18F-NaF PET/CT imaging versus 99mTc-MDP scintigraphy in assessing metastatic bone disease in patients with prostate cancer

Georgios Z. Papadakis, Kostas Marias, Corina Millo, Apostolos H. Karantanas

Abstract


Prostate Cancer (ProCa) is one of the most commonly encountered malignancies in men, with variable incidence and prevalence rates across different parts of the world. Despite being a slow-growing tumor, ProCa can be fatal when giving distant metastases, with the skeleton being the most common metastatic sites. Osseous metastases in ProCa patients can be osteoblastic, osteolytic or mixed, and are associated with significant morbidity. Thus, early detection and precise assessment of skeletal involvement is critical for prognosis and accurate management of ProCa patients. Conventional bone scintigraphy (BS) using 99m-Technetium (99mTc) labeled radiopharmaceuticals with planar imaging, SPECT imaging and hybrid SPECT/CT imaging, has been employed for decades in the assessment of metastatic bone disease of ProCa patients, due to its low cost and availability. However, skeletal imaging with modern hybrid PET/CT systems using 18Fluorine-Sodium fluoride (18F-NaF), exhibits superior diagnostic performance compared to conventional BS in addressing the task of evaluating bony involvement in ProCa patients. The superior spatial resolution of PET over SPECT, the preferable pharmacokinetics of 18F-NaF over 99mTc labeled agents, and the superior inherent quantitative capabilities of PET-imaging, result in the superior diagnostic performance of 18F-NaF PET/CT imaging, which is documented in the current review article. Given the widespread availability of PET/CT scanners, and the decreasing cost of 18F-NaF production, it is recommended that conventional BS should be replaced by the superior 18F-NaF PET/CT imaging in the work-up of ProCa patients.


Keywords


18F-NaF PET/CT imaging, 99mTc-MDP scintigraphy, Metastatic bone disease, Prostate Cancer

Full Text:

PDF

References


Bray F, Ferlay J, Soerjomataram I et al. Global cancer statistics 2018: GLOBOSCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68:394-424.

Cronin KA, Lake AJ, Scott S et al. Annual report to the nation on the status of cancer, Part II: National cancer statistics. Cancer 2018; 124(13):2785-2800.

Negoita S, Feuer EJ, Mariotto A et al. Annual report to the nation on the status of cancer, Part II: Recent changes in prostate cancer trends and disease characteristics. Cancer 2018;124(13):2801-2814.

Taitt HE. Global trends and prostate cancer: a review of incidence, detection, and mortality as influenced by race, ethnicity, and geographic location. Am J Mens Health 2018;12(6):1807-1823.

Attard G, Parker C, Edes RA et al. Prostate Cancer. Lancet 2016; 387: 70-82.

Ewing CM, Ray AM, Lange EM, et al. Germline mutations in HOXB13 and prostate-cancer risk. N Engl J Med 2012; 366: 141-49.

Breast cancer linkage consortium. cancer risks in BRCA2 mutation carriers. J Natl Cancer Inst 1999; 91: 1310-16.

Kote-Jarai Z, Leongamornlert D, Saunders E et al, for the UKGPCS Collaborators. BRCA2 is a moderate penetrance gene contributing to young-onset prostate cancer: implications for genetic testing in prostate cancer patients. Br J Cancer 2011; 105: 1230-34.

Chen H, Ewing CM, Zheng S et al. Genetic factors influencing prostate cancer in Norwegian men. Prostate 2018;78(3):186-192.

Perez-Cornago A, Key TJ, Allen NE et al. Prospective investigation of risk factors for prostate cancer in the UK Biobank cohort study. Br J Cancer 2017;117(10):1562-1571.

Bostwick DG, Burke HB, Djakiew D et al. Human prostate cancer risk factors. Cancer 2004;101(10 Suppl):2371-490.

American Cancer Society. Cancer facts & figures for African Americans 2016-2018. Atlanta: American Cancer Society, 2016.

Layne TM, Graubard BI, Ma X et al. Prostate cancer risk factors in black and white men in the NIH-AARPdiet and health study. Prostate Cancer Prostatic Dis 2019; 22(1): 91-100.

Gathirua-Mwangi WG, Zhang J. Dietary factors and risk of advanced prostate cancer. Eur J Cancer Prev 2014; 23(2): 96-109.

Pezaro C, Woo HH, Davis ID. Prostate cancer: measuring PSA. Intern Med J 2014; 44(5): 433-40.

Hatakeyama S, Yoneyama S, Tobisawa Y et al. Recent progress and perspectives on prostate cancer biomarkers. Int J Clin Oncol 2017; 22:214-221.

Saini S. PSA and beyond: alternative prostate cancer biomarkers. Cell Oncol (Dordr) 2016; 39(2): 97-106.

Leal J, Welton NJ, Martin RM et al. Estimating the sensitivity of a prostate cancer screening programme for different PSA cut-off levels: A UK case study. Cancer Epidemiol 2018; 52:99-105.

Koie T, Mitsuzuka K, Narita S et al. A solitary positive prostate cancer biopsy does not predict a unilateral lesion in radical prostatectomy specimens. Scand J Urol 2015, 49(2):103-107.

Hendriks RJ, van Oort IM, Schalken JA. Blood-based and urinary biomarkers: a review and comparison of novel biomarkers for detection and treatment decisions. Prostate Cancer Prostatic Dis 2017; 20(1): 12-19.

Verma S, Choyke PL, Eberhardt SC et al. The current state of MR Imaging - targeted biopsy techniques for detection of prostate cancer. Radiology 2017; 285 (2): 343-356.

Litwin MS, Tan H-J. The diagnosis and treatment of prostate cancer. JAMA 2017; 317(24): 2532-2542.

Epstein JI, Amin MB, Reuter VE et al. Contemporary Gleason grading of prostatic carcinoma. An update with discussion on practical issues to implement the 2014 international society of urological pathology (isup) consensus conference on Gleason grading of prostatic carcinoma. Am J Surg Pathol. 2017; 41(4): e1-e7.

Epstein JI. Gleason score 2-4 adenocarcinoma of the prostate on needle biopsy. Am J Surg Pathol 2000; 24(4):477-8.

Caverly TJ, Hayward RA, Reamer E et al. Presentation of benefits and harms in US Cancer Screening and Prevention Guidelines: Systematic Review. J Natl Cancer Inst 2016; 108: djv436.

Ahmed HU, El-Shater Bosaily A Brown LC et al. Diagnostic accuracy of multiparametric MRI and TRUS biopsy in prostate cancer (PROMIS): a paired validating confirmatory study. Lancet 2017; 389 (10071): 815-822.

Siddiqui MM, Rais-Bahrami S, Turkbey B et al. Comparison of MR/Ultrasound fusion-guided biopsy with ultrasound-guided biopsy for the diagnosis of prostate cancer. JAMA 2015; 313(4): 390-397.

Bitting RL, Schaeffer D, Somarelli JA et al. The role of epithelial plasticity in prostate cancer dissemination and treatment resistance. Cancer Metastasis Rev 2014;33(2-3):441-68.

Jin J-K, Dayyani F, Gallick GE. Steps in prostate progression that lead to bone metastasis. Int. J. Cancer 2011; 128: 2545-2561.

Gartrell BA and Saad F. Managing bone metastases and reducing skeletal related events in prostate cancer. Nat Rev Clin Oncol 2014;11(6):335-45.

Morrissey C and Vessella RI. The role of tumor microenvironment in prostate cancer bone metastasis. J Cell Biochem. 2007;101(4):873-86.

Van der Toom E, Verdone JE, Pienta KJ. Disseminated tumor cells and dormancy in prostate cancer metastasis. Curr Opin Biotechnol 2016; 40: 9-15.

Rubens RD. Bone metastases: incidence and complications. In: Mundy GR, Rubens RD. Cancer and the skeleton. London, England: Martin Dunitz, 2000; 33-42.

Ozulker T, Uzun AK, Ozulker F et al. Comparison of 18F-FDG-PET/CT with 99mTc-MDP bone scintigraphy for the detection of bone metastases in cancer patients. Nucl Med Commun 2010;31(6):597-603

Langsteger W, Rezaee A, Pirich C et al. 18F-NaF-PET/CT and 99mTc-MDP Bone scintigraphy in the detection of bone metastases in prostate cancer. Semin Nucl Med 2016; 46(6): 491-501.

Messiou C, Cook G, de Souza NM. Imaging metastatic bone disease from carcinoma of the prostate. Br J Cancer 2009;101(8):1225-32.

Mick CG, James T, Hill DJ et al. Molecular imaging in oncology: 18F-Sodium Fluoride PET imaging of osseous metastatic disease. AJR Am J Roentgenol 2014;203(2):263-71.

Kanishi D. 99mTc-MDP accumulation mechanisms in bone. Oral Surg Oral Med Oral Pathol 1993;75(2):239-46.

National institute for clinical excellence improving outcomes in urological cancer (2002). https://www.nice.org.uk/guidance/ csg2 Accessed 19 Jul 2016.

Briganti A, Passoni N, Ferrari M et al. When to perform bone scan in patients with newly diagnosed prostate cancer: external validation of the currently available guidelines and proposal of a novel risk stratification tool. Eur Urol 2010;57(4):551-8.

Ritenour CW, Abbott JT, Goodman M et al. The utilization of Gleason grade as the primary criterion for ordering nuclear bone scan in newly diagnosed prostate cancer patients. ScientificWorldJournal 2009; 9:1040-5.

Lin Y, Mao Q, Chen B et al. When to perform scintigraphy in patients with newly diagnosed prostate cancer? A retrospective study. BMC Urol 2017;17(1):41.

Even-Sapir E, Metser U, Mishani E et al. 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 2006; 47(2):287-97.

Damle NA, Bal C, Bandopadhyaya GP et al. The role of 18F-fluoride PET-CT in the detection of bone metastases in patients with breast, lung and prostate carcinoma: a comparison with 18F-FDG PET/CT and 99mTc-MDP bone scan. Jpn J Radiol 2013;31(4):262-9.

Rathke H, Afshar-Oromieh A, Giesel FL et al. Intraindividual comparison of 99mTc-Methylene Diphosphonate and prostate-specific membrane antigen ligand 99mTc-MIP-1427 in patients with osseous metastasized prostate cancer. J Nucl Med 2018;59(9):1373-1379.

Ciray I, Astrom G, Andreasson I et al. Evaluation of new sclerotic bone metastases in breast cancer patients during treatment. Acta Rad 2000; 41(2): 178-182.

Imbriaco M, Larson SM, Yeung HW et al. A new parameter for measuring metastatic bone involvement by prostate cancer: The bone scan index. Clin Cancer Res 1998; 4:1765-1772.

Meirelles GSP, Schroder H, Ravizzini et al. Prognostic value of baseline [18F] Fluorodeoxyglucose positron emission tomography and 99mTc-MDP bone scan in progressing metastatic prostate cancer. Clin Cancer Res 2010;16(24):6093-9.

Dennis ER, Jia X, Mezheritskiy IS et al. Bone scan index: a quantitative treatment response biomarker for castration resistant metastatic prostate cancer. J Clin Oncol 2012; 30(5): 519-24.

Reza M, Bjartell A, Ohlsson M et al: Bone sscan index as a prognostic imaging biomarker during androgen deprivation therapy. EJNMMI Res 2014; 4:58.

Zacho HD, Gade M, Mortensen JC et al. Bone Scan Index is an independent predictor of time to castration-resistant prostate cancer in newly diagnosed prostate cancer: A prospective study. Urology 2017; 108:135-141.

Poulsen MH, Rasmussen J, Edenbrandt L et al. Bone scan index predicts outcome in patients with metastatic hormone-sensitive prostate cancer. BJU Int 2016; 117(5): 748-53.

Armstrong AJ, Anand A, Edenbrandt L et al. Phase 3 Assessment of the automated bone scan index as a prognostic imaging biomarker of overall survival in men with metastatic castration-resistant prostate cancer. A secondary analysis of a randomized clinical trial. JAMA Oncol. 2018;4(7):944-951.

Li D, Lv H, Hao X et al. Prognostic value of bone scan index as imaging biomarker in metastatic prostate cancer: a meta-analysis. Oncotarget 2017; 8(48): 84449-84458.

Shen G, Deng H, Jia Z. Comparison of choline-PET/CT, MRI, SPECT, and bone scintigraphy in the diagnosis of bone metastasis in patients with prostate cancer: a meta-analysis. Skeletal Radiol 2014; 43:1503-1513.

O’Sullivan GJ, Carty FL, Cronin CG. Imaging of bone metastasis: an update. World J Radiol 2015; 28; 7(8): 202-211.

Römer W, Nömayr A, Uder M, Bautz W, Kuwert T. SPECTguided CT for evaluating foci of increased bone metabolism classified as indeterminate on SPECT in cancer patients. J Nucl Med. 2006; 47:1102-6.

Helyar V, Mohan HK, Barwick T, Livieratos L, Gnanasegaran G, Clarke SE, Fogelman I. The added value of multislice SPECT/CT in patients with equivocal bony metastasis from carcinoma of the prostate. Eur J Nucl Med Mol Imaging. 2010; 37:706-13.

Ndlovu X, George R, Ellmann A, Warwick J. Should SPECT-CT replace SPECT for the evaluation of equivocal bone scan lesions in patients with underlying malignancies? Nucl Med Commun. 2010; 31:659-65.

Zhang Y, Shi H, Gu Y, Xiu Y, Li B, Zhu W, Chen S, Yu H. Differential diagnostic value of single-photon emission computed tomography/spiral computed tomography with Tc-99m-methylene diphosphonate in patients with spinal lesions. Nucl Med Commun. 2011; 32:1194-200.

Jiang L, Han L, Tan H, Hu P, Zhang Y, Shi H. Diagnostic value of 99mTc-MDP SPECT/spiral CT in assessing indeterminate spinal solitary lesion of patients without malignant history. Ann Nucl Med. 2013; 27:460-7.

Cook GJ, Azad G, Padhani AR. Bone imaging in prostate cancer: the evolving roles of nuclear medicine and radiology. Clin Transl Imaging. 2016; 4:439-47.

Palmedo H, Marx C, Ebert A, et al. Whole-body SPECT/CT for bone scintigraphy: diagnostic value and effect on patient management in oncological patients. Eur J Nucl Med Mol Imaging. 2014;41(1):59-67.

Fonager RF, Zacho HD, Langkilde NC et al. Diagnostic test accuracy of 18F-sodium fluoride PET/CT, 99mTC - labelled diphosphonate SPECT/CT, and planar bone scintigraphy for diagnosis of bone metastases in newly diagnosed, high-risk prostate cancer. Am J Nucl Med Mol Imaging 2017;7(5):218-227

Sharma P, Dhull VS, Reddy RM et al. Hybrid SPECT-CT for characterizing isolated vertebral lesions observed by bone scintigraphy: Comparison with planar scintigraphy, SPECT, and CT. Diagn Interv Radiol 2013;19:33-40

Fleury V, Ferrer L, Colombié M, et al, Advantages of systematic trunk SPECT/CT to planar bone scan (PBS) in more than 300 patients with breast or prostate cancer. Oncotarget. 2018 Aug 3;9(60):31744-31752.

Araz M, Aras G, Kucuk ON. The role of 18F-NaF PET/CT in metastatic bone disease. J Bone Oncol 2015; 4:92-97

Blau M, Nagler W, Bender MA. Fluorine-18: a new isotope for bone scanning. J Nucl Med 1962; 3: 332-334.

Bastawrous S, Bhargava P, Behnia F et al. Newer PET application with an old tracer: role of 18F-NaF skeletal PET/CT in oncologic practice. Radiographics. 2014; 34(5):1295-316.

Papadakis GZ, Manikis GC, Karantanas AH, et al. 18F-NaF uptake by fibrous dysplasia bone lesions is positively associated with bone turnover markers (BTMs). Eur J Nucl Med Mol Imaging 2019; 45(Suppl 1): S241-S241.

Papadakis GZ, Manikis GC, Karantanas AH, et al. Prognostic utility of 18F-NaF PET/CT imaging for fractures in patients with fibrous dysplasia of bone. Eur J Nucl Med Mol Imaging 2019; 45(Suppl 1): S241-S242.

Papadakis G, Manikis G, Karantanas A, et al. Fibrous dysplasia related 18F-NaF activity in the spine is significantly higher in patients with scoliosis, compared to patients without spinal deformity. J Nucl Med 2019; 60 (suppl 1): 1298-1298.

Papadakis G, Manikis G, Karantanas A, et al. Positive Association between Fibrous Dysplasia (FD) related 18F-NaF activity and Bone Turnover Markers (BTMs). J Nucl Med 2019; 60 (suppl 1): 90-90.

Hartley I, McCarthy T, Papadakis G, et al. Treatment of Gorham-Stout Disease with Combination Sirolimus and Denosumab. J Endocr Soc. 2019; 3 (suppl 1) : MON-517.

Papadakis GZ, Jha S, Karantanas AH et al. Prospective evaluation of the application of 18F-NaF PET/CT imaging in melorheostosis Eur J Nucl Med Mol Imaging 2018; 45(Suppl 1): S231-S232.

Papadakis GZ, Jha S, Bhattacharyya T, et al. 18F-NaF PET/CT in extensive melorheostosis of the axial and appendicular skeleton with soft-tissue involvement. Clin Nucl Med 2017;42(7):537-539.

Papadakis GZ, Millo C, Bagci Uet al. 18F-NaF and 18F-FDG PET/CT in Gorham-Stout Disease. Clin Nucl Med 2016; 41(11):884-885.

Papadakis GZ, Millo C, Bagci U et al. Value of 18F-NaF PET/CT imaging in the assessment of Gorham-Stout disease activity. Eur J Nucl Med Mol Imaging 2016; 43(suppl 1): S597-S597.

Papadakis GZ, Manikis GC, Karantanas AH Et al. Application of 18F-NaF PET/CT imaging in fibrous dysplasia. Horm Res Paediatr 2017;88 (suppl 1):20-20.

Papadakis G, Manikis G, Karantanas A et al. Application of 18F-NaF PET/CT imaging in prognosis of fractures and treatment planning in patients with fibrous dysplasia. J Nucl Med 2017; 58 (suppl 1): 308-308.

Papadakis GZ, Manikis GC, Karantanas AH, et al., 18F-NaF PET/CT imaging in fibrous dysplasia of bone. J Bone Miner Res. 2019 ;34(9):1619-1631.

Segall G, Delbeke D, Stabin MG, et al. SNM practice guideline for sodium 18F-fluoride PET/CT bone scans 1.0. J Nucl Med 2010; 51(11):1813-20.

Vali R, Beheshti M, Waldenberger P et al. Assessment of malignant and benign bone lesions by static 18F-Fluoride PET-CT: Additional value of SUV!. J Nucl Med 2008; 49:150 (suppl 1).

Muzahir S, Jeraj R, Liu G et al. Differentiation of metastatic vs degenerative joint disease using semi-quantitative analysis with 18F-NaF PET/CT in castrate resistant prostate cancer patients. Am J Nucl Med Mol Imaging 2015; 5:162-8.

Simoncic U, Perlman S, Liu G et al. Comparison of NaF and FDG PET/CT for assessment of treatment response in castrate-resistant prostate cancers with osseous metastases. Clin Genitourin Cancer. 2015;13(1): e7-e17.

Jadvar H, Colletti PM. 18F-NaF/223RaCl2 theranostics in metastatic prostate cancer: treatment response assessment and prediction of outcome. Br J Radiol. 2018;91(1091):20170948.

Zukotynski KA, KimCK, GerbaudoVH et al. 18F-FDG-PET/CT and 18F-NaF-PET/CT in men with castrate-resistant prostate cancer. Am J Nucl Med Mol Imaging 2015; 5:72-82.

Apolo AB, Lindenberg L, Shih JH et al: Prospective study evaluating 18F-NaF-positron emission tomography/computed tomography (18F-NaF-PET/ CT) in predicting clinical outcomes and survival in advanced prostate cancer. J Nucl Med. 2016;57(6):886-892.

Poulsen MH, Petersen H, Hoilund-Carlsen PF et al. Spine metastases in prostate cancer: Comparison of technetium-99m-MDP whole - body bone scintigraphy, [18F] choline positron emission tomography (PET)/computed tomography (CT) and [18F] NaF PET/CT. BJU Int 2014;114: 818-823.

Vali R, Loidl W, Pirich C et al. Imaging of prostate cancer with PET/CT using 18F-Fluorocholine. Am J Nucl Med Mol Imaging. 2015; 5(2): 96-108.

Iagaru A, Mittra E, Dick DW et al. Prospective evaluation of 99mTc MDP scintigraphy, 18F NaF PET/CT, and 18F FDG PET/CT for detection of skeletal metastases. Mol Imaging Biol 2012; 14:252-259.

Minamimoto R, Loening A, Jamali M et al. Prospective Comparison of 99mTc-MDP Scintigraphy, Combined 18F-NaF and 18F-FDG PET/CT, and whole-body MRI in patients with breast and prostate cancer. J Nucl Med 2015; 56(12):1862-8.

Jambor I, Kuisma A, Ramadan S et al. Prospective evaluation of planar bone scintigraphy, SPECT, SPECT/CT, 18F-NaF PET/CT and whole body 1.5T MRI, including DWI, for the detection of bone metastases in high risk breast and prostate cancer patients: SKELETA clinical trial. Acta Oncol 2016; 55(1): 59-67.

Ceci F, Castellucci P, Fanti S. Current application and future perspectives of prostate specific membrane antigen PET imaging in prostate cancer. Q J Nucl Med Mol Imaging. 2019;63(1):7-18.

Treglia G, Pereira Mestre R, Ferrari M5, et al. Radiolabelled choline versus PSMA PET/CT in prostate cancer restaging: a meta-analysis. Am J Nucl Med Mol Imaging. 2019 15;9(2):127-139.




DOI: http://dx.doi.org/10.36162/hjr.v4i4.286

Refbacks

  • There are currently no refbacks.