Abstract
Theranostics refers to companion agents with identical or similar structure targeted to a specific biological entity for imaging and treatment. Although the concept has a long history with radioiodine in thyroidology, but it has experienced remarkable recent renaissance in management of neuroendocrine tumors and prostate cancer. Bone scintigraphy based on osteoblastic reaction and targeted radionuclide therapy with the alpha-particle calcium-mimetic agent, 223RaCl2, also form a theranostic model for imaging and treatment of osseous metastatic disease. Since the regulatory approval of 223RaCl2 in 2013, there has been accumulating evidence on the potential use of 18F-NaF PET scintigraphy in the assessment of response and prediction of outcome in males with metastatic castrate-resistant prostate cancer who undergo 223RaCl2 therapy. We review the 18F-NaF/223RaCl2 as theranostic companion in the management of prostate cancer with emphasis on the utility of 18F-NaF and other relevant PET radiotracers in the therapy response and prognosis assessments.
Theranostics had its beginning in diagnosis and treatment of thyroid diseases with radioiodine more than 75 years ago, although this specific designation was probably coined more recently.1 Theranostics is a systematic integration of targeted diagnostics and therapeutics as companion agents that facilitates precision medicine by identifying subset of patients who would benefit from a particular treatment based on imaging evidence for expression of the intended biological target. This fast-growing field, with more recent strides in care of patients with prostate cancer and neuroendocrine tumors, has been recognized as one of the priorities in future cancer research.2
There are now several approved treatments available for metastatic castrate-resistant prostate cancer (CRPC). These include drugs that target the androgen axis, such as abiraterone acetate (androgen synthesis inhibitor) and enzalutamide (androgen receptor antagonist), cabazitaxel (novel taxane chemotherapy), sipuleucel-T immunotherapy (cancer vaccine), and alpha particle therapy of bone lesions with 223RaCl2.3–5 The most optimal patient selection or drug sequencing criteria is not yet established and will need further understanding of the biological drivers of resistance to various treatments, cross-reactivity of treatments, and effect of tumor heterogeneity.6, 7 However, type of prior treatments, tumor burden, symptoms, performance status, comorbidities, and patient and physician preferences may also be other factors to consider.
Alpha particle therapy with 223RaCl2 targeted to osteoblastic metastatic bone lesions is the first-in-class of such treatments that has been shown to be safe and effective and was approved by the Food and Drug Administration (FDA) on May 15, 2013. 8–15 223RaCl2 is included in the US, UK and European guidelines and the nuts and bolts and practical considerations for clinical practice and patient management have been published.16–19
A recent study has demonstrated that 223RaCl2 can be safely combined with other treatments that are associated with improved outcome when compared to 223RaCl2 alone.20 Overall survival was also found to be longer in those patients who had normal range baseline alkaline phosphatase level, hemoglobin level 10 g dl−1 or greater, Eastern Cooperative Oncology Group Performance status of 0, and no pain in comparison to those patients who did not satisfy these parameters. This study suggested that 223RaCl2 might be used in asymptomatic patients (in contradistinction to the inclusion criterion of the ALSYMPCA trial) and with other treatments for improved outcome. In another retrospective investigation of 64 males with metastatic CRPC, a multivariate analysis showed that no prior chemotherapy, oligometastatic disease (≤5 bone metastases), baseline alkaline phosphatase <115 U l−1, and alkaline phosphatase response after 223RaCl2 therapy were predictive stratification factors of improved survival.21 In a recent investigation, a multivariable analysis showed that previous sipuleucel-T treatment (p = 0.009), hemoglobin ≥lower limit of normal (LLN) (p = 0.037), and absolute neutrophil count (ANC) ≥ LLN (p = 0.029) were significantly associated with the probability of completing the entire 6-cycle course of 223RaCl2 treatment.22 Treatment with 223RaCl2 may also benefit patients with hormone-sensitive bone metastases after radical prostatectomy. Overall, 223RaCl2 tends not to lead to sustained PSA decline but decrease in alkaline phosphatase is often observed. This is because PSA is also reflective of soft tissue disease while bone alkaline phosphatase is a more relevant parameter related to proliferating osteoblasts reacting to tumor cells.23
TREATMENT RESPONSE ASSESSMENT
Documentation of response to 223RaCl2 therapy with imaging is not well established and needs evaluation in the setting of available PET radiotracers and optimal timing for assessment after therapy.24, 25 18F-NaF PET, like 99mTc-based bone scintigraphy, may show treatment flare in responding lesions. In one case report in a patient receiving 223RaCl2 therapy for metastatic CRPC, both bone pain and PSA level increased initially but during follow-up bone scan showed evidence of response with marked decline in bone pain and PSA level26 (Figure 1). Keizman and colleagues suggest that radiological flare may be noted during the first 3 months of treatment and should not be considered as progression. Potential extraskeletal progression may be assessed with CT in case of PSA rise or other suspicious signs and symptoms.27 Kairemo and Joensuu evaluated retrospectively the use of 18F-NaF PET/CT in assessment of response after the first and sixth cycles of 223RaCl2 in 10 patients with metastatic CRPC.28 A modified version of PERCIST (PET Response Criteria in Solid Tumors) criteria was used. A reduction (range 6.9–68.4%) of 18F-NaF localization in bone lesions was noted after all 6 cycles of 223RaCl2 in accordance with decline in PSA level. Such decline was not seen after 1 cycle of therapy and in fact in 2 patients, flare might have been present.
Figure 1.
Radionuclide bone scan trend and response to 223RaCl2 therapy. (a) June 2008, metastatic prostate cancer diagnosis. (b) May 2010, disease progression on combined androgen blockade. (c) May 2011, disease progression following sipuleucel-T immunotherapy. (d) December 2011, disease progression just prior to docetaxel chemotherapy. (e) June 2013, widespread disease progression associated with severe diffuse bone pain on enzalutamide (“pre-223RaCl2”). (f) February 2014, dramatic theranostic clinical and bone scan response 2o months after completing six treatments of 223RaCl2 (“post-223RaCl2”). From McNamara et al.26 Courtesy of BioMed Central under the Creative Commons Attribution.26
The repeatability of 18F-NaF quantitation is important, if it is destined to be a useful imaging tool in the assessment of response to therapy. In a multi-center investigation of 35 males with metastatic CRPC, test-retest 18F-NaF PET/CT scan were performed within 3 ± 2 days apart.29 Using sufficiently large bone lesions (>1.5 cm3), the authors showed that at the lesion level, the coefficient of variation for maximum standardized uptake value (SUVmax) and SUVmean were 14.1 and 6.6%, respectively. At the patient level, the coefficient of variation for SUVmax and SUVmean were 12.0 and 5.3%, respectively. Despite these differences, the authors felt that 18F-NaF PET/CT repeatability level at a particular imaging site might be sufficient for clinical quantification of response of bone metastases to therapy. Another study showed that there could also be differences in relation to type of the PET imaging system, particularly with respect to use of time-of-flight (TOF) measurements.30 Osseous metastatic lesions showed higher SUVmean values (by about 2.5%), while normal soft tissues showed lower SUVmean with TOF. These studies suggest that it is most optimal to use the same type of imaging systems in follow-up of patients before and after therapy to reduce the effects of variability in lesion quantitative measures.
Murray et al investigated whether the bone lesion absorbed dose after administration of 223RaCl2 correlated with level of 18F-NaF uptake in 5 patients with metastatic CRPC.31 There was a high correlation (r = 0.96) between 223RaCl2 absorbed dose and18F-NaF uptake suggesting that those metastatic lesions with higher 18F-NaF uptake will also receive higher 223RaCl2 dose, in alignment with the general concept of theranostiscs with these paired agents.
Cook and colleagues performed 18F-NaF PET/CT in 5 patients with CRPC and only bone metastases before, at 6 weeks and at 12 weeks after 223RaCl2 therapy (100 KBq kg−1 at 0 and 6 weeks).32 The mean SUVmax of 5 selected bone metastases decreased at 12 weeks in correspondence to PSA decline in 3 patients and decline in alkaline phosphatase in 4 patients. This study suggested that 18F-NaF PET/CT may be useful in assessing response to 223RaCl2 therapy. Similar utility of 18F-NaF PET/CT in assessing response has been reported with chemotherapy and androgen receptor pathway inhibitors which can also be predictive of progression-free survival.33 In an imaging companion trial [American College of Radiology Imaging Network (ACRIN) 6687] to a multicenter metastatic CRPC tissue biomarker-guided therapeutic trial (NCT00918385), Yu et al showed statistically significant decline in SUVmaxavg (average of SUVmax of up to 5 lesions) in response to dasatinib, which also correlated with changes in bone alkaline phosphatase but not PSA levels.34
The extent of bone disease on bone scintigraphy, including 18F-NaF PET/CT, may have significant implications on experience of hematotoxicity and the number of 223RaCl2 cycles that may be tolerated with higher bone tumor load (i.e. less bone marrow reserve) associated with higher rate of Grade 3 or above toxicity and early therapy termination.35 Finally, a large cohort national study (National Oncologic PET Registry, NOPR) has shown that 18F-NaF PET is not only useful in treatment response assessment but also can impact management in up to 40% of patients.36 More recent NOPR data analysis revealed not only concordance between intended post-18F-NaF PET management plans and inferred management based on inquiry of Medicare claims but also an association between 18F-NaF PET results and subsequent hospice claims and patient survival.37, 38
PROGNOSIS AND PREDICTION OF OUTCOME
The baseline burden of metastatic disease has been demonstrated to be predictive of outcome. Jadvar et al showed in a prospective study of 87 patients with metastatic CRPC undergoing FDG PET/CT that the continuous parameter sum of SUVmax of up to 25 metastatic lesions (both bone and soft tissue) was significantly (p = 0.053) predictive of overall survival with a hazard ratio of 1.01 [95% confidence interval (CI), 1.001–1.02]. When sum of SUVmax was grouped into quartile ranges, there was poorer survival probability for the patients in the fourth-quartile range than for those in the first-quartile range, with a univariate hazard ratio of 3.8 [95% CI (1.8–7.9)].39 A recent investigation focusing only on bone metastases arrived at a similar conclusion that baseline skeletal tumor burden as demonstrated on 18F-NaF PET scintigraphy is a prognostic biomarker of overall survival and hematological toxicity in patients receiving 223RaCl2 therapy.40
Zukotynski and colleagues evaluated the prognostic utility of 18F-NaF and FDG in 9 patients with metastatic CRPC in a Phase 1 trial of abiraterone and cabozantinib therapy.41 The Kaplan-Meier survival analysis showed a statistically significant difference in progression free survival (PFS) among males with widespread FDG-avid, oligometastatic FDG-avid, and non-FDG-avid disease. Extent of osseous disease on 18F-NaF PET/CT was not predictive of treatment response. The authors concluded that FDG might be superior to 18F-NaF in the imaging evaluation of response to treatment with abiraterone and cabozantinib. However, in another relatively similar study evaluating zibotentan therapy, the authors suggested that FDG and 18F-NaF uptake levels in lesions may be spatially dislocated and both tracers provide useful complementary information.42
Apolo et al reported on a prospective pilot study of 18F-NaF PET/CT in predicting survival in patients with prostate cancer.43 The patient cohort included 30 males with and 30 males without bone metastases on 99mTc-based bone scintigraphy. 18F-NaF PET/CT was performed at baseline, 6-month and 12-month, and the finding were correlated with PSA and overall survival. 18F-NaF PET/CT detected more bone lesions than 99mTc-based bone scintigraphy. The number of lesions on PET (extent of disease) correlated significantly with outcome. In fact, measurement of burden of active osseous metastatic disease on18F-NaF PET/CT is feasible and has been shown to be predictive of overall survival and risk of skeletal related events in patients treated with 223RaCl2 44, 45 (Figure 2). In order to determine suitable semi-quantitative parameters for skeletal tumor burden, an SUVmax threshold of 10 was used to calculate the fluoride tumor volume within a whole-body volume of interest (FTV10) which excluded non-metastatic high activity sites such as renal collecting system, degenerative disease, and healing fractures. The total lesion fluoride uptake was calculated as a product of mean SUVmax and volume of interest (TLF10). The parameter TLF10 was not only reproducible but at a cutoff level of 8000 was able to discriminate survivors from non-survivors after 223RaCl2 therapy.45
Figure 2.
Contrasting patients with sequential 18F-NaF PET/CT before and after treatment with 223RaCl2. (a) Patient #1’s baseline 18F-NaFPET/CT demonstrates widespread osteoblastic metastases with high 18F-NaF uptake. (b) After treatment with 223RaCl2, patient #1 had significant signs of progression, with additional sites of osteoblastic metastases. Patient #2 (c) also had his baseline 18F-fluoride PET/CT demonstrating widespread osteoblastic metastases with high uptake. Fortunately, patient #2 responded to treatment with 223RaCl2 with marked skeletal tumor burden reduction (d). From: Rohren et al.44 © by the Society of Nuclear Medicine and Molecular Imaging, Inc.44
We assessed our one-year experience with 223RaCl2 after its approval for use.46 We observed that about one-quarter of our patient cohort of 25 were able to complete the entire six-dose treatment. Advancing soft tissue disease was the primary reason for cessation of therapy in the rest of the group. The adverse events were mild and manageable. We also noted that in responding patients, there was a decline in serum alkaline phosphatase, which was more common than a decline in PSA level. The investigators from MD Anderson Cancer Center reported retrospectively on a similar experience with 110 patients who underwent 223RaCl2 therapy.47 They noticed treatment-related significant reduction in alkaline phosphatase and pain score. On multivariate analysis, the total number of 223RaCl2 cycles and concomitant use of abiraterone were significantly associated with overall, progression-free, and bone event-free survivals.
COMPARATIVE UTILITY OF 18F-NAF TO OTHER PET RADIOTRACERS
Bone imaging with 18F-NaF is inherently indirect for identification of disease sites that have altered the bone microenvironment. It is of interest to decipher how bone imaging relates to direct imaging of marrow disease (particularly early involvement without major bony reactive changes) with the current and forthcoming PET agents. Beheshti and colleagues compared18F-NaF PET/CT and 18F-fluorocholine (FCH) in patients with prostate cancer.48 They concluded that although 18F-NaF PET/CT is highly sensitive for detection of bone metastases but FCH PET/CT provides greater specificity and can also detect early marrow involvement before there is significant osteoblastic reaction. FCH PET/CT may also be helpful in treatment response assessment and provide prognostic information (e.g. PSA progression) by monitoring changes in whole-body choline-avid tumor burden. In a cohort of 42 CRPC patients who underwent FCH PET/CT at baseline and 3 months after start of treatment (chemotherapy in 16, antiandrogens in 19, 223RaCl2 in 5, and sipuleucel-T in 2), a significant decline (≥30%) in net metabolically active tumor volume (MATV) (i.e. MATV response) was significantly associated with longer time to PSA progression in comparison to those patients who did not display a MATV response (481 vs 116 d, p = 0.0067), regardless of treatment type.49 Miyazaki et al reported similar results noting significant decline in MATV and total lesion activity (TLA) on FCH PET/CT in patients responding favorably to 223RaCl2 treatment.50 In one comparative study of 99mTc-MDP bone scintigraphy, 18F-NaF PET/CT and FCH PET/CT in 50 males with spinal metastases, the diagnostic performance of PET/CT studies were significantly higher than 99mTc-MDP bone scintigraphy prompting the investigators to question the value of 99mTc-MDP bone scintigraphy when PET/CT (with either 18F-NaF or FCH) is available.51 The sensitivity and specificity were 51 and 82% for 99mTc-MDP bone scintigraphy, 93 and 54% for 18F-NaF PET/CT, and 85 and 91% for FCH PET/CT with MRI used as standard of reference with characterizing bone lesions as likely malignant or benign. 18F-NaF PET/CT provides high sensitivity with FCH PET/CT improving the specificity and both have been suggested to be included in the relevant guidelines as alternative to 99mTc-MDP bone scintigraphy in this clinical setting.52, 53
Imaging with radiolabeled agents targeted to the prostate specific membrane antigen (PSMA) in conjunction with PET/CT has received much attention over the past few years and is anticipated to exert a major transformational change in management of prostate cancer.54 A recent prospective multi-center Australian investigation involving 431 patients with prostate cancer (primary staging of intermediate-high risk disease in 25% and restaging/biochemical recurrence in 75% of patients) reported an overall change in planned management of about half of the patients with 68Ga-PSMA-11 PET/CT.55 PSMA PET/CT may also be useful to more accurately assess the extent of disease prior to a planned treatment with 223RaCl2 based on bone scintigraphy. For example, the investigators from Münster, Germany, showed in a small cohort that 3 of their 4 patients with previously unknown visceral metastases (hence initially thought to be candidates for 223RaCl2 therapy) were in fact not suitable for such treatment and that 68Ga-PSMA PET/CT can be useful as gatekeeper in patient management in this clinical setting.56, 57 Ahmadzadehfar and colleagues also demonstrated that 68Ga-PSMA-11 can be a useful gatekeeper for 223RaCl2 treatment in order to select patients who might benefit most from such therapy58 (Figure 3). They also showed that PSA increase during therapy cycles is related to disease progression. The same group of investigators reported in a single case report that 68Ga-PSMA-11 can be useful in documenting the tumoricidal effect of 223RaCl2 therapy.59 Radiolabeled PSMA PET may also be advantageous and more sensitive over 18F-NaF for differentiation of treatment-induced “flare” from frank progressive disease as has been shown in single case reports.60, 61
Figure 3.
A 69-year-old patient with hormone-refractory prostate cancer (Gleason score, 8) who underwent bone scintigraphy (a) was referred for 223RaCl2 therapy. 68Ga-PSMA PET/CT (b) showed multifocal bone metastases concordant to bone scintigraphy. PSA level at time of PET imaging was 30 ng ml−1, and ALP was 207 U l−1. Patient underwent 6 cycles of therapy with 223RaCl2 therapy and 4 weeks later showed near-complete response by 68Ga-PMSA PET (c) and partial response by bone scintigraphy (d). PSA and ALP decreased to 2.95 ng ml−1 and 98 U l−1, respectively. From: Ahmadzadehfar et al.58 © by the Society of Nuclear Medicine and Molecular Imaging, Inc.58
CONCLUSIONS
Precision medicine is entirely aligned with theranostics, which is now emerging as an important systematic tool for targeted “see & treat” management of patients with cancer. Imaging with 18F-NaF PET/CT and radionuclide therapy with 223RaCl2 target osteoblastic metastatic bone lesions. Although the most optimal imaging method for assessing response to 223RaCl2 therapy is not well established, 18F-NaF PET can be useful if it is performed sufficiently after the end of treatment (about 2–3 months) to reduce the probability of treatment flare that can simulate progression. Pre-treatment total fluoride avid bone metastases correlate with patient outcome. The utility of 18F-NaF in comparison to other relevant PET radiotracers in the clinical setting of prostate cancer, particularly radiolabeled PSMA ligands, needs further exploration.
Contributor Information
Hossein Jadvar, Email: jadvar@med.usc.edu.
Patrick M Colletti, Email: colletti@usc.edu.
Funding
H Jadvar was supported in part by the National Institutes of Health grants R01-CA111613, R21-CA142426, R21-EB017568, and P30-CA014089.
REFERENCES
- 1. New directions in diagnostics. Med Device Technol 2000; 11: 33. [PubMed] [Google Scholar]
- 2. Jaffee EM, Dang CV, Agus DB, Alexander BM, Anderson KC, Ashworth A, et al. Future cancer research priorities in the USA: a Lancet Oncology Commission. Lancet Oncol 2017; 18: e653–e706. doi: 10.1016/S1470-2045(17)30698-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Crawford ED, Higano CS, Shore ND, Hussain M, Petrylak DP. Traeting patients with metastatic castration resistant prostate cancer: a comprehensive review of available therapies. J Urol 2015; 194: 1537–47. doi: 10.1016/j.juro.2015.06.106 [DOI] [PubMed] [Google Scholar]
- 4. Heidenreich A, Bastian PJ, Bellmunt J, Bolla M, Joniau S, van der Kwast T, et al. EAU guidelines on prostate cancer. Part II: treatment of advanced, relapsing, and castration-resistant prostate cancer. Eur Urol 2014; 65: 467–79. doi: 10.1016/j.eururo.2013.11.002 [DOI] [PubMed] [Google Scholar]
- 5. Fitzpatrick JM, Bellmunt J, Fizazi K, Heidenreich A, Sternberg CN, Tombal B, et al. Optimal management of metastatic castration-resistant prostate cancer: highlights from a European expert consensus panel. Eur J Cancer 2014; 50: 1617–27. doi: 10.1016/j.ejca.2014.03.010 [DOI] [PubMed] [Google Scholar]
- 6. Lorente D, Fizazi K, Sweeney C, de Bono JS. Optimal treatment sequence for metassativc castration-resistant prostate cancer. Eur Urol Focus 2016; 2: 488–98. doi: 10.1016/j.euf.2016.10.008 [DOI] [PubMed] [Google Scholar]
- 7. Galletti G, Leach BI, Lam L, Tagawa ST. Mechanisms of resistance to systemic therapy in metastatic castration-resistant prostate cancer. Cancer Treat Rev 2017; 57: 16–27. doi: 10.1016/j.ctrv.2017.04.008 [DOI] [PubMed] [Google Scholar]
- 8. Iagaru AH, Mittra E, Colletti PM, Jadvar H. Bone-targeted imaging and radionuclide therapy in prostate cancer. J Nucl Med 2016; 57(Suppl 3): 19S–24. doi: 10.2967/jnumed.115.170746 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Langsteger W, Rezaee A, Pirich C, Beheshti M. 18F-NaF-PET/CT and99mTc-MDP bone scintigraphy in the detection of bone metastases in prostate cancer. Semin Nucl Med 2016; 46: 491–501. doi: 10.1053/j.semnuclmed.2016.07.003 [DOI] [PubMed] [Google Scholar]
- 10. Jadvar H. Targeted radionuclide therapy: an evolution toward precision medicine. AJR Am J Roentgenol 2017; 209: 277–88. doi: 10.2214/AJR.17.18264 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Zukotynski K, Jadvar H, Capala J, Fahey F. Targeted radionuclide therapy: practical applications and future prospects. Biomark Cancer 2016; 8(Suppl 2): BIC.S31804. doi: 10.4137/BIC.S31804 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Parker C, Nilsson S, Heinrich D, Helle SI, O’Sullivan JM, Fosså SD, et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 2013; 369: 213–23. doi: 10.1056/NEJMoa1213755 [DOI] [PubMed] [Google Scholar]
- 13. Parker CC, Coleman RE, Sartor O, Vogelzang NJ, Bottomley D, Heinrich D. Three-year safety of radium-223 dichloride in patients with castration-resistant prostate cancer and symptomatic bone metastases rom phase 3 randomized Alpharadin in symptomatic prostate cancer trial. Eur Urol 2017; [Epub ahead of Print]. [DOI] [PubMed] [Google Scholar]
- 14. Kluetz PG, Pierce W, Maher VE, Zhang H, Tang S, Song P, et al. Radium Ra 223 dichloride injection: U.S. food and drug administration drug approval summary. Clin Cancer Res 2014; 20: 9–14. doi: 10.1158/1078-0432.CCR-13-2665 [DOI] [PubMed] [Google Scholar]
- 15. Buroni FE, Persico MG, Pasi F, Lodola L, Nano R, Aprile C. Radium-223: insight and perspectives in bone metastatic castration-resistant prostate cancer. Anticancer Res 2016; 36: 5719–30. doi: 10.21873/anticanres.11155 [DOI] [PubMed] [Google Scholar]
- 16. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: prostate cancer version 2. 2017. Available from: https://www.nccn.org/professionals/physician_gls/pdf/prostate.pdf [Accessed 23 November 2017]. [DOI] [PMC free article] [PubMed]
- 17. National Institute for Health and Care Excellence. Radium-223 dichloride for treating hormone-relapsed prostate cancer with bone metastases. 2016. Available from: https://www.nice.org.uk/guidance/ta412 [Published 28 September 2016].
- 18. Cornford P, Bellmunt J, Bolla M, Briers E, De Santis M, Gross T, et al. EAU-ESTRO-SIOG guidelines on prostate cancer. Part II: treatment of relapsing, metastatic, and castration-resistant prostate cancer. Eur Urol 2017; 71: 630–42. doi: 10.1016/j.eururo.2016.08.002 [DOI] [PubMed] [Google Scholar]
- 19. Du Y, Carrio I, De Vincentis G, Fanti S, Ilhan H, Mommsen C, et al. Practical recommendations for radium-223 treatment of metastatic castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 2017; 44: 1671–8. doi: 10.1007/s00259-017-3756-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Saad F, Carles J, Gillessen S, Heidenreich A, Heinrich D, Gratt J, et al. Radium-223 and concomitant therapies in patients with metastatic castration-resistant prostate cancer: an international, early access, open-label, single-arm phase 3b trial. Lancet Oncol 2016; 17: 1306–16. doi: 10.1016/S1470-2045(16)30173-5 [DOI] [PubMed] [Google Scholar]
- 21. Wong WW, Anderson EM, Mohammadi H, Daniels TB, Schild SE, Keole SR, et al. Factors associated with survival following Radium-223 treatment for meatsatic castration-resistant prostate cancer. Clin Genitourin Cancer 2017; 15: e969–e975 Epub ahead of print. doi: 10.1016/j.clgc.2017.04.016 [DOI] [PubMed] [Google Scholar]
- 22. McKay RR, Jacobus S, Fiorillo M, Ledet EM, Cotogna PM, Steinberger AE, et al. Radium-223 use in clinical practice and variables associated with completion of therapy. Clin Genitourin Cancer 2017; 15: e289–e298. doi: 10.1016/j.clgc.2016.08.015 [DOI] [PubMed] [Google Scholar]
- 23. Nome R, Hernes E, Bogsrud TV, Bjøro T, Fosså SD. Changes in prostate-specific antigen, markers of bone metabolism and bone scans after treatment with radium-223. Scand J Urol 2015; 49: 211–7. doi: 10.3109/21681805.2014.982169 [DOI] [PubMed] [Google Scholar]
- 24. Etchebehere E, Brito AE, Rezaee A, Langsteger W, Beheshti M. Therapy assessment of bone metastatic disease in the era of223radium. Eur J Nucl Med Mol Imaging 2017; 44(Suppl 1): 84–96. doi: 10.1007/s00259-017-3734-0 [DOI] [PubMed] [Google Scholar]
- 25. Bombardieri E, Setti L, Kirienko M, Antunovic L, Guglielmo P, Ciocia G. Which metabolic imaging, besides bone scan with 99mTc-phosphonates, for detecting and evaluating bone metastases in prostatic cancer patients? An open discussion. Q J Nucl Med Mol Imaging 2015; 59: 381–99. [PubMed] [Google Scholar]
- 26. McNamara MA, George DJ. Pain, PSA flare, and bone scan response in a patient with metastatic castration-resistant prostate cancer treated with radium-223, a case report. BMC Cancer 2015; 15: 371. doi: 10.1186/s12885-015-1390-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Keizman D, Fosboel MO, Reichegger H, Peer A, Rosenbaum E, Desax MC, et al. Imaging response during therapy with radium-223 for castration-resistant prostate cancer with bone metastases-analysis of an international multicenter database. Prostate Cancer Prostatic Dis 2017; 20: 289–93. doi: 10.1038/pcan.2017.6 [DOI] [PubMed] [Google Scholar]
- 28. Kairemo K, Joensuu T. Radium-223-dichloride in castration resistant metasdatic prostate cancer-preliminary results of the response evaluation using F-18-fluoride PET/CT. Diagnostics 2015; 5: 413–27. doi: 10.3390/diagnostics5040413 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Lin C, Bradshaw T, Perk T, Harmon S, Eickhoff J, Jallow N, et al. Repeatability of quantitative 18F-NaF PET: a multicenter study. J Nucl Med 2016; 57: 1872–9. doi: 10.2967/jnumed.116.177295 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Oldan JD, Turkington TG, Choudhury K, Chin BB. Quantitative differences in [18F] NaF PET/CT: TOF versus non-TOF measurements. Am J Nucl Med Mol Imaging 2015; 5: 504–14. [PMC free article] [PubMed] [Google Scholar]
- 31. Murray I, Chittenden SJ, Denis-Bacelar AM, Hindorf C, Parker CC, Chua S, et al. The potential of 223Ra and 18F-fluoride imaging to predict bone lesion response to treatment with 223Ra-dichloride in castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 2017; 44: 1832–1844. doi: 10.1007/s00259-017-3744-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Cook G, Parker C, Chua S, Johnson B, Aksnes AK, Lewington VJ. 18F-fluoride PET: changes in uptake as a method to assess response in bone metastases from castrate-resistant prostate cancer patients treated with 223Ra-chloride (Alpharadin). EJNMMI Res 2011; 1: 4. doi: 10.1186/2191-219X-1-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Harmon SA, Perk T, Lin C, Eickhoff J, Choyke PL, Dahut WL, et al. Quantitative assessment of early [18F]sodium fluoride positron emission tomography/computed tomography response to treatment in men with metastatic prostate cancer to bone. J Clin Oncol 2017; 35: 2829–37. doi: 10.1200/JCO.2017.72.2348 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Yu EY, Duan F, Muzi M, Deng X, Chin BB, Alumkal JJ, et al. Castration-resistant prostate cancer bone metastasis response measured by 18F-fluoride PET after treatment with dasatinib and correlation with progression-free survival: results from American College of Radiology Imaging Network 6687. J Nucl Med 2015; 56: 354–60. doi: 10.2967/jnumed.114.146936 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Miederer M, Thomas C, Beck J, Hampel C, Krieger C, Baqué PE, et al. Haematopoietic toxicity of radium-223 in patients with high skeletal tumour burden. Nuklearmedizin 2015; 54: 197–203. doi: 10.3413/Nukmed-0751-15-06 [DOI] [PubMed] [Google Scholar]
- 36. Hillner BE, Siegel BA, Hanna L, Duan F, Quinn B, Shields AF. 18F-fluoride PET used for treatment monitoring of systemic cancer therapy: results from the National Oncologic PET Registry. J Nucl Med 2015; 56: 222–8. doi: 10.2967/jnumed.114.150391 [DOI] [PubMed] [Google Scholar]
- 37. Hillner BE, Hanna L, Makineni R, Duan F, Shields AF, Subramaniam RM, et al. Intended versus inferred treatment after 18F-fluoride PET performed for evaluation of osseous metastatic disease in the National Oncologic PET Registry. J Nucl Med 2018; 59: 421–6. doi: 10.2967/jnumed.117.205047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Gareen IF, Hillner BE, Hanna L, Makineni R, Duan F, Shields AF, et al. Hospice admission and survival after 18F-fluoride PET performed for evaluation of osseous metastatic disease in the National Oncologic PET Registry. J Nucl Med 2018; 59: 427–33. doi: 10.2967/jnumed.117.205120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Jadvar H, Desai B, Ji L, Conti PS, Dorff TB, Groshen SG, et al. Baseline 18F-FDG PET/CT parameters as imaging biomarkers of overall survival in castrate-resistant metastatic prostate cancer. J Nucl Med 2013; 54: 1195–201. doi: 10.2967/jnumed.112.114116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Fosbøl MØ, Petersen PM, Kjaer A, Mortensen J. 223Ra therapy of advanced metastatic castration-resistant prostate cancer: quantitative assessment of skeletal tumor burden for prognostication of clinical outcome and hematologic toxicity. J Nucl Med 2018; 59: 596–602. doi: 10.2967/jnumed.117.195677 [DOI] [PubMed] [Google Scholar]
- 41. Zukotynski KA, Kim CK, Gerbaudo VH, Hainer J, Taplin ME, Kantoff P, 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. [PMC free article] [PubMed] [Google Scholar]
- 42. Simoncic U, Perlman S, Liu G, Staab MJ, Straus JE, Jeraj R. Comparison of NaF and FDG PET/CT for assessment of treatment response in castration-resistant prostate cancers with osseous metastases. Clin Genitourin Cancer 2015; 13: e7–e17. doi: 10.1016/j.clgc.2014.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Apolo AB, Lindenberg L, Shih JH, Mena E, Kim JW, Park JC, et al. Prospective study evaluating Na 18F PET/CT in predicting clinical outcomes and survival in advanced prostate cancer. J Nucl Med 2016; 57: 886–92. doi: 10.2967/jnumed.115.166512 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Rohren EM, Etchebehere EC, Araujo JC, Hobbs BP, Swanston NM, Everding M, et al. Determination of skeletal tumor burden on 18F-fluoride PET/CT. J Nucl Med 2015; 56: 1507–12. doi: 10.2967/jnumed.115.156026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Etchebehere EC, Araujo JC, Fox PS, Swanston NM, Macapinlac HA, Rohren EM. Prognostic factors in patients treated with 223Ra: the role of skeletal tumor burden on baseline 18F-fluoride PET/CT in predicting overall survival. J Nucl Med 2015; 56: 1177–84. doi: 10.2967/jnumed.115.158626 [DOI] [PubMed] [Google Scholar]
- 46. Jadvar H, Challa S, Quinn DI, Conti PS. One-Year postapproval clinical experience with radium-223 dichloride in patients with metastatic castrate-resistant prostate cancer. Cancer Biother Radiopharm 2015; 30: 195–9. doi: 10.1089/cbr.2014.1802 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Etchebehere EC, Milton DR, Araujo JC, Swanston NM, Macapinlac HA, Rohren EM. Factors affecting 223Ra therapy: clinical experience after 532 cycles from a single institution. Eur J Nucl Med Mol Imaging 2016; 43: 8–20. doi: 10.1007/s00259-015-3185-4 [DOI] [PubMed] [Google Scholar]
- 48. Beheshti M, Vali R, Waldenberger P, Fitz F, Nader M, Loidl W, et al. Detection of bone metastases in patients with prostate cancer by 18F fluorocholine and 18F fluoride PET-CT: a comparative study. Eur J Nucl Med Mol Imaging 2008; 35: 1766–74. doi: 10.1007/s00259-008-0788-z [DOI] [PubMed] [Google Scholar]
- 49. Lee J, Sato MM, Coel MN, Lee KH, Kwee SA. Prediction of PSA progression in castration-resistant prostate cancer based on treatment-associated change in tumor burden quantified by 18F-fluorocholine PET/CT. J Nucl Med 2016; 57: 1058–64. doi: 10.2967/jnumed.115.169177 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Miyazaki KS, Kuang Y, Kwee SA. Changes in skeletal tumor activity on 18F-choline PET/CT in patients receiving 223Radium radionuclide therapy for metastatic prostate cancer. Nucl Med Mol Imaging 2015; 49: 160–4. doi: 10.1007/s13139-014-0314-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Poulsen MH, Petersen H, Høilund-Carlsen PF, Jakobsen JS, Gerke O, Karstoft J, 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: 818–23. doi: 10.1111/bju.12599 [DOI] [PubMed] [Google Scholar]
- 52. Wondergem M, van der Zant FM, van der Ploeg T, Knol RJ. A literature review of 18F-fluoride PET/CT and 18F-choline or 11C-choline PET/CT for detection of bone metastases in patients with prostate cancer. Nucl Med Commun 2013; 34: 935–45. doi: 10.1097/MNM.0b013e328364918a [DOI] [PubMed] [Google Scholar]
- 53. Beheshti M, Rezaee A, Geinitz H, Loidl W, Pirich C, Langsteger W. Evaluation of prostate cancer bone metastases with 18F-NaF and 18F-fluorocholine PET/CT. J Nucl Med 2016; 57(Suppl 3): 55S–60. doi: 10.2967/jnumed.115.169730 [DOI] [PubMed] [Google Scholar]
- 54. Jadvar H, Ballas LK. PSMA PET: transformational change in prostate cancer management? J Nucl Med 2018; 59: 228–9. doi: 10.2967/jnumed.117.203604 [DOI] [PubMed] [Google Scholar]
- 55. Roach PJ, Francis R, Emmett L, Hsiao E, Kneebone A, Hruby G, et al. The impact of 68Ga-PSMA PET/CT on management intent in prostate cancer: results of an Australian prospective multicenter study. J Nucl Med 2018; 59: 82–8. doi: 10.2967/jnumed.117.197160 [DOI] [PubMed] [Google Scholar]
- 56. Bräuer A, Rahbar K, Konnert J, Bögemann M, Stegger L. Diagnostic value of additional68Ga-PSMA-PET before223Ra-dichloride therapy in patients with metastatic prostate carcinoma. Nuklearmedizin 2017; 56: 14–22. doi: 10.3413/Nukmed-0846-16-09 [DOI] [PubMed] [Google Scholar]
- 57. Bode A, Rahbar K, Konnert J, Bögemann M, Stegger L. Benefit of 68Ga-PSMA-PET/CT in patients considered for 223Ra-dichloride therapy. Clin Nucl Med 2016; 41: 951–2. doi: 10.1097/RLU.0000000000001374 [DOI] [PubMed] [Google Scholar]
- 58. Ahmadzadehfar H, Azgomi K, Hauser S, Wei X, Yordanova A, Gaertner FC, et al. 68Ga-PSMA-11 PET as gatekeeper for the treatment of metastatic prostate cancer with 223Ra: proof of concept. J Nucl Med 2017; 58: 438–44. doi: 10.2967/jnumed.116.178533 [DOI] [PubMed] [Google Scholar]
- 59. Ahmadzadehfar H, Schlenkhoff CD, Rogenhofer S, Yordanova A, Essler M. 68Ga-PSMA-11 PET represents the tumoricidal effect of 223Ra in a patient with castrate-resistant metastatic prostate cancer. Clin Nucl Med 2016; 41: 695–6. doi: 10.1097/RLU.0000000000001286 [DOI] [PubMed] [Google Scholar]
- 60. Rowe SP, Mana-Ay M, Javadi MS, Szabo Z, Leal JP, Pomper MG, et al. PSMA-based detection of prostate cancer bone lesions with 18F-DCFPyL: a sensitive alternative to 99mTc-MDP bone scan and NaF PET/CT? Clin Genitourin Cancer 2016; 14: e115–e118. doi: 10.1016/j.clgc.2015.09.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Uprimny C, Kroiss A, Nilica B, Buxbaum S, Decristoforo C, Horninger W, et al. 68Ga-PSMA ligand PET versus 18F-NaF PET: evaluation of response to 223Ra therapy in a prostate cancer patient. Eur J Nucl Med Mol Imaging 2015; 42: 362–3. doi: 10.1007/s00259-014-2922-4 [DOI] [PubMed] [Google Scholar]



