Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-nr4z6 Total loading time: 0 Render date: 2024-06-05T09:32:09.543Z Has data issue: false hasContentIssue false

11 - Nuclear medicine: diagnostic evaluation of metastatic disease

Published online by Cambridge University Press:  23 December 2009

Hedvig Hricak
Affiliation:
Memorial Sloan-Kettering Cancer Center
Peter Scardino
Affiliation:
Memorial Sloan-Kettering Cancer Center
Get access

Summary

Introduction

Nuclear medicine is a highly sensitive technique for detecting metastatic disease at sites throughout the body using radiotracers as biomarkers of cancer biology. Radioactive signals from these radiotracers are detected by sophisticated radioactivity-detector imaging systems including single-photon-emission computerized tomography (SPECT) and positron-emission tomography (PET). In this way nuclear medicine imaging is an example of “functional” or “molecular imaging” in which the imaging itself is based on a biochemical or physiologic property of the imaged tissue. For the patient with prostate cancer, nuclear medicine techniques, when used judiciously, are an important part of optimized cancer care; for example, bone scans are very commonly used to detect and follow up skeletal metastatic disease.

The clinical states model of Scher and Heller provides a useful guide to the possible progression of prostate cancer from a localized disease to widely spread tumor [1,2, 3] (Figure 11.1).

From the standpoint of natural history and chance of progression in the individual patient, prostate cancer is one of the most variable of all tumors. The cancer itself may behave in a highly indolent matter and be localized in the prostate gland for years, with the patient's death being due to other diseases. Then again, a localized tumor may rapidly progress, become widely metastatic within a short time and lead to death.

Fortunately, clinical trials are now resolving key management questions based on objective features of cancer histology and biochemistry.

Type
Chapter
Information
Prostate Cancer , pp. 177 - 194
Publisher: Cambridge University Press
Print publication year: 2008

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

Scher, H. I., Eisenberger, M., D'Amico, A. V., et al., Eligibility and outcomes reporting guidelines for clinical trials for patients in the state of a rising prostate-specific antigen: recommendations from the Prostate-Specific Antigen Working Group. J Clin Oncol, 22 (2004), 537–56.CrossRefGoogle ScholarPubMed
Scher, H. I., Heller, G., Clinical states in prostate cancer: toward a dynamic model of disease progression. Urology, 55 (2000), 323–7.CrossRefGoogle Scholar
Scher, H. I., Morris, M. J., Kelly, W. K., et al., Prostate cancer clinical trial end points: “RECIST”ing a step backwards. Clin Cancer Res, 11 (2005), 5223–32.CrossRefGoogle Scholar
Bill-Axelson, A., Holmberg, L., Ruutu, M., et al., Radical prostatectomy versus watchful waiting in early prostate cancer. N Engl J Med, 352 (2005), 1977–84.CrossRefGoogle ScholarPubMed
Holmberg, L., Bill-Axelson, A., Helgesen, F., et al., A randomized trial comparing radical prostatectomy with watchful waiting in early prostate cancer. N Engl J Med, 347 (2002), 781–9.CrossRefGoogle ScholarPubMed
Freedland, S. J., Humphreys, E. B., Mangold, L. A., et al., Risk of prostate cancer-specific mortality following biochemical recurrence after radical prostatectomy. JAMA, 294 (2005), 433–9.CrossRefGoogle ScholarPubMed
Freedland, S. J., Humphreys, E. B., Mangold, L. A., et al., Death in patients with recurrent prostate cancer after radical prostatectomy: prostate-specific antigen doubling time subgroups and their associated contributions to all-cause mortality. J Clin Oncol, 25 (2007), 1765–71.CrossRefGoogle ScholarPubMed
Sabbatini, P., Larson, S. M., Kremer, A., et al., Prognostic significance of extent of disease in bone in patients with androgen-independent prostate cancer. J Clin Oncol, 17 (1999), 948–57.CrossRefGoogle ScholarPubMed
Imbriaco, M., Larson, S. M., Yeung, H. W., et al., A new parameter for measuring metastatic bone involvement by prostate cancer: the Bone Scan Index. Clin Cancer Res, 4 (1998), 1765–72.Google ScholarPubMed
Morris, M. J., Akhurst, T., Osman, I., et al., Fluorinated deoxyglucose positron emission tomography imaging in progressive metastatic prostate cancer. Urology, 59 (2002), 913–18.CrossRefGoogle ScholarPubMed
Yeh, S. D., Imbriaco, M., Larson, S. M., et al., Detection of bony metastases of androgen-independent prostate cancer by PET-FDG. Nucl Med Biol, 23 (1996), 693–7.CrossRefGoogle ScholarPubMed
Schneider, J. A., Divgi, C. R., Scott, A. M., et al., Flare on bone scintigraphy following Taxol chemotherapy for metastatic breast cancer. J Nucl Med, 35 (1994), 1748–52.Google Scholar
Morris, M. J., Akhurst, T., Larson, S. M., et al., Fluorodeoxyglucose positron emission tomography as an outcome measure for castrate metastatic prostate cancer treated with antimicrotubule chemotherapy. Clin Cancer Res, 11 (2005), 3210–16.CrossRefGoogle ScholarPubMed
Young, H., Baum, R., Cremerius, U., et al., Measurement of clinical and subclinical tumour response using [18F]-fluorodeoxyglucose and positron emission tomography: review and 1999 EORTC recommendations. European Organization for Research and Treatment of Cancer (EORTC) PET Study Group. Eur J Cancer, 35 (1999), 1773–82.CrossRefGoogle Scholar
Schöder, H., Herrmann, K., Gönen, M., et al., 2-[18F]fluoro-2-deoxyglucose positron emission tomography for the detection of disease in patients with prostate-specific antigen relapse after radical prostatectomy. Clin Cancer Res, 11:13 (2005), 4761–9.CrossRefGoogle ScholarPubMed
Mohammed, A. A., Shergill, I. S., Vandal, M. T., et al., ProstaScint and its role in the diagnosis of prostate cancer. Expert Rev Mol Diagn, 7 (2007), 345–9.CrossRefGoogle Scholar
Haseman, M. K., Rosenthal, S. A., Kipper, S. L., et al., Central abdominal uptake of indium-111 capromab pendetide (ProstaScint) predicts for poor prognosis in patients with prostate cancer. Urology, 70 (2007), 303–8.CrossRefGoogle ScholarPubMed
Nagda, S. N., Mohideen, N., Lo, S. S., et al., Long-term follow-up of 111In-capromab pendetide (ProstaScint) scan as pretreatment assessment in patients who undergo salvage radiotherapy for rising prostate-specific antigen after radical prostatectomy for prostate cancer. Int J Radiat Oncol Biol Phys, 67:3 (2007), 834–40.CrossRefGoogle ScholarPubMed
Chen, C. D., Welsbie, D. S., Tran, C., et al., Molecular determinants of resistance to antiandrogen therapy. Nat Med, 10 (2004), 33–9.CrossRefGoogle ScholarPubMed
Bonasera, T. A., O'Neil, J. P., Xu, M., et al., Preclinical evaluation of fluorine-18-labeled androgen receptor ligands in baboons. J Nucl Med, 37 (1996), 1009–15.Google ScholarPubMed
Larson, S. M., Morris, M., Gunther, I., et al., Tumor localization of 16beta-18F-fluoro-5alpha-dihydrotestosterone versus 18F-FDG in patients with progressive, metastatic prostate cancer. J Nucl Med, 45 (2004), 366–73.Google ScholarPubMed
Dehdashti, F., Picus, J., Michalski, J. M., et al., Positron tomographic assessment of androgen receptors in prostatic carcinoma. Eur J Nucl Med Mol Imaging, 32 (2005), 344–50.CrossRefGoogle ScholarPubMed
Nilsson, S., Kalner, K., Ginman, C., et al., C-11 methionine positron emission tomography in the management of prostatic carcinoma. Antibody Immunoconj Radiopharm, 8 (1995), 23–38.Google Scholar
Nunez, R., Macapinlac, H. A., Yeung, H. W., et al., Combined 18F-FDG and 11C-methionine PET scans in patients with newly progressive metastatic prostate cancer. J Nucl Med, 43 (2002), 46–55.Google ScholarPubMed
Macapinlac, H. A., Humm, J. L., Akhurst, T., et al., Differential metabolism and pharmacokinetics of L-[1-(11)C]-methionine and 2-[(18)F] fluoro-2-deoxy-D-glucose (FDG) in androgen independent prostate cancer. Clin Positron Imaging, 2 (1999), 173–81.CrossRefGoogle Scholar
Schuster, D. M., Votaw, J. R., Nieh, P. T., et al., Initial experience with the radiotracer anti-1-amino-3-18F-fluorocyclobutane-1-carboxylic acid with PET/CT in prostate carcinoma. J Nucl Med, 48 (2007), 56–63.Google ScholarPubMed
Powles, T., Murray, I., Brock, C., et al., Molecular positron emission tomography and PET/CT imaging in urological malignancies. Eur Urol, 51 (2007), 1511–20; discussion 1520–1.CrossRefGoogle ScholarPubMed
Heinisch, M., Dirisamer, A., Loidl, W., et al., Positron emission tomography/computed tomography with F-18-fluorocholine for restaging of prostate cancer patients: meaningful at PSA <5 ng/ml?Mol Imaging Biol, 8 (2006), 43–8.CrossRefGoogle ScholarPubMed
Langsteger, W., Heinisch, M., Fogelman, I., The role of fluorodeoxyglucose, 18F-dihydroxyphenylalanine, 18F-choline, and 18F-fluoride in bone imaging with emphasis on prostate and breast. Semin Nucl Med, 36 (2006), 73–92.CrossRefGoogle ScholarPubMed
Vees, H., Buchegger, F., Albrecht, S., et al., 18F-choline and/or 11C-acetate positron emission tomography: detection of residual or progressive subclinical disease at very low prostate-specific antigen values (<1 ng/ml) after radical prostatectomy. BJU Int, 99 (2007), 1415–20.CrossRefGoogle ScholarPubMed
Oyama, N., Miller, T. R., Dehdashti, F., et al., 11C-acetate PET imaging of prostate cancer: detection of recurrent disease at PSA relapse. J Nucl Med, 44 (2003), 549–55.Google ScholarPubMed
Oyama, N., Kim, J., Jones, L. A., et al., MicroPET assessment of androgenic control of glucose and acetate uptake in the rat prostate and a prostate cancer tumor model. Nucl Med Biol, 29 (2002), 783–90.CrossRefGoogle Scholar
Oyama, N., Akino, H., Kanamaru, H., et al., 11C-acetate PET imaging of prostate cancer. J Nucl Med, 43 (2002), 181–6.Google ScholarPubMed
Smith-Jones, P. M., Vallabahajosula, S., Goldsmith, S. J., et al., In vitro characterization of radiolabeled monoclonal antibodies specific for the extracellular domain of prostate-specific membrane antigen. Cancer Res, 60 (2000), 5237–43.Google ScholarPubMed
Morris, M. J., Divgi, C. R., Pandit-Taskar, N., et al., Pilot trial of unlabeled and indium-111-labeled anti-prostate-specific membrane antigen antibody J591 for castrate metastatic prostate cancer. Clin Cancer Res, 11 (2005), 7454–61.CrossRefGoogle ScholarPubMed
Bander, N. H., Technology insight: monoclonal antibody imaging of prostate cancer. Nat Clin Pract Urol, 3 (2006), 216–25.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
×