Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-xfwgj Total loading time: 0 Render date: 2024-07-05T05:20:29.039Z Has data issue: false hasContentIssue false

46 - Molecular Imaging and Metastasis

from PART II - CLINICAL RESEARCH

Published online by Cambridge University Press:  05 June 2012

Yufang Hu
Affiliation:
University of California at Los Angeles, United States
Mai Johnson
Affiliation:
University of California at Los Angeles, United States
Frederic Pouliot
Affiliation:
University of California at Los Angeles, United States
Lily Wu
Affiliation:
University of California at Los Angeles, United States
David Lyden
Affiliation:
Weill Cornell Medical College, New York
Danny R. Welch
Affiliation:
Weill Cornell Medical College, New York
Bethan Psaila
Affiliation:
Imperial College of Medicine, London
Get access

Summary

With the advancement in modern genomic and proteomic technologies in the past decade, knowledge of the molecular and cellular mechanisms of cancer initiation and progression is expanding at an unprecedented rate. A prudent approach for clinicians and scientists would be to extract salient information and apply it to address significant challenges in the current practices of cancer management. An important issue is how best to query the molecular and physiological information relevant to cancer in patients. Molecular imaging is a particular useful technology in the pursuit of this quest, as it allows the visualization of critical molecular signaling pathways in action in living subjects, in a noninvasive and longitudinal manner. Metastasis, manifested often in the late stages of cancer (although most work today supports metastasis as an earlier event than previously recognized), is the main cause of mortality in patients with solid tumors. To be able to prevent or control metastasis is considered one of most significant challenges in clinical oncology.

Whole-body in vivo molecular imaging is ideally suited to assess the very complex process of metastasis, in which the location(s) and magnitude of disseminated lesions are changing in time. For cancer metastasis, the common imaging modalities employed for repetitive, noninvasive imaging include positron emission tomography (PET), computed tomography (CT), single photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), and optical imaging by bioluminescence (e.g., firefly luciferase [FL or Luc]) or fluorescence (e.g., green fluorescent protein [GFP]).

Type
Chapter
Information
Cancer Metastasis
Biologic Basis and Therapeutics
, pp. 516 - 537
Publisher: Cambridge University Press
Print publication year: 2011

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

Weber, WA (2006) Positron emission tomography as an imaging biomarker. J Clin Oncol. 24(20): 3282–3292.CrossRefGoogle ScholarPubMed
Massoud, TF, Gambhir, SS (2003) Molecular imaging in living subjects: seeing fundamental biological processes in a new light. Genes Dev. 17: 545–580.CrossRefGoogle Scholar
Weissleder, R, Pittet, MJ (2008) Imaging in the era of molecular oncology. Nature. 452: 580–589.CrossRefGoogle ScholarPubMed
Gambhir, S S, Yaghoubi, SS eds. (2010) Molecular Imaging with Reporter Genes. New York: Cambridge University Press.CrossRefGoogle Scholar
Liang, Q, Gotts, J, Satyamurthy, N et al. (2002) Noninvasive, repetitive, quantitative measurement of gene expression from a bicistronic message by positron emission tomography, following gene transfer with adenovirus. Mol Ther. 6: 73–82.CrossRefGoogle ScholarPubMed
Sun, X, Annala, AJ, Yaghoubi, SS et al. (2001) Quantitative imaging of gene induction in living animals. Gene Ther. 8: 1572–1579.CrossRefGoogle ScholarPubMed
Ponomarev, V, Doubrovin, M, Serganova, I et al. (2003) Cytoplasmically retargeted HSV1-tk/GFP reporter gene mutants for optimization of noninvasive molecular-genetic imaging. Neoplasia. 5: 245–254.CrossRefGoogle ScholarPubMed
Ray, P, Tsien, R, Gambhir, SS (2007) Construction and validation of improved triple fusion reporter gene vectors for molecular imaging of living subjects. Cancer Res. 67: 3085–3093.CrossRefGoogle ScholarPubMed
Paulmurugan, R, Umezawa, Y, Gambhir, SS (2002) Noninvasive imaging of protein-protein interactions in living subjects by using reporter protein complementation and reconstitution strategies. Proc Natl Acad Sci USA. 99: 15608–15613.CrossRefGoogle ScholarPubMed
Johnsson, N, Varshavsky, A (1994) Split ubiquitin as a sensor of protein interactions in vivo. Proc Natl Acad Sci USA. 91: 10340–10344.CrossRefGoogle ScholarPubMed
Pelletier, JN, Campbell-Valois, FX, Michnick, SW (1998) Oligomerization domain-directed reassembly of active dihydrofolate reductase from rationally designed fragments. Proc Natl Acad Sci USA. 95(21): 12141.CrossRefGoogle ScholarPubMed
Kaihara, A, Kawai, Y, Sato, M, Ozawa, T, Umezawa, Y (2003) Locating a protein-protein interaction in living cells via split Renilla luciferase complementation. Anal Chem. 75: 4176–4181.CrossRefGoogle ScholarPubMed
Luker, KE, Smith, MC, Luker, GD et al. (2004) Kinetics of regulated protein-protein interactions revealed with firefly luciferase complementation imaging in cells and living animals. Proc Natl Acad Sci USA. 101: 12288–12293.CrossRefGoogle ScholarPubMed
Luster, AD (1998) Chemokines – chemotactic cytokines that mediate inflammation. N Engl J Med. 338(7): 436–445.CrossRefGoogle ScholarPubMed
Christopher, MJ, Liu, F, Hilton, MJ, Long, F, Link, DC (2009) Suppression of CXCL12 production by bone marrow osteoblasts is a common and critical pathway for cytokine-induced mobilization. Blood. 114: 1331–1339.CrossRefGoogle ScholarPubMed
Muller, A, Homey, B, Soto, H et al. (2001) Involvement of chemokine receptors in breast cancer metastasis. Nature. 410: 50–56.CrossRefGoogle ScholarPubMed
Cabioglu, N, Sahin, AA, Morandi, P et al. (2009) Chemokine receptors in advanced breast cancer: differential expression in metastatic disease sites with diagnostic and therapeutic implications. Ann Oncol. 20: 1013–1019.CrossRefGoogle ScholarPubMed
Salvucci, O, Bouchard, A, Baccarelli, A et al. (2006) The role of CXCR4 receptor expression in breast cancer: a large tissue microarray study. Breast Cancer Res Treat. 97: 275–283.CrossRefGoogle ScholarPubMed
Luker, KE, Gupta, M, Luker, GD (2008) Imaging CXCR4 signaling with firefly luciferase complementation. Anal Chem. 80: 5565–5573.CrossRefGoogle ScholarPubMed
Balabanian, K, Levoye, A, Klemm, L et al. (2008) Leukocyte analysis from WHIM syndrome patients reveals a pivotal role for GRK3 in CXCR4 signaling. J Clin Invest. 118: 1074–1084.Google ScholarPubMed
Massoud, TF, Paulmurugan, R, De, A, Ray, P, Gambhir, SS (2007) Reporter gene imaging of protein-protein interactions in living subjects. Curr Opin Biotechnol. 18: 31–37.CrossRefGoogle ScholarPubMed
Iyer, M, Wu, L, Carey, M, Wang, Y, Smallwood, A, Gambhir, SS (2001) Two-step transcriptional amplification as a method for imaging reporter gene expression using weak promoters. Proc Natl Acad Sci USA. 98: 14595–14600.CrossRefGoogle ScholarPubMed
Johnson, M, Sato, M, Burton, J, Gambhir, SS, Carey, M, Wu, L (2005) Micro-PET/CT monitoring of herpes thymidine kinase suicide gene therapy in a prostate cancer xenograft: the advantage of a cell-specific transcriptional targeting approach. Mol Imaging. 4: 463–472.CrossRefGoogle Scholar
Ray, S, Paulmurugan, R, Patel, MR et al. (2008) Noninvasive imaging of therapeutic gene expression using a bidirectional transcriptional amplification strategy. Mol Ther. 16: 1848–1856.CrossRefGoogle ScholarPubMed
Huyn, ST, Burton, JB, Sato, M, Carey, M, Gambhir, SS, Wu, L (2009) A potent, imaging adenoviral vector driven by the cancer-selective mucin-1 promoter that targets breast cancer metastasis. Clin Cancer Res. 15: 3126–34.CrossRefGoogle ScholarPubMed
Burton, JB, Johnson, M, Sato, M et al. (2008) Adenovirus-mediated gene expression imaging to directly detect sentinel lymph node metastasis of prostate cancer. Nat Med. 14: 882–888.CrossRefGoogle ScholarPubMed
Adams, JY, Johnson, M, Sato, M et al. (2002) Visualization of advanced human prostate cancer lesions in living mice by a targeted gene transfer vector and optical imaging. Nat Med. 8: 891–897.CrossRefGoogle ScholarPubMed
Penuelas, I, Mazzolini, G, Boan, JF et al. (2005) Positron emission tomography imaging of adenoviral-mediated transgene expression in liver cancer patients. Gastroenterology. 128: 1787–1795.CrossRefGoogle ScholarPubMed
Barton, KN, Stricker, H, Brown, SL et al. (2008) Phase I study of noninvasive imaging of adenovirus-mediated gene expression in the human prostate. Mol Ther. 16: 1761–1769.CrossRefGoogle ScholarPubMed
Harisinghani, MG, Barentsz, J, Hahn, PF et al. (2003) Noninvasive detection of clinically occult lymph-node metastases in prostate cancer. N Engl J Med. 348: 2491–2499.CrossRefGoogle ScholarPubMed
Vassallo, P, Matei, C, Heston, WD, McLachlan, SJ, Koutcher, JA, Castellino, RA (1994) AMI-227-enhanced MR lymphography: usefulness for differentiating reactive from tumor-bearing lymph nodes. Radiology. 193: 501–506.CrossRefGoogle ScholarPubMed
Kim, S, Lim, YT, Soltesz, EG et al. (2004) Near-infrared fluorescent type II quantum dots for sentinel lymph node mapping. Nat Biotechnol. 22:93–97.CrossRefGoogle ScholarPubMed
Tanaka, E, Choi, HS, Fujii, H, Bawendi, MG, Frangioni, JV (2006) Image-guided oncologic surgery using invisible light: completed pre-clinical development for sentinel lymph node mapping. Ann Surg Oncol. 13: 1671–1681.CrossRefGoogle ScholarPubMed
Gao, X, Cui, Y, Levenson, RM, Chung, LW, Nie, S (2004) In vivo cancer targeting and imaging with semiconductor quantum dots. Nat Biotechnol. 22: 969–976.CrossRefGoogle ScholarPubMed
Morton, DL, Wen, DR, Wong, JH et al. (1992) Technical details of intraoperative lymphatic mapping for early-stage melanoma. Arch Surg. 127: 392–399.CrossRefGoogle ScholarPubMed
Keshtgar, MRS, Baum, M (2001) Axillary dissection over the years: Where to from here?World J Surg. 25: 761–766.CrossRefGoogle Scholar
Canavese, G, Catturich, A, Vecchio, C et al. (1998) Prognostic role of lymph-node level involvement in patients undergoing axillary dissection for breast cancer. Eur J Surg Oncol. 24: 104–109.CrossRefGoogle ScholarPubMed
Werner, JA, Dunne, AA, Ramaswamy, A et al. (2004) The sentinel node concept in head and neck cancer: solution for the controversies in the N0 neck? Head Neck. 26: 603–611.CrossRefGoogle ScholarPubMed
Chakera, AH, Drzewiecki, KT, Ingvar, C, Steiniche, T, Hesse, B (2006) Sentinel node imaging. Curr Med Imaging Rev. 2(3): 341–346.CrossRefGoogle Scholar
Alex, JC, Weaver, DL, Fairbank, JT, Rankin, BS, Krag, DN (1993) Gamma-probe-guided lymph node localization in malignant melanoma. Surg Oncol. 2: 303–308.CrossRefGoogle ScholarPubMed
Krag, DN, Weaver, DL, Alex, JC, Fairbank, JT (1993) Surgical resection and radiolocalization of the sentinel lymph node in breast cancer using a gamma probe. Surg Oncol. 2: 335–339; discussion 340.CrossRefGoogle ScholarPubMed
Roca, I, Caresia, AP, Gil-Moreno, A et al. (2005) Usefulness of sentinel lymph node detection in early stages of cervical cancer. Eur J Nucl Med Mol Imaging. 32: 1210–1216.CrossRefGoogle ScholarPubMed
Saha, S, Dan, AG, Viehl, CT, Zuber, M, Wiese, D (2005) Sentinel lymph node mapping in colon and rectal cancer: its impact on staging, limitations, and pitfalls. Cancer Treat Res. 127: 105–122.CrossRefGoogle ScholarPubMed
Sherif, A, Torre, M, Malmstrom, PU, Thorn, M (2001) Lymphatic mapping and detection of sentinel nodes in patients with bladder cancer. J Urol. 166: 812–815.CrossRefGoogle ScholarPubMed
Wawroschek, F, Vogt, H, Wengenmair, H et al. (2003) Prostate lymphoscintigraphy and radio-guided surgery for sentinel lymph node identification in prostate cancer. Technique and results of the first 350 cases. Urol Int. 70: 303–310.CrossRefGoogle ScholarPubMed
Ikomi, F, Hanna, GK, Schmid-Schonbein, GW (1999) Size- and surface-dependent uptake of colloid particles into the lymphatic system. Lymphology. 32: 90–102.Google ScholarPubMed
Ikomi, F, Hanna, GK, Schmid-Schonbein, GW (1995) Mechanism of colloidal particle uptake into the lymphatic system: basic study with percutaneous lymphography. Radiology. 196: 107–113.CrossRefGoogle ScholarPubMed
Szuba, A, Shin, SW, Strauss, W, Rockson, S (2003) The third circulation: radionuclide lymphoscintigraphy in the evaluation of lymphedema. Radionuclide Lymphoscintigraphy. 44: 43–57.Google ScholarPubMed
Islam, T, Harisinghani, MG (2009) Overview of nanoparticle use in cancer imaging. Cancer Biomark. 5: 61–67.CrossRefGoogle ScholarPubMed
Hricak, H, Choyke, PL, Eberhardt, SC, Leibel, SA, Scardino, PT (2007) Imaging prostate cancer: a multidisciplinary perspective. Radiology. 243: 28–53.CrossRefGoogle ScholarPubMed
Sharma, R, Wang, W, Rasmussen, JC et al. (2007) Quantitative imaging of lymph function. Am J Physiol Heart Circ Physiol. 292: H3109–H3118.CrossRefGoogle ScholarPubMed
Weissleder, R, Ntziachristos, V (2003) Shedding light onto live molecular targets. Nat Med. 9: 123–128.CrossRefGoogle ScholarPubMed
Kobayashi, H, Hama, Y, Koyama, Y et al. (2007) Simultaneous multicolor imaging of five different lymphatic basins using quantum dots. Nano Lett. 7: 1711–1716.CrossRefGoogle ScholarPubMed
Frangioni, JV (2003) In vivo near-infrared fluorescence imaging. Curr Opin Chem Biol. 7: 626–634.CrossRefGoogle ScholarPubMed
Michalet, X, Pinaud, FF, Bentolila, et al. (2005) Quantum dots for live cells, in vivo imaging, and diagnostics. Science. 307: 538–544.CrossRefGoogle ScholarPubMed
Stockwin, LH, Holmes, S (2003) The role of therapeutic antibodies in drug discovery. Biochem Soc Trans. 31: 433–436.CrossRefGoogle ScholarPubMed
Boswell, CA, Brechbiel, MW (2007) Development of radioimmunotherapeutic and diagnostic antibodies: an inside-out view. Nucl Med Biol. 34: 757–778.CrossRefGoogle Scholar
Wu, AM, Olafsen, T (2008) Antibodies for molecular imaging of cancer. Cancer J. 14: 191–197.CrossRefGoogle Scholar
Machac, J, Krynyckyi, B, Kim, C (2002) Peptide and antibody imaging in lung cancer. Semin Nucl Med. 32: 276–292.CrossRefGoogle ScholarPubMed
Wu, AM (2009) Antibodies and antimatter: the resurgence of immuno-PET. J Nucl Med. 50: 2–5.CrossRefGoogle ScholarPubMed
Gold, P, Freedman, SO (1965) Demonstration of tumor-specific antigens in human colonic carcinomata by immunological tolerance and absorption techniques. J Exp Med. 121: 439–462.CrossRefGoogle ScholarPubMed
Thomson, DM, Krupey, J, Freedman, SO, Gold, P (1969) The radioimmunoassay of circulating carcinoembryonic antigen of the human digestive system. Proc Natl Acad Sci USA. 64: 161–167.CrossRefGoogle ScholarPubMed
Duffy, MJ (2001) Carcinoembryonic antigen as a marker for colorectal cancer: is it clinically useful?Clin Chem 47: 624–630.Google ScholarPubMed
Goldstein, MJ, Mitchell, EP (2005) Carcinoembryonic antigen in the staging and follow-up of patients with colorectal cancer. Cancer Invest. 23: 338–351.CrossRefGoogle ScholarPubMed
Locker, GY, Hamilton, S, Harris, J et al. (2006) ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol. 24: 5313–5327.CrossRefGoogle ScholarPubMed
Libutti, SK, Alexander, HR Jr, Choyke, P et al. (2001) A prospective study of 2-[18F] fluoro-2-deoxy-D-glucose/positron emission tomography scan, 99mTc-labeled arcitumomab (CEA-scan), and blind second-look laparotomy for detecting colon cancer recurrence in patients with increasing carcinoembryonic antigen levels. Ann Surg Oncol. 8: 779–786.CrossRefGoogle Scholar
Willkomm, P, Bender, H, Bangard, M, Decker, P, Grunwald, F, Biersack, HJ (2000) FDG PET and immunoscintigraphy with 99mTc-labeled antibody fragments for detection of the recurrence of colorectal carcinoma. J Nucl Med 41: 1657–1663.Google ScholarPubMed
Hong, H, Zhang, Y, Sun, J, Cai, W (2009) Positron emission tomography imaging of prostate cancer. Amino Acids 39: 11–17.CrossRefGoogle ScholarPubMed
Bander, NH (2006) Technology insight: monoclonal antibody imaging of prostate cancer. Nat Clin Pract Urol. 3: 216–225.CrossRefGoogle ScholarPubMed
Wright, GL Jr, Grob, BM, Haley, C et al. (1996) Upregulation of prostate-specific membrane antigen after androgen-deprivation therapy. Urology. 48: 326–334.CrossRefGoogle ScholarPubMed
Bostwick, DG, Pacelli, A, Blute, M, Roche, P, Murphy, GP (1998) Prostate specific membrane antigen expression in prostatic intraepithelial neoplasia and adenocarcinoma: a study of 184 cases. Cancer. 82: 2256–2261.3.0.CO;2-S>CrossRefGoogle ScholarPubMed
Sweat, SD, Pacelli, A, Murphy, GP, Bostwick, DG (1998) Prostate-specific membrane antigen expression is greatest in prostate adenocarcinoma and lymph node metastases. Urology. 52: 637–640.CrossRefGoogle ScholarPubMed
Perk, LR, Stigter-van Walsum, M, Visser, GW et al. (2008) Quantitative PET imaging of Met-expressing human cancer xenografts with 89Zr-labelled monoclonal antibody DN30. Eur J Nucl Med Mol Imaging. 35: 1857–1867.CrossRefGoogle ScholarPubMed
Boccaccio, C, Comoglio, PM (2006) Invasive growth: a MET-driven genetic programme for cancer and stem cells. Nat Rev Cancer. 6: 637–645.CrossRefGoogle ScholarPubMed
Börjesson, PK, Jauw, YW, Boellaard, R et al (2006). Performance of immuno-positron emission tomography with zirconium-89-labeled chimeric monoclonal antibody U36 in the detection of lymph node metastases in head and neck cancer patients. Clin Cancer Res. 12: 2133–2140.CrossRefGoogle ScholarPubMed
Kenanova, V, Olafsen, T, Williams, et al. (2007) Radioiodinated versus radiometal-labeled anti-carcinoembryonic antigen single-chain Fv-Fc antibody fragments: optimal pharmacokinetics for therapy. Cancer Res. 67: 718–726.CrossRefGoogle ScholarPubMed
Olafsen, T, Kenanova, VE, Sundaresan, G et al. (2005) Optimizing radiolabeled engineered anti-p185HER2 antibody fragments for in vivo imaging. Cancer Res. 65: 5907–5916.CrossRefGoogle ScholarPubMed
Wu, AM, Senter, PD (2005) Arming antibodies: prospects and challenges for immunoconjugates. Nat Biotechnol. 23: 1137–1146.CrossRefGoogle ScholarPubMed
Wong, JY, Chu, DZ, Williams, et al. (2004) Pilot trial evaluating an 123I-labeled 80-kilodalton engineered anticarcinoembryonic antigen antibody fragment (cT84.66 minibody) in patients with colorectal cancer. Clin Cancer Res 10: 5014–5021.CrossRefGoogle ScholarPubMed
Hu, S, Shively, L, Raubitschek, A et al. (1996) Minibody: a novel engineered anti-carcinoembryonic antigen antibody fragment (single-chain Fv-CH3) which exhibits rapid, high-level targeting of xenografts. Cancer Res. 56: 3055–3061.Google ScholarPubMed
Olafsen, T, Cheung, CW, Yazaki, PJ et al. (2004) Covalent disulfide-linked anti-CEA diabody allows site-specific conjugation and radiolabeling for tumor targeting applications. Protein Eng Des Sel. 17: 21–27.CrossRefGoogle ScholarPubMed
Wong, JYC, Chu, DZ, Yamauchi, DM et al. (2000) A Phase I radioimmunotherapy trial evaluating 90yttrium-labeled anti-carcinoembryonic antigen (CEA) chimeric T84.66 in patients with metastatic CEA-producing malignancies. Clin Cancer Res. 6: 3855–3863.Google ScholarPubMed
Cai, W, Olafsen, T, Zhang, X et al. (2007) PET imaging of colorectal cancer in xenograft-bearing mice by use of an 18F-labeled T84.66 anti-carcinoembryonic antigen diabody. J Nucl Med. 48: 304–310.Google ScholarPubMed
Wu, AM, Yazaki, PJ, Tsai, S et al. (2000) High-resolution microPET imaging of carcinoembryonic antigen-positive xenografts by using a copper-64-labeled engineered antibody fragment. Proc Natl Acad Sci USA. 97: 8495–8500.CrossRefGoogle ScholarPubMed
Yazaki, PJ, Wu, AM, Tsai, SW et al. (2001) Tumor targeting of radiometal labeled anti-CEA recombinant T84.66 diabody and t84.66 minibody: comparison to radioiodinated fragments. Bioconjug Chem. 12: 220–228.CrossRefGoogle ScholarPubMed
Sundaresan, G, Yazaki, PJ, Shively, JE et al. (2003) 124I-labeled engineered anti-CEA minibodies and diabodies allow high-contrast, antigen-specific small-animal PET imaging of xenografts in athymic mice. J Nucl Med. 44: 1962–1969.Google ScholarPubMed
http://www.clinicaltrials.gov/ct2/show/NCT00647153?term=diabody&rank=1.
Brown, RS, Wahl, RL (1993) Overexpression of Glut-1 glucose transporter in human breast cancer. An immunohistochemical study. Cancer. 72: 2979–2985.Google ScholarPubMed
Schoder, H, Glass, EC, Pecking, AP et al. (2006) Molecular targeting of the lymphovascular system for imaging and therapy. Cancer Metastasis Rev. 25: 185–201.CrossRefGoogle ScholarPubMed
Warburg, O (1956) On the origin of cancer cells. Science. 123: 309–314.CrossRefGoogle ScholarPubMed
Paudyal, B, Oriuchi, N, Paudyal, P, Higuchi, T, Nakajima, T, Endo, K (2008) Expression of glucose transporters and hexokinase II in cholangiocellular carcinoma compared using [18F]-2-fluro-2-deoxy-D-glucose positron emission tomography. Cancer Sci. 99: 260–266.CrossRefGoogle Scholar
Younes, M, Brown, RW, Stephenson, M, Gondo, M, Cagle, PT (1997) Overexpression of Glut1 and Glut3 in stage I nonsmall cell lung carcinoma is associated with poor survival. Cancer. 80: 1046–1051.3.0.CO;2-7>CrossRefGoogle Scholar
Christofk, HR, Vander Heiden, MG, Harris, MH et al. (2008) The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 452: 230–233.CrossRefGoogle ScholarPubMed
Christofk, HR, Vander Heiden, MG, Wu, N, Asara, JM, Cantley, LC (2008) Pyruvate kinase M2 is a phosphotyrosine-binding protein. Nature. 452: 181–186.CrossRefGoogle ScholarPubMed
,American Cancer Society (2009). Facts & Figures 2009. American Cancer Society.Google Scholar
Avril, N, Rose, CA, Schelling, M et al. (2000) Breast imaging with positron emission tomography and fluorine-18 fluorodeoxyglucose: use and limitations. J Clin Oncol. 18: 3495–3502.CrossRefGoogle ScholarPubMed
Palmedo, H, Bender, H, Grunwald, F et al. (1997) Comparison of fluorine-18 fluorodeoxyglucose positron emission tomography and technetium-99m methoxyisobutylisonitrile scintimammography in the detection of breast tumours. Eur J Nucl Med. 24: 1138–1145.Google ScholarPubMed
Wahl, RL, Cody, RL, Hutchins, GD, Mudgett, EE (1991) Primary and metastatic breast carcinoma: initial clinical evaluation with PET with the radiolabeled glucose analogue 2-[F-18]-fluoro-2-deoxy-D-glucose. Radiology. 179: 765–770.CrossRefGoogle ScholarPubMed
Adler, LP, Faulhaber, PF, Schnur, KC, Al-Kasi, NL, Shenk, RR (1997) Axillary lymph node metastases: screening with [F-18]2-deoxy-2-fluoro-D-glucose (FDG) PET. Radiology. 203: 323–327.CrossRefGoogle Scholar
Greco, M, Crippa, F, Agresti, R et al. (2001) Axillary lymph node staging in breast cancer by 2-fluoro-2-deoxy-D-glucose-positron emission tomography: clinical evaluation and alternative management. J Natl Cancer Inst. 93: 630–635.CrossRefGoogle ScholarPubMed
Smith, IC, Ogston, KN, Whitford, P et al. (1998) Staging of the axilla in breast cancer: accurate in vivo assessment using positron emission tomography with 2-(fluorine-18)-fluoro-2-deoxy-D-glucose. Ann Surg. 228: 220–227.CrossRefGoogle ScholarPubMed
Utech, CI, Young, CS, Winter, PF (1996) Prospective evaluation of fluorine-18 fluorodeoxyclucose positron emission tomography in breast cancer for staging of the axilla related to surgery and immunocytochemistry. Eur J Nucl Med. 23: 1588–1593.CrossRefGoogle ScholarPubMed
Eubank, WB, Mankoff, DA, Vesselle, HJ et al. (2002) Detection of locoregional and distant recurrences in breast cancer patients by using FDG PET. Radiographics. 22: 5–17.CrossRefGoogle ScholarPubMed
Kao, CH, Hsieh, JF, Tsai, SC, Ho, YJ, Yen, RF (2000) Comparison and discrepancy of 18F-2-deoxyglucose positron emission tomography and Tc-99m MDP bone scan to detect bone metastases. Anticancer Res. 20: 2189–2192.Google ScholarPubMed
Kostakoglu, L, Goldsmith, SJ (2003) 18F-FDG PET evaluation of the response to therapy for lymphoma and for breast, lung, and colorectal carcinoma. J Nucl Med. 44: 224–239.Google ScholarPubMed
Borkar, S, Pandit-Taskar, N (2008) F-18 FDG uptake in cutaneous metastases from breast cancer. Clin Nucl Med. 33: 488–489.CrossRefGoogle ScholarPubMed
Evilevitch, V, Weber, WA, Tap, WD et al. (2008) Reduction of glucose metabolic activity is more accurate than change in size at predicting histopathologic response to neoadjuvant therapy in high-grade soft-tissue sarcomas. Clin Cancer Res. 14: 715–720.CrossRefGoogle ScholarPubMed
Brepoels, L, Stroobants, S (2008) PET scanning and prognosis in Hodgkin's lymphoma. Curr Opin Oncol. 20: 509–516.CrossRefGoogle ScholarPubMed
Gayed, I, Vu, T, Iyer, R et al. (2004) The role of 18F-FDG PET in staging and early prediction of response to therapy of recurrent gastrointestinal stromal tumors. J Nucl Med. 45: 17–21.Google ScholarPubMed
Dose Schwarz, J, Bader, M, Jenicke, L, Hemminger, G, Janicke, F, Avril, N (2005) Early prediction of response to chemotherapy in metastatic breast cancer using sequential 18F-FDG PET. J Nucl Med. 46: 1144–1150.Google ScholarPubMed
Zeisel, SH (1981) Dietary choline: biochemistry, physiology, and pharmacology. Annu Rev Nutr. 1: 95–121.CrossRefGoogle ScholarPubMed
Podo, F (1999) Tumour phospholipid metabolism. NMR Biomed. 12: 413–439.3.0.CO;2-U>CrossRefGoogle ScholarPubMed
Cimitan, M, Bortolus, R, Morassut, S et al. (2006) [18F] fluorocholine PET/CT imaging for the detection of recurrent prostate cancer at PSA relapse: experience in 100 consecutive patients. Eur J Nucl Med Mol Imaging. 33: 1387–1398.CrossRefGoogle ScholarPubMed
Langsteger, W, Heinisch, M, Fogelman, I (2006) The role of fluorodeoxyglucose, 18F-dihydroxyphenylalanine, 18F-choline, and 18F-fluoride in bone imaging with emphasis on prostate and breast. Semin Nucl Med. 36: 73–92.CrossRefGoogle ScholarPubMed
Reske, SN, Blumstein, NM, Neumaier, B et al. (2006) Imaging prostate cancer with 11C-choline PET/CT. J Nucl Med. 47: 1249–1254.Google ScholarPubMed
Beheshti, M, Vali, R, Waldenberger, P et al. (2009) The use of F-18 choline PET in the assessment of bone metastases in prostate cancer: correlation with morphological changes on CT. Mol Imaging Biol. 11: 446–54.CrossRefGoogle ScholarPubMed
Kotzerke, J, Volkmer, BG, Neumaier, B, Gschwend, JE, Hautmann, RE, Reske, SN (2002) Carbon-11 acetate positron emission tomography can detect local recurrence of prostate cancer. Eur J Nucl Med Mol Imaging. 29: 1380–1384.CrossRefGoogle ScholarPubMed
Kotzerke, J, Prang, J, Neumaier, B et al. (2000) Experience with carbon-11 choline positron emission tomography in prostate carcinoma. Eur J Nucl Med. 27: 1415–1419.CrossRefGoogle ScholarPubMed
Kotzerke, J, Gschwend, JE, Neumaier, B (2002) PET for prostate cancer imaging: still a quandary or the ultimate solution? J Nucl Med. 43: 200–202.Google ScholarPubMed
Hara, T, Kosaka, N, Kishi, H (1998) PET imaging of prostate cancer using carbon-11-choline. J Nucl Med. 39: 990–995.Google ScholarPubMed
Picchio, M, Messa, C, Landoni, C et al. (2003) Value of [11C]choline-positron emission tomography for re-staging prostate cancer: a comparison with [18F] fluorodeoxyglucose-positron emission tomography. J Urol. 169: 1337–1340.CrossRefGoogle Scholar
Roivainen, A, Forsback, S, Gronroos, T, Lehikoinen, P, Kahkonen, M, Sutinen, E, Minn, H: Blood metabolism of [methyl-11C]choline; implications for in vivo imaging with positron emission tomography. Eur J Nucl Med 2000, 27: 25–32.CrossRefGoogle ScholarPubMed
Yamaguchi, T, Lee, J, Uemura, H et al. (2005) Prostate cancer: a comparative study of 11C-choline PET and MR imaging combined with proton MR spectroscopy. Eur J Nucl Med Mol Imaging. 32: 742–748.CrossRefGoogle Scholar
Rinnab, L, Mottaghy, FM, Simon, J et al. (2008) [11C]Choline PET/CT for targeted salvage lymph node dissection in patients with biochemical recurrence after primary curative therapy for prostate cancer. Preliminary results of a prospective study. Urol Int. 81: 191–197.CrossRefGoogle ScholarPubMed
Blau, M, Nagler, W, Bender, MA (1962) Fluorine-18: a new isotope for bone scanning. J Nucl Med 3: 332–334.Google ScholarPubMed
Narita, N, Kato, K, Nakagaki, H, Ohno, N, Kameyama, Y, Weatherell, JA (1990) Distribution of fluoride concentration in the rat's bone. Calcif Tissue Int. 46: 200–204.CrossRefGoogle ScholarPubMed
Hsu, WK, Virk, MS, Feeley, BT, Stout, DB, Chatziioannou, AF, Lieberman, JR (2008) Characterization of osteolytic, osteoblastic, and mixed lesions in a prostate cancer mouse model using 18F-FDG and 18F-fluoride PET/CT. J Nucl Med. 49: 414–421.CrossRefGoogle Scholar
Even-Sapir, E, Metser, U, Mishani, E, Lievshitz, G, Lerman, H, Leibovitch, I (2006) The detection of bone metastases in patients with high-risk prostate cancer: 99mTc-MDP Planar bone scintigraphy, single- and multi-field-of-view SPECT, 18F-fluoride PET, and 18F-fluoride PET/CT. J Nucl Med. 47: 287–297.Google ScholarPubMed
Contag, CH, Bachmann, MH (2002) Advances in in vivo bioluminescence imaging of gene expression. Annu Rev Biomed Eng. 4: 235–260.CrossRefGoogle ScholarPubMed
Hogemann, D, Basilion, JP (2002) “Seeing inside the body”: MR imaging of gene expression. Eur J Nucl Med Mol Imaging. 29: 400–408.CrossRefGoogle Scholar
Groot-Wassink, T, Aboagye, EO, Glaser, M, Lemoine, NR, Vassaux, G (2002) Adenovirus biodistribution and noninvasive imaging of gene expression in vivo by positron emission tomography using human sodium/iodide symporter as reporter gene. Hum Gene Ther. 13: 1723–1735.CrossRefGoogle ScholarPubMed
Lamberts, SW, Bakker, WH, Reubi, JC, Krenning, EP (1990) Somatostatin-receptor imaging in the localization of endocrine tumors. N Engl J Med. 323: 1246–1249.CrossRefGoogle ScholarPubMed
Wong, DF, Wagner, HN Jr, Tune, et al. Positron emission tomography reveals elevated D2 dopamine receptors in drug-naive schizophrenics. Science. (1986) 234: 1558–1563.CrossRefGoogle ScholarPubMed
Gambhir, SS, Bauer, E, Black, ME et al. (2000) A mutant herpes simplex virus type 1 thymidine kinase reporter gene shows improved sensitivity for imaging reporter gene expression with positron emission tomography. Proc Natl Acad Sci USA. 97: 2785–2790.CrossRefGoogle ScholarPubMed
Tjuvajev, JG, Doubrovin, M, Akhurst, T et al. (2002) Comparison of radiolabeled nucleoside probes (FIAU, FHBG, and FHPG) for PET imaging of HSV1-tk gene expression. J Nucl Med. 43: 1072–1083.Google ScholarPubMed
Brakenhielm, E, Burton, JB, Johnson, M et al. (2007) Modulating metastasis by a lymphangiogenic switch in prostate cancer. Int J Cancer. 121: 2153–2161.CrossRefGoogle ScholarPubMed
Johnson, M, Sato, M, Burton, J et al. (2005) MicroPET/CT monitoring of herpes thymidine kinase suicide gene therapy in a prostate cancer xenograft: the advantage of a cell-specific transcriptional targeting approach. Molecular Imaging. 4: 463–472.CrossRefGoogle Scholar
Scattoni, V, Picchio, M, Suardi, N et al. (2007) Detection of lymph-node metastases with integrated [11C]choline PET/CT in patients with PSA failure after radical retropubic prostatectomy: results confirmed by open pelvic-retroperitoneal lymphadenectomy. Eur Urol. 52: 423–429.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×