-MB-435/HAL (human breast carcinoma) cells were identified and tracked longitudinally. Tumor size and location were verified by histologic evaluation. Tumor volumes were calculated from the three-dimensional volumetric data, with individual liver metastases showing exponential growth. The importance of volumetric imaging to reduce uncertainty in tumor volume measurement was shown by comparing three-dimensional segmented volumes with volumes estimated from diameter measurements and the assumption of an ellipsoid shape. The utility of high-frequency ultrasound imaging in the evaluation of therapeutic interventions was established with a doxorubicin treatment trial. These results show that three-dimensional high-frequency ultrasound imaging may be particularly well suited for the quantitative assessment of metastatic progression and the evaluation of chemotherapeutics in preclinical liver metastasis models.
Collapse
MESH Headings
- Animals
- Cell Growth Processes/physiology
- Cell Line, Tumor
- Disease Models, Animal
- HT29 Cells
- Humans
- Liver Neoplasms, Experimental/diagnostic imaging
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/secondary
- Melanoma, Experimental/diagnostic imaging
- Melanoma, Experimental/pathology
- Melanoma, Experimental/secondary
- Mice
- Mice, Inbred C57BL
- Mice, SCID
- Necrosis
- Ultrasonography
Collapse
Affiliation(s)
- Kevin C Graham
- Department of Medical Biophysics, The University of Western Ontario, London, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
221
|
Xiong T, Zhang Z, Liu BF, Zeng S, Chen Y, Chu J, Luo Q. In vivo optical imaging of human adenoid cystic carcinoma cell metastasis. Oral Oncol 2005; 41:709-15. [PMID: 15935724 DOI: 10.1016/j.oraloncology.2005.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2005] [Accepted: 03/20/2005] [Indexed: 10/25/2022]
Abstract
A noninvasive, whole-body, real-time fluorescence optical imaging of stable high-level green fluorescent protein (GFP)-expressing human adenoid cystic carcinoma (ACC-M-GFP) was demonstrated for in vivo visualization of metastatic behavior in nude mice. Five-week-old female nude mice were injected with ACC-M-GFP in the primary organ: submandibular gland. Metastases were only visualized by GFP expression in the lung. However, metastatic lesions of ACC-M-GFP in the lung, muscle, bladder and bony were found by imaging of GFP expression in intact mice through tail vein injection of ACC-M-GFP cells. The construction of highly fluorescent and stable GFP transfectants of ACC-M has revealed the multi-organ metastatic capability of ACC-M cells through this optical imaging.
Collapse
Affiliation(s)
- Tao Xiong
- The Key Laboratory of Biomedical Photonics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China.
| | | | | | | | | | | | | |
Collapse
|
222
|
Yano S, Kuroda S, Lee JB, Shichinohe H, Seki T, Ikeda J, Nishimura G, Hida K, Tamura M, Iwasaki Y. In Vivo Fluorescence Tracking of Bone Marrow Stromal Cells Transplanted into a Pneumatic Injury Model of Rat Spinal Cord. J Neurotrauma 2005; 22:907-18. [PMID: 16083357 DOI: 10.1089/neu.2005.22.907] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Recent experimental studies have shown that bone marrow stromal cells (BMSC) differentiate into neural cells and reduce neurological deficits when transplanted into traumatized spinal cord. These findings have been derived primarily from histological analyses. We conducted a study directed chiefly at developing a non-invasive system for tracking BMSC transplanted into the spinal cord of living animals. In this study, we induced spinal cord injury (SCI) in rats with a pneumatic device. BMSC were harvested from transgenic mice expressing green fluorescence protein (BMSC-GFP), and were transplanted stereotactically into a control group of rats without SCI (n = 6) and a group with SCI (n = 3). At 2 and 4 weeks after transplantation, the dura mater was exposed and green fluorescence derived from the transplanted BMSC-GFP was observed. The distribution and differentiation of the transplanted cells were subsequently evaluated with immunohistochemistry. Green fluorescence could be detected around the transplantation site in three of six of the control rats. In all three rats subjected to SCI, green fluorescence was shown to spread from the site of BMSC-GFP injection toward the injury site, suggesting that the transplanted cells had migrated toward the lesion within the 4-week post-transplantation period. Histological evaluation suggested that the detected green fluorescence was emitted by cells that had distributed in the dorsal white matter, and demonstrated that some of the transplanted cells expressed neuronal or astrocytic markers. These results suggest the possibility of tracking BMSC transplanted into the spinal cord in living animals. Such noninvasive bioimaging techniques would be valuable for monitoring the fate of these transplanted cells and assessing the safety and efficacy of their transplantation.
Collapse
Affiliation(s)
- Shunsuke Yano
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
223
|
Yang M, Luiken G, Baranov E, Hoffman RM. Facile whole-body imaging of internal fluorescent tumors in mice with an LED flashlight. Biotechniques 2005; 39:170, 172. [PMID: 16116787 DOI: 10.2144/05392bm02] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Meng Yang
- AntiCancer, Inc., San Diego, CA 92111, USA
| | | | | | | |
Collapse
|
224
|
Ntziachristos V, Ripoll J, Wang LV, Weissleder R. Looking and listening to light: the evolution of whole-body photonic imaging. Nat Biotechnol 2005; 23:313-20. [PMID: 15765087 DOI: 10.1038/nbt1074] [Citation(s) in RCA: 963] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Optical imaging of live animals has grown into an important tool in biomedical research as advances in photonic technology and reporter strategies have led to widespread exploration of biological processes in vivo. Although much attention has been paid to microscopy, macroscopic imaging has allowed small-animal imaging with larger fields of view (from several millimeters to several centimeters depending on implementation). Photographic methods have been the mainstay for fluorescence and bioluminescence macroscopy in whole animals, but emphasis is shifting to photonic methods that use tomographic principles to noninvasively image optical contrast at depths of several millimeters to centimeters with high sensitivity and sub-millimeter to millimeter resolution. Recent theoretical and instrumentation advances allow the use of large data sets and multiple projections and offer practical systems for quantitative, three-dimensional whole-body images. For photonic imaging to fully realize its potential, however, further progress will be needed in refining optical inversion methods and data acquisition techniques.
Collapse
Affiliation(s)
- Vasilis Ntziachristos
- Center for Molecular Imaging Research, Massachusetts General Hospital & Harvard Medical School CNY149, 13th street 5406, Charlestown, Massachusetts 02129, USA.
| | | | | | | |
Collapse
|
225
|
Zacharakis G, Ripoll J, Weissleder R, Ntziachristos V. Fluorescent protein tomography scanner for small animal imaging. IEEE TRANSACTIONS ON MEDICAL IMAGING 2005; 24:878-85. [PMID: 16011317 DOI: 10.1109/tmi.2004.843254] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Microscopy of fluorescent proteins has enabled unprecedented insights into visualizing gene expression in living systems. Imaging deeper into animals, however, has been limited due to the lack of accurate imaging methods for the visible. We present a novel system designed to perform tomographic imaging of fluorescent proteins through whole animals. The tomographic method employed a multiangle, multiprojection illumination scheme, while detection was achieved using a highly sensitive charge-coupled device camera with appropriate filters. Light propagation was modeled using a modified solution to the diffusion equation to account for the high absorption and high scattering of tissue at the visible wavelengths. We show that the technique can quantitatively detect fluorescence with sub millimeter spatial resolution both in phantoms and in tissues. We conclude that the method could be applied in tomographic imaging of fluorescent proteins for in vivo targeting of different diseases and abnormalities.
Collapse
Affiliation(s)
- Giannis Zacharakis
- Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| | | | | | | |
Collapse
|
226
|
Hoffman RM. Advantages of multi-color fluorescent proteins for whole-body and in vivo cellular imaging. JOURNAL OF BIOMEDICAL OPTICS 2005; 10:41202. [PMID: 16178626 DOI: 10.1117/1.1992485] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The revolution of in vivo cancer biology enabled by fluorescent proteins is described. The high extinction coefficients, quantum yields, and unique spectral properties of fluorescent proteins have been taken advantage of in order to visualize, in real time, the important aspects of cancer in living animals, including tumor cell trafficking, invasion, metastasis, and angiogenesis. Fluorescent proteins enable whole-body imaging of tumors on internal organs. These multicolored proteins have allowed the color-coding of cancer cells growing in vivo with distinction of different cell types, including host from tumor, with single-cell resolution.
Collapse
Affiliation(s)
- Robert M Hoffman
- AntiCancer, Inc, 7917 Ostrow Street, San Diego, California 92111, USA.
| |
Collapse
|
227
|
|
228
|
Graves EE, Yessayan D, Turner G, Weissleder R, Ntziachristos V. Validation of in vivo fluorochrome concentrations measured using fluorescence molecular tomography. JOURNAL OF BIOMEDICAL OPTICS 2005; 10:44019. [PMID: 16178652 DOI: 10.1117/1.1993427] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Fluorescence molecular tomography (FMT) has emerged as a means of quantitatively imaging fluorescent molecular probes in three dimensions in living systems. To assess the accuracy of FMT in vivo, translucent plastic tubes containing a turbid solution with a known concentration of Cy 5.5 fluorescent dye are constructed and implanted subcutaneously in nude mice, simulating the presence of a tumor accumulating a fluorescent molecular reporter. Comparisons between measurements of fluorescent tubes made before and after implantation demonstrate that the accuracy of FMT reported for homogeneous phantoms extends to the in vivo situation. The sensitivity of FMT to background fluorescence is tested by imaging fluorescent tubes in mice injected with Cy 5.5-labeled Annexin V. For small tube fluorochrome concentrations, the presence of background fluorescence results in increases in the reconstructed concentration. This phenomenon is counteracted by applying a simple subtraction correction to the measured fluorescence data. The effects of varying tumor photon absorption are simulated by imaging fluorescent tubes with varying ink concentrations, and are found to be minor. These findings demonstrate the in vivo quantitative accuracy of fluorescence tomography, and encourage further development of this imaging modality as well as application of FMT in molecular imaging studies using fluorescent reporters.
Collapse
Affiliation(s)
- Edward E Graves
- Stanford University School of Medicine, Department of Radiation Oncology, Stanford, California 94305, USA.
| | | | | | | | | |
Collapse
|
229
|
Mansfield JR, Gossage KW, Hoyt CC, Levenson RM. Autofluorescence removal, multiplexing, and automated analysis methods for in-vivo fluorescence imaging. JOURNAL OF BIOMEDICAL OPTICS 2005; 10:41207. [PMID: 16178631 DOI: 10.1117/1.2032458] [Citation(s) in RCA: 213] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The ability to image and quantitate fluorescently labeled markers in vivo has generally been limited by autofluorescence of the tissue. Skin, in particular, has a strong autofluorescence signal, particularly when excited in the blue or green wavelengths. Fluorescence labels with emission wavelengths in the near-infrared are more amenable to deep-tissue imaging, because both scattering and autofluorescence are reduced as wavelengths are increased, but even in these spectral regions, autofluorescence can still limit sensitivity. Multispectral imaging (MSI), however, can remove the signal degradation caused by autofluorescence while adding enhanced multiplexing capabilities. While the availability of spectral "libraries" makes multispectral analysis routine for well-characterized samples, new software tools have been developed that greatly simplify the application of MSI to novel specimens.
Collapse
Affiliation(s)
- James R Mansfield
- Cambridge Research and Instrumentation, Inc., 35-B Cabot Road, Woburn, Massachusetts 01801, USA
| | | | | | | |
Collapse
|
230
|
Amoh Y, Yang M, Li L, Reynoso J, Bouvet M, Moossa AR, Katsuoka K, Hoffman RM. Nestin-linked green fluorescent protein transgenic nude mouse for imaging human tumor angiogenesis. Cancer Res 2005; 65:5352-7. [PMID: 15958583 DOI: 10.1158/0008-5472.can-05-0821] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We report here a novel transgenic nude mouse for the visualization of human tumor angiogenesis. We have recently shown that the neural stem cell marker nestin is expressed in hair follicle stem cells and blood vessel networks in the skin of C57/B6 transgenic mice with nestin regulatory element-driven green fluorescent protein (ND-GFP). Others have shown ND-GFP is expressed in the brain, pancreas, and testes in these mice. In the present study, the nestin ND-GFP gene was crossed into nude mice on the C57/B6 background to obtain ND-GFP nude mice. ND-GFP was expressed in the brain, spinal cord, pancreas, stomach, esophagus, heart, lung, blood vessels of glomeruli, blood vessels of skeletal muscle, testes, hair follicles, and blood vessel network in the skin of ND-GFP nude mice. Human lung cancer, pancreatic cancer, and colon cancer cell lines as well as a murine melanoma cell line and breast cancer tumor cell line expressing red fluorescent protein were implanted orthotopically, and a red fluorescent protein-expressing human fibrosarcoma was implanted s.c. in the ND-GFP nude mice. These tumors grew extensively in the ND-GFP mice. ND-GFP was highly expressed in proliferating endothelial cells and nascent blood vessels in the growing tumors, visualized by dual-color fluorescence imaging. Results of immunohistochemical staining showed that CD31 was expressed in the ND-GFP-expressing nascent blood vessels. The ND-GFP transgenic nude mouse model enables the visualization of nascent angiogenesis in human and mouse tumor progression. These results suggest that this model is useful for the imaging of the angiogenesis of human as well as rodent tumors and visualization of the efficacy of angiogenetic inhibitors.
Collapse
|
231
|
Nakajima M, Takeda M, Kobayashi M, Suzuki S, Ohuchi N. Nano-sized fluorescent particles as new tracers for sentinel node detection: experimental model for decision of appropriate size and wavelength. Cancer Sci 2005; 96:353-6. [PMID: 15958058 PMCID: PMC11159817 DOI: 10.1111/j.1349-7006.2005.00053.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The concepts of made-to-order and low-invasiveness medicines are becoming widely accepted. A treatment for cancer, with minimum invasive surgery and without lymph nodes dissection based on sentinel lymph node (SN) navigation surgery, would adhere to these concepts. Dyes and/or radioisotopes are employed for SN detection in standard methods, however, each detection method has advantages and disadvantages. To make up for the disadvantages, we aimed at developing a new non-invasive method using fluorescent beads of uniform nano-size that could efficiently visualize SN from outside the body, and conducted experiments to determine the appropriate size and fluorescent wavelength. We examined various bead sizes and fluorescent wavelengths. The sizes were 20, 40, 100 and 200 nm. The fluorescent peak wavelengths of the beads were yellow-green (515 nm), dark red (680 nm), far red (720 nm) and infrared (755 nm). The beads were subcutaneously injected into the foot pad of the hind leg of a rat, and followed by laser scanning of the inguinal area for fluorescence observation. The beads exhibited different times for the fluorescence detection according to their sizes and wavelength. The 40 nm beads were considered to be the most appropriate size for SN detection in rats. The wavelength of near infrared was effective for avoiding attenuation by the tissue. In conclusion, we confirmed that uniformly nano-sized fluorescent beads have the potential to be an alternative to existing tracers in the detection of the SN in animal experiments if we select the appropriate particle size and wavelength.
Collapse
Affiliation(s)
- Morio Nakajima
- Division of Surgical Oncology, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Sendai 980-8574, Japan
| | | | | | | | | |
Collapse
|
232
|
Peters CM, Ghilardi JR, Keyser CP, Kubota K, Lindsay TH, Luger NM, Mach DB, Schwei MJ, Sevcik MA, Mantyh PW. Tumor-induced injury of primary afferent sensory nerve fibers in bone cancer pain. Exp Neurol 2005; 193:85-100. [PMID: 15817267 DOI: 10.1016/j.expneurol.2004.11.028] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2004] [Revised: 11/11/2004] [Accepted: 11/18/2004] [Indexed: 10/25/2022]
Abstract
Bone is the most common site of chronic pain in patients with metastatic cancer. What remains unclear are the mechanisms that generate this pain and why bone cancer pain can be so severe and refractory to treatment with opioids. Here we show that following injection and confinement of NCTC 2472 osteolytic tumor cells within the mouse femur, tumor cells sensitize and injure the unmyelinated and myelinated sensory fibers that innervate the marrow and mineralized bone. This tumor-induced injury of sensory nerve fibers is accompanied by an increase in ongoing and movement-evoked pain behaviors, an upregulation of activating transcription factor 3 (ATF3) and galanin by sensory neurons that innervate the tumor-bearing femur, upregulation of glial fibrillary acidic protein (GFAP) and hypertrophy of satellite cells surrounding sensory neuron cell bodies within the ipsilateral dorsal root ganglia (DRG), and macrophage infiltration of the DRG ipsilateral to the tumor-bearing femur. Similar neurochemical changes have been described following peripheral nerve injury and in other non-cancerous neuropathic pain states. Chronic treatment with gabapentin did not influence tumor growth, tumor-induced bone destruction or the tumor-induced neurochemical reorganization that occurs in sensory neurons or the spinal cord, but it did attenuate both ongoing and movement-evoked bone cancer-related pain behaviors. These results suggest that even when the tumor is confined within the bone, a component of bone cancer pain is due to tumor-induced injury to primary afferent nerve fibers that innervate the tumor-bearing bone. Tumor-derived, inflammatory, and neuropathic mechanisms may therefore be simultaneously driving this chronic pain state.
Collapse
Affiliation(s)
- Christopher M Peters
- Neurosystems Center and Department of Preventive Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Chung A, Wachsmann-Hogiu S, Zhao T, Xiong Y, Joseph A, Farkas DL. Advanced Optical Imaging Requiring No Contrast Agents—A New Armamentarium for Medicine and Surgery. ACTA ACUST UNITED AC 2005; 62:365-70. [PMID: 15890230 DOI: 10.1016/j.cursur.2004.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2004] [Accepted: 12/07/2004] [Indexed: 11/20/2022]
Affiliation(s)
- Alice Chung
- Minimally Invasive Surgical Technologies Institute, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA.
| | | | | | | | | | | |
Collapse
|
234
|
Abstract
With an increasing understanding of the molecular basis of disease, various new imaging targets have recently been defined that potentially allow for an early, sensitive, and specific diagnosis of disease or monitoring of treatment response. Different approaches to depict these molecular structures in vivo are currently being explored by the molecular imaging community. We briefly review methodologies for molecular imaging by magnetic resonance imaging and optical methods. Special emphasis is put on different contrast agent designs (e.g., targeted and smart probes). New technical developments in optical imaging are briefly discussed. In addition, current research results are put into a clinical perspective to elucidate the potential merits one might expect from this new research field.
Collapse
Affiliation(s)
- T Persigehl
- Department of Clinical Radiology, University Hospital Muenster, Albert-Schweitzer-Str. 33, D-48129 Münster, Germany
| | | | | |
Collapse
|
235
|
Tannous BA, Kim DE, Fernandez JL, Weissleder R, Breakefield XO. Codon-Optimized Gaussia Luciferase cDNA for Mammalian Gene Expression in Culture and in Vivo. Mol Ther 2005; 11:435-43. [PMID: 15727940 DOI: 10.1016/j.ymthe.2004.10.016] [Citation(s) in RCA: 539] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2004] [Accepted: 10/27/2004] [Indexed: 01/12/2023] Open
Abstract
Photoproteins have played a major role in advancing our understanding of biological processes. A broader array of biocompatible, nontoxic, and novel reporters can serve to expand this potential. Here we describe the properties of a luciferase from the copepod marine organism Gaussia princeps. It is a monomeric protein composed of 185 aa (19.9 kDa) with a short coding sequence (555 bp) making it suitable for viral vectors. The humanized form of Gaussia luciferase (hGLuc) was efficiently expressed in mammalian cells following delivery by HSV-1 amplicon vectors. It was found to be nontoxic and naturally secreted, with flash bioluminescence characteristics similar to those of other coelenterazine luciferases. hGLuc generated over 1000-fold higher bioluminescent signal intensity from live cells together with their immediate environment and over 100-fold higher intensity from viable cells alone (not including secreted luciferase) or cell lysates, compared to humanized forms of firefly (hFLuc) and Renilla (hRLuc) luciferases expressed under similar conditions. Furthermore, hGLuc showed 200-fold higher signal intensity than hRLuc and intensity comparable to that of hFLuc in vivo under standard imaging conditions. Gaussia luciferase provides a sensitive means of imaging gene delivery and other events in living cells in culture and in vivo, with a unique combination of features including high signal intensity, secretion, and ATP independence, thus being able to report from the cells and their environment in real time.
Collapse
Affiliation(s)
- Bakhos A Tannous
- Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129, USA.
| | | | | | | | | |
Collapse
|
236
|
Xu Y, Sun HC, Tian B, Li Y, Chen J, Chen J, Gao DM, Xue Q, Tang ZY. Establishment of green fluorescent protein-expressing hepatocellular carcinoma cell lines with different metastatic potential: relevant models for in vivo monitoring of metastasis and angiogenesis. J Cancer Res Clin Oncol 2005; 130:375-82. [PMID: 15133661 DOI: 10.1007/s00432-004-0551-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE To establish stable green fluorescent protein (GFP)-expressing metastatic human hepatocellular carcinoma (HCC) cell lines with different metastatic potential for long-term in vivo studies of metastasis and angiogenesis. METHODS The pIRES2-EGFP vector, which contains an enhanced GFP gene, was transfected into MHCC97-H and MHCC97-L, HCC cell lines with different metastatic potential. The stability of GFP expression, basic biological characteristics, invasion abilities in vitro, and spontaneous metastasis in vivo of the new cell lines (MHCC97-HG and MHCC97-LG) were studied. Microvessel density (MVD) of orthotopic implanted tumors was compared by anti-CD31 immunohistochemical staining, and real-time angiogenesis and metastasis of GFP-transfected tumors were detected by intravital fluorescent microscope. RESULTS The GFP-transfected cell lines stably expressed green fluorescence in the absence of G418 over a 36-day period. Compared with the parental cell lines, they exhibited no distinct differences in biological characteristics. MHCC97-HG showed more aggressive invasion and spontaneous metastatic behavior than MHCC97-LG, and even its parental cell line, MHCC97-H (P<0.01). MVD levels induced by MHCC97-HG orthotopic implanted tumors were significantly higher than MHCC97-LG (P<0.01). Real-time angiogenesis and sequential steps of metastasis could be detected clearly under intravital fluorescent microscope. CONCLUSIONS These two stable GFP-expressing HCC cell lines with the same genetic background and different metastatic potential were established, which could be useful models for monitoring metastasis and angiogenesis of HCC.
Collapse
Affiliation(s)
- Yang Xu
- Liver Cancer Institute and Zhong Shan Hospital, Fudan University, 136 Yi Xue Yuan Road, 200032 Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Yang WQ, Senger DL, Lun XQ, Muzik H, Shi ZQ, Dyck RH, Norman K, Brasher PMA, Rewcastle NB, George D, Stewart D, Lee PWK, Forsyth PA. Reovirus as an experimental therapeutic for brain and leptomeningeal metastases from breast cancer. Gene Ther 2005; 11:1579-89. [PMID: 15372068 DOI: 10.1038/sj.gt.3302319] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Brain and leptomeningeal metastases are common in breast cancer patients and our current treatments are ineffective. Reovirus type 3 is a replication competent, naturally occurring virus that usurps the activated Ras-signaling pathway (or an element thereof) of tumor cells and lyses them but leaves normal cells relatively unaffected. In this study we evaluated reovirus as an experimental therapeutic in models of central nervous system (CNS) metastasis from breast cancer. We found all breast cancer cell lines tested were susceptible to reovirus, with > 50% of these cells lysed within 72 h of infection. In vivo neurotoxicity studies showed only mild local inflammation at the injection site and mild communicating hydrocephalus with neither diffuse encephalitis nor behavioral abnormalities at the therapeutically effective dose of reovirus (intracranial) (ie 10(7) plaque-forming units) or one dose level higher. In vivo, a single intratumoral administration of reovirus significantly reduced the size of tumors established from two human breast cancer cell lines and significantly prolonged survival. Intrathecal administration of reovirus also remarkably prolonged survival in an immunocompetent racine model of leptomeningeal metastases. These data suggest that the evaluation of reovirus as an experimental therapeutic for CNS metastases from breast cancer is warranted.
Collapse
Affiliation(s)
- W Q Yang
- Department of Oncology, University of Calgary, Tom Baker Cancer Centre, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Mayer-Kuckuk P, Menon LG, Blasberg RG, Bertino JR, Banerjee D. Role of reporter gene imaging in molecular and cellular biology. Biol Chem 2005; 385:353-61. [PMID: 15195994 DOI: 10.1515/bc.2004.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Molecular imaging, including reporter gene methods, provides a unique opportunity to study biology in a living subject, thereby allowing physiological events to be monitored in an intact microenvironment. This review takes a molecular and cell biology perspective on recent studies which utilize reporter gene imaging as a tool to non-invasively monitor specific molecular biology pathways in vivo. Studies in rodent models demonstrate the feasibility of reporter gene imaging to visualize and measure key cellular pathways, such as transcription, translation and protein-protein interactions. The review indicates that molecular imaging is likely to be useful in the translation of molecular biology to medicine and biotechnological applications.
Collapse
Affiliation(s)
- Philipp Mayer-Kuckuk
- Department of Pharmacology, The Cancer Institute of New Jersey, UMDNJ, New Brunswick, NJ 08903, USA.
| | | | | | | | | |
Collapse
|
239
|
Zhao M, Yang M, Li XM, Jiang P, Baranov E, Li S, Xu M, Penman S, Hoffman RM. Tumor-targeting bacterial therapy with amino acid auxotrophs of GFP-expressing Salmonella typhimurium. Proc Natl Acad Sci U S A 2005; 102:755-60. [PMID: 15644448 PMCID: PMC545558 DOI: 10.1073/pnas.0408422102] [Citation(s) in RCA: 389] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Here we report a genetically modified bacteria strain, Salmonella typhimurium A1, selected for anticancer activity in vivo. The strain grows in tumor xenografts. In sharp contrast, normal tissue is cleared of these bacteria even in immunodeficient athymic mice. S. typhimurium A1 is auxotrophic (Leu/Arg-dependent) but apparently receives sufficient support from the neoplastic tissue to grow locally. Whether additional genetic lesions are present is not known. In in vitro infection, the GFP-expressing bacteria grew in the cytoplasm of PC-3 human prostate cancer cells and caused nuclear destruction. These effects were visualized in cells labeled with GFP in the nucleus and red fluorescent protein in the cytoplasm. In vivo, the bacteria caused tumor inhibition and regression of xenografts visualized by whole-body imaging. The bacteria, introduced i.v. or intratumorally, invaded and replicated intracellularly in PC-3 prostate cancer cells labeled with red fluorescent protein grafted into nude mice. By day 15, S. typhimurium A1 was undetectable in the liver, lung, spleen, and kidney, but it continued to proliferate in the PC-3 tumor, which stopped growing. When the bacteria were injected intratumorally, the tumor completely regressed by day 20. There were no obvious adverse effects on the host when the bacteria were injected by either route. The S. typhimurium A1 strain grew throughout the tumor, including viable malignant tissue. This result is in marked contrast to bacteria previously tried for cancer therapy that were confined to necrotic areas of the tumor, which may account, in part, for the strain's unique antitumor efficacy.
Collapse
Affiliation(s)
- Ming Zhao
- AntiCancer, 7917 Ostrow Street, San Diego, CA 92111-3604, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Molecular Imaging in Drug Discovery and Development: Potential and Limitations of Nonnuclear Methods. Mol Imaging Biol 2005. [DOI: 10.1007/s11307-005-0954-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
241
|
Gross S, Piwnica-Worms D. Monitoring proteasome activity in cellulo and in living animals by bioluminescent imaging: technical considerations for design and use of genetically encoded reporters. Methods Enzymol 2005; 399:512-30. [PMID: 16338379 DOI: 10.1016/s0076-6879(05)99035-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The ubiquitin-proteasome pathway is the central mediator of regulated proteolysis, instrumental for switching on and off a variety of signaling cascades. Deregulation of proteasomal activity or improper substrate recognition and processing by the ubiquitin-proteasome machinery may lead to cancer, stroke, chronic inflammation, and neurodegenerative diseases. Quantifying total and substrate-specific proteasome activity in intact cells and living animals would enable analysis in vivo of proteasomal regulation and facilitate the screening and validation of potential modulators of the proteasome or its substrates. We discuss examples of tetra-ubiquitin or IkappaBalpha fused to firefly luciferase as genetically encoded reporters for monitoring total and IkappaBalpha-specific proteasomal activity by bioluminescence imaging. Such technology enables repetitive, temporally resolved, and regionally targeted assessment of proteasomal activity in vivo.
Collapse
Affiliation(s)
- Shimon Gross
- Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
242
|
Rudin M, Rausch M, Stoeckli M. Molecular Imaging in Drug Discovery and Development: Potential and Limitations of Nonnuclear Methods. Mol Imaging Biol 2005; 7:5-13. [PMID: 15912270 DOI: 10.1007/s11307-004-0954-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Noninvasive conventional imaging methods are established technologies in modern drug discovery and development providing valuable morphological, physiological, and metabolic information to characterize disease phenotypes, to evaluate the efficacy of therapy and to identify and develop potential biomarkers for clinical drug evaluation. The development of target-specific or molecular imaging has added a new dimension: molecular events such as the target expression, the drug-target interaction, or the activation of signal transduction pathways can be studied in the intact organism with high spatial and temporal resolution. Molecular imaging is inherently a multimodality approach. In this article, we review the role of molecular imaging for drug discovery and development focusing on nonnuclear imaging methods, i.e., magnetic resonance imaging (MRI) and optical imaging techniques based on fluorescence and bioluminescence readouts. Examples discussed are direct visualization of target expression using target-specific ligands or reporter genes, pathway imaging, and cell-trafficking studies.
Collapse
Affiliation(s)
- Markus Rudin
- DTC/Analytical and Imaging Sciences, Novartis Institute for Biomedical Research, WSJ-386.2.02, CH-4002, Basel, Switzerland.
| | | | | |
Collapse
|
243
|
Abstract
This chapter describes a new cell biology where the behavior of individual cells can be visualized in the living animal. Previously it has been demonstrated that fluorescent proteins can be used for whole-body imaging of metastatic tumor growth, bacterial infection, and gene expression. An example of the new cell biology is dual-color fluorescence imaging using red fluorescent protein (RFP)-expressing tumors transplanted in green fluorescent protein (GFP)-expressing transgenic mice. These models show with great clarity the details of tumor-stroma interactions and especially tumor-induced angiogenesis, tumor-infiltrating lymphocytes, stromal fibroblasts, and macrophages. Another example is the color coding of cells with RFP or GFP such that both cell types can be simultaneously visualized in vivo. Stem cells can also be visualized and tracked in vivo. Mice in which the regulatory elements of the stem cell marker nestin drive GFP expression enable nascent vasculature to be visualized interacting with transplanted RFP-expressing cancer cells. Nestin-driven GFP expression can also be used to visualize hair follicle stem cells. Dual-color cells expressing GFP in the nucleus and RFP in the cytoplasm enable real-time visualization of nuclear-cytoplasm dynamics including cell cycle events and apoptosis. Highly elongated cancer cells in capillaries in living mice were observed within skin flaps. The migration velocities of the cancer cells in the capillaries were measured by capturing images of the dual-color fluorescent cells over time. The cells in the capillaries elongated to fit the width of these vessels. The use of the dual-color cancer cells differentially labeled in the cytoplasm and nucleus and associated fluorescent imaging provide a powerful tool to understand the mechanism of cancer cell migration and deformation in small vessels.
Collapse
|
244
|
Abstract
Significant progress has been made recently in the variety of ways that cancer can be non-invasively imaged in murine tumour models. The development and continued refinement of specialized hardware for an array of small animal imaging methodologies are only partly responsible. So too has been the development of new imaging techniques and materials that enable specific, highly sensitive and quantitative measurement of a wide range of tumour-related parameters. Included amongst these new materials are imaging probes that selectively accumulate in tumours, or that become activated by tumour-specific molecules in vivo. Other tumour imaging strategies have been developed that rely upon the detection of reporter transgene expression in vivo, and these too have made a significant impact on both the versatility and the specificity of tumour imaging in living mice. The biological implications resulting from these latest advances are presented here, with particular emphasis on those associated with MRI, PET, SPECT, BLI, and fluorescence-based imaging modalities. Taken together, these advances in tumour imaging are set to have a profound impact on our basic understanding of in vivo tumour biology and will radically alter the application of mouse tumour models in the laboratory.
Collapse
Affiliation(s)
- Scott K Lyons
- Oncology Department, Xenogen Corporation, Alameda, CA 94501, USA.
| |
Collapse
|
245
|
Stanziale SF, Stiles BM, Bhargava A, Kerns SA, Kalakonda N, Fong Y. Oncolytic herpes simplex virus-1 mutant expressing green fluorescent protein can detect and treat peritoneal cancer. Hum Gene Ther 2004; 15:609-18. [PMID: 15212719 DOI: 10.1089/104303404323142051] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
NV1066 is a herpes simplex virus-1 (HSV-1) oncolytic mutant that contains the gene for enhanced green fluorescent protein (EGFP). We sought to determine (1) whether NV1066 is effective against human peritoneal cancer, (2) whether EGFP is detectable in an animal model of gastric cancer, and (3) whether EGFP expression can be used to assess oncolytic therapy in a minimally invasive, laparoscopic system. The current study demonstrates that NV1066 is cytotoxic to OCUM human gastric cancer cells in vitro and in an in vivo model of disseminated peritoneal gastric cancer. In vitro this human gastric cancer cell line is sensitive to NV1066. Lysis occurs in a dose-dependent fashion, achieving near-complete lysis even at multiplicities of infection (MOIs) as low as 0.01 by 7 days. NV1066 also replicates within OCUM cells and induces expression of GFP in a dose-dependent manner. At MOIs of 0.01 to 1, EGFP expression is seen by flow cytometry in 100% of OCUM cells within 5 days after infection. NV1066 effectively treats OCUM carcinomatosis in an in vivo model. After intraperitoneal administration of NV1066, macroscopic tumor foci express EGFP by direct laparoscopy with the appropriate fluorescent filtering. Noncancerous organs are not infected and do not express EGFP. We conclude that NV1066 has significant oncolytic activity in vitro and in vivo and reliably induces EGFP expression in infected tumor cells. Furthermore, EGFP expression in intraperitoneal tumors can be visualized laparoscopically, allowing detection and localization of viral gene therapy.
Collapse
Affiliation(s)
- Stephen F Stanziale
- Department of Surgery, Hepatobiliary Division, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
246
|
Staton CA, Stribbling SM, Tazzyman S, Hughes R, Brown NJ, Lewis CE. Current methods for assaying angiogenesis in vitro and in vivo. Int J Exp Pathol 2004; 85:233-48. [PMID: 15379956 PMCID: PMC2517524 DOI: 10.1111/j.0959-9673.2004.00396.x] [Citation(s) in RCA: 245] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis, the development of new blood vessels from an existing vasculature, is essential in normal developmental processes and in numerous pathologies, including diabetic retinopathy, psoriasis and tumour growth and metastases. One of the problems faced by angiogenesis researchers has been the difficulty of finding suitable methods for assessing the effects of regulators of the angiogenic response. The ideal assay would be reliable, technically straightforward, easily quantifiable and, most importantly, physiologically relevant. Here, we review the advantages and limitations of the principal assays in use, including those for the proliferation, migration and differentiation of endothelial cells in vitro, vessel outgrowth from organ cultures and in vivo assays such as sponge implantation, corneal, chamber, zebrafish, chick chorioallantoic membrane (CAM) and tumour angiogenesis models.
Collapse
Affiliation(s)
- Carolyn A Staton
- Tumour Targeting Group University of Sheffield Medical School, Sheffield, UK
| | | | | | | | | | | |
Collapse
|
247
|
Abstract
Accurate and rapid detection of tumors is of great importance for interrogating the molecular basis of cancer pathogenesis, preventing the onset of complications, and implementing a tailored therapeutic regimen. In this era of molecular medicine, molecular probes that respond to, or target molecular processes are indispensable. Although numerous imaging modalities have been developed for visualizing pathologic conditions, the high sensitivity and relatively innocuous low energy radiation of optical imaging method makes it attractive for molecular imaging. While many human diseases have been studied successfully by using intrinsic optical properties of normal and pathologic tissues, molecular imaging of the expression of aberrant genes, proteins, and other pathophysiologic processes would be enhanced by the use of highly specific exogenous molecular beacons. This review focuses on the development of receptor-specific molecular probes for optical imaging of tumors. Particularly, bioconjugates of probes that absorb and fluoresce in the near infrared wavelengths between 750 and 900 nm will be reviewed.
Collapse
Affiliation(s)
- Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, 4525 Scot Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
248
|
Chen HH, Zhan X, Kumar A, Du X, Hammond H, Cheng L, Yang X. Detection of dual-gene expression in arteries using an optical imaging method. JOURNAL OF BIOMEDICAL OPTICS 2004; 9:1223-9. [PMID: 15568943 DOI: 10.1117/1.1803842] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
We evaluate the in vivo use of an optical imaging method to detect the vascular expression of green fluorescent protein (GFP) or red fluorescent protein (RFP), and to detect the simultaneous expression of GFP and RFP after transduction into arteries by a dual-promoter lentiviral vector driving their concurrent expression. This method involves using a charge-coupled device camera to detect fluorescence, a fiber optic probe to transmit light, and optical filters to distinguish each marker. In animal models, these vectors are locally delivered to target arteries, whereas the gene for a nonfluorescent cell-surface protein is transduced into contralateral arteries as the sham control. The images show distinct areas of bright fluorescence from GFP and RFP along the target arteries on excitation; no exogenous fluorescence is observed in the controls. Measured signal intensities from arteries transduced with the single- and dual-promoter vectors exceed the autofluorescence signal from the controls. Transgene expression of GFP and RFP in vivo is confirmed with confocal microscopy. We demonstrate the use of an optical imaging method to concurrently detect two distinct fluorescent proteins, potentially permitting the expression of multiple transgenes and their localization in the vasculature to be monitored.
Collapse
Affiliation(s)
- Hunter H Chen
- Johns Hopkins University, Department of Biomedical Engineering, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
249
|
Umeoka T, Kawashima T, Kagawa S, Teraishi F, Taki M, Nishizaki M, Kyo S, Nagai K, Urata Y, Tanaka N, Fujiwara T. Visualization of intrathoracically disseminated solid tumors in mice with optical imaging by telomerase-specific amplification of a transferred green fluorescent protein gene. Cancer Res 2004; 64:6259-65. [PMID: 15342413 DOI: 10.1158/0008-5472.can-04-1335] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Currently available methods for detection of tumors in vivo such as X-ray, computed tomography, and ultrasonography are noninvasive and have been well studied; the images, however, are not specific for tumors. Direct optical imaging of tumor cells in vivo that can clearly distinguish them from surrounding normal tissues may be clinically useful. Here, we describe a new approach to visualizing tumors whose fluorescence can be detected using tumor-specific replication-competent adenovirus (OBP-301, Telomelysin) in combination with Ad-GFP, a replication-deficient adenovirus expressing green fluorescent protein (GFP). Human telomerase reverse transcriptase is the catalytic subunit of telomerase, which is highly active in cancer cells but quiescent in most normal somatic cells. We constructed an adenovirus 5 vector in which the human telomerase reverse transcriptase promoter element drives expression of E1A and E1B genes linked with an internal ribosome entry site and showed that OBP-301 replicated efficiently in human cancer cells, but not in normal cells such as human fibroblasts. When the human lung and colon cancer cell lines were infected with Ad-GFP at a low multiplicity of infection, GFP expression could not be detected under a fluorescence microscope; in the presence of OBP-301, however, Ad-GFP replicated in these tumor cells and showed strong green signals. In contrast, coinfection with OBP-301 and Ad-GFP did not show any signals in normal cells such as fibroblasts and vascular endothelial cells. We also found that established subcutaneous tumors could be visualized after intratumoral injection of OBP-301 and Ad-GFP. A549 human lung tumors and SW620 human colon tumors transplanted into BALB/c nu/nu mice were intratumorally injected with 8 x 10(5) plaque-forming units of Ad-GFP in combination with 8 x 10(6) plaque-forming units of OBP-301. Within 3 days of treatment, the fluorescence of the expressed GFP became visible by a three-chip color cooled charged-coupled device camera in these tumors, whereas intratumoral injection of Ad-GFP alone could not induce GFP fluorescence. Moreover, intrathoracic administration of Ad-GFP and OBP-301 could visualize disseminated A549 tumor nodules in mice after intrathoracic implantation. Our results indicate that intratumoral or intrathoracic injection of Ad-GFP in combination with OBP-301 might be a useful diagnostic method that provides a foundation for future clinical application.
Collapse
Affiliation(s)
- Tatsuo Umeoka
- Division of Surgical Oncology, Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Abstract
Multi-modality, noninvasive in vivo imaging is increasingly being used in molecular-genetic studies and will soon become the standard approach for reporter gene imaging studies in small animals. The coupling of nuclear and optical reporter genes, as described here, represents only the beginning of a far wider application of this technology in the future. Optical imaging and optical reporter systems are cost-effective and time-efficient; they require less resources and space than PET or MRI, and are particularly well suited for imaging small animals, such as mice. Optical reporter systems are also very useful for the quantification and selection of transduced cells using FACS, and for performing in vitro assays to validate the function and sensitivity of constitutive and specific-inducible reporter systems. However, optical imaging techniques are limited by depth of light penetration and do not yet provide optimal quantitative or tomographic information. These issues are not limiting for PET- or MRI-based reporter systems, and PET- and MRI-based animal studies are more easily generalized to human applications. Many of the shortcomings of each modality alone can be overcome by the use of dual- or triple-modality reporter constructs that incorporate the opportunity for PET, fluorescence and bioluminescence imaging.
Collapse
Affiliation(s)
- Ronald G Blasberg
- Memorial Sloan Kettering Cancer Center, Nueurology and Radiology, 1275 York Ave, Box 52, New York, NY 10021, USA.
| |
Collapse
|