ArticlePDF Available

Comparison of PSMA-based 18F-DCFBC PET/CT to Conventional Imaging Modalities for Detection of Hormone-Sensitive and Castration-Resistant Metastatic Prostate Cancer

Authors:

Abstract and Figures

Methods: Seventeen patients were prospectively enrolled (nine HNPC and eight CRPC); 16 had CIM evidence of new or progressive metastatic prostate cancer and one had high clinical suspicion of metastatic disease. DCFBC PET/CT imaging was obtained with two successive PET scans starting at two hours post-injection. Patients were imaged with CIM at approximately the time of PET. A lesion-by-lesion analysis of PET to CIM was performed in the context of either HNPC or CRPC. The patients were followed with available clinical imaging as a reference standard to determine the true nature of identified lesions on PET and CIM. Results: On the lesion-by-lesion analysis, DCFBC PET was able to detect a larger number of lesions (592 positive with 63 equivocal) than CIM (520 positive with 61 equivocal) overall, in both HNPC and CRPC patients. DCFBC PET detection of lymph nodes, bone lesions, and visceral lesions was superior to CIM. When intrapatient clustering effects were taken into account, DCFBC PET was estimated to be positive in a large proportion of lesions that would be negative or equivocal on CIM (0.45). On follow-up, the sensitivity of DCFBC PET (0.92) was superior to CIM (0.71). DCFBC tumor uptake was increased at the later time PET time point (~ 2.5 hr post-injection) with background uptake showing a decreasing trend on later PET. Conclusion: PET imaging with DCFBC, a small molecule PSMA-targeted radiotracer, detects more lesions than CIM and promised to diagnose and stage patients with metastatic prostate cancer more accurately than current imaging methods.
Content may be subject to copyright.
Title: Comparison of PSMA-based 18F-DCFBC PET/CT to Conventional Imaging Modalities for
Detection of Hormone-Naïve and Castration-Resistant Metastatic Prostate Cancer
Authors: Steven P. Rowe1, Katarzyna J. Macura1,2,3, Anthony Ciarallo1, Esther Mena1, Amanda
Blackford2, Rosa Nadal2, Emmanuel S. Antonarakis2, Mario Eisenberger2, Michael Carducci2,
Ashley Ross3, Philip W. Kantoff4, Daniel P. Holt1, Robert F. Dannals1, Ronnie C. Mease1,
Martin G. Pomper1, and Steve Y. Cho1,5
Author Affiliations: 1The Russell H. Morgan Department of Radiology and Radiological
Science, 2Department of Medical Oncology, and 3The James Buchanan Brady Urological
Institute and Department of Urology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
4Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA. 5Present address:
Department of Radiology, University of Wisconsin School of Medicine and Public Health,
Madison, WI, USA.
First Author:
Steven P. Rowe, M.D., Ph.D.
Resident
The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins
Hospital
601 N. Caroline St., Room 3150, Baltimore, MD 21287
(p) (410) 955-6500; srowe8@jhmi.edu
Corresponding Author:
Steve Y. Cho, M.D.
1111 Highland Avenue, WIMR1 Rm 7139
Madison, WI 53705
Office: (608) 263-5048
Fax: (608) 265-7390
Email: scho@uwhealth.org
Running Title: 18F-DCFBC PET/CT of Metastatic Prostate Cancer
Journal of Nuclear Medicine, published on October 22, 2015 as doi:10.2967/jnumed.115.163782
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
1
ABSTRACT
Conventional imaging modalities (CIM) have limited sensitivity and specificity for detection of
metastatic prostate cancer. We examined the potential of a first-in-class radiofluorinated small-
molecule inhibitor of prostate-specific membrane antigen (PSMA), 18F-DCFBC (DCFBC), to
detect metastatic hormone-naïve (HNPC) and castration-resistant prostate cancer (CRPC).
Methods: Seventeen patients were prospectively enrolled (nine HNPC and eight CRPC); 16 had
CIM evidence of new or progressive metastatic prostate cancer and one had high clinical
suspicion of metastatic disease. DCFBC PET/CT imaging was obtained with two successive
PET scans starting at two hours post-injection. Patients were imaged with CIM at approximately
the time of PET. A lesion-by-lesion analysis of PET to CIM was performed in the context of
either HNPC or CRPC. The patients were followed with available clinical imaging as a
reference standard to determine the true nature of identified lesions on PET and CIM.
Results: On the lesion-by-lesion analysis, DCFBC PET was able to detect a larger number of
lesions (592 positive with 63 equivocal) than CIM (520 positive with 61 equivocal) overall, in
both HNPC and CRPC patients. DCFBC PET detection of lymph nodes, bone lesions, and
visceral lesions was superior to CIM. When intrapatient clustering effects were taken into
account, DCFBC PET was estimated to be positive in a large proportion of lesions that would be
negative or equivocal on CIM (0.45). On follow-up, the sensitivity of DCFBC PET (0.92) was
superior to CIM (0.71). DCFBC tumor uptake was increased at the later time PET time point (~
2.5 hr post-injection) with background uptake showing a decreasing trend on later PET.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
2
Conclusions: PET imaging with DCFBC, a small molecule PSMA-targeted radiotracer, detects
more lesions than CIM and promised to diagnose and stage patients with metastatic prostate
cancer more accurately than current imaging methods.
Key Words: Prostate-specific membrane antigen, metastatic prostate cancer, positron emission
tomography, computed tomography, bone scan.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
3
INTRODUCTION
Prostate cancer is common, representing the most frequent cancer diagnosis and second
most frequent cause of cancer-related death in men in the United States (1). Many men who
undergo curative therapy for primary prostate cancer will suffer recurrent/metastatic disease.
After a patient demonstrates biochemical recurrence, with newly-appearing or increasing
prostate specific antigen (PSA) blood levels, subsequent treatments such as androgen deprivation
or cytotoxic chemotherapy are often deferred until there has been unequivocal new or
progressive metastatic disease on imaging. That emphasizes the need for imaging of metastatic
prostate cancer to be highly sensitive and specific in order to ensure that patients are treated
appropriately in a timely manner.
Patients suffering biochemical recurrence may be imaged with the conventional imaging
modalities (CIM) of 99mTc-methylene diphosphonate (MDP) bone scan (BS) and contrast-
enhanced CT (CECT) of the chest, abdomen, and pelvis.. There are important limitations to the
sensitivity and specificity of CIM including small (less than 1 cm short axis) lymph nodes that
are not definitively characterized as on CECT; primarily lytic bone lesions that may have little
uptake on BS and be occult on CECT until significant trabecular or cortical destruction has
occurred; and areas of degenerative bone change that are sclerotic on CECT and have high
uptake on BS and that can be mistaken for, or obscure, osteoblastic osseous metastases.
Partly as a result of those limitations, there has been interest in the development of
functional imaging tools for the detection of metastatic prostate cancer. 18F-fluorodeoxyglucose
(FDG) PET/CT, despite widespread use in a variety of cancers, has generally proven to be
problematic in this setting. An array of additional PET radiotracers has been investigated in
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
4
metastatic prostate cancer including those targeting fatty acid metabolism (11C-choline, 18F-
fluorocholine, and 11C-acetate) (2-9) and amino acid transport (anti-1-amino-3-18F-
fluorocyclobutane-1-carboxylic acid, 18F-FACBC) (10-12). Additional radiotracers targeting the
prostate-specific membrane antigen (PSMA) include small-molecule (13-18) and antibody (19-
21) agents . Gastrin releasing peptide (GRP) (22), and glutamine (23, 24) targeted radiotracers
are also being developed.
PSMA is an attractive target for imaging prostate cancer as it is expressed in the vast
majority of prostate cancers and histological studies have associated high PSMA expression with
metastatic spread (25, 26), castration resistance (27-29), and expression levels may be predictive
of progression (30, 31). Our previous work has shown that a radiofluorinated small-molecule
inhibitor of PSMA, N-[N-[(S)-1,3-dicarboxypropyl]carbamoyl]-4-[18F]fluorobenzyl-L-cysteine
(DCFBC, Figure 1), was able to concentrate in PSMA-expressing tumors in pre-clinical studies
(14), to identify sites of metastatic disease clinically (13), and to localize at sites of high-grade
primary prostate cancer (32). For this study, we evaluated the ability of DCFBC PET/CT to
identify sites of bone, lymph node, and visceral soft tissue metastatic disease in comparison to
CIM. The study cohort consisted of both hormone-naïve and castration-resistant metastatic
prostate cancer patients.
MATERIALS AND METHODS
Patient Population and Selection
Our hospital’s Institutional Review Board (IRB) approved this study under the auspices
of a Food and Drug Administration exploratory investigation new drug application (eIND
108943). This clinical trial was registered in ClinicalTrials.gov (Identifier: NCT01815515).
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
5
Written, informed consent was obtained from all participating patients. Inclusion criteria for this
study included histologic confirmation of prostate cancer, radiologic evidence of new or
progressive metastatic disease on anatomic or functional imaging and rising prostate specific
antigen (PSA) serum levels on two observations at least one week apart. Exclusion criteria
included the patient being treated with an investigational drug, biologic, or device within 14 days
of DCFBC administration; iniation of new prostate cancer therapy within 14 days of DCFBC
administration; initiation of new therapy for progressive metastatic disease since radiographic
documentation of progression; serum creatinine or total bilirubin greater than 3 times the upper
limit of normal; or liver transaminases greater than 5 times the upper limit of normal. These
baseline laboratory values were obtained to ensure patients were appropriately healthy enough to
reasonably participate in the study. Patients had CECT of the chest, abdomen, and pelvis (single,
venous phase) and planar bone scan within 28 days of DCFBC PET.
Seventeen patients were prospectively enrolled and imaged with DCFBC PET/CT
between May 2013 and May 2014. Patients were followed up to one year with subsequent
imaging examinations obtained at the discretion of the treating medical oncologists.
Radiochemistry
2-[3-(1-Carboxy-2-mercapto-ethyl)-ureido]-pentanedioic acid was synthesized as
previously detailed (33). Non-radioactive DCFBC was prepared according to a modification of a
published protocol with conformation to current good manufacturing practice (14). DCFBC
(radiolabeled) was prepared according to published protocols (13, 34). Specific activity range of
administered DCFBC was 18,837 ± 7,095 mCi/µmol.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
6
PET/CT Protocol
Patients were asked to remain nil per os (except for water and some medications) for at
least 6 hours prior to the administration of DCFBC. As other investigators have noted the ability
of folate to act as a substrate for PSMA (35-37), we asked that patients not take multivitamins or
folate supplements on the day of DCFBC PET/CT imaging. Blood was drawn and sent for
serum folate, red blood cell folate, and testosterone levels (Table 1).
DCFBC PET/CT images were acquired on a Discovery DRX PET/CT scanner (GE
Healthcare, Waukesha, WI) operating in 3D emission mode with CT-derived attenuation
correction. A bolus injection of 10 ± 1 mCi (370 ± 37 MBq) of DCFBC was administered
intravenously. Two hours post-injection, a whole body (WB, from the top of the skull through
the mid-thighs) CT was obtained [120 kVp, 80 mA maximum (auto-adjusting)] followed by an
initial WB PET acquisition beginning at the mid-thighs with 4 minutes and 15 seconds per bed
position (early time point). Given our earlier experience with DCFBC from the first-in-man
study, we suspected that imaging at a later time point after radiotracer injection might yield
improved tumor uptake and decreased background. Accordingly, immediately following the
initial PET acquisition, a second WB acquisition was obtained, again starting from the mid-
thighs and occurring approximately 2.5 hours post-injection (late time point). PET images were
reconstructed using a clinical ordered subset expectation maximization (OSEM) algorithm.
Image Analysis
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
7
DCFBC PET/CT and BS images were centrally reviewed by 3 expert nuclear medicine
readers (AC, EM, and SYC) who reached a group consensus on the lesions for each scan.
Analyses of the PET/CT and BS images on any one patient were performed at least one week
apart to minimize any bias that might occur in the interpretation of either the PET/CT or BS
based on results from the other study; although the number of patients was relatively small, a
large number of lesions were identified (see results section), decreasing the likelihood that
individual lesions would be recalled and mentally correlated by the central reviewers. CECT
images were centrally reviewed by 2 expert readers (SPR and KJM) who were blinded to the
results of the PET and BS studies and who also reached a group consensus read on each scan.
Visual analysis of DCFBC uptake on the PET/CT scans was performed on a 3-point scale
(1 = negative/below adjacent background, 2 = equivocal/approximately at adjacent background,
and 3 = positive/above adjacent background) on both the early and late time points on a General
Electric Advantage Workstation (GE Healthcare, Waukesha, WI). Maximum standardized
uptake values (SUVmax) corrected for lean body mass were obtained from both time points. For
lesions identified on other modalities that lacked discrete DCFBC uptake, regions of interest
(ROIs) were drawn at corresponding sites on the PET images to derive SUVmax levels for these
lesions. One patient had diffusely infiltrating, biopsy-proven liver metastases that was
interpreted as such by central review of the CECT and was negative (and hence not identified) on
DCFBC PET; this patient’s liver was considered a single lesion for purposes of analysis and
SUVmax was determined from the most confluent focus of disease in the liver. To measure
background, average SUVs (SUVavg) were obtained from ascending aorta blood pool activity,
liver parenchyma in the non-disease-involved right lobe of the liver, within a vertebral body not
involved with disease, and within the right gluteal muscles.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
8
The BS and CECT images were analyzed on our institution’s standard clinical viewing
software, UltraVisual (Emageon, Birmingham, Alabama, USA). For both modalities, lesions
were again classified on a 3-point scale. For lymph nodes on the CECT scans, a short axis
measurement less than 1 cm was considered negative and a short axis measurement greater than
1 cm was considered positive.
During follow-up of these patients, any available imaging was reviewed by the
appropriate central reviewers. Those lesions that demonstrated subjectively determined
progression or response to therapy on the follow-up studies were considered to be true positive
lesions for purposes of calculating sensitivity. Lesions that remained unchanged were
considered equivocal, and sensitivity was calculated with these equivocal lesions grouped with
either the positive or negative lesions in separate analyses. One patient entered hospice and
subsequently died of his metastatic disease after being imaged with DCFBC PET but before any
imaging follow-up could be completed; the nature of his lesions was established in consultation
between the central imaging reviewers and medical oncologists.
Statistical Analysis
Each lesion was classified as positive, negative, or equivocal by DCFBC PET/CT,
CECT, BS, and combined CIM. The proportion of agreement between modalities was estimated
using intercept-only logistic regression models with a generalized estimating equation (GEE)
approach to account for intra-patient correlation of multiple lesions. In addition to an overall
modality-based analysis, the proportion of agreement was also estimated for lesions based on
location (i.e. lymph node, bone, or visceral soft tissue) as well as patient castrate status (hormone
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
9
naïve versus resistant). Sensitivity was calculated based on follow-up imaging findings using the
GEE intercept-only approach described above. Differences in continuously measured
parameters including SUVmax were estimated with linear regression models using GEE.
Analyses were completed with R version 3.1.2 (38).
RESULTS
Study Population Baseline Imaging
Sixteen out of 17 patients met all inclusion criteria; one patient lacked definite evidence
of new or progressive metastatic disease on imaging, but there was a strong clinical suspicion
that he would have detectable disease with DCFBC PET and the IRB granted an exemption.
Selected clinical and demographic data for the 17 imaged patients are included in Table 1. Of
the 17 patients imaged with DCFBC PET/CT, complete contemporaneous CIM (both CECT of
the chest, abdomen, and pelvis as well as planar WB BS) was available for all but 3 patients.
One patient had a history of severe allergy to iodinated contrast and was imaged with a non-
contrast CT. A second patient had a follow-up CECT at an outside institution but we were not
able to obtain this scan for central review. The third patient had an outside BS, but the images
provided could not be obtained in DICOM format for adequate interpretation.
Imaging Findings
In aggregate, between DCFBC PET and CIM, 714 metastatic lesions were detected on at
least one modality (per patient: median 17 lesions, range 4 to 237). Positive DCFBC PET uptake
was observed visually in 592 lesions with 63 additional lesions deemed equivocal. Overall, for
diagnostic CT, 402 lesions were determined to be positive with an additional 41 determined to be
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
10
equivocal. For BS, 303 lesions were positive and 29 were equivocal. In sum total, 520 lesions
were positive with CIM with a further 61 equivocal lesions.
As shown in Figure 2A, the median and range of SUVmax for DCFBC-positive metastatic
lesions demonstrated higher uptake at the later time point (p < 0.001). The measured
background PET SUVavg trended lower on the later PET time point, though again it was not
statistically significant (Figure 2B). When comparing DCFBC PET-positive metastatic lesions
in patients with HNPC and CRPC, we did not observe a statistically significant difference in PET
SUVmax in the lesions from the two patient populations (p = 0.81 for the early time point and p =
0.57 for the late time point). There was no difference in visual detection of metastatic lesions on
early and later time point PET acquisitions, thus lesion positive/negative/equivocal status
between the two acquisitions was unchanged for all detected lesions.
Statistical Analysis
The general estimating equation estimates for lesion detection by modality are detailed in
Table 2 (the actual number of discrete lesions seen on each modality are included in
Supplemental Table 1). DCFBC PET was able to identify more definitive lesions than CIM.
The estimated proportion of all detected metastatic lesions that would be positive with DCFBC
PET but negative or equivocal with CIM is 0.44 (95% confidence interval (CI) 0.28 – 0.61).
The estimated proportion of lesions that would be positive on CIM but negative or equivocal on
DCFBC PET were 0.08 (95% CI 0.04 – 0.16). The estimated proportions for different types of
metastatic sites are detailed in Table 2.
Despite the concern that high folate levels (defined in our hospital laboratory as >24
ng/mL serum folate) could potentially interfere with DCFBC uptake in cells expressing PSMA,
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
11
the range of number of lesions detected in patients with high folate was similar to the range in
patients with normal folate levels (range 16 – 172 in patients with high folate versus 4 – 237 in
patients with normal folate) with a higher median number of lesions in patients with high folate
(47 in patients with high folate versus 13.5 in patients with normal folate).
Of the original 17 patients recruited, 12 had adequate imaging follow-up to assess for
progression, response, or stability of the lesions originally identified. This follow-up was
generally with conventional imaging only, although a single patient did have a follow-up
research PET scan with a PSMA-targeted radiotracer. Central review of the follow-up imaging
was performed with individual lesions subjectively determined as progressing/responding to
therapy (true lesions) or remaining unchanged (equivocal). Table 3 details the available imaging
and time to follow-up for each patient as well as the intercurrent therapy each received.
Maximum time to follow-up was one year (median time to follow-up was 4 months with range
from 1 month to 1 year). The estimates for sensitivity of DCFBC PET for true metastatic
lesions, with equivocal lesions considered negative for metastasis, was 0.92 (95% CI 0.80 –
0.97) as compared to a sensitivity 0.64 (95% CI 0.41 – 0.82) for CECT, 0.40 (95% CI 0.20 –
0.65) for BS, and 0.71 (95% CI 0.49 – 0.86) for combined CIM (Table 4).
Pertinent examples of imaging findings with DCFBC are shown in Figures 3 – 6 and
Supplemental Figure 1, as detailed in the accompanying figure legends.
DISCUSSION
As noted in the introduction, significant progress has been made in the development of
PET radiotracers for molecular imaging of metastatic prostate cancer, many of which have
demonstrated promise for improving detection relative to CIM. We have presented prospective,
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
12
systematic evidence of the superior sensitivityof the small-molecule PSMA inhibitor DCFBC for
detecting lesions in metastatic prostate cancer patients.
Of particular importance, patients with either HNPC or CRPC were reliably imaged with
DCFBC PET with no statistically significant difference in the observed SUVmax ranges for
metastatic lesions. Given previously published data that had suggested increased PSMA
expression with low androgen signaling, it was of concern that lesions in CRPC patients might
have shown low uptake of a PSMA-targeted radiotracer. Recent clinical data from 68Ga-labeled
PSMA PET radiotracers has also demonstrated that metastatic lesions in CRPC express enough
PSMA to be reliably detected (15, 16).
It is noteworthy that DCFBC PET was capable of showing definitive focal radiotracer
uptake at sites of involvement that are often problematic in the interpretation of conventional
imaging. Sclerotic lesions in the spine on CECT with corresponding MDP uptake on BS may be
interpreted as indeterminate for metastatic involvement versus degenerative change (Figure 3).
Predominantly lytic or mixed bone lesions that can be subtle or are not visualized on CIM can
also be well visualized on DCFBC PET (Figure 4). Furthermore, lymph node metastases that are
too small to definitively identify with CIM can show focal DCFBC uptake (Figure 5).
Analysis of uptake at the early versus late time points suggests that the late time point
produced both improved tumor uptake and decreased background distribution of DCFBC. It is
possible that even later imaging may further improve image quality, although the relatively high
degree of activity within blood pool for DCFBC is likely to persist to at least some degree. The
late time point in this study (approximately 2.5 – 3 hours post injection) is likely to represent a
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
13
suitable compromise between optimizing image quality while preserving reasonable clinical
work-flow.
Potential limitations of DCFBC PET/CT became apparent over the course of this study.
A small number of densely sclerotic bone lesions were much more apparent on CIM (Figure 6).
Although the dense sclerosis and high MDP uptake are indicative of significant bony reaction to
the presence of tumor cells, it may be that these lesions have relatively few metastatic prostate
cancer cells and therefore a diminished ability to sequester PSMA-targeted radiotracers. A
second potential pitfall we observed with DCFBC was in the context of liver metastases, which
were not well seen (Supplemental Figure 1).The reason for this was not immediately apparent,
although we suspect that while metastases are PSMA-avid, signal from such lesions is
overwhelmed by background.
At the initiation of the study, a small minority of patients could not or did not receive
complete baseline CIM, preventing the most complete possible analysis. An additional
significant limitation of this study was the lack of histopathologic truth standard; although we
have attempted to mitigate this by using the surrogate of response to therapy/disease progression
on follow-up imaging to assess for true lesions, this approach remains limited in that lesions
identified on DCFBC PET imaging were necessarily compared to follow-up conventional
imaging. It can also be noted that BS imaging in this study was only planar and tomographic
bone scintigraphy and/or Na18F PET would likely have detected more bone lesions, potentially
narrowing the sensitivity difference between DCFBC PET and CIM.
We recently conducted an initial clinical study with a second generation 18F-labeled
PSMA ligand, 2-(3-(1-carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl)-ureido)-
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
14
pentanedioic acid (DCFPyL), a chemically and mechanistically similar compound to DCFBC but
with higher binding affinity for PSMA and lower activity within blood pool (39). We expect
DCFPyL, as well as additional refinements to other PSMA-binding radiotracers, to address some
of the limitations we have observed. There have been promising results as well for Gallium-68
PSMA radiotracers (15, 16), with advantages easier radiochemistry inherent in a generator
produced Gallium-68 without need for a cyclotron and potential integration to theranostic
applications. We favor the use of Fluorine-18 PSMA agents however, due to ease of distribution
utilizing pre-existing networks for 18F-FDG and improved spatial resolution and more accurate
quantitation inherent in the shorter positron range and higher positron yield of Fluorine-18 versus
Gallium-68 (40). Nonetheless, the systematic prospective evaluation of DCFBC presented here
indicates the promise of PET imaging of PSMA in general as a means to improve detection of
metastatic prostate cancer.
CONCLUSION
PSMA-based PET/CT imaging with DCFBC can detect more metastatic prostate cancer
lesions than the current standard of clinical imaging with CECT and BS in patients with either
HNPC or CRPC. PSMA-targeted imaging offers promise in more accurate identification of the
presence and extent of metastatic prostate cancer.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
15
DISCLOSURES
None.
ACKNOWLEDGMENTS
We acknowledge funding from the Prostate Cancer Foundation – Young Investigator
Award, RSNA Research & Education Foundation – Research Scholar Award, EB006351,
CA184228, CA183031, and CA134675. We thank Akimosa Jeffrey-Kwanisai and Yavette
Morton for providing dedicated clinical coordination for this trial.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
16
REFERENCES
1. Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin. 2014;64:9-29.
2. Bauman G, Belhocine T, Kovacs M, Ward A, Beheshti M, Rachinsky I. 18F-fluorocholine for
prostate cancer imaging: a systematic review of the literature. Prostate Cancer Prostatic Dis.
2012;15:45-55.
3. Beheshti M, Treglia G, Zakavi SR, et al. Application of 11C-acetate positron-emission tomography
(PET) imaging in prostate cancer: systematic review and meta-analysis of the literature. British Journal of
Urology International. 2013;13 AUG 2013.
4. Evangelista L, Guttilla A, Zattoni F, Muzzio PC. Utility of choline positron emission
tomography/computed tomography for lymph node involvement identification in intermediate- to high-
risk prostate cancer: a systematic literature review and meta-analysis. Eur Urol. 2013;63:1040-1048.
5. Evangelista L, Zattoni F, Guttilla A, Saladini G, Colletti PM, Rubello D. Choline PET or PET/CT and
biochemical relapse of prostate cancer: a systematic review and meta-analysis. Clin Nucl Med.
2013;38:305-314.
6. Fuccio C, Rubello D, Castellucci P, Marzola MC, Fanti S. Choline PET/CT for prostate cancer: main
clinical applications. Eur J Radiol. 2011;80:e50-56.
7. Jadvar H. Prostate cancer: PET with 18F-FDG, 18F- or 11C-acetate, and 18F- or 11C-choline. J
Nucl Med. 2011;52:81-89.
8. Mertens K, Slaets D, Lambert B, Acou M, De Vos F, Goethals I. PET with (18)F-labelled choline-
based tracers for tumour imaging: a review of the literature. Eur J Nucl Med Mol Imaging. 2010;37:2188-
2193.
9. Umbehr MH, Muntener M, Hany T, Sulser T, Bachmann LM. The role of 11C-choline and 18F-
fluorocholine positron emission tomography (PET) and PET/CT in prostate cancer: a systematic review
and meta-analysis. Eur Urol. 2013;64:106-117.
10. Schuster DM, Votaw JR, Nieh PT, et al. Initial experience with the radiotracer anti-1-amino-3-
18F-fluorocyclobutane-1-carboxylic acid with PET/CT in prostate carcinoma. J Nucl Med. 2007;48:56-63.
11. Schuster DM, Savir-Baruch B, Nieh PT, et al. Detection of recurrent prostate carcinoma with anti-
1-amino-3-18F-fluorocyclobutane-1-carboxylic acid PET/CT and 111In-capromab pendetide SPECT/CT.
Radiology. 2011;259:852-861.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
17
12. Mena E, Turkbey B, Mani H, et al. 11C-Acetate PET/CT in localized prostate cancer: a study with
MRI and histopathologic correlation. J Nucl Med. 2012;53:538-545.
13. Cho SY, Gage KL, Mease RC, et al. Biodistribution, Tumor Detection, and Radiation Dosimetry of
18F-DCFBC, a Low-Molecular-Weight Inhibitor of Prostate-Specific Membrane Antigen, in Patients with
Metastatic Prostate Cancer. J Nucl Med. 2012;53:1883-1891.
14. Mease RC, Dusich CL, Foss CA, et al. N-[N-[(S)-1,3-Dicarboxypropyl]carbamoyl]-4-
[18F]fluorobenzyl-L-cysteine, [18F]DCFBC: a new imaging probe for prostate cancer. Clin Cancer Res.
2008;14:3036-3043.
15. Afshar-Oromieh A, Malcher A, Eder M, et al. PET imaging with a [68Ga]gallium-labelled PSMA
ligand for the diagnosis of prostate cancer: biodistribution in humans and first evaluation of tumour
lesions. Eur J Nucl Med Mol Imaging. 2013;40:486-495.
16. Afshar-Oromieh A, Avtzi E, Giesel FL, 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.
17. Barrett JA, Coleman RE, Goldsmith SJ, et al. First-in-man evaluation of 2 high-affinity PSMA-avid
small molecules for imaging prostate cancer. J Nucl Med. 2013;54:380-387.
18. Vallabhajosula S, Nikolopoulou A, Babich JW, et al. 99mTc-labeled small-molecule inhibitors of
prostate-specific membrane antigen: pharmacokinetics and biodistribution studies in healthy subjects
and patients with metastatic prostate cancer. J Nucl Med. 2014;55:1791-1798.
19. Tagawa ST, Milowsky MI, Morris M, et al. Phase II study of Lutetium-177-labeled anti-prostate-
specific membrane antigen monoclonal antibody J591 for metastatic castration-resistant prostate
cancer. Clin Cancer Res. 2013;19:5182-5191.
20. Viola-Villegas NT, Sevak KK, Carlin SD, et al. Noninvasive Imaging of PSMA in Prostate Tumors
with (89)Zr-Labeled huJ591 Engineered Antibody Fragments: The Faster Alternatives. Mol Pharm.
2014;11:3965-3973.
21. Osborne JR, Green DA, Spratt DE, et al. A prospective pilot study of (89)Zr-J591/prostate specific
membrane antigen positron emission tomography in men with localized prostate cancer undergoing
radical prostatectomy. J Urol. 2014;191:1439-1445.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
18
22. Wieser G, Mansi R, Grosu AL, et al. Positron emission tomography (PET) imaging of prostate
cancer with a gastrin releasing peptide receptor antagonist--from mice to men. Theranostics.
2014;4:412-419.
23. Lieberman BP, Ploessl K, Wang L, et al. PET imaging of glutaminolysis in tumors by 18F-(2S,4R)4-
fluoroglutamine. J Nucl Med. 2011;52:1947-1955.
24. Venneti S, Dunphy MP, Zhang H, et al. Glutamine-based PET imaging facilitates enhanced
metabolic evaluation of gliomas in vivo. Sci Transl Med. 2015;7:274ra217.
25. Sweat SD, Pacelli A, Murphy GP, Bostwick DG. Prostate-specific membrane antigen expression is
greatest in prostate adenocarcinoma and lymph node metastases. Urology. 1998;52:637-640.
26. Chang SS, Reuter VE, Heston WD, Gaudin PB. Comparison of anti-prostate-specific membrane
antigen antibodies and other immunomarkers in metastatic prostate carcinoma. Urology. 2001;57:1179-
1183.
27. Wright GL, Jr., Grob BM, Haley C, et al. Upregulation of prostate-specific membrane antigen
after androgen-deprivation therapy. Urology. 1996;48:326-334.
28. Evans MJ, Smith-Jones PM, Wongvipat J, et al. Noninvasive measurement of androgen receptor
signaling with a positron-emitting radiopharmaceutical that targets prostate-specific membrane antigen.
Proc Natl Acad Sci U S A. 2011;108:9578-9582.
29. Noss KR, Wolfe SA, Grimes SR. Upregulation of prostate specific membrane antigen/folate
hydrolase transcription by an enhancer. Gene. 2002;285:247-256.
30. Perner S, Hofer MD, Kim R, et al. Prostate-specific membrane antigen expression as a predictor
of prostate cancer progression. Hum Pathol. 2007;38:696-701.
31. Ross JS, Sheehan CE, Fisher HA, et al. Correlation of primary tumor prostate-specific membrane
antigen expression with disease recurrence in prostate cancer. Clin Cancer Res. 2003;9:6357-6362.
32. Rowe SP, Gage KL, Faraj SF, et al. 18F-DCFBC PET/CT for PSMA-Based Detection and
Characterization of Primary Prostate Cancer. J Nucl Med. 2015;56:1003-1010.
33. Kozikowski AP, Nan F, Conti P, et al. Design of remarkably simple, yet potent urea-based
inhibitors of glutamate carboxypeptidase II (NAALADase). J Med Chem. 2001;44:298-301.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
19
34. Holt DP RH, Mathews WB, Horti A, Mease R, Dannals RF. A semi-automated microwave
chemistry system for complete radiosynthesis, purification, and formulation of F-18 radiotracers. J Label
Compd Radiopharm. 2011;54.
35. Foss CA, Mease RC, Cho SY, Kim HJ, Pomper MG. GCPII imaging and cancer. Curr Med Chem.
2012;19:1346-1359.
36. Yao V, Berkman CE, Choi JK, O'Keefe DS, Bacich DJ. Expression of prostate-specific membrane
antigen (PSMA), increases cell folate uptake and proliferation and suggests a novel role for PSMA in the
uptake of the non-polyglutamated folate, folic acid. Prostate. 2010;70:305-316.
37. Yao V, Parwani A, Maier C, Heston WD, Bacich DJ. Moderate expression of prostate-specific
membrane antigen, a tissue differentiation antigen and folate hydrolase, facilitates prostate
carcinogenesis. Cancer Res. 2008;68:9070-9077.
38. Team RC, ed. R: A language and environment for statistical computing. Vienna, Austria.; 2014.
39. Szabo Z, Mena E, Rowe SP, et al. Initial Evaluation of [F]DCFPyL for Prostate-Specific Membrane
Antigen (PSMA)-Targeted PET Imaging of Prostate Cancer. Mol Imaging Biol. 2015.
40. Sanchez-Crespo A. Comparison of Gallium-68 and Fluorine-18 imaging characteristics in positron
emission tomography. Appl Radiat Isot. 2013;76:55-62.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
20
Figure 1. Chemical structure of DCBFC, a first in class radiofluorinated inhibitor of PSMA.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
21
Figure 2. Box plot of SUVmax for DCFBC PET positive metastatic lesions by location and
patient’s androgen-resistant status (A), and boxplot of SUVavg for various regions of background
physiologic uptake (B).
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
22
Figure 3. Anterior projection planar BS (A), DCFBC PET MIP (B), axial CT (C), and axial
DCFBC PET/CT fusion (D) images from a patient thought to have degenerative arthritic changes
at a site of MDP uptake on bone scan (black arrowhead in (A)). However, intense focal DCFBC
uptake was also noted at this site that progressed on follow-up corresponding to a rise in PSA
(black and white arrowheads in B to D).
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
23
Figure 4. DCFBC PET MIP (A), axial CECT (B) and axial fused DCFBC PET/CT (D) images
from a patient with a subtle lytic bone lesion on CT that corresponded to intense DCFBC uptake
in the right posterolateral T5 vertebral body and progressed on follow-up as patient’s PSA level
continued to rise (black and white arrowheads in A to D).
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
24
Figure 5. DCFBC PET MIP (A), axial CECT (B) and axial fused DCFBC PET/CT (C) images
demonstrating intense DCFBC uptake in multiple small pelvic lymph nodes that had been
deemed too small to be definitively disease involved on CECT (black and white arrowheads in A
to D). The lymph nodes decreased in size on follow-up imaging and correlated with a fall in
patient’s PSA level to undetectable.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
25
Figure 6. Posterior projection planar BS (A), DCFBC PET MIP (posterior view, B), axial CT
(C), axial DCFBC PET (D), and axial fused DCFBC PET/CT (E) images from a patient who was
post-prostatectomy with rising PSA and was naïve to systemic ADT and chemotherapy. Imaging
demonstrates intense MDP uptake on BS and corresponding dense sclerosis on CT of the right
scapula without significant DCFBC uptake (black and white arrowheads in A to E). This lesion
progressed in extent to involve more of the scapula on follow-up imaging in correlation with
rising PSA level in this patient.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
26
Table 1.
Patient
Number
Age PSA
(ng/mL)
Serum
folate
(ng/mL;
normal
2.5-20)
Red cell
Folate
(ng/mL;
normal
160-855)
Testosterone
(ng/dL)
Prior Prostate Cancer Therapy
1 72 81.8 >24 478 <20 Prostatectomy, external beam
radiation to the pelvis, androgen
deprivation
2 83 11.6 14.6 429 245 External beam radiation to the pelvis
3 61 38.9 >24 559 <20 External beam radiation to the
pelvis, androgen deprivation
4 70 8.3 12 268 247 Prostatectomy
5 68 67.6 13.6 361 465 None
6 76 31.4 >24 627 <20 Prostatectomy, androgen
deprivation
7 59 99.6 >24 433 <20 Androgen deprivation, docetaxel,
external beam radiation to the spine
8 69 95.8 18.1 486 <20 External beam radiation to the
pelvis, androgen deprivation
9 69 6.7 17.1 359 <20 Androgen deprivation
10 61 48.3 16.6 N/A 331 Prostatectomy, external beam
radiation to the pelvis
11 73 98.8 >24 N/A 280 Prostatectomy
12 62 564.5 11.8 N/A <20 Androgen deprivation, docetaxel,
tasquinimod, external beam
radiation to the right hip, radium-
223
13 55 62.1 10.3 N/A 442 Prostatectomy
14 58 3.5 11.8 584 273 Prostatectomy, external beam
radiation to the pelvis
15 77 316.1 14.2 860 <20 Androgen deprivation, abiraterone
16 75 6.7 10.4 666 746 Prostate brachytherapy
17 75 83.6 22 1049 538 External beam radiation to the pelvis
Table 1. Selected clinical and demographic data on the patients imaged in this study.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
1
Table 2.
All patients HNPC patients CRPC patients
Modality All
lesions
Lymph
node
lesions
Bone
lesions
Visceral
lesions
All
lesions
Lymph
node
lesions
Bone
lesions
Visceral
lesions
All
lesions
Lymph
node
lesions
Bone
lesions
Visceral
lesions
PET CT BS
Pos Neg/Eq _ 0.30
(0.17 -
0.48)
0.39
(0.21 -
0.62)
0.24
(0.11 -
0.46)
0.18
(0.06 -
0.42)
0.40
(0.20 -
0.65)
0.33
(0.08 -
0.73)
0.34
(0.12 -
0.67)
0.23
(0.07 -
0.56)
0.22
(0.10 -
0.42)
0.50
(0.45 -
0.54)
0.16
(0.05 -
0.42)
0.12
(0.02 -
0.49)
Pos _ Neg/Eq
0.44
(0. 28 -
0.61)
N/A 0.22
(0.12 -
0.36)
N/A 0.55
(0.32 -
0.76)
N/A 0.28
(0.12 -
0.52)
N/A 0.31
(0.14 -
0.57)
N/A 0.18
(0.07 -
0.38)
N/A
Pos Neg/Eq*
0.44
(0.28 -
0.61)
0.90
(0.75 -
0.96)
0.22
(0.12 -
0.36)
0.41
(0.17 -
0.69)
0.55
(0.32 -
0.76)
0.84
(0.44 -
0.97)
0.28
(0.12 -
0.52)
0.39
(0.11 -
0.77)
0.31
(0.14 -
0.57)
0.93
(0.83 -
0.97)
0.18
(0.07 -
0.38)
0.42
(0.12 -
0.80)
Neg/Eq Pos _ 0.07
(0.04 -
0.14)
0.07
(0.01 -
0.39)
0.09
(0.05 -
0.17)
0.05
(0.01 -
0.28)
0.06
(0.01 -
0.24)
0.17
(0.02 -
0.63)
0.07
(0.02 -
0.21)
0.00
(0.00 -
0.00)
0.08
(0.04 -
0.16)
0.00
(0.00 -
0.00)
0.10
(0.04 -
0.21)
0.08
(0.01 -
0.46)
Neg/Eq _ Pos 0.03
(0.01 -
0.08)
N/A 0.05
(0.02 -
0.12)
N/A 0.03
(0.01 -
0.19)
N/A 0.06
(0.01 -
0.29)
N/A 0.03
(0.01 -
0.08)
N/A 0.04
(0.02 -
0.11)
N/A
Neg/Eq Pos*
0.08
(0.04 -
0.16)
0.07
(0.01 -
0.39)
0.10
(0.06 -
0.18)
0.05
(0.01 -
0.28)
0.07
(0.01 -
0.27)
0.17
(0.02 -
0.63)
0.08
(0.02 -
0.27)
0.00
(0.00 -
0.00)
0.09
(0.05 -
0.17)
0.00
(0.00 -
0.00)
0.11
(0.06 -
0.21)
0.08
(0.01 -
0.46)
* Combined CIM (CT and BS)
Table 2. Estimated proportion of agreement in metastatic lesions detection between the PET and CIM, accounting for intrapatient
clustering effects by GEE regression model analysis. 95% confidence intervals are included in parentheses.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
1
Table 3.
Patient Number Therapy following
DCFBC PET
Time to imaging
follow-up
Follow-up imaging
modalities available
1 Started sipuleucel-T 6 months Na18F PET/CT
2 Started androgen
deprivation
4 months BS
3 Continued androgen
deprivation
N/A N/A
4 Started androgen
deprivation
2 months CECT, BS
5 Started androgen
deprivation
6 months CECT, BS
6 Started cabazitaxel 4 months CECT, BS
7 Entered hospice N/A N/A
8 Continued androgen
deprivation
N/A N/A
9 External beam
radiation to the
pelvis, continued
androgen deprivation
4 months CECT, BS
10 Started androgen
deprivation
N/A N/A
11 Started androgen
deprivation
6 months CECT, BS
12 Continued radium-
223
3 months CECT, BS
13 Started androgen
deprivation
1 year PSMA PET/CT
14 External beam
radiation to pelvic
lymph node, started
nelfinavir
3 months CECT
15 Continued androgen
deprivation, started
veliparib
3 months CECT, BS
16 No follow-up
information available
N/A N/A
17 Started androgen
deprivation
1 month CECT
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
2
Table 3. List of prostate cancer therapies received by the patients in this study in the follow-up
period after DCFBC PET imaging. The time to follow-up and available modalities at follow-up
are also noted. Patient 13 underwent follow-up PET/CT imaging with a different PSMA targeted
radiotracer (18F-DCFPyL) than the one primarily described here.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
3
Table 4.
Modality Sensitivity
[Equivocal Lesions
Considered Negative]
(95% CI)
Sensitivity
[Equivocal Lesions
Considered Positive]
(95% CI)
DCFBC PET 0.92 (0.80 – 0.97) 0.88 (0.70 – 0.96)
CECT 0.64 (0.41 – 0.82) 0.77 (0.58 – 0.89)
BS 0.40 (0.20 – 0.65) 0.43 (0.25 – 0.63)
CIM
(BS and CECT)
0.71 (0.49 – 0.86) 0.82 (0.60 – 0.93)
Table 4. Sensitivity, with equivocal lesions considered either positive or negative for metastases
in two separate analyses, for DCFBC PET and CIM as estimated by GEE regression model
analysis. The 95% confidence intervals are included in parentheses.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
Doi: 10.2967/jnumed.115.163782
Published online: October 22, 2015.
J Nucl Med.
C Mease, Martin G. Pomper and Steve Y Cho
Antonarakis, Mario Eisenberger, Michael Carducci, Ashley Ross, Philip Kantoff, Daniel P Holt, Robert F Dannals, Ronnie
Steven P. Rowe, Katarzyna J Macura, Anthony Ciarallo, Esther Mena, Amanda Blackford, Rosa Nadal, Emmanuel
Cancer
for Detection of Hormone-Sensitive and Castration-Resistant Metastatic Prostate
F-DCFBC PET/CT to Conventional Imaging Modalities
18
Comparison of PSMA-based
http://jnm.snmjournals.org/content/early/2015/10/21/jnumed.115.163782
This article and updated information are available at:
http://jnm.snmjournals.org/site/subscriptions/online.xhtml
Information about subscriptions to JNM can be found at:
http://jnm.snmjournals.org/site/misc/permission.xhtml
Information about reproducing figures, tables, or other portions of this article can be found online at:
and the final, published version.
proofreading, and author review. This process may lead to differences between the accepted version of the manuscript
ahead of print area, they will be prepared for print and online publication, which includes copyediting, typesetting,JNM
copyedited, nor have they appeared in a print or online issue of the journal. Once the accepted manuscripts appear in the
. They have not beenJNM ahead of print articles have been peer reviewed and accepted for publication in JNM
(Print ISSN: 0161-5505, Online ISSN: 2159-662X)
1850 Samuel Morse Drive, Reston, VA 20190.
SNMMI | Society of Nuclear Medicine and Molecular Imaging
is published monthly.The Journal of Nuclear Medicine
© Copyright 2015 SNMMI; all rights reserved.
by on February 21, 2017. For personal use only. jnm.snmjournals.org Downloaded from
... [54] In one study, PSMA PET detected sites of recurrence in 44% of patients with a negative choline PET. [54] Novel [55,56] 18 F-DCFBC was shown to have an improved sensitivity (90%) compared to CT scan (64%), Tc99 mMDP bone scan (40%), and CT + bone scan combined (71%) in patients with known metastatic disease in both hormone sensitive and castrate resistant settings. [55] The study was, however, limited as there was no histopathologic confirmation, only confirmation based on lesion response to treatment. ...
... [54] Novel [55,56] 18 F-DCFBC was shown to have an improved sensitivity (90%) compared to CT scan (64%), Tc99 mMDP bone scan (40%), and CT + bone scan combined (71%) in patients with known metastatic disease in both hormone sensitive and castrate resistant settings. [55] The study was, however, limited as there was no histopathologic confirmation, only confirmation based on lesion response to treatment. [55] In a direct comparison study involving 14 patients with recurrent prostate cancer, staging shown by 18 F-DCFPyL was equivalent to that with 68 Ga-PSMA PET. ...
... [55] The study was, however, limited as there was no histopathologic confirmation, only confirmation based on lesion response to treatment. [55] In a direct comparison study involving 14 patients with recurrent prostate cancer, staging shown by 18 F-DCFPyL was equivalent to that with 68 Ga-PSMA PET. [57] A study in 2017 showed 18 F-DCFPyL to be superior to conventional imaging methods, detecting 131 sites of cancer in patients with known metastatic disease, compared to only 45 sites detected on x-ray, bone scan, and CT. ...
Article
Full-text available
Accurate detection of metastatic prostate cancer in the setting of preoperative staging as well as posttreatment recurrence is crucial to provide patients with appropriate and timely treatment of their disease. This has traditionally been accomplished with a combination of computed tomography, magnetic resonance imaging, and bone scan. Recently, more novel imaging techniques have been developed to help improve the detection of advanced and metastatic prostate cancer. This review discusses the efficacy of the traditional imaging modalities as well as the novel imaging techniques in detecting metastatic prostate cancer. Articles discussed were gathered through a formal PubMed search.
... Many clinical trials using PSMA-targeted small molecules or nanomaterials for the treatment of prostate cancer have yielded promising results. [7][8][9] PSMA, a protein with folate substrate hydrolase enzymatic activity, cleaves the amide bond of N-acetylaspartate and hydrolyzes extracellular polyglutamic acid to monoglutamic acid folate, which can be used by the cell. 10 Folate-coupled nanoparticles may be a promising PSMA-targeting nanocarrier as reported by several studies. ...
Article
Full-text available
Background Prostate cancer (PCa) ranks second in the incidence of all malignancies in male worldwide. The presence of multi-organ metastases and tumor heterogeneity often leads to unsatisfactory outcomes of conventional radiotherapy treatments. This study aimed to develop a novel folate-targeted nanohydroxyapatite (nHA) coupling to deliver adriamycin (Doxorubicin, DOX), ³²P, and 99mTc simultaneously for the diagnosis and treatment of prostate-specific membrane antigen (PSMA) positive prostate cancer. Methods The spherical nHA was prepared by the biomimetic method and characterized. Folic acid (FA) was coupled to nHA with polyethylene glycol (PEG), and the grafting ratio of PEG-nHA and FA-PEG-nHA was determined by the thermogravimetric analysis (TGA) method. In addition, ³²P, 99mTc, and DOX were loaded on nHA by physisorption. And the labeling rate and stability of radionuclides were measured by a γ-counter. The loading and release of DOX at different pH were determined by the dialysis method. Targeting of FA-PEG-nHA loaded with 99mTc was verified by in vivo SPECT imaging. In vitro anti-tumor effect of ³²P/DOX-FA-PEG-nHA was assessed with apoptosis assay. The safety of the nano-drugs was verified by histopathological analysis. Results The SEM images showed that the synthesized nHA was spherical with uniform particle size (average diameter of about 100nm). The grafting ratio is about 10% for PEG and about 20% for FA. The drug loading and the delayed release of DOX at different pH confirmed its long-term therapeutic ability. The labeling of ³²P and 99mTc was stable and the labeling rate was great. SPECT showed that FA-PEG-nHA showed well in vivo tumor targeting and less damage to normal tissues. Conclusion FA-targeted nHA loaded with ³²P, 99mTc, and DOX may be a new diagnostic and therapeutic strategy for targeting PSMA-positive prostate cancer tumors, which may achieve better therapeutic results while circumventing the severe toxic side effects of conventional chemotherapeutic agents.
... A 27% greater accuracy of the PSMA-PET/CT was found in the Phase 3 clinical trial by Hofman et al. when compared with the conventional imaging techniques, which translated into modulations in the management (Hofman et al., 2020). At present, there is also solid evidence of the restaging efficacy of PSMA-PET/CT in men with CRPC (Pandit-Taskar et al., 2015;Pyka et al., 2016;Rowe et al., 2016). Figure 2 showed the imagings of a 56-year-old patient with an initial diagnosis as nmCRPC according to the results from CIMs, for whom PSMA-PET detected evidence of distant metastatic lesions on the left fifth rib. ...
Article
Full-text available
Castration-resistant prostate cancer (CRPC) is a therapy-resistant and lethal form of prostate cancer as well as a therapeutic challenge. Prostate-specific membrane antigen (PSMA) has been proved as a promising molecular target for optimizing the theranostics for CRPC patients. When combined with PSMA radiotracers, novel molecular imaging techniques such as positron emission tomography (PET) can provide more accurate and expedient identification of metastases when compared with conventional imaging techniques. Based on the PSMA-based PET scans, the accurate visualization of local and disseminative lesions may help in metastasis-directed therapy. Moreover, the combination of 68Ga-labeled PSMA-based PET imaging and radiotherapy using PSMA radioligand therapy (RLT) becomes a novel treatment option for CRPC patients. The existing studies have demonstrated this therapeutic strategy as an effective and well-tolerated therapy among CRPC patients. PSMA-based PET imaging can accurately detect CRPC lesions and describe their molecular features with quantitative parameters, which can be used to select the best choice of treatments, monitor the response, and predict the outcome of RLT. This review discussed the current and potential role of PSMA‐based imaging and RLT in the diagnosis, treatment, and prediction of prognosis of CRPC.
... The lack of sensitivity of conventional imaging (5) has limited the understanding of disease spread in BCR. With the advent of positron emitting (PET) probes targeting prostate-specific membrane antigen (PSMA), molecular imaging has yielded new insights into PCa recurrence (6), with improved sensitivity and specificity that far exceeds conventional imaging and earlier types of PET agents (7). 18 F-DCFPyL is a recent U.S Food and Drug Administration (FDA)-approved PSMA-PET agent with high affinity for PCa (8,9). ...
Article
Our objective was to investigate the factors predicting scan positivity and disease location in patients with biochemical recurrence (BCR) of prostate cancer (PCa) after primary local therapy using prostate-specific membrane antigen-targeted 18F-DCFPyL PET/CT. Methods: This was a 2-institution study including 245 BCR PCa patients after primary local therapy and negative results on conventional imaging. The patients underwent 18F-DCFPyL PET/CT. We tested for correlations of lesion detection rate and disease location with tumor characteristics, time from initial therapy, prostate-specific antigen (PSA) level, and PSA doubling time (PSAdt). Multivariate logistic regression analyses were used to determine predictors of a positive scan. Regression-based coefficients were used to develop nomograms predicting scan positivity and extrapelvic disease. Results: Overall, 79.2% (194/245) of patients had a positive 18F-DCFPyL PET/CT result, with detection rates of 48.2% (27/56), 74.3% (26/35), 84% (37/44), 96.7% (59/61), and 91.8% (45/49) for PSAs of <0.5, 0.5 to <1.0, 1.0 to <2.0, 2.0 to <5.0, and ≥5.0 ng/mL, respectively. Patients with lesions confined to the pelvis had lower PSAs than those with distant sites (1.6 ± 3.5 vs. 3.0 ± 6.3 ng/mL, P < 0.001). In patients treated with prostatectomy (n = 195), 24.1% (47/195) had a negative scan result, 46.1% (90/195) showed intrapelvic disease, and 29.7% (58/195) showed extrapelvic disease. In the postradiation subgroup (n = 50), 18F-DCFPyL PET/CT was always negative at a PSA lower than 1.0 ng/mL and extrapelvic disease was seen only when PSA was greater than 2.0 ng/mL. At multivariate analysis, PSA and PSAdt were independent predictive factors of scan positivity and the presence of extrapelvic disease in postsurgical patients, with area under the curve of 78% and 76%, respectively. PSA and PSAdt were independent predictors of the presence of extrapelvic disease in the postradiation cohort, with area under the curve of 85%. Time from treatment to scan was significantly longer for prostatectomy-bed-only recurrences than for those with bone or visceral disease (6.2 ± 6.4 vs. 2.4 ± 1.3 y, P < 0.001). Conclusion: 18F-DCFPyL PET/CT offers high detection rates in BCR PCa patients. PSA and PSAdt are able to predict scan positivity and disease location. Furthermore, the presence of bone or visceral lesions is associated with shorter intervals from treatment than are prostate-bed-only recurrences. These tools might guide clinicians to select the most suitable candidates for 18F-DCFPyL PET/CT imaging.
Article
Full-text available
Purpose This study aimed to compare the diagnostic efficiency of ¹⁸ F-DCFPyL PET/CT imaging and 99m Tc-MDP SPECT/CT bone imaging for the detection of bone metastases in prostate cancer. Methods A retrospective analysis was conducted on 31 patients with confirmed prostate cancer between September 2020 and September 2022 at China-Japan Union Hospital of Jilin University. All patients underwent ¹⁸ F-DCFPyL PET/CT and 99m Tc-MDP SPECT/CT bone imaging. The gold standard was the pathology or Best Valuable Comparator (BVC) result based on clinical follow-up. Diagnostic performance indicators, including sensitivity, specificity, accuracy, positive predictive value (PPV), and negative predictive value (NPV), were analyzed at both the patient and lesion levels. The paired sample chi-square test was used to compare the two imaging methods. Receiver operating characteristic (ROC) curves were plotted, and the area under the curve (AUC) was calculated for each method. The AUC values were compared using the Z -test, and a p -value < 0.05 was considered statistically significant. Results Of the 31 prostate cancer patients, 18 were diagnosed with bone metastases, with a total of 84 bone metastatic lesions. At the patient level, ¹⁸ F-DCFPyL PET/CT imaging showed superior diagnostic performance compared to 99m Tc-MDP SPECT/CT bone imaging in all indicators: sensitivity (100% vs. 77.8%, p < 0.01), specificity (92.3% vs. 69.2%, p < 0.05), accuracy (96.8% vs. 74.2%, p < 0.01), PPV (94.7% vs. 77.8%, p < 0.01), and NPV (100% vs. 69.2%, p < 0.01). The AUC values for ¹⁸ F-DCFPyL PET/CT imaging and 99m Tc-MDP SPECT/CT bone imaging were 0.962 and 0.735 ( Z = 2.168, p < 0.05). At the lesion level, ¹⁸ F-DCFPyL PET/CT imaging showed superior diagnostic performance compared to 99m Tc-MDP SPECT/CT bone imaging in all indicators: sensitivity (97.6% vs. 72.6%, p < 0.01), specificity (95.7% vs. 73.9%, p < 0.01), accuracy (97.2% vs. 72.9%, p < 0.01), PPV (98.8% vs. 91.0%, p < 0.01), and NPV (91.7% vs. 42.5%, p < 0.01). The AUC values for ¹⁸ F-DCFPyL PET/CT imaging and 99m Tc-MDP SPECT/CT bone imaging were 0.966 and 0.733 ( Z = 3.541, p < 0.001). Conclusion Compared with 99m Tc-MDP SPECT/CT bone imaging, ¹⁸ F-DCFPyL PET/CT imaging demonstrated higher diagnostic efficiency for bone metastases in prostate cancer, and it can more accurately determine the presence of bone metastases. It is an important supplement to imaging examination for prostate cancer patients and has great potential and broad application prospects.
Article
Full-text available
Prostate-specific membrane antigen (PSMA) is the most extensively studied biomarker and antigen of prostate cancer. It is overexpressed in almost all prostate cancers, and the expression level increases with prostate cancer progression. PSMA is also highly expressed in the neovasculature of solid tumours including prostate cancer. As a result, numerous PSMA-specific ligands have been discovered for prostate cancer diagnosis and therapy, and one of them has been approved for clinical use. Moreover, a number of other PSMA-specific ligands are currently evaluated in clinical studies. In this review we discuss four major types of PSMA-specific ligands, including antibody, aptamer, peptide, and small molecule inhibitor. Their emerging applications in prostate cancer diagnosis, targeted drug delivery, and therapy are also discussed.
Article
Context Prostate-specific membrane antigen (PSMA) is a promising molecular target for prostate cancer (PCa) that has allowed the development of a novel diagnostic approach to PCA in the primary and recurrent settings. Objective To summarize available data and recommendations regarding the use of PSMA in newly diagnosed and recurrent PCa via a narrative review. Evidence acquisition A literature review was conducted using MEDLINE (via PubMed) and Scopus. The search strategy included meta-analyses, reviews, and original studies on staging and restaging with ⁶⁸Ga-PSMA positron emission tomography (PET)/computed tomography (CT). Evidence synthesis Studies comparing PSMA-targeted imaging and conventional imaging suggest superior performance of PSMA-targeted imaging in primary and recurrent PCa, albeit with several clinically relevant limitations. Pretreatment ⁶⁸Ga-PSMA PET/CT allowed more accurate PCa staging in compared to routine practice for high-risk cases, and identified a number of otherwise unknown metastatic lesions. In biochemically recurrent PCa, PSMA PET can reveal sites of recurrence with greater sensitivity and specificity than conventional imaging, potentially detecting a major proportion of occult disease. This review will help providers in applying the most up-to-date and relevant literature to (1) determine which patients truly have oligometastatic disease and (2) ascertain who is most likely to experience a meaningful response to local consolidation in the biochemical recurrence setting. Conclusions Data on PSMA diagnostic studies in primary and recurrent PCa highlight the accuracy and clinical application of PSMA PET. While this review and the evidence to date might lead to a perception of superiority in metastasis directed therapy, fundamental lack of phase III clinical trials with clinically meaningful outcomes are yet to be determined. Patient summary PSMA (prostate-specific membrane antigen) scans have shown great promise for initial evaluation of prostate cancer (PCa) and in detection of PCa recurrence. The benefits are more apparent for initial staging of PCa. There are more limited clinical trial results for PCa recurrence on how best to use this new technique to guide cancer treatment.
Article
Much of the modern growth in nuclear medicine has been driven by PET imaging of prostate-specific membrane antigen (PSMA) in men with prostate cancer. Fluorine-18 is the ideal PET radionuclide with a moderately long half-life, high positron yield, low positron energy, and cyclotron-based production. 18F-DCFPyL is the first Food and Drug Administration-approved compound in this class. In this review, we cover a number of aspects of radiofluorinated PSMA PET agents, including their historical development, the early clinical trials, key multicenter registration trials, emerging clinical agents, new compounds that are entering human use, and future directions for the field.
Article
Prostate cancer (PCa) is the third most common cancer diagnosed in the world. Since its first identification in 1987 and its first molecular cloning in 1993, prostate-specific membrane antigen (PSMA) has been developed as a theragnostic imaging biomarker and therapeutic agent for PCa. For metastatic castration-resistant PCa, PSMA-based PET imaging can be applied to the monitoring of disease and response assessment with PSMA-based therapeutics. This novel imaging modality is bringing new insights into diagnosis, stratification, and clinical decision-making and treatment.
Article
Full-text available
We previously demonstrated the ability to detect metastatic prostate cancer using (18)F-DCFBC (DCFBC), a low-molecular-weight radiotracer that targets the prostate-specific membrane antigen (PSMA). PSMA has been shown to be associated with higher Gleason grade and more aggressive disease. An imaging biomarker able to detect clinically significant high-grade primary prostate cancer reliably would address an unmet clinical need by allowing for risk-adapted patient management. We enrolled 13 patients with primary prostate cancer who were imaged with DCFBC PET prior to scheduled prostatectomy, with 12 of these patients also undergoing pelvic prostate MRI. Prostate DCFBC PET was correlated with MRI and histological and immunohistochemical (IHC) analysis on a prostate segment (12 regions) and dominant lesion basis. There were no incidental extraprostatic findings on PET suspicious for metastatic disease. MRI was more sensitive than DCFBC PET for detection of primary prostate cancer on a per-segment (sensitivities of 0.17 and 0.39 for PET and MRI for non-stringent analysis and 0.10 and 0.35 for stringent analysis, respectively) and per-dominant lesion analysis (sensitivity of 0.46 and 0.92 for PET and MRI, respectively). However, DCFBC PET was more specific than MRI by per-segment analysis (specificity of 0.96 and 0.89 for PET and MRI for non-stringent analysis and 1.00 versus 0.91 for stringent analysis, respectively) and highly specific for detection of high-grade lesions greater than or equal to 1.1 mL in size (Gleason 8 and 9). DCFBC uptake in tumors was positively correlated with Gleason score (ρ = 0.64, PSMA expression (ρ = 0.47) and PSA (ρ = 0.52). There was significantly lower DCFBC uptake in benign prostatic hypertrophy (BPH) compared to primary tumors (median SUVmax 2.2 versus SUVmax 3.5; P = 0.004). Although the sensitivity of DCFBC for primary prostate cancer was less than MRI, DCFBC PET was able to detect the more clinically significant high-grade tumors (Gleason score 8 and 9) with higher specificity than MRI. In particular, there was relatively low DCFBC PET uptake in BPH lesions compared to cancer in the prostate, which may allow for more specific detection of primary prostate cancer by DCFBC PET. This study demonstrates the utility of PSMA-based PET for detection of primary prostate cancer, which may be used in conjunction with MRI to guide biopsy and identify clinically significant, aggressive disease non-invasively. Copyright © 2015 by the Society of Nuclear Medicine and Molecular Imaging, Inc.
Article
Full-text available
Since the introduction of positron emission tomography (PET) imaging with (68)Ga-PSMA-HBED-CC (=(68)Ga-DKFZ-PSMA-11), this method has been regarded as a significant step forward in the diagnosis of recurrent prostate cancer (PCa). However, published data exist for small patient cohorts only. The aim of this evaluation was to analyse the diagnostic value of (68)Ga-PSMA-ligand PET/CT in a large cohort and the influence of several possibly interacting variables. We performed a retrospective analysis in 319 patients who underwent (68)Ga-PSMA-ligand PET/CT from 2011 to 2014. Potential influences of several factors such as prostate-specific antigen (PSA) level and doubling time (DT), Gleason score (GSC), androgen deprivation therapy (ADT), age and amount of injected tracer were evaluated. Histological verification was performed in 42 patients after the (68)Ga-PSMA-ligand PET/CT. Tracer uptake was measured in 901 representative tumour lesions. In 82.8 % of the patients at least one lesion indicative of PCa was detected. Tumor-detection was positively associated with PSA level and ADT. GSC and PSA-DT were not associated with tumor-detection. The average maximum standardized uptake value (SUVmax) of tumour lesions was 13.3 ± 14.6 (0.7-122.5). Amongst lesions investigated by histology, 30 were false-negative in 4 different patients, and all other lesions (n = 416) were true-positive or true-negative. A lesion-based analysis of sensitivity, specificity, negative predictive value (NPV) and positive predictive value (PPV) revealed values of 76.6 %, 100 %, 91.4 % and 100 %. A patient-based analysis revealed a sensitivity of 88.1 %. Of 116 patients available for follow-up, 50 received local therapy after (68)Ga-PSMA-ligand PET/CT. (68)Ga-PSMA-ligand PET/CT can detect recurrent PCa in a high number of patients. In addition, the radiotracer is highly specific for PCa. Tumour detection is positively associated with PSA and ADT. (68)Ga-PSMA-ligand PET/CT can help delay systemic therapy of PCa.
Article
Full-text available
Unlabelled: Prostate-specific membrane antigen (PSMA) is a well-established target for developing radiopharmaceuticals for imaging and therapy of prostate cancer (PCa). We have recently reported that novel (99m)Tc-labeled small-molecule PSMA inhibitors bind with high affinity to PSMA-positive tumor cells in vitro and localize in PCa xenografts. This study reports the first, to our knowledge, human data in men with metastatic PCa and in healthy male subjects. Methods: Under an exploratory investigational new drug, using a cross-over design, we compared the pharmacokinetics, biodistribution, and tumor uptake of (99m)Tc-MIP-1404 and (99m)Tc-MIP-1405 in 6 healthy men and 6 men with radiographic evidence of metastatic PCa. Whole-body images were obtained at 10 min and 1, 2, 4, and 24 h. SPECT was performed between 3 and 4 h after injection. Results: Both agents cleared the blood rapidly, with MIP-1404 demonstrating significantly lower urinary activity (7%) than MIP-1405 (26%). Both agents showed persistent uptake in the salivary, lacrimal, and parotid glands. Uptake in the liver and kidney was acceptable for imaging at 1-2 h. In men with PCa, both agents rapidly localized in bone and lymph node lesions as early as 1 h. SPECT demonstrated excellent lesion contrast. Good correlation was seen with bone scanning; however, more lesions were demonstrated with (99m)Tc-MIP-1404 and (99m)Tc-MIP-1405. The high-contrast images exhibited tumor-to-background ratios from 3:1 to 9:1 at 4 and 20 h. Conclusion: Compared with the standard-of-care bone scanning, (99m)Tc-MIP-1404 and (99m)Tc-MIP-1405 identified most bone metastatic lesions and rapidly detected soft-tissue PCa lesions including subcentimeter lymph nodes. Because (99m)Tc-MIP-1404 has minimal activity in the bladder, further work is planned to correlate imaging findings with histopathology in patients with high-risk metastatic PCa.
Article
Full-text available
Ex vivo studies have shown that the gastrin releasing peptide receptor (GRPr) is overexpressed on almost all primary prostate cancers, making it a promising target for prostate cancer imaging and targeted radiotherapy.Methods: Biodistribution, dosimetry and tumor uptake of the GRPr antagonist 64Cu-CB-TE2A-AR06 [(64Cu-4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo(6.6.2)hexadecane)-PEG4-D-Phe-Gln-Trp-Ala-Val-Gly-His-Sta-LeuNH2] were studied by PET/CT in four patients with newly diagnosed prostate cancer (T1c-T2b, Gleason 6-7).Results: No adverse events were observed after injection of 64Cu-CB-TE2A-AR06. Three of four tumors were visualized with high contrast [tumor-to-prostate ratio > 4 at 4 hours (h) post injection (p.i.)], one small tumor (T1c, < 5% tumor on biopsy specimens) showed moderate contrast (tumor-to-prostate ratio at 4 h: 1.9). Radioactivity was cleared by the kidneys and only the pancreas demonstrated significant accumulation of radioactivity, which rapidly decreased over time.Conclusion: 64Cu-CB-TE2A-AR06 shows very favorable characteristics for imaging prostate cancer. Future studies evaluating 64Cu-CB-TE2A-AR06 PET/CT for prostate cancer detection, staging, active surveillance, and radiation treatment planning are necessary.
Article
Prostate-specific membrane antigen (PSMA) is a recognized target for imaging prostate cancer. Here we present initial safety, biodistribution, and radiation dosimetry results with [(18)F]DCFPyL, a second-generation fluorine-18-labeled small-molecule PSMA inhibitor, in patients with prostate cancer. Biodistribution was evaluated using sequential positron-emission tomography (PET) scans in nine patients with prostate cancer. Time-activity curves from the most avid tumor foci were determined. The radiation dose to selected organs was estimated using OLINDA/EXM. No major radiotracer-specific adverse events were observed. Physiologic accumulation was observed in known sites of PSMA expression. Accumulation in putative sites of prostate cancer was observed (SUVmax up to >100, and tumor-to-blood ratios up to >50). The effective radiation dose from [(18)F]DCFPyL was 0.0139 mGy/MBq or 5 mGy (0.5 rem) from an injected dose of 370 MBq (10 mCi). [(18)F]DCFPyL is safe with biodistribution as expected, and its accumulation is high in presumed primary and metastatic foci. The radiation dose from [(18)F]DCFPyL is similar to that from other PET radiotracers.
Article
Glucose and glutamine are the two principal nutrients that cancer cells use to proliferate and survive. Many cancers show altered glucose metabolism, which constitutes the basis for in vivo positron emission tomography (PET) imaging with (18)F-fluorodeoxyglucose ((18)F-FDG). However, (18)F-FDG is ineffective in evaluating gliomas because of high background uptake in the brain. Glutamine metabolism is also altered in many cancers, and we demonstrate that PET imaging in vivo with the glutamine analog 4-(18)F-(2S,4R)-fluoroglutamine ((18)F-FGln) shows high uptake in gliomas but low background brain uptake, facilitating clear tumor delineation. Chemo/radiation therapy reduced (18)F-FGln tumor avidity, corresponding with decreased tumor burden. (18)F-FGln uptake was not observed in animals with a permeable blood-brain barrier or neuroinflammation. We translated these findings to human subjects, where (18)F-FGln showed high tumor/background ratios with minimal uptake in the surrounding brain in human glioma patients with progressive disease. These data suggest that (18)F-FGln is avidly taken up by gliomas, can be used to assess metabolic nutrient uptake in gliomas in vivo, and may serve as a valuable tool in the clinical management of gliomas. Copyright © 2015, American Association for the Advancement of Science.
Article
</sup>Engineered antibody fragments offer faster delivery with retained tumor specificity and rapid clearance from non-tumor tissues. Here, we demonstrate that positron emission tomography (PET)-based detection of prostate specific membrane antigen (PSMA) in prostatic tumor models using engineered bivalent antibodies built on single chain fragments (scFv) derived from the intact antibody, huJ591, offer similar tumor delineating properties but with the advantage of rapid targeting and imaging. <sup>89</sup>Zr-radiolabeled huJ591 scFv (dimeric scFv-C<sub>H</sub>3; <sup>89</sup>Zr-Mb) and cysteine-diabodies (dimeric scFv; <sup>89</sup>Zr-Cys-Db) demonstrated internalization and similar Kds (~2 nM) compared to <sup>89</sup>Zr-huJ591 in PSMA(+) cells. Tissue distribution assays established the specificities of both <sup>89</sup>Zr-Mb and <sup>89</sup>Zr-Cys-Db for PSMA(+) xenografts (6.2 ± 2.5 %ID/g and 10.2 ± 3.4 %ID/g at 12 h p.i. respectively), while minimal accumulation in PSMA(-) tumors was observed. From the PET images, <sup>89</sup>Zr-Mb and <sup>89</sup>Zr-Cys-Db exhibited faster blood clearance than the parent huJ591 while tumor-to-muscle ratios for all probes show comparable values across all time points. Ex vivo autoradiography and histology assessed the distribution of the probes within the tumor. Imaging PSMA-expressing prostate tumors with smaller antibody fragments offer rapid tumor accumulation and accelerated clearance; hence, shortened wait periods between tracer administration and high-contrast tumor imaging and lower dose-related toxicity is potentially realized.
Article
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States in the current year and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data were collected by the National Cancer Institute, the Centers for Disease Control and Prevention, and the North American Association of Central Cancer Registries and mortality data were collected by the National Center for Health Statistics. A total of 1,665,540 new cancer cases and 585,720 cancer deaths are projected to occur in the United States in 2014. During the most recent 5 years for which there are data (2006-2010), delay-adjusted cancer incidence rates declined slightly in men (by 0.6% per year) and were stable in women, while cancer death rates decreased by 1.8% per year in men and by 1.4% per year in women. The combined cancer death rate (deaths per 100,000 population) has been continuously declining for 2 decades, from a peak of 215.1 in 1991 to 171.8 in 2010. This 20% decline translates to the avoidance of approximately 1,340,400 cancer deaths (952,700 among men and 387,700 among women) during this time period. The magnitude of the decline in cancer death rates from 1991 to 2010 varies substantially by age, race, and sex, ranging from no decline among white women aged 80 years and older to a 55% decline among black men aged 40 years to 49 years. Notably, black men experienced the largest drop within every 10-year age group. Further progress can be accelerated by applying existing cancer control knowledge across all segments of the population. CA Cancer J Clin 2014. (©) 2014 American Cancer Society, Inc.
Article
The goal of this pilot study was to explore the feasibility of (89)Zr-labeled J591 monoclonal antibody positron-emission tomography (PET) of localized prostate cancer (PCa). Prior to scheduled radical prostatectomy, eleven patients were injected intravenously with (89)Zr-J591 followed 6 days later by whole-body PET. Patients underwent surgery the day after imaging and the specimens were imaged by ex vivo micro-PET and custom 3 Tesla magnetic resonance scanner coil. PET imaging studies and histopathology were correlated. Median age was 61 years (range, 47-68 years), median PSA was 5.2 ng/mL (3.5-12.0 ng/mL), and median biopsy Gleason score of the 11 index lesions was 7 (range, 7-9). On histopathology, a total of 22 lesions were identified. Median lesion size was 5.5 mm (range, 2-21 mm) and median post-RP Gleason score was 7 (range, 6-9). On in vivo PET, 8 of 11 index lesions were identified (72.7%). Lesion identification improved with increasing lesion size for in vivo (p<0.0001) and ex vivo PET imaging (p<0.0001), and increasing Gleason score (ex vivo PET imaging, p=0.01; in vivo, p=0.14). SUV appeared to correlate with increased Gleason score but was not significant (p=0.19). This is the first report describing (89)Zr-J591/PSMA PET imaging in localized PCa. In this setting, (89)Zr-J591-PET binds to tumor foci in situ and identifies primarily Gleason ≥7 and larger sized tumors, likely corresponding to clinically significant disease warranting definitive therapy. Future work is planned in a larger clinical validation trial to better define the utility of (89)Zr-J591-PET in localized PCa.