Next Article in Journal
Prediction of Deep Myometrial Infiltration, Clinical Risk Category, Histological Type, and Lymphovascular Space Invasion in Women with Endometrial Cancer Based on Clinical and T2-Weighted MRI Radiomic Features
Next Article in Special Issue
An Apparent Diffusion Coefficient-Based Machine Learning Model Can Improve Prostate Cancer Detection in the Grey Area of the Prostate Imaging Reporting and Data System Category 3: A Single-Centre Experience
Previous Article in Journal
Dendritic Cell Subsets in Melanoma: Pathophysiology, Clinical Prognosis and Therapeutic Exploitation
Previous Article in Special Issue
Histology and PSMA Expression on Immunohistochemistry in High-Risk Prostate Cancer Patients: Comparison with 68Ga-PSMA PET/CT Features in Primary Staging
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Retrospective Multicenter Analysis of the Incidence of Bone-Only Disease at PSMA PET/CT in Castration Resistant Prostate Cancer Patients

1
Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
2
Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
3
Nuclear Medicine Department, IRCCS Azienda Ospedaliero—Universitaria di Bologna, 40138 Bologna, Italy
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(8), 2208; https://doi.org/10.3390/cancers15082208
Submission received: 1 March 2023 / Revised: 30 March 2023 / Accepted: 6 April 2023 / Published: 8 April 2023
(This article belongs to the Special Issue PET/CT in Prostate Cancer)

Abstract

:

Simple Summary

This study aimed to determine how often castration-resistant prostate cancer spreads only to the bones and to identify factors that predict this spread. The researchers looked at PSMA PET scans from 179 men. They found that 20% had cancer that had spread only to their bones, with the most common sites being the spine, ribs, and hips. Men who were diagnosed with lymph-node spread at diagnosis or who had only received hormone treatment were more likely to have bone-only spread. These findings suggest that men with bone-only disease could benefit from personalized treatment that specifically targets the bone. The study highlights the importance of PSMA PET scans in accurately detecting prostate cancer and helping doctors choose the best treatment.

Abstract

PSMA PET/CT has unprecedented accuracy for localization of initial or recurrent prostate cancer (PC), which can be applied in a metastasis-directed therapy approach. PSMA PET/CT (PET) also has a role in the selection of patients for metastasis-directed therapy or radioligand therapy and therapy assessment in CRPC patients. The purpose of this multicenter retrospective study was to determine the incidence of bone-only metastasis in CRPC patients who underwent PSMA PET/CT for restaging, as well as identifying potential predictors of bone-only PET positivity. The study analyzed data from 179 patients from two centers in Essen and Bologna. Results showed that 20.1% of the patients had PSMA uptake only in the bone, with the most frequent lesions located in the vertebrae, ribs, and hip bone. Half half of the patients showed oligo disease in bone and may benefit from a bone-metastasis-directed therapy. Initial positive nodal status and solitary ADT were shown to be negative predictors of osseous metastasis. The role of PSMA PET/TC in this patient population needs to be further explored in terms of its role in the evaluation and adoption of bone-specific therapies.

1. Introduction

Prostate cancer (PC) is a major cause of morbidity and mortality in men worldwide; in the United States, approximately 34,500 prostate cancer deaths have been estimated for 2022, a significant proportion of which were at the metastatic stage [1]; specifically, bone disease is difficult to treat and comes with considerable reduction in quality of life and survival [2]. In Europe, although the trend for prostate cancer specific mortality is decreasing [3], it is still a high burden disease.
PSMA PET/CT, both with 68Gallium or 18Fluorine, has shown unprecedented accuracy for localization of initial or recurrent prostate cancer [4], and it is now a widely used imaging technique in the clinical setting given its high accuracy for staging [5,6] and restaging of patients with PC in a biochemical relapse (BCR) context [7,8,9].
The introduction of PET/CT with more specific tracers for prostate cancer (Choline, Fluciclovine and PSMA) led to a metastasis-directed therapy (MDT) approach, to delay systemic therapy [8,10,11]. There are still limited data available regarding the outcomes consequent on MDT guided by PSMA PET/CT imaging [12], but there is hope this imaging technique can lead to an improvement of overall survival (OS) and progression-free survival (PFS) in these set of patients.
PSMA PET/CT is increasingly being utilized for both initial staging and restaging of prostate cancer, including in patients with castration-resistant prostate cancer (CRPC) [13], and for identifying candidates for radioligand therapy with Lu-PSMA. However, the clinical significance of PSMA PET/CT findings in patients with CRPC remains uncertain. As stated in the EAU guidelines, all the therapeutic recommendations are based on conventional imaging only [14]. Patients with metastatic CRPC (mCRPC) and osseous predominance can benefit from bone-specific therapies both as active treatment and as protective agent for symptomatic skeletal events: The only approved bone-specific drug that is associated with a survival benefit is [223Ra]RaCl2 (Ra223). In 2018, the European Medicines Agency (EMA) concluded its review of Ra223, and given safety issues, has recommended restricting its use to patients who have had two previous treatments for metastatic prostate cancer [15]. The role of PSMA PET/CT in this setting is still unclear.
To avoid skeletal-related events in patients with bone metastasis, various treatments can be used, such as radiotherapy on the bone lesion and the use of bisphosphonates and RANK ligand inhibitors [14]. For RANK ligand inhibitors, in particular, in non-metastatic CRPC (nmCRPC) for conventional imaging, denosumab has been associated with increased bone-metastasis-free survival compared to placebo [16].
Given the high predominance of bone metastasis in CRPC patients and their possible exclusive presence [17], with the current study we wanted to retrospectively analyze the incidence of bone-only PSMA uptake in a population of consecutive CRPC patients who underwent PSMA PET/CT for restaging. The secondary endpoint of the study was to evaluate the presence of potential predictors of bone-only PSMA PET/CT positivity.

2. Materials and Methods

This is a multicentric, retrospective observational cohort study in patients with CRPC—defined in accordance with the EAU guidelines [14]: castrate serum testosterone < 50 ng/dL plus either biochemical progression, defined as PSA increase, or radiological progression, defined as the appearance of new lesions either on bone scan or soft tissue lesion at CT—which consecutively performed their first 68Ga-PSMA PET/CT between January 2019 and December 2019 in two high-volume Nuclear Medicine centers (Essen, Germany, and Bologna, Italy). A total of 68Ga-PSMA PET/CT images were reviewed according to PROMISE Criteria [18].
A univariate analysis with Fisher analysis was performed between bone-only metastasis population and other CRPC patients for age (at diagnosis and at PSMA PET/CT), nodal involvement at primary treatment, ISUP, iPSA, PSA, PSAdt and PSAvel, time elapsed between primary treatment and the PET scan and number of systemic treatments performed during CRPC status before PET.
All patients gave written informed consent to undergo PSMA PET. This retrospective study was approved by the respective ethic review boards (Essen: 19-8570-BO; Bologna: 244/2016/O/Oss) and necessity for study specific consent was waived.

3. Results

A total of 179 patients were considered for analysis (65 from Bologna and 114 from Essen); 36 out of 179 patients (20.1%) showed PSMA uptake only in the bone.
Of these 36 patients, the median age at diagnosis was 64 y (IQR, 58.0–69.0 y), median age at PET/CT was 74.0 y (IQR, 67.0–78.3 y), median ISUP grade was 4 and median iPSA was 10.0 ng/mL (IQR, 6.3–36.9 ng/mL). Median PSA before PET/CT was 5.16 ng/mL (IQR, 1.5–12.0 ng/mL), PSAdt 2.8 months (IQR 2.0–5.1 months) and PSAvel 8.4 ng/mL/year (IQR 2.9–35.9). Median time elapsed between primary treatment and the PET scan was 6 years (IQR 4–10) and the median number of systemic treatments performed during CRPC status was 1 (Summarized in Table 1).
In a per-patient analysis, 50.0% of the patients (18/36) showed focal PSMA uptake in three lesions or fewer (oligometastatic), 41.7% (15/36) showed focal PSMA uptake in more than three lesions, and 8.3% (3/36) showed a diffuse bone marrow PSMA uptake.
In Table 2, it is interesting to note that out of the 18 patients with oligometastatic disease, 6/18 (33.3%) had a focal uptake in only one rib with either a PSMA uptake greater than the liver or a corresponding CT correlate. It can also be noted that 72.2% of the PSMA uptake of most representative lesions corresponded to CT pathological sclerotic findings while 8.3% had no corresponding finding on CT, and 19.4% were in correspondence of lytic lesions on CT.
In a per-patient analysis, the areas of increased PSMA uptake were localized: 30.6% (11/36) sacrum (median SUVmax 19.2 g/mL, IQR 8.9–24.3); 44.4% (16/36) hip bone (median SUVmax = 23.6 g/mL, IQR 12.5–24.3); 55.6% (20/36) vertebrae (median SUVmax = 23.5 g/mL, IQR 16.4–35.8); 36.1% (13/36) sternum (median SUVmax = 13.2 g/mL, IQR 6.7–23.9); 55.6% (20/36) ribs (median SUVmax = 13.8 g/mL, IQR 7.8–79.3); 22.2% (8/36) femur (median SUVmax = 16.8 g/mL, IQR 9.0–23.7); 19.4% (7/36) humerus (median SUVmax = 14.9 g/mL, IQR 9.8–21.9); 27.8% (10/36) scapula (median SUVmax = 17.9 g/mL, IQR 9.9–24.6); 13.9% (5/36) clavicula (median SUVmax = 17.2 g/mL, IQR 8.1–23.3); and 13.9% (5/36) in the skull (median SUVmax = 15.2 g/mL, IQR 11.6–21.3) (summarized in Table 3).
Regarding the secondary aim of the study (predictors of bone-only PSMA PET/CT positivity), at the univariate analysis, we observed that pN1 patients at presentation were more likely to show bone-only disease at PSMA PET/CT (p < 0.01) if compared with other CRPC patients. Moreover, patients who received only ADT as primary treatment were more likely to show bone-only uptake at PSMA PET/CT when classified as CRPC (p < 0.01).
The other analyzed factors (age both at diagnosis and at PSMA PET/CT, ISUP, iPSA, PSA, PSAdt, and PSAvel, time elapsed between primary treatment and the PET scan and number of systemic treatments performed during CRPC status before PET) did not show any statistical significance.
The p-values retrieved by the Fisher exact test are summarized in Table 4.

4. Discussion

To our knowledge, this is the first study evaluating the incidence of bone-only PSMA focal uptake in the CRPC population. In this analysis, the incidence of bone-only uptake was 20% of all the patients who received a PSMA PET during their CRPC status.
In a per-patient analysis, we found that half of the patients showed an oligometastatic spread of the disease only (three or fewer focal PSMA lesions, according to PROMISE Criteria [18]).
Two recent phase II randomized trials, ORIOLE [19] and STOMP [20], showed interesting results for MDT.
The ORIOLE used only stereotactic ablative radiation (SABR) in the treatment arm, whereas the STOMP trial included patients whose metastases were treated with different techniques, either radiotherapy or surgery.
Regarding this difference in treatments, both trials showed that for patients with oligometastatic hormone-sensitive PC, MTD is safe and improves androgen-deprivation-therapy (ADT)-free survival when compared to surveillance only.
However, neither of the studies used PSMA PET/CT as an inclusion criterion: STOMP used Choline PET/CT, whereas ORIOLE used conventional imaging. The latter one, for patients randomized in the treatment SABR arm, performed a PSMA PET/CT and the investigative team was blinded to the PSMA PET/CT results: this resulted in 16 out of 36 patients with PSMA avid lesions not being included in the treatment field [19]. The median progression-free survival (PFS) (rise of PSA or radiologic progression or initiation of ADT) was unreached among participants with no untreated lesions vs. 11.8 months among participants with any untreated lesions [19].
It has now been reported in the literature that PSMA PET/CT has a higher sensitivity than CT and bone scan, leading to a change in patient management [21,22], especially in the staging setting.
A recent retrospective multicenter study by Zamboglou et al. [23] used PSMA PET/CT to guide salvage radiotherapy, and the metastasis-free survival at 4 years was 83% of the 815 patients.
These findings support the use of PSMA PET/CT for MTD; the concept that oligometastatic disease may have a different biological potential without a fully developed metastatic power has arisen since a 1995 study by Hellmann et al. [24], and at this stage there may still be the possibility to have a curative intent for PC [19].
In this context, it may be hypothesized that these patients could benefit from an MDT approach, not only for pain control but in order to delay as much as clinically possible the introduction of advanced systemic therapies and their adverse reaction.
The studies cited, however, had a different population: they had a hormone-sensitive prostate cancer population, whereas our study investigated a CRPC population. To this day, it has not been investigated whether the CRPC population could benefit from an MDT approach.
In a per-patient analysis, roughly 40% of the patients showed a disseminated disease with more than three focal PSMA uptakes in bone segments; at this point of the disease, both for STOMP and for ORIOLE inclusion criteria [19,20], these patients should be considered for a systemic approach including the introduction of bone-protective agents such as bisphosphonates or RANK ligand inhibitors.
A total of 72.2% of patients had a PSMA uptake of the most representative lesion on sclerotic findings at corresponding CT images for patients with multiple symptomatic sclerotic bone metastases, therapy with 223Ra can be considered, according to the selection criteria of the phase III ALSYMPCA trial [25]. Patients eligible for 223Ra therapy have to show more than six lesions at bone scintigraphy, which is now well known to be less sensitive than PSMA PET/CT. In the ALSYMPCA trial, having fewer than six lesions on bone scintigraphy was an unfavorable risk for 223Ra therapy; it is not yet known if the use of PSMA PET/CT could better select the patients, given that having six bone lesions at PSMA PET/CT may not correspond to six lesions at bone scan. At the moment, there is only one registered clinical trial (NCT04951817) evaluating the change of lesion detection number at PSMA PET/CT in patients selected with bone scan for 223Ra therapy [26].
With the 2018 EMA safety concerns, the prescription for this therapy has to be crafted carefully, and future analysis of these concerns is needed.
A total of 8.3% of the patients had no corresponding pathological CT images underneath their most representative lesion, and the question is still open of what the best treatment option for patients without any morphological corresponding finding could be.
It is well known that PSMA PET/CT can have false-positive bone findings, which may lead to inadequate therapy; however, experienced nuclear medicine physicians are able to correctly detect and identify the lesions as unspecific [27].
It has already been shown that using PSMA PET/CT leads to an upstaging in a majority of CRPC patients non-metastatic by conventional imaging [28].
Fendler et al. [29] conducted a retrospective analysis of 200 PSMA PET/CT scans in patients with non-metastatic castration-resistant prostate cancer (nmCRPC), which revealed that 98% of the patients analyzed (196/200) had a positive result on PSMA PET/CT. Among these patients, 16% (31/200) were found to have metastatic disease without evidence of local or pelvic recurrence.
All these patients may benefit from a combined approach of MTD and bisphosphonates or RANK ligand inhibitors.
To this day, it is not clear if the use of bisphosphonates provided early in the course of cancer will be able to prevent the formation of bone metastases or otherwise [30].
A high PSMA uptake demonstrated by bone lesions shows that patients with disseminated bone disease could benefit from a bone-directed and/or PSMA-directed therapy, by VISION Criteria [13]. Patients with a high osseous metastatic burden in PSMA PET/CT have an overall worse prognosis and a higher risk of myelotoxicity when treated with 177Lutetium-PSMA-617 (Lu PSMA) [31]. However, the recent publication of an interim analysis of the RALU study shows the safety and feasibility of the sequential use of first Ra223 (Ra223 and then Lu PSMA [32], both myelotoxic therapies). Moreover, the findings from the WARMTH study showed that a prior therapy with 223Ra had a positive impact on overall survival in patients who were subsequently treated with LuPSMA [33]. Therefore, this patient population can potentially benefit from accurate selection by PSMA PET/CT.
Patients with a more advanced stage at diagnosis (pN1) or treated with ADT only were more likely to show bone-only PSMA uptake when a PSMA PET/CT was performed during their CRPC status. These patients may be considered at a higher risk of showing bone metastasis and may be considered for a more aggressive therapy since diagnosis. A systematic review investigating the role of neoadjuvant ARSI therapy in unfavorable intermediate- and high-risk prostate cancer showed that a pathological complete response is rarely attained but promising results are obtained in terms of positive surgical margins [34]. A more recent publication showed similar results [35] and is awaiting publications of clinical trials [36]. None of the studies cited had PSMA PET/CT as staging diagnostic criteria.
There are various limitations to our study. First, its retrospective design involved collecting clinical information from the nuclear medicine file’s notes and relying on the information provided by the clinician during the examination’s booking. Other limitations are the small sample size considered for analysis, heterogeneous CRPC population (both early and advanced), and lack of follow-up.

5. Conclusions

In this retrospective study, the incidence of PSMA PET/CT bone-only disease in a CRPC population was 20%, showing that PSMA PET/CT reliably identifies bone metastases in CRPC patients, opening the door for bone-targeted therapies and potentially improving treatment sequences. Initial positive nodal status and solitary ADT were shown to be negative predictors of osseous metastasis. The role of PSMA PET/CT in this patient population needs to be further explored in terms of its role in the evaluation and adoption of bone-specific therapies.

Author Contributions

Conceptualization, S.F., K.H. and F.S.; methodology, F.S. and W.P.F.; formal analysis, F.S.; investigation, F.S; data curation, F.S., C.B. and F.B.; writing—original draft preparation, F.S.; writing—review and editing, P.C., W.P.F., C.B. and A.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Ethics Committee of University Hospital Essen (protocol code: 19-8570-BO) and IRCCS AOU of Bologna (protocol code: 244/2016/O/Oss).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data are not available due to privacy and ethics restrictions.

Conflicts of Interest

F.S. declares no conflict of interest. W.P.F. reports fees from SOFIE Bioscience (research funding), Janssen (consultant, speaker), Calyx (consultant, image review), Bayer (consultant, speaker, research funding), Novartis (speaker), and Telix (speaker) outside of the submitted work. C.B. reports fees from ABX (consultant) and Janssen (speakers bureau). P.C. declares no conflict of interest. F.B. declares no conflict of interest. A.F. declares no conflict of interest. K.H. reports personal fees from Bayer, personal fees and other from Sofie Biosciences, personal fees from SIRTEX, non-financial support from ABX, personal fees from Adacap, personal fees from Curium, personal fees from Endocyte, grants and personal fees from BTG, personal fees from IPSEN, personal fees from Siemens Healthineers, personal fees from GE Healthcare, personal fees from Amgen, personal fees from Novartis, personal fees from ymabs, personal fees from Aktis Oncology, personal fees from Theragnostics, personal fees from Pharma15, personal fees from Debiopharm, personal fees from AstraZeneca, personal fees from Janssen. S.F. reports fees from Novartis (consulting or advisory), ANMI (consulting or advisory), Astellas (consulting or advisory), Bayer (advisory and non-financial support), Blue Earth Diagnostics (consulting or advisory), GE Healthcare (consulting or advisory and non-financial support), Jenssen (consulting or advisory), Sofie Biosciences (consulting or advisory), AAA (non-financial support), Curium (non-financial support), Tema Sinergie (non-financial support), Sanofi (non-financial support), and Telix (non-financial support).

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Broder, M.S.; Gutierrez, B.; Cherepanov, D.; Linhares, Y. Burden of Skeletal-Related Events in Prostate Cancer: Unmet Need in Pain Improvement. Support. Care Cancer 2015, 23, 237–247. [Google Scholar] [CrossRef]
  3. Dalmartello, M.; La Vecchia, C.; Bertuccio, P.; Boffetta, P.; Levi, F.; Negri, E.; Malvezzi, M. European Cancer Mortality Predictions for the Year 2022 with Focus on Ovarian Cancer. Ann. Oncol. 2022, 33, 330–339. [Google Scholar] [CrossRef]
  4. Hope, T.A.; Goodman, J.Z.; Allen, I.E.; Calais, J.; Fendler, W.P.; Carroll, P.R. Metaanalysis of 68Ga-PSMA-11 PET Accuracy for the Detection of Prostate Cancer Validated by Histopathology. J. Nucl. Med. 2019, 60, 786–793. [Google Scholar] [CrossRef] [Green Version]
  5. van Kalmthout, L.W.M.; van Melick, H.H.E.; Lavalaye, J.; Meijer, R.P.; Kooistra, A.; de Klerk, J.M.H.; Braat, A.J.A.T.; Kaldeway, H.P.; de Bruin, P.C.; de Keizer, B.; et al. Prospective Validation of Gallium-68 Prostate Specific Membrane Antigen-Positron Emission Tomography/Computerized Tomography for Primary Staging of Prostate Cancer. J. Urol. 2020, 203, 537–545. [Google Scholar] [CrossRef]
  6. Jansen, B.H.E.; Bodar, Y.J.L.; Zwezerijnen, G.J.C.; Meijer, D.; van der Voorn, J.P.; Nieuwenhuijzen, J.A.; Wondergem, M.; Roeleveld, T.A.; Boellaard, R.; Hoekstra, O.S.; et al. Pelvic Lymph-Node Staging with 18F-DCFPyL PET/CT Prior to Extended Pelvic Lymph-Node Dissection in Primary Prostate Cancer-the SALT Trial. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 509–520. [Google Scholar] [CrossRef] [PubMed]
  7. Perera, M.; Papa, N.; Christidis, D.; Wetherell, D.; Hofman, M.S.; Murphy, D.G.; Bolton, D.; Lawrentschuk, N. Sensitivity, Specificity, and Predictors of Positive 68Ga-Prostate-Specific Membrane Antigen Positron Emission Tomography in Advanced Prostate Cancer: A Systematic Review and Meta-Analysis. Eur. Urol. 2016, 70, 926–937. [Google Scholar] [CrossRef] [PubMed]
  8. Morigi, J.J.; Stricker, P.D.; van Leeuwen, P.J.; Tang, R.; Ho, B.; Nguyen, Q.; Hruby, G.; Fogarty, G.; Jagavkar, R.; Kneebone, A.; et al. Prospective Comparison of 18F-Fluoromethylcholine Versus 68Ga-PSMA PET/CT in Prostate Cancer Patients Who Have Rising PSA After Curative Treatment and Are Being Considered for Targeted Therapy. J. Nucl. Med. 2015, 56, 1185–1190. [Google Scholar] [CrossRef] [Green Version]
  9. Afshar-Oromieh, A.; Zechmann, C.M.; Malcher, A.; Eder, M.; Eisenhut, M.; Linhart, H.G.; Holland-Letz, T.; Hadaschik, B.A.; Giesel, F.L.; Debus, J.; et al. Comparison of PET Imaging with a (68)Ga-Labelled PSMA Ligand and (18)F-Choline-Based PET/CT for the Diagnosis of Recurrent Prostate Cancer. Eur. J. Nucl. Med. Mol. Imaging 2014, 41, 11–20. [Google Scholar] [CrossRef] [Green Version]
  10. Steuber, T.; Jilg, C.; Tennstedt, P.; De Bruycker, A.; Tilki, D.; Decaestecker, K.; Zilli, T.; Jereczek-Fossa, B.A.; Wetterauer, U.; Grosu, A.L.; et al. Standard of Care Versus Metastases-Directed Therapy for PET-Detected Nodal Oligorecurrent Prostate Cancer Following Multimodality Treatment: A Multi-Institutional Case-Control Study. Eur. Urol. Focus 2019, 5, 1007–1013. [Google Scholar] [CrossRef] [Green Version]
  11. Calais, J.; Ceci, F.; Eiber, M.; Hope, T.A.; Hofman, M.S.; Rischpler, C.; Bach-Gansmo, T.; Nanni, C.; Savir-Baruch, B.; Elashoff, D.; et al. 18F-Fluciclovine PET-CT and 68Ga-PSMA-11 PET-CT in Patients with Early Biochemical Recurrence after Prostatectomy: A Prospective, Single-Centre, Single-Arm, Comparative Imaging Trial. Lancet Oncol. 2019, 20, 1286–1294. [Google Scholar] [CrossRef]
  12. Mottet, P.N.; Cornford, R.C.N.; van den Bergh, E.; Expert Patient Advocate (European Prostate Cancer Coalition/Europa UOMO); De Santis, S.; Gillessen, J.; Grummet, A.M.; Henry, T.H.; van der Kwast, T.B.; Lam, M.D.; et al. EAU Guidelines. Prostate Cancer; EAU Guidelines Office: Arnhem, The Netherlands, 2022; ISBN 978-94-92671-16-5. Available online: http://uroweb.org/guidelines/compilations-of-all-guidelines/ (accessed on 23 February 2023).
  13. Sartor, O.; de Bono, J.; Chi, K.N.; Fizazi, K.; Herrmann, K.; Rahbar, K.; Tagawa, S.T.; Nordquist, L.T.; Vaishampayan, N.; El-Haddad, G.; et al. Lutetium-177-PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2021, 385, 1091–1103. [Google Scholar] [CrossRef]
  14. EAU Guidelines. Prostate Cancer. Uroweb. Available online: https://uroweb.org/guidelines/prostate-cancer (accessed on 29 January 2023).
  15. EMA. EMA Restricts Use of Prostate Cancer Medicine Xofigo. Available online: https://www.ema.europa.eu/en/news/ema-restricts-use-prostate-cancer-medicine-xofigo (accessed on 23 February 2023).
  16. Fizazi, K.; Carducci, M.; Smith, M.; Damião, R.; Brown, J.; Karsh, L.; Milecki, P.; Shore, N.; Rader, M.; Wang, H.; et al. Denosumab versus Zoledronic Acid for Treatment of Bone Metastases in Men with Castration-Resistant Prostate Cancer: A Randomised, Double-Blind Study. Lancet 2011, 377, 813–822. [Google Scholar] [CrossRef] [Green Version]
  17. Skolarus, T.A.; Wolf, A.M.D.; Erb, N.L.; Brooks, D.D.; Rivers, B.M.; Underwood, W.; Salner, A.L.; Zelefsky, M.J.; Aragon-Ching, J.B.; Slovin, S.F.; et al. American Cancer Society Prostate Cancer Survivorship Care Guidelines. CA Cancer J. Clin. 2014, 64, 225–249. [Google Scholar] [CrossRef] [Green Version]
  18. Eiber, M.; Herrmann, K.; Calais, J.; Hadaschik, B.; Giesel, F.L.; Hartenbach, M.; Hope, T.; Reiter, R.; Maurer, T.; Weber, W.A.; et al. Prostate Cancer Molecular Imaging Standardized Evaluation (PROMISE): Proposed MiTNM Classification for the Interpretation of PSMA-Ligand PET/CT. J. Nucl. Med. 2018, 59, 469–478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Phillips, R.; Shi, W.Y.; Deek, M.; Radwan, N.; Lim, S.J.; Antonarakis, E.S.; Rowe, S.P.; Ross, A.E.; Gorin, M.A.; Deville, C.; et al. Outcomes of Observation vs. Stereotactic Ablative Radiation for Oligometastatic Prostate Cancer: The ORIOLE Phase 2 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 650–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Ost, P.; Reynders, D.; Decaestecker, K.; Fonteyne, V.; Lumen, N.; De Bruycker, A.; Lambert, B.; Delrue, L.; Bultijnck, R.; Claeys, T.; et al. Surveillance or Metastasis-Directed Therapy for Oligometastatic Prostate Cancer Recurrence: A Prospective, Randomized, Multicenter Phase II Trial. J. Clin. Oncol. 2018, 36, 446–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Hofman, M.S.; Lawrentschuk, N.; Francis, R.J.; Tang, C.; Vela, I.; Thomas, P.; Rutherford, N.; Martin, J.M.; Frydenberg, M.; Shakher, R.; et al. Prostate-Specific Membrane Antigen PET-CT in Patients with High-Risk Prostate Cancer before Curative-Intent Surgery or Radiotherapy (ProPSMA): A Prospective, Randomised, Multicentre Study. Lancet 2020, 395, 1208–1216. [Google Scholar] [CrossRef]
  22. Pepe, P.; Pennisi, M. Should 68Ga-PSMA PET/CT Replace CT and Bone Scan in Clinical Staging of High-Risk Prostate Cancer? Anticancer Res. 2022, 42, 1495–1498. [Google Scholar] [CrossRef]
  23. Zamboglou, C.; Strouthos, I.; Sahlmann, J.; Farolfi, A.; Serani, F.; Medici, F.; Cavallini, L.; Morganti, A.G.; Trapp, C.; Koerber, S.A.; et al. Metastasis-Free Survival and Patterns of Distant Metastatic Disease After Prostate-Specific Membrane Antigen Positron Emission Tomography (PSMA-PET)-Guided Salvage Radiation Therapy in Recurrent or Persistent Prostate Cancer After Prostatectomy. Int. J. Radiat. Oncol. Biol. Phys. 2022, 113, 1015–1024. [Google Scholar] [CrossRef]
  24. Hellman, S.; Weichselbaum, R.R. Oligometastases. J. Clin. Oncol. 1995, 13, 8–10. [Google Scholar] [CrossRef] [PubMed]
  25. Parker, C.; Nilsson, S.; Heinrich, D.; Helle, S.I.; O’Sullivan, J.M.; Fosså, S.D.; Chodacki, A.; Wiechno, P.; Logue, J.; Seke, M.; et al. Alpha Emitter Radium-223 and Survival in Metastatic Prostate Cancer. N. Engl. J. Med. 2013, 369, 213–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Tseng, J.-R. Using 68Ga-PSMA PET/CT for the Assessment of Osseous Tumor Burden in MCRPC Patients Undergoing 223Ra-Dichloride Injections, 2022. Available online: https://clinicaltrials.gov/ (accessed on 21 February 2023).
  27. Seifert, R.; Telli, T.; Opitz, M.; Barbato, F.; Berliner, C.; Nader, M.; Umutlu, L.; Stuschke, M.; Hadaschik, B.; Herrmann, K.; et al. Non-Specific PSMA-1007 Bone Uptake Evaluated through PSMA-11 PET, Bone Scan and MRI Triple Validation in Patients with Biochemical Recurrence of Prostate Cancer. J. Nucl. Med. 2022, 64. [Google Scholar] [CrossRef]
  28. Farolfi, A.; Hirmas, N.; Gafita, A.; Weber, M.; Barbato, F.; Wetter, A.; Mei, R.; Pianori, D.; Hadaschik, B.; Herrmann, K.; et al. Identification of PCWG3 Target Populations Is More Accurate and Reproducible with PSMA PET Than with Conventional Imaging: A Multicenter Retrospective Study. J. Nucl. Med. 2021, 62, 675–678. [Google Scholar] [CrossRef] [PubMed]
  29. Fendler, W.P.; Weber, M.; Iravani, A.; Hofman, M.S.; Calais, J.; Czernin, J.; Ilhan, H.; Saad, F.; Small, E.J.; Smith, M.R.; et al. Prostate-Specific Membrane Antigen Ligand Positron Emission Tomography in Men with Nonmetastatic Castration-Resistant Prostate Cancer. Clin. Cancer Res. 2019, 25, 7448–7454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Neville-Webbe, H.L.; Coleman, R.E. Bisphosphonates and RANK Ligand Inhibitors for the Treatment and Prevention of Metastatic Bone Disease. Eur. J. Cancer 2010, 46, 1211–1222. [Google Scholar] [CrossRef] [PubMed]
  31. Gafita, A.; Calais, J.; Grogan, T.R.; Hadaschik, B.; Wang, H.; Weber, M.; Sandhu, S.; Kratochwil, C.; Esfandiari, R.; Tauber, R.; et al. Nomograms to Predict Outcomes after 177Lu-PSMA Therapy in Men with Metastatic Castration-Resistant Prostate Cancer: An International, Multicentre, Retrospective Study. Lancet Oncol. 2021, 22, 1115–1125. [Google Scholar] [CrossRef] [PubMed]
  32. Rahbar, K.; Essler, M.; Pabst, K.M.; Eiber, M.; la Fougère, C.P.; Prasad, V.; Rassek, P.; Hasa, E.; Dittmann, H.; Bundschuh, R.A.; et al. Safety and Survival Outcomes of Lutetium-177-Prostate-Specific Membrane Antigen Therapy in Patients with Metastatic Castration-Resistant Prostate Cancer with Prior Radium-223 Treatment: The RALU Study. J. Nucl. Med. 2022, 64, 574–578. [Google Scholar] [CrossRef]
  33. Ahmadzadehfar, H.; Matern, R.; Baum, R.P.; Seifert, R.; Kessel, K.; Bögemann, M.; Kratochwil, C.; Rathke, H.; Ilhan, H.; Svirydenka, H.; et al. The Impact of the Extent of the Bone Involvement on Overall Survival and Toxicity in MCRPC Patients Receiving [177Lu]Lu-PSMA-617: A WARMTH Multicentre Study. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 4067–4076. [Google Scholar] [CrossRef]
  34. Devos, G.; Vansevenant, B.; De Meerleer, G.; Clinckaert, A.; Devlies, W.; Claessens, F.; Graefen, M.; Steuber, T.; Briganti, A.; de la Taille, A.; et al. Neoadjuvant Treatment with Androgen Receptor Signaling Inhibitors Prior to Radical Prostatectomy: A Systematic Review. World J. Urol. 2021, 39, 3177–3185. [Google Scholar] [CrossRef]
  35. McKay, R.R.; Xie, W.; Ye, H.; Fennessy, F.M.; Zhang, Z.; Lis, R.; Calagua, C.; Rathkopf, D.; Laudone, V.P.; Bubley, G.J.; et al. Results of a Randomized Phase II Trial of Intense Androgen Deprivation Therapy Prior to Radical Prostatectomy in Men with High-Risk Localized Prostate Cancer. J. Urol. 2021, 206, 80–87. [Google Scholar] [CrossRef] [PubMed]
  36. Schweizer, M.A. Phase II Neoadjuvant Study of Apalutamide, Abiraterone Acetate, Prednisone, Degarelix and Indomethacin in Men with Localized Prostate Cancer Pre-Prostatectomy, 2021. Available online: https://clinicaltrials.gov/ (accessed on 26 February 2023).
Table 1. Patient characteristics.
Table 1. Patient characteristics.
CharacteristicMedianIQR
Age at diagnosis (y)64.058.0–69.0
Age at PSMA PET/CT (y)74.067.0–78.3
ISUP Grade4-
Initial PSA (ng/mL)10.06.3–36.9
PSA before PET/CT (ng/mL)5.161.5–12.0
PSA doubling time (months)2.82.0–5.1
PSA velocity (ng/mL/year)8.42.9–35.9
Time elapsed primary therapy to PSMA PET/CT (y)64–10
Systemic treatment performed1-
Table 2. Per patient localizations of PSMA bone uptake.
Table 2. Per patient localizations of PSMA bone uptake.
PatientUnifocal vs. Oligometastatic vs. Disseminated vs. Diffuse Bone Marrow InvolvementNumber of LesionsLocationHighest SuvmaxCt Findings for Representative Lesions
1uni1rib9.3lytic
2uni1rib4.5sclerotic
3diss4sacrum, sternum, ribs11.9sclerotic
4oligo2vertebrae, rib27none
5oligo2vertebrae19.9none
6uni1skull17.2lytic
7uni1rib10sclerotic
8uni1rib26.3sclerotic
9uni1vertebrae25.4sclerotic
10diss5hip bone, femur, ribs8.2sclerotic
11diss7hip bone, femur, homerus, scapula8.2sclerotic
12oligo3sacrum, femur10.3sclerotic
13diss>10hip bone, vertebrae, sternum43.9sclerotic
14diss>10vertebrae, sternum, ribs, scapula74.8none
15diss10hip bone, vertebrae, sternum, ribs, skull28sclerotic
16diss>10sacrum, hip bone, femur, vertebrae, sternum, ribs, scapula, skull70.2sclerotic
17uni1vertebrae14.6sclerotic
18diss>10sacrum, hip bone, verterbrae, ribs scapula29.9lytic
19diss5vertebrae, sternum,30.2sclerotic
20oligo2sacrum, hip bone13.2sclerotic
21dmidiffuse bone marrowdiffuse75.4sclerotic
22uni1hip bone32.4sclerotic
23diss>10sacrum, hip bone, femur, vertebrae, sternum, ribs, scapula15.4sclerotic
24diss5hip bone, femur, vertebrae2.9sclerotic
25uni1hip bone30.4sclerotic
26uni1rib8.6sclerotic
27uni1hip bone3.9lytic
28diss>10hip bone, vertebrae, sternum, ribs, homerus, scapula, clavicula37.9sclerotic
29dmidiffuse bone marrowdiffuse22.9sclerotic
30oligo2vertebrae, hip bone144.8sclerotic
31diss6vertebrae, sternum, ribs, homerus, scapula, clavicula4.5none
32diss>10sacrum, hip bone, femur, vertebrae, sternum, ribs, scapula167.7sclerotic
33uni1sacrum26.9lytic
34dmidiffuse bone marrowdiffuse25.9sclerotic
35uni1rib3.9sclerotic
36diss5hip bone, vertebrae, ribs43.9lytic
Uni = unifocal uptake; Oligo = oligometastatic disease; Diss = disseminated disease; Dmi = diffuse bone marrow involvement.
Table 3. Summary of per-patient localization and SUVmax.
Table 3. Summary of per-patient localization and SUVmax.
Location% of Patients (n)Median SUVmax (g/mL)IQR (g/mL)
Vertebrae55.6% (20/36)23.516.4–35.8
Ribs55.6% (20/36)13.87.8–79.3
Hip bone44.4% (16/36)23.612.5–24.3
Sternum36.1% (13/36)13.26.7–23.9
Sacrum30.6% (11/36)19.28.9–24.3
Scapula27.8% (10/36)17.99.9–24.6
Femur22.2% (8/36)16.89.0–23.7
Humerus19.4% (7/36)14.99.8–21.9
Clavicula13.9% (5/36)17.28.1–23.3
Skull13.9% (5/36)15.211.6–21.3
Table 4. Univariate analysis of the variables taken into consideration.
Table 4. Univariate analysis of the variables taken into consideration.
VariableFisher’s Exact Test p Value
pN1 at primary treatment0.009
ADT as single therapy0.005
Age at diagnosis0.050
Age at PSMA PET/CT0.045
ISUP0.535
initial PSA0.242
PSA at PSMA PET/CT0.326
PSAdt0.829
PSAvel0.526
Time to PSMA PET/CT from primary treatment0.045
Number of systemic treatments performed0.444
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Serani, F.; Fendler, W.P.; Castellucci, P.; Berliner, C.; Barbato, F.; Herrmann, K.; Farolfi, A.; Fanti, S. A Retrospective Multicenter Analysis of the Incidence of Bone-Only Disease at PSMA PET/CT in Castration Resistant Prostate Cancer Patients. Cancers 2023, 15, 2208. https://doi.org/10.3390/cancers15082208

AMA Style

Serani F, Fendler WP, Castellucci P, Berliner C, Barbato F, Herrmann K, Farolfi A, Fanti S. A Retrospective Multicenter Analysis of the Incidence of Bone-Only Disease at PSMA PET/CT in Castration Resistant Prostate Cancer Patients. Cancers. 2023; 15(8):2208. https://doi.org/10.3390/cancers15082208

Chicago/Turabian Style

Serani, Francesca, Wolfgang P. Fendler, Paolo Castellucci, Christoph Berliner, Francesco Barbato, Ken Herrmann, Andrea Farolfi, and Stefano Fanti. 2023. "A Retrospective Multicenter Analysis of the Incidence of Bone-Only Disease at PSMA PET/CT in Castration Resistant Prostate Cancer Patients" Cancers 15, no. 8: 2208. https://doi.org/10.3390/cancers15082208

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop