Hostname: page-component-76fb5796d-zzh7m Total loading time: 0 Render date: 2024-04-25T17:47:48.490Z Has data issue: false hasContentIssue false

Molecular imaging agents: impact on diagnosis and therapeutics in oncology

Published online by Cambridge University Press:  15 July 2010

Marc E. Seaman
Affiliation:
Department of Biomedical Engineering, University of Virginia, Box 800759 Health System, Charlottesville, VA 22908, USA.
Gianmarco Contino
Affiliation:
Department of Medicine, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA 02114, USA. Division of General Surgery, European Institute of Oncology and University of Milan, Milan, Italy.
Nabeel Bardeesy
Affiliation:
Department of Medicine, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA 02114, USA.
Kimberly A. Kelly*
Affiliation:
Department of Biomedical Engineering, University of Virginia, Box 800759 Health System, Charlottesville, VA 22908, USA.
*
*Corresponding author: Kimberly A. Kelly, Department of Biomedical Engineering, University of Virginia, Box 800759 Health System, Charlottesville, VA 22908, USA. E-mail: kak3x@eservices.virginia.edu

Abstract

Imaging has become a crucial tool in oncology throughout the course of disease detection and management, and is an integral part of clinical trials. Anatomical and functional imaging led the way, providing valuable information used in the diagnosis of disease, including data regarding the size and location of the tumour and on physiological processes such as blood flow and perfusion. As understanding of cancer pathogenesis has advanced through the identification of genetic, biochemical and cellular alterations in evolving tumours, emphasis has been put on developing methods to detect and serially monitor such alterations. This class of approaches is referred to as molecular imaging. Molecular imaging offers the potential for increasingly sensitive and specific visualisation and quantification of biological processes at the cellular and molecular level. These approaches have become established as essential tools for cancer research, early cancer detection and staging, and monitoring and predicting response to targeted therapies. Here, we discuss recent advances in the development of molecular imaging agents and their implementation in basic cancer research as well as in more rationalised approaches to cancer care.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2010

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Evans, P.M. (2008) Anatomical imaging for radiotherapy. Physics in Medicine and Biology 53, R151–191CrossRefGoogle ScholarPubMed
2Baka, S., Clamp, A.R. and Jayson, G.C. (2006) A review of the latest clinical compounds to inhibit VEGF in pathological angiogenesis. Expert Opinion on Therapeutic Targets 10, 867-876CrossRefGoogle ScholarPubMed
3Tang, Y. et al. (2005) In vivo assessment of RAS-dependent maintenance of tumor angiogenesis by real-time magnetic resonance imaging. Cancer Research 65, 8324-8330CrossRefGoogle ScholarPubMed
4Batchelor, T.T. et al. (2007) AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell 11, 83-95CrossRefGoogle ScholarPubMed
5Tong, R.T. et al. (2004) Vascular normalization by vascular endothelial growth factor receptor 2 blockade induces a pressure gradient across the vasculature and improves drug penetration in tumors. Cancer Research 64, 3731-3736CrossRefGoogle ScholarPubMed
6Mankoff, D.A. (2007) A definition of molecular imaging. Journal of Nuclear Medicine 48, 18N, 21NGoogle ScholarPubMed
7Naik, S., Moss, B.L. and Piwnica-Worms, D. (2008) Imaging cellular networks and protein-protein interactions in vivo. In Molecular Imaging in Oncology (1st edn) (Pomper, M.G. and Gelovani, J.G., eds), pp. 391-403, Informa Healthcare USA, Inc., New York City, NY, USACrossRefGoogle Scholar
8Donati, O.F. et al. (2010) 18F-FDG-PET and MRI in patients with malignancies of the liver and pancreas. Accuracy of retrospective multimodality image registration by using the CT-component of PET/CT. Nuklearmedizin 49, 106-114CrossRefGoogle Scholar
9Antoch, G. et al. (2004) Accuracy of whole-body dual-modality fluorine-18–2-fluoro-2-deoxy-D-glucose positron emission tomography and computed tomography (FDG-PET/CT) for tumor staging in solid tumors: comparison with CT and PET. Journal of Clinical Oncology 22, 4357-4368CrossRefGoogle ScholarPubMed
10Barwick, T. et al. (2010) Single photon emission computed tomography (SPECT)/computed tomography using iodine-123 in patients with differentiated thyroid cancer: additional value over whole body planar imaging and SPECT. European Journal of Endocrinology 162, 1131-1139CrossRefGoogle Scholar
11Mishra, A. et al. (2006) A new class of Gd-based DO3A-ethylamine-derived targeted contrast agents for MR and optical imaging. Bioconjugate Chemistry 17, 773-780CrossRefGoogle ScholarPubMed
12Huber, M.M. et al. (1998) Fluorescently detectable magnetic resonance imaging agents. Bioconjugate Chemistry 9, 242-249CrossRefGoogle ScholarPubMed
13Kelly, K.A., Reynolds, F. and Kristof, K.R. (2008) High-throughput screening for probe development. In Molecular Imaging in Oncology (1st edn) (Pomper, M.G. and Gelovani, J.G., eds), pp. 179-188, Informa Healthcare USA, Inc., New York City, NY, USACrossRefGoogle Scholar
14Warburg, O. (1956) On the origin of cancer cells. Science 123, 309-314CrossRefGoogle ScholarPubMed
15Taegtmeyer, H. and Dilsizian, V. (2008) Imaging myocardial metabolism and ischemic memory. Nature Clinical Practice Cardiovascular Medicine 5 Suppl 2, S42–48CrossRefGoogle Scholar
16Matsui, T. et al. (2009) Inflammatory cytokines and hypoxia contribute to 18F-FDG uptake by cells involved in pannus formation in rheumatoid arthritis. Journal of Nuclear Medicine 50, 920-926CrossRefGoogle ScholarPubMed
17Rubin, D.T. et al. (2009) Positron emission tomography (PET) used to image subclinical inflammation associated with ulcerative colitis (UC) in remission. Inflammatory Bowel Diseases 15, 750-755CrossRefGoogle ScholarPubMed
18Takuma, K., Kamisawa, T. and Itoi, T. (2010) Positive response to steroid therapy for autoimmune pancreatitis evaluated with fluorodeoxyglucose positron emission tomography. Clinical Gastroenterology and Hepatology 8, e54–55CrossRefGoogle ScholarPubMed
19Safran, M. et al. (2006) Mouse model for noninvasive imaging of HIF prolyl hydroxylase activity: assessment of an oral agent that stimulates erythropoietin production. Proceedings of the National Academy of Sciences of the United States of America 103, 105-110CrossRefGoogle ScholarPubMed
20Karin, M. et al. (2002) NF-kappaB in cancer: from innocent bystander to major culprit. Nature Reviews Cancer 2, 301-310CrossRefGoogle ScholarPubMed
21Li, Q. and Verma, I.M. (2002) NF-kappaB regulation in the immune system. Nature Reviews Immunology 2, 725-734CrossRefGoogle ScholarPubMed
22Aggarwal, B.B. (2004) Nuclear factor-kappaB: the enemy within. Cancer Cell 6, 203-208CrossRefGoogle Scholar
23Ghosh, S. and Karin, M. (2002) Missing pieces in the NF-kappaB puzzle. Cell 109 Suppl, S81-96CrossRefGoogle ScholarPubMed
24Karin, M. and Ben-Neriah, Y. (2000) Phosphorylation meets ubiquitination: the control of NF-[kappa]B activity. Annual Review of Immunology 18, 621-663CrossRefGoogle Scholar
25Hayden, M.S. and Ghosh, S. (2004) Signaling to NF-kappaB. Genes and Development 18, 2195-2224CrossRefGoogle ScholarPubMed
26Gross, S. and Piwnica-Worms, D. (2005) Real-time imaging of ligand-induced IKK activation in intact cells and in living mice. Nature Methods 2, 607-614CrossRefGoogle ScholarPubMed
27Shaner, N.C., Steinbach, P.A. and Tsien, R.Y. (2005) A guide to choosing fluorescent proteins. Nature Methods 2, 905-909CrossRefGoogle ScholarPubMed
28Li, I.T., Pham, E. and Truong, K. (2006) Protein biosensors based on the principle of fluorescence resonance energy transfer for monitoring cellular dynamics. Biotechnology Letters 28, 1971-1982CrossRefGoogle Scholar
29Takemoto, K. et al. (2003) Spatio-temporal activation of caspase revealed by indicator that is insensitive to environmental effects. Journal of Cell Biology 160, 235-243CrossRefGoogle ScholarPubMed
30Zhou, F. et al. (2010) Intravital Imaging of tumor apoptosis with FRET probes during tumor therapy. Molecular Imaging and Biology 12, 63-70CrossRefGoogle ScholarPubMed
31Blasberg, R.G. and Gelovani-Tjuvajev, J. (2002) In vivo molecular-genetic imaging. Journal of Cellular Biochemistry Suppl 39, 172-183CrossRefGoogle ScholarPubMed
32Tjuvajev, J.G. et al. (1996) Noninvasive imaging of herpes virus thymidine kinase gene transfer and expression: a potential method for monitoring clinical gene therapy. Cancer Research 56, 4087-4095Google ScholarPubMed
33Culver, K.W. et al. (1992) In vivo gene transfer with retroviral vector-producer cells for treatment of experimental brain tumors. Science 256, 1550-1552CrossRefGoogle ScholarPubMed
34Moolten, F.L. et al. (1990) Lymphoma regression induced by ganciclovir in mice bearing a herpes thymidine kinase transgene. Human Gene Therapy 1, 125-134CrossRefGoogle ScholarPubMed
35Ram, Z. et al. (1997) Therapy of malignant brain tumors by intratumoral implantation of retroviral vector-producing cells. Nature Medicine 3, 1354-1361CrossRefGoogle ScholarPubMed
36Alauddin, M.M. et al. (2007) In vivo evaluation of 2'-deoxy-2'-[(18)F]fluoro-5-iodo-1-beta-D-arabinofuranosyluracil ([18F]FIAU) and 2'-deoxy-2'-[18F]fluoro-5-ethyl-1-beta-D-arabinofuranosyluracil ([18F]FEAU) as markers for suicide gene expression. European Journal of Nuclear Medicine and Molecular Imaging 34, 822-829CrossRefGoogle ScholarPubMed
37Bengtsson, N.E. et al. (2010) lacZ as a genetic reporter for real-time MRI. Magnetic Resonance in Medicine 63, 745-753CrossRefGoogle ScholarPubMed
38Heuermann, K. and Cosgrove, J. (2001) S-Gal: an autoclavable dye for color selection of cloned DNA inserts. Biotechniques 30, 1142-1147CrossRefGoogle ScholarPubMed
39American Cancer Society (2007) Cancer Facts & Figures 2007, American Cancer Society, Atlanta, GA, USA. http://www.cancer.org/docroot/STT/content/stt_1x_cancer_facts__figures_2007.aspGoogle Scholar
40Wallace, M.B. and Keisslich, R. (2010) Advances in endoscopic imaging of colorectal neoplasia. Gastroenterology 138, 2140-2150CrossRefGoogle ScholarPubMed
41Rex, D.K. et al. (1997) Colonoscopic miss rates of adenomas determined by back-to-back colonoscopies. Gastroenterology 112, 24-28CrossRefGoogle ScholarPubMed
42Rembacken, B.J. et al. (2000) Flat and depressed colonic neoplasms: a prospective study of 1000 colonoscopies in the UK. Lancet 355, 1211-1214CrossRefGoogle ScholarPubMed
43Krok, K.L. and Lichtenstein, G.R. (2004) Colorectal cancer in inflammatory bowel disease. Current Opinion in Gastroenterology 20, 43-48CrossRefGoogle ScholarPubMed
44Sloane, B.F. et al. (2005) Cathepsin B and tumor proteolysis: contribution of the tumor microenvironment. Seminars in Cancer Biology 15, 149-157CrossRefGoogle ScholarPubMed
45Joyce, J.A. et al. (2004) Cathepsin cysteine proteases are effectors of invasive growth and angiogenesis during multistage tumorigenesis. Cancer Cell 5, 443-453CrossRefGoogle ScholarPubMed
46Gounaris, E. et al. (2008) Live imaging of cysteine-cathepsin activity reveals dynamics of focal inflammation, angiogenesis, and polyp growth. PLoS One 3, e2916CrossRefGoogle ScholarPubMed
47Goetz, M. et al. (2010) In vivo molecular imaging of colorectal cancer with confocal endomicroscopy by targeting epidermal growth factor receptor. Gastroenterology 138, 435-446CrossRefGoogle ScholarPubMed
48Keller, R. et al. (2002) Fluorescence endoscopy using a fluorescein-labeled monoclonal antibody against carcinoembryonic antigen in patients with colorectal carcinoma and adenoma. Endoscopy 34, 801-807CrossRefGoogle Scholar
49Kelly, K.A. and Jones, D.A. (2003) Isolation of a colon tumor specific binding peptide using phage display selection. Neoplasia 5, 437-444CrossRefGoogle ScholarPubMed
50Kelly, K. et al. (2004) Detection of invasive colon cancer using a novel, targeted, library-derived fluorescent peptide. Cancer Research 64, 6247-6251CrossRefGoogle ScholarPubMed
51Hsiung, P.L. et al. (2008) Detection of colonic dysplasia in vivo using a targeted heptapeptide and confocal microendoscopy. Nature Medicine 14, 454-458CrossRefGoogle ScholarPubMed
52Ross, J.S. and Fletcher, J.A. (1998) The HER-2/neu oncogene in breast cancer: prognostic factor, predictive factor, and target for therapy. Oncologist 3, 237-252CrossRefGoogle ScholarPubMed
53Weiner, L.M. et al. (1995) Phase I trial of 2B1, a bispecific monoclonal antibody targeting c-erbB-2 and Fc gamma RIII. Cancer Research 55, 4586-4593Google ScholarPubMed
54Disis, M.L. et al. (1999) Generation of immunity to the HER-2/neu oncogenic protein in patients with breast and ovarian cancer using a peptide-based vaccine. Clinical Cancer Research 5, 1289-1297Google Scholar
55Harris, J.D. et al. (1994) Gene therapy for cancer using tumour-specific prodrug activation. Gene Therapy 1, 170-175Google ScholarPubMed
56Baselga, J. et al. (1998) Recombinant humanized anti-HER2 antibody (Herceptin) enhances the antitumor activity of paclitaxel and doxorubicin against HER2/neu overexpressing human breast cancer xenografts. Cancer Research 58, 2825-2831Google ScholarPubMed
57Cobleigh, M.A. et al. (1999) Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. Journal of Clinical Oncology 17, 2639-2648CrossRefGoogle ScholarPubMed
58Seidman, A. et al. (2002) Cardiac dysfunction in the trastuzumab clinical trials experience. Journal of Clinical Oncology 20, 1215-1221CrossRefGoogle ScholarPubMed
59Kumar, S.R., Quinn, T.P. and Deutscher, S.L. (2007) Evaluation of an 111In-radiolabeled peptide as a targeting and imaging agent for ErbB-2 receptor expressing breast carcinomas. Clinical Cancer Research 13, 6070-6079CrossRefGoogle ScholarPubMed
60Li, D. et al. (2004) Pancreatic cancer. Lancet 363, 1049-1057CrossRefGoogle ScholarPubMed
61Jemal, A. et al. (2003) Cancer statistics, 2003. CA: A Cancer Journal for Clinicians 53, 5-26Google ScholarPubMed
62Gillen, S. et al. (2010) Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and meta-analysis of response and resection percentages. PLoS Medicine 7, e1000267CrossRefGoogle ScholarPubMed
63Kelly, K.A. et al. (2008) Targeted nanoparticles for imaging incipient pancreatic ductal adenocarcinoma. PLoS Medicine 5, e85CrossRefGoogle ScholarPubMed
64Bardeesy, N. et al. (2006) Both p16(Ink4a) and the p19(Arf)-p53 pathway constrain progression of pancreatic adenocarcinoma in the mouse. Proceedings of the National Academy of Sciences of the United States of America 103, 5947-5952CrossRefGoogle ScholarPubMed
65Wilson, J.M.G. and Jungner, G. (1968) Principles and Practice of Screening for Disease. Public Health Papers 34, 1-163Google Scholar
66Andermann, A. et al. (2008) Revisiting Wilson and Jungner in the genomic age: a review of screening criteria over the past 40 years. Bulletin of the World Health Organization 86, 317-319CrossRefGoogle Scholar
67Desar, I.M. et al. (2009) Beyond RECIST: molecular and functional imaging techniques for evaluation of response to targeted therapy. Cancer Treatment Reviews 35, 309-321CrossRefGoogle ScholarPubMed
68Jackson, D.B. (2009) Molecular perspectives on the non-responder phenomenon. Drug Discovery Today 14, 373-379CrossRefGoogle ScholarPubMed
69Minchinton, A.I. and Tannock, I.F. (2006) Drug penetration in solid tumours. Nature Reviews Cancer 6, 583-592CrossRefGoogle ScholarPubMed
70Lupold, S.E. et al. (2002) Identification and characterization of nuclease-stabilized RNA molecules that bind human prostate cancer cells via the prostate-specific membrane antigen. Cancer Research 62, 4029-4033Google ScholarPubMed
71Sarker, D. and Workman, P. (2007) Pharmacodynamic biomarkers for molecular cancer therapeutics. Advances in Cancer Research 96, 213-268CrossRefGoogle ScholarPubMed
72Thomas, G.V. et al. (2006) Hypoxia-inducible factor determines sensitivity to inhibitors of mTOR in kidney cancer. Nature Medicine 12, 122-127CrossRefGoogle ScholarPubMed
73Man, T.K. et al. (2005) Expression profiles of osteosarcoma that can predict response to chemotherapy. Cancer Research 65, 8142-8150CrossRefGoogle ScholarPubMed
74Cortesi, L. et al. (2009) Identification of protein clusters predictive of response to chemotherapy in breast cancer patients. Journal of Proteome Research 8, 4916-4933CrossRefGoogle ScholarPubMed
75Kuukasjarvi, T. et al. (1996) Loss of estrogen receptor in recurrent breast cancer is associated with poor response to endocrine therapy. Journal of Clinical Oncology 14, 2584-2589CrossRefGoogle ScholarPubMed
76Sundararajan, L. et al. (2007) 18F-Fluoroestradiol. Seminars in Nuclear Medicine 37, 470-476CrossRefGoogle ScholarPubMed
77Engelman, J.A. et al. (2008) Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nature Medicine 14, 1351-1356CrossRefGoogle ScholarPubMed
78Lee, T.-Y. et al. (2008) Physiological and functional imaging with CT: techniques and applications. In Molecular Imaging in Oncology (1st edn) (Pomper, M.G. and Gelovani, J.G., eds), pp. 139-156, Informa Healthcare USA, Inc., New York City, NY, USACrossRefGoogle Scholar
79Dose Schwarz, J. et al. (2005) Early prediction of response to chemotherapy in metastatic breast cancer using sequential 18F-FDG PET. Journal of Nuclear Medicine 46, 1144-1150Google ScholarPubMed
80Sohn, H.J. et al. (2008) [18F]Fluorothymidine positron emission tomography before and 7 days after gefitinib treatment predicts response in patients with advanced adenocarcinoma of the lung. Clinical Cancer Research 14, 7423-7429CrossRefGoogle Scholar
81Jain, R.K. (2002) Tumor angiogenesis and accessibility: role of vascular endothelial growth factor. Seminars in Oncology 29, 3-9CrossRefGoogle ScholarPubMed
82Brooks, P.C., Clark, R.A. and Cheresh, D.A. (1994) Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science 264, 569-571CrossRefGoogle ScholarPubMed
83Beer, A.J. et al. (2006) PET-based human dosimetry of 18F-galacto-RGD, a new radiotracer for imaging alpha v beta3 expression. Journal of Nuclear Medicine 47, 763-769Google ScholarPubMed
84Bach-Gansmo, T. et al. (2006) Integrin receptor imaging of breast cancer: a proof-of-concept study to evaluate 99mTc-NC100692. Journal of Nuclear Medicine 47, 1434-1439Google ScholarPubMed
85Dayton, P.A. and Borden, M.A. (2008) Ultrasound instrumentation and techniques. In Molecular Imaging in Oncology (1st edn) (Pomper, M.G. and Gelovani, J.G., eds), pp. 109-119, Informa Healthcare USA, Inc., New York City, NY, USACrossRefGoogle Scholar
86Wahl, R.L. et al. (2009) From RECIST to PERCIST: evolving considerations for PET response criteria in solid tumors. Journal of Nuclear Medicine 50 Suppl 1, 122S-150SCrossRefGoogle Scholar
87Frangioni, J.V. (2006) Translating in vivo diagnostics into clinical reality. Nature Biotechnology 24, 909-913CrossRefGoogle ScholarPubMed
88Wagenaar, D.J. and Patt, B.E. (2008) Multimodality imaging instrumentation and techniques. In Molecular Imaging in Oncology (1st edn) (Pomper, M.G. and Gelovani, J.G., eds), pp. 157-168, Informa Healthcare USA, Inc., New York City, NY, USACrossRefGoogle Scholar
Pomper, M.G. and Gelovani, J.G., eds (2008) Molecular Imaging in Oncology, Informa Healthcare USA, Inc., New York, NY, USACrossRefGoogle Scholar
Frangioni, J.V. (2006) Translating in vivo diagnostics into clinical reality. Nature Biotechnology 24, 909-913CrossRefGoogle ScholarPubMed
Pomper, M.G. and Gelovani, J.G., eds (2008) Molecular Imaging in Oncology, Informa Healthcare USA, Inc., New York, NY, USACrossRefGoogle Scholar
Frangioni, J.V. (2006) Translating in vivo diagnostics into clinical reality. Nature Biotechnology 24, 909-913CrossRefGoogle ScholarPubMed