Skip to main content
Korean Journal of Radiology logoLink to Korean Journal of Radiology
. 2023 Feb 23;24(6):574–589. doi: 10.3348/kjr.2022.1002

The Application of Radiolabeled Targeted Molecular Probes for the Diagnosis and Treatment of Prostate Cancer

Luyi Cheng 1, Tianshuo Yang 1, Jun Zhang 2, Feng Gao 3, Lingyun Yang 4, Weijing Tao 1,
PMCID: PMC10248352  PMID: 37271211

Abstract

Radiopharmaceuticals targeting prostate-specific membrane antigens (PSMA) are essential for the diagnosis, evaluation, and treatment of prostate cancer (PCa), particularly metastatic castration-resistant PCa, for which conventional treatment is ineffective. These molecular probes include [68Ga]PSMA, [18F]PSMA, [Al18F]PSMA, [99mTc]PSMA, and [89Zr]PSMA, which are widely used for diagnosis, and [177Lu]PSMA and [225Ac]PSMA, which are used for treatment. There are also new types of radiopharmaceuticals. Due to the differentiation and heterogeneity of tumor cells, a subtype of PCa with an extremely poor prognosis, referred to as neuroendocrine prostate cancer (NEPC), has emerged, and its diagnosis and treatment present great challenges. To improve the detection rate of NEPC and prolong patient survival, many researchers have investigated the use of relevant radiopharmaceuticals as targeted molecular probes for the detection and treatment of NEPC lesions, including DOTA-TOC and DOTA-TATE for somatostatin receptors, 4A06 for CUB domain-containing protein 1, and FDG. This review focused on the specific molecular targets and various radionuclides that have been developed for PCa in recent years, including those mentioned above and several others, and aimed to provide valuable up-to-date information and research ideas for future studies.

Keywords: Prostate cancer, Prostate specific membrane antigen, Neuroendocrine prostate cancer, Radionuclide imaging, Radionuclide treatment

INTRODUCTION

Prostate cancer (PCa) arises from prostate epithelial cells and is the most common malignancy of the male genitourinary system (7.1% of all cancers) and the second leading cause of cancer-related deaths in men [1]. Current treatment options for early-stage PCa include prostatectomy or local radiotherapy, which are generally effective. Nevertheless, approximately 35% of PCa patients develop biochemical recurrence, for which the diagnostic criterion is elevated prostate-specific antigen (PSA) levels. Moreover, a significant proportion of these patients also exhibit distant metastases [2]. Prostate-specific membrane antigen (PSMA) shows significantly higher expression in primary and metastatic lesions than in normal tissues and has become an important molecular target in PCa.

Androgen receptor (AR)-targeted therapy is required when PCa progresses or recurs. However, although such treatment can temporarily control tumor progression (usually for 2–3 years), it inevitably promotes the adaptation of tumor cells to low-androgen conditions, resulting in highly aggressive (and lethal) metastatic castration-resistant prostate cancer (mCRPC) [3]. AR pathway inhibition, which is widely used clinically, leads to the neuroendocrine differentiation of PCa cells, promoting the more general castration-resistant prostate cancer (CRPC) subtype, namely, neuroendocrine prostate cancer (NEPC) [4]. NEPC is unresponsive to AR-targeted therapy and is characterized by decreased PSMA expression [5].

Radionuclide-targeted molecular probes can facilitate the precise positioning and treatment of lesions and effectively prolong progression-free and overall survival [6,7,8]. Therefore, radiopharmaceuticals play an important role in the detection and treatment of primary and metastatic PCa lesions, particularly those in the mCRPC and NEPC stages. Currently, there are various radionuclide molecular probes targeting PCa. In this review, we divided these molecular probes into two main categories: PSMA and non-PSMA. Non-PSMA molecular probes are further classified into those targeting NEPC and others. It is hoped that this review involving dozens of radiopharmaceuticals (Tables 1, 2) will help to improve the overall understanding of available radionuclide molecular probes for PCa.

Table 1. The Structural Formula, Indication, Advantages, and Disadvantages of Various Radiopharmaceuticals in PCa.

Radiopharmaceuticals Formula of Ligand Indication Advantage Limitation
Imaging
68Ga-PSMA-11 N,N’-bis(2-hydroxy-5-(carboxyethyl)Benzyl] ethylenediamineN,N’-diacetic acid Primary and recurrent adenocarcinoma of prostate and its metastatic lesions 1. Clear blood and organs quickly 1. Half-life short
2. Low liver accumulation 2. Slightly low spatial resolution
68Ga-PSMA-617 2-[3-(1-Carboxy-5-{3-naphthalen-2-yl-2-[(4-{[2-(4,7,10-tris-carboxymethyl-1,4,7,10-tetraaza-cyclododec-1-yl)-acetylamino]-methyl}-cyclohexanecarbonyl)-amino]-propionylamino}-pentyl)-ureido]-pentanedioic acid Primary and recurrent adenocarcinoma of prostate and its metastatic lesions 1. The kidneys excrete quickly High intake of salivary glands and intestines
2. High ratio of tumor to background tissue
3. Ligands can be attached to therapeutic nuclides
68Ga-PSMA-I&T (2S)-2-[[(1S)-1-carboxy-5-[[8-[[(5R)-5-carboxy-5-[[(2R)-2-[[(2R)-2-[[4-carboxy-4-[4,7,10-tris(carboxymethyl)-1,4,7,10-tetrazacyclododec-1-yl]butanoyl]amino]-3-(4-hydroxy-3-iodophenyl)propanoyl]amino]-3-phenylpropanoyl]amino]pentyl] amino]-8-oxooctanoyl]amino]pentyl]carbamoylamino]pentanedioic acid Primary and recurrent adenocarcinoma of prostate and its metastatic lesions 1. Low background uptake in liver and spleen High renal intake
2. Ligands can be attached to therapeutic nuclides
68Ga-iPSMA-BN HO-Glu-CO-Lys{Nal-Cys[Pyr-Gln-Lys(maleimidobutyl)-Leu-Gly-AsnGln-Trp-Ala-Val-Gly-His-Leu-Met-NH2]-DOTA}-OH Primary and recurrent adenocarcinoma of prostate and its metastatic lesions Detect liver metastases because of low liver background Low hepatobiliary clearance
68Ga-PSMA-TO-1 (3S,7S,12S,19R,26R,29R,32R,35S)-19-(3,5-dicarboxybenzamido)-32-(4-hydroxybenzyl)-29,35-bis(naphthalen-2-ylmethyl)-5,10,17,20,28,31,34,37-octaoxo-40-(4,7,10-tris(carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)-4,6,11,16,21,27,30,33,36-nonaazatetracontane-1,3,7,12,26,40-hexacarboxylic acid All lesions from adenocarcinoma of prostate and NEPC 1. Higher lesions’ uptake value than 68Ga-PSMA-I&T High renal uptake
2. Liver metastases because of low background in liver
3. Detect various differentiated prostate cancer
4. Ligands can be attached to therapeutic nuclides
68Ga-NeoBOMB1 2-[4-[2-[[4-[[2-[2-[[(2R)-1-[[(2S)-5-amino-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[2-[[(2S)-1-(2,6-dimethylheptan-4-ylamino)-3-(1H-imidazol-5-yl)-1-oxopropan-2-yl]amino]-2-oxoethyl]amino]-3-methyl-1-oxobutan-2-yl]amino]-1-oxopropan-2-yl]amino]-3-(1H-indol-3-yl)-1-oxopropan-2-yl]amino]-1,5-dioxopentan-2-yl]amino]-1-oxo-3-phenylpropan-2-yl]amino]-2-oxoethoxy]acetyl]amino]phenyl] methylamino]-2-oxoethyl]-7,10-bis(carboxylatomethyl)-1,4,7,10-tetrazacyclododec-1,4,7-triyl]triacetic acid All lesions from adenocarcinoma of prostate and NEPC 1. Retain longer in tumor -
2. High metabolic stability
3. Rapid renal excretion
4. Detect various differentiated prostate cancer
68Ga-DOTA-NT-20.3 Ac-Lys(DOTA)-Pro-Me-Arg-Arg-Pro-Tyr-Tle-Leu-OH NEPC Exhibit high stability and retention within tumors Low sensitivity for adenocarcinoma of prostate
68Ga-RM26 D-Phe–Gln–Trp–Ala–Val–Gly–His–Sta–Leu–NH2 NEPC High tumor-to-background ratio Low sensitivity for adenocarcinoma of prostate
18F-PSMA-1007 2-(3-(1-carboxyl-5-(6-[18F]pyridine-3-carbonyl) -amino)-Amyl)-urea)-glutaric acid Adenocarcinoma of prostate 1. More sensitive to bone marrow metastasis Difficult to observe liver lesions
2. Lesions near the bladder and ureter
1818F-DCFPyL 2-(3-{1-carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid Adenocarcinoma of prostate 1. Low background of blood pool High intake of salivary glands, liver, small intestine
2. Detect prostate lesions with low grade and small volume
18F-JK-PSMA-7 2-MeO-2-(3-{1-carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid Adenocarcinoma of prostate Compared with 18F-DCFPyL: Low detection rate when PSA < 0.3 ng
1. High image quality
2. High small PSMA-expressing foci sensitivity
Al18F-PSMA-BCH (((S)-5-((S)-2-((1r,4S)-4-((2-(4,7-bis(carboxymethyl)-1,4,7-triazonan-1-yl)acetamido)methyl)cyclohexane-1-carboxamido)-3-(naphthalen-2-yl)propanamido)-1-carboxypentyl)carbamoyl)-L-glutamic acid Adenocarcinoma of prostate Easy to prepare Compared to 68Ga-PSMA-11 and 18F-PSMA-1007: Low detection rate of lesions
Al18F-PSMA-11 N,N‘-bis(2-hydroxy-5-(carboxyethyl)Benzyl] ethylenediamineN,N’-diacetic acid Adenocarcinoma of prostate Easy to prepare Poor stability at room temperature
99mTc-MIP-1404 ((7S,12S,16S)-1-(1-(2-(bis(carboxymethyl)amino)-2-oxoethyl)-1H-imidazol-2-yl)-2-((1-(2-(bis(carboxymethyl)amino)-2-oxoethyl)-1H-imidazol-2-yl)methyl)-9,14-dioxo-2,8,13,15-tetraazaoctadecane-7,12,16,18-tetracarboxylic acid Adenocarcinoma of prostate 1. Can delay imaging Low detection rate of liver lesions
2. Detect the gland and pelvis lesions in early stage because of minimal urinary excretion
99mTc-MIP-1405 (7S,14S,18S)-7-amino-1-(1-(carboxymethyl)-1H-imidazol-2-yl)-2-((1(carboxymethyl)-1H-imidazol-2-yl)methyl)-8,16-dioxo-2,9,15,17tetraazaicosane-14,18,20-tricarboxylic acid Adenocarcinoma of prostate Compared with 99mTc-MIP-1404: Less sensitive to liver lesions than 99mTc-MIP-1404
The whole-body clearance was significantly greater
89Zr-PSMA-Df* - Adenocarcinoma of prostate Compared with 68Ga-PSMA-11 and 18F-JKPSMA-7: Difficult to prepare
1. Long half-life
2. High tumor-to-background ratio
3. High sensitivity of metastatic lymph nodes near the ureter
89Zr-df-IAB2M* - Adenocarcinoma of prostate 1. Rapid bio-distribution with efficient target penetration Difficult to prepare
2. Facilitate earlier lesion detection
89Zr-4A06* - All lesions from adenocarcinoma of prostate and NEPC 1. High sensitivity Difficult to prepare
18F-FDG 2-Fluoro-2-deoxy-D-glucose NEPC 1. Easy to prepare Not specific
2. NEPC can be detected
18F-FSPG (S)-4-(3-[18F]fluoropropyl)-L-glutamate Adenocarcinoma of prostate 1. Excrete rapidly through the kidney Difficult to observe lesions near the bladder and ureter
2. Low background activity
3. Reflect tumor redox status and antioxidant capacity to predict tumor chemotherapy resistance
Treatment
177Lu-PSMA-617 Like 68Ga-PSMA-617 mCRPC Integration of diagnosis and treatment Excessive lacrimal gland intake
177Lu-EB-PSMA-617 (((1S)-5-((2S)-2-(4-((2-((1-(2-(((R)-6-((4’-((Z)-(8-amino-1-hydroxy-5,7-disulfonaphthalen-2-yl)diazenyl)-3,3’-dimethyl-[1,1’-biphenyl]-4-yl)amino)-6-oxo-5-(2-(4,7,10-tris (carboxymethyl)-1,4,7,10-tetraazacyclododecan-1-yl)acetamido)hexyl)amino)-2-oxoethyl)-2,5-dioxopyrrolidin-3-yl)thio)acetamido)methyl)cyclohexane-1-carboxamido)-3-(naphthalen-2-yl)propanamido)-1-carboxypentyl)carbamoyl)-L-glutamic acid mCRPC Compared with 177Lu-PSMA-617: Excessive lacrimal gland intake
1. Longer half-life
2. The maximum therapeutic effect with the lowest dose
177Lu-PSMA-I&T Like 68Ga-PSMA-11 mCRPC Integration of diagnosis and treatment Nephrotoxicity
177Lu-PSMA-TO-1 Like 68Ga-PSMA-TO-1 mCRPC and NEPC Compared with 177Lu-PSMA-617: 1. Nephrotoxicity
1. Long circulation time and improve the therapeutic effect 2. Hematotoxicity
2. Therapy various differentiated prostate cancer 3. Bone marrow involvement
177Lu-4A06* mCRPC and NEPC 1. Can better inhibit and eliminate mCRPC lesions -
2. Therapy various differentiated prostate cancer
225Ac-PSMA-617 Like 68Ga-PSMA-617 mCRPC 1. Long physical half-life and high linear energy of α nuclides Xerostomia
2. Better therapeutic effect than 177Lu-PSMA-617
225Ac-PSMA-TO-1 Like 68Ga-PSMA-TO-1 mCRPC and NEPC 1. Long physical half-life and high linear energy of α nuclides Nephrotoxicity
2. Better therapeutic effect than 225Ac-PSMA-617
3. Therapy various differentiated prostate cancer
90Y-DOTA-TOC 1,4,7,10-tetraazacyclododecane-N,N’,N’’,N’’’-tetra-acetic acid-D-Phe1-Tyr3-Octreotide NEPC Improve the treatment effect of neuroendocrine tumors Hematotoxicity

*There is no formula for antibodies. PCa = prostate cancer, NEPC = neuroendocrine prostate cancer, mCRPC = metastatic castration-resistant prostate cancer, PSMA = prostate-specific membrane antigens, PSA = prostate-specific antigen, I&T = imaging and treatment

Table 2. Clinical Significance of Radionuclides in Patients with Prostate Cancer.

Radiopharmaceuticals Clinical Significance
[68Ga]Ga-PSMA-11 They can detect more than 80% of clinically unpredictable biochemical relapses, which could be benefit for early diagnosis, staging and curative effect of prostatic adenocarcinoma and guide clinicians to formulate personalized treatment plans. Among them, [68Ga]Ga-PSMA-I&T can detect high-grade prostatic adenocarcinoma (GS 8 or above, PSA > 10 ng/mL) sensitively and [68Ga]Ga-iPSMA-BN accumulates the least in the body.
[68Ga]Ga-PSMA-617
[68Ga]Ga-PSMA-I&T
[68Ga]Ga-iPSMA-BN
[68Ga]Ga-PSMA-TO-1 It can improve the accuracy of early diagnosis, staging and curative effect of prostatic adenocarcinoma under the delayed imaging.
[68Ga]NeoBOMB1 It stays in the tumor longer than other organs, so it can detect the prostatic adenocarcinoma and NEPC more accurately.
[68Ga]Ga-DOTA-NT-20.3 They can make early diagnosis and curative effect of NEPC, including the PRRT treatment.
[68Ga]RM26
[18F]F-PSMA-1007 They can sensitively detect small periurethral lesions of prostatic adenocarcinoma.
[18F]DCFPyL
[18F]F-JK-PSMA-7
Al[18F]F-PSMA-BCH In the absence of the Ge/Ga generator, they are easy to be prepared, which is beneficial to examination of more prostatic adenocarcinoma patient.
Al[18F]F-PSMA-11
[99mTc]Tc-MIP-1404 Radiation dose of patients with prostatic adenocarcinoma can be reduced in the examination with single-photon radionuclides.
[99mTc]Tc-MIP-1405
[89Zr]Zr-PSMA-Df It can improve the detection rate of lymph node metastasis near ureter, which is conducive to the accurate staging of the prostatic adenocarcinoma.
[89Zr]Zr-df-IAB2M It can perform delayed imaging and can rapid from the body quickly.
[89Zr]4A06 They can detect neuroendocrine prostate cancer to distinguish between different types of prostate cancer.
[18F]FDG
[18F]FSPG It can reflect tumor redox status and antioxidant capacity, and predict tumor chemotherapy resistance of prostatic adenocarcinoma.
[177Lu]Lu -PSMA-617 These radionuclides are suitable for patients with prostatic adenocarcinoma who have already developed mCRPC.
[177Lu]Lu-EB-PSMA-617
[177Lu]Lu -PSMA-I&T
[177Lu]Lu-PSMA-TO-1 They can therapy prostatic adenocarcinoma and NEPC at all stages.
[177Lu]4A06
[177Lu]NeoBOMB1
[225Ac]Ac-PSMA-617 They can therapy lesions that are insensitive to 177Lu-lableded pharmaceuticals.
[225Ac]Ac-PSMA-TO-1
[90Y]Y-DOTA-TOC It can therapy patients with NEPC at all stages.

There is almost no difference in the clinical significance of nuclides in the same cell. GS = Gleason score, PSA = prostate-specific antigen, NEPC = neuroendocrine prostate cancer, PRRT = peptide receptor radionuclide therapy, mCRPC = metastatic castration-resistant prostate cancer

PSMA

PSMA is a 100kDa type II transmembrane protein [9,10,11], with glutamate carboxypeptidase and folate hydrolase activity [12,13]. PSMA expression is increased 100- to 1000-fold in PCa cells compared to that in normal cells and is correlated with PCa grade based on the Gleason score (GS), which is an independent predictor of PCa progression [14,15,16]. Monoclonal antibodies and small-molecule inhibitors have been found to effectively bind to the extracellular portion of PSMA, where most small-molecule compounds, such as PSMA-617, PSMA-I&T (I&T: imaging and treatment), MIP-1404, and MIP-1405, are rapidly excreted by cells, which reduces the radiation exposure. PSMA acts as an important site for the coupling of radionuclides, allowing for the detection and treatment of PCa lesions. The following is a summary of the radiopharmaceuticals that target PSMA in the diagnosis and treatment of PCa.

68Ga-PSMA

Gallium-68 is a short half-life positron radionuclide (half-life:68 min) used for positron emission tomography (PET) imaging [17]. In general, 68Ga-labeled PSMA PET performs significantly better than conventional imaging examinations because of its exceptionally high sensitivity and specificity for primary and metastatic PCa lesions [18]. [68Ga]PSMA PET has been reported to detect 35.4% more PCa lesions than MRI [19,20]. Additionally, compared to 18F-choline and 11C-choline, [68Ga]PSMA PET increases the detection of local recurrence and metastases to the lymph nodes and bone by 14% [21,22]. Furthermore, when PSA levels are very low, [68Ga]PSMA PET can accurately detect PCa recurrent lesions biochemically [9,23]. [68Ga]PSMA PET can also be used for accurate TNM staging, which helps guide appropriate treatment strategies for patients [24,25,26].

Common PSMA ligands coupled to gallium-68 include PSMA-11, PSMA-617, and PSMA-I&T. These are all small-molecule inhibitors. For small molecule inhibitors, the strong zinc-binding motif of the enzyme’s active site and the glutaric acid portion of the P1′ position bound to the S1′ pocket are critical [27,28]. In PSMA-11, the lipophilicity between the radionuclide and the PSMA-active group (glutamate-urea-lysine) is enhanced via the conjugated HBED-CC group. This has now become the preferred chelate for gallium-68 PSMA tracers [27,29,30]. [68Ga]Ga-PSMA-11 significantly accumulates in the spleen and salivary glands and is equally eliminated via the renal and hepatobiliary routes. When [68Ga]Ga-PSMA-11 PET is used for the detection of PCa lesions, the positive predictive value was consistently greater than 0.8 and 0.75 for pelvic nodal metastasis [31,32]. [68Ga]Ga-PSMA-11 positron emission tomography-computed tomography (PET-CT) detected PCa recurrence in 86% of patients who did not meet the definition of biochemical recurrence [33,34].

PSMA-617 is another PSMA ligand that connects radionuclides via conjugated DOTA chelates. DOTA-conjugated PSMA-617 was labeled with 68Ga for PET imaging and 177Lu/225Ac for nuclide therapy. However, because [68Ga] Ga-PSMA-617 is rapidly excreted (mainly through the kidneys), it can interfere with the diagnosis of primary and periurethral PCa lesions. Therefore, this tracer is primarily used to evaluate metastatic PCa. PSMA-I&T is similar to PSMA-617, and can be coupled to multiple radionuclides. However, it uses the chelating compound DATAGA, as opposed to DOTA, for PSMA-617. [68Ga]Ga-PSMA-I&T has a low background uptake in the liver and spleen. Therefore, [68Ga]Ga-PSMA-I&T shows greater sensitivity for the detection of primary and metastatic periurethral PCa lesions compared with [68Ga]Ga-PSMA-617, especially for high-grade PCa (GS 8 or above, PSA > 10 ng/mL). However, the kidneys have a much higher uptake of PSMA-I&T than PSMA-617, which is unfavorable for treatment [35,36,37].

PSMA-TO-1 (tumor-optimized-1) and iPSMA-BN (iPSMA-Lys3-bombesin) are relatively rare gallium-labeled PSMA ligands. PSMA-TO-1 was developed to have a prolonged circulation time using an extended linker with additional naphthyl groups to increase protein binding in the blood and promote lipophilicity. Therefore, it has a higher uptake in the primary and metastatic lesions than PSMA-617 and PSMA-I&T. Liver metastases show higher tracer uptake of [68Ga]Ga-PSMA-TO-1 than of [68Ga]Ga-PSMA-11 (mean standard uptake value [SUVmean]: 6.0 vs. 4.0). After 120 min, the SUVmean of [68Ga]Ga-PSMA-TO-1 in metastases increased to 8.0 (up to 33%) [38]. Furthermore, there have been few comparative studies of PSMA-TO-1 and other gallium-labeled PSMA tracers. iPSMA-BN includes a heterodimer with the sequence HO-Glu-CO-Lys-OH and was recently designed to target PSMA and gastrin-releasing peptide receptor (GRPR), which are overexpressed in different stages of PCa. iPSMA-BN can be labeled with 68Ga and 177Lu, making it possible to integrate diagnosis and treatment. [68Ga]Ga-iPSMA-BN showed faster blood clearance than [68Ga]Ga-PSMA-11 (half-life in the blood = 2.64 min vs. 6.5 min). [68Ga]Ga-iPSMA-BN clearly visualized the pancreas and is eliminated mainly via the kidneys, with low hepatobiliary clearance and low salivary gland uptake.

18F-PSMA

18F is produced by a cyclotron and has a higher yield and longer half-life than 68Ga (110 min vs. 68 min) [39,40]. 18F-labeled PSMA ligands have the potential for centralized production and distribution, thus enabling cost savings. PSMA-1007 is a novel PSMA ligand with Glu-urea-Lys targeting the PSMA enzyme pocket S1′ and a naphthalenyl linkage region thought to co-target the hydrophobic accessory pocket S1. The main difference is that the radiolabeled molecule in the carboxyl group of the DOTA chelating agent is replaced by two glutamic acids [27,41]. [18F]F-PSMA-1007 is excreted mainly in hepatic bile and minimally in urine, facilitating a high detection rate of lesions that are near the bladder and ureter. Moreover, [18F]F-PSMA-1007 offers advantages in terms of imaging quality and sensitivity, making it a promising candidate for clinical applications [42,43,44]. Another 18F labeled PSMA-specific small molecule imaging agent, 2-(3-[45]-ureido)-pentanedioic acid ([18F]DCFPyL), has also been developed based on the Glu-urea-Lys motif. This molecule is characterized by high affinity and favorable pharmacokinetics in vivo [23,45,46], thus allowing the earlier detection of local recurrence, even at lower PSA levels [47]. The novel probe, JK-PSMA-7, was identified by screening multiple DCFPyL analogs, with the main difference being the addition of a methoxy group to the pyridine ring. [18F] F-JK-PSMA-7 is rapidly excreted via the kidneys [48,49] and yields high-quality images, allowing the detection of small PSMA-expressing foci with high sensitivity [50,51] (Fig. 1).

Fig. 1. Maximum intensity projections (MIPs), axial images of positron emission tomography (PET), and axial fusion images of PET-computed tomography (CT) using [68Ga]Ga-PSMA-11 (A, B, C) and [18F]F-PSMA-1007 (D, E, F) in a 67-year-old male with Gleason 8 and PSA 4.9 ng/mL. The MIP image of [68Ga]Ga-PSMA-11 (A) (arrow) shows significant uptake in the bladder and left ureter, while urinary excretion of [18F]F-PSMA-1007 (F) is virtually absent. Lesions in the left prostate lobe are visible on both scans (arrowheads in B, D). However, the second lesion in the right lobe is only visible under [18F]F-PSMA-1007 (arrow in D), and has been pathologically confirmed to be a malignant lesion. Reprinted with permission from Kuten et al. [39] (J Nucl Med 2020;61:527-532; https://doi.org/10.2967/jnumed.119.234187). PSMA = prostate-specific membrane antigen.

Fig. 1

Al18F-PSMA

Al18F was developed as an 18F-labeling technique that allows convenient 18F-labeling and requires less time under mild conditions. Al[18F]F-PSMA-BCH can be prepared at a reasonable yield within 30 min and is mostly a stable complex with the macrocyclic NODA chelate and Al18F2þ. Al[18F]F-PSMA-BCH shows promising imaging capabilities for PCa with appropriate radiation exposure. Al[18F]F-PSMA-BCH is highly hydrophilic. The uptake of Al[18F]F-PSMA-BCH is dependent on PSMA levels in cells and tumors. Al[18F]F-PSMA-BCH accumulates in the kidneys and can be significantly blocked by ZJ-43(a PSMA inhibitor) because of the high hydrophilicity of Al[18F]F-PSMA-BCH and high PSMA levels in the kidneys. The maximum standard uptake values (SUVmax) for patients with high-risk PCa (GS ≥ 8) are significantly higher than those for patients with intermediate-risk PCa. This is similar to the findings of a previous study on [68Ga]Ga-PSMA-617 [52]. The SUVmax and SUVmean of Al[18F]F-PSMA-BCH in PCa lesions are significantly increased after 1 h and 2 h. Al[18F]F-PSMA-BCH is almost non-toxic and more economical for patients [41,52]. Al[18F]F-PSMA-11 is another PSMA radioligand, and its uptake in the kidneys is greatly reduced compared to [68Ga]Ga-PSMA-11 both at 1 h and 4 h. Meanwhile, Al[18F]F-PSMA-11 develops well in tumors with high PSMA expression and is influenced by its own molar activity. The higher the molar activity, the better the developmental effect. However, studies have found that the uptake of Al[18F]F-PSMA-11 is higher in bone and lower in PSMA-positive tumors than that of both [68Ga]Ga-PSMA-11 and [18F]F-PSMA-1007 [53,54].

99mTc-PSMA

Single-photon emission computed tomography CT (SPECT-CT) is a more cost-effective imaging method than PET-CT. Technetium-99m has better physical properties for gamma probe measurements, thus reducing the radiation exposure for both patients and medical personnel [55]. 99mTc-labeled PSMA ligands, including [99mTc]Tc-MIP-1404 and [99mTc]Tc-MIP-1405 (both of which utilize an imidazole with a carboxylate substitution), have favorable pharmacokinetic properties. Compared to [99mTc]Tc-MIP-1405, [99mTc]Tc-MIP-1404 has shown a greater uptake in the liver and spleen, owing to its higher ratio of carboxyl groups. The phase III clinical trials for [99mTc]Tc-MIP-1404, also known as Treforsta, have recently been completed [27]. Compared to [68Ga]PSMA, delayed SPECT-CT imaging using [99mTc]PSMA can ensure the choice of subsequent treatment strategies [56,57]. Furthermore, scintigraphy with [99mTc]MDP is limited by factors, such as low PSA levels, long PSA doubling times, and osteolytic lesions. Thus, many examination results may be equivocal. In this regard, [99mTc]PSMA is superior to MDP and more sensitive for the detection of visceral metastases [58,59].

89Zr-PSMA

Ligand internalization is a vital prerequisite for tracer accumulation in PCa lesions. Experimental data suggest that the internalization of PSMA ligands gradually increases over 24 h. Currently, the commonly used PSMA tracers have a short radioactive half-life, necessitating PET imaging within 3 h of injection. To overcome this limitation, a new ligand, 89Zr-labeled PSMA tracer ([89Zr]Zr-PSMA-Df) was explored. The long half-life of 89Zr (77 h) and the prolonged ligand internalization period allow image acquisition several days after the tracer injection. Compared to [68Ga]Ga-PSMA-11 and [18F]F-JK-PSMA-7, [89Zr]Zr-PSMA-Df shows a higher tumor-to-background ratio and increases the detection rate of metastatic lymph nodes near the ureter. Additionally, [89Zr]Zr-PSMA-Df can detect [68Ga]Ga-PSMA-11 or [18F]F-JK-PSMA-7 intake-free lesions, thus improving the detection rate (57%) of biochemically recurrent lesions [60].

Most PSMA ligands described above are small-molecule inhibitors of PSMA. An 85-Kd vesicle, IAB2M, has also been developed. This molecule is a de-immunizing monoclonal antibody that binds to the extracellular region of PSMA. The performance of [89Zr]Zr-df-IAB2M PET in detecting intra- and extra-prostatic lesions supports its use in clinical patient management for radical prostatectomy, pelvic lymphadenectomy, radiotherapy, and systemic therapy [61].

177Lu-PSMA

Radioligand therapy (RLT) targeting PSMA effectively controls PCa progression at the mCRPC stage [62,63]. 177Lu is a commonly used β radionuclide, and [177Lu]Lu-PSMA-617 and [177Lu]Lu-PSMA-I&T have been used to treat mCRPC [64]. In one study, PSA levels decreased by 65% following [177Lu]PSMA treatment, compared with a 37% PSA reduction after traditional treatment [65]. Although ligand studies for 177Lu labeling are ongoing, PSMA ligands with a DOTA-BnSCN structure show higher uptake and internalization, whereas better tumor uptake and retention have been demonstrated for bromophenyl-modified ligands and PSMA ligands with linear linkers [66,67]. RLT can cause xerostomia due to the high intake of [177Lu]PSMA by the salivary glands. Furthermore, the rapid excretion of [177Lu]PSMA from the body results in 177Lu-contaminated waste. To solve this problem, researchers proposed the introduction of Evans Blue dye into serum albumin to extend the circulation half-life of [177Lu]Lu-PSMA-617 in vivo to obtain [177Lu]Lu-EB-PSMA-617, which reached the maximum therapeutic effect at the lowest dose.

However, more than half of the patients with mCRPC treated with PSMA RLT ultimately failed treatment. The reasons for disease progression or lesion recurrence may include insufficient radiation dose delivery or radioresistance. One potential strategy for increasing tumor radiation doses is to extend the PSMA ligand circulation time. Tumor uptake was higher with PSMA-TO-1 than with PSMA-617 at all measured time points after 1h. The absorption of [177Lu]Lu-PSMA-TO-1 was 26 times greater than that of [177Lu]Lu-PSMA-617, suggesting long-term nephrotoxicity. Because PSMA-TO-1 is a long-circulating peptide, higher bone marrow doses are expected. While this higher dose could pose a greater risk of hematotoxicity and greater bone marrow exposure, dose delivery may be effective for treating bone marrow involvement [38].

225Ac-PSMA

225Ac is an α radionuclide with a longer physical half-life and higher linear energy than α nuclides, and can cause DNA double-strand breaks and cell death. [225Ac]PSMA is potentially highly effective against tumors. For example, lesions that do not respond to the [177Lu]PSMA treatment can be eradicated by using [225Ac]PSMA (Fig. 2). Additionally, [225Ac]PSMA treatment can benefit patients with mCRPC who has developed diffuse red bone marrow infiltration and fail to respond to other therapies [68,69]. Although, xerostomia is one of the adverse effects of [225Ac]PSMA treatment , the treatment is generally tolerated by the patients, and the incidence of xerostomia is reduced by applying ice [70,71]. Similar to 177Lu, 225Ac can be conjugated to PSMA ligands (PSMA-617, PSMA-I, PSMA-T, and PSMA-TO-1) and used for the treatment of mCRPC. The survival benefit conferred to mice treated with [225Ac]Ac-PSMA-TO-1 was statistically significant compared with that observed in mice treated with [225Ac]Ac-PSMA-617 [38].

Fig. 2. [68Ga]Ga-PSMA-11 positron emission tomography-computed tomography (PET-CT) scans of a patient with prostate cancer before and after radionuclide therapy. A: [68Ga]Ga-PSMA-11 PET-CT imaging before radionuclide therapy. B: Progression is observed after 2 cycles of treatment with beta-emitting [177Lu]Lu-PSMA-617. Promising treatment results after two (C) and three (D) cycles of treatment with α-emitting [225Ac]Ac-PSMA-617. Reprinted with permission from Kratochwil et al. [68] (J Nucl Med 2016;57:1941-1944; https://doi.org/10.2967/jnumed.116.178673).

Fig. 2

Many other nuclides are under development, such as [18F]F-PSMA-7Q [72], [111In]In-PSMA-617 [73], [68Ga]Ga-P16-093 [74], 68Ga-NGUL [75], and dual-targeted nuclides, such as [64Cu]Cu-FP-L1, which target both PSMA and fibroblast activator protein inhibitors (FAPI) [76].

NEPC Radionuclide Imaging and Therapy

PSMA-labeled radionuclides still play an important role in PCa. PCa cells express both adenocarcinoma and neuroendocrine differentiation markers. In contrast, this is not the case for NEPC, which expresses neuroendocrine differentiation only. Moreover, NEPC usually appears in the later stages of CRPC treatment and is characterized by small-cell morphology, downregulation of AR expression, and upregulation of neuroendocrine markers. In contrast to PCa adenocarcinoma, treatment-induced NEPC has aggressive tumor features, including large tumor size and a predisposition to bone and visceral metastases. Furthermore, treatment-induced NEPC has an inferior prognosis and is unresponsive to androgen deprivation therapy. The most common causes of NEPC development are the loss of the tumor suppressors RB1 and TP53 and the activation of oncogenic drivers, combined with significant epigenetic changes that further promote tumor proliferation and neuroendocrine lineage pathways [77]. In most NEPC cases, hormone depletion, p53 deletion, and lineage plasticity inhibit the PSMA gene, FOLH1. However, other molecular probes for NEPC differ from PSMA in prostate adenocarcinoma.

SSTR-2-Targeted Radionuclide Imaging and Therapy

There are five somatostatin receptor (SSTR) subtypes (SSTRs 1–5). Of these, SSTR2 is expressed specifically in neuroendocrine tumors (NETs). SSTR is a powerful target for NET-targeted therapy. NEPC cells show elevated expression of SSTR-2 and downregulation of PMSA [77,78,79]. Many studies have found that NEPC lesions show high radioactive uptake in SSTR-targeted PET [80] but no radioactive uptake in PSMA-targeted PET [81]. High-affinity ligands for SSTR, including TOC, NOC, and TATE, can be used to target NETs. 68Ga-labeled DOTA-TOC and DOTA-TATE have been widely used for the PET imaging of tumors. Additionally, peptide receptor radionuclide therapy (PRRT) with [90Y]Y-DOTA-TOC and [177Lu]Lu-DOTA-TATE can improve treatment efficacy for NETs [4,45].

CDCP1-Targeted Radionuclide Imaging and Therapy

Overexpression of the single-pass transmembrane protein, CUB domain-containing protein 1 (CDCP1), is significantly associated with phosphatase and tensin homolog deleted on chromosome ten (PTEN) gene loss and a more aggressive PCa phenotype. CDCP1 expression was detected in 90% of mCRPC biopsies. To quantify the number of CDCP1 receptors per cell, researchers have used 4A06, a monoclonal recombinant human antibody that recognizes the ectodomain of full-length or cleaved CDCP1. The tumor-autonomous expression of CDCP1 in mCRPC can be detected using [89Zr]4A06 PET. Moreover, [177Lu]4A06 RLT inhibits and eliminates mCRPC lesions [82]. Therefore, combining CDCP1-targeted RLT with standard treatment for mCRPC could be a potentially more efficacious clinical treatment strategy.

NT-20.3-Targeted Radionuclide Imaging

High expression levels of neurohypotensin receptor subtype 1 (NTR1) are associated with neuroendocrine differentiation in PCa, which makes NTR1 a potential target for NEPC imaging. Recent studies have reported positive NTR1 expression in 91.8% of PCa tissues, including all PSMA-negative tissues. [68Ga]Ga-DOTA-NT-20.3, can be used as a targeted radionuclide for the detection of NEPC because of its high affinity for NTR1 and favorable distribution and kinetics in the body [83]. In PC3 xenografts expressing NTR1, high-contrast [68Ga]Ga-DOTA-NT-20.3 images indicate the potential to detect low or neuroendocrine differentiation in PCa. Furthermore, NT-20.3 exhibits high stability and retention within tumors, which is conducive to its application in PRRT for mCRPC at later stages.

[18F]FDG-PET Imaging

Some high-grade aggressive NETs frequently lose SSTR expression. FDG is a glucose analog that shows a high level of uptake by cells with high glycolysis rates. [18F]FDG-PET is widely used to detect tumors. The degree of [18F]FDG uptake reflects the level of glucose metabolism in viable tumor cells, with highly aggressive malignancies showing higher levels of [18F]FDG uptake [45,84]. Studies have demonstrated that NEPC cells show increased glucose uptake owing to the increased expression of glucose transporters. More glucose is brought into the tumor cells and is phosphorylated by hexokinase [81]. Importantly, glucokinase levels are 5-fold higher in PSMA-negative tumors than in AR-positive tumors, leading to increased uptake and deposition of [18F]FDG within NEPC cells [81,85]. Research has found that eecurrent NEPC lesions show an increased [18F]FDG uptake and a decreased PSMA expression. These lesions are characterized by high metabolic activity, rapid progression, and poor prognosis [86,87]. Although [18F]-FDG-PET is inefficient in detecting PCa lesions, it can be beneficial in detecting NEPC lesions [88] (Fig. 3).

Fig. 3. Maximum intensity projection (MIP) of positron emission tomography (PET) and trans-axial fusion images of PET-computed tomography (CT) scans of a 53-year-old male with post-operative relapse of his prostate cancer. Al[18F]F-PSMA PET-CT shows no uptake in the recurrent lesions (A, D). [18F]FDG PET-CT shows multiple lesions with high uptake throughout the body (B, E). After six cycles of chemotherapy, [18F]FDG PET-CT demonstrates that multiple lesions are significantly reduced in size and radioactive uptake (C, F).

Fig. 3

Other Molecular Probes for PCa

Fibroblast Activator Protein Inhibitors

Fibroblast activator protein (FAP) is a 97 kDa type II transmembrane serine protease that is expressed at low or undetectable levels in normal tissues but at high levels in a variety of cancers, including 90% epithelial tumors [89,90]. Recently, quinoline-based FAPIs have been developed as promising imaging probes for various solid tumors, including PCa. FAP-targeted radionuclide imaging and treatment can overcome tumor heterogeneity and the limitations associated with insufficient PSMA expression. However, their application may be limited to highly differentiated PCa [86]. Additionally, FAPI-targeted RLT has shown therapeutic potential in PSMA-negative mCRPC [91].

FSPG

L-glutamate is an unnatural amino acid that is upregulated in many cancers and can reflect tumor redox status and antioxidant capacity, and predict resistance to chemotherapy [92,93]. The glutamate derivative, (S)-4-(3-18F-fluoropropyl)-L-glutamate (FSPG), is rapidly excreted through the kidneys and has low background activity, providing high contrast for tumor imaging [93]. [18F]-labeled FSPG ([18F]FSPG) has been used in clinical imaging of hepatocellular carcinoma, non-small cell lung cancer, PCa, and intracranial malignancies [94].

Gastrin-Releasing Peptide Receptor Antagonists

Although some NEPCs express little PSMA, they express GRPR. RM26 is a peptide skeleton-modified lactopin analog that serves as a high-affinity antagonist of GRPR, a member of the G protein-coupled receptor family of urotin receptors. Endogenous receptor expression is observed in the pancreas, whereas only low expression levels are detected in both normal and proliferative prostate tissues [7,95]. Studies have confirmed that [68Ga]RM26-PET showed high specific uptake in tumors and a high tumor-to-background ratio [96,97]. [68Ga]RM26-PET is of significant value for detecting primary and metastatic PCa lesions because GRPR expression is high in early-stage PCa. However, GRPR expression decreases as PCa progresses [98]. NOTA-DUPA-RM26 heterodimers that bind to both GRPR and PSMA are produced in PCa. These [68Ga]- and [111In]-labeled NOTA-DUPA-RM26 dimers can be used for simultaneous PSMA- and GRPR-targeted PET and SPECT imaging to improve the diagnostic accuracy of PCa [98,99].

NeoBOMB1 is a novel DOTA-coupled GRPR antagonist with a high affinity for GRPR and excellent in vivo stability. NeoBOMB1 can be labeled as [68Ga] and [177Lu]. Biodistribution studies with [68Ga]NeoBOMB1 have shown high tumor uptake, leading to a clear visualization of the tumor on PET-CT scans. These data suggest that [177Lu]NeoBOMB1 has strong specificity. It is also proven as a treatment for the currently known types of PCa, and has n a good tumor-kidney ratio [100].

Summary

Radionuclide-labeled PSMA ligands are currently the most commonly used nuclide drugs in clinical practice. These drugs have replaced traditional imaging methods, allowing accurate TNM staging of PCa and providing a basis for the selection of a suitable treatment plan. Tracers excreted via the hepatobiliary route can highlight the bladder and periurethral lesions more clearly. Furthermore, promising progress has been made in the treatment of advanced mCRPC using [177Lu]/[225Ac]-PSMA.

However, PSMA is not a perfect marker. There can be some physiological uptake of ligands and approximately 10% of primary PCa cases lack PSMA expression, which can arise from lesions with insufficient PSMA expression or from lesions developing into NEPC because of therapeutic AR resistance. Some NEPC targets have recently been used for NEPC imaging and treatment. Additionally, dual-targeting of PSMA and FAP by heterodimers has shown that combining different markers improved the specificity, sensitivity, and accuracy of PCa imaging and treatment. PRRT and RLT have opened new avenues for the treatment of NEPC. Furthermore, numerous radiopharmaceuticals and targeted molecular probes have continuously improved the detection rate of PCa lesions, and are being used for mCRPC, which will continue to be the focus of future research.

Acknowledgments

We thanks for the permissions from Springer, European Journal of Nuclear Medicine Molecular Imaging, Society of Nuclear Medicine and Molecular Imaging regarding permission to use figure/table in Kuten et al. [39], J Nucl Med 2020;61:527-532. We also thanks for the support from the Department of Urology and Pathology in the Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University. We thank James P. Mahaffey, PhD, from Liwen Bianji (Edanz) (www.liwenbianji.cn) for editing the English expression of this manuscript.

Footnotes

Conflicts of Interest: The authors have no potential conflicts of interest to disclose.

Author Contributions:
  • Conceptualization: Weijing Tao.
  • Funding acquisition: Weijing Tao.
  • Methodology: Tianshuo Yang.
  • Supervision: Weijing Tao.
  • Writing—original draft: Luyi Cheng.
  • Writing—review & editing: Jun Zhang, Feng Gao, Lingyun Yang.

Funding Statement: This study was funded by Huai’an Science and Technology Project (grant no. HAB202017 to Weijing Tao), and the innovation key talents Project of the hospital (grant no. ZC202208 to Weijing Tao).

References

  • 1.Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7–33. doi: 10.3322/caac.21708. [DOI] [PubMed] [Google Scholar]
  • 2.Dreyfuss AD, Ahn GS, Barsky AR, Gillman JA, Vapiwala N, Pantel AR. 18F-fluciclovine PET/CT in therapeutic decision making for prostate cancer: a large single-center practice-based analysis. Clin Nucl Med. 2021;46:187–194. doi: 10.1097/RLU.0000000000003444. [DOI] [PubMed] [Google Scholar]
  • 3.Tse BWC, Volpert M, Ratther E, Stylianou N, Nouri M, McGowan K, et al. Neuropilin-1 is upregulated in the adaptive response of prostate tumors to androgen-targeted therapies and is prognostic of metastatic progression and patient mortality. Oncogene. 2017;36:3417–3427. doi: 10.1038/onc.2016.482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bakht MK, Derecichei I, Li Y, Ferraiuolo RM, Dunning M, Oh SW, et al. Neuroendocrine differentiation of prostate cancer leads to PSMA suppression. Endocr Relat Cancer. 2018;26:131–146. doi: 10.1530/ERC-18-0226. [DOI] [PubMed] [Google Scholar]
  • 5.Vellky JE, Ricke WA. Development and prevalence of castration-resistant prostate cancer subtypes. Neoplasia. 2020;22:566–575. doi: 10.1016/j.neo.2020.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Li R, Ravizzini GC, Gorin MA, Maurer T, Eiber M, Cooperberg MR, et al. The use of PET/CT in prostate cancer. Prostate Cancer Prostatic Dis. 2018;21:4–21. doi: 10.1038/s41391-017-0007-8. [DOI] [PubMed] [Google Scholar]
  • 7.Lundmark F, Abouzayed A, Mitran B, Rinne SS, Varasteh Z, Larhed M, et al. Heterodimeric radiotracer targeting PSMA and GRPR for imaging of prostate cancer-optimization of the affinity towards PSMA by linker modification in murine model. Pharmaceutics. 2020;12:614. doi: 10.3390/pharmaceutics12070614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Langbein T, Weber WA, Eiber M. Future of theranostics: an outlook on precision oncology in nuclear medicine. J Nucl Med. 2019;60(Suppl 2):13S–19S. doi: 10.2967/jnumed.118.220566. [DOI] [PubMed] [Google Scholar]
  • 9.Ruschoff JH, Ferraro DA, Muehlematter UJ, Laudicella R, Hermanns T, Rodewald AK, et al. What’s behind (68)Ga-PSMA-11 uptake in primary prostate cancer PET? Investigation of histopathological parameters and immunohistochemical PSMA expression patterns. Eur J Nucl Med Mol Imaging. 2021;48:4042–4053. doi: 10.1007/s00259-021-05501-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ferraro DA, Rüschoff JH, Muehlematter UJ, Kranzbuhler B, Müller J, Messerli M, et al. Immunohistochemical PSMA expression patterns of primary prostate cancer tissue are associated with the detection rate of biochemical recurrence with (68)Ga-PSMA-11-PET. Theranostics. 2020;10:6082–6094. doi: 10.7150/thno.44584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Siow A, Kowalczyk R, Brimble MA, Harris PWR. Evolution of peptide-based prostate-specific membrane antigen (PSMA) inhibitors: an approach to novel prostate cancer therapeutics. Curr Med Chem. 2021;28:3713–3752. doi: 10.2174/0929867327666201006153847. [DOI] [PubMed] [Google Scholar]
  • 12.Afshar-Oromieh A, Sattler LP, Steiger K, Holland-Letz T, da Cunha ML, Mier W, et al. Tracer uptake in mediastinal and paraaortal thoracic lymph nodes as a potential pitfall in image interpretation of PSMA ligand PET/CT. Eur J Nucl Med Mol Imaging. 2018;45:1179–1187. doi: 10.1007/s00259-018-3965-8. [DOI] [PubMed] [Google Scholar]
  • 13.Parsi M, Desai MH, Desai D, Singhal S, Khandwala PM, Potdar RR. PSMA: a game changer in the diagnosis and treatment of advanced prostate cancer. Med Oncol. 2021;38:89. doi: 10.1007/s12032-021-01537-3. [DOI] [PubMed] [Google Scholar]
  • 14.Klingenberg S, Jochumsen MR, Ulhøi BP, Fredsøe J, Sørensen KD, Borre M, et al. (68)Ga-PSMA PET/CT for primary lymph node and distant metastasis NM staging of high-risk prostate cancer. J Nucl Med. 2021;62:214–220. doi: 10.2967/jnumed.120.245605. [DOI] [PubMed] [Google Scholar]
  • 15.Wester HJ, Schottelius M. PSMA-targeted radiopharmaceuticals for imaging and therapy. Semin Nucl Med. 2019;49:302–312. doi: 10.1053/j.semnuclmed.2019.02.008. [DOI] [PubMed] [Google Scholar]
  • 16.Wang F, Li Z, Feng X, Yang D, Lin M. Advances in PSMA-targeted therapy for prostate cancer. Prostate Cancer Prostatic Dis. 2022;25:11–26. doi: 10.1038/s41391-021-00394-5. [DOI] [PubMed] [Google Scholar]
  • 17.van Boxtel W, Lütje S, van Engen-van Grunsven ICH, Verhaegh GW, Schalken JA, Jonker MA, et al. (68)Ga-PSMA-HBED-CC PET/CT imaging for adenoid cystic carcinoma and salivary duct carcinoma: a phase 2 imaging study. Theranostics. 2020;10:2273–2283. doi: 10.7150/thno.38501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Okamoto I, Nokihara H, Nomura S, Niho S, Sugawara S, Horinouchi H, et al. Comparison of carboplatin plus pemetrexed followed by maintenance pemetrexed with docetaxel monotherapy in elderly patients with advanced nonsquamous non-small cell lung cancer: a phase 3 randomized clinical trial. JAMA Oncol. 2020;6:e196828. doi: 10.1001/jamaoncol.2019.6828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Domachevsky L, Goldberg N, Bernstine H, Nidam M, Groshar D. Quantitative characterisation of clinically significant intra-prostatic cancer by prostate-specific membrane antigen (PSMA) expression and cell density on PSMA-11. Eur Radiol. 2018;28:5275–5283. doi: 10.1007/s00330-018-5484-1. [DOI] [PubMed] [Google Scholar]
  • 20.Alberto M, Yim A, Papa N, Siva S, Ischia J, Touijer K, et al. Role of PSMA PET-guided metastases-directed therapy in oligometastatic recurrent prostate cancer. Front Oncol. 2022;12:929444. doi: 10.3389/fonc.2022.929444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rauscher I, Düwel C, Haller B, Rischpler C, Heck MM, Gschwend JE, et al. Efficacy, predictive factors, and prediction nomograms for (68)Ga-labeled prostate-specific membrane antigen-ligand positron-emission tomography/computed tomography in early biochemical recurrent prostate cancer after radical prostatectomy. Eur Urol. 2018;73:656–661. doi: 10.1016/j.eururo.2018.01.006. [DOI] [PubMed] [Google Scholar]
  • 22.Iglesias-Gato D, Thysell E, Tyanova S, Crnalic S, Santos A, Lima TS, et al. The proteome of prostate cancer bone metastasis reveals heterogeneity with prognostic implications. Clin Cancer Res. 2018;24:5433–5444. doi: 10.1158/1078-0432.CCR-18-1229. [DOI] [PubMed] [Google Scholar]
  • 23.Plichta KA, Graves SA, Buatti JM. Prostate-specific membrane antigen (PSMA) theranostics for treatment of oligometastatic prostate cancer. Int J Mol Sci. 2021;22:12095. doi: 10.3390/ijms222212095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Murthy V, Sonni I, Jariwala N, Juarez R, Reiter RE, Raman SS, et al. The role of PSMA PET/CT and PET/MRI in the initial staging of prostate cancer. Eur Urol Focus. 2021;7:258–266. doi: 10.1016/j.euf.2021.01.016. [DOI] [PubMed] [Google Scholar]
  • 25.Barbato F, Fendler WP, Rauscher I, Herrmann K, Wetter A, Ferdinandus J, et al. PSMA-PET for the assessment of metastatic hormone-sensitive prostate cancer volume of disease. J Nucl Med. 2021;62:1747–1750. doi: 10.2967/jnumed.121.262120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Dekalo S, Kuten J, Mintz I, Fahoum I, Gitstein G, Keizman D, et al. Preoperative 68Ga-PSMA PET/CT defines a subgroup of high-risk prostate cancer patients with favorable outcomes after radical prostatectomy and lymph node dissection. Prostate Cancer Prostatic Dis. 2021;24:910–916. doi: 10.1038/s41391-021-00347-y. [DOI] [PubMed] [Google Scholar]
  • 27.Pastorino S, Riondato M, Uccelli L, Giovacchini G, Giovannini E, Duce V, et al. Toward the discovery and development of PSMA targeted inhibitors for nuclear medicine applications. Curr Radiopharm. 2020;13:63–79. doi: 10.2174/1874471012666190729151540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Jeitner TM, Babich JW, Kelly JM. Advances in PSMA theranostics. Transl Oncol. 2022;22:101450. doi: 10.1016/j.tranon.2022.101450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Witkowska-Patena E, Mazurek A, Dziuk M. 68Ga-PSMA PET/CT imaging in recurrent prostate cancer: where are we now? Cent European J Urol. 2017;70:37–43. doi: 10.5173/ceju.2017.947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Zhang LL, Li WC, Xu Z, Jiang N, Zang SM, Xu LW, et al. (68)Ga-PSMA PET/CT targeted biopsy for the diagnosis of clinically significant prostate cancer compared with transrectal ultrasound guided biopsy: a prospective randomized single-centre study. Eur J Nucl Med Mol Imaging. 2021;48:483–492. doi: 10.1007/s00259-020-04863-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fendler WP, Calais J, Eiber M, Flavell RR, Mishoe A, Feng FY, et al. Assessment of 68Ga-PSMA-11 PET accuracy in localizing recurrent prostate cancer: a prospective single-arm clinical trial. JAMA Oncol. 2019;5:856–863. doi: 10.1001/jamaoncol.2019.0096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Fütterer JJ, Nagarajah J. Research highlight: (68)Ga-PSMA-11 PET imaging for pelvic nodal metastasis in prostate cancer. Korean J Radiol. 2022;23:293–294. doi: 10.3348/kjr.2021.0938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sonni I, Eiber M, Fendler WP, Alano RM, Vangala SS, Kishan AU, et al. Impact of (68)Ga-PSMA-11 PET/CT on staging and management of prostate cancer patients in various clinical settings: a prospective single-center study. J Nucl Med. 2020;61:1153–1160. doi: 10.2967/jnumed.119.237602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Karyağar S, Güven O, Karyağar SS, Arici S, Selvi O, Geredeli Ç, et al. Can 68Ga-PSMA PET/CT-derived prostate-specific membrane antigen expression parameters predict prostate-specific antigen response to enzalutamide treatment? Nucl Med Commun. 2021;42:1011–1016. doi: 10.1097/MNM.0000000000001431. [DOI] [PubMed] [Google Scholar]
  • 35.Meyrick DP, Asokendaran M, Skelly LA, Lenzo NP, Henderson A. The role of 68Ga-PSMA-I&T PET/CT in the pretreatment staging of primary prostate cancer. Nucl Med Commun. 2017;38:956–963. doi: 10.1097/MNM.0000000000000738. [DOI] [PubMed] [Google Scholar]
  • 36.Cytawa W, Seitz AK, Kircher S, Fukushima K, Tran-Gia J, Schirbel A, et al. (68)Ga-PSMA I&T PET/CT for primary staging of prostate cancer. Eur J Nucl Med Mol Imaging. 2020;47:168–177. doi: 10.1007/s00259-019-04524-z. [DOI] [PubMed] [Google Scholar]
  • 37.de Kouchkovsky I, Aggarwal R, Hope TA. Prostate-specific membrane antigen (PSMA)-based imaging in localized and advanced prostate cancer: a narrative review. Transl Androl Urol. 2021;10:3130–3143. doi: 10.21037/tau-20-1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Meyer C, Prasad V, Stuparu A, Kletting P, Glatting G, Miksch J, et al. Comparison of PSMA-TO-1 and PSMA-617 labeled with gallium-68, lutetium-177 and actinium-225. EJNMMI Res. 2022;12:65. doi: 10.1186/s13550-022-00935-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kuten J, Fahoum I, Savin Z, Shamni O, Gitstein G, Hershkovitz D, et al. Head-to-head comparison of (68)Ga-PSMA-11 with (18)F-PSMA-1007 PET/CT in staging prostate cancer using histopathology and immunohistochemical analysis as a reference standard. J Nucl Med. 2020;61:527–532. doi: 10.2967/jnumed.119.234187. [DOI] [PubMed] [Google Scholar]
  • 40.Piron S, Verhoeven J, Vanhove C, De Vos F. Recent advancements in (18)F-labeled PSMA targeting PET radiopharmaceuticals. Nucl Med Biol. 2022;106-107:29–51. doi: 10.1016/j.nucmedbio.2021.12.005. [DOI] [PubMed] [Google Scholar]
  • 41.Evangelista L, Maurer T, van der Poel H, Alongi F, Kunikowska J, Laudicella R, et al. [(68)Ga]Ga-PSMA versus [(18)F]PSMA positron emission tomography/computed tomography in the staging of primary and recurrent prostate cancer. A systematic review of the literature. Eur Urol Oncol. 2022;5:273–282. doi: 10.1016/j.euo.2022.03.004. [DOI] [PubMed] [Google Scholar]
  • 42.Sprute K, Kramer V, Koerber SA, Meneses M, Fernandez R, Soza-Ried C, 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. doi: 10.2967/jnumed.120.246363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Giesel FL, Knorr K, Spohn F, Will L, Maurer T, Flechsig P, et al. Detection efficacy of 18F-PSMA-1007 PET/CT in 251 patients with biochemical recurrence of prostate cancer after radical prostatectomy. J Nucl Med. 2019;60:362–368. doi: 10.2967/jnumed.118.212233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Giesel FL, Hadaschik B, Cardinale J, Radtke J, Vinsensia M, Lehnert W, et al. F-18 labelled PSMA-1007: biodistribution, radiation dosimetry and histopathological validation of tumor lesions in prostate cancer patients. Eur J Nucl Med Mol Imaging. 2017;44:678–688. doi: 10.1007/s00259-016-3573-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Iravani A, Parihar AS, Akhurst T, Hicks RJ. Molecular imaging phenotyping for selecting and monitoring radioligand therapy of neuroendocrine neoplasms. Cancer Imaging. 2022;22:25. doi: 10.1186/s40644-022-00465-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Pan KH, Wang JF, Wang CY, Nikzad AA, Kong FQ, Jian L, et al. Evaluation of 18F-DCFPyL PSMA PET/CT for prostate cancer: a meta-analysis. Front Oncol. 2020;10:597422. doi: 10.3389/fonc.2020.597422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sun J, Lin Y, Wei X, Ouyang J, Huang Y, Ling Z. Performance of 18F-DCFPyL PET/CT imaging in early detection of biochemically recurrent prostate cancer: a systematic review and meta-analysis. Front Oncol. 2021;11:649171. doi: 10.3389/fonc.2021.649171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hohberg M, Kobe C, Krapf P, Täger P, Hammes J, Dietlein F, et al. Biodistribution and radiation dosimetry of [(18)F]-JK-PSMA-7 as a novel prostate-specific membrane antigen-specific ligand for PET/CT imaging of prostate cancer. EJNMMI Res. 2019;9:66. doi: 10.1186/s13550-019-0540-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Morris MJ, Rowe SP, Gorin MA, Saperstein L, Pouliot F, Josephson D, et al. Diagnostic performance of (18)F-DCFPyL-PET/CT in men with biochemically recurrent prostate cancer: results from the CONDOR phase III, multicenter study. Clin Cancer Res. 2021;27:3674–3682. doi: 10.1158/1078-0432.CCR-20-4573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zlatopolskiy BD, Endepols H, Krapf P, Guliyev M, Urusova EA, Richarz R, et al. Discovery of (18)F-JK-PSMA-7, a PET probe for the detection of small PSMA-positive lesions. J Nucl Med. 2019;60:817–823. doi: 10.2967/jnumed.118.218495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Dietlein F, Hohberg M, Kobe C, Zlatopolskiy BD, Krapf P, Endepols H, et al. An (18)F-labeled PSMA ligand for PET/CT of prostate cancer: first-in-humans observational study and clinical experience with (18)F-JK-PSMA-7 during the first year of application. J Nucl Med. 2020;61:202–209. doi: 10.2967/jnumed.119.229542. [DOI] [PubMed] [Google Scholar]
  • 52.Liu T, Liu C, Xu X, Liu F, Guo X, Li N, et al. Preclinical evaluation and pilot clinical study of Al(18)F-PSMA-BCH for prostate cancer PET imaging. J Nucl Med. 2019;60:1284–1292. doi: 10.2967/jnumed.118.221671. [DOI] [PubMed] [Google Scholar]
  • 53.Piron S, Verhoeven J, De Coster E, Descamps B, Kersemans K, Pieters L, et al. Impact of the molar activity and PSMA expression level on [(18)F]AlF-PSMA-11 uptake in prostate cancer. Sci Rep. 2021;11:22623. doi: 10.1038/s41598-021-02104-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ioppolo JA, Nezich RA, Richardson KL, Morandeau L, Leedman PJ, Price RI. Direct in vivo comparison of [(18)F]PSMA-1007 with [(68)Ga]Ga-PSMA-11 and [(18)F]AlF-PSMA-11 in mice bearing PSMA-expressing xenografts. Appl Radiat Isot. 2020;161:109164. doi: 10.1016/j.apradiso.2020.109164. [DOI] [PubMed] [Google Scholar]
  • 55.Mix M, Schultze-Seemann W, von Büren M, Sigle A, Omrane MA, Grabbert MT, et al. (99m)Tc-labelled PSMA ligand for radio-guided surgery in nodal metastatic prostate cancer: proof of principle. EJNMMI Res. 2021;11:22. doi: 10.1186/s13550-021-00762-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Fallahi B, Khademi N, Karamzade-Ziarati N, Fard-Esfahani A, Emami-Ardekani A, Farzanefar S, et al. 99mTc-PSMA SPECT/CT versus 68Ga-PSMA PET/CT in the evaluation of metastatic prostate cancer. Clin Nucl Med. 2021;46:e68–e74. doi: 10.1097/RLU.0000000000003410. [DOI] [PubMed] [Google Scholar]
  • 57.Sergieva S, Mangaldgiev R, Dimcheva M, Nedev K, Zahariev Z, Robev B. SPECT-CT imaging with [99mTc]PSMA-T4 in patients with recurrent prostate cancer. Nucl Med Rev Cent East Eur. 2021;24:70–81. doi: 10.5603/NMR.2021.0018. [DOI] [PubMed] [Google Scholar]
  • 58.Kabunda J, Gabela L, Kalinda C, Aldous C, Pillay V, Nyakale N. Comparing 99mTc-PSMA to 99mTc-MDP in prostate cancer staging of the skeletal system. Clin Nucl Med. 2021;46:562–568. doi: 10.1097/RLU.0000000000003702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Werner P, Neumann C, Eiber M, Wester HJ, Schottelius M. [(99cm)Tc]Tc-PSMA-I&S-SPECT/CT: experience in prostate cancer imaging in an outpatient center. EJNMMI Res. 2020;10:45. doi: 10.1186/s13550-020-00635-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Dietlein F, Kobe C, Vázquez SM, Fischer T, Endepols H, Hohberg M, et al. An 89Zr-labeled PSMA tracer for PET/CT imaging of prostate cancer patients. J Nucl Med. 2022;63:573–583. doi: 10.2967/jnumed.121.262290. [DOI] [PubMed] [Google Scholar]
  • 61.Vlachostergios PJ, Niaz MJ, Thomas C, Christos PJ, Osborne JR, Margolis DJA, et al. Pilot study of the diagnostic utility of (89) Zr-df-IAB2M and (68) Ga-PSMA-11 PET imaging and multiparametric MRI in localized prostate cancer. Prostate. 2022;82:483–492. doi: 10.1002/pros.24294. [DOI] [PubMed] [Google Scholar]
  • 62.Barrio M, Fendler WP, Czernin J, Herrmann K. Prostate specific membrane antigen (PSMA) ligands for diagnosis and therapy of prostate cancer. Expert Rev Mol Diagn. 2016;16:1177–1188. doi: 10.1080/14737159.2016.1243057. [DOI] [PubMed] [Google Scholar]
  • 63.Rahbar K, Afshar-Oromieh A, Jadvar H, Ahmadzadehfar H. PSMA Theranostics: current status and future directions. Mol Imaging. 2018;17:1536012118776068. doi: 10.1177/1536012118776068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hennrich U, Eder M. [(177)Lu]Lu-PSMA-617 (Pluvicto(TM)): The first FDA-approved radiotherapeutical for treatment of prostate cancer. Pharmaceuticals (Basel) 2022;15:1292. doi: 10.3390/ph15101292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hofman MS, Emmett L, Sandhu S, Iravani A, Joshua AM, Goh JC, et al. [177Lu]Lu-PSMA-617 versus cabazitaxel in patients with metastatic castration-resistant prostate cancer (TheraP): a randomised, open-label, phase 2 trial. Lancet. 2021;397:797–804. doi: 10.1016/S0140-6736(21)00237-3. [DOI] [PubMed] [Google Scholar]
  • 66.Banerjee SR, Kumar V, Lisok A, Chen J, Minn I, Brummet M, et al. (177)Lu-labeled low-molecular-weight agents for PSMA-targeted radiopharmaceutical therapy. Eur J Nucl Med Mol Imaging. 2019;46:2545–2557. doi: 10.1007/s00259-019-04434-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Schuchardt C, Zhang J, Kulkarni HR, Chen X, Müller D, Baum RP. Prostate-specific membrane antigen radioligand therapy using (177)Lu-PSMA I&T and (177)Lu-PSMA-617 in patients with metastatic castration-resistant prostate cancer: comparison of safety, biodistribution, and dosimetry. J Nucl Med. 2022;63:1199–1207. doi: 10.2967/jnumed.121.262713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kratochwil C, Bruchertseifer F, Giesel FL, Weis M, Verburg FA, Mottaghy F, et al. 225Ac-PSMA-617 for PSMA-targeted alpha-radiation therapy of metastatic castration-resistant prostate cancer. J Nucl Med. 2016;57:1941–1944. doi: 10.2967/jnumed.116.178673. [DOI] [PubMed] [Google Scholar]
  • 69.Alam MR, Singh SB, Thapaliya S, Shrestha S, Deo S, Khanal K. A review of 177Lutetium-PSMA and 225Actinium-PSMA as emerging theranostic agents in prostate cancer. Cureus. 2022;14:e29369. doi: 10.7759/cureus.29369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sanli Y, Kuyumcu S, Simsek DH, Büyükkaya F, Civan C, Isik EG, et al. 225Ac-prostate-specific membrane antigen therapy for castration-resistant prostate cancer: a single-center experience. Clin Nucl Med. 2021;46:943–951. doi: 10.1097/RLU.0000000000003925. [DOI] [PubMed] [Google Scholar]
  • 71.Kratochwil C, Bruchertseifer F, Rathke H, Bronzel M, Apostolidis C, Weichert W, et al. Targeted alpha-therapy of metastatic castration-resistant prostate cancer with (225)Ac-PSMA-617: dosimetry estimate and empiric dose finding. J Nucl Med. 2017;58:1624–1631. doi: 10.2967/jnumed.117.191395. [DOI] [PubMed] [Google Scholar]
  • 72.Liu Y, Zhang X, Liu J, Zhang J, Xu B. Prospective intraindividual comparison of 18F-PSMA-7Q and 18F-DCFPyL PET/CT in patients with newly diagnosed prostate cancer. Nucl Med Commun. 2022;43:725–730. doi: 10.1097/MNM.0000000000001564. [DOI] [PubMed] [Google Scholar]
  • 73.Mix M, Reichel K, Stoykow C, Bartholomä M, Drendel V, Gourni E, et al. Performance of (111)In-labelled PSMA ligand in patients with nodal metastatic prostate cancer: correlation between tracer uptake and histopathology from lymphadenectomy. Eur J Nucl Med Mol Imaging. 2018;45:2062–2070. doi: 10.1007/s00259-018-4094-0. [DOI] [PubMed] [Google Scholar]
  • 74.Green MA, Hutchins GD, Bahler CD, Tann M, Mathias CJ, Territo W, et al. [(68)Ga]Ga-P16-093 as a PSMA-targeted PET radiopharmaceutical for detection of cancer: initial evaluation and comparison with [(68)Ga]Ga-PSMA-11 in prostate cancer patients presenting with biochemical recurrence. Mol Imaging Biol. 2020;22:752–763. doi: 10.1007/s11307-019-01421-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Suh M, Ryoo HG, Kang KW, Jeong JM, Jeong CW, Kwak C, et al. Phase I clinical trial of prostate-specific membrane antigen-targeting (68)Ga-NGUL PET/CT in healthy volunteers and patients with prostate cancer. Korean J Radiol. 2022;23:911–920. doi: 10.3348/kjr.2022.0176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Boinapally S, Lisok A, Lofland G, Minn I, Yan Y, Jiang Z, et al. Hetero-bivalent agents targeting FAP and PSMA. Eur J Nucl Med Mol Imaging. 2022;49:4369–4381. doi: 10.1007/s00259-022-05933-3. [DOI] [PubMed] [Google Scholar]
  • 77.Yamada Y, Beltran H. Clinical and biological features of neuroendocrine prostate cancer. Curr Oncol Rep. 2021;23:15. doi: 10.1007/s11912-020-01003-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Parida GK, Tripathy S, Datta Gupta S, Singhal A, Kumar R, Bal C, et al. Adenocarcinoma prostate with neuroendocrine differentiation: potential utility of 18F-FDG PET/CT and 68Ga-DOTANOC PET/CT over 68Ga-PSMA PET/CT. Clin Nucl Med. 2018;43:248–249. doi: 10.1097/RLU.0000000000002013. [DOI] [PubMed] [Google Scholar]
  • 79.Arora K, Barbieri CE. Molecular subtypes of prostate cancer. Curr Oncol Rep. 2018;20:58. doi: 10.1007/s11912-018-0707-9. [DOI] [PubMed] [Google Scholar]
  • 80.Kitajima K, Yamamoto S, Ikeda M, Yamasaki T, Kawanaka Y, Komoto H, et al. Pelvic MRI, FDG-PET/CT, and somatostatin receptor scintigraphy findings of treatment-related neuroendocrine-differentiated prostate cancer. Case Rep Oncol. 2021;14:397–402. doi: 10.1159/000511070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Bakht MK, Lovnicki JM, Tubman J, Stringer KF, Chiaramonte J, Reynolds MR, et al. Differential expression of glucose transporters and hexokinases in prostate cancer with a neuroendocrine gene signature: a mechanistic perspective for (18)F-FDG imaging of PSMA-suppressed tumors. J Nucl Med. 2020;61:904–910. doi: 10.2967/jnumed.119.231068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Zhao N, Chopra S, Trepka K, Wang YH, Sakhamuri S, Hooshdaran N, et al. CUB domain-containing protein 1 (CDCP1) is a target for radioligand therapy in castration-resistant prostate cancer, including PSMA null disease. Clin Cancer Res. 2022;28:3066–3075. doi: 10.1158/1078-0432.CCR-21-3858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Wu W, Yu F, Zhang P, Bu T, Fu J, Ai S, et al. (68)Ga-DOTA-NT-20.3 neurotensin receptor 1 positron emission tomography imaging as a surrogate for neuroendocrine differentiation of prostate cancer. J Nucl Med. 2022;63:1394–1400. doi: 10.2967/jnumed.121.263132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wang B, Liu C, Wei Y, Meng J, Zhang Y, Gan H, et al. A Prospective Trial of (68)Ga-PSMA and (18)F-FDG PET/CT in nonmetastatic prostate cancer patients with an early PSA progression during castration. Clin Cancer Res. 2020;26:4551–4558. doi: 10.1158/1078-0432.CCR-20-0587. [DOI] [PubMed] [Google Scholar]
  • 85.Zhou X, Li Y, Jiang X, Wang X, Chen S, Shen T, et al. Intra-individual comparison of 18F-PSMA-1007 and 18F-FDG PET/CT in the evaluation of patients with prostate cancer. Front Oncol. 2020;10:585213. doi: 10.3389/fonc.2020.585213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Khreish F, Rosar F, Kratochwil C, Giesel FL, Haberkorn U, Ezziddin S. Positive FAPI-PET/CT in a metastatic castration-resistant prostate cancer patient with PSMA-negative/FDG-positive disease. Eur J Nucl Med Mol Imaging. 2020;47:2040–2041. doi: 10.1007/s00259-019-04623-x. [DOI] [PubMed] [Google Scholar]
  • 87.Shen K, Liu B, Zhou X, Ji Y, Chen L, Wang Q, et al. The evolving role of (18)F-FDG PET/CT in diagnosis and prognosis prediction in progressive prostate cancer. Front Oncol. 2021;11:683793. doi: 10.3389/fonc.2021.683793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Chen R, Wang Y, Shi Y, Zhu Y, Xu L, Huang G, et al. Diagnostic value of (18)F-FDG PET/CT in patients with biochemical recurrent prostate cancer and negative (68)Ga-PSMA PET/CT. Eur J Nucl Med Mol Imaging. 2021;48:2970–2977. doi: 10.1007/s00259-021-05221-6. [DOI] [PubMed] [Google Scholar]
  • 89.Kesch C, Yirga L, Dendl K, Handke A, Darr C, Krafft U, et al. High fibroblast-activation-protein expression in castration-resistant prostate cancer supports the use of FAPI-molecular theranostics. Eur J Nucl Med Mol Imaging. 2021;49:385–389. doi: 10.1007/s00259-021-05423-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kratochwil C, Flechsig P, Lindner T, Abderrahim L, Altmann A, Mier W, et al. (68)Ga-FAPI PET/CT: tracer uptake in 28 different kinds of cancer. J Nucl Med. 2019;60:801–805. doi: 10.2967/jnumed.119.227967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Isik EG, Has-Simsek D, Sanli O, Sanli Y, Kuyumcu S. Fibroblast activation protein-targeted PET imaging of metastatic castration-resistant prostate cancer compared with 68Ga-PSMA and 18F-FDG PET/CT. Clin Nucl Med. 2022;47:e54–e55. doi: 10.1097/RLU.0000000000003837. [DOI] [PubMed] [Google Scholar]
  • 92.Park SY, Na SJ, Kumar M, Mosci C, Wardak M, Koglin N, et al. Clinical evaluation of (4S)-4-(3-[(18)F]Fluoropropyl)-L-glutamate ((18)F-FSPG) for PET/CT imaging in patients with newly diagnosed and recurrent prostate cancer. Clin Cancer Res. 2020;26:5380–5387. doi: 10.1158/1078-0432.CCR-20-0644. [DOI] [PubMed] [Google Scholar]
  • 93.Edwards R, Greenwood HE, McRobbie G, Khan I, Witney TH. Robust and facile automated radiosynthesis of [(18)F]FSPG on the GE FASTlab. Mol Imaging Biol. 2021;23:854–864. doi: 10.1007/s11307-021-01609-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Shih KT, Huang YY, Yang CY, Cheng MF, Tien YW, Shiue CY, et al. Synthesis and analysis of 4-(3-fluoropropyl)-glutamic acid stereoisomers to determine the stereochemical purity of (4S)-4-(3-[18F]fluoropropyl)-L-glutamic acid ([18F]FSPG) for clinical use. PLoS One. 2020;15:e0243831. doi: 10.1371/journal.pone.0243831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Mitran B, Rinne SS, Konijnenberg MW, Maina T, Nock BA, Altai M, et al. Trastuzumab cotreatment improves survival of mice with PC-3 prostate cancer xenografts treated with the GRPR antagonist (177) Lu-DOTAGA-PEG2 -RM26. Int J Cancer. 2019;145:3347–3358. doi: 10.1002/ijc.32401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Fox JJ, Gavane SC, Blanc-Autran E, Nehmeh S, Gönen M, Beattie B, et al. Positron emission tomography/computed tomography-based assessments of androgen receptor expression and glycolytic activity as a prognostic biomarker for metastatic castration-resistant prostate cancer. JAMA Oncol. 2018;4:217–224. doi: 10.1001/jamaoncol.2017.3588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Cheng S, Lang L, Wang Z, Jacobson O, Yung B, Zhu G, et al. Positron emission tomography imaging of prostate cancer with Ga-68-labeled gastrin-releasing peptide receptor agonist BBN7-14 and antagonist RM26. Bioconjug Chem. 2018;29:410–419. doi: 10.1021/acs.bioconjchem.7b00726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Mitran B, Varasteh Z, Abouzayed A, Rinne SS, Puuvuori E, De Rosa M, et al. Bispecific GRPR-antagonistic anti-PSMA/GRPR heterodimer for PET and SPECT diagnostic imaging of prostate cancer. Cancers (Basel) 2019;11:1371. doi: 10.3390/cancers11091371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Abouzayed A, Tano H, Nagy Á, Rinne SS, Wadeea F, Kumar S, et al. Preclinical evaluation of the GRPR-targeting antagonist RM26 conjugated to the albumin-binding domain for GRPR-targeting therapy of cancer. Pharmaceutics. 2020;12:977. doi: 10.3390/pharmaceutics12100977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Dalm SU, Bakker IL, de Blois E, Doeswijk GN, Konijnenberg MW, Orlandi F, et al. 68Ga/177Lu-NeoBOMB1, a novel radiolabeled GRPR antagonist for theranostic use in oncology. J Nucl Med. 2017;58:293–299. doi: 10.2967/jnumed.116.176636. [DOI] [PubMed] [Google Scholar]

Articles from Korean Journal of Radiology are provided here courtesy of Korean Society of Radiology

RESOURCES