Next Article in Journal
Marfan Syndrome: Enhanced Diagnostic Tools and Follow-up Management Strategies
Previous Article in Journal
Performance Analysis of Segmentation and Classification of CT-Scanned Ovarian Tumours Using U-Net and Deep Convolutional Neural Networks
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Up-to-Date Imaging and Diagnostic Techniques for Prostate Cancer: A Literature Review

Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Diagnostics 2023, 13(13), 2283; https://doi.org/10.3390/diagnostics13132283
Submission received: 18 May 2023 / Revised: 29 June 2023 / Accepted: 30 June 2023 / Published: 5 July 2023
(This article belongs to the Section Medical Imaging and Theranostics)

Abstract

:
Prostate cancer (PCa) faces great challenges in early diagnosis, which often leads not only to unnecessary, invasive procedures, but to over-diagnosis and treatment as well, thus highlighting the need for modern PCa diagnostic techniques. The review aims to provide an up-to-date summary of chronologically existing diagnostic approaches for PCa, as well as their potential to improve clinically significant PCa (csPCa) diagnosis and to reduce the proliferation and monitoring of PCa. Our review demonstrates the primary outcomes of the most significant studies and makes comparisons across the diagnostic efficacies of different PCa tests. Since prostate biopsy, the current mainstream PCa diagnosis, is an invasive procedure with a high risk of post-biopsy complications, it is vital we dig out specific, sensitive, and accurate diagnostic approaches in PCa and conduct more studies with milestone findings and comparable sample sizes to validate and corroborate the findings.

1. Introduction

Prostate cancer (PCa) is one of the most prevalent diagnosed cancers globally with 1,276,106 new cases in 2018, and a prominent reason for tumor-related mortality in men [1,2]. The incidence increases with each decade of age, and therefore, 59% of men over 79 years of age have suffered from PCa [3]. Generally, most of the prostate cancers are indolent in nature, but others are invasive and are diagnosed either when already metastasized or rapidly spread beyond the prostate, resulting in morbidity and potential prostate cancer-specific mortality [4].
PCa mortality has lowered in the past decades, mainly due to the widespread application of preliminary exams [5]. Transrectal/transperineal prostate biopsy is the current mainstream prostate cancer diagnosis technique in most areas of the world, one which is conducted for a raised prostate specific antigen (PSA) value and/or abnormal findings on digital-rectal examination [6]. Established screening methods based on PSA level could significantly improve the rates of early diagnosis, and almost 90% of PCa can be localized clinically at the time of its diagnosis [7]. PSA screening is currently suggested for all males over 50 years, based on the European Society of Medical Oncology (ESMO) recommendations from 2020. Nevertheless, secreted by prostate epithelial cells, PSA is considered as an organ-specific rather than a cancer-specific marker. Because of its organ-specific nature, preoperative serum PSA has been considered as a predictor for tumor volume in PCa patients undergoing radical prostatectomy (RP). In recent decades, early detection of PCa and its clinical management have become debatable topics, because the current primary non-invasive prostate cancer screening methods have resulted in an unsatisfactory amount of unnecessary prostate biopsy cases. It is commonly well-accepted that clinically significant PCa or high-grade PCa (GS (Gleason score) ≥ 7) benefit the most from treatment, which includes either radiotherapy or radical surgery, and thus the risk of overdiagnosis and overtreatment of indolent PCa restricts PSA implementation at a population level [8,9]. Serum total PSA level could be abnormally increased due to benign prostatic hyperplasia, infection, etc. [10]. On the other hand, data from the Prostate Cancer Prevention Trial (PCPT) highlighted that 14.9% of PCa in men occurs with PSA levels lower than 4.0 ng/mL burden and Gleason scores of seven or higher [11]. Recent studies that have applied an exacting reference criterion have demonstrated that over half of men with clinically significant prostate cancer were incorrectly diagnosed when exposed to transrectal ultrasound (TRUS) biopsy [7]. Biopsies are an invasive procedure that has a high risk of post-biopsy complications, including per rectal bleeding, hematuria, and sepsis. Therefore, it is necessary to find a new and effective approach in order to diagnose clinically significant PCa (csPCa).
In this literature review, we discuss the indications and imaging techniques for prostate biopsy, summarize recent data concerning the diagnostic performance of several modern techniques, and compare the diagnostic and prognostic utility of prostate cancer tests.

2. Discussion

2.1. Conventional Imaging

For local lesions, ultrasound has the advantage of being non-ionizing and repeatable. The TRUS probe, which is in close proximity to the prostate gland, provides clearer images compared to computed tomography (CT) or magnetic resonance imaging (MRI), and has thus become a commonly utilized imaging diagnostic technique for prostate cancer. Currently, the ultrasound methods mainly include two- and three-dimensional TRUS, doppler ultrasound, ultrasound quantification, and acoustic radiation force impulse imaging. Two-dimensional (2D) ultrasound is a classic technique for prostate imaging, but it can only detect a small number of PCa cases (11~35%) [12]. Only 17~57% of the hypoechoic nodules displayed in the 2D ultrasound images are malignant [13], indicating poor sensitivity and specificity. However, due to the clear imaging and easy operation, it is still the preferred method for guiding systematic prostate biopsy in clinical practice. Three-dimensional (3D) ultrasound enables axial and sagittal imaging, and coronal and three-dimensional images are reconstructed by computer. Sedelaar et al. noted that, compared with 2D ultrasound, 3D ultrasound has increased sensitivity: 74% vs. 85%; but this sensitivity is associated with a decrease in specificity: 52% vs. 41% [14]. Mitterberger et al. discovered that the sensitivity and specificity of 3D ultrasound for extraprostatic lesions were 84% and 96%, respectively, and among the 16 patients, 14 were classified as T3b, indicating its role in clinical staging of PCa [15]. In recent years, several ultrasound quantification techniques have emerged for the diagnosis of prostate lesions, including the automated urologic diagnostic expert system (AUDEX), computer-aided transrectal ultrasound (C-TRUS) technology, and HistoScanning. Simmons et al. performed a comparative analysis of HistoScanning ultrasound results and pathology following RP for prostate cancer, indicating a sensitivity and specificity of 90% and 72%, respectively, for HistoScanning [16]. Doppler ultrasound enables the visualization of blood flow and its velocity in PCa, thereby reflecting the extent of PCa proliferation and its malignancy [17]. It encompasses two modes: color Doppler flow imaging (CDFI) and power Doppler ultrasonography (PDU). Zhao et al.’s study suggested a significant improvement in the cancer detection rate (CDR) of PDU compared to TRUS for patients with a PSA greater than 10 ng/mL [18]. The aforementioned techniques have achieved a certain success in visualizing prostate cancer. However, given the existence of interobserver bias, ultrasound is still mainly used as the guidance for prostate biopsy.
Regarding metastases, conventional imaging methods mainly include CT, bone scintigraphy (BS), and whole-body MRI (WB MRI). CT is of limited value in detecting intraprostatic lesion and local staging, but of use in metastases of lymph node or bone. CT or MRI nodal staging relies on assessment of lymph node morphological criteria such as size or shape [19]. The metastatic node may be of normal size, while the enlargement is possibly due to reactive hyperplasia caused by infection or inflammatory reaction. However, AUA guidelines still recommend CT for intermediate- to high-risk PCa patients, because of its easy accessibility and low cost. A radionuclide BS following the injection of a technetium-99m (99mTc) tracer is currently the standard and most widely used method of evaluating bone metastases of intermediate- to high-risk PCa [20,21]. A meta-analysis comparing choline-PET/CT, MRI, SPECT, and BS for diagnosing bone metastases showed a pooled sensitivity of 79% (95% CI: 73–83%), and a specificity of 82% (95% CI: 78–85%), respectively, at patient level for BS [22]. However, with the emergence of new techniques such as 68Ga-PSMA-PET/CT, it is expected that CT and BS are to be gradually phased out. Pasoglou et al. indicated that WB MRI showed higher sensitivity and specificity than did combined BS [23]. WB MRI provides high soft-tissue contrast and can display anatomical details with great precision, while avoiding ionizing radiation. It is noteworthy that existing research suggests that the sensitivity of WB MRI may be slightly lower than that of PET [24,25]. However, more data is needed, especially in evaluating the potential complementary roles of WB MRI and PET.

2.2. mpMRI

The hypoechoic lesions on TRUS are not specific to PCa; thus, the accuracy of TRUS in guiding prostate biopsy depends on the operator’s knowledge and experience [26,27]. Since MRI-guided prostate biopsy was first conducted by D’Amico in 2000 [28], it has been proven, through high-quality research, to detect more csPCa with fewer biopsy cores than does system biopsy [29]. The standard multiparametric magnetic resonance imaging (mpMRI) protocol is the combination of multiple planars, including T2-weighted imaging (T2WI), diffusion-weighted imaging (DWI), and dynamic contrast-enhanced (DCE) sequences [30]. mpMRI is assessed through the Prostate Imaging-Reporting and Data System (PI-RADS) and is currently revolutionizing the PCa diagnostic pathway [29]. Under the guidance of mpMRI, radiologists can detect, score, and stage lesions that may correspond to csPCa, the statuses of which can later be verified through histopathological grading [7]. Previous study has proved that mpMRI is a promising tool, with sensitivity ranged between 44% and 93% and specificity ranged between 38% and 94% [31]. Wide variations in sensitivity and specificity can be explained by different acquisition protocols, different thresholds being applied, different reference criteria used, biopsy inaccuracies, and the varied experience levels of the radiologists with PI-RADS v2. Multiple guidelines have recommended the routine application of mpMRI in biopsy-naïve patients with suspected PCa, and there is an ongoing implementation process in clinical practice [32].
Patients with a previous negative biopsy could also benefit from mpMRI as a method for omitting unnecessary repeat biopsy. Oishi et al. indicated that men with negative mpMRI, a previously negative biopsy, and a PSA density (PSAd) lower that 0.15 ng/mL/cc can safely omit rebiopsy [33]. The EAU/EANM/ESTRO/ESUR/SIOG guidelines strongly suggested conducting the mpMRI before repeat biopsy to select a target lesion in repeat biopsy settings [20]. All in all, 64% of urologists thought mpMRI was useful in detecting PCa for biopsy-naïve men, while 97% regarded it as valuable in men with a negative biopsy [34].
Nevertheless, significant interobserver variability remains to be a non-ignorable drawback for MRI [35], leading, in the literature, to overall heterogeneous findings for accuracy [36]. A multicenter, multi-reader study with six expert prostate radiologists detected moderate reproducibility for PIRADS version 2, but also found “considerable inter-reader variation” [37]. Even so, radiologists should have the requisite training, experience, and clinical volumes to routinely read prostate MRI images to maintain adequate expertise. In recent years, efforts have been devoted to exploring whether mpMRI could allow better evaluation of PCa aggressiveness, through both conventional statistics metrics [38,39] and higher-order texture features based on T2WI and apparent diffusion coefficient (ADC) maps [40,41,42]. Several important characteristics, including tumor grade and size, have been proved to affect conspicuity on mpMRI; tumor location and its association with mpMRI visibility remain undetermined facets of this area [43]. Therefore, it is necessary to investigate additional techniques for adequate management of the technique.
Bi-parametric (bp) MRI (a combination of T2WI and DWI) has also been introduced as a substitute for mpMRI with gadolinium-enhanced sequences, reducing the costs and potential side effects of gadolinium-based contrast agents [30,44,45]. As of yet, multiple studies have shown that the application of bpMRI protocols would not significantly decrease PCa detection rates, and that it is comparable to mpMRI protocols [46,47]. Based on PI-RADS V2.1, the role of the DCE sequence is only helpful for score 3 lesions in the peripheral zone (PZ) [48]. Nevertheless, evidence on their diagnostic performance is insufficient, and potential limitations of abbreviated protocols (such as an increased number of equivalent findings) should be investigated. Besides, DCE-MRI is itself a fairly applicable strategy to show neovascularization, and one normally applied to acquire kinetic information of image intensity enhancement. Whether the role of DCE can be replaced by other parameters is a proposition that still requires high quality research for proof.

2.3. Combination with Biomarkers

When considering advancements in the MRI pathway, it is apparent that the overall detection rate for csPCa, specifically defined as ISUP Grade Group (GG) 2, is approximately 38–40% [7,29,49]. This rate may seem relatively low, primarily due to the considerably lower detection rate observed for PI-RADS 3 lesions (12% in the PRECISION trial) compared to PI-RADS 4/5 lesions (60% and 83%, respectively) [29]. Studies have indicated that forgoing biopsies in patients who exhibit no lesions on an MRI could potentially result in missing approximately 5–11% of all incidences of csPCa [50,51]. Conversely, conducting biopsies in all patients with equivocal MRI findings (PI-RADS 3 lesions considered positive by guidelines [52]), may lead to a csPCa diagnosis in approximately 3–50% of these patients [50,51,53,54]. PI-RADS 3 is considered critical for several reasons. Firstly, the detection rate for PI-RADS 3 lesions remains relatively low, leading to a higher likelihood of men undergoing unnecessary biopsies. Secondly, the interpretation of PI-RADS 3 lesions can vary depending on the experience of the radiologist, resulting in inconsistencies in the number of lesions categorized as PI-RADS 3. In such circumstances, laboratory diagnostic biomarkers can play a role complementary to that of MRI in the diagnosis of csPCa.
According to a study by Friesbie et al., the implementation of a PSAd threshold of ≥0.1 ng/mL/cc demonstrated an improvement in the detection of csPCa, by 7% for PI-RADS 3, 17% for PI-RADS 4, and 15% for PI-RADS 5, respectively, on a per-patient basis [55]. The IMRIE study retrospectively analyzed 2642 men and showed that incorporating the standard PSAd threshold of ≥0.15 ng/mL/cc into the MRI pathway resulted in increased sensitivity and negative predictive value (NPV) for GG ≥ 2 (ranging from 87.3% to 96.6% and from 87.5% to 90.6%, respectively). Moreover, the utilization of a PSAd of 0.12 ng/mL/mL further improved sensitivity and NPV [56]. Preliminary studies have indicated the potential benefits of incorporating PSAd into the decision-making process for repeat biopsy in cases of active surveillance or during follow-up for men with a negative targeted biopsy [57].
Besides PSAd, other advanced laboratory biomarkers, such as 4Kscore and the prostate health index (PHI), have been utilized in conjunction with mpMRI to further augment the detection of csPCa. The 4Kscore test encompasses the assessment of four kallikreins (total PSA (tPSA), free PSA (fPSA), intact PSA, and human kallikrein 2 (hK2)), in conjunction with age, findings from digital rectal examination (DRE), and a record of previous prostate biopsy. In a retrospective series conducted by Wagasker et al., a nomogram was proposed combining 4Kscore with mpMRI, one which demonstrated promising results with area under the curve (AUC) values of 0.84 for any PCa, 0.88 for csPCa, and 0.86 for ≥GG3 PCa [58]. PHI is a biomarker which combines tPSA, percentage fPSA, and −2proPSA. Nomograms developed by Siddiqui et al. incorporating serum biomarker data (PHI, %fPSA, or tPSA) and mpMRI along with other clinical variables have demonstrated high accuracy in both training cohorts and independent cohorts. In the independent validation cohort, a nomogram for ≥GG2 PCa using PHI as a biomarker resulted in a reduction of 39.1% unnecessary biopsies (143/366) while only missing 0.8% of csPCa (1/124), with a biopsy threshold of 20% probability of csPCa. Indeed, these biomarkers are readily available and do not involve further tests combined with imaging. However, it is worth noting that despite their potential, these biomarkers may still be underutilized in clinical practice, and further studies are necessary.

2.4. Fusion Targeted Biopsy

Recent trends and evidence advocate pre-biopsy MRI with selective targeting of suspected malignant lesions under the guidance of MRI/ultrasound (US) and a targeted biopsy (TB) approach for its advantage to elevate the detection rate of csPCa, with a reduction in the overdiagnosis of clinically insignificant cancers [7,29]. This strategy allows for lesion-directed mpMRI TB and optimal planning of a biopsy. Through this approach, men who had positive results on the mpMRI underwent an MRI-targeted biopsy with the application of real-time ultrasonographic guidance, a technique which could allow MRI targets to be visualized on the ultrasound [29]. Recent research has proved that mpMRI and mpMRI/TRUS fusion targeted biopsy have demonstrated good accuracy in the diagnosis of csPCa, especially for the cancer located at the anterior zone of the gland [59]. As the PRECISION study indicated, MRI-ultrasound (US) fusion biopsy significantly outperforms systematic biopsy regarding CDR: 38% vs. 26% with p = 0.005 [29]. Generally fusion prostate biopsy (FPB) can be carried out in the form of TB, which is based only on acquiring specimens from suspicious lesions, or in combined biopsy (CB), in which 10- to 12-core standard biopsy (SB) is performed in addition to TB [60]. During CB, as expected, the quantity of cores sampled per target and the total biopsy time are increased [61]. Thus, the current controversy is centered on the question of whether one should continue doing the systematic biopsy along with MRI guided biopsy.
The findings of J.P. Radtke et al. prove that a combined TB+SB provides improved csPCa detection rates over either systematic or MRI-targeted biopsy or mpMRI alone, and that 97% of csPCa incidence has been detected by combined TB + SB [62]. However, this strategy is related to an increased quantity of biopsy cores, and frequently, with increased detection of indolent disease [63,64], and we are still far from safely selecting patients who might benefit from MRI-TB alone, relying on the combination of patient characteristics and mpMRI parameters. Therefore, the combination of MRI-TB and TRUS-guided biopsy (TRUS-Bx) should strongly be recommended as the best available approach for reducing the risk of csPCa misdiagnosis and the option offering the most reliable depiction of PCa multifocality.
To date, three approaches have been introduced: cognitive fusion TB (COG-TB), software-based fusion TB (FUS-TB), and in-bore or in-gantry TB (IB-TB). The first approach is “cognitive” targeting, where the physician conducting a transrectal US-guided biopsy reviews the MR imaging results before the procedure and applies this knowledge to identify the most suitable area for TB, as guided by US images [65]. The second approach involves superimposing the MRI images onto the TRUS images after paired landmarks are generated in both through MRI-TRUS fusion platforms. The third approach, in-bore MRI target biopsy (MRI-TB) is performed in the MRI suite through real-time MRI guidance [66].
COG-TB is a more cost-effective and accessible targeted biopsy strategy, especially for small institutions, or those without fusion software or equipment for MRI in-bore biopsy; however, primarily according to the operator’s tumor identification, COG-TB needs a higher level of experience and a more easily followed template to reduce operator variability, and there are insufficient data on the optimal template and predictors for the detection rate of COG-TB. It has been proven that, compared with computer fusion biopsy, cognitive fusion is characterized by a lower cancer confirmation rate for lesions located in the anterior zone or in the transition zone of the prostate [59,67,68,69]. Puech et al. compared COG-TB vs. FUS-TB and detected no difference in cancer detection rate [70]. While most research findings are derived from data from developed countries, there are a few studies from low- and middle-income countries that analyze the impact of COG-TB through mpMRI data on the detection of clinically significant cancer. At the same time, compared against software, further study is needed to pave the way for incorporating MRI-targeted COG-TB, at least until mpMRI fusion biopsy is more widely available.
Progress in the fields of information technology and artificial intelligence has resulted in the development of software platforms that support clinical diagnosis and decision-making through patient data from personalized medicine. The MRI-US fusion biopsy platforms have several advantages, including real-time overlay of the MRI and ultrasound images (with precision similar to the in-bore targeted biopsy), possibility of concurrent systematic sampling, and shorter duration of the procedure when compared with in-bore sampling [64,71]. Multiple versions of targeted biopsy software exist and are capable of conducting biopsies of suspicious regions on the prostate MP-MRI. Uncertainty about the effect of different software-based imaging processing techniques remains to be explored [72]. MRI-TRUS software fusion is also considered less expensive than would be an MRI-guided biopsy (MRGB), and it can be conducted in a shorter time [73].
The European Association of Urologists guidelines suggest that in biopsy-naïve patients with a suspect lesion, systematic TRUS-Bx should be followed by MRGB directed at the MRI-suspicious areas, while in patients with a persisting clinical suspicion of PCa after having gone through a systematic TRUS-Bx with prior negative results, only MRGB targeted to the lesion is recommended [74]. MRGB allows real-time control of the sampling correctness, potentially decreasing errors during the targeting process [75]. Nevertheless, in-bore biopsies fail to allow for concurrent systematic sampling because of time limitation; thus, lesions missed by the prebiopsy cannot be detected by mpMRI through this modality. Another shortcoming is the limited space within the MRI bore, thus limiting the range of motion of the physicians within the magnet. Additionally, the research sample for the in-bore technique is relatively small compared to other trials on mpMRI-TB. This is mainly caused by the fact that in-bore targeted biopsy is neither widely available nor widely applied.

2.5. PET/CT

The sensitivity of conventional imaging approach such as CT, MRI, or BS often fails to detect sites of relapse and/or metastasis [76]. Molecular imaging of prostate cancer has demonstrated good results and allows whole-body assessment of tumor biology. In recent decades, positron emission tomography (PET) techniques have arisen as an encouraging tool for PCa detection, tumor staging, and treatment planning, including metastatic castration-resistant prostate adenocarcinoma [77,78]. PET combined with CT or MRI can help to localize suspicious lesions in the prostate gland through prostate-specific radiotracers (i.e., 18F-fluorodeoxyglucose (FDG), Fluorine-18-labeled sodium fluoride (18F-NaF), choline—labelled with either 18F and 11C, 18F-Fluciclovine, 18F-16b-fluoro-5a-dihydrotestosterone (18F-FDHT), and prostate specific membrane antigen (PSMA) ligands labelled with 68Ga or 18F), thus providing a valuable tool for the diagnosis of cancer, and for the initial staging of the disease [79]. Different radiotracers for PET imaging have been explored in recent years and each tracer has different uptake features. With the occurrence of nuclear tracers, PET could map changes in function and metabolism rather than in anatomy only [80].
18F-FDG PET/CT in the initial staging of PCa is controversial [81,82], since the uptake of FDG is generally low in PCa cells compared with other malignant cells, which causes difficulty in separating malignant from benign tissue [83,84]. The NCCN guidelines recommend against 18F-FDG PET/CT in the initial staging of PCa, but instead advise evaluating biochemical recurrence (BCR) or metastasis [85]. However, H. Jadvar has insisted that FDG PET/CT may be of use in the diagnosis and staging of primary tumors with GS > 7, given the tendency to display high FDG uptake [81].
The uptake of 18F-NaF does not directly allow for the visualization of the presence of tumor cells. Instead, it reflects the increased blood flow, osteoblastic activity, and bone remodeling that are associated with osseous metastases. In comparison to BS, 18F-NaF demonstrates increased bone uptake and a more rapid clearance from soft tissues (owing to minimal serum protein binding). This leads to enhanced bone-to-background contrast and shorter examination duration [86]. At present, there is insufficient evidence to endorse, for clinical benefit, the routine use of 18F-NaF PET/CT instead of BS. Additionally, the bone-only detection capability of 18F-NaF PET/CT presents a limitation, making it a less attractive option in the current age of developing targeted tracers for molecular imaging of prostate cancer which can simultaneously detect extra-skeletal disease.
Radiolabeled choline is an extensively studied tracer in the restaging of prostate cancer in BCR [87]. Choline is an essential element of phospholipids in the cellular wall, and the elevated uptake of choline leads to increased metabolism of the cell membrane components of malignant tumors [88]. Up to now, the EAU has suggested PET imaging with choline derivatives for BCR after radical prostatectomy with a PSA serum level ≥ 1 ng/mL [89].
18F-Fluciclovine PET/CT has also shown promise in the detection of recurrent prostate cancer, with significant impact on subsequent treatment planning, and it has been approved by the Food and Drug Administration (FDA) and the European Commission for patients with elevated PSAs for BCR following prior treatment [90]. The FDA approval was due to its high diagnostic performance and the histologically confirmed data in patients with BCR, with a 68% detection rate on a per-patient basis, a 62% positive predictive value on a per-lesion basis (greater than 90% for extra prostatic disease), and a 70% specificity level for lesions [91]. Fluorine-18-labeled fluciclovine PET/CT has also been demonstrated to be effective for evaluating distant metastases in recurrent PCa [91,92]. Detectability of F-18 fluciclovine PET/CT is generally increased as the PSA level is elevated, and it is more sensitive at PSA level > 1 ng/mL with rapid PSA kinetics [93]. As for PSA levels less than or equal to 1 ng/mL, detection rates range only from 21.0% to 46.4% [94,95,96]. Correspondingly, up to now, there has still been no absolute lower-level cutoff for PSA value that has been explored as an indication for 18F-fluciclovine PET/CT which would provide guidance to referring physicians (urologists, radiation oncologists, and medical oncologists) in determining which patients might benefit most from imaging.
In the context of advanced CRPC resistant to initial conventional ADT, 18F-FDHT holds significant promise. This tracer specifically targets the androgen receptor (AR), which, along with its natural ligands, testosterone and 5a-dihydrotestosterone, plays a crucial role in male sexual differentiation. However, the use of 18F-FDHT PET/CT is currently limited to investigational research purposes, and it has not yet been approved for clinical routine use. Preliminary studies investigating the use of 18F-FDHT PET/CT in patients with CRPC have demonstrated safety, feasibility, accurate detection of lesions, and a correlation with survival outcomes [97,98].
To date, no single imaging approach has demonstrated optimal diagnostic performance in the evaluation of metastatic lymph nodes. Studies with the PET tracers 18F-choline and 11C-choline have indicated similarly high specificities, but low sensitivities have ranged from 40% to 50% [99]. Because of the low-spatial resolution of PET imaging (approximately 5 mm in clinical scanners), choline PET/CT has been found to be unable to diagnose sub-centimeter node metastasis. Beheshti et al. demonstrated a sensitivity of 66% and specificity of 96% for lymph nodes larger than 5 mm when applying 18F-choline PET/CT [100]. It is important to clarify the advantages and disadvantages of 18F-choline PET/CT use for imaging tests of PCa patients.

2.6. PSMA PET

Prostate-specific membrane antigen (PSMA) is a type II transmembrane glycoprotein receptor with folate hydrolase activity and glutamate carboxypeptidase activity. PSMA expression is elevated significantly in higher-grade prostate tumor cells and other solid cancers, including advanced salivary gland cancer, glioblastoma, thyroid cancer, hepatocellular carcinoma, and clear cell renal carcinoma [100]. PSMA can easily penetrate tissues and diffuse within solid tumor lesions and thus reflect the metastasis situation [101,102,103]. Compared with mpMRI, a distinct advantage of 68Ga-PSMA PET scans is that PSMA is overexpressed up to 1000-fold in prostate malignancies compared to benign tissues, which theoretically makes PSMA PET scans relatively specific for malignant transformation compared to mpMRI [104].
Fendler et al. assessed 21 patients for the accuracy of 68Ga-PSMA- PET/CT in localizing a tumor in the prostate and surrounding tissue and detected a significantly higher SUVmax in histopathologically positive segments (11.8 ± 7.6) when compared with negative segments (4.9 ± 2.9; p < 0.001) [105]. 68Ga-PSMA PET/CT is also a prevalently applied modality in patients with BCR, as it has been proved to be superior to conventional diagnostic imaging for locating recurrent disease. Although a BCR after RP is defined by a PSA value > 0.2 ng/mL based on guidance [106], and PSMA PET/CT imaging is suggested for patients with a PSA value > 0.2 ng/mL according to the EAU guidelines [107], the patient group with pre-scan PSA values < 0.2 ng/mL could also be included in determining the effects on the outcome.
PSMA-PET/CT has shown its accuracy in restaging patients in either the local, nodal, or metastatic setting. 68Ga-PSMA has demonstrated greater diagnostic accuracy and a competitive advantage over 18F-fluciclovine in detecting recurrences at PSA levels down to less than 0.5 ng/mL [108,109]. A systematic review including 4790 patients demonstrated patient-based PCa BCR detection rates of 33% and 45% at PSA levels < 0.2 and 0.2–0.49 ng/mL, respectively [110]. Guevelou et al. summarized the potential impact of restaging according to PSMA-PET/CT changes in the management of recurrent prostate cancer after RP [111].
Currently, there is also convincing evidence on the re-staging efficacy of Ga-68-PSMA-11-PET in men with non-metastatic castration-resistant prostate cancer (CRPC). Fendler et al. performed a multicenter, retrospective study in 200 CRPC patients with serum PSA levels over 2 ng/mL, and/or a Gleason score ≥ 8, in whom conventional imaging showed the absence of metastasis; the Ga-68-PSMA-11-PET/CT demonstrated positive findings in 98% of these patients. In addition, in 55% of the patients who suffered disease recurrence, metastases were found in the extra-pelvic lymph nodes (39%), bone (24%), and visceral organs (6%) [112]. 68Ga-PSMA PET/CT imaging has also demonstrated impressive early results, to the extent that some consider it to be the reference standard for the detection of lymphatic metastases [113,114]. Metastatic lymph nodes can appear with non-specific characteristics on MRI; thus, traditional imaging modality will always fail to detect lymphatic metastases [115]. The higher diagnostic accuracy of 68Ga-PSMA-11 PET/CT over mpMRI for pelvic lymph node staging prior to radical prostatectomy in patients with intermediate- to high-risk PCa were confirmed according to the most recent evidence, with a sensitivity of 71% and a specificity of 92% [116].
However, up to now, there has still been no recommendation on the routine application of 68Ga-PSMA-11 PET/CT imaging for the initial staging of PCa according to the current EAU-EANM-ESTRO-ESUR-SIOG Guidelines [20]. The EAU guidelines still rate their recommendation for PSMA-PET/CT in the setting of BCR after radical prostatectomy as “weak”, and fluciclovine-PET/CT is endorsed where PSMA-PET/CT is not available. Because most of the studies exploring PCa primary detection and initial staging before definitive therapy are retrospective design and composed of intermediate to high-risk PCa patients, these tumors are more likely to overexpress PSMA; thus, a potential bias and overestimated accuracy is possible [117,118,119,120]. Some studies have proved the limitation of 68Ga-PSMA PET in detecting low- and intermediate-risk PCa, one which is caused by the low prevalence of extra-prostatic lesions [121]. As expected, some small lesions are missed due to the limited spatial resolution of PET and the presence of background activity in the urinary tract. Additionally, 68Ga-PSMA-11 is metabolized through the urinary system and thus accumulates in the urinary bladder.
At the standard imaging time 100 min after injection, this could result in obscuration of local recurrence in the prostatic fossa by overlaying activity in the urinary bladder. Radiopharmaceutical 18F-PSMA-1007 is a novel PSMA-based radiopharmaceutical that has multiple advantages compared with 68Ga-PSMA-11 [122]. 18F-PSMA-1007 is not excreted from the kidneys in the first few hours after injection, which is potentially beneficial in the detection of local recurrences [123]. In addition, the end-point positron energy of [18F]-F is much lower than that of [68Ga]-Ga (0.65 vs. 1.90 MeV), which could decrease the positron range in tissue and thus improve spatial resolution [124]. Nevertheless, Rauscher et al. have indicated that, despite the aforementioned superiority of [18F]-PSMA-1007 over [68Ga]-PSMA-11, they also noticed a high incidence of unspecific bone uptake—which could lead to over-staging—in a significant number of patients [125].

2.7. PET-Target

mpMRI based fusion biopsy often misses some foci located at the transition and central zones, and the specificity of mpMRI for detecting PCa also decreases its diagnostic efficacy as a triage tool for biopsy. PET is also a useful tool for improving the accuracy of imaging-guided biopsy in PCa, one which mainly includes three approaches: PSMA PET/CT-TRUS software-assisted fusion biopsy, transgluteal PSMA PET/CT-targeted prostate biopsy, and cognitive PSMA PET/CT-TRUS-targeted prostate biopsy. Up to now, PET/CT-biopsy guidance has recommended its use only in patients with previous negative biopsy [126], but it is considered a promising potential tool for future diagnosis [127]. The process of PSMA PET/CT-TRUS software-assisted fusion-targeted prostate biopsy includes selection of suspected PCa lesions by PSMA PET/CT, incorporation of the PSMA PET/CT data into real-time TRUS by an imaging fusion system, and then the acquisition of biopsy samples, as guided through the fused TRUS imaging in real-time. Transgluteal PSMA PET/CT-targeted prostate biopsy means identifying PCa lesions through PSMA PET/CT imaging and then taking two to four biopsy specimens by single-puncture percutaneous transgluteal method under real-time CT guidance, as initially introduced by Zhang et al. in May 2020 [128]. When applied to cognitive PSMA PET/CT-TRUS-targeted prostate biopsy, the surgeon needs to view the location of a target lesion identified by PSMA PET/CT imaging and then translates the suspected PCa lesion sites to be targeted by TRUS-guided biopsy through various mental processes, such as memory, measurement calculation, three-dimensional spatial reasoning, and pattern recognition.
Simopoulos et al. first introduced a successful case of 68Ga-PSMA PET/CT and MRI/ultrasound-guided prostate biopsy [129]. Subsequently, Westenfelder et al. successfully detected csPCa (GS 4 + 3) through 68Ga-PSMA PET/MR-plus-ultrasound guided biopsy in a patient with a previous negative prostate biopsy and an MRI result [130]. Limited available evidence has shown the superior performance of PSMA-PET in comparison with mpMRI for lesion characterization and intra-prostatic staging [127,131,132]. Caracciolo et al. highlighted that PSMA-PET/MRI has a high accuracy for detecting csPCa, and is a promising tool for the selection of patients with suspicion of PCa and preceding negative biopsy or contraindications to MRI [133]. Ferraro et al. demonstrated that 68Ga-PSMA-11 PET/MRI-guided biopsy had a high accuracy for detecting csPCa with patient-based sensitivity, specificity, negative and positive predictive value, and accuracy of 96%, 81%, 93%, 89%, and 90%, respectively [134]. These preliminary results indicated that PSMA PET could be a useful tool in identifying and defining malignant lesions before prostate biopsy. Fendler et al. highlighted that lesions with SUVmax > 6.5 for PCa diagnosis would lead to a sensitivity of 67% and specificity of 92% when performing target biopsy [105]. SUVmax reflects the tumor expression of PSMA, with higher grade tumors (Gleason score > 7) usually relating to a much higher SUVmax value, varying from 16 to 21, as compared with intermediate and lower grade tumors corresponding to SUVmax values of 8.2–8.8 and 5.9–9.6, respectively [135,136].
In the study of R. Kumar et al., postprocedural complications were reported in five of fifty-six (9%) participants and were minor (i.e., hematuria, hematospermia, and gluteal pain), with no participant suffering from a postprocedural infection [137]. The transgluteal modality could decrease the risk of infection and the need for multiple prostatic capsule biopsy. 68Ga-PSMA PET/CT use in this context could significantly reduce unnecessary biopsies and related complications. Nevertheless, literature on PSMA-PET/CT-guided biopsy remains sparse. Few studies have made a direct comparison between outcome of mpMRI-guided biopsy and PSMA-PET/CT-guided biopsy for the detection of localized PCa. Besides, all the image tracers up to now have been labeled with 68Ga, and thus the value of 18F-PSMA PET/CT-TB remains to be investigated. More clinical studies applying PET/MRI-guided biopsy are recommended, and these may have better performance due to the superior spatial resolution provided by MRI. PET-guided biopsy could then be used when mpMRI is contraindicated or equivocal, or when an MRI-guided biopsy is negative for cancer despite high clinical suspicion.
The combination of mpMRI and PSMA-PET could offer complementary information, and the simultaneous use of both imaging approaches offers significantly augmented sensitivity and specificity for intraprostatic tumor mass delineation, and thereby generates an innovative imaging modality capable of overcoming the pitfalls of traditional imaging, and, potentially, helping clinicians in the management of PCa [138]. In addition, improved detection accuracy could help patient selection for salvage therapy and assist in guiding individualized treatments. 18F-FDG PET/ MRI could offer the anatomical imaging benefits of MRI over CT and the molecular imaging of 18F-FDG PET, without radiation or CT beam-hardening imaging artifacts. Furthermore, PSMA-PET/MRI enables whole-body staging at a lower radiation dose and the guidance of prostate biopsies in a single hospital visit, which is superior to the conduction of both a pre-biopsy mpMRI and a staging PSMA PET/CT.

2.8. PET/MRI

Multiple experience-based reports of PET/MRI have now been published. The simultaneous application of PET and mpMRI offers metabolic, structural, and functional imaging information regarding PCa status in a whole-body single-session test, along with better soft tissue contrast and reduced radiation exposure in comparison to PET/CT [139].
Eiber et al. demonstrated an overall detection rate of 84% in 75 patients with median PSA levels of 2.6 ng/mL through 11C-choline PET/MRI in recurrent PCa (range 0.2–88 ng/mL), and thus indicated a significant advantage for 68Ga-PSMA-PET/MRI over PET/CT and mpMRI modality alone for detecting malignant intraprostatic foci [117,140]. Hope et al. conducted a meta-analysis of 68Ga-PSMA-11 PET accuracy for the detection of PCa, with a sensitivity and specificity of 74% and 96% [141].
68Ga-PSMA-11 PET/MRI has demonstrated higher sensitivity and specificity than have either mpMRI or 68Ga-PSMA-11 PET imaging performed separately for detecting intraprostatic tumors and pelvic lymph nodes, especially for patients with extremely low levels of PSA (<0.5 ng/mL). Kranzbühler et al. published a retrospective study and reported a detection rate of 65% in patients with PSA values between 0.2 and 0.5 ng/mL [142]. In comparison, mpMRI has a reported sensitivity of 61% and specificity of 58.7% for the detection of local recurrence [143]. K.M. Selnæs et al. suggested that the quantity of equivocal findings was decreased when MR images were assessed combined with PET uptake [144]. PSMA-PET/MRI showed a patient-based accuracy of 90% for detecting csPCa, according to a prospective single-center study, with a sensitivity of 96% and specificity of 81% [134]. This is higher than the conventional mpMRI accuracy indicated in most studies applying template biopsy as a reference standard, including the PROMIS trial, which demonstrated sensitivity and specificity of 93% and 41%, respectively [7].
Very few studies so far have made a direct comparison between PSMA-PET/CT and PET/MRI for biopsy guidance. All of the literature up to now has been based on tests performed with patients with intermediate- to high-risk disease, which is not the patient population in which increased detection sensitivity is required. Thereby, further work in the active surveillance population is still needed. Domachevsky et al. compared 68Ga-PSMA PET/MRI with same-day late 68Ga-PSMA PET/CT according to lesion detectability in a mixed group of patients for initial staging of high-risk PCa patients (n = 13) and suspected recurrence (n = 8) and obtained a comparable agreement between the two imaging approaches [145]. I. Jambor et al. indicated that the use of neither PET/MRI nor mpMRI was not able to detect pelvic lymph node metastases smaller than 8 mm [146].
However, up to now, very limited data has been available as to its role in clinical setting, mainly owing to the high cost of PET/MRI. Besides, two aspects still need further investigation. First, MRI-positive lesions may show unapparent or low uptake in PET images, which may lead to misdiagnosis of some MRI-negative lesions as positive, if we regard all apparent uptake lesions as positive in PET images. Second, the cut-off value varies across different medical centers. Therefore, each study has been performed under different execution standards.

2.9. Radiomics

In recent years, radiomics has been explored as a means of adding value to diagnostic pathways and patient management. Radiomics is the transformation of medical images into high-dimension mineable data through the extraction of quantitative features, with the ultimate goal of applying these features to glean valuable diagnostic or prognostic information that can guide clinical decision making [147]. Radiomics provides a non-invasive and low-cost automated technique for the evaluation of tumor properties according to MR images. Thus, radiomics could offer additional data that are often not visible to the naked eye. Multiple approaches for csPCa segmentation or classification through deep-learning networks or radiomics methods have been reported in the literature [148,149,150]. One of the essential steps in radiomics is the obtaining of prospective protocol-based good-quality images. Multiple previous studies have already demonstrated that a radiomics-based machine learning method could help not only for the quantification of imaging results, but also has the potential to identify pathological findings without visible abnormalities. Another benefit is that radiomics examines whole tumors, as opposed to biopsy schedules, which tend to sampling errors caused by intra-tumoral heterogeneity [151]. Through radiomics analysis, noninvasive MR-based techniques could predict PCa aggressiveness prior to biopsy or surgery. Recent studies have investigated the value of radiomics according to MRI in the differentiation of PCa from benign prostate tissue [152]. The ability of MRI to visualize the whole tumor volume, in conjunction with ongoing attempts to standardize image acquisition parameters, offers the potential to explore the ability of quantitative image-derived features, or radiomics, to accelerate the development of accurate and reproducible predictors of disease progression on AS. Ginsburg et al. considered ADC radiomics indicators to be highly relevant for their transition zone classifier [153]. A quantitative radiomics method named the morphology, asymmetry, physiology, and size (MAPS) feature model reached an accuracy of 87% for prostate cancer detection [154]. S.J. Hectors et al. also demonstrated the additional value of MRI texture analysis for the identification of csPCa in PI-RADS 3 lesions, with a resulting sensitivity of 75% [155]. Min et al. and Zhang et al. have also assessed the outcomes of an mpMRI-based radiomics signature for csPCa diagnosis, with impressive results [156,157]. Multiple previous studies have applied radiomics analysis to the automation of PCa diagnosis and risk stratification [158].
Recently, M.C.F. Cysouw et al. indicated in a preliminary report the additional and potential value of PET-derived radiomics features through a machine-learning extraction method in a sample of patients with high-risk PCa [159].
However, in practice, one of the biggest problems of radiomics is generation [147]. The data on radiomics learning methods for the evaluation of the predictive or prognostic value of PSMA-PET are still unconvincing. The role of prostate MRI radiomics, from the whole prostate gland (WG) and/or from MRI-based suspicious lesions, remains the subject of controversy. This is mainly because the majority of the radiomics studies validated their methods through splitting their original dataset into training and validation subsets, while only a few studies have conducted a validation via an external set [158,160]. Radiomics analyses could probably help in treatment optimization, but this should be examined in large patient populations first.

2.10. Cost-Effectiveness

The overdiagnosis of PCa leading to overtreatment, as well as the potential underdiagnosis of csPCa, represent inefficient utilizations of limited healthcare resources. The potential advantages of diagnostics may be evident in men whose prostate cancer is likely to progress. However, other men can be subjected to unnecessary tests and treatments, resulting in both economic costs to healthcare resources and harm to patients. Hence, determining whether the potential benefits of diagnostics outweigh the costs and harms necessitates a formal comparison of the costs and consequences of different courses of action through an economic evaluation. Reports have shown that a strategy using MRI may be cost-effective compared with systematic TRUS-Bx, not only in patients with prior negative TRUS-Bx [36], but also in biopsy-naïve populations [161]. Faria et al. conducted a cost-effectiveness study utilizing data from the PROMIS study, which demonstrated that the utilization of mpMRI followed by up to two targeted TRUS-Bx procedures was a cost-effective strategy for the early detection of PCa [162]. PSMA PET/CT is expected to incur higher costs compared to CT+BS when it comes to the primary staging of PCa. However, it is deemed cost-effective, particularly in newly diagnosed patients with intermediate- to high-risk PCa, as it helps to avoid unnecessary treatments [163]. The cost-effectiveness analysis conducted by Subramanian et al. indicates that PSMA PET/CT should be regarded as a viable alternative to both 18F-Fluciclovine PET/CT and standard imaging modalities for prostate cancer staging [164]. Regarding the BCR of PCa, both PSMA PET/CT and PSMA PET/MRI have been considered cost-effective approaches. Gordon et al. have demonstrated that the utilization of PSMA PET/MRI as a cost-effective alternative to the standard CT and BS staging scans for BCR can help to prevent unnecessary or ineffective local therapies and guide increasingly precise and targeted treatments [165]. However, according to the findings of Parikh et al., for PSMA PET/CT to no longer be considered cost-effective in PCa patients with BCR, its price would need to be set at or exceed $14,004 [166]. The evidence suggests that PSMA PET may, in the future, become an indispensable tool in the diagnosis and management of prostate cancer. However, currently, there is a lack of comparative cost-effectiveness studies between PSMA PET/CT and mpMRI. This may be due to the predominant focus of PSMA PET/CT cost-effectiveness research on lymph node metastasis and BCR, while studies on mpMRI’s cost-effectiveness tend to concentrate on intraprostatic lesions. Simultaneously, the issue of whether diagnostics for PCa are cost-effective in a particular country remains unclear. Therefore, any recommendations provided to decision-makers should take into consideration the current country-specific data.

3. Conclusions

The diagnostic techniques for prostate cancer develop with time. Since prostate biopsy is an invasive procedure which may cause post-biopsy complications, it is essential to discover non-invasive and increasingly accurate diagnostic approaches.
In this literature review, we have summarized the up-to-date existing techniques. (Table 1) For intraprostatic lesions, while conventional imaging techniques such as US and CT play their roles, mpMRI and various types of targeted biopsies are becoming increasingly important. Considering the csPCa detection rate, it is still recommended to utilize mpMRI for individuals who are biopsy-naïve or who require repeated biopsy. Simultaneously, the role of biomarkers in conjunction with mpMRI, as proposed in recent years, cannot be overlooked. For metastatic lesions and BCR, BS is the most accessible and widely employed method. Emerging PET diagnostic methods such as PSMA-PET and PET/MRI demonstrate promising diagnostic efficacy. However, due to cost constraints and lack of experimental data, their widespread application remains limited in the short term. Similarly, with the progress of artificial intelligence and machine learning, radiomics will play an increasingly significant role in future diagnostics.
Regarding the current state of affairs, the combined employment of MRI-TB and TRUS-Bx has been widely adopted for biopsy purposes to reduce misdiagnosis, and BS is widely used for screening metastases outside the prostate. In the future, we eagerly anticipate the emergence of novel imaging diagnostics that may gradually replace them.

Author Contributions

M.Z. and Z.L. reviewed the literature, collected the data, and drafted the manuscript, W.Y., T.F., and Z.M. revised and edited the manuscript critically, S.J., L.W., H.Z. and Y.C. supervised the manuscript, M.Z. proofread the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by National High Level Hospital Clinical Research Funding. Project No.: [2022-PUMCH-B-009][2022-PUMCH-A-063].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare that there is no conflict of interest regarding the publication of this paper.

References

  1. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Mathers, C.; Parkin, D.M.; Piñeros, M.; Znaor, A.; Bray, F. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int. J. Cancer 2019, 144, 1941–1953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  3. Bell, K.J.; Del Mar, C.; Wright, G.; Dickinson, J.; Glasziou, P. Prevalence of incidental prostate cancer: A systematic review of autopsy studies. Int. J. Cancer 2015, 137, 1749–1757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Musunuru, H.B.; Yamamoto, T.; Klotz, L.; Ghanem, G.; Mamedov, A.; Sethukavalan, P.; Jethava, V.; Jain, S.; Zhang, L.; Vesprini, D.; et al. Active Surveillance for Intermediate Risk Prostate Cancer: Survival Outcomes in the Sunnybrook Experience. J. Urol. 2016, 196, 1651–1658. [Google Scholar] [CrossRef] [PubMed]
  5. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer Statistics, 2017. CA Cancer J. Clin. 2017, 67, 7–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Johansen, T.E.B.; Zahl, P.-H.; Baco, E.; Bartoletti, R.; Bonkat, G.; Bruyere, F.; Cai, T.; Cek, M.; Kulchavenya, E.; Köves, B.; et al. Antibiotic resistance, hospitalizations, and mortality related to prostate biopsy: First report from the Norwegian Patient Registry. World J. Urol. 2020, 38, 17–26. [Google Scholar] [CrossRef]
  7. Ahmed, H.U.; El-Shater Bosaily, A.; Brown, L.C.; Gabe, R.; Kaplan, R.; Parmar, M.K.; Collaco-Moraes, Y.; Ward, K.; Hindley, R.G.; Freeman, A.; et al. Diagnostic accuracy of multi-parametric MRI and TRUS biopsy in prostate cancer (PROMIS): A paired validating confirmatory study. Lancet 2017, 389, 815–822. [Google Scholar] [CrossRef] [Green Version]
  8. Welch, H.G.; Albertsen, P.C. Reconsidering Prostate Cancer Mortality—The Future of PSA Screening. N. Engl. J. Med. 2020, 382, 1557–1563. [Google Scholar] [CrossRef]
  9. Grossman, D.C.; Curry, S.J.; Owens, D.K.; Bibbins-Domingo, K.; Caughey, A.B.; Davidson, K.W.; Doubeni, C.A.; Ebell, M.; Epling, J.W., Jr.; Kemper, A.R. Screening for Prostate Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2018, 319, 1901–1913. [Google Scholar]
  10. Duffy, M.J. Biomarkers for prostate cancer: Prostate-specific antigen and beyond. Clin. Chem. Lab. Med. 2019, 58, 326–339. [Google Scholar] [CrossRef] [Green Version]
  11. Thompson, I.M.; Pauler, D.K.; Goodman, P.J.; Tangen, C.M.; Lucia, M.S.; Parnes, H.L.; Minasian, L.M.; Ford, L.G.; Lippman, S.M.; Crawford, E.D.; et al. Prevalence of prostate cancer among men with a prostate-specific antigen level < or =4.0 ng per milliliter. N. Engl. J. Med. 2004, 350, 2239–2246. [Google Scholar] [PubMed] [Green Version]
  12. Beemsterboer, P.M.; Kranse, R.; de Koning, H.J.; Habbema, J.D.; Schröder, F.H. Changing role of 3 screening modalities in the European randomized study of screening for prostate cancer (Rotterdam). Int. J. Cancer 1999, 84, 437–441. [Google Scholar] [CrossRef]
  13. Engelbrecht, M.R.; Barentsz, J.O.; Jager, G.J.; van der Graaf, M.; Heerschap, A.; Sedelaar, J.P.; Aarnink, R.G.; de la Rosette, J.J. Prostate cancer staging using imaging. BJU Int. 2000, 86 (Suppl. 1), 123–134. [Google Scholar] [CrossRef] [PubMed]
  14. Sedelaar, J.P.; van Leenders, G.J.; Hulsbergen-van de Kaa, C.A.; van der Poel, H.G.; van der Laak, J.A.; Debruyne, F.M.; Wijkstra, H.; de la Rosette, J.J. Microvessel density: Correlation between contrast ultrasonography and histology of prostate cancer. Eur. Urol. 2001, 40, 285–293. [Google Scholar] [CrossRef]
  15. Mitterberger, M.; Pinggera, G.M.; Pallwein, L.; Gradl, J.; Frauscher, F.; Bartsch, G.; Strasser, H.; Akkad, T.; Horninger, W. The value of three-dimensional transrectal ultrasonography in staging prostate cancer. BJU Int. 2007, 100, 47–50. [Google Scholar] [CrossRef] [PubMed]
  16. Simmons, L.A.; Autier, P.; Zát’ura, F.; Braeckman, J.; Peltier, A.; Romic, I.; Stenzl, A.; Treurnicht, K.; Walker, T.; Nir, D. Detection, localisation and characterisation of prostate cancer by prostate HistoScanning(™). BJU Int. 2012, 110, 28–35. [Google Scholar] [CrossRef]
  17. Erbersdobler, A.; Isbarn, H.; Dix, K.; Steiner, I.; Schlomm, T.; Mirlacher, M.; Sauter, G.; Haese, A. Prognostic value of microvessel density in prostate cancer: A tissue microarray study. World J. Urol. 2010, 28, 687–692. [Google Scholar] [CrossRef]
  18. Zhao, H.X.; Zhu, Q.; Wang, Z.C. Detection of prostate cancer with three-dimensional transrectal ultrasound: Correlation with biopsy results. Br. J. Radiol. 2012, 85, 714–719. [Google Scholar] [CrossRef] [Green Version]
  19. Padhani, A.R.; Lecouvet, F.E.; Tunariu, N.; Koh, D.M.; De Keyzer, F.; Collins, D.J.; Sala, E.; Fanti, S.; Vargas, H.A.; Petralia, G.; et al. Rationale for Modernising Imaging in Advanced Prostate Cancer. Eur. Urol. Focus 2017, 3, 223–239. [Google Scholar] [CrossRef] [Green Version]
  20. Mottet, N.; van den Bergh, R.C.N.; Briers, E.; Van den Broeck, T.; Cumberbatch, M.G.; De Santis, M.; Fanti, S.; Fossati, N.; Gandaglia, G.; Gillessen, S.; et al. EAU-EANM-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer-2020 Update. Part 1: Screening, Diagnosis, and Local Treatment with Curative Intent. Eur. Urol. 2021, 79, 243–262. [Google Scholar] [CrossRef]
  21. Donohoe, K.J.; Cohen, E.J.; Giammarile, F.; Grady, E.; Greenspan, B.S.; Henkin, R.E.; Millstine, J.; Smith, G.T.; Srinivas, S.; Kauffman, J.; et al. Appropriate Use Criteria for Bone Scintigraphy in Prostate and Breast Cancer: Summary and Excerpts. J. Nucl. Med. 2017, 58, 14n–17n. [Google Scholar] [PubMed]
  22. Shen, G.; Deng, H.; Hu, S.; Jia, Z. Comparison of choline-PET/CT, MRI, SPECT, and bone scintigraphy in the diagnosis of bone metastases in patients with prostate cancer: A meta-analysis. Skelet. Radiol 2014, 43, 1503–1513. [Google Scholar] [CrossRef] [PubMed]
  23. Pasoglou, V.; Larbi, A.; Collette, L.; Annet, L.; Jamar, F.; Machiels, J.P.; Michoux, N.; Vande Berg, B.C.; Tombal, B.; Lecouvet, F.E. One-step TNM staging of high-risk prostate cancer using magnetic resonance imaging (MRI): Toward an upfront simplified “all-in-one” imaging approach? Prostate 2014, 74, 469–477. [Google Scholar] [CrossRef]
  24. Metser, U.; Chua, S.; Ho, B.; Punwani, S.; Johnston, E.; Pouliot, F.; Tau, N.; Hawsawy, A.; Anconina, R.; Bauman, G.; et al. The Contribution of Multiparametric Pelvic and Whole-Body MRI to Interpretation of (18)F-Fluoromethylcholine or (68)Ga-HBED-CC PSMA-11 PET/CT in Patients with Biochemical Failure After Radical Prostatectomy. J. Nucl. Med. 2019, 60, 1253–1258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Zacho, H.D.; Nielsen, J.B.; Afshar-Oromieh, A.; Haberkorn, U.; de Souza, N.; De Paepe, K.; Dettmann, K.; Langkilde, N.C.; Haarmark, C.; Fisker, R.V.; et al. Prospective comparison of (68)Ga-PSMA PET/CT, (18)F-sodium fluoride PET/CT and diffusion weighted-MRI at for the detection of bone metastases in biochemically recurrent prostate cancer. Eur. J. Nucl. Med. Mol Imaging 2018, 45, 1884–1897. [Google Scholar] [CrossRef]
  26. Ukimura, O.; de Castro Abreu, A.L.; Gill, I.S.; Shoji, S.; Hung, A.J.; Bahn, D. Image visibility of cancer to enhance targeting precision and spatial mapping biopsy for focal therapy of prostate cancer. BJU Int. 2013, 111, E354–E364. [Google Scholar] [CrossRef]
  27. Ukimura, O.; Marien, A.; Palmer, S.; Villers, A.; Aron, M.; de Castro Abreu, A.L.; Leslie, S.; Shoji, S.; Matsugasumi, T.; Gross, M.; et al. Trans-rectal ultrasound visibility of prostate lesions identified by magnetic resonance imaging increases accuracy of image-fusion targeted biopsies. World J. Urol. 2015, 33, 1669–1676. [Google Scholar] [CrossRef]
  28. D’Amico, A.V.; Tempany, C.M.; Cormack, R.; Hata, N.; Jinzaki, M.; Tuncali, K.; Weinstein, M.; Richie, J.P. Transperineal magnetic resonance image guided prostate biopsy. J. Urol. 2000, 164, 385–387. [Google Scholar] [CrossRef]
  29. Kasivisvanathan, V.; Rannikko, A.S.; Borghi, M.; Panebianco, V.; Mynderse, L.A.; Vaarala, M.H.; Briganti, A.; Budäus, L.; Hellawell, G.; Hindley, R.G.; et al. MRI-Targeted or Standard Biopsy for Prostate-Cancer Diagnosis. N. Engl. J. Med. 2018, 378, 1767–1777. [Google Scholar] [CrossRef]
  30. Turkbey, B.; Rosenkrantz, A.B.; Haider, M.A.; Padhani, A.R.; Villeirs, G.; Macura, K.J.; Tempany, C.M.; Choyke, P.L.; Cornud, F.; Margolis, D.J.; et al. Prostate Imaging Reporting and Data System Version 2.1: 2019 Update of Prostate Imaging Reporting and Data System Version 2. Eur. Urol. 2019, 76, 340–351. [Google Scholar] [CrossRef]
  31. Moldovan, P.C.; Van den Broeck, T.; Sylvester, R.; Marconi, L.; Bellmunt, J.; van den Bergh, R.C.N.; Bolla, M.; Briers, E.; Cumberbatch, M.G.; Fossati, N.; et al. What Is the Negative Predictive Value of Multiparametric Magnetic Resonance Imaging in Excluding Prostate Cancer at Biopsy? A Systematic Review and Meta-analysis from the European Association of Urology Prostate Cancer Guidelines Panel. Eur. Urol. 2017, 72, 250–266. [Google Scholar] [CrossRef] [PubMed]
  32. Israël, B.; Immerzeel, J.; van der Leest, M.; Hannink, G.; Zámecnik, P.; Bomers, J.; Schoots, I.G.; van Basten, J.P.; Debruyne, F.; van Oort, I.; et al. Clinical implementation of pre-biopsy magnetic resonance imaging pathways for the diagnosis of prostate cancer. BJU Int. 2022, 129, 480–490. [Google Scholar] [CrossRef] [PubMed]
  33. Oishi, M.; Shin, T.; Ohe, C.; Nassiri, N.; Palmer, S.L.; Aron, M.; Ashrafi, A.N.; Cacciamani, G.E.; Chen, F.; Duddalwar, V.; et al. Which Patients with Negative Magnetic Resonance Imaging Can Safely Avoid Biopsy for Prostate Cancer? J. Urol. 2019, 201, 268–276. [Google Scholar] [CrossRef] [PubMed]
  34. Stanzione, A.; Creta, M.; Imbriaco, M.; La Rocca, R.; Capece, M.; Esposito, F.; Imbimbo, C.; Fusco, F.; Celentano, G.; Napolitano, L.; et al. Attitudes and perceptions towards multiparametric magnetic resonance imaging of the prostate: A national survey among Italian urologists. Arch. Ital. Urol. Androl. 2020, 92, 291–296. [Google Scholar] [CrossRef]
  35. Muller, B.G.; Shih, J.H.; Sankineni, S.; Marko, J.; Rais-Bahrami, S.; George, A.K.; de la Rosette, J.J.; Merino, M.J.; Wood, B.J.; Pinto, P.; et al. Prostate Cancer: Interobserver Agreement and Accuracy with the Revised Prostate Imaging Reporting and Data System at Multiparametric MR Imaging. Radiology 2015, 277, 741–750. [Google Scholar] [CrossRef] [Green Version]
  36. Mowatt, G.; Scotland, G.; Boachie, C.; Cruickshank, M.; Ford, J.A.; Fraser, C.; Kurban, L.; Lam, T.B.; Padhani, A.R.; Royle, J.; et al. The diagnostic accuracy and cost-effectiveness of magnetic resonance spectroscopy and enhanced magnetic resonance imaging techniques in aiding the localisation of prostate abnormalities for biopsy: A systematic review and economic evaluation. Health Technol. Assess. 2013, 17, 1–281. [Google Scholar] [CrossRef] [Green Version]
  37. Rosenkrantz, A.B.; Ginocchio, L.A.; Cornfeld, D.; Froemming, A.T.; Gupta, R.T.; Turkbey, B.; Westphalen, A.C.; Babb, J.S.; Margolis, D.J. Interobserver Reproducibility of the PI-RADS Version 2 Lexicon: A Multicenter Study of Six Experienced Prostate Radiologists. Radiology 2016, 280, 793–804. [Google Scholar] [CrossRef] [Green Version]
  38. Vargas, H.A.; Akin, O.; Franiel, T.; Mazaheri, Y.; Zheng, J.; Moskowitz, C.; Udo, K.; Eastham, J.; Hricak, H. Diffusion-weighted endorectal MR imaging at 3 T for prostate cancer: Tumor detection and assessment of aggressiveness. Radiology 2011, 259, 775–784. [Google Scholar] [CrossRef] [Green Version]
  39. Jie, C.; Rongbo, L.; Ping, T. The value of diffusion-weighted imaging in the detection of prostate cancer: A meta-analysis. Eur. Radiol. 2014, 24, 1929–1941. [Google Scholar] [CrossRef] [Green Version]
  40. Rosenkrantz, A.B.; Triolo, M.J.; Melamed, J.; Rusinek, H.; Taneja, S.S.; Deng, F.M. Whole-lesion apparent diffusion coefficient metrics as a marker of percentage Gleason 4 component within Gleason 7 prostate cancer at radical prostatectomy. J. Magn. Reson Imaging 2015, 41, 708–714. [Google Scholar] [CrossRef] [Green Version]
  41. Vignati, A.; Mazzetti, S.; Giannini, V.; Russo, F.; Bollito, E.; Porpiglia, F.; Stasi, M.; Regge, D. Texture features on T2-weighted magnetic resonance imaging: New potential biomarkers for prostate cancer aggressiveness. Phys. Med. Biol. 2015, 60, 2685–2701. [Google Scholar] [CrossRef] [PubMed]
  42. Rozenberg, R.; Thornhill, R.E.; Flood, T.A.; Hakim, S.W.; Lim, C.; Schieda, N. Whole-Tumor Quantitative Apparent Diffusion Coefficient Histogram and Texture Analysis to Predict Gleason Score Upgrading in Intermediate-Risk 3 + 4 = 7 Prostate Cancer. AJR Am. J. Roentgenol. 2016, 206, 775–782. [Google Scholar] [CrossRef] [PubMed]
  43. Huebner, N.A.; Korn, S.; Resch, I.; Grubmüller, B.; Gross, T.; Gale, R.; Kramer, G.; Poetsch, N.; Clauser, P.; Haitel, A.; et al. Visibility of significant prostate cancer on multiparametric magnetic resonance imaging (MRI)-do we still need contrast media? Eur. Radiol. 2021, 31, 3754–3764. [Google Scholar] [CrossRef] [PubMed]
  44. Wei, C.G.; Chen, T.; Zhang, Y.Y.; Pan, P.; Dai, G.C.; Yu, H.C.; Yang, S.; Jiang, Z.; Tu, J.; Lu, Z.H.; et al. Biparametric prostate MRI and clinical indicators predict clinically significant prostate cancer in men with “gray zone” PSA levels. Eur. J. Radiol. 2020, 127, 108977. [Google Scholar] [CrossRef]
  45. Kuhl, C.K.; Bruhn, R.; Krämer, N.; Nebelung, S.; Heidenreich, A.; Schrading, S. Abbreviated Biparametric Prostate MR Imaging in Men with Elevated Prostate-specific Antigen. Radiology 2017, 285, 493–505. [Google Scholar] [CrossRef] [Green Version]
  46. Sherrer, R.L.; Glaser, Z.A.; Gordetsky, J.B.; Nix, J.W.; Porter, K.K.; Rais-Bahrami, S. Comparison of biparametric MRI to full multiparametric MRI for detection of clinically significant prostate cancer. Prostate Cancer Prostatic Dis. 2019, 22, 331–336. [Google Scholar] [CrossRef]
  47. Palumbo, P.; Manetta, R.; Izzo, A.; Bruno, F.; Arrigoni, F.; De Filippo, M.; Splendiani, A.; Di Cesare, E.; Masciocchi, C.; Barile, A. Biparametric (bp) and multiparametric (mp) magnetic resonance imaging (MRI) approach to prostate cancer disease: A narrative review of current debate on dynamic contrast enhancement. Gland. Surg. 2020, 9, 2235–2247. [Google Scholar] [CrossRef]
  48. Rudolph, M.M.; Baur, A.D.J.; Cash, H.; Haas, M.; Mahjoub, S.; Hartenstein, A.; Hamm, C.A.; Beetz, N.L.; Konietschke, F.; Hamm, B.; et al. Diagnostic performance of PI-RADS version 2.1 compared to version 2.0 for detection of peripheral and transition zone prostate cancer. Sci. Rep. 2020, 10, 15982. [Google Scholar] [CrossRef]
  49. Rouvière, O.; Puech, P.; Renard-Penna, R.; Claudon, M.; Roy, C.; Mège-Lechevallier, F.; Decaussin-Petrucci, M.; Dubreuil-Chambardel, M.; Magaud, L.; Remontet, L.; et al. Use of prostate systematic and targeted biopsy on the basis of multiparametric MRI in biopsy-naive patients (MRI-FIRST): A prospective, multicentre, paired diagnostic study. Lancet Oncol. 2019, 20, 100–109. [Google Scholar] [CrossRef]
  50. Drost, F.-J.H.; Osses, D.; Nieboer, D.; Bangma, C.H.; Steyerberg, E.W.; Roobol, M.J.; Schoots, I.G. Prostate Magnetic Resonance Imaging, with or Without Magnetic Resonance Imaging-targeted Biopsy, and Systematic Biopsy for Detecting Prostate Cancer: A Cochrane Systematic Review and Meta-analysis. Eur. Urol. 2020, 77, 78–94. [Google Scholar] [CrossRef]
  51. Jambor, I.; Verho, J.; Ettala, O.; Knaapila, J.; Taimen, P.; Syvänen, K.T.; Kiviniemi, A.; Kähkönen, E.; Perez, I.M.; Seppänen, M.; et al. Validation of IMPROD biparametric MRI in men with clinically suspected prostate cancer: A prospective multi-institutional trial. PLoS Med. 2019, 16, e1002813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Weinreb, J.C.; Barentsz, J.O.; Choyke, P.L.; Cornud, F.; Haider, M.A.; Macura, K.J.; Margolis, D.; Schnall, M.D.; Shtern, F.; Tempany, C.M.; et al. PI-RADS Prostate Imaging—Reporting and Data System: 2015, Version 2. Eur. Urol. 2016, 69, 16–40. [Google Scholar] [CrossRef] [PubMed]
  53. Schoots, I.G. MRI in early prostate cancer detection: How to manage indeterminate or equivocal PI-RADS 3 lesions? Transl. Androl. Urol. 2018, 7, 70–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Maggi, M.; Panebianco, V.; Mosca, A.; Salciccia, S.; Gentilucci, A.; Di Pierro, G.; Busetto, G.M.; Barchetti, G.; Campa, R.; Sperduti, I.; et al. Prostate Imaging Reporting and Data System 3 Category Cases at Multiparametric Magnetic Resonance for Prostate Cancer: A Systematic Review and Meta-analysis. Eur. Urol. Focus 2020, 6, 463–478. [Google Scholar] [CrossRef] [PubMed]
  55. Frisbie, J.W.; Van Besien, A.J.; Lee, A.; Xu, L.; Wang, S.; Choksi, A.; Afzal, M.A.; Naslund, M.J.; Lane, B.; Wong, J.; et al. PSA density is complementary to prostate MP-MRI PI-RADS scoring system for risk stratification of clinically significant prostate cancer. Prostate Cancer Prostatic Dis. 2022, 26, 347–352. [Google Scholar] [CrossRef] [PubMed]
  56. Stonier, T.; Simson, N.; Shah, T.; Lobo, N.; Amer, T.; Lee, S.M.; Bass, E.; Chau, E.; Grey, A.; McCartan, N.; et al. The “Is mpMRI Enough” or IMRIE Study: A Multicentre Evaluation of Prebiopsy Multiparametric Magnetic Resonance Imaging Compared with Biopsy. Eur. Urol. Focus 2021, 7, 1027–1034. [Google Scholar] [CrossRef]
  57. Stavrinides, V.; Papageorgiou, G.; Danks, D.; Giganti, F.; Pashayan, N.; Trock, B.; Freeman, A.; Hu, Y.; Whitaker, H.; Allen, C.; et al. Mapping PSA density to outcome of MRI-based active surveillance for prostate cancer through joint longitudinal-survival models. Prostate Cancer Prostatic Dis. 2021, 24, 1028–1031. [Google Scholar] [CrossRef]
  58. Wagaskar, V.G.; Sobotka, S.; Ratnani, P.; Young, J.; Lantz, A.; Parekh, S.; Falagario, U.G.; Li, L.; Lewis, S.; Haines, K., 3rd; et al. A 4K score/MRI-based nomogram for predicting prostate cancer, clinically significant prostate cancer, and unfavorable prostate cancer. Cancer Rep. 2021, 4, e1357. [Google Scholar]
  59. Pepe, P.; Garufi, A.; Priolo, G.; Pennisi, M. Transperineal Versus Transrectal MRI/TRUS Fusion Targeted Biopsy: Detection Rate of Clinically Significant Prostate Cancer. Clin. Genitourin. Cancer 2017, 15, e33–e36. [Google Scholar] [CrossRef]
  60. Ahdoot, M.; Wilbur, A.R.; Reese, S.E.; Lebastchi, A.H.; Mehralivand, S.; Gomella, P.T.; Bloom, J.; Gurram, S.; Siddiqui, M.; Pinsky, P.; et al. MRI-Targeted, Systematic, and Combined Biopsy for Prostate Cancer Diagnosis. N. Engl. J. Med. 2020, 382, 917–928. [Google Scholar] [CrossRef]
  61. Demirtaş, A.; Sönmez, G.; Tombul, Ş.T.; Demirtaş, T. Comparison of pain levels in fusion prostate biopsy and standard TRUS-Guided biopsy. Int. Braz. J. Urol. 2020, 46, 557–562. [Google Scholar] [CrossRef] [PubMed]
  62. Radtke, J.P.; Schwab, C.; Wolf, M.B.; Freitag, M.T.; Alt, C.D.; Kesch, C.; Popeneciu, I.V.; Huettenbrink, C.; Gasch, C.; Klein, T.; et al. Multiparametric Magnetic Resonance Imaging (MRI) and MRI-Transrectal Ultrasound Fusion Biopsy for Index Tumor Detection: Correlation with Radical Prostatectomy Specimen. Eur. Urol. 2016, 70, 846–853. [Google Scholar] [CrossRef]
  63. Schoots, I.G.; Roobol, M.J.; Nieboer, D.; Bangma, C.H.; Steyerberg, E.W.; Hunink, M.G. Magnetic resonance imaging-targeted biopsy may enhance the diagnostic accuracy of significant prostate cancer detection compared to standard transrectal ultrasound-guided biopsy: A systematic review and meta-analysis. Eur. Urol. 2015, 68, 438–450. [Google Scholar] [CrossRef] [PubMed]
  64. Wegelin, O.; van Melick, H.H.E.; Hooft, L.; Bosch, J.; Reitsma, H.B.; Barentsz, J.O.; Somford, D.M. Comparing Three Different Techniques for Magnetic Resonance Imaging-targeted Prostate Biopsies: A Systematic Review of In-bore versus Magnetic Resonance Imaging-transrectal Ultrasound fusion versus Cognitive Registration. Is There a Preferred Technique? Eur. Urol. 2017, 71, 517–531. [Google Scholar] [CrossRef] [PubMed]
  65. Ouzzane, A.; Puech, P.; Lemaitre, L.; Leroy, X.; Nevoux, P.; Betrouni, N.; Haber, G.P.; Villers, A. Combined multiparametric MRI and targeted biopsies improve anterior prostate cancer detection, staging, and grading. Urology 2011, 78, 1356–1362. [Google Scholar] [CrossRef]
  66. Overduin, C.G.; Fütterer, J.J.; Barentsz, J.O. MRI-guided biopsy for prostate cancer detection: A systematic review of current clinical results. Curr. Urol. Rep. 2013, 14, 209–213. [Google Scholar] [CrossRef]
  67. Lee, D.J.; Recabal, P.; Sjoberg, D.D.; Thong, A.; Lee, J.K.; Eastham, J.A.; Scardino, P.T.; Vargas, H.A.; Coleman, J.; Ehdaie, B. Comparative Effectiveness of Targeted Prostate Biopsy Using Magnetic Resonance Imaging Ultrasound Fusion Software and Visual Targeting: A Prospective Study. J. Urol. 2016, 196, 697–702. [Google Scholar] [CrossRef] [Green Version]
  68. Winoker, J.S.; Wajswol, E.; Falagario, U.; Maritini, A.; Moshier, E.; Voutsinas, N.; Knauer, C.J.; Sfakianos, J.P.; Lewis, S.C.; Taouli, B.A.; et al. Transperineal Versus Transrectal Targeted Biopsy With Use of Electromagnetically-tracked MR/US Fusion Guidance Platform for the Detection of Clinically Significant Prostate Cancer. Urology 2020, 146, 278–286. [Google Scholar] [CrossRef]
  69. Salagierski, M.; Kania, P.; Wierzchołowski, W.; Poźniak-Balicka, R. The role of a template-assisted cognitive transperineal prostate biopsy technique in patients with benign transrectal prostate biopsies: A preliminary experience. Cent Eur. J. Urol. 2019, 72, 15–18. [Google Scholar]
  70. Puech, P.; Rouvière, O.; Renard-Penna, R.; Villers, A.; Devos, P.; Colombel, M.; Bitker, M.O.; Leroy, X.; Mège-Lechevallier, F.; Comperat, E.; et al. Prostate cancer diagnosis: Multiparametric MR-targeted biopsy with cognitive and transrectal US-MR fusion guidance versus systematic biopsy--prospective multicenter study. Radiology 2013, 268, 461–469. [Google Scholar] [CrossRef]
  71. Arsov, C.; Rabenalt, R.; Quentin, M.; Hiester, A.; Blondin, D.; Albers, P.; Antoch, G.; Schimmöller, L. Comparison of patient comfort between MR-guided in-bore and MRI/ultrasound fusion-guided prostate biopsies within a prospective randomized trial. World J. Urol. 2016, 34, 215–220. [Google Scholar] [CrossRef]
  72. Wysock, J.S.; Rosenkrantz, A.B.; Huang, W.C.; Stifelman, M.D.; Lepor, H.; Deng, F.M.; Melamed, J.; Taneja, S.S. A prospective, blinded comparison of magnetic resonance (MR) imaging-ultrasound fusion and visual estimation in the performance of MR-targeted prostate biopsy: The PROFUS trial. Eur. Urol. 2014, 66, 343–351. [Google Scholar] [CrossRef] [PubMed]
  73. Ghai, S.; Trachtenberg, J. MRI-guided biopsies and minimally invasive therapy for prostate cancer. Indian J. Urol. 2015, 31, 209–216. [Google Scholar] [CrossRef] [PubMed]
  74. Lam, T.B.L.; MacLennan, S.; Willemse, P.M.; Mason, M.D.; Plass, K.; Shepherd, R.; Baanders, R.; Bangma, C.H.; Bjartell, A.; Bossi, A.; et al. EAU-EANM-ESTRO-ESUR-SIOG Prostate Cancer Guideline Panel Consensus Statements for Deferred Treatment with Curative Intent for Localised Prostate Cancer from an International Collaborative Study (DETECTIVE Study). Eur. Urol. 2019, 76, 790–813. [Google Scholar] [CrossRef] [Green Version]
  75. Schiavina, R.; Vagnoni, V.; D’Agostino, D.; Borghesi, M.; Salvaggio, A.; Giampaoli, M.; Pultrone, C.V.; Saraceni, G.; Gaudiano, C.; Vigo, M.; et al. “In-bore” MRI-guided Prostate Biopsy Using an Endorectal Nonmagnetic Device: A Prospective Study of 70 Consecutive Patients. Clin. Genitourin. Cancer 2017, 15, 417–427. [Google Scholar] [CrossRef]
  76. Crocerossa, F.; Marchioni, M.; Novara, G.; Carbonara, U.; Ferro, M.; Russo, G.I.; Porpiglia, F.; Di Nicola, M.; Damiano, R.; Autorino, R.; et al. Detection Rate of Prostate Specific Membrane Antigen Tracers for Positron Emission Tomography/Computerized Tomography in Prostate Cancer Biochemical Recurrence: A Systematic Review and Network Meta-Analysis. J. Urol. 2021, 205, 356–369. [Google Scholar] [CrossRef]
  77. de Kouchkovsky, I.; Aggarwal, R.; Hope, T.A. Prostate-specific membrane antigen (PSMA)-based imaging in localized and advanced prostate cancer: A narrative review. Transl. Androl. Urol. 2021, 10, 3130–3143. [Google Scholar] [CrossRef] [PubMed]
  78. Hupe, M.C.; Philippi, C.; Roth, D.; Kümpers, C.; Ribbat-Idel, J.; Becker, F.; Joerg, V.; Duensing, S.; Lubczyk, V.H.; Kirfel, J.; et al. Expression of Prostate-Specific Membrane Antigen (PSMA) on Biopsies Is an Independent Risk Stratifier of Prostate Cancer Patients at Time of Initial Diagnosis. Front. Oncol. 2018, 8, 623. [Google Scholar] [CrossRef]
  79. Evangelista, L.; Zattoni, F.; Cassarino, G.; Artioli, P.; Cecchin, D.; Dal Moro, F.; Zucchetta, P. PET/MRI in prostate cancer: A systematic review and meta-analysis. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 859–873. [Google Scholar] [CrossRef]
  80. Cook, G.J.; Fogelman, I. The role of positron emission tomography in the management of bone metastases. Cancer 2000, 88 (Suppl. 12), 2927–2933. [Google Scholar] [CrossRef]
  81. Jadvar, H. Is There Use for FDG-PET in Prostate Cancer? Semin. Nucl. Med. 2016, 46, 502–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Hess, S.; Blomberg, B.A.; Zhu, H.J.; Høilund-Carlsen, P.F.; Alavi, A. The pivotal role of FDG-PET/CT in modern medicine. Acad. Radiol. 2014, 21, 232–249. [Google Scholar] [CrossRef]
  83. Jadvar, H. Imaging evaluation of prostate cancer with 18F-fluorodeoxyglucose PET/CT: Utility and limitations. Eur. J. Nucl. Med. Mol. Imaging 2013, 40 (Suppl. 1), S5–S10. [Google Scholar] [CrossRef]
  84. Watanabe, H.; Kanematsu, M.; Kondo, H.; Kako, N.; Yamamoto, N.; Yamada, T.; Goshima, S.; Hoshi, H.; Bae, K.T. Preoperative detection of prostate cancer: A comparison with 11C-choline PET, 18F-fluorodeoxyglucose PET and MR imaging. J. Magn. Reson Imaging 2010, 31, 1151–1156. [Google Scholar] [CrossRef]
  85. Carroll, P.H.; Mohler, J.L. NCCN Guidelines Updates: Prostate Cancer and Prostate Cancer Early Detection. J. Natl. Compr. Cancer Netw. 2018, 16, 620–623. [Google Scholar] [CrossRef] [Green Version]
  86. Segall, G.; Delbeke, D.; Stabin, M.G.; Even-Sapir, E.; Fair, J.; Sajdak, R.; Smith, G.T. SNM practice guideline for sodium 18F-fluoride PET/CT bone scans 1.0. J. Nucl. Med. 2010, 51, 1813–1820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Fanti, S.; Minozzi, S.; Castellucci, P.; Balduzzi, S.; Herrmann, K.; Krause, B.J.; Oyen, W.; Chiti, A. PET/CT with (11)C-choline for evaluation of prostate cancer patients with biochemical recurrence: Meta-analysis and critical review of available data. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, 55–69. [Google Scholar] [CrossRef] [PubMed]
  88. Zeisel, S.H. Dietary choline: Biochemistry, physiology, and pharmacology. Annu. Rev. Nutr. 1981, 1, 95–121. [Google Scholar] [CrossRef]
  89. Cornford, P.; Bellmunt, J.; Bolla, M.; Briers, E.; De Santis, M.; Gross, T.; Henry, A.M.; Joniau, S.; Lam, T.B.; Mason, M.D.; et al. EAU-ESTRO-SIOG Guidelines on Prostate Cancer. Part II: Treatment of Relapsing, Metastatic, and Castration-Resistant Prostate Cancer. Eur. Urol. 2017, 71, 630–642. [Google Scholar] [CrossRef]
  90. Rais-Bahrami, S.; Efstathiou, J.A.; Turnbull, C.M.; Camper, S.B.; Kenwright, A.; Schuster, D.M.; Scarsbrook, A.F. (18)F-Fluciclovine PET/CT performance in biochemical recurrence of prostate cancer: A systematic review. Prostate Cancer Prostatic Dis. 2021, 24, 997–1006. [Google Scholar] [CrossRef]
  91. Bach-Gansmo, T.; Nanni, C.; Nieh, P.T.; Zanoni, L.; Bogsrud, T.V.; Sletten, H.; Korsan, K.A.; Kieboom, J.; Tade, F.I.; Odewole, O.; et al. Multisite Experience of the Safety, Detection Rate and Diagnostic Performance of Fluciclovine ((18)F) Positron Emission Tomography/Computerized Tomography Imaging in the Staging of Biochemically Recurrent Prostate Cancer. J. Urol. 2017, 19 Pt 1, 676–683. [Google Scholar] [CrossRef] [Green Version]
  92. Andriole, G.L.; Kostakoglu, L.; Chau, A.; Duan, F.; Mahmood, U.; Mankoff, D.A.; Schuster, D.M.; Siegel, B.A. The Impact of Positron Emission Tomography with 18F-Fluciclovine on the Treatment of Biochemical Recurrence of Prostate Cancer: Results from the LOCATE Trial. J. Urol. 2019, 201, 322–331. [Google Scholar] [CrossRef]
  93. Savir-Baruch, B.; Banks, K.P.; McConathy, J.E.; Molchanova-Cook, O.P.; Parent, E.E.; Takalkar, A.; Tulchinsky, M.; Yu, J.Q.; Subramaniam, R.M.; Schuster, D.M. ACR-ACNM Practice Parameter for the Performance of Fluorine-18 Fluciclovine-PET/CT for Recurrent Prostate Cancer. Clin. Nucl. Med. 2018, 43, 909–917. [Google Scholar] [CrossRef] [Green Version]
  94. England, J.R.; Paluch, J.; Ballas, L.K.; Jadva, H. 18F-Fluciclovine PET/CT Detection of Recurrent Prostate Carcinoma in Patients With Serum PSA ≤ 1 ng/mL After Definitive Primary Treatment. Clin. Nucl. Med. 2019, 44, e128–e132. [Google Scholar] [CrossRef] [PubMed]
  95. Nanni, C.; Schiavina, R.; Brunocilla, E.; Boschi, S.; Borghesi, M.; Zanoni, L.; Pettinato, C.; Martorana, G.; Fanti, S. 18F-Fluciclovine PET/CT for the Detection of Prostate Cancer Relapse: A Comparison to 11C-Choline PET/CT. Clin. Nucl. Med. 2015, 40, e386–e391. [Google Scholar] [CrossRef] [PubMed]
  96. Odewole, O.A.; Tade, F.I.; Nieh, P.T.; Savir-Baruch, B.; Jani, A.B.; Master, V.A.; Rossi, P.J.; Halkar, R.K.; Osunkoya, A.O.; Akin-Akintayo, O.; et al. Recurrent prostate cancer detection with anti-3-[(18)F]FACBC PET/CT: Comparison with CT. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, 1773–1783. [Google Scholar] [CrossRef] [Green Version]
  97. Larson, S.M.; Morris, M.; Gunther, I.; Beattie, B.; Humm, J.L.; Akhurst, T.A.; Finn, R.D.; Erdi, Y.; Pentlow, K.; Dyke, J.; et al. Tumor localization of 16beta-18F-fluoro-5alpha-dihydrotestosterone versus 18F-FDG in patients with progressive, metastatic prostate cancer. J. Nucl. Med. 2004, 45, 366–373. [Google Scholar]
  98. Vargas, H.A.; Kramer, G.M.; Scott, A.M.; Weickhardt, A.; Meier, A.A.; Parada, N.; Beattie, B.J.; Humm, J.L.; Staton, K.D.; Zanzonico, P.B.; et al. Reproducibility and Repeatability of Semiquantitative (18)F-Fluorodihydrotestosterone Uptake Metrics in Castration-Resistant Prostate Cancer Metastases: A Prospective Multicenter Study. J. Nucl. Med. 2018, 59, 1516–1523. [Google Scholar] [CrossRef] [Green Version]
  99. Evangelista, L.; Guttilla, A.; Zattoni, F.; Muzzio, P.C.; Zattoni, F. 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. [Google Scholar] [CrossRef] [PubMed]
  100. Beheshti, M.; Imamovic, L.; Broinger, G.; Vali, R.; Waldenberger, P.; Stoiber, F.; Nader, M.; Gruy, B.; Janetschek, G.; Langsteger, W. 18F choline PET/CT in the preoperative staging of prostate cancer in patients with intermediate or high risk of extracapsular disease: A prospective study of 130 patients. Radiology 2010, 254, 925–933. [Google Scholar] [CrossRef] [Green Version]
  101. Liu, C.; Liu, T.; Zhang, N.; Liu, Y.; Li, N.; Du, P.; Yang, Y.; Liu, M.; Gong, K.; Yang, X.; et al. 68Ga-PSMA-617 PET/CT: A promising new technique for predicting risk stratification and metastatic risk of prostate cancer patients. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 1852–1861. [Google Scholar] [CrossRef]
  102. Sweat, S.D.; Pacelli, A.; Murphy, G.P.; Bostwick, D.G. Prostate-specific membrane antigen expression is greatest in prostate adenocarcinoma and lymph node metastases. Urology 1998, 52, 637–640. [Google Scholar] [CrossRef]
  103. Marchal, C.; Redondo, M.; Padilla, M.; Caballero, J.; Rodrigo, I.; García, J.; Quian, J.; Boswick, D.G. Expression of prostate specific membrane antigen (PSMA) in prostatic adenocarcinoma and prostatic intraepithelial neoplasia. Histol. Histopathol. 2004, 19, 715–718. [Google Scholar]
  104. Lauri, C.; Chiurchioni, L.; Russo, V.M.; Zannini, L.; Signore, A. PSMA Expression in Solid Tumors beyond the Prostate Gland: Ready for Theranostic Applications? J. Clin. Med. 2022, 11, 6590. [Google Scholar] [CrossRef] [PubMed]
  105. Fendler, W.P.; Schmidt, D.F.; Wenter, V.; Thierfelder, K.M.; Zach, C.; Stief, C.; Bartenstein, P.; Kirchner, T.; Gildehaus, F.J.; Gratzke, C.; et al. 68Ga-PSMA PET/CT Detects the Location and Extent of Primary Prostate Cancer. J. Nucl. Med. 2016, 57, 1720–1725. [Google Scholar] [CrossRef] [Green Version]
  106. Tourinho-Barbosa, R.; Srougi, V.; Nunes-Silva, I.; Baghdadi, M.; Rembeyo, G.; Eiffel, S.S.; Barret, E.; Rozet, F.; Galiano, M.; Cathelineau, X.; et al. Biochemical recurrence after radical prostatectomy: What does it mean? Int. Braz. J. Urol. 2018, 44, 14–21. [Google Scholar] [CrossRef] [Green Version]
  107. Hoffmann, M.A.; von Eyben, F.E.; Fischer, N.; Rosar, J. Müller-Hübenthal, F.; Buchholz, H.G.; Wieler, H.J.; Schreckenberger, M. Comparison of [(18)F]PSMA-1007 with [(68)Ga]Ga-PSMA-11 PET/CT in Restaging of Prostate Cancer Patients with PSA Relapse. Cancers 2022, 14, 1479. [Google Scholar] [CrossRef] [PubMed]
  108. Calais, J.; Ceci, F.; Eiber, M.; Hope, T.A.; Hofman, M.S.; Rischpler, C.; Bach-Gansmo, T.; Nanni, C.; Savir-Baruch, B.; Elashoff, D.; et al. (18)F-fluciclovine PET-CT and (68)Ga-PSMA-11 PET-CT in patients with early biochemical recurrence after prostatectomy: A prospective, single-centre, single-arm, comparative imaging trial. Lancet Oncol. 2019, 20, 1286–1294. [Google Scholar] [CrossRef] [PubMed]
  109. Tan, N.; Oyoyo, U.; Bavadian, N.; Ferguson, N.; Mukkamala, A.; Calais, J.; Davenport, M.S. PSMA-targeted Radiotracers versus (18)F Fluciclovine for the Detection of Prostate Cancer Biochemical Recurrence after Definitive Therapy: A Systematic Review and Meta-Analysis. Radiology 2020, 296, 44–55. [Google Scholar] [CrossRef] [PubMed]
  110. Perera, M.; Papa, N.; Roberts, M.; Williams, M.; Udovicich, C.; Vela, I.; Christidis, D.; Bolton, D.; Hofman, M.S.; Lawrentschuk, N.; et al. Gallium-68 Prostate-specific Membrane Antigen Positron Emission Tomography in Advanced Prostate Cancer-Updated Diagnostic Utility, Sensitivity, Specificity, and Distribution of Prostate-specific Membrane Antigen-avid Lesions: A Systematic Review and Meta-analysis. Eur. Urol. 2020, 77, 403–417. [Google Scholar]
  111. le Guevelou, J.; Achard, V.; Mainta, I.; Zaidi, H.; Garibotto, V.; Latorzeff, I.; Sargos, P.; Ménard, C.; Zilli, T. PET/CT-Based Salvage Radiotherapy for Recurrent Prostate Cancer After Radical Prostatectomy: Impact on Treatment Management and Future Directions. Front. Oncol. 2021, 11, 742093. [Google Scholar] [CrossRef]
  112. Fendler, W.P.; Weber, M.; Iravani, A.; Hofman, M.S.; Calais, J.; Czernin, J.; Ilhan, H.; Saad, F.; Small, E.J.; Smith, M.R.; et al. Prostate-Specific Membrane Antigen Ligand Positron Emission Tomography in Men with Nonmetastatic Castration-Resistant Prostate Cancer. Clin. Cancer Res. 2019, 25, 7448–7454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Jilg, C.A.; Drendel, V.; Rischke, H.C.; Beck, T.I.; Reichel, K.; Krönig, M.; Wetterauer, U.; Schultze-Seemann, W.; Meyer, P.T.; Vach, W. Detection Rate of (18)F-Choline PET/CT and (68)Ga-PSMA-HBED-CC PET/CT for Prostate Cancer Lymph Node Metastases with Direct Link from PET to Histopathology: Dependence on the Size of Tumor Deposits in Lymph Nodes. J. Nucl. Med. 2019, 60, 971–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Schmidt-Hegemann, N.S.; Eze, C.; Li, M.; Rogowski, P.; Schaefer, C.; Stief, C.; Buchner, A.; Zamboglou, C.; Fendler, W.P.; Ganswindt, U.; et al. Impact of (68)Ga-PSMA PET/CT on the Radiotherapeutic Approach to Prostate Cancer in Comparison to CT: A Retrospective Analysis. J. Nucl. Med. 2019, 60, 963–970. [Google Scholar] [CrossRef] [Green Version]
  115. Giesel, F.L.; Will, L.; Kesch, C.; Freitag, M.; Kremer, C.; Merkle, J.; Neels, O.C.; Cardinale, J.; Hadaschik, B.; Hohenfellner, M.; et al. Biochemical Recurrence of Prostate Cancer: Initial Results with [(18)F]PSMA-1007 PET/CT. J. Nucl. Med. 2018, 59, 632–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Wang, X.; Wen, Q.; Zhang, H.; Ji, B. Head-to-Head Comparison of (68)Ga-PSMA-11 PET/CT and Multiparametric MRI for Pelvic Lymph Node Staging Prior to Radical Prostatectomy in Patients With Intermediate to High-Risk Prostate Cancer: A Meta-Analysis. Front. Oncol. 2021, 11, 737989. [Google Scholar] [CrossRef] [PubMed]
  117. Eiber, M.; Weirich, G.; Holzapfel, K.; Souvatzoglou, M.; Haller, B.; Rauscher, I.; Beer, A.J.; Wester, H.J.; Gschwend, J.; Schwaiger, M.; et al. Simultaneous (68)Ga-PSMA HBED-CC PET/MRI Improves the Localization of Primary Prostate Cancer. Eur. Urol. 2016, 70, 829–836. [Google Scholar] [CrossRef]
  118. Park, S.Y.; Zacharias, C.; Harrison, C.; Fan, R.E.; Kunder, C.; Hatami, N.; Giesel, F.; Ghanouni, P.; Daniel, B.; Loening, A.M.; et al. Gallium 68 PSMA-11 PET/MR Imaging in Patients with Intermediate- or High-Risk Prostate Cancer. Radiology 2018, 288, 495–505. [Google Scholar] [CrossRef] [Green Version]
  119. Hofman, M.S.; Lawrentschuk, N.; Francis, R.J.; Tang, C.; Vela, I.; Thomas, P.; Rutherford, N.; Martin, J.M.; Frydenberg, M.; Shakher, R.; et al. Prostate-specific membrane antigen PET-CT in patients with high-risk prostate cancer before curative-intent surgery or radiotherapy (proPSMA): A prospective, randomised, multicentre study. Lancet 2020, 395, 1208–1216. [Google Scholar] [CrossRef]
  120. Klingenberg, S.; Jochumsen, M.R.; Ulhøi, B.P.; Fredsøe, J.; Sørensen, K.D.; Borre, M.; Bouchelouche, K. (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. [Google Scholar] [CrossRef]
  121. Cytawa, W.; Seitz, A.K.; Kircher, S.; Fukushima, K.; Tran-Gia, J.; Schirbel, A.; Bandurski, T.; Lass, P.; Krebs, M.; Połom, W.; 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. [Google Scholar] [PubMed]
  122. Kuten, J.; Fahoum, I.; Savin, Z.; Shamni, O.; Gitstein, G.; Hershkovitz, D.; Mabjeesh, N.J.; Yossepowitch, O.; Mishani, E.; Even-Sapir, E. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Alberts, I.; Niklas-Hünermund, J.; Sachpekidis, C.; Zacho, H.D.; Mingels, C.; Dijkstra, L.; Bohn, K.P.; Läppchen, T.; Gourni, E.; Rominger, A.; et al. Combination of Forced Diuresis with Additional Late Imaging in (68)Ga-PSMA-11 PET/CT: Effects on Lesion Visibility and Radiotracer Uptake. J. Nucl. Med. 2021, 62, 1252–1257. [Google Scholar] [CrossRef]
  124. Kesch, C.; Kratochwil, C.; Mier, W.; Kopka, K.; Giesel, F.L. (68)Ga or (18)F for Prostate Cancer Imaging? J. Nucl. Med. 2017, 58, 687–688. [Google Scholar] [CrossRef] [Green Version]
  125. Rauscher, I.; Krönke, M.; König, M.; Gafita, A.; Maurer, T.; Horn, T.; Schiller, K.; Weber, W.; Eiber, M. Matched-Pair Comparison of (68)Ga-PSMA-11 PET/CT and (18)F-PSMA-1007 PET/CT: Frequency of Pitfalls and Detection Efficacy in Biochemical Recurrence After Radical Prostatectomy. J. Nucl. Med. 2020, 61, 51–57. [Google Scholar] [CrossRef]
  126. Fendler, W.P.; Eiber, M.; Beheshti, M.; Bomanji, J.; Ceci, F.; Cho, S.; Giesel, F.; Haberkorn, U.; Hope, T.A.; Kopka, K.; et al. (68)Ga-PSMA PET/CT: Joint EANM and SNMMI procedure guideline for prostate cancer imaging: Version 1.0. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 1014–1024. [Google Scholar] [CrossRef] [PubMed]
  127. Donato, P.; Morton, A.; Yaxley, J.; Ranasinghe, S.; Teloken, P.E.; Kyle, S.; Coughlin, G.; Esler, R.; Dunglison, N.; Gardiner, R.A.; et al. (68)Ga-PSMA PET/CT better characterises localised prostate cancer after MRI and transperineal prostate biopsy: Is (68)Ga-PSMA PET/CT guided biopsy the future? Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 1843–1851. [Google Scholar] [CrossRef]
  128. Zhang, L.L.; Li, W.C.; Xu, Z.; Jiang, N.; Zang, S.M.; Xu, L.W.; Huang, W.B.; Wang, F.; Sun, H.B. (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. [Google Scholar] [CrossRef]
  129. Simopoulos, D.N.; Natarajan, S.; Jones, T.A.; Fendler, W.P.; Sisk, A.E., Jr.; Marks, L.S. Targeted Prostate Biopsy Using (68)Gallium PSMA-PET/CT for Image Guidance. Urol. Case Rep. 2017, 14, 11–14. [Google Scholar] [CrossRef]
  130. Westenfelder, K.M.; Lentes, B.; Rackerseder, J.; Navab, N.; Gschwend, J.E.; Eiber, M.; Maurer, T. Gallium-68 HBED-CC-PSMA Positron Emission Tomography/Magnetic Resonance Imaging for Prostate Fusion Biopsy. Clin. Genitourin. Cancer 2018, 16, 245–247. [Google Scholar] [CrossRef]
  131. Donato, P.; Roberts, M.J.; Morton, A.; Kyle, S.; Coughlin, G.; Esler, R.; Dunglison, N.; Gardiner, R.A.; Yaxley, J. Improved specificity with (68)Ga PSMA PET/CT to detect clinically significant lesions “invisible” on multiparametric MRI of the prostate: A single institution comparative analysis with radical prostatectomy histology. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 20–30. [Google Scholar] [CrossRef]
  132. Woythal, N.; Arsenic, R.; Kempkensteffen, C.; Miller, K.; Janssen, J.C.; Huang, K.; Makowski, M.R.; Brenner, W.; Prasad, V. Immunohistochemical Validation of PSMA Expression Measured by (68)Ga-PSMA PET/CT in Primary Prostate Cancer. J. Nucl. Med. 2018, 59, 238–243. [Google Scholar] [CrossRef] [Green Version]
  133. Caracciolo, M.; Castello, A.; Urso, L.; Borgia, F.; Ortolan, N.; Uccelli, L.; Cittanti, C.; Castellani, M.; Bartolomei, M.; Lazzeri, M.; et al. The Role of [(68)Ga]PSMA PET/CT for Clinical Suspicion of Prostate Cancer in Patients with or without Previous Negative Biopsy: A Systematic Review. Cancers 2022, 14, 5036. [Google Scholar] [CrossRef]
  134. Ferraro, D.A.; Becker, A.S.; Kranzbühler, B.; Mebert, I.; Baltensperger, A.; Zeimpekis, K.G.; Grünig, H.; Messerli, M.; Rupp, N.J.; Rueschoff, J.H.; et al. Diagnostic performance of (68)Ga-PSMA-11 PET/MRI-guided biopsy in patients with suspected prostate cancer: A prospective single-center study. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 3315–3324. [Google Scholar] [CrossRef] [PubMed]
  135. Uprimny, C.; Kroiss, A.S.; Decristoforo, C.; Fritz, J.; von Guggenberg, E.; Kendler, D.; Scarpa, L.; di Santo, G.; Roig, L.G.; Maffey-Steffan, J.; et al. (68)Ga-PSMA-11 PET/CT in primary staging of prostate cancer: PSA and Gleason score predict the intensity of tracer accumulation in the primary tumour. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 941–949. [Google Scholar] [CrossRef] [PubMed]
  136. Sathekge, M.; Lengana, T.; Maes, A.; Vorster, M.; Zeevaart, J.; Lawal, I.; Ebenhan, T.; Van de Wiele, C. (68)Ga-PSMA-11 PET/CT in primary staging of prostate carcinoma: Preliminary results on differences between black and white South-Africans. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 226–234. [Google Scholar] [CrossRef] [Green Version]
  137. Kumar, R.; Singh, S.K.; Mittal, B.R.; Vadi, S.K.; Kakkar, N.; Singh, H.; Krishnaraju, V.S.; Kumar, S.; Bhattacharya, A. Safety and Diagnostic Yield of (68)Ga Prostate-specific Membrane Antigen PET/CT-guided Robotic-assisted Transgluteal Prostatic Biopsy. Radiology 2022, 303, 392–398. [Google Scholar] [CrossRef]
  138. Spohn, S.; Jaegle, C.; Fassbender, T.F.; Sprave, T.; Gkika, E.; Nicolay, N.H.; Bock, M.; Ruf, J.; Benndorf, M.; Gratzke, C.; et al. Intraindividual comparison between (68)Ga-PSMA-PET/CT and mpMRI for intraprostatic tumor delineation in patients with primary prostate cancer: A retrospective analysis in 101 patients. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 2796–2803. [Google Scholar] [CrossRef] [PubMed]
  139. Ponisio, M.R.; McConathy, J.; Laforest, R.; Khanna, G. Evaluation of diagnostic performance of whole-body simultaneous PET/MRI in pediatric lymphoma. Pediatr. Radiol. 2016, 46, 1258–1268. [Google Scholar] [CrossRef] [Green Version]
  140. Eiber, M.; Rauscher, I.; Souvatzoglou, M.; Maurer, T.; Schwaiger, M.; Holzapfel, K.; Beer, A.J. Prospective head-to-head comparison of (11)C-choline-PET/MR and (11)C-choline-PET/CT for restaging of biochemical recurrent prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 2179–2188. [Google Scholar] [CrossRef]
  141. Hope, T.A.; Goodman, J.Z.; Allen, I.E.; Calais, J.; Fendler, W.P.; Carroll, P.R. Metaanalysis of (68)Ga-PSMA-11 PET Accuracy for the Detection of Prostate Cancer Validated by Histopathology. J. Nucl. Med. 2019, 60, 786–793. [Google Scholar] [CrossRef] [Green Version]
  142. Kranzbühler, B.; Müller, J.; Becker, A.S.; Garcia Schüler, H.I.; Muehlematter, U.; Fankhauser, C.D.; Kedzia, S.; Guckenberger, M.; Kaufmann, P.A.; Eberli, D.; et al. Detection Rate and Localization of Prostate Cancer Recurrence Using (68)Ga-PSMA-11 PET/MRI in Patients with Low PSA Values ≤ 0.5 ng/mL. J. Nucl. Med. 2020, 61, 194–201. [Google Scholar] [CrossRef] [Green Version]
  143. Breen, W.G.; Stish, B.J.; Harmsen, W.S.; Froemming, A.T.; Mynderse, L.A.; Choo, C.R.; Davis, B.J.; Pisansky, T.M. The prognostic value, sensitivity, and specificity of multiparametric magnetic resonance imaging before salvage radiotherapy for prostate cancer. Radiother. Oncol. 2021, 161, 9–15. [Google Scholar] [CrossRef] [PubMed]
  144. Selnæs, K.M.; Krüger-Stokke, B.; Elschot, M.; Johansen, H.; Steen, P.A.; Langørgen, S.; Aksnessæther, B.Y.; Indrebø, G.; Sjøbakk, T.A.E.; Tessem, M.B.; et al. Detection of Recurrent Prostate Cancer With (18)F-Fluciclovine PET/MRI. Front. Oncol. 2020, 10, 582092. [Google Scholar] [CrossRef]
  145. Domachevsky, L.; Bernstine, H.; Goldberg, N.; Nidam, M.; Stern, D.; Sosna, J.; Groshar, D. Early (68)GA-PSMA PET/MRI acquisition: Assessment of lesion detectability and PET metrics in patients with prostate cancer undergoing same-day late PET/CT. Clin. Radiol. 2017, 72, 944–950. [Google Scholar] [CrossRef] [PubMed]
  146. Jambor, I.; Kuisma, A.; Kähkönen, E.; Kemppainen, J.; Merisaari, H.; Eskola, O.; Teuho, J.; Perez, I.M.; Pesola, M.; Aronen, H.J.; et al. Prospective evaluation of (18)F-FACBC PET/CT and PET/MRI versus multiparametric MRI in intermediate- to high-risk prostate cancer patients (FLUCIPRO trial). Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 355–364. [Google Scholar] [CrossRef] [PubMed]
  147. van Timmeren, J.E.; Cester, D.; Tanadini-Lang, S.; Alkadhi, H.; Baessler, B. Radiomics in medical imaging-”how-to” guide and critical reflection. Insights Imaging 2020, 11, 91. [Google Scholar] [CrossRef] [PubMed]
  148. Dai, Z.; Carver, E.; Liu, C.; Lee, J.; Feldman, A.; Zong, W.; Pantelic, M.; Elshaikh, M.; Wen, N. Segmentation of the Prostatic Gland and the Intraprostatic Lesions on Multiparametic Magnetic Resonance Imaging Using Mask Region-Based Convolutional Neural Networks. Adv. Radiat. Oncol. 2020, 5, 473–481. [Google Scholar] [CrossRef] [Green Version]
  149. Arif, M.; Schoots, I.G.; Castillo Tovar, J.; Bangma, C.H.; Krestin, G.P.; Roobol, M.J.; Niessen, W.; Veenland, J.F. Clinically significant prostate cancer detection and segmentation in low-risk patients using a convolutional neural network on multi-parametric MRI. Eur. Radiol. 2020, 30, 6582–6592. [Google Scholar] [CrossRef]
  150. Pellicer-Valero, O.J.; Marenco Jiménez, J.L.; Gonzalez-Perez, V.; Casanova Ramón-Borja, J.L.; Martín García, I.; Barrios Benito, M.; Pelechano Gómez, P.; Rubio-Briones, J.; Rupérez, M.J.; Martín-Guerrero, J.D. Deep learning for fully automatic detection, segmentation, and Gleason grade estimation of prostate cancer in multiparametric magnetic resonance images. Sci. Rep. 2022, 12, 2975. [Google Scholar] [CrossRef]
  151. Gillies, R.J.; Kinahan, P.E.; Hricak, H. Radiomics: Images Are More than Pictures, They Are Data. Radiology 2016, 278, 563–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Xu, M.; Fang, M.; Zou, J.; Yang, S.; Yu, D.; Zhong, L.; Hu, C.; Zang, Y.; Dong, D.; Tian, J.; et al. Using biparametric MRI radiomics signature to differentiate between benign and malignant prostate lesions. Eur. J. Radiol. 2019, 114, 38–44. [Google Scholar] [CrossRef] [PubMed]
  153. Ginsburg, S.B.; Algohary, A.; Pahwa, S.; Gulani, V.; Ponsky, L.; Aronen, H.J.; Boström, P.J.; Böhm, M.; Haynes, A.M.; Brenner, P.; et al. Radiomic features for prostate cancer detection on MRI differ between the transition and peripheral zones: Preliminary findings from a multi-institutional study. J. Magn. Reson Imaging 2017, 46, 184–193. [Google Scholar] [CrossRef] [PubMed]
  154. Cameron, A.; Khalvati, F.; Haider, M.A.; Wong, A. MAPS: A Quantitative Radiomics Approach for Prostate Cancer Detection. IEEE Trans. Biomed. Eng. 2016, 63, 1145–1156. [Google Scholar] [CrossRef] [PubMed]
  155. Hectors, S.J.; Chen, C.; Chen, J.; Wang, J.; Gordon, S.; Yu, M.; Al Hussein Al Awamlh, B.; Sabuncu, M.R.; Margolis, D.J.A.; Hu, J.C. Magnetic Resonance Imaging Radiomics-Based Machine Learning Prediction of Clinically Significant Prostate Cancer in Equivocal PI-RADS 3 Lesions. J. Magn. Reson Imaging 2021, 54, 1466–1473. [Google Scholar] [CrossRef]
  156. Min, X.; Li, M.; Dong, D.; Feng, Z.; Zhang, P.; Ke, Z.; You, H.; Han, F.; Ma, H.; Tian, J.; et al. Multi-parametric MRI-based radiomics signature for discriminating between clinically significant and insignificant prostate cancer: Cross-validation of a machine learning method. Eur. J. Radiol. 2019, 115, 16–21. [Google Scholar] [CrossRef] [Green Version]
  157. Zhang, Y.; Chen, W.; Yue, X.; Shen, J.; Gao, C.; Pang, P.; Cui, F.; Xu, M. Development of a Novel, Multi-Parametric, MRI-Based Radiomic Nomogram for Differentiating Between Clinically Significant and Insignificant Prostate Cancer. Front. Oncol. 2020, 10, 888. [Google Scholar] [CrossRef]
  158. Stanzione, A.; Gambardella, M.; Cuocolo, R.; Ponsiglione, A.; Romeo, V.; Imbriaco, M. Prostate MRI radiomics: A systematic review and radiomic quality score assessment. Eur. J. Radiol. 2020, 129, 109095. [Google Scholar] [CrossRef]
  159. Cysouw, M.C.F.; Jansen, B.H.E.; van de Brug, T.; Oprea-Lager, D.E.; Pfaehler, E.; de Vries, B.M.; van Moorselaar, R.J.A.; Hoekstra, O.S.; Vis, A.N.; Boellaard, R. Machine learning-based analysis of [18F]DCFPyL PET radiomics for risk stratification in primary prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 340–349. [Google Scholar] [CrossRef]
  160. Transin, S.; Souchon, R.; Gonindard-Melodelima, C.; de Rozario, R.; Walker, P.; Funes de la Vega, M.; Loffroy, R.; Cormier, L.; Rouvière, O. Computer-aided diagnosis system for characterizing ISUP grade≥2 prostate cancers at multiparametric MRI: A cross-vendor evaluation. Diagn Interv Imaging 2019, 100, 801–811. [Google Scholar] [CrossRef]
  161. de Rooij, M.; Crienen, S.; Witjes, J.A.; Barentsz, J.O.; Rovers, M.M.; Grutters, J.P.C. Cost-effectiveness of Magnetic Resonance (MR) Imaging and MR-guided Targeted Biopsy Versus Systematic Transrectal Ultrasound–Guided Biopsy in Diagnosing Prostate Cancer: A Modelling Study from a Health Care Perspective. Eur. Urol. 2014, 66, 430–436. [Google Scholar] [CrossRef]
  162. Faria, R.; Soares, M.O.; Spackman, E.; Ahmed, H.U.; Brown, L.C.; Kaplan, R.; Emberton, M.; Sculpher, M.J. Optimising the Diagnosis of Prostate Cancer in the Era of Multiparametric Magnetic Resonance Imaging: A Cost-effectiveness Analysis Based on the Prostate MR Imaging Study (PROMIS). Eur. Urol. 2018, 73, 23–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Song, R.; Jeet, V.; Sharma, R.; Hoyle, M.; Parkinson, B. Cost-Effectiveness Analysis of Prostate-Specific Membrane Antigen (PSMA) Positron Emission Tomography/Computed Tomography (PET/CT) for the Primary Staging of Prostate Cancer in Australia. PharmacoEconomics 2022, 40, 807–821. [Google Scholar] [CrossRef] [PubMed]
  164. Subramanian, K.; Martinez, J.; Castellanos, S.H.; Ivanidze, J.; Nagar, H.; Nicholson, S.; Youn, T.; Nauseef, J.T.; Tagawa, S.; Osborne, J.R. Complex implementation factors demonstrated when evaluating cost-effectiveness and monitoring racial disparities associated with [18F]DCFPyL PET/CT in prostate cancer men. Sci. Rep. 2023, 13, 8321. [Google Scholar] [CrossRef] [PubMed]
  165. Gordon, L.G.; Elliott, T.M.; Joshi, A.; Williams, E.D.; Vela, I. Exploratory cost-effectiveness analysis of 68Gallium-PSMA PET/MRI-based imaging in patients with biochemical recurrence of prostate cancer. Clin. Amp; Exp. Metastasis 2020, 37, 305–312. [Google Scholar] [CrossRef] [PubMed]
  166. Parikh, N.R.; Johnson, D.; Raldow, A.; Steinberg, M.L.; Czernin, J.; Nickols, N.G.; Calais, J.; Kishan, A.U.; Royce, T.J. Cost-effectiveness of 68Ga-PSMA-11 PET/CT in Prostate Cancer Patients with Biochemical Recurrence. Int. J. Radiat. Oncol. Biol. Phys. 2020, 108, S144–S145. [Google Scholar] [CrossRef]
Table 1. Summaries of the up-to-date imaging and diagnostic techniques for prostate cancer.
Table 1. Summaries of the up-to-date imaging and diagnostic techniques for prostate cancer.
MethodsStudiesYearTotal PatientsCharactersUsage
2D TRUSBeemsterboer, P.M., et al. [12]19998600Clear imaging and easy operation;
poor SE and SP.
Preferred method for guiding SB
CT——————Use in metastases of lymph node and bone;
easy accessibility and low cost.
AUA recommendations for intermediate- to high-risk PCa.
BS——————For metastases: SE 79% (95% CI: 73–83%), SP 82% (95% CI: 78–85%) [22].The standard and most-used method for bone metastases of intermediate- to high-risk PCa.
mpMRIUkimura, O., et al. [27]
Kasivisvanathan, V., et al. [29]
Oishi, M., et al. [33]
2015–2019762SE ranged between 44% and 93%,
SP ranged between 38% and 94% [31];
interobserver variability.
Routine application in biopsy-naïve patients;
conduction before repeat biopsy to select target lesions.
mpMRI TB + SBAhdoot, M., et al. [60]
Radtke, J.P., et al. [62]
2017–20202223Detection rate of 97% for csPCa, superior to mpMRI (85%), TB (78%), and SB (88%) [62];
increase of the quantity of cores and total biopsy time.
Recommended as the best available approach to reduce the csPCa misdiagnosis.
PET/CT
(18 F-Fluciclovine)
Bach-Gansmo, T., et al. [91]
Andriole, G.L., et al. [92]
England, J.R., et al. [94]
Nanni, C., et al. [95]
Odewole, O.A., et al. [96]
2015–2019940For patients with BCR, 68% detection rate, 62% PPV, 70% SP [91].FDA and European Commission approval in patients with elevated PSA for BCR;
no lower-level cutoff for PSA explored as an indication.
PSMA PETLiu, C., et al. [101]
Fendler, W.P., et al. [105]
Hoffmann, M.A., et al. [107]
Calais, J., et al. [108]
Fendler, W.P., et al. [112]
Jilg, C.A., et al. [113]
Schmidt-Hegemann, N.S., et al. [114]
Giesel, F.L., et al. [115]
Hofman, M.S., et al. [119]
Klingenberg, S., et al. [120]
Cytawa, W., et al. [121]
Donato, P., et al. [131]
2016–20221986Patient-based PCa BCR detection rates of 33% and 45% at PSA < 0.2 and 0.2–0.49 ng/mL [110];
for pelvic lymph node staging prior to RP in intermediate- to high-risk PCa, SE 71%, SP 92% [116].
For patients with a >0.2 ng/mL PSA according to EAU guidelines;
no recommendations for initial staging.
PET-targetDonato, P., et al. [127]
Zhang, L.L., et al. [128]
Ferraro, D.A., et al. [134]
Kumar, R., et al. [137]
2020–2022384Detecting csPCa with patient-based SE 96%, SP 81%, NPV 93%, PPV 89%, and accuracy 90% [134];
decrease in unnecessary biopsies and complications.
Only recommended in patients with previous negative biopsy;
a promising tool in the future diagnosis.
PET/MRIEiber, M., et al. [117]
Park, S.Y., et al. [118]
Eiber, M., et al. [140]
Kranzbühler, B., et al. [142]
Jambor, I., et al. [146]
2016–2020253SE 74%, SP 96% [141];
higher accuracy, especially for PSA < 0.5 ng/mL.
Limitations as to the cost and lack of data.
SE = sensitivity; SP = specificity; PPV = positive predictive value; NPV = negative predictive value; PCa = prostate cancer; csPCa = clinically significant prostate cancer; BCR = biochemical recurrence; PSA = prostate specific antigen; RP= radical prostatectomy; 2D TRUS = two-dimensional transrectal ultrasound; CT = computed tomography; BS = bone scintigraphy; mpMRI = multiparametric magnetic resonance imaging; TB = targeted biopsy; SB = systematic biopsy; PET/CT = positron emission tomography computed tomography; PSMA = prostate-specific membrane antigen; PET/MRI = positron emission tomography magnetic resonance imaging.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhu, M.; Liang, Z.; Feng, T.; Mai, Z.; Jin, S.; Wu, L.; Zhou, H.; Chen, Y.; Yan, W. Up-to-Date Imaging and Diagnostic Techniques for Prostate Cancer: A Literature Review. Diagnostics 2023, 13, 2283. https://doi.org/10.3390/diagnostics13132283

AMA Style

Zhu M, Liang Z, Feng T, Mai Z, Jin S, Wu L, Zhou H, Chen Y, Yan W. Up-to-Date Imaging and Diagnostic Techniques for Prostate Cancer: A Literature Review. Diagnostics. 2023; 13(13):2283. https://doi.org/10.3390/diagnostics13132283

Chicago/Turabian Style

Zhu, Ming, Zhen Liang, Tianrui Feng, Zhipeng Mai, Shijie Jin, Liyi Wu, Huashan Zhou, Yuliang Chen, and Weigang Yan. 2023. "Up-to-Date Imaging and Diagnostic Techniques for Prostate Cancer: A Literature Review" Diagnostics 13, no. 13: 2283. https://doi.org/10.3390/diagnostics13132283

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

Article Metrics

Back to TopTop