ArticlePDF AvailableLiterature Review

PSMA PET-CT in the Diagnosis and Staging of Prostate Cancer

Authors:

Abstract and Figures

Prostate cancer is the most common cancer and the second leading cause of cancer death in men. The imaging assessment and treatment of prostate cancer has vastly improved over the past decade. The introduction of PSMA PET-CT has improved the detection of loco-regional and metastatic disease. PSMA PET-CT also has a role in the primary diagnosis and staging, in detecting biochemical recurrence after curative treatment and in metastasis-directed therapy. In this paper we review the role of PSMA PET-CT in prostate cancer.
Content may be subject to copyright.
Citation: Combes, A.D.; Palma, C.A.;
Calopedos, R.; Wen, L.; Woo, H.;
Fulham, M.; Leslie, S. PSMA PET-CT
in the Diagnosis and Staging of
Prostate Cancer. Diagnostics 2022,12,
2594. https://doi.org/10.3390/
diagnostics12112594
Academic Editors: Helle D. Zacho
and Kirsten Bouchelouche
Received: 5 October 2022
Accepted: 24 October 2022
Published: 26 October 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
diagnostics
Review
PSMA PET-CT in the Diagnosis and Staging of Prostate Cancer
Alexander D. Combes 1, Catalina A. Palma 1, Ross Calopedos 1, Lingfeng Wen 2,3, Henry Woo 4,5,
Michael Fulham 2,4 and Scott Leslie 1,4,5,6,*
1Department of Urology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
2Department of Molecular Imaging, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
3Faculty of Engineering and Computer Science, University of Sydney, Sydney, NSW 2006, Australia
4Faculty of Medicine, University of Sydney, Sydney, NSW 2006, Australia
5Department of Urology, Chris O’Brien Lifehouse, Sydney, NSW 2050, Australia
6RPA Institute of Academic Surgery, Sydney, NSW 2050, Australia
*Correspondence: scott.leslie@me.com; Tel.: +61-2-8815-4931
Abstract:
Prostate cancer is the most common cancer and the second leading cause of cancer death
in men. The imaging assessment and treatment of prostate cancer has vastly improved over the
past decade. The introduction of PSMA PET-CT has improved the detection of loco-regional and
metastatic disease. PSMA PET-CT also has a role in the primary diagnosis and staging, in detecting
biochemical recurrence after curative treatment and in metastasis-directed therapy. In this paper we
review the role of PSMA PET-CT in prostate cancer.
Keywords: prostate cancer; imaging; PSMA PET
1. Introduction
Since the advent of PSA testing, prostate cancer management has been fast evolving
and heavily debated. This is in part due to the high prevalence but protracted course of the
disease, coupled with our understanding of tumour biology—an enigma that is inherently
limited by technology used to assess it. PSMA PET-CT is an example of technology that has
influenced our practice. It has resulted in a divergence from traditional algorithms, opened
the realm of theranostics and highlighted as many uncertainties as it has improvements.
By all measures, PCa is the most common cancer in men and one of the most common
cause of cancer deaths amongst men [
1
,
2
]. Routine PSA testing was met with resistance
by opponents touting PCa is an indolent disease that men usually die with, rather than
from [
3
,
4
]. This is not a surprising sentiment considering conflicting mortality benefits from
screening and early treatment published in international literature [
5
7
]. Unfortunately, the
resultant drops in PSA testing also correlated with increased prostate cancer mortality [8].
Furthermore, societal impact of men living with metastatic disease is unequivocal. Approx-
imately, two million men each year are diagnosed with prostate cancer with 10 million
men living with the disease and 700,000 of these living with metastatic disease [9]. Analy-
sis of conflicting organisational policies and management algorithms requires historical
examination of clinical practices.
Traditionally, prostate cancer risk stratification relied predominantly on PSA level &
kinetics, digital rectal examination (DRE), and Gleason score obtained from non-targeted
‘sextant’ template biopsy [
10
]. PSA screening led to early treatment of prostate cancer,
leading to a decline in mortality, but the limited specificity of PSA and DRE likely resulted
in overdiagnosis and overtreatment [
10
]. Furthermore, the classic TRUS sextant biopsy
probably missed a proportion of clinically significant cancers and detected cancers that we
now know are unlikely to warrant immediate treatment [
11
]. In addition to a high overall
error rate in accurate diagnosis of prostate cancer, staging of PCa was also limited by the
poor accuracy of CT and bone scan. Prior to development of MRI and PSMA PET-CT, a
variety of nomograms were then developed to help risk stratify patients diagnosed with
Diagnostics 2022,12, 2594. https://doi.org/10.3390/diagnostics12112594 https://www.mdpi.com/journal/diagnostics
Diagnostics 2022,12, 2594 2 of 25
PCa. For example, the NCCN guidelines use PSA, Gleason score, and clinical stage to
stratify PCa into ‘very low’, ‘low’, ‘intermediate’, ‘high’, and ‘very high’ risk categories,
which guide clinical management before and after definitive therapy [
12
]. Within the limits
of our understanding then, changing nomenclatures and evolving nomograms sought to
better guide our practice.
With the same momentum, the focus partly diverged from the traditional categories
and subclassifications, which are founded on light-microscopy glandular architecture and
serum PSA levels [
13
]. The international community embarked on a deeper molecular and
genetic understanding of PCa, the results of which are difficult to transplant into current
practice [
14
]. Notably, a variety of serum, urine, and biopsy biomarkers have also been
approved to more accurately identify men with high-grade PCa [
15
]. Similarly, imaging
technology evolved to investigate more than cross-sectional architecture alone. The advent
of multi-parametric MRI enriched the risk stratification algorithm in this way. Improved
anatomical detail coupled with sequences used to differentiate tissue characteristics of
internal structures has allowed targeted sampling that has been shown to yield more
clinically significant disease [
16
,
17
]. Some have even proposed the combination of Pi-RADS
score and mRNA urine test to improve PCa detection further [18].
Combining progress in molecular oncology and imaging technology, PSMA PET-CT
represents the next step in our pursuit to better treat men with PCa. It has similarly
superseded traditional staging techniques of CT and bone scan and has been shown to
enhance local staging of MRI. By coupling bio-functional and anatomical information, we
can more accurately detail the extent of a patient’s disease. The broad application of its
utility in modern PCa management will be outlined in the article. For reference PSMA
PET-CT will refer to 68GA- PSMA PET-CT in this article unless specified otherwise
2. Positron Emission Tomography-Computed Tomography (PET-CT)
PET began in the 1980s and employed the tracer-kinetic assay method and tomographic
image reconstruction to provide images of ‘function’. The first PET scanners were built ‘in-
house’ in various research institutions in the northern hemisphere. PET-CT was introduced
as a clinical tool in late 2000 and replaced PET-only scanners. Positrons are positive
electrons, which are emitted from the nucleus of positron emitters (
18
F,
15
O,
11
C,
13
N), have
short half-lives and are usually produced by medical cyclotrons. The positron emitters
are ‘tagged’ or attached to compounds using complex synthetic modules to produce a
PET radiopharmaceutical. The PET radiopharmaceutical then, usually after intravenous
injection in humans in trace amounts (hence, the term ‘PET tracer’), participates but does
not perturb, a biochemical process of interest such as glucose metabolism or receptor
uptake. Positron emitters decay to a stable state by the emission of a positron, which
when in tissue collides with an innocent bystander electron; this collision results in the
annihilation of the electrons and the generation of two 511 keV gamma rays (photons)
that are then detected by crystal detectors in the PET tomograph [
19
]. In prostate cancer,
the most commonly used PET radiopharmaceutical is
68
Ga-PSMA;
68
Ga has a half-life of
68 min
.
68
Ga is usually produced from a
68
Ga-
68
Ga generator with a few exceptions—our
institution and a few other sites in the world produce
68
Ga in a cyclotron [
20
]. In clinical
practice, PET-CT imaging is usually undertaken at a set time, after the intravenous injection
of the PET radiopharmaceutical, referred to as the ‘uptake time’. The uptake time for
68
Ga-PSMA in our institution is 50 min. The uptake of the PET radiopharmaceutical in the
tissue can be quantified by kinetic analysis but this requires continues scanning after the
injection of the PET radiopharmaceutical and this consumes valuable time on the scanner.
So, for the clinic, a semi-quantitative measurement of the uptake, the standardized uptake
value (SUV), that relates uptake to the injected activity of the PET radiopharmaceutical, the
patient’s weight and to time, is used.
Since 2000 there have been progressive improvements in the PET and CT technologies—
smaller more efficient crystals, digital photomultiplier tubes in PET; faster CT scanners with
64- and 128-images slices; better reconstruction techniques, improvements in ‘time-of-flight’
Diagnostics 2022,12, 2594 3 of 25
timing and continuous bed motion. The most recent advance has been the development of
long field of view PET-CT scanners or ‘total body’ scanners. Siemens Healthineers have
introduced the Biograph Vision Quadra (the ‘Quadra’) a scanner with a 106 cm z-axis
field-of-view (FOV) and United Imaging, the Explorer with a 200 cm FOV. Both scanners
have markedly improved sensitivity enabling faster scans, lower injected doses of PET
radiopharmaceuticals and superior image quality. There are less than a dozen such scanners
in use at the present time. The majority of the images used in this paper are from our
Biograph Vision Quadra which was the 2nd such device installed and it went ‘live’ in May
2021. The sensitivity of the Quadra is 16
×
that of conventional PET-CT scanners and it
allows the simultaneous acquisition of data throughout the z-axis extent of the scanner.
3. PSMA PET-CT
PSMA is a surface receptor antigen expressed in prostate tissue and tumour-associated
neovasculature [
21
]. It is a glutamate carboxypeptidase type II non-secreted transmembrane
protein comprising 750 amino acids. Antibodies that bind to the extracellular domain of
PSMA were developed. The first was humanised IgG monoclonal antibody ‘J591
0
, followed
by the DKFZ- PSMA-617 ligand and the peptide-linker unit DOTAGA-(I-y)fk (Sub-KuE),
termed PSMA-I & T [
22
] and these were then radio-labelled. Most published studies report
68-
Ga labelled compound but recently
18
F-labelled PSMA have been used [
23
]. Of relevance
to the interpretation of PSMA PET-CT scans, PSMA expression is found in normal prostatic
tissue albeit to a mild degree, in the salivary and lacrimal glands, nasal space and larynx,
liver, spleen, bowel, the kidneys and the sympathetic ganglia (Figure 1) (20). PSMA uptake
is also found in other tumors including glioblastoma, thyroid, breast, lung, colon and renal
carcinomas. PSMA uptake is also seen in benign tumors—haemangiomas, thyroid and
adrenal adenomas, schwannomas and desmoid tumors—and also in reactive/inflammatory
conditions and Paget’s disease [
21
]. PSMA is overexpressed in almost all PCa by around
100–1000 times the normal level, however the exact reason for this remains unclear [
24
,
25
].
Current hypothesis suggests that functionally PSMA has a role in folate metabolism, with
the extra-cellular unit hydrolyzing glutamated folates released by PCa cells, which are then
utilised to enhance proliferation in PCa cells [
26
]. It’s utility in the diagnosis and staging of
prostate cancer has since become an extremely advantageous imaging modality furthering
international urological practice.
Diagnostics 2022, 12, x FOR PEER REVIEW 4 of 27
(a)
(b)
Figure 1. PSMA PET-CT scan showing normal distribution of PSMA-11. 60 yr old M—Gleason 6
prostate cancer on active surveillance for past 2 years; PSA 7.2; MR imaging—low grade changes
PIRADS 2. PSMA PET-CT:
68
Ga-PSMA - 204 MBq; uptake 52 min; BMI = 29.7; Wt 90 Kg; coronal PET
and corresponding CT slices (soft tissue windows) from left to right. (a)physiological uptake in
lacrimal, submandibular salivary glands, parotid glands, retropharyngeal soft tissue, liver, bowel,
part of spleen and pooling of tracer in bladder; mild reactive tracer uptake in groin and axillary
nodes. (b)—physiological uptake in head and neck; reactive uptake axillary nodes; focal uptake in
apex of prostate gland anterior below the bladder SUV = 9.1; marked uptake/excretion of tracer in
both kidneys.
Figure 1. Cont.
Diagnostics 2022,12, 2594 4 of 25
Diagnostics 2022, 12, x FOR PEER REVIEW 4 of 27
(a)
(b)
Figure 1. PSMA PET-CT scan showing normal distribution of PSMA-11. 60 yr old M—Gleason 6
prostate cancer on active surveillance for past 2 years; PSA 7.2; MR imaging—low grade changes
PIRADS 2. PSMA PET-CT:
68
Ga-PSMA - 204 MBq; uptake 52 min; BMI = 29.7; Wt 90 Kg; coronal PET
and corresponding CT slices (soft tissue windows) from left to right. (a)physiological uptake in
lacrimal, submandibular salivary glands, parotid glands, retropharyngeal soft tissue, liver, bowel,
part of spleen and pooling of tracer in bladder; mild reactive tracer uptake in groin and axillary
nodes. (b)—physiological uptake in head and neck; reactive uptake axillary nodes; focal uptake in
apex of prostate gland anterior below the bladder SUV = 9.1; marked uptake/excretion of tracer in
both kidneys.
Figure 1.
PSMA PET-CT scan showing normal distribution of PSMA-11. 60 yr old M—Gleason
6 prostate
cancer on active surveillance for past 2 years; PSA 7.2; MR imaging—low grade changes
PIRADS 2. PSMA PET-CT:
68
Ga-PSMA - 204 MBq; uptake 52 min; BMI = 29.7; Wt 90 Kg; coronal PET
and corresponding CT slices (soft tissue windows) from left to right. (
a
)—physiological uptake in
lacrimal, submandibular salivary glands, parotid glands, retropharyngeal soft tissue, liver, bowel,
part of spleen and pooling of tracer in bladder; mild reactive tracer uptake in groin and axillary
nodes. (
b
)—physiological uptake in head and neck; reactive uptake axillary nodes; focal uptake in
apex of prostate gland anterior below the bladder SUV = 9.1; marked uptake/excretion of tracer in
both kidneys.
4. Diagnosis
Multiparametric MRI (mpMRI) has improved our ability to select who will benefit from
a prostate biopsy and how to get best yield from that biopsy. The technique has reported
sensitivities of up to 91% for grade group
2 PCa and 95% for grade
group 3
[
27
]. A
meta-analysis of over 15 studies involving men with suspected PCa, showed the average
positive predictive value (PPV) in mpMRI alone for ISUP
2 with PIRADS score of 3,4
and 5 were 16%, 59% and 85%, respectively [
28
]. Several prospective trials including the
PRECISION trial also found that MRI-targeted biopsy had far superior levels of detecting
PCa with ISUP 2 compared to the systematic biopsy approach or MRI alone [29,30].
Like any new technology, the application of mpMRI is not perfect. Ambiguity still
exists about whether biopsy is appropriate for PI-RADS 3 lesions. Specifically, PI-RADS
v2.1 includes modifications in the assessment of lesions in the central zone and the anterior
fibromuscular stroma, evaluation of the transition zone, and revision of the criteria for
characterizing lesions as 2 or 3 on DWI [
17
]. Despite this, accurate interpretation of very
large hyperplastic transitional zones remains difficult. Prostatitis is a common and false-
positive/false-negative rates as high as 14% for ISUP
3 PCa have been reported, indicating
that significant prostate cancer can be missed for 1 in 7 patients [
31
]. Additionally, cases of
MRI-invisible lesions have been reported [
17
]. PET imaging is being used to clarify difficult
cases and augment this space.
Previous nuclear medicine studies using FDG-PET lacked the sensitivity and specificity
to successfully diagnose clinically significant PCa. A large retrospective study investigating
47,109 men who underwent FDG-PET over a 10-year period found 1335 patients with
incidental FDG uptake in the prostate. Only 1 patient with a normal PSA (<2.5 ng/mL)
Diagnostics 2022,12, 2594 5 of 25
and just 40 patients with elevated PSA out of 93 had biopsy confirmed PCa [
32
]. It was not
until the advent of antibodies to PSMA and thereafter the introduction of PSMA PET-CT,
have we been able to identify localised prostate cancer with sufficient accuracy and avoid
excessive and unnecessary prostatic biopsies.
A recent meta-analysis also showed that PSMA PET-CT had pooled sensitivity and
specificity of 0.97 and 0.66, respectively, and a negative likelihood ratio of 0.05 for the
initial detection of PCa in patients with clinical suspicion, using histopathology as the
reference standard [
33
]. The low negative likelihood ratio in this analysis suggests that
PSMA PET-CT may be used to rule out disease in patients with a clinical suspicion of PCa,
potentially avoiding unnecessary biopsies [
33
]. A retrospective analysis comparing PSMA
PET-CT to mpMRI investigated 144 patients who underwent both imaging modalities,
finding that both had excellent rates of detecting PCa but a higher sensitivity for clinically
significant PCa through PSMA PET-CT when compared to mpMRI (94.85% vs. 86.03%;
p= 0.022
) [
34
]. A similar but smaller retrospective study also found PSMA PET-CT to be
more accurate than mpMRI in the diagnosis of PCa, whilst also showing higher rates of
detection of localised bilateral disease and multifocal disease [
35
]. Further studies have
also shown the added benefit PSMA PET-CT can have in intermediate risk PCa (ISUP grade
2 and 3
) demonstrating its higher sensitivity, specificity, PPV and Negative Predictive Value
(NPV) than mpMRI alone [
36
]. One potential advantage of PSMA PET-CT over mpMRI
is that interpretation is not influenced by biopsy-related artefacts such as haemorrhage
or inflammation. Another advantage is clarification of indeterminate lesions within large,
hyperplastic transitional zones, which might be easily missed on biopsy.
A combination of MRI and PSMA PET-CT is also being used to improve the detection
of localised PCa. The PRIMARY prospective clinical trial assessed 291 patients with
suspected prostate cancer using MRI, PSMA-PET-CT or MRI + PSMA PET-CT. Included
patients were MRI and biopsy-naïve men with a raised PSA or suspicious DRE [
37
]. Patients
underwent PSMA PET-CT and MRI, and each were analysed separately. Patients proceeded
to systematic transperineal prostate biopsies, with recommended minimum 18 cores and
additional targeted biopsies when possible. Study found 67% of patients had a positive
MRI (PIRADS 3–5), 73% had positive PSMA PET-CT and 81% patients were positive for
clinically significant PCa when both modalities were used. They found the NPV for these
in clinically significant PCa were 72, 80 and 91% respectively. Similarly, the false negative
rate improved from 17% for MRI compared to 10% using PSMA PET-CT and just 3% using
a combination of MRI and PSMA PET-CT. Of the five patient’s combined MRI and PSMA
PET-CT missed, four had ISUP 2 malignancies and one had ISUP 3 malignancy [
37
]. In
select patients, there appears to be benefits of combining the improved specificity offered
by PET imaging with the improved tumour localization offered by MRI.
5. Local Staging
Accurate assessment of T-stage is crucial to provision of the most appropriate treat-
ment, thereby improving BCR-free survival. A clear understanding of the relationship
between suspected cancer and key local structures (seminal vesicles, neurovascular bundle,
prostate apex, bladder neck and rectum) is paramount for surgical and IMRT planning.
Traditionally, this is detail gleaned on MRI using direct qualitative signs, such as irregular
bulging or disruption of the prostate capsule [
38
]. However, identification of more subtle
signs rely on subjective assessment of neurovascular symmetry and focal low signal inten-
sity in SVs or peri-prostatic fat. MRI has a known low sensitivity for focal (or microscopic)
EPE [
39
]. A recent study further comparing PSMA PET-CT to MRI for identification of
Extracapsular extension (ECE) and seminal vesical invasion (SVI) found that PSMA PET-
CT had a higher sensitivity for detection for ECE compared to MRI (78 vs. 54%) but no
significant difference in SVI (75 vs. 67%) [
40
]. Another study by Skawran et al. (2022) found
that there was similar sensitivities (58% vs. 61%) and specificities (81% vs. 81%) between
the two modalities [
41
]. When slight modifications in dissection can have such profound
Diagnostics 2022,12, 2594 6 of 25
functional and oncological implications, it is not unreasonable that more certainty provided
by PSMA PET-CT is warranted in equivocal cases where nerve sparing surgery is planned.
6. Conventional Staging
The advent of PSMA PET-CT has highlighted the limitations of CT and general
nuclear medicine bone scans (BSs) in staging of PCa. When comparing PSMA PET-CT
to conventional CT and BS, Hofman et al., (2020) reported, in a randomised controlled
trial, that PSMA PET-CT was superior to CT and BS in detecting LNM (92% vs. 65%),
sensitivity (85% vs. 38%) and specificity (98% vs. 91%). Furthermore, there were more
equivocal lesions with CT and BS than with PSMA PET-CT (23% vs. 7%) and a higher
radiation exposure in CT and BS compared to PSMA PET-CT (19.2 mSv vs. 8,4 mSv) [
42
].
The recent meta-analysis by Wang et al., (2021), compared the detection of LNM using
multiparametric prostatic MRI (mpMRI) and PSMA PET-CT, showing PSMA PET-CT to
have superior sensitivity (71% vs. 40%) and similar specificity (92% vs. 92%) (46). They
also demonstrated that PSMA PET-CT was able to detect smaller lymph nodes with an
average avid diameter of 7 mm compared to 11.3 mm using MRI [
43
]. Several studies have
compared the predictive ability of PSMA PET-CT with their histopathological outcomes.
Across these studies the sensitivities had a wide range on a per lesion analysis, varying
from 33–92%, and specificities ranging from 82–100% (Figure 2) [4448].
One of the limiting factors for the utility and accuracy of PSMA PET-CT has been the
variability and size of LNM. In the study by Budaus et al., (2016), the median size of detected
vs. undetected LNM using PSMA PET-CT differed by a mean of 9 mm resulting in 67% of
patients in their study, identified as negative for LNM based on PSMA PET-CT, ultimately
returning as positive on final histopathology [
44
]. Similarly,
Van Leeuwen et al., (2017)
,
found that the mean diameter of true positive lymph nodes were around 5 mm larger than
those with false negative LNM and concluded that almost all (91%) of LNM <5 mm in
diameter and all LNM <2 mm in diameter were undetectable by PSMA PET-CT [
44
,
48
].
Furthermore, Van Kalmthout et al., (2020) performed a prospective multicentre trial on
patients with newly diagnosed PCa with negative Bone scans and MSKCC >10% of nodal
involvement. In their study 97 patients underwent ePLND for increased risk of LNI and
found that PSMA PET-CT had a 42% sensitivity and 91% specificity [
49
]. This highlights
that PSMA PET-CT is an improvement in imaging for the detection of LNM but not a
perfect imaging modality and is not reliable for LNM <5 mm in size.
Despite this, PSMA PET-CT represents the most accurate means of staging prostate
cancer and is impacting our management of PCa. The proPSMA trial, a multicentre ran-
domised two-armed clinical trial of 302 patients, directly compared conventional (CT plus
Bone scan) staging to PSMA PET-CT as first line imaging in patients with high risk PCa
(PSA > 20 ng/mL, ISUP3-5 or/and clinical stage T3 disease) [
42
]. The study aimed to
determine if PSMA PET-CT had superior accuracy than conventional imaging for the identi-
fication of pelvic LN or distant metastases, using a predefined reference standard consisting
of histopathology, imaging and biochemistry at a 6 month follow up. The study found
that PSMA PET-CT had 27% greater accuracy than conventional imaging (
91% vs. 65%
),
with conventional imaging demonstrating lower sensitivity (
38% vs. 85%
) and specificity
(
91% vs. 98%
). The superior accuracy of PSMA PET-CT was also demonstrated in patient
subsets with pelvic nodal disease and distant metastases. Importantly, PSMA PET-CT
altered management (28% vs. 15%) and had lower rates of equivocal findings (
7% vs. 23%
).
This study also added cross-over second line imaging, which showed a management
change in 27% of patients who had PSMA PET-CT as second-line, compared to 5% of
patients who had conventional imaging as second line. These changes entailed a change of
surgical technique (7.4%), usually the addition of an ePLND, and change in radiotherapy
dose (7.4%), mostly an increase in the amount and field of radiotherapy [
42
]. Karagiannis
et al., (2022), also found that PSMA PET-CT modified radiotherapy treatment plans in
approximately 60% of their patients, usually finding further locoregional or metastatic
disease and thus implementing additional systemic therapy [50].
Diagnostics 2022,12, 2594 7 of 25
Diagnostics 2022, 12, x FOR PEER REVIEW 7 of 27
(a)
(b)
(c)
Figure 2. Cont.
Diagnostics 2022,12, 2594 8 of 25
Diagnostics 2022, 12, x FOR PEER REVIEW 8 of 27
(d)
(e)
(f)
Figure 2. Cont.
Diagnostics 2022,12, 2594 9 of 25
Diagnostics 2022, 12, x FOR PEER REVIEW 9 of 27
(g)
Figure 2. Primary staging. 70 yr old M; PSA 42; Gleason 4 + 5 = 9; MR scan—PIRADS 6 at right apex;
PIRADS 4 at left midzone. PSMA PET-CT:
68
Ga-PSMA - 216 MBq; uptake 48 min; BMI = 30.5; Wt 83
Kg. (a): Coronal images—focal uptake midline apex anterior SUV = 26.1 with smaller focus SUV-5.4
right midzone; bulky disease in abdominal nodes; metastasis left humerus; left para-aortic node in
thorax; bony metastasis left side of S1. (b,c): enlarged coronal and transaxial images of left common
iliac/para-aortic nodal disease and uptake in apex of gland; transaxial images—node measures 14
mm SUV = 40.1; X-hairs show location. (d,e): enlarged coronal and transaxial images of right pelvic
nodal that measures 1.4 mm SUV = 5.1; uptake in right midzone and apex as well as disease in left
midzone of gland; X-hairs show location. (f,g): enlarged coronal images of para-aortic/para-oesoph-
ageal nodal disease in mediastinum; node measures 5.5 mm, SUV = 17.2; X-hairs show location.
One of the limiting factors for the utility and accuracy of PSMA PET-CT has been the
variability and size of LNM. In the study by Budaus et al., (2016), the median size of de-
tected vs. undetected LNM using PSMA PET-CT differed by a mean of 9 mm resulting in
67% of patients in their study, identified as negative for LNM based on PSMA PET-CT,
ultimately returning as positive on final histopathology [44]. Similarly, Van Leeuwen et
al., (2017), found that the mean diameter of true positive lymph nodes were around 5 mm
larger than those with false negative LNM and concluded that almost all (91%) of LNM
<5 mm in diameter and all LNM <2 mm in diameter were undetectable by PSMA PET-CT
[44,48]. Furthermore, Van Kalmthout et al., (2020) performed a prospective multicentre
trial on patients with newly diagnosed PCa with negative Bone scans and MSKCC >10%
of nodal involvement. In their study 97 patients underwent ePLND for increased risk of
LNI and found that PSMA PET-CT had a 42% sensitivity and 91% specificity [49]. This
highlights that PSMA PET-CT is an improvement in imaging for the detection of LNM
but not a perfect imaging modality and is not reliable for LNM <5 mm in size.
Despite this, PSMA PET-CT represents the most accurate means of staging prostate
cancer and is impacting our management of PCa. The proPSMA trial, a multicentre ran-
domised two-armed clinical trial of 302 patients, directly compared conventional (CT plus
Bone scan) staging to PSMA PET-CT as first line imaging in patients with high risk PCa
(PSA >20 ng/mL, ISUP3-5 or/and clinical stage T3 disease) [42]. The study aimed to deter-
mine if PSMA PET-CT had superior accuracy than conventional imaging for the identifi-
cation of pelvic LN or distant metastases, using a predefined reference standard consisting
of histopathology, imaging and biochemistry at a 6 month follow up. The study found
that PSMA PET-CT had 27% greater accuracy than conventional imaging (91% vs. 65%),
with conventional imaging demonstrating lower sensitivity (38% vs. 85%) and specificity
(91% vs. 98%). The superior accuracy of PSMA PET-CT was also demonstrated in patient
subsets with pelvic nodal disease and distant metastases. Importantly, PSMA PET-CT
Figure 2.
Primary staging. 70 yr old M; PSA 42; Gleason 4 + 5 = 9; MR scan—PIRADS 6 at right
apex; PIRADS 4 at left midzone. PSMA PET-CT:
68
Ga-PSMA - 216 MBq; uptake 48 min; BMI = 30.5;
Wt 83 Kg. (
a
): Coronal images—focal uptake midline apex anterior SUV = 26.1 with smaller focus
SUV-5.4 right midzone; bulky disease in abdominal nodes; metastasis left humerus; left para-aortic
node in thorax; bony metastasis left side of S1. (
b
,
c
): enlarged coronal and transaxial images of
left common iliac/para-aortic nodal disease and uptake in apex of gland; transaxial images—node
measures 14 mm SUV = 40.1; X-hairs show location. (
d
,
e
): enlarged coronal and transaxial images
of right pelvic nodal that measures 1.4 mm SUV = 5.1; uptake in right midzone and apex as well
as disease in left midzone of gland; X-hairs show location. (
f
,
g
): enlarged coronal images of para-
aortic/para-oesophageal nodal disease in mediastinum; node measures 5.5 mm, SUV = 17.2; X-hairs
show location.
7. ePLND in the Era of PSMA PET-CT
It is not clear, however, if ePLND should still be based on a nomogram calculation
of an individual’s risk of LNM or PSMA PET-CT findings alone. Traditionally, PLND is
considered only valuable for prognosis as correctly identified pN1 patients typically benefit
from adjuvant therapies [
51
]. This is supported by research that has failed to demonstrate
an oncological benefit for lymphadenectomy or its extent [
52
,
53
].With the new lens of
PSMA PET-CT, new studies have demonstrated 50% of suspected LNM lie outside the
boundary of ePLND [
54
]. While not vogue, an ePLND in appropriately selected patients
may remove all positive nodes, thereby maximising local disease control.
Despite the higher accuracy of PSMA PET-CT, it seems clinical factors should still
guide decision to perform ePLND. A recent meta-analysis demonstrates that the sensitivity
of nodal staging does not exceed 60% [
55
]. PSMA radio-guided surgery is also being
investigated and has demonstrated some success in intra-operative identification of LNM
that are too small for the spatial resolution of PSMA PET-CT or MRI [
56
]. The EAU currently
recommends the use of novel nomogram developed by Gandaglia et al. [
57
], which includes
mpMRI targeted biopsy results as one of its parameters, along with PSA, clinical stage
and maximum diameter of index lesion on mpMRI. This nomogram had an AUC of
86%, compared to 82% for Briganti 2017 nomogram. Utilising a cut-off of 7% to identify
candidates for ePLND, this nomogram missed 1.6% of patients with LNM (compared to
4.6% if using Briganti 2017 nomogram). Interestingly, Meijer et al., (2021) assessed the
predictive performance of the commonly used Briganti and MSKCC nomograms with the
addition of PSMA PET-CT and found a substantial improvement in discriminative ability,
from AUCs of 0.76 to 0.82 for Briganti 2019, and 0.71 to 0.77 for MSKCC [58].
Diagnostics 2022,12, 2594 10 of 25
8. Metastatic PCa and PSMA PET-CT
European Association of Urology guidelines currently recommend the use of PSMA
PET-CT for assessment of metastases [
59
]. In population-based studies PCa most commonly
spreads to bone (84%), distant lymph nodes (10.6%), liver (10.2%), and thorax (9.1%) [
60
].
Approximately 18% of men have multiple metastatic sites involved (Figure 3). About
10% of patients with PCa have bone metastasis at presentation, and 33% of the remaining
patients will develop metastases during follow-up [
61
]. Traditional imaging modalities
may be helpful in evaluating distant metastases, with CT able to detect sclerotic bone
lesions and visceral metastases, however CT has been reported to be positive in only 14%
of cases [
62
]. BS has been up until recently the most widely used method to detect bone
metastases in clinical practice due to its low cost. It can detect bone metastases with good
sensitivity and can carry out whole body skeletal examination, however it is non-specific,
with inflammation confounding metastatic deposits. Moreover, it reportedly only has a
positive rate of 5% when PSA < 7 ng/mL [
62
], making it an imaging technique more suited
to patients with very high PSA ranges and suspected late-stage disease.
A recent meta-analysis [
63
] aimed to compare the detection of bone metastases of PCa
between PSMA PET-CT, NaF-PET-CT, choline-PET-CT, MRI and BS. They demonstrated
PSMA and NaF-PET had higher pooled sensitivities on a per-patient basis (0.97 and 0.96
respectively) than choline-PET, MRI and BS (0.87, 0.91, 0.86). Further prospective trails
have showed similar results with greater sensitivity (96% vs. 73%) and better specificity
(99% vs. 84%) in the detection of skeletal metastases [
64
,
65
]. This improvement has
profound implications in the management of small volume metastatic disease invisible on
conventional staging.
Liver metastases typically occur in systemic, late stage disease, however there are cases
of patients with liver metastases as the only metastatic site, thus early detection remains
important for treatment decisions. There is evidence that PCa metastases to the liver are
associated with neuroendocrine characteristic [
66
], and this malignant pattern might lead to
the loss of PSMA-expression [
67
], which would hamper the visualisation of liver metastases
by PSMA PET-CT. In a retrospective study, Damjanovic et al. (2019) reviewed 739 PCa
patients for hepatic metastases using PSMA PET-CT together with CT or MRI. A total of
17 patients
had hepatic metastases, with 15 patients (83.3%) demonstrating PSMA-positive
metastases, two patients (11.1%) PSMA-negative metastases, and one patient (5.6%) had
mixed metastases [
68
]. This study was limited by lack of histopathological confirmation of
results as no liver biopsies were performed, however it shows that while PSMA PET-CT
remains robust at 83.3% detection of PCa liver metastases, it’s limitation lies in the reliance
of cellular expression of PSMA, which can be lost with disease progression and tumour
dedifferentiation.
Pulmonary metastases are considered the second most common extra-nodal metastatic
site for PCa in autopsy studies (lung 46% vs. bone 90%) [
69
], and its reliable detection
as PCa, as opposed to a concurrent primary lung malignancy or benign process, is of
high clinical importance for staging and management. Retrospective studies have found
PSMA PET-CT to detect 72.5% of pulmonary metastases [
70
]. The PSMA PET-CT negative
lesions (27.5%) were postulated to be secondary to loss of PSMA due to neuroendocrine
transdifferentiation (confirmed histologically in a single case where the metastatic deposit
was biopsied [
70
]), however Pryka et al., (2016) demonstrated that due to high PSMA uptake
in lung cancer, PSMA PET-CT was unable to differentiate between a lung primary lesion
and PCa metastasis [
71
]. The utility of PSMA PET-CT in assessment of lung lesions might
be further restricted, as benign lesions such as areas of bronchiectasis [
72
], sarcoidosis [
73
]
and tuberculosis [71].
Despite PSMA PET-CT improving the detection rate in early recurrence, there are
clinical challenges to its use, primarily due to technical shortcomings including a short
half-life (
68
Ga has a physical half-life of only 68 min [
74
]) and limited availability of
68
Ga.
It is known from PSMA PET-CT studies with different ligands that PCa lesions are shown
with better contrast and higher tracer uptake after longer uptake times (eg. 3 h, rather than
Diagnostics 2022,12, 2594 11 of 25
1 h after injection which is the standard protocol) [
75
77
]. Thus, imaging with a ligand with
longer half-life and higher activity (such as
18
F(Fluorine)-PSMA-provides for higher lesion
uptake and superior clearance of background activity. Interestingly,
18
F-PSMA-1007 has
also been found to have less urinary activity than PSMA PET-CT, which would improve its
differentiation of local recurrence and regional lymph node metastases from ureter/bladder
activity, and decrease rate of false positives [76,78].
Diagnostics 2022, 12, x FOR PEER REVIEW 11 of 27
(a)
(b)
Figure 3. Cont.
Diagnostics 2022,12, 2594 12 of 25
Diagnostics 2022, 12, x FOR PEER REVIEW 12 of 27
(c)
Figure 3. Recurrent disease. 74 yr old M; radical prostatectomy 8 yrs prior; extensive para-aortic and
upper pelvic lymph node involvement on CT; severe back pain; PSA 60. PSMA PET-CT:
68
Ga-PSMA-
216 MBq; uptake 48 min; BMI = 31.9; Wt 89 kg. (a,b): Coronal and sagittal images show bulky nodal
disease with markedly increased uptake (SUV = 68.2) in abdomen and pelvis, hepatic metastases,
multiple bony metastases in pelvis, lower limbs, sternum, ribs and vertebral column. (c): enlarged
transaxial images of lower thorax/upper abdomen show tracer avid foci in segments 7 (SUV = 8.8),
4 (SUV = 8.7) of liver; X-hairs on lesion in right 10th rib, SUV = 14.6; CT on bone windows shows
small region of sclerosis vs. extensive uptake on PET.
A recent meta-analysis [63] aimed to compare the detection of bone metastases of
PCa between PSMA PET-CT, NaF-PET-CT, choline-PET-CT, MRI and BS. They demon-
strated PSMA and NaF-PET had higher pooled sensitivities on a per-patient basis (0.97
and 0.96 respectively) than choline-PET, MRI and BS (0.87, 0.91, 0.86). Further prospective
trails have showed similar results with greater sensitivity (96% vs. 73%) and better speci-
ficity (99% vs. 84%) in the detection of skeletal metastases [64,65]. This improvement has
profound implications in the management of small volume metastatic disease invisible on
conventional staging.
Liver metastases typically occur in systemic, late stage disease, however there are
cases of patients with liver metastases as the only metastatic site, thus early detection re-
mains important for treatment decisions. There is evidence that PCa metastases to the liver
are associated with neuroendocrine characteristic [66], and this malignant pattern might
lead to the loss of PSMA-expression [67], which would hamper the visualisation of liver
metastases by PSMA PET-CT. In a retrospective study, Damjanovic et al. (2019) reviewed
739 PCa patients for hepatic metastases using PSMA PET-CT together with CT or MRI. A
total of 17 patients had hepatic metastases, with 15 patients (83.3%) demonstrating PSMA-
positive metastases, two patients (11.1%) PSMA-negative metastases, and one patient
(5.6%) had mixed metastases [68]. This study was limited by lack of histopathological con-
firmation of results as no liver biopsies were performed, however it shows that while
PSMA PET-CT remains robust at 83.3% detection of PCa liver metastases, it’s limitation
lies in the reliance of cellular expression of PSMA, which can be lost with disease progres-
sion and tumour dedifferentiation.
Pulmonary metastases are considered the second most common extra-nodal meta-
static site for PCa in autopsy studies (lung 46% vs. bone 90%) [69], and its reliable detec-
tion as PCa, as opposed to a concurrent primary lung malignancy or benign process, is of
high clinical importance for staging and management. Retrospective studies have found
PSMA PET-CT to detect 72.5% of pulmonary metastases [70]. The PSMA PET-CT negative
lesions (27.5%) were postulated to be secondary to loss of PSMA due to neuroendocrine
transdifferentiation (confirmed histologically in a single case where the metastatic deposit
was biopsied [70]), however Pryka et al., (2016) demonstrated that due to high PSMA up-
take in lung cancer, PSMA PET-CT was unable to differentiate between a lung primary
lesion and PCa metastasis [71]. The utility of PSMA PET-CT in assessment of lung lesions
Figure 3.
Recurrent disease. 74 yr old M; radical prostatectomy 8 yrs prior; extensive para-aortic and
upper pelvic lymph node involvement on CT; severe back pain; PSA 60. PSMA PET-CT:
68
Ga-PSMA-
216 MBq; uptake 48 min; BMI = 31.9; Wt 89 kg. (
a
,
b
): Coronal and sagittal images show bulky nodal
disease with markedly increased uptake (SUV = 68.2) in abdomen and pelvis, hepatic metastases,
multiple bony metastases in pelvis, lower limbs, sternum, ribs and vertebral column. (
c
): enlarged
transaxial images of lower thorax/upper abdomen show tracer avid foci in segments 7 (SUV = 8.8),
4 (SUV = 8.7) of liver; X-hairs on lesion in right 10th rib, SUV = 14.6; CT on bone windows shows
small region of sclerosis vs. extensive uptake on PET.
Furthermore, as mentioned prior, PSMA is not exclusively expressed in PCa. This
uptake in other parts of the body can potentially increase the difficulty of interpretation
of PSMA PET-CT in suspected metastatic disease. A comprehensive prospective trial by
Fendler et al., (2021) found that in patients post radiotherapy or RP who met criteria for
BCR, (PSA > 0.2 mg/mL post RP or PSA > 2 ng/mL above nadir following radiation
therapy) 17 of 217 patients (8%) had a false positive PSMA PET-CT Of these, almost two-
thirds occurred in the context of suspected recurrence in the prostate post radiotherapy.
Other causes for false positives included one case of primary lung cancer, one bronchogenic
cyst, one prostatic abscess and two cases of fibrosis [79].
Another important consideration is the small proportion of reportedly negative PSMA
PET-CT in the context of raised PSA [
80
] (e.g., PSA > 10 ng/mL, where negative PSMA
PET-CT was 4% [
81
]) and metastatic hepatic and pulmonary lesions which are PSMA
PET-CT negative. According to literature almost all prostatic adenocarcinomas will express
PSMA [
82
], however there is a subpopulation that lacks strong PSMA tracer uptake, includ-
ing men with neuroendocrine histology. Those men with advanced, castration resistant
disease, may have areas of de-differentiation and loss of PSMA expression [
25
]. False
negatives are also more common in patients with lower serum PSA values, or slower PSA
kinetics. For purely intraductal carcinoma, which represents around 0.3% of all prostate
cancers [
83
], the sensitivity of PSMA PET-CT has been questioned. Intraductal PCa has been
shown to have a lower PSA expression by 30% and thus may make detecting intraductal
PCa more difficult [
84
]. No specific studies have reviewed the efficacy of PSMA PET-CT in
intraductal PCa, however several articles express concerns over their accuracy and suggest
the addition of mpMRI or FDG PET to more accurately stage and monitor patients [
85
,
86
].
9. Stage Shift & Evolution of Oligometastatic Prostate Cancer
The current treatment paradigm for patients with rising PSA after maximal local thera-
pies with negative conventional imaging is non-curative, consisting of systemic treatments.
Amongst others, this algorithm is based on the results of the CHAARTED and LATITUDE
trials, in which conventional imaging was used to detect metastatic disease [
87
89
]. In
other words, patients with molecular PSMA-identified only oligo-recurrent or de novo
synchronous oligometastatic disease were not included in these studies. Therefore, the
Diagnostics 2022,12, 2594 13 of 25
recommendations of these seminal papers need to be interpreted carefully in patients with
positive PSMA PET-CT but negative conventional imaging.
Similarly, many patients in previous literature considered to have high-risk localised
disease were probably oligometastatic. While there is limited data in this space, we know
that men with de novo oligometastatic disease in the H-arm of the STAMPEDE trial derived
a 10% 3-year OS benefit from local radical radiotherapy in addition to systemic treatment,
compared to those who received systemic treatment alone [
90
]. It is not unreasonable to
extrapolate and expect similar outcomes for cytoreductive prostatectomy in this setting.
10. PSMA PET-CT and MDT (Metastasis Directed Therapy)
Metastatic PCa is becoming more accurately diagnosed and detected earlier through
imaging such as PSMA PET-CT. Metastatic directed therapy is a newer concept aiming
at improving outcomes for patients with oligometastases or metastatic disease. Histori-
cally, metastatic PCa was managed with chemotherapy, androgen biosynthesis inhibition,
androgen receptor inhibition or radium 223 [
91
]. However, several techniques have been
established, specifically targeting metastases. Salvage ePLND has been shown to delay
the development of a new clinical recurrence [
92
,
93
]. Yet, in studies with longer term
follow
>5 years
, the efficacy and reduction in BCR is not as promising as once thought
and therefore, salvage PLND should be perceived as a technique to delay BCR rather
than a cure [
94
]. Essentially, salvage PLND is a form of metastectomy and cure for PCa is
unlikely to be achieved with patients likely requiring salvage ADT and or chemotherapy
and ultimately progressing towards CRPC.
Stereotactic Body Radiotherapy (SBRT) is another metastatic directed therapy which
has been enhanced through the use of PSMA PET-CT. Several authors have shown higher
disease free survival rates (64% vs. 34%), and lower long term requirement of ADT admin-
istration when using
68
GA- PSMA PET vs.
18
F-Choline for directed oligometastatic PCa
treatment [
95
,
96
]. Similar benefits of using PSMA PET-CT to target skeletal oligometastatic
disease have been demonstrated with over 40% of patients showing no evidence of disease
progression [
97
]. Furthermore, elective nodal radiotherapy has also been shown to have a
potential benefit for survival and decrease BCR in a recent systematic review. De Meerleer
et al., (2021) found that patients with high risk PCa and evidence of pathologically positive
pelvic lymph nodes predominately diagnosed through PSMA PET-CT had a substantial
benefit with elective nodal radiotherapy, with minimal grade III or higher toxic effects [
98
].
11. Role of PSMA PET-CT in Biochemical Recurrence after Curative Treatment
The recurrence of PCa is defined by a rise in serum PSA level, termed biochemical
recurrence (BCR), which occurs in 20–30% of patients after radical prostatectomy and up
to 60% of patients after radiotherapy [
99
]. On average BCR precedes the appearance of
clinical metastasis by 8 years [
99
]. The definition of BCR is a serum PSA over 0.2 ng/mL
in two separate tests after prostatectomy, or an absolute rise in PSA level of 2 ng/mL
over the posttreatment PSA nadir following radiotherapy (ASTRO-Phoenix consensus
definition) [
100
]. Apart from a rise in PSA, it is difficult to detect early recurrence as
symptoms are absent with low disease burden. Treatment strategies for PCa recurrence
varies according to the presence of local recurrence, loco-regional lymph node involvement,
or metastases to distant organs or bone [
101
], with treatment options including local salvage
therapy, systemic therapy, surveillance or androgen deprivation. Thus, it is important for
patients with BCR to have early and correct identification of the extent of recurrent disease
to guide management decisions.
Previously, guidelines have recommend traditional imaging modalities such as CT,
BS and MRI in the setting of BCR, however they are limited in their ability to detect
small lesions, with limited sensitivity at lower PSA levels (PSA < 2 ng/mL) [
59
]. CT has
poor anatomical resolution in the treated prostate bed, and unless recurrence is large, it
is of limited use. One meta-analysis found CT sensitivity was 0.42 (0.26–0.56 95% CI)
and specificity was 0.82 (0.8–0.83 95% CI), similarly for MRI, the sensitivity was 0.39
Diagnostics 2022,12, 2594 14 of 25
(0.22–0.56 95% CI)
and specificity was 0.82 (0.79–0.83 95% CI) [
102
]. Indeed, anatomical
imaging techniques depend solely on morphological features for identifying recurrent
disease, which is insufficient when 80% of nodal metastases from PCa are in non-enlarged
lymph nodes [
103
]. BS has proven to be inadequate for early BCR assessment, with 4%
of scans positive with PSA < 10 ng/mL [
104
]. FDG PET-CT has low sensitivity for BCR,
with only 28% detection when PSA is <1.5 ng/mL.
18
F- or 11C-choline PET-CT is only of
useful when PSA is >2.0 ng/mL, and 11C-choline PET demonstrating only 21% detection
rate when PSA < 0.4 ng/mL. Comparison between 11-C-choline,
18
F-fluciclovine and Ga-
PMA PET-CT in patients with BCR, have demonstrated that PSMA PET-CT has a superior
detection rate [105].
PSMA PET-CT has been found to outperform traditional imaging methods in its
detection of recurrent disease in several retrospective studies and meta-analyses, In the
largest retrospective (unblinded) study by Eiber et al. (2015) 248 patients were iden-
tified and reviewed with BCR. They found that the detection rate of recurrence using
PSMA PET-CT was 89.5% with a median PSA 1.99 ng/mL, and 57.9% when the PSA
was
<0.5 ng/mL [106]
. Similarly, a study by Afshar-Oromieh et al. (2015, 319 patients)
looked at BCR for patients post prostatectomy, radiotherapy, ADT or a combination of
these. They found a detection rate for BCR with PSA 1.1–2.0 ng/mL was 71%, PSA of
0.21 to <0.5 was 50%, and
PSA < 0.2 ng/mL
was 47.1% as well as a patient-based sen-
sitivity of 88.1% [
75
].
Fendler et al., (2019)
performed a prospective multicentre trial of
635 men with BCR-PCa, found a direct correlation between detection rate as serum PSA
increased:
38% for <0.5 ng/mL
, 57% for 0.5 to <1.0, 84% for 1.0 to <2.0 ng/mL, 86% for 2.0
to
<5.0 ng/mL
, and 97% for
5.0 ng/mL [
107
]. The overall detection rate was 75%. Their
results found an overall positive predictive value for
68
Ga-PSMA-11 PET of 0.92 (95% CI,
0.88–0.95) [
75
]. A proportion of these patients underwent histopathological correlation and,
with similar results to Afshar-Oromieh et al. (2015), the PSMA PET-CT positive predictive
value for localisation of PCa was >0.8 for a median PSA of 2.1 ng/mL [75].
To date there have been multiple meta-analyses assessing the utility of PSMA PET-
CT [
108
113
] to detect recurrence. In patients with BCR, the proportion of positive PSMA
PET-CT scans consistently increased with higher PSA. For PSA categories 0–0.19,
0.2–0.49
,
0.5–0.99, 1–1.99, and >2 ng/mL, the percentages of positive scans were 33%, 45%, 59%,
75%, and 95%, respectively. Significantly, PSMA PET-CT improves the detection of recur-
rence at low PSA (<0.5 ng/mL), and has high sensitivity (75%) and specificity (99%) on
meta-analysis of pooled data (Figure 4) [
108
]. Furthermore, a meta-analysis by Kimura
et al., (2020) looking at accuracy for PSMA PET-CT performed prior to salvage lymph
node dissection for nodal recurrence in PCa patients with BCR (462 patients, 14 studies)
found sensitivity using lesion based analysis of 0.84 (95%CI: 0.61–0.95) and specificity 0.97
(95% CI: 0.95–0.99) [113].
Shortcomings notwithstanding, uptake of PSMA PET-CT by clinicians has been enthu-
siastic, with detectable impact on patient management. A prospective study demonstrated
the change in therapeutic planning in 62% of cases with BCR after definitive management,
with PSMA PET-CT detecting unsuspected: local relapse in 27% patients, lymph node
spread 39%, distant metastases 16% [
114
]. A retrospective single centre study, found PSMA
PET-CT changed staging in 32% of patients, with 23% upstaged, and 9% downstaged after
PSMA PET-CT (Figure 5) [
115
], compared to conventional imaging, leading to changes in
treatment strategies. Meta-analysis results are more conservative, with 54% of patients re-
porting change in intended management after PSMA PET-CT [
116
], with changes including
proportionally higher rates of radiotherapy (56% to 61%), surgery (1% to 7%), focal therapy
(1% to 2%) and multimodal treatment (2% to 6%), and lower rates of systemic therapy (26%
to 12%) and no treatment (14% to 11%) after PSMA PET-CT in patients with BCR.
PSMA PET-CT for BCR has identified differences in sites of recurrence when compar-
ing BCR post radiotherapy and prostatectomy. Lawal et al. (2021) identified 247 patients
with BCR post external beam radiotherapy (EBRT) or RP. When comparing EBRT and
RP, the site of recurrence on PSMA PET-CT was 67.4% vs. 43.4% at the prostate bed,
Diagnostics 2022,12, 2594 15 of 25
17.4% vs. 16.5%
at SV, 37.2% vs. 34.7% in pelvic nodes and 9.3% vs. 18.3% to bone, respec-
tively, [
117
]. Similar results were demonstrated in a review by Armstrong et al. (2020),
which also found that patients who underwent RP had a lower local recurrence rate than
those who had radiotherapy (21.3% vs. 63.3%) but shorter time from initial therapy to
recurrence (27.7 months vs. 54.6 months). They also found that recurrence rates were lower
in RP patients compared to those that received radiotherapy for extrapelvic metastasis
(20.0% vs. 46.8%) [118].
Diagnostics 2022, 12, x FOR PEER REVIEW 16 of 27
(b)
Figure 4. Biochemical recurrence—serial scans. 58 yr old Mradical prostatectomy and node dis-
Section 10 yrs prior; scans done in July 2020 when PSA 0.24 and Sep 2021 when PSA 0.38. (a): PSMA
PET-CT: July 2020-
68
Ga-PSMA-226 MBq; uptake 49 min; BMI = 35.1; Wt 105 kg; coronal images and
transaxial images at level of prostate bed show tracer excretion; rest of study clear. (b): PSMA PET-
CT: Sep 2021-
68
Ga-PSMA-238 MBq; uptake 50 min; BMI = 35.2; Wt 107 kg; coronal images and trans-
axial images at level of prostate bed show tracer excretion but with a new small focus of uptake
SUV= 6.0 in left side of prostate bed; rest of study clear.
Shortcomings notwithstanding, uptake of PSMA PET-CT by clinicians has been en-
thusiastic, with detectable impact on patient management. A prospective study demon-
strated the change in therapeutic planning in 62% of cases with BCR after definitive man-
agement, with PSMA PET-CT detecting unsuspected: local relapse in 27% patients, lymph
node spread 39%, distant metastases 16% [114]. A retrospective single centre study, found
PSMA PET-CT changed staging in 32% of patients, with 23% upstaged, and 9% down-
staged after PSMA PET-CT (Figure 5) [115], compared to conventional imaging, leading
to changes in treatment strategies. Meta-analysis results are more conservative, with 54%
of patients reporting change in intended management after PSMA PET-CT [116], with
changes including proportionally higher rates of radiotherapy (56% to 61%), surgery (1%
to 7%), focal therapy (1% to 2%) and multimodal treatment (2% to 6%), and lower rates of
systemic therapy (26% to 12%) and no treatment (14% to 11%) after PSMA PET-CT in pa-
tients with BCR.
Figure 4.
Biochemical recurrence—serial scans. 58 yr old M—radical prostatectomy and node
disSection 10 yrs prior; scans done in July 2020 when PSA 0.24 and Sep 2021 when PSA 0.38. (
a
):
PSMA PET-CT: July 2020-
68
Ga-PSMA-226 MBq; uptake 49 min; BMI = 35.1; Wt 105 kg; coronal images
and transaxial images at level of prostate bed show tracer excretion; rest of study clear. (
b
): PSMA
PET-CT: Sep 2021-
68
Ga-PSMA-238 MBq; uptake 50 min; BMI = 35.2; Wt 107 kg; coronal images and
transaxial images at level of prostate bed show tracer excretion but with a new small focus of uptake
SUV= 6.0 in left side of prostate bed; rest of study clear.
Diagnostics 2022,12, 2594 16 of 25
In patients with BCR, PSMA PET-CT is currently being used as the gold standard
imaging modality to identify residual cancer and guide planning of salvage radiotherapy
(SRT) planning [
119
]. Interestingly, retrospective studies using PSMA PET-CT of patients
with biochemical persistence (BCP) post-radical prostatectomy have found the obturator,
mesorectal and pre-sacral nodes at highest risk of containing residual disease [
120
]. In
this cohort of patients, SRT represents one of mainstays of treatment although the role of
concurrent ADT is still being investigated. A retrospective study by Rogowski et al. (2022),
demonstrated that PSMA PET-CT based SRT for lymph node recurrence yielded BCR-free
survival of 81%, 72% and 66% at 1, 2 and 3 years, respectively, [121].
Diagnostics 2022, 12, x FOR PEER REVIEW 17 of 27
(a)
(b)
Figure 5. Cont.
Diagnostics 2022,12, 2594 17 of 25
Diagnostics 2022, 12, x FOR PEER REVIEW 18 of 27
(c)
(d)
Figure 5. Biochemical recurrence—negative scan; normal distribution of tracer. 75 yr old M—radical
prostatectomy 7 yrs prior; PSA 0.42. PSMA PET-CT:
68
Ga-PSMA - 219 MBq; uptake 48 min; BMI =
22.6; Wt 63 kg. (a,b): coronal images show physiological uptake in parotid glands, bowel and kid-
neys and tracer pooling in bladder; X-hairs show uptake in cervical (SUV = 2.3) and sacral (SUV =
2.5) nerve roots and sympathetic ganglia in upper abdomen SUV = 4.6. (c,d): enlarged coronal, sag-
ittal and transaxial images of prostate bed; X-hairs identify locations; no abnormal tracer uptake but
pooling of trace in bladder problematic; Note—scale in sagittal images on (c) has been deliberately
altered to ensure there are no adjacent focal regions of uptake to suggest local tumor recurrence.
PSMA PET-CT for BCR has identified differences in sites of recurrence when com-
paring BCR post radiotherapy and prostatectomy. Lawal et al. (2021) identified 247 pa-
tients with BCR post external beam radiotherapy (EBRT) or RP. When comparing EBRT
and RP, the site of recurrence on PSMA PET-CT was 67.4% vs. 43.4% at the prostate bed,
17.4% vs. 16.5% at SV, 37.2% vs. 34.7% in pelvic nodes and 9.3% vs. 18.3% to bone, respec-
tively, [117]. Similar results were demonstrated in a review by Armstrong et al. (2020),
which also found that patients who underwent RP had a lower local recurrence rate than
those who had radiotherapy (21.3% vs. 63.3%) but shorter time from initial therapy to
recurrence (27.7 months vs. 54.6 months). They also found that recurrence rates were
lower in RP patients compared to those that received radiotherapy for extrapelvic metas-
tasis (20.0% vs. 46.8%) [118].
In patients with BCR, PSMA PET-CT is currently being used as the gold standard
imaging modality to identify residual cancer and guide planning of salvage radiotherapy
(SRT) planning [119]. Interestingly, retrospective studies using PSMA PET-CT of patients
with biochemical persistence (BCP) post-radical prostatectomy have found the obturator,
Figure 5.
Biochemical recurrence—negative scan; normal distribution of tracer. 75 yr old M—radical
prostatectomy 7 yrs prior; PSA 0.42. PSMA PET-CT:
68
Ga-PSMA - 219 MBq; uptake 48 min;
BMI = 22.6
;
Wt 63 kg. (
a
,
b
): coronal images show physiological uptake in parotid glands, bowel and kidneys and
tracer pooling in bladder; X-hairs show uptake in cervical (SUV = 2.3) and sacral (SUV = 2.5) nerve
roots and sympathetic ganglia in upper abdomen SUV = 4.6. (
c
,
d
): enlarged coronal, sagittal and
transaxial images of prostate bed; X-hairs identify locations; no abnormal tracer uptake but pooling
of trace in bladder problematic; Note—scale in sagittal images on (
c
) has been deliberately altered to
ensure there are no adjacent focal regions of uptake to suggest local tumor recurrence.
12. Limitations of PSMA PET-CT
Despite it being a non-invasive, readily performed, swift process there are some
limitations and side effects of PET and whole-body PET-CT. PET does require exposing
patients to radiation, however, this dose is relatively low at approximately 2.0 mSv for
PSMA PET-CT alone and an additional 22 mSv if whole body PET-CT was performed
totaling approximately 24 mSv [
107
]. Serial radiation exposure through CT has been
shown to have and estimated lifetime attributable cancer mortality risk of approximately
2% with annual exposure for 30 consecutive years [
122
]. Most patients would receive
one or two PET-CT’s in their lifetime thus making the cancer mortality risk negligible.
Interpretation of PET imaging may be difficult in patients who are unable to lie still for
their scan, potentially requiring reimaging or sedation to improve image quality as well
as potential for interdepartmental and international variation in protocol timing of tracer
administration and SUV measurement [
123
]. Furthermore, reliance on the production
Diagnostics 2022,12, 2594 18 of 25
of radiotracer allows PSMA PET-CT to be susceptible to broader changes in the global
community with supply chains dependent on a cohesive global community.
From a clinical perspective, PSMA PET-CT has been well documented to detect PCa
with positive scans seen in the majority of patients with suspected cancer (approximately
83%) and shown to be highly specific. Despite superior accuracy compared to cross-
sectional imaging, false negatives have also been described including small nodal metas-
tases under the special resolution of PET (i.e., <5 mm), ISUP < 3 tumours and a minority of
prostate cancers [
124
]. Mannweiler et al. found that 5% of primary prostate cancer and 15%
of prostate cancer mets were PSMA-negative in immunohistochemistry [125].
Furthermore, stage migration based on the accuracy of PSMA PET-CT has also be-
come an area of interest with patients being upstaged now representing more favourable
disease compared to others in the new stage grouping—apparent rates of survival im-
prove, yet there is no impact on individual patient outcome (also known as the Will
Rogers phenomenon).
13. PSMA PET-CT in the Future
Moving forward, it is likely that PSMA PET-CT will provide even more accurate diag-
nosis and staging for PCa. This could occur through the development of new radioligands.
18
F-PSMA-1007 is one of the newly developed radioligands that may improve the accuracy
of diagnosis and staging. It has different properties to PSMA PET-CT including a longer
half-life and reduced urinary clearance [
126
]. Initial studies show benefit at identifying
small LNM (
3 mm) with a sensitivity of 82% and specificity of 99.6% [
127
]. However, the
rate of detection of benign lesions in thyroid, non-specific lymph nodes and bones requires
careful interpretation of the PET images to avoid false positive results [128].
Furthermore, radiotheranostics and the use of radioligands with specific uptake target-
ing PSMA have been proposed to treat PCa. These new radioligands have been recognised
to deliver a sustained treatment to the local microenvironment without causing harm to
surrounding tissue [
129
,
130
]. As mentioned earlier, PSMA is not exclusively expressed
on prostatic tissue and can be identified in other malignant and non-malignant processes
throughout the body. Lutetium-177-PSMA-617 (
177
LuPSMA) is a radioligand which deliv-
ers beta particle radiation selectively to PSMA positive cells and their direct microenviron-
ment. The VISION trial followed 831 patients with castrate resistant prostate cancer and
randomly assigned them to the
177
LuPSMA arm or the control group. They showed an
increase in overall survival from 11.3 to 15.3 months (p< 0.001), decrease in PSA level by
50–80% and delayed imaging-based progression through CT, MRI or BS. This treatment
did have significant side effects with 53% of patients suffering grade 3 or higher adverse
events compared to 38% of the control arm (134). Further PSMA ligands using copper
(
64
Cu) and scandium (
44
SC) have also been proposed with different radioligand properties
such as significant tumour uptake and retention after 24 h, appearing to be promising
alternatives [
131
]. Yet, all of these are still in their infancy but provide a glimpse of the
future for PSMA PET-CT and PCa.
Another area PSMA may prove to be useful is through the tracking of radioactively
tagged LNM in intraoperative PLND. After performing PSMA PET-CT and defining pa-
tients with high risk prostate cancer suspicious for positive pelvic lymph nodes, a radio-
tracer is injected prior to surgery that emit specific fluorescence or gamma-emitting tracers
such as
99m
Tc or
111
InPSMA, which are then able to be detected through direct vision or
gamma-emitting probe detectors [
132
,
133
]. Although in its infancy, real time diagnosis
and treatment of positive lymph nodes accessible during operative management has the
potential to decrease BCR and increase overall survival. In addition to this, xenograft
models have been used to assess the utility of alpha particle emitting analogues of
213
Bi-L1
and
225
Ac-L1. Several studies suggest comprehensive cell uptake on a micrometastatic
level and therapeutic effect on PSMA positive cells with minimal toxicity. [134,135].
In summary, PSMA PET-CT is now routinely used in the evaluation of prostate cancer
in the context of primary staging and suspected tumour recurrence. More specific PET
Diagnostics 2022,12, 2594 19 of 25
radiopharmaceuticals my improve both assessments and provide additional benefits in
theranostics. PET-MR with the ability to perform simultaneous PET and MR acquisitions
may be the imaging modality of choice in the future.
Author Contributions:
Conceptualization, S.L., A.D.C. and C.A.P.; resources, S.L., L.W., M.F.;
writing—original draft preparation, A.D.C., C.A.P.; writing—review and editing, S.L., A.D.C., C.A.P.,
R.C.; supervision, S.L., H.W.; project administration S.L., A.D.C., C.A.P.; funding acquisition. All
authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement:
Informed consent was obtained from all subjects involved in
the study
.
Data Availability Statement: Not applicable.
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2018. CA: A Cancer J. Clin. 2018,68, 7–30. [CrossRef] [PubMed]
2.
Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN
Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: Cancer J. Clin.
2021
,71, 209–249. [CrossRef]
[PubMed]
3.
Moyer, V.A. Screening for prostate cancer: U.S. Preventive Services Task Force recommendation statement. Ann. Intern. Med.
2012,157, 120–134. [CrossRef] [PubMed]
4.
Royal Australian College of General Practitioners; National Preventive and Community Medicine Committee. Guidelines for
Preventive Activities in General Practice, 9th ed.; RACGP: East Melbourne, Australia, 2016.
5.
Pickles, K.; Carter, S.M.; Rychetnik, L.; Entwistle, V.A. Doctors’ perspectives on PSA testing illuminate established differences in
prostate cancer screening rates between Australia and the UK: A qualitative study. BMJ Open 2016,6, e011932. [CrossRef]
6.
Andriole, G.L.; Crawford, E.D.; Grubb, R.L.; Buys, S.S.; Chia, D.; Church, T.R.; Fouad, M.N.; Gelmann, E.P.; Kvale, P.A.;
Reding, D.J.; et al.
Mortality Results from a Randomized Prostate-Cancer Screening Trial. N. Engl. J. Med.
2009
,360, 1310–1319.
[CrossRef]
7.
Schröder, F.H.; Hugosson, J.; Roobol, M.J.; Tammela, T.L.J.; Ciatto, S.; Nelen, V.; Kwiatkowski, M.; Lujan, M.; Lilja, H.;
Zappa, M.; et al
. Screening and Prostate-Cancer Mortality in a Randomized European Study. N. Engl. J. Med.
2009
,
360, 1320–1328
.
[CrossRef]
8.
Hugosson, J.; Roobol, M.J.; Månsson, M.; Tammela, T.L.J.; Zappa, M.; Nelen, V.; Kwiatkowski, M.; Lujan, M.; Carlsson, S.V.;
Talala, K.M.; et al.
A 16-yr Follow-up of the European Randomized study of Screening for Prostate Cancer. Eur. Urol.
2019
,
76, 43–51. [CrossRef]
9.
James, S.L.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.
Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries
and territories, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet
2018
,392, 1789–1858.
[CrossRef]
10.
Thompson, I.M.; Ankerst, D.P.; Chi, C.; Goodman, P.J.; Tangen, C.M.; Lucia, M.S.; Feng, Z.; Parnes, H.L.; Coltman, C.A., Jr.
Assessing prostate cancer risk: Results from the Prostate Cancer Prevention Trial. J. Natl. Cancer Inst.
2006
,98, 529–534. [CrossRef]
11. Shariat, S.F.; Roehrborn, C.G. Using biopsy to detect prostate cancer. Rev. Urol. 2008,10, 262–280.
12.
Schaeffer, E.; Srinivas, S.; Antonarakis, E.S.; Armstrong, A.J.; Bekelman, J.E.; Cheng, H.; D’Amico, A.V.; Davis, B.J.; Desai, N.;
Dorff, T.; et al. NCCN Guidelines Insights: Prostate Cancer, Version 1.2021. J. Natl. Compr Cancer Netw.
2021
,19, 134–143.
[CrossRef] [PubMed]
13.
Pierorazio, P.M.; Walsh, P.C.; Partin, A.W.; Epstein, J.I. Prognostic Gleason grade grouping: Data based on the modified Gleason
scoring system. BJU Int. 2013,111, 753–760. [CrossRef] [PubMed]
14.
Leyten, G.H.; Hessels, D.; Smit, F.P.; Jannink, S.A.; de Jong, H.; Melchers, W.J.; Cornel, E.B.; de Reijke, T.M.; Vergunst, H.;
Kil, P.; et al.
Identification of a Candidate Gene Panel for the Early Diagnosis of Prostate Cancer. Clin. Cancer Res.
2015
,
21, 3061–3070. [CrossRef] [PubMed]
15.
Van Neste, L.; Hendriks, R.J.; Dijkstra, S.; Trooskens, G.; Cornel, E.B.; Jannink, S.A.; de Jong, H.; Hessels, D.; Smit, F.P.;
Melchers, W.J.; et al.
Detection of High-grade Prostate Cancer Using a Urinary Molecular Biomarker-Based Risk Score. Eur. Urol.
2016,70, 740–748. [CrossRef]
16.
Ahmed, H.U.; El-Shater Bosaily, A.; Brown, L.C.; Gabe, R.; Kaplan, R.; Parmar, M.K.; Collaco-Moraes, Y.; Ward, K.;
Hindley, R.G.
;
Freeman, A.; et al. Diagnostic accuracy of multi-parametric MRI and TRUS biopsy in prostate cancer (PROMIS): A paired
validating confirmatory study. Lancet 2017,389, 815–822. [CrossRef]
Diagnostics 2022,12, 2594 20 of 25
17.
Purysko, A.S.; Baroni, R.H.; Giganti, F.; Costa, D.; Renard-Penna, R.; Kim, C.K.; Raman, S.S. PI-RADS Version 2.1: A Critical
Review, From the AJR Special Series on Radiology Reporting and Data Systems. Am. J. Roentgenol. 2020,216, 20–32. [CrossRef]
18.
Katzendorn, O.; Klot, C.; Mahjoub, S.; Faraj Tabrizi, P.; Harke, N.; Tezval, H.; Hellms, S.; Hennenlotter, J.; Baig, M.;
Stenzl, A.; et al.
Combination of PI-RADS score and mRNA urine test—A novel scoring system for improved detection of prostate cancer.
PLoS ONE 2022,17, e0271981. [CrossRef]
19. Berger, A. How does it work? Positron emission tomography. BMJ 2003,326, 1449. [CrossRef]
20.
Rodnick, M.E.; Sollert, C.; Stark, D.; Clark, M.; Katsifis, A.; Hockley, B.G.; Parr, D.C.; Frigell, J.; Henderson, B.D.;
Abghari-Gerst, M.; et al.
Cyclotron-based production of 68Ga, [68Ga]GaCl3, and [68Ga]Ga-PSMA-11 from a liquid target.
EJNMMI Radiopharm. Chem. 2020,5, 25. [CrossRef]
21.
Silver, D.A.; Pellicer, I.; Fair, W.R.; Heston, W.D.; Cordon-Cardo, C. Prostate-specific membrane antigen expression in normal and
malignant human tissues. Clin. Cancer Res. 1997,3, 81–85.
22.
Nargund, V.; Al Hashmi, D.; Kumar, P.; Gordon, S.; Otitie, U.; Ellison, D.; Carroll, M.; Baithun, S.; Britton, K.E. Imaging with
radiolabelled monoclonal antibody (MUJ591) to prostate-specific membrane antigen in staging of clinically localized prostatic
carcinoma: Comparison with clinical, surgical and histological staging. BJU Int. 2005,95, 1232–1236. [CrossRef] [PubMed]
23.
Werner, R.A.; Derlin, T.; Lapa, C.; Sheikbahaei, S.; Higuchi, T.; Giesel, F.L.; Behr, S.; Drzezga, A.; Kimura, H.; Buck, A.K.; et al.
18
F-Labeled, PSMA-Targeted Radiotracers: Leveraging the Advantages of Radiofluorination for Prostate Cancer Molecular
Imaging. Theranostics 2020,10, 1–16. [CrossRef] [PubMed]
24.
Zhao, R.; Li, Y.; Nie, L.; Qin, K.; Zhang, H.; Shi, H. The meta-analysis of the effect of 68Ga-PSMA-PET/CT diagnosis of prostatic
cancer compared with bone scan. Medicine 2021,100, e25417. [CrossRef]
25.
Hofman, M.S.; Hicks, R.J.; Maurer, T.; Eiber, M. Prostate-specific Membrane Antigen PET: Clinical Utility in Prostate Cancer,
Normal Patterns, Pearls, and Pitfalls. Radiographics 2018,38, 200–217. [CrossRef] [PubMed]
26.
O’Keefe, D.S.; Bacich, D.J.; Huang, S.S.; Heston, W.D.W. A Perspective on the Evolving Story of PSMA Biology, PSMA-Based
Imaging, and Endoradiotherapeutic Strategies. J. Nucl. Med. 2018,59, 1007–1013. [CrossRef] [PubMed]
27.
Drost, F.H.; Osses, D.F.; Nieboer, D.; Steyerberg, E.W.; Bangma, C.H.; Roobol, M.J.; Schoots, I.G. Prostate MRI, with or without
MRI-targeted biopsy, and systematic biopsy for detecting prostate cancer. Cochrane Database Syst. Rev.
2019
,4, Cd012663.
[CrossRef]
28.
Barkovich, E.J.; Shankar, P.R.; Westphalen, A.C. A Systematic Review of the Existing Prostate Imaging Reporting and Data System
Version 2 (PI-RADSv2) Literature and Subset Meta-Analysis of PI-RADSv2 Categories Stratified by Gleason Scores. AJR Am. J.
Roentgenol. 2019,212, 847–854. [CrossRef]
29.
Kasivisvanathan, V.; Rannikko, A.S.; Borghi, M.; Panebianco, V.; Mynderse, L.A.; Vaarala, M.H.; Briganti, A.; Budäus, L.;
Hellawell, G.
; Hindley, R.G.; et al. MRI-Targeted or Standard Biopsy for Prostate-Cancer Diagnosis. N. Engl. J. Med.
2018
,
378, 1767–1777. [CrossRef]
30.
Rouvière, O.; Puech, P.; Renard-Penna, R.; Claudon, M.; Roy, C.; Mège-Lechevallier, F.; Decaussin-Petrucci, M.; Dubreuil-
Chambardel, M.; Magaud, L.; Remontet, L.; et al. Use of prostate systematic and targeted biopsy on the basis of multiparametric
MRI in biopsy-naive patients (MRI-FIRST): A prospective, multicentre, paired diagnostic study. Lancet Oncol.
2019
,20, 100–109.
[CrossRef]
31.
Borofsky, S.; George, A.K.; Gaur, S.; Bernardo, M.; Greer, M.D.; Mertan, F.V.; Taffel, M.; Moreno, V.; Merino, M.J.; Wood, B.J.; et al.
What Are We Missing? False-Negative Cancers at Multiparametric MR Imaging of the Prostate. Radiology
2018
,286, 186–195.
[CrossRef]
32.
Kwon, T.; Jeong, I.G.; You, D.; Hong, J.H.; Ahn, H.; Kim, C.S. Prevalence and clinical significance of incidental
18
F-fluoro-2-
deoxyglucose uptake in prostate. Korean J. Urol. 2015,56, 288–294. [CrossRef] [PubMed]
33.
Satapathy, S.; Singh, H.; Kumar, R.; Mittal, B.R. Diagnostic Accuracy of
68
Ga-PSMA PET/CT for Initial Detection in Patients
With Suspected Prostate Cancer: A Systematic Review and Meta-Analysis. AJR Am. J. Roentgenol.
2021
,216, 599–607. [CrossRef]
[PubMed]
34.
Donato, P.; Morton, A.; Yaxley, J.; Ranasinghe, S.; Teloken, P.E.; Kyle, S.; Coughlin, G.; Esler, R.; Dunglison, N.; Gardiner, R.A.; et al.
68
Ga-PSMA PET/CT better characterises localised prostate cancer after MRI and transperineal prostate biopsy: Is
68
Ga-PSMA
PET/CT guided biopsy the future? Eur. J. Nucl. Med. Mol. Imaging 2020,47, 1843–1851. [CrossRef]
35.
Donato, P.; Roberts, M.J.; Morton, A.; Kyle, S.; Coughlin, G.; Esler, R.; Dunglison, N.; Gardiner, R.A.; Yaxley, J. Improved specificity
with
68
Ga PSMA PET/CT to detect clinically significant lesions “invisible” on multiparametric MRI of the prostate: A single
institution comparative analysis with radical prostatectomy histology. Eur. J. Nucl. Med. Mol. Imaging
2019
,46, 20–30. [CrossRef]
36.
Scheltema, M.J.; Chang, J.I.; Stricker, P.D.; van Leeuwen, P.J.; Nguyen, Q.A.; Ho, B.; Delprado, W.; Lee, J.; Thompson, J.E.;
Cusick, T.; et al.
Diagnostic accuracy of
68
Ga-prostate-specific membrane antigen (PSMA) positron-emission tomography (PET)
and multiparametric (mp)MRI to detect intermediate-grade intra-prostatic prostate cancer using whole-mount pathology: Impact
of the addition of 68Ga-PSMA PET to mpMRI. BJU Int. 2019,124 (Suppl. 1), 42–49. [CrossRef]
37.
Emmett, L.; Buteau, J.; Papa, N.; Moon, D.; Thompson, J.; Roberts, M.J.; Rasiah, K.; Pattison, D.A.; Yaxley, J.; Thomas, P.; et al. The
Additive Diagnostic Value of Prostate-specific Membrane Antigen Positron Emission Tomography Computed Tomography to
Multiparametric Magnetic Resonance Imaging Triage in the Diagnosis of Prostate Cancer (PRIMARY): A Prospective Multicentre
Study. Eur. Urol. 2021,80, 682–689. [CrossRef] [PubMed]
Diagnostics 2022,12, 2594 21 of 25
38.
Barentsz, J.O.; Richenberg, J.; Clements, R.; Choyke, P.; Verma, S.; Villeirs, G.; Rouviere, O.; Logager, V.; Fütterer, J.J. ESUR prostate
MR guidelines 2012. Eur. Radiol. 2012,22, 746–757. [CrossRef]
39.
Soeterik, T.F.W.; van Melick, H.H.E.; Dijksman, L.M.; Biesma, D.H.; Witjes, J.A.; van Basten, J.A. Multiparametric Magnetic
Resonance Imaging Should Be Preferred Over Digital Rectal Examination for Prostate Cancer Local Staging and Disease Risk
Classification. Urology 2021,147, 205–212. [CrossRef]
40.
Chen, M.; Zhang, Q.; Zhang, C.; Zhou, Y.H.; Zhao, X.; Fu, Y.; Gao, J.; Zhang, B.; Wang, F.; Qiu, X.; et al. Comparison of
68
Ga-prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) and multi-
parametric magnetic resonance imaging (MRI) in the evaluation of tumor extension of primary prostate cancer. Transl. Androl.
Urol. 2020,9, 382–390. [CrossRef]
41.
Skawran, S.M.; Sanchez, V.; Ghafoor, S.; Hötker, A.M.; Burger, I.A.; Huellner, M.W.; Eberli, D.; Donati, O.F. Primary staging in
patients with intermediate- and high-risk prostate cancer: Multiparametric MRI and
68
Ga-PSMA-PET/MRI—What is the value of
quantitative data from multiparametric MRI alone or in conjunction with clinical information? Eur. J. Radiol.
2022
,146, 110044.
[CrossRef]
42.
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. [CrossRef]
43.
Wang, X.; Wen, Q.; Zhang, H.; Ji, B. Head-to-Head Comparison of 68Ga-PSMA-11 PET/CT and Multiparametric MRI for Pelvic
Lymph Node Staging Prior to Radical Prostatectomy in Patients With Intermediate to High-Risk Prostate Cancer: A Meta-Analysis.
Front. Oncol. 2021,11, 737989. [CrossRef] [PubMed]
44.
Budäus, L.; Leyh-Bannurah, S.R.; Salomon, G.; Michl, U.; Heinzer, H.; Huland, H.; Graefen, M.; Steuber, T.; Rosenbaum, C. Initial
Experience of
68
Ga-PSMA PET/CT Imaging in High-risk Prostate Cancer Patients Prior to Radical Prostatectomy. Eur. Urol.
2016
,
69, 393–396. [CrossRef] [PubMed]
45.
Herlemann, A.; Wenter, V.; Kretschmer, A.; Thierfelder, K.M.; Bartenstein, P.; Faber, C.; Gildehaus, F.J.; Stief, C.G.; Gratzke, C.;
Fendler, W.P.
68
Ga-PSMA Positron Emission Tomography/Computed Tomography Provides Accurate Staging of Lymph Node
Regions Prior to Lymph Node Dissection in Patients with Prostate Cancer. Eur. Urol. 2016,70, 553–557. [CrossRef]
46.
Maurer, T.; Gschwend, J.E.; Rauscher, I.; Souvatzoglou, M.; Haller, B.; Weirich, G.; Wester, H.J.; Heck, M.; Kübler, H.;
Beer, A.J.; et al
.
Diagnostic Efficacy of
68
Gallium-PSMA Positron Emission Tomography Compared to Conventional Imaging for Lymph Node
Staging of 130 Consecutive Patients with Intermediate to High Risk Prostate Cancer. J. Urol. 2016,195, 1436–1443. [CrossRef]
47.
Rahbar, K.; Weckesser, M.; Huss, S.; Semjonow, A.; Breyholz, H.J.; Schrader, A.J.; Schäfers, M.; Bögemann, M. Correlation of
Intraprostatic Tumor Extent with
68
Ga-PSMA Distribution in Patients with Prostate Cancer. J. Nucl. Med.
2016
,57, 563–567.
[CrossRef]
48.
van Leeuwen, P.J.; Emmett, L.; Ho, B.; Delprado, W.; Ting, F.; Nguyen, Q.; Stricker, P.D. Prospective evaluation of 68Gallium-
prostate-specific membrane antigen positron emission tomography/computed tomography for preoperative lymph node staging
in prostate cancer. BJU Int. 2017,119, 209–215. [CrossRef]
49.
van Kalmthout, L.W.M.; van Melick, H.H.E.; Lavalaye, J.; Meijer, R.P.; Kooistra, A.; de Klerk, J.M.H.; Braat, A.; 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. [CrossRef]
50.
Karagiannis, V.; Wichmann, V.; Saarinen, J.; Eigeliene, N.; Andersen, H.; Jekunen, A. Radiotherapy treatment modification for
prostate cancer patients based on PSMA-PET/CT. Radiat. Oncol. 2022,17, 19. [CrossRef]
51.
Marra, G.; Valerio, M.; Heidegger, I.; Tsaur, I.; Mathieu, R.; Ceci, F.; Ploussard, G.; van den Bergh, R.C.N.; Kretschmer, A.;
Thibault, C.; et al. Management of Patients with Node-positive Prostate Cancer at Radical Prostatectomy and Pelvic Lymph Node
Dissection: A Systematic Review. Eur. Urol. Oncol. 2020,3, 565–581. [CrossRef]
52.
Fossati, N.; Willemse, P.M.; Van den Broeck, T.; van den Bergh, R.C.N.; Yuan, C.Y.; Briers, E.; Bellmunt, J.; Bolla, M.; Cornford, P.;
De Santis, M.; et al. The Benefits and Harms of Different Extents of Lymph Node Dissection During Radical Prostatectomy for
Prostate Cancer: A Systematic Review. Eur. Urol. 2017,72, 84–109. [CrossRef] [PubMed]
53.
Lestingi, J.F.P.; Guglielmetti, G.B.; Trinh, Q.D.; Coelho, R.F.; Pontes, J., Jr.; Bastos, D.A.; Cordeiro, M.D.; Sarkis, A.S.; Faraj, S.F.;
Mitre, A.I.; et al. Extended Versus Limited Pelvic Lymph Node Dissection During Radical Prostatectomy for Intermediate- and
High-risk Prostate Cancer: Early Oncological Outcomes from a Randomized Phase 3 Trial. Eur. Urol.
2021
,79, 595–604. [CrossRef]
[PubMed]
54.
Yaxley, J.W.; Raveenthiran, S.; Nouhaud, F.X.; Samaratunga, H.; Yaxley, W.J.; Coughlin, G.; Yaxley, A.J.; Gianduzzo, T.;
Kua, B.;
McEwan, L.; et al. Risk of metastatic disease on
68
gallium-prostate-specific membrane antigen positron emission tomogra-
phy/computed tomography scan for primary staging of 1253 men at the diagnosis of prostate cancer. BJU Int.
2019
,124, 401–407.
[CrossRef] [PubMed]
55.
Stabile, A.; Pellegrino, A.; Mazzone, E.; Cannoletta, D.; de Angelis, M.; Barletta, F.; Scuderi, S.; Cucchiara, V.; Gandaglia, G.;
Raggi, D.; et a
l. Can Negative Prostate-specific Membrane Antigen Positron Emission Tomography/Computed Tomography
Avoid the Need for Pelvic Lymph Node Dissection in Newly Diagnosed Prostate Cancer Patients? A Systematic Review and
Meta-analysis with Backup Histology as Reference Standard. Eur. Urol. Oncol. 2022,5, 1–17. [CrossRef] [PubMed]
56.
Gandaglia, G.; Mazzone, E.; Stabile, A.; Pellegrino, A.; Cucchiara, V.; Barletta, F.; Scuderi, S.; Robesti, D.; Leni, R.;
Samanes Gajate, A.M.; et al.
Prostate-specific membrane antigen Radioguided Surgery to Detect Nodal Metastases in Primary
Diagnostics 2022,12, 2594 22 of 25
Prostate Cancer Patients Undergoing Robot-assisted Radical Prostatectomy and Extended Pelvic Lymph Node Dissection: Results
of a Planned Interim Analysis of a Prospective Phase 2 Study. Eur. Urol. 2022,82, 411–418. [CrossRef]
57.
Gandaglia, G.; Fossati, N.; Zaffuto, E.; Bandini, M.; Dell’Oglio, P.; Bravi, C.A.; Fallara, G.; Pellegrino, F.; Nocera, L.;
Karakiewicz, P.I.; et al
. Development and Internal Validation of a Novel Model to Identify the Candidates for Extended Pelvic
Lymph Node Dissection in Prostate Cancer. Eur. Urol. 2017,72, 632–640. [CrossRef]
58.
Meijer, D.; van Leeuwen, P.J.; Roberts, M.J.; Siriwardana, A.R.; Morton, A.; Yaxley, J.W.; Samaratunga, H.; Emmett, L.;
van de Ven, P.M.
; van der Poel, H.G.; et al. External Validation and Addition of Prostate-specific Membrane Antigen Positron
Emission Tomography to the Most Frequently Used Nomograms for the Prediction of Pelvic Lymph-node Metastases: An
International Multicenter Study. Eur. Urol. 2021,80, 234–242. [CrossRef]
59.
Guidelines EAU. Prostate Cancer. 2022. Available online: https://uroweb.org/guidelines/prostate-cancer/chapter/treatment
(accessed on 18 June 2022).
60.
Gandaglia, G.; Abdollah, F.; Schiffmann, J.; Trudeau, V.; Shariat, S.F.; Kim, S.P.; Perrotte, P.; Montorsi, F.; Briganti, A.;
Trinh, Q.D.; et al.
Distribution of metastatic sites in patients with prostate cancer: A population-based analysis. Prostate
2014
,
74, 210–216. [CrossRef]
61.
Pasoglou, V.; Michoux, N.; Van Damme, J.; Van Nieuwenhove, S.; Halut, M.; Triqueneaux, P.; Tombal, B.; Lecouvet, F.E. Pattern of
metastatic deposit in recurrent prostate cancer: A whole-body MRI-based assessment of lesion distribution and effect of primary
treatment. World J. Urol. 2019,37, 2585–2595. [CrossRef]
62.
Kane, C.J.; Amling, C.L.; Johnstone, P.A.; Pak, N.; Lance, R.S.; Thrasher, J.B.; Foley, J.P.; Riffenburgh, R.H.; Moul, J.W. Limited
value of bone scintigraphy and computed tomography in assessing biochemical failure after radical prostatectomy. Urology
2003
,
61, 607–611. [CrossRef]
63.
Zhou, X.; Jiang, X.; Liu, L.; Wang, X.; Li, C.; Yao, Y.; Kou, Y.; Shen, J.; Shen, T.; Li, Z.; et al. Evaluation of
18
F-PSMA-1007 PET/CT
in prostate cancer patients with biochemical recurrence after radical prostatectomy. Transl. Oncol.
2022
,15, 101292. [CrossRef]
[PubMed]
64.
Lengana, T.; Lawal, I.O.; Boshomane, T.G.; Popoola, G.O.; Mokoala, K.M.G.; Moshokoa, E.; Maes, A.; Mokgoro, N.P.;
Van de Wiele, C.;
Vorster, M.; et al.
68
Ga-PSMA PET/CT Replacing Bone Scan in the Initial Staging of Skeletal Metastasis in
Prostate Cancer: A Fait Accompli? Clin. Genitourin. Cancer 2018,16, 392–401. [CrossRef] [PubMed]
65.
Regula, N.; Kostaras, V.; Johansson, S.; Trampal, C.; Lindström, E.; Lubberink, M.; Iyer, V.; Velikyan, I.; Sörensen, J. Comparison of
68
Ga-PSMA PET/CT with fluoride PET/CT for detection of bone metastatic disease in prostate cancer. Eur. J. Hybrid Imaging
2022,6, 5. [CrossRef] [PubMed]
66.
Pouessel, D.; Gallet, B.; Bibeau, F.; Avancès, C.; Iborra, F.; Sénesse, P.; Culine, S. Liver metastases in prostate carcinoma: Clinical
characteristics and outcome. BJU Int. 2007,99, 807–811. [CrossRef]
67.
Parimi, V.; Goyal, R.; Poropatich, K.; Yang, X.J. Neuroendocrine differentiation of prostate cancer: A review. Am. J. Clin. Exp. Urol.
2014,2, 273–285.
68.
Damjanovic, J.; Janssen, J.C.; Prasad, V.; Diederichs, G.; Walter, T.; Brenner, W.; Makowski, M.R.
68
Ga-PSMA-PET/CT for the
evaluation of liver metastases in patients with prostate cancer. Cancer Imaging 2019,19, 37. [CrossRef]
69.
Bubendorf, L.; Schopfer, A.; Wagner, U.; Sauter, G.; Moch, H.; Willi, N.; Gasser, T.C.; Mihatsch, M.J. Metastatic patterns of prostate
cancer: An autopsy study of 1,589 patients. Hum. Pathol. 2000,31, 578–583. [CrossRef]
70.
Damjanovic, J.; Janssen, J.C.; Furth, C.; Diederichs, G.; Walter, T.; Amthauer, H.; Makowski, M.R.
68
Ga-PSMA-PET/CT for the
evaluation of pulmonary metastases and opacities in patients with prostate cancer. Cancer Imaging 2018,18, 20. [CrossRef]
71.
Pyka, T.; Weirich, G.; Einspieler, I.; Maurer, T.; Theisen, J.; Hatzichristodoulou, G.; Schwamborn, K.; Schwaiger, M.; Eiber, M.
68
Ga-PSMA-HBED-CC PET for Differential Diagnosis of Suggestive Lung Lesions in Patients with Prostate Cancer. J. Nucl. Med.
2016,57, 367–371. [CrossRef]
72.
Bouchelouche, K.; Vendelbo, M.H. Pulmonary Opacities and Bronchiectasis Avid on
68
Ga-PSMA PET. Clin. Nucl. Med.
2017
,
42, e216–e217. [CrossRef]
73.
Ardies, P.J.; Gykiere, P.; Goethals, L.; De Mey, J.; De Geeter, F.; Everaert, H. PSMA Uptake in Mediastinal Sarcoidosis. Clin. Nucl.
Med. 2017,42, 303–305. [CrossRef] [PubMed]
74.
Kesch, C.; Kratochwil, C.; Mier, W.; Kopka, K.; Giesel, F.L.
68
Ga or
18
F for Prostate Cancer Imaging? J. Nucl. Med.
2017
,
58, 687–688
.
[CrossRef] [PubMed]
75.
Afshar-Oromieh, A.; Avtzi, E.; Giesel, F.L.; Holland-Letz, T.; Linhart, H.G.; Eder, M.; Eisenhut, M.; Boxler, S.; Hadaschik, B.A.;
Kratochwil, C.; et al. The diagnostic value of PET/CT imaging with the
68
Ga-labelled PSMA ligand HBED-CC in the diagnosis of
recurrent prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 2015,42, 197–209. [CrossRef] [PubMed]
76.
Rahbar, K.; Afshar-Oromieh, A.; Bogemann, M.; Wagner, S.; Schafers, M.; Stegger, L.; Weckesser, M.
18
F-PSMA-1007 PET/CT at 60
and 120 minutes in patients with prostate cancer: Biodistribution, tumour detection and activity kinetics. Eur. J. Nucl. Med. Mol.
Imaging 2018,45, 1329–1334. [CrossRef] [PubMed]
77.
Afshar-Oromieh, A.; Hetzheim, H.; Kubler, W.; Kratochwil, C.; Giesel, F.L.; Hope, T.A.; Eder, M.; Eisenhut, M.; Kopka, K.;
Haberkorn, U. Radiation dosimetry of
68
Ga-PSMA-11 (HBED-CC) and preliminary evaluation of optimal imaging timing. Eur. J.
Nucl. Med. Mol. Imaging 2016,43, 1611–1620. [CrossRef]
Diagnostics 2022,12, 2594 23 of 25
78.
Rahbar, K.; Weckesser, M.; Ahmadzadehfar, H.; Schafers, M.; Stegger, L.; Bogemann, M. Advantage of
18
F-PSMA-1007 over
68
Ga-PSMA-11 PET imaging for differentiation of local recurrence vs. urinary tracer excretion. Eur. J. Nucl. Med. Mol. Imaging
2018,45, 1076–1077. [CrossRef]
79.
Fendler, W.P.; Calais, J.; Eiber, M.; Simko, J.P.; Kurhanewicz, J.; Santos, R.D.; Feng, F.Y.; Reiter, R.E.; Rettig, M.B.;
Nickols, N.G.; et al.
False positive PSMA PET for tumor remnants in the irradiated prostate and other interpretation pitfalls in a prospective multi-
center trial. Eur. J. Nucl. Med. Mol. Imaging 2021,48, 501–508. [CrossRef]
80.
Rosenzweig, B.; Haramaty, R.; Davidson, T.; Lazarovich, A.; Shvero, A.; Haifler, M.; Gal, J.; Golan, S.; Shpitzer, S.;
Hoffman, A.; et al.
Very Low Prostate PET/CT PSMA Uptake May Be Misleading in Staging Radical Prostatectomy Candidates. J.
Pers. Med. 2022,12, 410. [CrossRef]
81.
Afshar-Oromieh, A.; Holland-Letz, T.; Giesel, F.L.; Kratochwil, C.; Mier, W.; Haufe, S.; Debus, N.; Eder, M.; Eisenhut, M.;
Schäfer, M.; et al.
Diagnostic performance of
68
Ga-PSMA-11 (HBED-CC) PET/CT in patients with recurrent prostate cancer:
Evaluation in 1007 patients. Eur. J. Nucl. Med. Mol. Imaging 2017,44, 1258–1268. [CrossRef]
82.
Ross, J.S.; Sheehan, C.E.; Fisher, H.A.; Kaufman, R.P., Jr.; Kaur, P.; Gray, K.; Webb, I.; Gray, G.S.; Mosher, R.; Kallakury, B.V.
Correlation of primary tumor prostate-specific membrane antigen expression with disease recurrence in prostate cancer. Clin.
Cancer Res. 2003,9, 6357–6362.
83.
Watts, K.; Li, J.; Magi-Galluzzi, C.; Zhou, M. Incidence and clinicopathological characteristics of intraductal carcinoma detected in
prostate biopsies: A prospective cohort study. Histopathology 2013,63, 574–579. [CrossRef] [PubMed]
84.
Morgan, T.M.; Welty, C.J.; Vakar-Lopez, F.; Lin, D.W.; Wright, J.L. Ductal adenocarcinoma of the prostate: Increased mortality risk
and decreased serum prostate specific antigen. J. Urol. 2010,184, 2303–2307. [CrossRef] [PubMed]
85.
Ranasinha, N.; Omer, A.; Philippou, Y.; Harriss, E.; Davies, L.; Chow, K.; Chetta, P.M.; Erickson, A.; Rajakumar, T.;
Mills, I.G.; et al.
Ductal adenocarcinoma of the prostate: A systematic review and meta-analysis of incidence, presentation, prognosis, and
management. BJUI Compass 2021,2, 13–23. [CrossRef] [PubMed]
86.
McEwan, L.M.; Wong, D.; Yaxley, J. Flourodeoxyglucose positron emission tomography scan may be helpful in the case of ductal
variant prostate cancer when prostate specific membrane antigen ligand positron emission tomography scan is negative. J. Med.
Imaging Radiat. Oncol. 2017,61, 503–505. [CrossRef]
87.
Fizazi, K.; Tran, N.; Fein, L.; Matsubara, N.; Rodriguez-Antolin, A.; Alekseev, B.Y.; Özgüro˘glu, M.; Ye, D.; Feyerabend, S.;
Protheroe, A. Abiraterone acetate plus prednisone in patients with newly diagnosed high-risk metastatic castration-sensitive
prostate cancer (LATITUDE): Final overall survival analysis of a randomised, double-blind, phase 3 trial. Lancet Oncol.
2019
,
20, 686–700. [CrossRef]
88.
Sweeney, C.J.; Chen, Y.-H.; Carducci, M.; Liu, G.; Jarrard, D.F.; Eisenberger, M.; Wong, Y.-N.; Hahn, N.; Kohli, M.;
Cooney, M.M.; et al
. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer. N. Engl. J. Med.
2015
,
373, 737–746. [CrossRef]
89.
Kyriakopoulos, C.E.; Chen, Y.H.; Carducci, M.A.; Liu, G.; Jarrard, D.F.; Hahn, N.M.; Shevrin, D.H.; Dreicer, R.; Hussain, M.;
Eisenberger, M.; et al. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer: Long-Term Survival Analysis
of the Randomized Phase III E3805 CHAARTED Trial. J. Clin. Oncol. 2018,36, 1080–1087. [CrossRef]
90.
Parker, C.C.; James, N.D.; Brawley, C.D.; Clarke, N.W.; Hoyle, A.P.; Ali, A.; Ritchie, A.W.S.; Attard, G.; Chowdhury, S.;
Cross, W.; et al
. Radiotherapy to the primary tumour for newly diagnosed, metastatic prostate cancer (STAMPEDE): A ran-
domised controlled phase 3 trial. Lancet 2018,392, 2353–2366. [CrossRef]
91.
Henkenberens, C.; Derlin, T.; Bengel, F.; Ross, T.L.; Kuczyk, M.A.; Giordano, F.A.; Sarria, G.R.; Schmeel, L.C.; Christiansen, H.;
von Klot, C.A.J. Efficacy of PSMA PET-Guided Radiotherapy for Oligometastatic Castrate-Resistant Prostate Cancer. Front. Oncol.
2021,11, 664225. [CrossRef]
92.
Osmonov, D.K.; Aksenov, A.V.; Boller, A.; Kalz, A.; Heimann, D.; Janssen, I.; Jünemann, K.P. Extended salvage pelvic lymph node
dissection in patients with recurrent prostate cancer. Adv. Urol. 2014,2014, 321619. [CrossRef]
93.
Kolontarev, K.; Govorov, A.; Kasyan, G.; Rasner, P.; Vasiliev, A.; Pushkar, D. Extended robotic salvage lymphadenectomy in
patients with ‘node-only’ prostate cancer recurrence: Initial experience. Cent. Eur. J. Urol. 2018,71, 162–167. [CrossRef]
94.
Bravi, C.A.; Fossati, N.; Gandaglia, G.; Suardi, N.; Mazzone, E.; Robesti, D.; Osmonov, D.; Juenemann, K.P.; Boeri, L.; Jeffrey
Karnes, R.; et al. Long-term Outcomes of Salvage Lymph Node Dissection for Nodal Recurrence of Prostate Cancer After Radical
Prostatectomy: Not as Good as Previously Thought. Eur. Urol. 2020,78, 661–669. [CrossRef] [PubMed]
95.
Mazzola, R.; Francolini, G.; Triggiani, L.; Napoli, G.; Cuccia, F.; Nicosia, L.; Livi, L.; Magrini, S.M.; Salgarello, M.;
Alongi, F.
Metastasis-directed Therapy (SBRT) Guided by PET-CT
18
F-CHOLINE Versus PET-CT
68
Ga-PSMA in Castration-sensitive
Oligorecurrent Prostate Cancer: A Comparative Analysis of Effectiveness. Clin. Genitourin. Cancer
2021
,19, 230–236. [CrossRef]
96.
Tu, X.; Zhang, C.; Liu, Z.; Shen, G.; Wu, X.; Nie, L.; Chang, T.; Xu, H.; Bao, Y.; Yang, L.; et al. The Role of 68Ga-PSMA Positron
Emission Tomography/Computerized Tomography for Preoperative Lymph Node Staging in Intermediate/High Risk Patients
With Prostate Cancer: A Diagnostic Meta-Analysis. Front. Oncol. 2020,10, 1365. [CrossRef] [PubMed]
97.
Rogowski, P.; Trapp, C.; von Bestenbostel, R.; Schmidt-Hegemann, N.-S.; Shi, R.; Ilhan, H.; Kretschmer, A.; Stief, C.;
Ganswindt, U.
;
Belka, C.; et al. Outcomes of metastasis-directed therapy of bone oligometastatic prostate cancer. Radiat. Oncol.
2021
,16, 125.
[CrossRef] [PubMed]
98.
De Meerleer, G.; Berghen, C.; Briganti, A.; Vulsteke, C.; Murray, J.; Joniau, S.; Leliveld, A.M.; Cozzarini, C.; Decaestecker, K.;
Rans, K.; et al. Elective nodal radiotherapy in prostate cancer. Lancet Oncol. 2021,22, e348–e357. [CrossRef]
Diagnostics 2022,12, 2594 24 of 25
99.
Artibani, W.; Porcaro, A.B.; De Marco, V.; Cerruto, M.A.; Siracusano, S. Management of Biochemical Recurrence after Primary
Curative Treatment for Prostate Cancer: A Review. Urol. Int. 2018,100, 251–262. [CrossRef]
100.
Roach, M., 3rd; Hanks, G.; Thames, H., Jr.; Schellhammer, P.; Shipley, W.U.; Sokol, G.H.; Sandler, H. Defining biochemical failure
following radiotherapy with or without hormonal therapy in men with clinically localized prostate cancer: Recommendations of
the RTOG-ASTRO Phoenix Consensus Conference. Int. J. Radiat. Oncol. Biol. Phys. 2006,65, 965–974. [CrossRef]
101.
Fanti, S.; Minozzi, S.; Antoch, G.; Banks, I.; Briganti, A.; Carrio, I.; Chiti, A.; Clarke, N.; Eiber, M.; De Bono, J.; et al. Consensus on
molecular imaging and theranostics in prostate cancer. Lancet Oncol. 2018,19, e696–e708. [CrossRef]
102.
Trabulsi, E.J.; Rumble, R.B.; Jadvar, H.; Hope, T.; Pomper, M.; Turkbey, B.; Rosenkrantz, A.B.; Verma, S.; Margolis, D.J.;
Froemming, A.; et al.
Optimum Imaging Strategies for Advanced Prostate Cancer: ASCO Guideline. J. Clin. Oncol.
2020
,
38, 1963–1996. [CrossRef]
103.
Créhange, G.; Chen, C.P.; Hsu, C.C.; Kased, N.; Coakley, F.V.; Kurhanewicz, J.; Roach, M., 3rd. Management of prostate cancer
patients with lymph node involvement: A rapidly evolving paradigm. Cancer Treat. Rev.
2012
,38, 956–967. [CrossRef] [PubMed]
104.
Dotan, Z.A.; Bianco, F.J., Jr.; Rabbani, F.; Eastham, J.A.; Fearn, P.; Scher, H.I.; Kelly, K.W.; Chen, H.N.; Schöder, H.; Hricak, H.; et al.
Pattern of prostate-specific antigen (PSA) failure dictates the probability of a positive bone scan in patients with an increasing
PSA after radical prostatectomy. J. Clin. Oncol. 2005,23, 1962–1968. [CrossRef] [PubMed]
105.
Sathianathen, N.J.; Butaney, M.; Konety, B.R. The utility of PET-based imaging for prostate cancer biochemical recurrence: A
systematic review and meta-analysis. World J. Urol. 2019,37, 1239–1249. [CrossRef]
106.
Eiber, M.; Maurer, T.; Souvatzoglou, M.; Beer, A.J.; Ruffani, A.; Haller, B.; Graner, F.P.; Kübler, H.; Haberkorn, U.;
Eisenhut, M.; et al.
Evaluation of Hybrid
68
Ga-PSMA Ligand PET/CT in 248 Patients with Biochemical Recurrence After Radical Prostatectomy. J.
Nucl. Med. 2015,56, 668–674. [CrossRef] [PubMed]
107.
Fendler, W.P.; Eiber, M.; Beheshti, M.; Bomanji, J.; Ceci, F.; Cho, S.; Giesel, F.; Haberkorn, U.; Hope, T.A.; Kopka, K.; et al.
68
Ga-PSMA PET/CT: Joint EANM and SNMMI procedure guideline for prostate cancer imaging: Version 1.0. Eur. J. Nucl. Med.
Mol. Imaging 2017,44, 1014–1024. [CrossRef]
108.
Perera, M.; Papa, N.; Roberts, M.; Williams, M.; Udovicich, C.; Vela, I.; Christidis, D.; Bolton, D.; Hofman, M.S.;
Lawrentschuk, N.; et al
. Gallium-68 Prostate-specific Membrane Antigen Positron Emission Tomography in Advanced
Prostate Cancer-Updated Diagnostic Utility, Sensitivity, Specificity, and Distribution of Prostate-specific Membrane Antigen-avid
Lesions: A Systematic Review and Meta-analysis. Eur. Urol. 2020,77, 403–417. [CrossRef]
109.
von Eyben, F.E.; Picchio, M.; von Eyben, R.; Rhee, H.; Bauman, G.
68
Ga-Labeled Prostate-specific Membrane Antigen Ligand
Positron Emission Tomography/Computed Tomography for Prostate Cancer: A Systematic Review and Meta-analysis. Eur. Urol.
Focus 2018,4, 686–693. [CrossRef]
110.
Tan, N.; Oyoyo, U.; Bavadian, N.; Ferguson, N.; Mukkamala, A.; Calais, J.; Davenport, M.S. PSMA-targeted Radiotracers versus
18
F Fluciclovine for the Detection of Prostate Cancer Biochemical Recurrence after Definitive Therapy: A Systematic Review and
Meta-Analysis. Radiology 2020,296, 44–55. [CrossRef]
111.
Hope, T.A.; Goodman, J.Z.; Allen, I.E.; Calais, J.; Fendler, W.P.; Carroll, P.R. Metaanalysis of
68
Ga-PSMA-11 PET Accuracy for the
Detection of Prostate Cancer Validated by Histopathology. J. Nucl. Med. 2019,60, 786–793. [CrossRef]
112.
Matushita, C.S.; da Silva, A.M.M.; Schuck, P.N.; Bardisserotto, M.; Piant, D.B.; Pereira, J.L.; Cerci, J.J.; Coura-Filho, G.B.;
Esteves, F.P.
; Amorim, B.J.; et al.
68
Ga-Prostate-specific membrane antigen (psma) positron emission tomography (pet) in prostate
cancer: A systematic review and meta-analysis. Int. Braz J. Urol. 2021,47, 705–729. [CrossRef]
113.
Kimura, S.; Abufaraj, M.; Janisch, F.; Iwata, T.; Parizi, M.K.; Foerster, B.; Fossati, N.; Briganti, A.; Egawa, S.; Hartenbach, M.; et al.
Performance of [
68
Ga] Ga-PSMA 11 PET for detecting prostate cancer in the lymph nodes before salvage lymph node dissection:
A systematic review and meta-analysis. Prostate Cancer Prostatic Dis. 2020,23, 1–10. [CrossRef] [PubMed]
114.
Roach, P.J.; Francis, R.; Emmett, L.; Hsiao, E.; Kneebone, A.; Hruby, G.; Eade, T.; Nguyen, Q.A.; Thompson, B.D.; Cusick, T.; et al.
The Impact of
68
Ga-PSMA PET/CT on Management Intent in Prostate Cancer: Results of an Australian Prospective Multicenter
Study. J. Nucl. Med. 2018,59, 82–88. [CrossRef]
115.
Donswijk, M.L.; van Leeuwen, P.J.; Vegt, E.; Cheung, Z.; Heijmink, S.W.T.P.J.; van der Poel, H.G.; Stokkel, M.P.M. Clinical impact
of PSMA PET/CT in primary prostate cancer compared to conventional nodal and distant staging: A retrospective single center
study. BMC Cancer 2020,20, 723. [CrossRef] [PubMed]
116.
Han, S.; Woo, S.; Kim, Y.J.; Suh, C.H. Impact of
68
Ga-PSMA PET on the Management of Patients with Prostate Cancer: A
Systematic Review and Meta-analysis. Eur. Urol. 2018,74, 179–190. [CrossRef] [PubMed]
117.
Lawal, I.O.; Lengana, T.; Popoola, G.O.; Orunmuyi, A.T.; Kgatle, M.M.; Mokoala, K.M.G.; Sathekge, M.M. Pattern of Prostate
Cancer Recurrence Assessed by
68
Ga-PSMA-11 PET/CT in Men Treated with Primary Local Therapy. J. Clin. Med.
2021
,10, 3883.
[CrossRef]
118.
Armstrong, W.; Thin, P.; Alano, R.; Nguyen, K.; Booker, K.; Gartmann, J.; Lok, V.; Lira, S.; Sonni, I.; Czernin, J.; et al. Incidence
of local prostate cancer recurrence by
68
Ga-PSMA-11 PET/CT is higher after definitive Radiation Therapy than after Radical
Prostatectomy: A single center post-hoc retrospective analysis of recurrence patterns of 787 patients. J. Nucl. Med.
2020
,
61 (Suppl. 1), 41.
119.
Mottet, N.; Bellmunt, J.; Bolla, M.; Briers, E.; Cumberbatch, M.G.; De Santis, M.; Fossati, N.; Gross, T.; Henry, A.M.; Joniau, S.; et al.
EAU-ESTRO-SIOG Guidelines on Prostate Cancer. Part 1: Screening, Diagnosis, and Local Treatment with Curative Intent. Eur.
Urol. 2017,71, 618–629. [CrossRef]
Diagnostics 2022,12, 2594 25 of 25
120.
Barbosa, F.G.; Queiroz, M.A.; Nunes, R.F.; Viana, P.C.C.; Marin, J.F.G.; Cerri, G.G.; Buchpiguel, C.A. Revisiting Prostate Cancer
Recurrence with PSMA PET: Atlas of Typical and Atypical Patterns of Spread. Radiographics 2019,39, 186–212. [CrossRef]
121.
Rogowski, P.; Trapp, C.; von Bestenbostel, R.; Eze, C.; Ganswindt, U.; Li, M.; Unterrainer,M.; Zacherl, M.J.; Ilhan, H.;
Beyer, L.; et a
l.
Outcome after PSMA-PET/CT-based salvage radiotherapy for nodal recurrence after radical prostatectomy. Eur. J. Nucl. Med.
Mol. Imaging 2022,49, 1417–1428. [CrossRef]
122.
Brenner, D.J.; Elliston, C.D. Estimated radiation risks potentially associated with full-body CT screening. Radiology
2004
,
232, 735–738. [CrossRef]
123. Kapoor, M.; Kasi, A. PET Scanning. In StatPearls; University of Kansas: Lawrence, KS, USA, 2022.
124.
Afshar-Oromieh, A.; Malcher, A.; Eder, M.; Eisenhut, M.; Linhart, H.G.; Hadaschik, B.A.; Holland-Letz, T.; Giesel, F.L.;
Kratochwil, C.;
Haufe, S.; et al. PET imaging with a [
68
Ga]gallium-labelled PSMA ligand for the diagnosis of prostate can-
cer: Biodistribution in humans and first evaluation of tumour lesions. Eur. J. Nucl. Med. Mol. Imaging
2013
,40, 486–495. [CrossRef]
[PubMed]
125.
Mannweiler, S.; Amersdorfer, P.; Trajanoski, S.; Terrett, J.A.; King, D.; Mehes, G. Heterogeneity of prostate-specific membrane
antigen (PSMA) expression in prostate carcinoma with distant metastasis. Pathol. Oncol. Res.
2009
,15, 167–172. [CrossRef]
[PubMed]
126.
Cardinale, J.; Schäfer, M.; Benešová, M.; Bauder-Wüst, U.; Leotta, K.; Eder, M.; Neels, O.C.; Haberkorn, U.; Giesel, F.L.; Kopka, K.
Preclinical Evaluation of
18
F-PSMA-1007, a New Prostate-Specific Membrane Antigen Ligand for Prostate Cancer Imaging. J.
Nucl. Med. 2017,58, 425–431. [CrossRef] [PubMed]
127.
Sprute, K.; Kramer, V.; Koerber, S.A.; Meneses, M.; Fernandez, R.; Soza-Ried, C.; Eiber, M.; Weber, W.A.; Rauscher, I.;
Rahbar, K.; et al
. Diagnostic Accuracy of
18
F-PSMA-1007 PET/CT Imaging for Lymph Node Staging of Prostate Carcinoma in
Primary and Biochemical Recurrence. J. Nucl. Med. 2021,62, 208–213. [CrossRef]
128.
Rauscher, I.; Krönke, M.; König, M.; Gafita, A.; Maurer, T.; Horn, T.; Schiller, K.; Weber, W.; Eiber, M. Matched-Pair Comparison of
68
Ga-PSMA-11 PET/CT and
18
F-PSMA-1007 PET/CT: Frequency of Pitfalls and Detection Efficacy in Biochemical Recurrence
After Radical Prostatectomy. J. Nucl. Med. 2020,61, 51–57. [CrossRef] [PubMed]
129.
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. [CrossRef]
130.
Rowe, S.P.; Gorin, M.A.; Pomper, M.G. Imaging of prostate-specific membrane antigen with small-molecule PET radiotracers:
From the bench to advanced clinical applications. Annu. Rev. Med. 2019,70, 461–477. [CrossRef]
131.
McInnes, L.; Zia, N.; Cullinane, C.; Van Zuylekom, J.; Jackson, S.; Stoner, J.; Haskali, M.; Roselt, P.; Van Dam, E.; Harris, M.; et al.
A Cu-64/Cu-67 bifunctional PSMA ligand as a theranostic for prostate cancer. J. Nucl. Med. 2020,61, 1215.
132.
Jilg, C.A.; Reichel, K.; Stoykow, C.; Rischke, H.C.; Bartholomä, M.; Drendel, V.; von Büren, M.; Schultze-Seemann, W.; Meyer, P.T.;
Mix, M. Results from extended lymphadenectomies with [
111
In]PSMA-617 for intraoperative detection of PSMA-PET/CT-positive
nodal metastatic prostate cancer. EJNMMI Res. 2020,10, 17. [CrossRef]
133.
Derks, Y.H.W.; Löwik, D.; Sedelaar, J.P.M.; Gotthardt, M.; Boerman, O.C.; Rijpkema, M.; Lütje, S.; Heskamp, S. PSMA-targeting
agents for radio- and fluorescence-guided prostate cancer surgery. Theranostics 2019,9, 6824–6839. [CrossRef]
134.
Banerjee, S.R.; Lisok, A.; Minn, I.; Josefsson, A.; Kumar, V.; Brummet, M.; Boinapally, S.; Brayton, C.; Mease, R.C.;
Sgouros, G.; et al.
Preclinical Evaluation of
213
Bi- and
225
Ac-Labeled Low-Molecular-Weight Compounds for Radiopharmaceutical Therapy of
Prostate Cancer. J. Nucl. Med. 2021,62, 980–988. [CrossRef] [PubMed]
135.
AJCC TNM Staging. Prostate Conditions Education Council. Available online: https://www.cancer.org/cancer/prostate-cancer/
detection-diagnosis-staging/staging.html (accessed on 14 March 2022).
... In the treatment of high-risk PCa, the cancer stage, along with GS affects the treatment decision. MpMRI and PSMA PET/CT are currently used diagnostic tools for local and distant staging [13]. When both tests are used together, the ability to predict extracapsular extension is increased [14,15]. ...
Article
Full-text available
Background To investigate the predictable parameters associated with downgrading in patients with a Gleason score (GS) 8 (4+4) in prostate biopsy after radical prostatectomy. Methods We retrospectively analyzed 62 patients with a GS of 4+4 on prostate biopsy who underwent robotic radical prostatectomy between 2017 and 2022. Results 38 of 62 (61.2%) were downgraded. In multivariable logistic regression model, Ga-68 prostate-specific membrane antigen (PSMA) positron-emission tomography (PET)/computed tomography (CT) SUV max was independent predictor of downgrading (OR 0.904; p = 0.011) and a Logistic Regression model was constructed using the following formula: Y = 1.465–0.95 (PSMA PET/CT SUV max). The model using this variable correctly predicted the downgrading in 72.6% of patients. The AUC for PSMA PET/CT SUV max was 0.709 the cut off being 8.8. A subgroup analysis was performed in 37 patients who had no other European Association of Urology (EAU) high risk features. 25 out of 37 (67.5%) were downgraded, and 21 of these 25 had organ confined disease. Low PSMA SUV max (<8.1) and percentage of GS 4+4 biopsy cores to cancer bearing cores (45.0%) were independently associated with downgrading to GS 7. Conclusion PSMA PET/CT can be used to predict downgrading in patients with GS 4+4 PCa. Patients with GS 4+4 disease, but no other EAU high risk features, low percentage of GS 4+4 biopsy cores to cancer bearing cores, and a low PSMA PET/CT SUV max are associated with a high likelihood of the cancer reclassification to intermediate risk group.
... La correcta etapificación del CaP es fundamental para seleccionar adecuadamente el tratamiento más preciso para cada paciente. Las técnicas convencionales han mostrado poca precisión para la detección de metástasis ganglionares 3 . ...
... Стоит отметить, что для визуализации РПЖ разработано и применяется наибольшее количество радиофармацевтических препаратов (РФП), которые условно можно разделить на метаболические и рецепторные агенты, а также радиомеченые антитела. В настоящее время наиболее широко применяемыми РФП для позитронно-эмиссионной томографии, совмещенной с компьютерной томографией (ПЭТ/КТ), являются такие рецепторные препараты, как лиганды к простатическому специфическому мембранному антигену (ПСМА), экспрессия которого значительно повышена при наиболее часто встречаемых фенотипах РПЖ [1,2]. ...
Article
It is established that prostate-specific membrane antigen (PSMA), despite its name, is expressed in many tissues other than the prostate gland, both within physiological conditions and in various pathological processes. Additionally, apart from prostate cancer, other malignant tumors are characterized by increased PSMA expression which, according to many authors, is associated with neoangiogenesis. These factors are reflected in the results of PSMA-radioligand imaging and require comprehensive approach to image interpretation including evaluation of computed tomography and magnetic resonance semiotics. In addition, rare cases of distant visceral prostate cancer metastasis in the form of solitary lesions also should be considered during interpretation of the results of radiologic imaging including positron emission tomography/computed tomography. We present two clinical cases in which positron emission tomography/computed tomography revealed solitary foci of pathological 18F-PSMA-1007 uptake outside the areas of typical metastatic spread (with exception of advanced disease) of prostate cancer, specifically in the stomach wall and the left cerebellar hemisphere. In the first case histological examination results revealed a metachronous low grade neuroendocrine tumor of the stomach, in the second case a metastatic lesion of the cerebellum was diagnosed as part of the underlying disease.
... The advent of prostate-specific radiotracers (notably prostate-specific membrane antigen [PSMA]) allows the detection of previously occult micrometastatic disease. 4 For localized PCa not requiring WPRT or to boost the primary disease, brachytherapy-the temporary or permanent placement of sealed radioactive sources ("seeds")-is an appropriate treatment option. Accurate permanent seed placement in low dose rate (LDR) brachytherapy requires ultrasound guidance, while temporary seed placement in high dose rate (HDR) brachytherapy can be accomplished with ultrasound or computed tomography (CT) guidance. ...
Article
Full-text available
Prostate cancer remains a significant global health concern, necessitating continuous research and innovation in treatment modalities. This review explores the currently employed techniques in radiation dose planning and tumor irradiation in the context of prostate cancer management. In addition, we delve into the nuances of expected posttreatment magnetic resonance imaging (MRI) appearances within the gland or in the prostate bed, postradiation tumor recurrence, and its mimics. Radiation therapy (RT) has evolved as a cornerstone in prostate cancer treatment, offering both curative and palliative solutions. Recent developments have seen the emergence of advanced techniques such as intensity-modulated radiation therapy (IMRT) and stereotactic body radiation therapy (SBRT), allowing for precise targeting of cancer cells while minimizing damage to surrounding healthy tissue. The avoidance of normal tissue dose through more conformal dose distribution as in IMRT or proton therapy, improved imaging modalities as in multiparametric magnetic resonance imaging (mpMRI) and prostate positron emission tomography (PET), interventional separation of critical structures from the prostate target, and many other techniques can greatly reduce the side effects of RT. These advancements enhance treatment efficacy and reduce the risk of side effects, promoting improved patient outcomes.
... Prostate cancer (PCa) is the most commonly diagnosed malignancy in men; although, conventional imaging with computed tomography (CT) and technetium-based bone scan are widely used for staging, recently, cumulative evidence indicates that prostate-specific membrane antigen (PSMA) positron-emission tomography (PET/CT) should be a centerpiece of diagnosis and staging for intermediate/high risk patients (1)(2)(3)(4)(5). Although, PSMA PET/CT seems about 27% more accurate than conventional imaging (6) ...
Article
Full-text available
Introduction: To evaluate the accuracy of PSMA PET/CT in the diagnosis and clinical staging of prostatic ductal adenocarcinoma (DAC). Materials and methods: Two Caucasian men 58 and 62 years old were admitted to our Department for dysuria: the patients had not familiarity for prostate cancer (PCa), PSA values were 5.6 and 2.8 ng/ml, digital rectal examination was positive, multiparametric magnetic resonance image (mpMRI) showed for both the presence of an index lesion PIRADS score 5. The patients underwent extended transperineal prostate biopsy combined with four mpMRI/TRUS fusion biopsy under sedation and antibiotic prophylaxis; biopsy histology demonstrated the presence of a mixed PCa characterized by DAC and acinar PCa (Grade Group 4/Gleason score 8). The patients underwent clinical staging performing lung and abdominal CT, bone scan and fluoride 18 (18F) PSMA PET/CT. Results: Conventional imaging was negative for distant metastases; 18F-PSMA PET/CT showed in both patients an intraprostatic lesion characterized by a standardized uptake value (SUVmax) equal to 4.6 and 4.9 in the absence of distant lesions suspicious for metastases. Following multidisciplinary evaluation, the patients underwent radical prostatectomy plus extended pelvic lymphadenectomy. Definitive specimen showed the presence in both cases of a mixed pT3bN1 PCa (ductal plus acinar pattern Grade Group 4) with positive surgical margins, neuronal invasion, and nodes metastases (5/20 and 6/24, respectively). Post-operative PSA in the two patients was 0.8 and 0.3 ng/ml, therefore patients underwent adjuvant therapy. Conclusions: Conventional imaging and PSMA PET/CT could result inadequate in clinical staging of DAC, the use of more imaging data (i.e. mpMRI and/or F-18 FDG) could improve overall accuracy.
... The tumor uptake, which represents PSMA expression, is highly correlated with the aggressiveness of the primary prostatic tumor 48,49 . Although 68 Ga-PSMA-PET/CT is recommended to improve the clinical staging of high-risk PCa and disease recurrence [50][51][52][53][54][55] recently, PSMA PET/ CT has been proposed for the diagnosis of PCa by targeted biopsy [56][57][58][59] and in men enrolled in active Surveillance protocol [60][61][62][63] . A PET/CT scan suspicious for PCa results from a combination of factors, such as homogeneity and intensity of PSMA expression, tumor volume, and grade. ...
Article
Full-text available
Multiparametric magnetic resonance imaging (mpMRI) has improved systematic prostate biopsy procedures in the diagnosis of clinically significant prostate cancer (csPCa) by reducing the number of unnecessary biopsies; numerous level one evidence studies have confirmed the accuracy of MRI-targeted biopsy, but, still today, systematic prostate biopsy is recommended to reduce the 15-20% false negative rate of mpMRI. New advanced imaging has been proposed to detect suspicious lesions and perform targeted biopsies especially when mpMRI cannot be performed. Transrectal ultrasound (TRUS) modalities are emerging as methods with greater sensitivity and specificity for the detection of PCa compared to the traditional TRUS; these techniques include elastography and contrast-enhanced ultrasound, as well as improved B-mode and Doppler techniques. These modalities can be combined to define a novel ultrasound approach: multiparametric ultrasound (mpUS). More recently, micro-ultrasound (MicroUS) and prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) have demonstrated to be sensitive for the detection of primary prostatic lesions resulting highly correlated with the aggressiveness of the primary prostatic tumor. In parallel, artificial intelligence is advancing and is set out to deeply change both radiology and pathology. In this study we address the role, advantages and shortcomings of novel imaging techniques for Pca, and discuss future directions including the applications of artificial intelligence–based techniques to imaging as well as histology. The significance of these findings for the practicing pathologist is discussed. [read full article]
... Over the last decade, the arsenal of minimally invasive methods to assess PCA and its trajectory has vastly expanded. Functional imaging using PSMA-ligand PET/CT has been shown to stage PCA with unprecedented, disease management-changing detection rates (7,8,28,29) and to yield prognostic and predictive information on therapy responses to local and systemic approaches alike (26). In parallel, the quantification of blood-derived cfDNA has been shown repeatedly to be a valuable biomarker of PCA aggressiveness and response to taxane-based chemotherapies (24,25). ...
Article
Full-text available
Functional imaging with prostate-specific membrane antigen (PSMA) ligands has emerged as the standard imaging method for prostate cancer (PCA). In parallel, the analysis of blood-derived, cell-free DNA (cfDNA) has been shown to be a promising quantitative biomarker of PCA aggressiveness and patient outcome. This study aimed to evaluate the relationship and prognostic value of cfDNA concentrations and the PSMA-positive tumor volume (PSMA-TV) in men with PCA undergoing [68Ga]Ga-PSMA-11 PET/CT imaging. Methods: We recruited 148 men with histologically proven PCA (mean age, 70.7 ± 7.7 y) who underwent [68Ga]Ga-PSMA-11 PET/CT (184.9 ± 18.9 MBq) and blood sampling between March 2019 and August 2021. Among these, 74 (50.0%) had hormone-sensitive PCA and 74 (50.0%) had castration-resistant PCA (CRPC). All patients provided written informed consent before blood sample collection and imaging. The cfDNA was extracted and quantified, and PSMA-expressing tumor lesions were delineated to extract the PSMA-TVs. The Spearman coefficient assessed correlations between PSMA-TV and cfDNA concentrations and cfDNA's relation with clinical parameters. The Kruskal-Wallis test examined the mean cfDNA concentration differences based on PSMA-TV quartiles for significantly correlated patient groups. Log-rank and multivariate Cox regression analyses evaluated the prognostic significance of high and low cfDNA and PSMA-TV levels for overall survival. Results: Weak positive correlations were found between cfDNA concentration and PSMA-TV in the overall group (r = 0.16, P = 0.049) and the CRPC group (r = 0.31, P = 0.007) but not in hormone-sensitive PCA patients (r = -0.024, P = 0.837). In the CRPC cohort, cfDNA concentrations significantly differed between PSMA-TV quartiles 4 and 1 (P = 0.002) and between quartiles 4 and 2 (P = 0.016). Survival outcomes were associated with PSMA-TV (P < 0.0001, P = 0.004) but not cfDNA (P = 0.174, P = 0.12), as per the log-rank and Cox regression analysis. Conclusion: These findings suggest that cfDNA might serve as a biomarker of advanced, aggressive CRPC but does not reliably reflect total tumor burden or prognosis. In comparison, [68Ga]Ga-PSMA-11 PET/CT provides a highly granular and prognostic assessment of tumor burden across the spectrum of PCA disease progression.
Article
Objective To investigate the relationship between intraprostatic ⁶⁸ Ga-prostate-specific membrane antigen (PSMA) uptake values and volumetric parameters derived from early pelvic and standard-time whole-body ⁶⁸ Ga-PSMA PET/computed tomography (CT) images in untreated prostate cancer (PCa) patients, and to assess the predictive significance of these data in relation to disease prognosis, comparing them with the Gleason score, clinical risk classification and the presence of metastatic disease detected in 68Ga-PSMA PET/CT imaging. Methods Eighty-one newly diagnosed PCa patients underwent early phase pelvic imaging at the 5 th minute and standard time whole-body imaging at the 60 th minute. Various threshold values were used in intraprostatic delineations to compute maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), intraprostatic PSMA tumor volume and intraprostatic total lesion PSMA uptake. Correlations between early and standard time measurements, as well as changes in SUV parameters over time, were examined. The association of these values with Gleason score, clinical risk status (National Comprehensive Cancer Network), and metastatic disease was explored. Results SUVmax measurements from both early and standard time images distinguished all three groups (clinical risk scores, Gleason score and metastatic group), with standard imaging demonstrating statistical superiority in receiver operating characteristic analyses. Strong correlations were observed between early and standard-time PET parameters. Changes in intraprostatic SUVmax and SUVmean values over time did not exhibit predictive value. Conclusion Although intraprostatic PSMA PET parameters generally aligned at both early and standard times, parameters obtained from standard time images showed more robust correlations with clinical risk scores, Gleason score and metastasis status in newly diagnosed, untreated PCa patients.
Article
Full-text available
Simple Summary The contemporary development of radiomics offers an opportune methodology for the interpretation of prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT). While both technologies are relatively new for consideration of clinical integration, the present exploration seeks to review current literature on their intersection. Review of twenty-three peer-reviewed articles revealed promising results for the use of PSMA PET/CT-derived radiomics in the prediction of biopsy Gleason score, adverse pathology, and treatment outcomes for prostate cancer (PC). Clinical integration of these findings, however, are limited by lack of biologic validation and reproducible methodology. Abstract The clinical integration of prostate membrane specific antigen (PSMA) positron emission tomography and computed tomography (PET/CT) scans represents potential for advanced data analysis techniques in prostate cancer (PC) prognostication. Among these tools is the use of radiomics, a computer-based method of extracting and quantitatively analyzing subvisual features in medical imaging. Within this context, the present review seeks to summarize the current literature on the use of PSMA PET/CT-derived radiomics in PC risk stratification. A stepwise literature search of publications from 2017 to 2023 was performed. Of 23 articles on PSMA PET/CT-derived prostate radiomics, PC diagnosis, prediction of biopsy Gleason score (GS), prediction of adverse pathology, and treatment outcomes were the primary endpoints of 4 (17.4%), 5 (21.7%), 7 (30.4%), and 7 (30.4%) studies, respectively. In predicting PC diagnosis, PSMA PET/CT-derived models performed well, with receiver operator characteristic curve area under the curve (ROC-AUC) values of 0.85–0.925. Similarly, in the prediction of biopsy and surgical pathology results, ROC-AUC values had ranges of 0.719–0.84 and 0.84–0.95, respectively. Finally, prediction of recurrence, progression, or survival following treatment was explored in nine studies, with ROC-AUC ranging 0.698–0.90. Of the 23 studies included in this review, 2 (8.7%) included external validation. While explorations of PSMA PET/CT-derived radiomic models are immature in follow-up and experience, these results represent great potential for future investigation and exploration. Prior to consideration for clinical use, however, rigorous validation in feature reproducibility and biologic validation of radiomic signatures must be prioritized.
Article
Full-text available
Available tests to detect clinically significant prostate cancer frequently lead to overdiagnosis and overtreatment. Our study assessed the feasibility of combining a urinary biomarker-based risk score (SelectMDx ® ) and multiparametric MRI outcomes in order to identify patients with prostate cancer on prostate biopsy with increased accuracy and reliability. Samples of 74 men with suspicion of prostate cancer and available multiparametric MRI were analysed in a prospective cross-sectional study design. First-voided urine for determination of HOXC6 and DLX1 mRNA levels was collected after digital rectal examination and prior to MRI/ultrasound fusion-guided prostate biopsy. All multiparametric MRI images were centrally reviewed by two experienced radiologists blinded for urine test results and biopsy outcome. The PI-RADS v2 was used. SelectMDx ® score, PI-RADS and Gleason Sore were obtained. Associations between Gleason Score, PI-RADS scores and SelectMDx ® were assessed using ANOVA and t-test. Sensitivity and specificity were assessed and evaluated as area-under-the-curve of the receiver operating characteristic. Upon biopsy, 59.5% of patients were diagnosed with prostate cancer, whereby 40.6% had high-grade prostate cancer (GS ≥ 7a). SelectMDx ® scores were significantly higher for patients with positive biopsy findings (49.07 ± 25.99% vs. 22.00 ± 26.43%; p < 0.001). SelectMDx ® scores increased with higher PI-RADS scores. Combining SelectMDx ® , history of prior biopsy with benign histology and PI-RADS scores into a novel scoring system led to significant prostate cancer detection rates with tiered detection rate of 39%, 58%, 81% and 100% for Gleason grade group II, III, IV, and V, respectively. The area-under-the-curve for our novel sum score in receiver operating characteristic analysis was 0.84. The synergistic combination of two non-invasive tests into a sum score with increased sensitivity may help avoiding unnecessary biopsies for initial prostate cancer diagnosis. For confirmation, further prospective studies with larger sample sizes and univariate and multivariate regression analyses and decision curve analyses are required.
Article
Full-text available
Purpose: to evaluate a unique subpopulation of radical prostatectomy (RP) candidates with "negative" prostate 68Ga-labeled prostate-specific membrane antigen (PSMA) positron emission tomography (PET) computed tomography (CT) imaging scans and to characterize the clinical implications of misleading findings. Materials and methods: This case-control retrospective study compared the final histological outcomes of patients with "negative" pre-RP PSMA PET/CT prostate scans (with a prostate maximal standardized uptake value [SUVmax] below the physiologic uptake) to those with an "intense" prostatic tracer uptake (with a SUVmax above the physiologic uptake). The patients underwent an RP between March 2015 and July 2019 in five academic centers. Data on the demographics, comorbidities, prostate-specific antigen (PSA) and rectal exam findings, prior biopsies, imaging results, biopsies, and RP histology results were collected. Results: Ninety-seven of the 392 patients who underwent an RP had PSMA PET/CT imaging preoperatively. Fifty-two (54%) had a "negative" uptake (in the study group), and 45 (46%) had a "positive" uptake (in the control group). Only the lesion size and SUVmax values on the PSMA PET/CT differed between the groups preoperatively. On the histological analysis, only the ISUP score, seminal vesicles invasion, T stage, and positive margin rates differed between the groups (p < 0.05), while 50 (96%) study group patients harbored clinically significant disease (ISUP ≥ 2), with an extra-prostatic disease in 24 (46%), perineural invasion in 35 (67%), and positive lymph nodes in 4 (8%). Conclusions: Disease aggressiveness generally correlated with an intense PSMA uptake on the preoperative PSMA PET/CT, but a subpopulation of patients with clinically significant cancer and aggressive characteristics showed a deceptively weak PSMA uptake. These data raise a concern about the unqualified application of PSMA PET/CT for staging RP candidates.
Article
Full-text available
Background ¹⁸F-NaF positron emission tomography/computed tomography (fluoride PET/CT) is considered the most sensitive technique to detect bone metastasis in prostate cancer (PCa). ⁶⁸Ga-PSMA-11 (PSMA) PET/CT is increasingly used for staging of PCa. This study primarily aimed to compare the diagnostic performance of fluoride PET/CT and gallium-based PSMA PET/CT in identifying bone metastasis followed by a comparison of PSMA PET/CT with contrast-enhanced CT (CE-CT) in identifying soft tissue lesions as a secondary objective. Methods Twenty-eight PCa patients with high suspicion of disseminated disease following curative treatment were prospectively evaluated. PET/CT examinations using fluoride and PSMA were performed. All suspicious bone lesions were counted, and the tracer uptake was measured as standardized uptake values (SUV) for both tracers. In patients with multiple findings, ten bone lesions with highest SUVmax were selected from which identical lesions from both scans were considered for direct comparison of SUVmax. Soft tissue findings of local and lymph node lesions from CE-CT were compared with PSMA PET/CT. Results Both scans were negative for bone lesions in 7 patients (25%). Of 699 lesions consistent with skeletal metastasis in 21 patients on fluoride PET/CT, PSMA PET/CT identified 579 lesions (83%). In 69 identical bone lesions fluoride PET/CT showed significantly higher uptake (mean SUVmax: 73.1 ± 36.8) compared to PSMA PET/CT (34.5 ± 31.4; p < 0.001). Compared to CE-CT, PSMA PET/CT showed better diagnostic performance in locating local (96% vs 61%, p = 0.004) and lymph node (94% vs 46%, p < 0.001) metastasis. Conclusion In this prospective comparative study, PSMA PET/CT detected the majority of bone lesions that were positive on fluoride PET/CT. Further, this study indicates better diagnostic performance of PSMA PET/CT to locate soft tissue lesions compared to CE-CT.
Article
Full-text available
Background Prostate cancer is the most common cancer among men, and its diagnosis and treatment are improving. Our study evaluated how PSMA-PET/CT prior to treatment planning might improve the optimal management of prostate cancer radiotherapy. Methods This retrospective pilot study included 43 prostate cancer (PCa) patients referred to our radiation oncologist department, from the urology department, for radiation therapy. 18F-PSMA-PET/CT was ordered by the radiation oncologists mainly due to the lack of resent image staging. The patients were divided into three different groups according to their initially planned treatments: radical radiation therapy (RT) (newly diagnosed PCa patients), salvage RT (patients with biochemical recurrence after radical prostatectomy), or oligometastatic RT (oligometastatic PCa patients with good response after systemic treatment). Results Following PSMA-PET/CT, the initially planned RT was changed for 60.5% of the patients due to new findings (metastases and/or recurrent disease). The final treatment choice was effected by PSMA-PET/CT outcome in 60.5% (26/43) of the patients, and in 50% (16/32) of patients, the radiation treatment plan changed following PSMA-PET/CT. Only 39.5% (17/43) of the patients who underwent PSMA-PET/CT were treated according to their initial treatment plans. Conclusions Our results indicate that PSMA-PET/CT impacts treatment decisions and the selection of RT as well as adjuvant treatment protocols in the management of prostate cancer.
Article
Full-text available
Purpose Prostate-specific membrane antigen (PSMA) ligands targeting has shown promising results in staging of prostate cancer (PCa). The aim of present study was to evaluate the value of ¹⁸F-PSMA-1007 PET/CT in PCa patients with biochemical recurrence. Methods 71 patients with PCa after radical prostatectomy (RP) were included in the present study. Median prostate-specific antigen (PSA) level was 1.27 ng/mL (range 0.01–67.40 ng/mL, n = 69). All patients underwent whole-body PET/CT imaging after injection of 333±38 MBq ¹⁸F-PSMA-1007. The distribution of PSMA-positive lesions was assessed. The influence of PSA level, androgen deprivation therapy and primary Gleason score on PSMA-positive finding and uptake of ¹⁸F-PSMA-1007 were evaluated. Results 56 (79%) patients showed at least one pathological finding on ¹⁸F-PSMA-1007 PET/CT. The rates of positive scans were 50%, 80%, 100%, 100% among patients with PSA levels ≤0.5, 0.51–1.0, 1.1–2.0 and >2.0 ng/mL, respectively. The median Gleason score was 8 (range 7–10), and higher Gleason score (≤7 vs. ≥8) leads to higher detection rates (58.3% (14/24) vs. 88.9% (32/36), P = 0.006). The median SUVmax of positive findings in patients with PSA levels ≤0.5, 0.51–1.0, 1.1–2.0 and >2.0 ng/mL were 4.51, 4.27, 11.50 and 14.08, respectively. The median SUVmax in patients with PSA level >2.0 ng/mL was significantly higher than that in patients with PSA ≤2.0 ng/mL (14.08 vs. 6.13, P<0.001). Conclusion ¹⁸F-PSMA-1007 PET/CT demonstrated a high detection rate for patients with a raised PSA level after radical prostatectomy even in patients with extremely low PSA level (eg. PSA level ≤0.5 ng/mL), which was essential for further clinical management for PCa patients.
Article
Full-text available
Purpose Comparing mpMRI and ⁶⁸Ga-PSMA-PET/MRI in primary staging of PCa and investigating the value of quantitative mpMRI-measurements for prediction of extracapsular extension and N-metastases. Methods Patients with PCa undergoing ⁶⁸Ga-PSMA-PET/MRI and mpMRI during January 2016 to February 2019 were retrospectively included. Two readers each on ⁶⁸Ga-PSMA-PET/MRI or mpMRI rated extraprostatic extension (≥T3) and regional lymph-node-metastasis (N1) on a Likert-scale. A fifth reader measured tumor volume, maximum diameter, and capsular contact length on mpMRI. Probability of lymph-node-metastasis was additionally calculated using the 2018 Briganti model. Interobserver-agreement was assessed by squared Cohen‘s kappa, and diagnostic accuracy was determined using radical prostatectomy (n=35/49) as reference standard. Results 49 patients (median age 66 years [IQR: 61–72 years]) were evaluated. Interobserver-agreement for mpMRI and ⁶⁸Ga-PSMA-PET/MRI was: ≥T3: κ=0.58/0.47; N1: κ=0.55/0.92. Diagnostic accuracy for mpMRI vs ⁶⁸Ga-PSMA-PET/MRI readers for ≥T3 was AUC: 0.72, 0.62 vs 0.71, 0.72 (p>.38) and for N1 was AUC: 0.39, 0.55 vs 0.72, 0.78 (p<0.01). Quantitative parameters delivered diagnostic accuracies of: AUC: 0.70-0.72 for ≥T3. The 2018 Briganti model achieved an AUC of 0.89 for N1. Conclusions Interreader-agreement regarding ≥ T3 was similar for mpMRI and ⁶⁸Ga-PSMA-PET/MRI while for N1 it was higher for ⁶⁸Ga-PSMA-PET/MRI. Diagnostic accuracy was comparable for ≥T3 while for N1 it was higher in ⁶⁸Ga-PSMA-PET/MRI and the 2018 Briganti model. Combining clinical data and quantitative data from mpMRI in the 2018 Briganti model yielded the highest AUC for prediction of lymph node metastasis and may aid in selecting patients who will benefit from ⁶⁸Ga-PSMA-PET/MRI for primary staging.
Article
Full-text available
Purpose To compare the diagnostic performance of ⁶⁸Ga-PSMA-11 PET/CT and mpMRI for pelvic lymph node staging prior to radical prostatectomy in prostate cancer (PCa) patients based on per patient data. Methods PubMed and Embase databases were searched until October 2020 for eligible studies evaluating head-to-head comparison of ⁶⁸Ga-PSMA-PET/CT and mpMRI for the detection of pelvic lymph node metastases (PLNMs) using pelvic lymph node dissection (PLND) as gold standard. The pooled sensitivity, specificity, and area under the summary receiver-operating characteristics curve (AUC) were determined for the two imaging modalities. Results Nine studies with 640 patients were included. The pooled sensitivity, specificity, and AUC for ⁶⁸Ga-PSMA-11 PET/CT vs. mpMRI were 0.71 (95% CI: 0.48–0.86) vs. 0.40 (95% CI: 0.16–0.71), 0.92 (95% CI: 0.88–0.95) vs. 0.92 (95% CI: 0.80–0.97), and 0.92 (95% CI: 0.88–0.95) vs. 0.82 (95% CI: 0.79–0.86), respectively. There was substantial heterogeneity for both imaging modalities, and meta-regression analysis revealed that the number of patients, prevalence of PLNMs, PSA level, reference standard, and risk classification might be the potential causes of heterogeneity. Conclusion This meta-analysis of head-to-head comparison studies confirms that there is a trend toward a higher sensitivity and diagnostic accuracy of ⁶⁸Ga-PSMA-11 PET/CT compared to mpMRI for the detection of PLNMs in PCa patients. Nevertheless, according to current guidelines, PLND still needs to be recommended in case of negative results from ⁶⁸Ga-PSMA-11 PET/CT due to significant risk of malignancy.
Article
Full-text available
Purpose Nodal recurrent prostate cancer (PCa) represents a common state of disease, amenable to local therapy. PSMA-PET/CT detects PCa recurrence at low PSA levels. The aim of this study was to evaluate the outcome of PSMA-PET/CT-based salvage radiotherapy (sRT) for lymph node (LN) recurrence. Methods A total of 100 consecutive patients treated with PSMA-PET/CT-based salvage elective nodal radiotherapy (sENRT) for LN recurrence were retrospectively examined. Patients underwent PSMA-PET/CT scan due to biochemical persistence (bcP, 76%) or biochemical recurrence (bcR, 24%) after radical prostatectomy (RP). Biochemical recurrence-free survival (BRFS) defined as PSA < post-RT nadir + 0.2 ng/ml and distant metastasis-free survival (DMFS) were calculated using the Kaplan–Meier method and uni- and multivariate analysis was performed. Results Median follow-up was 37 months. Median PSA at PSMA-PET/CT was 1.7 ng/ml (range 0.1–40.1) in patients with bcP and 1.4 ng/ml (range 0.3–5.1) in patients with bcR. PSMA-PET/CT detected 1, 2, and 3 or more LN metastases in 35%, 23%, and 42%, respectively. Eighty-three percent had only pelvic, 2% had only paraaortic, and 15% had pelvic and paraaortic LN metastases. Cumulatively, a total dose converted to EQD21.5 Gy of 66 Gy (60–70 Gy) was delivered to the prostatic fossa, 70 Gy (66–72 Gy) to the local recurrence, if present, 65.1 Gy (56–66 Gy) to PET-positive lymph nodes, and 47.5 Gy (42.4–50.9 Gy) to the lymphatic pathways. Concomitant androgen deprivation therapy (ADT) was administered in 83% of patients. One-, 2-, and 3-year BRFS was 80.7%, 71.6%, and 65.8%, respectively. One-, 2-, and 3-year DMFS was 91.6%, 79.1%, and 66.4%, respectively. In multivariate analysis, concomitant ADT, longer ADT duration (≥ 12 vs. < 12 months) and LN localization (pelvic vs. paraaortic) were associated with improved BRFS and concomitant ADT and lower PSA value before sRT (< 1 vs. > 1 ng/ml) with improved DMFS, respectively. No such association was seen for the number of affected lymph nodes. Conclusions Overall, the present analysis shows that the so far, unmatched sensitivity and specificity of PSMA-PET/CT translates in comparably high BRFS and DMFS after PSMA-PET/CT-based sENRT for patients with PCa LN recurrence. Concomitant ADT, duration of ADT, PSA value before sRT, and localization of LN metastases were significant factors for improved outcome.
Article
Background Extended pelvic nodal dissection (ePLND) represents the gold standard for nodal staging in prostate cancer (PCa). Prostate-specific membrane antigen (PSMA) radioguided surgery (RGS) could identify lymph node invasion (LNI) during robot-assisted radical prostatectomy (RARP). Objective To report the planned interim analyses of a phase 2 prospective study (NCT04832958) aimed at describing PSMA-RGS during RARP. Design, setting, and participants A phase 2 trial aimed at enrolling 100 patients with intermediate- or high-risk cN0cM0 PCa at conventional imaging with a risk of LNI of >5% was conducted. Overall, 18 patients were enrolled between June 2021 and March 2022. Among them, 12 patients underwent PSMA-RGS and represented the study cohort. Surgical procedure All patients received ⁶⁸Ga-PSMA positron emission tomography (PET)/magnetic resonance imaging; 99mTc-PSMA-I&S was synthesised and administered intravenously the day before surgery, followed by single-photon emission computed tomography/computed tomography. A Drop-In gamma probe was used for in vivo measurements. All positive lesions (count rate ≥2 compared with background) were excised and ePLND was performed. Measurements Side effects, perioperative outcomes, and performance characteristics of robot-assisted PSMA-RGS for LNI were measured. Results and limitations Overall, four (33%), six (50%), and two (17%) patients had intermediate-risk, high-risk, and locally advanced PCa. Overall, two (17%) patients had pathologic nodal uptake at PSMA PET. The median operative time, blood loss, and length of stay were 230 min, 100 ml, and 5 d, respectively. No adverse events and intraoperative complications were recorded. One patient experienced a 30-d complication (Clavien-Dindo 2; 8.3%). Overall, three (25%) patients had LNI at ePLND. At per-region analyses on 96 nodal areas, sensitivity, specificity, positive predictive value, and negative predictive value of PSMA-RGS were 63%, 99%, 83%, and 96%, respectively. On a per-patient level, sensitivity, specificity, positive predictive value, and negative predictive values of PSMA-RGS were 67%, 100%, 100%, and 90%, respectively. Conclusions Robot-assisted PSMA-RGS in primary staging is a safe and feasible procedure characterised by acceptable specificity but suboptimal sensitivity, missing micrometastatic nodal disease. Patient summary Prostate-specific membrane antigen radioguided robot-assisted surgery is a safe and feasible procedure for the intraoperative identification of nodal metastases in cN0cM0 prostate cancer patients undergoing robot-assisted radical prostatectomy with extended pelvic lymph node dissection. However, this approach might still miss micrometastatic nodal dissemination.
Article
Context: The role of positron emission tomography/computed tomography (PET/CT) with prostate-specific membrane antigen (PSMA) in the primary staging for patients with prostate cancer (PCa) is still debated. Objective: To analyze published studies reporting the accuracy of PSMA PET/CT for detecting lymph node invasion (LNI) at pelvic lymph node dissection (PLND). Evidence acquisition: A search of PubMed/MEDLINE, Cochrane library's Central, EMBASE and Scopus databases, from inception to May 2021, was conducted. The primary outcome was to evaluate the sensitivity, specificity, positive (PPV) and negative (NPV) predictive values of PSMA PET/CT in detecting LNI on a per-patient level. As a secondary outcome, NPV of PET PSMA was tested on a per-node-level analysis. Detection rates were pooled using random-effect models. Preplanned subgroup analyses tested the diagnostic accuracy after stratification for the preoperative risk group. PPV and NPV variation over LNI prevalence was evaluated. Only studies including extended PLND (ePLND) as the reference standard test were included. Evidence synthesis: Twenty-seven studies, with a total of 2832 participants, were included in quantitative synthesis. The sensitivity, specificity, PPV, and NPV of PSMA PET/CT for LNI were, respectively, 58% (95% confidence interval [CI] 50-66%), 95% (95% CI 93-97%), 79% (95% CI 72-85%), and 87% (95% CI 84-89%), with overall moderate heterogeneity between studies. At bivariate analysis, the diagnostic accuracy of PSMA PET/CT estimated through summary receiver operating characteristic-derived area under the curve was 84% (95% CI 81-87%). On a per-node level, NPV of PET PSMA was 97% (95% CI 96-99%). At subgroup analyses, according to preoperative risk groups, sensitivity, specificity, PPV, and NPV were 51%, 93%, 73%, and 81%, respectively, in high-risk patients. Over the LNI prevalence range of 5-40%, PPV increased from 59% to 91%, while NPV decreased from 99% to 84%. Conclusions: PSMA PET/CT scan provides promising accuracy in the field of primary nodal staging for PCa. The high NPV in men with a lower risk of LNI might be clinically useful to reduce the number of unnecessary PLND procedures performed. Conversely, in high-risk patients, negative PSMA PET/CT cannot replace staging ePLND. Patient summary: In this systematic review and meta-analysis, we demonstrated that prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) scan may optimize the primary nodal staging and surgical management of prostate cancer patients candidate to radical prostatectomy. The high negative predictive value in men with a lower risk of lymph node invasion might be clinically useful for reducing the number of useless pelvic lymph node dissection (PLND) procedures performed. Conversely, in high-risk patients, negative PSMA PET/CT cannot allow avoiding of PLND.