Skip to main content

Main menu

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI

User menu

  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Nuclear Medicine
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Journal of Nuclear Medicine

Advanced Search

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • View or Listen to JNM Podcast
  • Visit JNM on Facebook
  • Join JNM on LinkedIn
  • Follow JNM on Twitter
  • Subscribe to our RSS feeds
Research ArticleContinuing Education

Treatment Response Evaluation in Prostate Cancer Using PSMA PET/CT

Andrei Gafita, Jennifer A. Schroeder, Francesco Ceci, Jorge D. Oldan, Amir H. Khandani, Frederic E. Lecouvet, Lilja B. Solnes and Steven P. Rowe
Journal of Nuclear Medicine June 2025, jnumed.124.268071; DOI: https://doi.org/10.2967/jnumed.124.268071
Andrei Gafita
1Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland;
2Johns Hopkins Theranostics Center, Baltimore, Maryland;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jennifer A. Schroeder
3Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, North Carolina;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Francesco Ceci
4Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milan, Italy; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jorge D. Oldan
3Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, North Carolina;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Amir H. Khandani
3Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, North Carolina;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Frederic E. Lecouvet
5Department of Medical Imaging, Institut de Recherche Expérimentale et Clinique, Institut du Cancer Roi Albert II, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lilja B. Solnes
1Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland;
2Johns Hopkins Theranostics Center, Baltimore, Maryland;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Steven P. Rowe
3Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, North Carolina;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

In recent years, there has been a headlong rush into the use of prostate-specific membrane antigen (PSMA)–targeted PET for the staging and restaging of men with prostate cancer (PC). To date, there have been regulatory approvals for PSMA PET for purposes of initial staging, recurrence, and establishing eligibility for PSMA-targeted radiopharmaceutical therapy. Conventional imaging modalities, including bone scan and CT, are inadequate for identifying sites of PC in a variety of clinical scenarios. Further, current standardized response assessment approaches based on either conventional imaging or PET radiotracers that lack sensitivity for PC are inappropriate for response assessment in men with PC. There is currently no specific regulatory approval for the use of PSMA PET for response assessment. In the context of the use of PSMA-targeted radiopharmaceutical therapy and other cytotoxic therapeutic approaches, both the PSMA PET progression criteria and RECIP 1.0 have been shown to have value and to provide prognostic information. However, the role of those criteria is less clear for patients who are being treated with agents targeting the androgen signaling axis, given variable changes in PSMA expression. Ultimately, there may be key roles for machine learning and artificial intelligence in identifying imaging biomarkers based on changes in PSMA PET uptake during therapy.

  • PSMA
  • prostate-specific membrane antigen
  • prostate cancer
  • radiopharmaceutical therapy
  • response assessment
  • PPP
  • RECIP

Prostate cancer (PC) is the second leading cause of cancer-related deaths among men in the United States (1). According to the American Cancer Society’s projections, 1,466,718 new cases of PC were anticipated worldwide for 2024 (1). The 5-y survival rate for patients with early, localized PC exceeds 99%; however, that rate plummets to 30% for patients with metastases (2). The treatment landscape of metastatic PC has evolved significantly over the past 2 decades. Multiple phase 3 trials have led to new drug approvals and rapid changes in the therapeutic armamentarium for PC (e.g., hormonal therapy, chemotherapy, radiopharmaceuticals, immunotherapy, and targeted therapies). Therapies initially developed in later stages of the disease (e.g., metastatic castration-resistant prostate cancer [mCRPC]) have started to move earlier in the PC trajectory, with new standards of care for metastatic castration-sensitive PC and non-mCRPC (3).

Overall, in oncology, determining response to therapy or defining disease progression is considered a pillar in the management of patients with cancer, especially when varying treatment options are available for the treating physician. In PC, conventional imaging using bone scans and CT has been the mainstay of radiographic follow-up in patients with PC (4).

The U.S. Food and Drug Administration (FDA) has approved the prostate-specific membrane antigen (PSMA)–targeted agents 68Ga-PSMA-11, 18F-DCFPyL, and 18F-rhPSMA-7.3 for PET imaging (PSMA PET) for multiple indications (5). Current guidelines specifically recommend PSMA PET with those radiotracers in patients with PC and suspected metastases who are candidates for initial definitive therapy and who have suspected recurrence based on elevated prostate specific antigen (PSA) level to determine eligibility for treatment with PSMA-targeted radiopharmaceutical therapy such as 177Lu-PSMA-617 (6–8). PSMA PET has demonstrated higher detection accuracy in PC when compared with conventional imaging (9); however, its superiority for treatment response evaluation purposes has not been established to the best of our knowledge. Response evaluation methods have been developed for PSMA PET that distinguish partial response (PR), complete response (CR), progressive disease (PD), or stable disease (SD) (10–12). Evidence is emerging regarding the prognostic value of response categorization of metastatic disease using PSMA PET in patients with PC being treated with either androgen receptor pathway inhibitors (ARPIs) (13,14), taxane (15), or 177Lu-PSMA radiopharmaceutical therapy (16). Recent imaging guidelines recommend a baseline PSMA PET to be performed before the initiation of systemic therapies, and follow-up PSMA PET should be performed when the results are expected to change clinical management (17,18). An end-of-treatment scan at least 3 mo after systemic treatment has concluded.

An important topic discussed further in this article is the postulated flare phenomenon caused by increased PSMA expression after initiation of androgen-deprivation therapy (ADT) or ARPI in the castration-resistant population, whereas in hormone-sensitive disease, there may be decreased uptake which could underestimate disease burden (19). Further studies are needed to fully clarify the role of PSMA PET in response assessment during hormonal therapy in PC.

This article aims to discuss the present literature on the role of PSMA PET for treatment response evaluation for PC and provide guidance on its further development.

ROLE OF PSMA-TARGETED IMAGING IN PC

PSMA is a molecular target of great interest and importance in the imaging of PC. PSMA is a surface membrane glycoprotein that is highly expressed on PC cells (20). Although that antigen was discovered as early as 1993, it was as recent as December of 2020 that the first FDA-approved PSMA-targeted PET imaging radiotracer (68Ga-PSMA-11) was cleared for clinical use in the United States. That development was contemporaneous to the ProPSMA trial and other studies showing that PSMA PET was superior to conventional imaging (CT and bone scan) for evaluating men with PC (9,21–23). Now multiple different PSMA agents are widely available, and there is a plethora of data correlating PSMA expression in PC with aggressive behavior, increased risk of progression, and development of castration resistance (24). Although other tissues do express PSMA (kidney, nervous system, small bowel, and salivary and lacrimal glands), they either do so to a lesser extent or represent tissues that are uncommonly involved with PC metastases. The result is a high-contrast form of imaging that is sensitive and specific and thus ideally suited for the evaluation of PC (20).

PSMA PET has been studied in multiple different clinical settings, with resulting data that support its role in the evaluation of PC at multiple time points in the disease trajectory, including, but not limited to, initial staging, recurrence, and mCRPC (22,25–27). Indeed, PSMA PET has demonstrated an increased detection rate compared with conventional imaging and, as a result, can lead to a change in management in a significant number of cases (22). Previously within the United States, efforts had been directed toward obtaining regulatory approval for these indications and with much success. However, as posttherapy PSMA PET examinations become more common, especially after the advent and FDA approval of PSMA-targeted radiotherapy, the focus in the scientific and clinical community has shifted to its potential roles in outcome prediction and response evaluation. One such example of the potential prognostic value of PSMA PET is demonstrated in a 2019 paper that showed that even a false-negative PSMA PET correlates with better disease-free survival than does a true-positive scan (28).

PSMA is expressed in normal prostate cells but is overexpressed in PC cells. In addition, PSMA shows coordinated expression with vascular endothelial growth factor receptor, a key driver of tumor-induced angiogenesis, suggesting PSMA as a potential indirect marker of neoangiogenesis in nonprostate solid tumors (29,30). This suggests that PSMA can be a dynamic biomarker with a strong prognostic value, as confirmed by some recent studies pointing to a correlation between a positive PSMA PET and the clinical outcome of the patient (31). The proPSMA study suggests a prognostic value of PSMA PET identifying metastatic disease early, thus influencing treatment decisions and potentially reducing the risk of recurrence (32). By improving accuracy in PC staging, PSMA PET could lead to better-tailored treatments and improved outcomes for patients at high risk of recurrence. PSMA PET has played a key role in stage migration of patients with metastatic disease in the hormone-sensitive setting, namely identifying high-volume patients eligible for ARPI (33,34) and more accurately delineating low-volume patients who may benefit from metastasis-directed therapy (35).

In the setting of PSMA-targeted therapeutic radiopharmaceuticals, PSMA PET is required to select patients for radiopharmaceutical therapy and, hence, to identify candidates who are most likely to respond to the therapy. In that context, an analysis of PSMA PET imaging parameters as predictive and prognostic biomarkers in the patient population of the TheraP study demonstrated that the SUVmean from PSMA PET was predictive of a higher likelihood of response to 177Lu-PSMA-617 therapy (36). Concordant results were observed from an exploratory secondary analysis of the VISION trial: it was found that baseline whole-body (WB) tumor SUVmean from PSMA PET was the strongest predictor of the efficacy of 177Lu-PSMA-617. Participants with higher tumor SUVmean levels showed greater improvements in radiographic progression-free survival (PFS) and overall survival (OS), but the benefit of the treatment was observed across all SUVmean quartiles (37).

Fewer studies have explored the use of PSMA PET to monitor treatment response to PSMA-targeted therapeutic radiopharmaceuticals. The LuPIN trial identified that an increase in the quantitative parameter total tumor volume (TTV) on posttreatment PSMA PET was a strong prognostic biomarker for early disease progression and shorter OS, regardless of PSA levels (38). Emerging evidence supports the use of PSMA PET as a prognostic biomarker, refining patient management and therapeutic response models. By accurately mapping metastatic burden and tumor heterogeneity, PSMA PET can provide insight into disease aggressiveness, aiding in the prediction of outcomes for patients with PC, both in the hormone-sensitive and castration-resistant settings.

THERAPEUTIC RESPONSE EVALUATION IN PC

Objective response assessment criteria are needed to demonstrate the efficacy of a treatment. Although these criteria do not always replace the physical exam and other less objective criteria completely, they often largely complement those approaches and sometimes replace them entirely. In PC, serial serum PSA levels have been traditionally used in response assessment. A rise in PSA in a patient with a history of treated PC is considered as an indicator of disease recurrence or progression. It should be noted that a single-time-point PSA measurement is sometimes not sufficient to draw a definitive conclusion, and repeat PSA measurements may be needed for more definitive conclusions (39). Another, more recently developed blood-based biomarker is circulating tumor DNA; whereas data are still being developed, they indicate that circulating tumor DNA at baseline can probably serve as a prognostic biomarker, and changes in the circulating tumor DNA after treatment can probably be used as an early predictor of both time to progression and survival (40).

The main advantage of imaging-based over biochemical tumor markers is their ability to localize tumor sites along with the ability to assess expression of the biomarker at each site (i.e., intrapatient heterogeneity) and to measure biomarker-expressing tumor volume. Knowing the location of the tumor can serve as a prognostic factor and help determine the optimal treatment approach (systemic vs. localized) and treatment goal and intensity (dosimetry) as well as the need of concomitant medication to combat side effects. In the case of PC, the presence of brain or mesenteric metastases might trigger the prophylactic administration of steroids. And if a patient has extensive bone involvement, closer attention to blood counts at the time of treatment helps determine whether the patient may not be a candidate for PSMA-targeted therapeutic radiopharmaceuticals altogether or if shorter-term check of blood counts are required.

Current versions of proposed standardized reporting frameworks for PSMA PET have begun to incorporate aspects of response assessment (41,42), although as efforts are made to harmonize the proposed systems, the results may prove cumbersome (43). Such efforts are in the early stages of development and will ultimately need to be added to various PET-viewing software packages.

The historical guideline-recommended imaging modalities for the detection of metastases in advanced PC and for the assessment of their response to treatment are 99mTc-based bone scintigraphy (BS) for bone lesions and contrast-enhanced abdomen and pelvis CT or pelvis MRI for nodal, visceral, and bone lesions (4). However, all of those modalities have significant limitations for lesion detection and especially for response assessment.

BS has a sensitivity of 79%–88% and a specificity of 75%–82% for detecting bone lesions (44,45). The proportion of equivocal and false-negative findings on BS is problematic, with the latter probably being responsible for futile radical treatments (46), and a patient with widespread metastatic disease may be falsely identified as having oligometastatic disease (35). Evaluation of therapy response using BS also suffers limitations. A first important pitfall is the possible observation of a flare phenomenon in BS performed 8–12 wk after treatment initiation, consisting of the appearance of new osteoblastic foci on a first follow-up scan, which in reality represents a sign of favorable response to treatment. Such pseudoprogression cannot be distinguished from true disease progression without longer-term follow-up (47). The Prostate Cancer Working Group 3 (PCWG3) criteria suggest that all patients who have at least 2 new lesions at a first follow-up BS require a confirmatory BS after more than 6 wk of treatment continuation (4). BS progression is only confirmed if 2 or more new lesions are seen on the confirmatory BS (2 + 2 rule). Therefore, a change in treatment on the basis of progression can only occur after at least 14 wk of treatment (depending on the reassessment schedule).

Further, only the appearance of new lesions is considered, and an increase in the extent of preexisting lesions cannot be used as an objective criterion for BS progression, further limiting the value of BS in identifying disease progression. Moreover, diffuse metastatic bone disease on BS (i.e., a superscan), often observed in advanced PC, cannot be assessed for response because new disease cannot be distinguished on an already diffusely increased tracer uptake background. Lastly, BS assessment of response distinguishes only 2 categories (PD or non-PD) and cannot positively identify response because reduction and resolution of bone uptake takes a long time to occur, limiting the timeliness of readouts (48).

In contrast to its diagnostic performance in the assessment of visceral lesions, CT has limitations in the detection of node and bone lesions and in the assessment of their response to treatment. For nodes, CT only considers abnormal lymph nodes on the basis of size criteria and has both low sensitivity (42%) and limited low specificity (82%) (49). For the assessment of response to treatment, RECIST 1.1 and PCWG3 criteria only consider nodes larger than 15 mm in the short axis as pathologic and measurable (4,50). Nodules with a short axis larger than 10 mm but smaller than 15 mm are considered pathologic but nonmeasurable or a nontarget. For nodal and visceral lesions, RECIST 1.1 categorizes findings as CR, PR, SD, or PD. For bone, CT cannot detect lesions before significant osteolysis or sclerosis has occurred. RECIST considers bone lesions to be nonmeasurable, and their response to treatment cannot be assessed unless they have measurable extraosseous spread (50).

Although these multiple limitations argue against the use of BS and CT/MRI for response assessment, those methods are still commonly used because of their wide availability and use in many pivotal clinical trials (51). However, there is currently no evidence that disease staging and response stratification based on PSMA PET adds value for therapeutic decisions and, most importantly, for improving patient outcomes.

PET FOR RESPONSE EVALUATION

PERCIST is a response framework introduced in 2009 (52) using FDG PET/CT in different cancers. Briefly, the SUVpeak of the most avid tumor corrected for lean body mass (SUL), within a 1-cm3 volume, is measured in each scan (i.e., the SULpeak of the scan). It must also be more significantly avid than a background organ (usually liver), or the lesion is considered not measurable. Specifically, SULpeak greater than 1.5 times the mean plus 2 SD of the SUL of a 3-cm–diameter volume of interest in the liver, or twice the mean plus 2 SD in the thoracic aorta, can be considered measurable (53). A partial metabolic response is a reduction of at least 30% from baseline and at least 0.8 SUL. For stable metabolic disease, SULpeak changes must be within 30% of the baseline value. Progressive metabolic disease is defined as an increase of at least 30% and 0.8 SUL or the occurrence of new lesions, or there must be unequivocal progression in nontarget lesions (53). CR requires resolution of uptake within target lesions (to less than liver and similar to blood pool) and the absence of new lesions.

PERCIST was designed for FDG, so its applicability to PSMA is unclear (54). There are concerns about loss of PSMA expression by aggressive tumors creating false-positive responses by lowering PSMA uptake (55), which has been observed in a challenge trial with repeat 177Lu-PSMA-I&T (56) with rising PSA despite declining PSMA uptake. A study of test–retest variability for 68Ga-PSMA-11 showed an excessive 50% variability (greater than the values that have been reported for 18F-DCFPyL (57,58)), but this was of SUVmax rather than the SULpeak used in PERCIST, so its applicability is unclear (59). A 2021 European Association of Urology and European Association of Nuclear Medicine consensus statement suggested finding new detectability thresholds given the differences with FDG (18) and declined to endorse PERCIST for PSMA for the same reasons, preferring tumor volume where possible.

Nonetheless, PERCIST has been used without further modification with 68Ga-PSMA-11 to assess 177Lu-labeled theranostic PSMA agents (60,61), 225Ac-labeled theranostic agents (62), or external-beam radiation (63,64). There are also numerous modified PERCISTs, often using the PERCIST cutoffs of 30% with variables other than SULpeak, sometimes not explicitly differentiating themselves from orthodox PERCIST. One study simply used body weight because of reproducibility concerns (65). A study of patients being treated with docetaxel used summed SUVmean of the 2 hottest lesions in each organ system instead (66) and was later cited in a renal cell PSMA study (67), a trial of 177Lu-PSMA-617 (68), and a recent study of rechallenge with 177Lu-PSMA-I&T (56). Equivalents to metabolic tumor volume and total lesion glycolysis with a variety of names have been used to assess 225Ac-PSMA-617 (69) and 177Lu-PSMA-I&T (61,70).

Despite any limitations, PERCIST may be better than RECIST for PSMA PET. A study of 88 patients with a mixture of treatments showed PERCIST (and European Organisation for Research and Treatment of Cancer criteria) detected more progression than RECIST 1.1 (71). PERCIST in 23 patients treated with 177Lu-labeled PSMA therapy (72) showed better agreement with biochemical response (72) than did RECIST. PERCIST (and other PET metrics) correlated with biochemical response, though not survival, in a study of 42 patients on a mixture of systemic therapies (73). A study of 72 patients receiving chemohormonal therapy showed better performance for PERCIST and the European Associating of Urologists and European Association of Nuclear Medicine criteria (which use total volume) than RECIST or findings on multiparametric MRI (74).

However, WB volumetric biomarkers may be more useful still. Thirty-nine patients with mCRPC who were treated with 177Lu-PSMA-617 showed poor agreement between PERCIST (or variant measures using SUVmax or SUVpeak) and biochemical response or survival outcome, though the correlation was ultimately positive (70). Nineteen patients with mCRPC who were treated with 177Lu-PSMA-I&T showed metrics of PSMA-expressing tumor volume and overall expression (analogous to metabolic tumor volume and total lesion glycolysis) correlated better than PERCIST with patient survival (61) and were relatively stable on 2 different software platforms (75). Another study compared a modified PERCIST using metabolic tumor volume (therein described as TTV) to PSA response and OS in 55 patients with mCRPC who were treated with 177Lu-PSMA, showing better correlation than RECIST or SUVmean with OS and PSA changes (70). Another study of 51 patients treated with 177Lu-PSMA-617 showed total lesion PSMA divided by healthy liver tissue (again, a TLG analog) had better correlation with PFS and performance status (68) than the modified PERCIST, which used summed SUVpeak. Other metrics such as the percentage of bone volume affected by tumors (possibly incorporating level of uptake) have been suggested and may be more reproducible and faster (63).

PSMA PET–SPECIFIC CRITERIA FOR RESPONSE EVALUATION IN PC

The PSMA PET progression (PPP) criteria were introduced as an initial effort to standardize the assessment of imaging-based response in metastatic PC using PSMA PET (12). The PPP criteria integrate biochemical and genetics data into the response assessment, which limits their use for calculation of imaging-based–only PFS. The PPP criteria derive progression as the appearance of at least 2 new lesions, which mirrors the PCWG3 criteria for progression on BS (Table 1) (4). It was shown retrospectively that progression on PSMA PET according to the modified PPP criteria after completion of 177Lu-PSMA-617 therapy was significantly associated with shorter OS (median OS, 7.0 vs. 29.0 mo) (76).

View this table:
  • View inline
  • View popup
TABLE 1.

Definition of PPP Criteria

RECIP 1.0 was proposed as the first evidence-based standardized framework for response evaluation in PC using PSMA PET. Compared with PPP, which uses individual lesion measurements, RECIP 1.0 accounts for changes in TTV, capturing the entire extent of disease. That has clinical relevance, especially during treatment of advanced PC in which heterogeneous response by individual metastatic lesions is quite common, although it must be kept in mind that this heterogeneity might be of prognostic significance. RECIP 1.0 categorizes scans into CR, PR, SD, and PD on the basis of changes in PSMA-positive TTV and the appearance of new lesion(s) (Table 2). RECIP PD requires both the occurrence of new lesions and a minimum significant increase in tumor volume (>20% increase), whereas RECIP PR requires both the absence of new lesions and a substantial decrease (>30% reduction) of TTV. The appearance of new lesions and a concomitant decrease in TTV is classified as stable disease. In a retrospective, multicenter study of 124 patients, PR versus SD versus PD by RECIP 1.0 was associated with OS (median OS, 21.7, 13.1, and 8.3 mo, respectively) (11). RECIP 1.0 can be determined using 2 approaches, that is, visually by physicians (visual RECIP) or quantitatively using tumor segmentation software (quantitative RECIP) (77). Both visual and quantitative RECIP 1.0 achieved an excellent interreader agreement (83% and 92%, respectively). Agreement between visual and quantitative RECIP 1.0 was observed in 95% of cases, indicating that the 2 methods can be used interchangeably in both clinical trials and clinical routine. Although it was initially developed in patients with late-stage mCRPC, RECIP 1.0 was successfully validated in patients with early-stage PC with biochemical recurrence after definitive therapy (78), during the use of ARPI in early-stage mCRPC (13), and during 223Ra therapy (79). PROMISE V2 has proposed PPP criteria for response evaluation in early PC and RECIP 1.0 for late-stage mCRPC based on the evidence available in 2023 (41). A case example of treatment response in PC using PSMA PET/CT evaluated by PPP and RECIP 1.0 is given in Figure 1.

View this table:
  • View inline
  • View popup
TABLE 2.

Definition of RECIP 1.0

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Case example of treatment response in metastatic PC using PSMA PET/CT. 72-y-old male with mCRPC previously treated with ARPIs and taxanes. Baseline 18F-DCFPyL PET/CT (A) from June 2022 showed diffuse skeleton involvement and pelvic lymph nodes. Follow-up 18F-DCFPyL PET/CT (B) from May 2023 after 6 cycles of 177Lu-PSMA-617 showed mixed response with majority of lesions decreasing in size and intensity, however, with occurrence of ≥2 new lesions (e.g., right femur and spine). Response classified as PD according to PPP criteria (≥2 new lesions) and SD according to RECIP 1.0 (≥1 new lesion without significant change in total tumor volume). Patient died 14.5 mo after 177Lu-PSMA-617 initiation.

PSMA PET FOR RESPONSE EVALUATION DURING PSMA THERANOSTICS

On the basis of the positive results from the VISION trial (80), 177Lu-PSMA-617 was approved by the U.S. FDA in patients with mCRPC who progressed on ARPI and taxane-based chemotherapy. The prerequisite for being eligible for 177Lu-PSMA-617 is the presence of PSMA-positive lesions on the screening PSMA PET scan. An important question in the community is how to evaluate treatment response during PSMA-targeted therapeutic radiopharmaceuticals, especially considering that baseline PSMA PET imaging is available as a standard-of-care procedure.

According to the current Society of Nuclear Medicine and Molecular Imaging and European Association of Nuclear Medicine guidelines, interim PSMA PET scans should be conducted every 12 wk during PSMA-targeted therapeutic radiopharmaceutical administration to ensure optimal follow-up and treatment response assessment (81). However, the current FDA label does not include response assessment or monitoring, so such imaging is likely performed much less often in the United States relative to other regions such as Europe and Australia. Several retrospective studies have demonstrated the prognostic value of interim PSMA PET performed after 2 cycles of therapy (11,76,77). One report found the end-of-treatment PSMA PET after the last therapy cycle is associated with OS (82). Patients with RECIP PD had significantly shorter survival compared with those with RECIP non-PD (median OS, 10.9 mo vs. not reached; P = 0.002).

Posttherapy SPECT imaging using 1 of the γ-photopeaks from the 177Lu radionuclide has the potential to evolve as an imaging response biomarker for PSMA-targeted therapeutic radiopharmaceuticals, in addition to its value in dosimetry (83). SPECT at 24 h after 177Lu-PSMA administration represents a potentially cost-effective alternative to interim PSMA PET. 177Lu is a β-therapy that also emits 11% γ-rays, which can be used to derive WB tomographic images similar to PSMA PET. That allows image quantitation and evaluation after every treatment dose. Previous studies investigated the role of posttherapeutic SPECT for response evaluation during 177Lu-PSMA treatment. John et al. (84) demonstrated that TTV derived from SPECT after 2 cycles of 177Lu-PSMA-I&T in 96 patients with mCRPC is prognostic for PFS but not for OS. Song et al. (85) found that changes in tumor volume quantified on digital WB SPECT imaging performed 1–5 h after a second dose of 177Lu-PSMA-617 is prognostic for OS. In a multicenter retrospective study, changes in TTV on SPECT after 2 cycles were associated with OS (hazard ratio, 0.28; P < 0.001) (86).

Nevertheless, attention should be paid particularly to new or progressive lesions at morphologic examination without PSMA uptake. PC metastases that have lost PSMA expression can be identified using a second imaging modality such as diagnostic CT or MRI or FDG PET (81). Emerging data support the broader use of WB MRI, including diffusion-weighted imaging sequences, for disease burden assessment and response monitoring, although its relative role in the standard of care for patients with advanced PC is yet to be fully defined (87–90). WB MRI can detect bone metastases with higher sensitivity than BS and CT and with a performance close to that of PSMA PET (91,92). In contrast to BS, WB MRI allows an accurate assessment and categorization of treatment response of bone, node, and visceral metastases in a RECIST-like fashion (PD, SD, but also PR and CR) using the Metastasis Reporting Data System for Prostate Cancer criteria (93–95). As it does not depend on the (persistent) expression of a tracer, WB MRI offers a good and consistent one-size-fits-all solution for patients, allowing assessment of bone, node, and visceral lesions, regardless of tracer expression (96). The technique is gaining acceptance because of reasonable acquisition times (<30 min) using the Dixon technique and deep learning image reconstruction (90,94). However, WB MRI can have limited applicability in patients with claustrophobia and cardiac devices, and some patients may still not tolerate the acquisition time.

PSMA PET FOR RESPONSE EVALUATION WITH NON-PSMA–TARGETED THERAPIES

Assessing chemotherapy response is essential for determining the management of metastatic hormone-sensitive PC and mCRPC. At present, therapy monitoring relies on the use of imaging combined with serum PSA levels (4). However, PSA can be unreliable as it provides an indirect estimation of the volume of disease, and new metastatic sites might appear even when PSA is decreasing. Several studies have reported discordance between serum PSA trends and PET imaging results (especially when PSA decreased while PET showed appearance of new lesions) (97). Interestingly, WB MRI–based studies reported the same observation (98). In addition, some specific sites of metastases in patients with mCRPC are associated with different OS: visceral metastases (especially lung and liver) are associated with increased mortality compared with bone metastases (99). But radiographic response assessments also have limitations. For example, RECIST 1.1 for sclerotic bone appearance on CT images, since it can have limited PSMA uptake, may represent treated sites of disease and often lack a soft-tissue component (50).

Although PSMA PET is primarily used for PSMA-targeted therapies, emerging data suggest its potential role in monitoring responses to cytotoxic agents including taxane-based chemotherapy (docetaxel and cabazitaxel). There are currently no specific validated response criteria for PSMA PET in this context. However, the cutoff of 30% change in TTV in PSMA-expressing disease, as established by the consensus statements on PSMA PET response assessment criteria (18), may have value. Those studies showed that a change in PSMA expression correlated closely with PSA trends, and that those criteria could be used as an independent predictive biomarker for OS (15). Other studies have used the same cutoff applied to the summed SUVmean (66).

However, the utility of end-of-treatment versus interim PSMA PET is still under investigation, and clear guidelines for timing remain to be established. Although evidence supporting the use of PSMA PET in evaluating chemotherapy response is growing, further research is needed to validate standardized protocols and optimize timing to maximize clinical utility.

The use of agents that target the androgen signaling axis creates specific challenges with PSMA PET for response assessment. In the preclinical setting, it has been observed that an interruption of the androgen signaling axis with agents such as androgen-deprivation therapy or second-generation antiandrogen drugs can drive increased expression of PSMA even while the tumor may be responding (100). Although concordant results were noted in an initial case of a patient starting androgen-deprivation therapy reported by Hope et al. (101), subsequent studies have demonstrated that the overall in vivo process is more complicated (14,102,103). Despite the complexity of the data, there are likely imaging biomarkers from serial PSMA PET scans that are associated with PFS and OS and of potential utility in patient prognostication (14). Further study is certainly needed to establish the role of PSMA PET for response assessment in the context of treatment with androgen-axis–targeted agents.

PSMA PET is being explored to evaluate responses to other systemic therapies, including poly(ADP-ribose) polymerase inhibitors (PARPi) and 223Ra. For PARPi, data on PSMA PET as a response biomarker are sparse, but early studies suggest it could play a role in evaluating the therapeutic effects of PARPi in metastatic PC, particularly when combined with other agents. The ongoing exploration of these systemic therapies with PSMA PET highlights the potential for more personalized, data-driven treatment adjustments based on real-time imaging results.

223Ra-dichloride is an approved bone-targeting radiotracer for patients with mCRPC with bone metastases after 2 systemic therapy lines (104). However, since its approval, no imaging method has been firmly established to reliably monitor treatment response. Although there are emerging methods being tested, such as PSMA PET, none are yet considered the gold standard. A study has suggested a role in using PSMA PET response criteria (both RECIP 1.0 and PPP) in predicting OS after 3 cycles of 223Ra, but such an approach needs validation in larger studies (79).

POTENTIAL ROLE OF PSMA PET IN TREATMENT RESPONSE FOR LOCAL THERAPIES

In cases of presumed early-stage PC localized to the gland (initially staged with PSMA PET) that were treated with primary radiation therapy, posttherapy PSMA PET demonstrated residual prostatic disease in 39% cases or new PD, including pelvic nodes and extrapelvic disease, in approximately 7% and 33% of cases, respectively (105). PSA before the PSMA PET scan was the only factor that correlated with PSMA-positive findings, with many patients demonstrating positive findings even in cases with extremely low PSA after therapy (>0.2 ng/mL, or velocity of >1 ng/mL/y) (106). Even more notable is the fact that patients with higher posttherapy PSA levels (>10 ng/mL) were more likely to demonstrate new distant disease at the time of repeat PSMA PET (105).

Although early imaging is crucial for early identification of metastatic disease, in patients with definitive radiation therapy, including brachytherapy and external-beam therapy, to the prostate gland, posttherapy imaging by PSMA PET should be optimally timed to minimize false positives due to local inflammation. One paper attempted to characterize pitfalls of postradiation therapy PSMA PET findings in patients with biochemical recurrence as defined by a PSA rise of more than 2 ng/mL above nadir after definitive radiation. That study demonstrated that 77% of those patients had true recurrence, and that the false-positive rate was less than 10% (confirmed by pathology). The time since the last therapy in patients with false positives ranged from 1 to 20 y, but the majority of false-positive results were from indolent tumor remnants with treatment effects as seen in patients who were approximately 1 y out from therapy (107). This result may be due to the fact that radiation effects are long-lasting and may not affect cell death until the next cellular division, thus leaving detectable PSMA-expressing cells that are not viable. Indeed, complete histologic resolution after those therapies may take several years (108).

Currently, there are no substantial data to suggest what constitutes an ideal window for imaging with PSMA PET in a patient after radiation therapy, although the above-noted study suggests that PSMA PET has improved specificity when used at least 1 y out from therapy. It also warns against aggressive salvage therapy for a PSMA-positive finding in the gland less than 1 y from therapy, as those findings may represent a false positive. Such patients may benefit from repeat biopsy to assess the need for additional intervention. In observation studies, that patient group with residual indolent or posttreatment adenocarcinoma had similar outcomes as patients with negative biopsies (109,110). However, it could be argued that using PSMA PET within a 1-y posttherapy period still holds value in identifying early sites of extraprostatic recurrence that could be targeted with curable intent. There are currently no specific response assessment criteria that address lesions within the irradiated gland and how they might differ from extraprostatic lesions. One frequent question in daily practice is whether residual focal prostatic uptake with lower PSMA uptake than in the liver should be considered a sign of residual disease. Estimating residual intraprostatic tumor volume is not as easy as measuring an avid node. As such, response assessment of isolated intraprostatic recurrence is a topic that ideally could be assessed in future trials.

If indeed there is residual path-proven viable disease in the gland correlating to PSMA PET–positive findings, then there is literature supporting the feasibility and safety of PSMA PET avidity being used in planning for reirradiation in patients with locally recurrent PC. In this patient population, repeat gland radiation therapy resulted in complete PET response in 92% of patients (111). However, questions remain regarding the optimal manner by which to volumetrically delineate tumor within the prostate gland (112).

These findings together adjudicate the role of PSMA PET for use in the evaluation of recurrent disease in patients with low PSA and previously localized disease. By imaging with exquisitely sensitive PSMA PET tracers at lower PSA levels, we can identify early recurrent disease both inside and outside of the gland that would have gone undiagnosed by conventional methods and identify those patients who can potentially be cured by salvage therapies.

REGULATORY AND REIMBURSEMENT LANDSCAPE

The processes for regulatory approval and reimbursement for PET radiotracers are very different in various countries. For example, in the United States, PET radiotracers must go through FDA approval to be clinically used. That approval process is initiated by a commercial or noncommercial (in-house radiopharmacy) manufacturer, based on data from a clinical trials. Hence, indications are clearly stated in the approval notice and package insert. Medicare may decide to pay for PET radiotracers or not, but if they pay, it is usually for the indications approved by the FDA. Private and public insurance carriers usually follow Medicare guidelines. In the past few years, Medicare has been paying only for 3 y for new imaging radiotracers, so-called pass-through status, and after that, the cost of the radiotracer is bundled into the reimbursement for image acquisition; however, that situation was at least temporarily rectified by recent new guidance from the Center for Medicare and Medicaid Services (113).

In Europe, the practice of radiopharmaceutical administration is regulated at a national level, and there are 3 main routes to get a radiopharmaceutical into the clinic for human use: marketing authorization, clinical trial, and the magistral approach (114). A marketing authorization is very expensive and takes years to obtain. A clinical trial is less expensive but takes at least a year. In contrast, the magistral approach is the least expensive and can take only a matter of months. A marketing authorization may be used for both established and new radiotracers. A clinical trial is mainly for novel tracers (first-in-human studies) and for new or additional uses of an already established tracer. The magistral approach is for known radiopharmaceuticals that have been previously tested in humans with available preclinical data. The payment for PET imaging agents and therapeutic radiotracers varies significantly among countries in Europe, and a complete discussion is beyond the scope of this article.

CONCLUSION

The rapid adoption of PSMA-targeted diagnostic radiotracers has made it imperative that response assessment for men with PC be adapted to that new imaging modality. Although there are numerous approaches to objective imaging response, including RECIST, PCWG3, PERCIST, European Organisation for Research and Treatment of Cancer, PPP, and RECIP, there is not yet a unifying response assessment based on PSMA PET. In addition to an objective set of response criteria, it is also likely that careful assessment of PSMA PET data will uncover imaging biomarkers that will go beyond objective response and will instead provide prognostic information (14). Ultimately, it will likely require advanced imaging methodologies including machine learning and artificial intelligence to uncover those biomarkers (115–117).

Footnotes

  • ↵* Contributed equally to this work.

  • Published online Jun. 5, 2025.

  • Learning Objectives: On successful completion of this activity, participants should be able to describe (1) the common roles of PSMA PET for imaging of prostate cancer; (2) the use of PET as a therapy response evaluation tool; and (3) the PPP and RECIP criteria for PSMA-based response assessment in prostate cancer.

  • Financial Disclosure: Dr. Gafita is a consultant and on the speaker’s bureau for Novartis; is a consultant for Blue Earth Diagnostics, NucsAI, and Lilly; and has investment interest in NucsAI. Dr. Schroeder received a research grant from Lantheus Medical Imaging. Dr. Ceci is a consultant, on the speaker’s bureau, and on the advisory board and steering committee for Novartis and Curium; is a consultant and on the speaker’s bureau for Bayer, GE HealthCare, and Telix; is a consultant and has received grants from Hermes; is a consultant for NucsAI; and is a consultant and has investment interest in Radiantis Research. Dr. Oldan was a one-time panel consultant for Telix Pharmaceuticals. Dr. Lecouvet is a consultant, has received a research grant, and is on the speakers’ bureau of GE HealthCare. Dr. Solnes has research grants from Novartis, Lantheus, Perspective Therapeutics, and Cellectar and is on the advisory board of Novartis. Dr. Rowe is a consultant, has received a research grant, and is on the speakers’ bureau for Lantheus Medical Imaging; is a consultant for Imaging Endpoints; and is an owner/investor in D&D Pharmatech. The authors of this article have indicated no other relevant relationships that could be perceived as a real or apparent conflict of interest.

  • CE Credit: SNMMI is accredited by the Accreditation Council for Continuing Medical Education (ACCME), the Accreditation Council for Pharmacy Education (ACPE), and the American Registry for Radiologic Technologists (ARRT) and Nuclear Medicine Technology Certification Board (NMTCB) to sponsor continuing education for physicians, pharmacists, and nuclear medicine technologists. You may make 3 attempts to pass the test and must answer 80% of the questions correctly to receive credit—number of credits awarded will be determined by the length of the article. Participants can access this activity through the SNMMI website (http://www.snmmilearningcenter.org) through July 2028. Additional details such as the number of credits issued per article, expiration dates, financial disclosure information, and the process to earn CE credit can also be found in the SNMMI Learning Center.

  • © 2025 by the Society of Nuclear Medicine and Molecular Imaging.

REFERENCES

  1. 1.↵
    1. Siegel RL,
    2. Giaquinto AN,
    3. Jemal A
    . Cancer statistics, 2024. CA Cancer J Clin. 2024;74:12–49.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Siegel RL,
    2. Miller KD,
    3. Jemal A
    . Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Yamada Y,
    2. Beltran H
    . The treatment landscape of metastatic prostate cancer. Cancer Lett. 2021;519:20–29.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Scher HI,
    2. Morris MJ,
    3. Stadler WM,
    4. et al
    .; Prostate Cancer Clinical Trials Working Group 3. Trial design and objectives for castration-resistant prostate cancer: updated recommendations from the Prostate Cancer Clinical Trials Working Group 3. J Clin Oncol. 2016;34:1402–1418.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Hennrich U,
    2. Eder M
    . [68Ga]Ga-PSMA-11: the first FDA-approved 68Ga-radiopharmaceutical for PET imaging of prostate cancer. Pharmaceuticals (Basel). 2021;14.
  6. 6.↵
    FDA approves second PSMA-targeted PET imaging drug for men with prostate cancer. U.S. Food and Drug Administration website. https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-second-psma-targeted-pet-imaging-drug-men-prostate-cancer. Published May 27, 2021. Accessed February 21, 2024.
  7. 7.
    FDA approves first PSMA-targeted PET imaging drug for men with prostate cancer. U.S. Food and Drug Administration website. https://www.fda.gov/news-events/press-announcements/fda-approves-first-psma-targeted-pet-imaging-drug-men-prostate-cancer. Published December 1, 2020. Accessed Feburary 21, 2024.
  8. 8.↵
    Drug Trials Snapshot: POSLUMA. U.S. Food and Drug Administration website. https://www.fda.gov/drugs/drug-approvals-and-databases/drug-trials-snapshot-posluma. Published January 16, 2024. Accessed February 21, 2024.
  9. 9.↵
    1. Hofman MS,
    2. Lawrentschuk N,
    3. Francis RJ,
    4. et al
    .; proPSMA Study Group Collaborators. 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.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Swiha M,
    2. Gafita A,
    3. Nguyen A,
    4. Emmett L
    . Treatment response imaging in prostate cancer. PET Clin. 2024;19:417–430.
    OpenUrlPubMed
  11. 11.↵
    1. Gafita A,
    2. Rauscher I,
    3. Weber M,
    4. et al
    . Novel framework for treatment response evaluation using PSMA PET/CT in patients with metastatic castration-resistant prostate cancer (RECIP 1.0): an international multicenter study. J Nucl Med. 2022;63:1651–1658.
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    1. Fanti S,
    2. Hadaschik B,
    3. Herrmann K
    . Proposal for systemic-therapy response-assessment criteria at the time of PSMA PET/CT imaging: the PSMA PET progression criteria. J Nucl Med. 2020;61:678–682.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    1. Shagera QA,
    2. Karfis I,
    3. Kristanto P,
    4. et al
    . PSMA PET/CT for response assessment and overall survival prediction in patients with metastatic castration-resistant prostate cancer treated with androgen receptor pathway inhibitors. J Nucl Med. 2023;64:1869–1875.
    OpenUrlAbstract/FREE Full Text
  14. 14.↵
    1. Zukotynski KA,
    2. Emmenegger U,
    3. Hotte S,
    4. et al
    . Prospective, single-arm trial evaluating changes in uptake patterns on prostate-specific membrane antigen–targeted 18F-DCFPyL PET/CT in patients with castration-resistant prostate cancer starting abiraterone or enzalutamide. J Nucl Med. 2021;62:1430–1437.
    OpenUrlPubMed
  15. 15.↵
    1. Shagera QA,
    2. Artigas C,
    3. Karfis I,
    4. et al
    . 68Ga-PSMA PET/CT for response assessment and outcome prediction in metastatic prostate cancer patients treated with taxane-based chemotherapy. J Nucl Med. 2022;63:1191–1198.
    OpenUrlAbstract/FREE Full Text
  16. 16.↵
    1. Kurth J,
    2. Kretzschmar J,
    3. Aladwan H,
    4. et al
    . Evaluation of [68Ga]Ga-PSMA PET/CT for therapy response assessment of [177Lu]Lu-PSMA radioligand therapy in metastasized castration refractory prostate cancer and correlation with survival. Nucl Med Commun. 2021;42:1217–1226.
    OpenUrlPubMed
  17. 17.↵
    1. Fendler WP,
    2. Eiber M,
    3. Beheshti M,
    4. et al
    . PSMA PET/CT: joint EANM procedure guideline/SNMMI procedure standard for prostate cancer imaging 2.0. Eur J Nucl Med Mol Imaging. 2023;50:1466–1486.
    OpenUrlPubMed
  18. 18.↵
    1. Fanti S,
    2. Goffin K,
    3. Hadaschik BA,
    4. et al
    . Consensus statements on PSMA PET/CT response assessment criteria in prostate cancer. Eur J Nucl Med Mol Imaging. 2021;48:469–476.
    OpenUrlPubMed
  19. 19.↵
    1. Emmett L,
    2. Yin C,
    3. Crumbaker M,
    4. et al
    . Rapid modulation of PSMA expression by androgen deprivation: serial 68Ga-PSMA-11 PET in men with hormone-sensitive and castrate-resistant prostate cancer commencing androgen blockade. J Nucl Med. 2019;60:950–954.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    1. Ristau BT,
    2. O’Keefe DS,
    3. Bacich DJ
    . The prostate-specific membrane antigen: lessons and current clinical implications from 20 years of research. Urol Oncol. 2014;32:272–279.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Carlucci G,
    2. Ippisch R,
    3. Slavik R,
    4. Mishoe A,
    5. Blecha J,
    6. Zhu S
    . 68Ga-PSMA-11 NDA approval: a novel and successful academic partnership. J Nucl Med. 2021;62:149–155.
    OpenUrlAbstract/FREE Full Text
  22. 22.↵
    1. Lawhn-Heath C,
    2. Salavati A,
    3. Behr SC,
    4. et al
    . Prostate-specific membrane antigen PET in prostate cancer. Radiology. 2021;299:248–260.
    OpenUrlPubMed
  23. 23.↵
    1. Rowe SP,
    2. Macura KJ,
    3. Mena E,
    4. et al
    . PSMA-based [18F]DCFPyL PET/CT is superior to conventional imaging for lesion detection in patients with metastatic prostate cancer. Mol Imaging Biol. 2016;18:411–419.
    OpenUrlCrossRefPubMed
  24. 24.↵
    1. Rowe SP,
    2. Gorin MA,
    3. Pomper MG
    . Imaging of prostate-specific membrane antigen with small-molecule PET radiotracers: from the bench to advanced clinical applications. Annu Rev Med. 2019;70:461–477.
    OpenUrlPubMed
  25. 25.↵
    1. Hope TA,
    2. Goodman JZ,
    3. Allen IE,
    4. Calais J,
    5. Fendler WP,
    6. Carroll PR
    . Metaanalysis of 68Ga-PSMA-11 PET accuracy for the detection of prostate cancer validated by histopathology. J Nucl Med. 2019;60:786–793.
    OpenUrlAbstract/FREE Full Text
  26. 26.
    1. Perera M,
    2. Papa N,
    3. Roberts M,
    4. 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.
    OpenUrlCrossRefPubMed
  27. 27.↵
    1. Fendler WP,
    2. Calais J,
    3. Eiber M,
    4. et al
    . Assessment of 68Ga-PSMA-11 PET accuracy in localizing recurrent prostate cancer: a prospective single-arm clinical trial. JAMA. Oncol. 2019;5:856–863.
    OpenUrlPubMed
  28. 28.↵
    1. van Leeuwen PJ,
    2. Donswijk M,
    3. Nandurkar R,
    4. et al
    . Gallium-68-prostate-specific membrane antigen (68Ga-PSMA) positron emission tomography (PET)/computed tomography (CT) predicts complete biochemical response from radical prostatectomy and lymph node dissection in intermediate- and high-risk prostate cancer. BJU Int. 2019;124:62–68.
    OpenUrlPubMed
  29. 29.↵
    1. O’Keefe DS,
    2. Bacich DJ,
    3. Huang SS,
    4. Heston WDW
    . A perspective on the evolving story of PSMA biology, PSMA-based imaging, and endoradiotherapeutic strategies. J Nucl Med. 2018;59:1007–1013.
    OpenUrlCrossRefPubMed
  30. 30.↵
    1. Tsui P,
    2. Rubenstein M,
    3. Guinan P
    . Correlation between PSMA and VEGF expression as markers for LNCaP tumor angiogenesis. J Biomed Biotechnol. 2005;2005:287–290.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Bianchi L,
    2. Ceci F,
    3. Costa F,
    4. et al
    . The impact of PSMA-PET on oncologic control in prostate cancer patients who experienced psa persistence or recurrence. Cancers (Basel). 2022;15:274.
    OpenUrlPubMed
  32. 32.↵
    1. Hofman MS,
    2. Kasivisvanathan V,
    3. Link E,
    4. et al
    . Baseline nodal status on 68Ga-PSMA-11 positron emission tomography/computed tomography in men with intermediate- to high-risk prostate cancer is prognostic for treatment failure: follow-up of the proPSMA trial. Eur Urol Oncol. November 28, 2024 [Epub ahead of print].
  33. 33.↵
    1. Francini E,
    2. Gray KP,
    3. Xie W,
    4. et al
    . Time of metastatic disease presentation and volume of disease are prognostic for metastatic hormone sensitive prostate cancer (mHSPC). Prostate. 2018;78:889–895.
    OpenUrlPubMed
  34. 34.↵
    1. Henríquez I,
    2. Malave B,
    3. Campos FL,
    4. et al
    . PSMA PET/CT SUVmax as a prognostic biomarker in patients with metachronous metastatic hormone-sensitive prostate cancer (mHSPC). Clin Transl Oncol. 2025;27:706–715.
    OpenUrlPubMed
  35. 35.↵
    1. Phillips R,
    2. Shi WY,
    3. Deek M,
    4. et al
    . Outcomes of observation vs stereotactic ablative radiation for oligometastatic prostate cancer: the ORIOLE phase 2 randomized clinical trial. JAMA. Oncol. 2020;6:650–659.
    OpenUrlPubMed
  36. 36.↵
    1. Buteau JP,
    2. Martin AJ,
    3. Emmett L,
    4. et al
    .; TheraP Trial Investigators and the Australian and New Zealand Urogenital and Prostate Cancer Trials Group. PSMA and FDG-PET as predictive and prognostic biomarkers in patients given [177Lu]Lu-PSMA-617 versus cabazitaxel for metastatic castration-resistant prostate cancer (TheraP): a biomarker analysis from a randomised, open-label, phase 2 trial. Lancet Oncol. 2022;23:1389–1397.
    OpenUrlCrossRefPubMed
  37. 37.↵
    1. Hofman MS,
    2. Emmett L,
    3. Sandhu S,
    4. et al
    . Overall survival with [177Lu]Lu-PSMA-617 versus cabazitaxel in metastatic castration-resistant prostate cancer (TheraP): secondary outcomes of a randomised, open-label, phase 2 trial. Lancet Oncol. 2023.
  38. 38.↵
    1. Pathmanandavel S,
    2. Crumbaker M,
    3. Nguyen A,
    4. et al
    . The prognostic value of posttreatment 68Ga-PSMA-11 PET/CT and 18F-FDG PET/CT in metastatic castration-resistant prostate cancer treated with 177Lu-PSMA-617 and NOX66 in a phase I/II trial (LuPIN). J Nucl Med. 2023;64:69–74.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    1. Kind F,
    2. Fassbender TF,
    3. Andrieux G,
    4. Boerries M,
    5. Meyer PT,
    6. Ruf J
    . Early PSA change after [177Lu]PSMA-617 radioligand therapy as a predicator of biochemical response and overall survival. Cancers (Basel). 2021;14:149.
    OpenUrlPubMed
  40. 40.↵
    1. Sartor O
    . Circulating tumor DNA biomarkers for response assessment in prostate cancer. Clin Cancer Res. 2023;29:2745–2747.
    OpenUrlPubMed
  41. 41.↵
    1. Seifert R,
    2. Emmett L,
    3. Rowe SP,
    4. et al
    . Second version of the prostate cancer molecular imaging standardized evaluation framework including response evaluation for clinical trials (PROMISE V2). Eur Urol. 2023;83:405–412.
    OpenUrlPubMed
  42. 42.↵
    1. Werner RA,
    2. Hartrampf PE,
    3. Fendler WP,
    4. et al
    . Prostate-specific membrane antigen reporting and data system version 2.0. Eur Urol. 2023;84:491–502.
    OpenUrlPubMed
  43. 43.↵
    1. Ceci F,
    2. Oprea-Lager DE,
    3. Emmett L,
    4. et al
    . E-PSMA: the EANM standardized reporting guidelines v1.0 for PSMA-PET. Eur J Nucl Med Mol Imaging. 2021;48:1626–1638.
    OpenUrlPubMed
  44. 44.↵
    1. Shen CT,
    2. Qiu ZL,
    3. Han TT,
    4. Luo QY
    . Performance of 18F-fluoride PET or PET/CT for the detection of bone metastases: a meta-analysis. Clin Nucl Med. 2015;40:103–110.
    OpenUrlCrossRefPubMed
  45. 45.↵
    1. Shen G,
    2. Deng H,
    3. Hu S,
    4. 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. Skeletal Radiol. 2014;43:1503–1513.
    OpenUrlCrossRefPubMed
  46. 46.↵
    1. Lecouvet FE,
    2. Oprea-Lager DE,
    3. Liu Y,
    4. et al
    . Use of modern imaging methods to facilitate trials of metastasis-directed therapy for oligometastatic disease in prostate cancer: a consensus recommendation from the EORTC imaging group. Lancet Oncol. 2018;19:e534–e545.
    OpenUrlCrossRefPubMed
  47. 47.↵
    1. Ryan CJ,
    2. Shah S,
    3. Efstathiou E,
    4. et al
    . Phase II study of abiraterone acetate in chemotherapy-naive metastatic castration-resistant prostate cancer displaying bone flare discordant with serologic response. Clin Cancer Res. 2011;17:4854–4861.
    OpenUrlAbstract/FREE Full Text
  48. 48.↵
    1. Padhani AR,
    2. Lecouvet FE,
    3. Tunariu N,
    4. et al
    . Rationale for modernising imaging in advanced prostate cancer. Eur Urol Focus. 2017;3:223–239.
    OpenUrlPubMed
  49. 49.↵
    1. Hövels AM,
    2. Heesakkers RA,
    3. Adang EM,
    4. et al
    . The diagnostic accuracy of CT and MRI in the staging of pelvic lymph nodes in patients with prostate cancer: a meta-analysis. Clin Radiol. 2008;63:387–395.
    OpenUrlCrossRefPubMed
  50. 50.↵
    1. Eisenhauer EA,
    2. Therasse P,
    3. Bogaerts J,
    4. et al
    . New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–247.
    OpenUrlCrossRefPubMed
  51. 51.↵
    1. Gillessen S,
    2. Omlin A,
    3. Attard G,
    4. et al
    . Management of patients with advanced prostate cancer: recommendations of the St Gallen Advanced Prostate Cancer Consensus Conference (APCCC) 2015. Ann Oncol. 2019;30:e3.
    OpenUrlPubMed
  52. 52.↵
    1. Wahl RL,
    2. Jacene H,
    3. Kasamon Y,
    4. Lodge MA
    . From RECIST to PERCIST: evolving considerations for PET response criteria in solid tumors. J Nucl Med. 2009;50(suppl 1):122S–150S.
    OpenUrlAbstract/FREE Full Text
  53. 53.↵
    1. O JH,
    2. Lodge MA,
    3. Wahl RL
    . Practical PERCIST: a simplified guide to PET response criteria in solid tumors 1.0. Radiology. 2016;280:576–584.
    OpenUrlPubMed
  54. 54.↵
    1. Hodler J,
    2. Kubik-Huch RA,
    3. von Schulthess GK
    1. Burger IA,
    2. Beets-Tan RGH
    . Therapy monitoring of oncologic disease in the abdomen (including PET/CT). In: Hodler J, Kubik-Huch RA, von Schulthess GK, eds. Diseases of the Abdomen and Pelvis 2018-2021: Diagnostic Imaging - IDKD Book. Springer; 2018:35–47.
  55. 55.↵
    1. Bakht MK,
    2. Derecichei I,
    3. Li Y,
    4. et al
    . Neuroendocrine differentiation of prostate cancer leads to PSMA suppression. Endocr Relat Cancer. 2018;26:131–146.
    OpenUrlCrossRefPubMed
  56. 56.↵
    1. Santo G,
    2. Santo GD,
    3. Sviridenko A,
    4. et al
    . Efficacy and safety of rechallenge with [177Lu]Lu-PSMA-I&T radioligand therapy in metastatic castration resistant prostate cancer. Eur J Nucl Med Mol Imaging. 2024;52:354–365.
    OpenUrlPubMed
  57. 57.↵
    1. Jansen BHE,
    2. Cysouw MCF,
    3. Vis AN,
    4. et al
    . Repeatability of quantitative 18F-DCFPyL PET/CT measurements in metastatic prostate cancer. J Nucl Med. 2020;61:1320–1325.
    OpenUrlPubMed
  58. 58.↵
    1. Werner RA,
    2. Habacha B,
    3. Lutje S,
    4. et al
    . High SUVs have more robust repeatability in patients with metastatic prostate cancer: results from a prospective test–retest cohort imaged with 18F-DCFPyL. Mol Imaging. 2022;2022:7056983.
    OpenUrlPubMed
  59. 59.↵
    1. Roef MJ,
    2. Rijnsdorp S,
    3. Brouwer C,
    4. Wyndaele DN,
    5. Arends AJ
    . Evaluation of quantitative Ga-68 PSMA PET/CT repeatability of recurrent prostate cancer lesions using both OSEM and Bayesian penalized likelihood reconstruction algorithms. Diagnostics (Basel). 2021;11:1100.
    OpenUrlPubMed
  60. 60.↵
    1. Yadav MP,
    2. Ballal S,
    3. Tripathi M,
    4. et al
    . 177Lu-DKFZ-PSMA-617 therapy in metastatic castration resistant prostate cancer: safety, efficacy, and quality of life assessment. Eur J Nucl Med Mol Imaging. 2017;44:81–91.
    OpenUrlPubMed
  61. 61.↵
    1. Acar E,
    2. Özdoğan Ö,
    3. Aksu A,
    4. Derebek E,
    5. Bekiş R,
    6. Çapa Kaya G
    . The use of molecular volumetric parameters for the evaluation of Lu-177 PSMA I&T therapy response and survival. Ann Nucl Med. 2019;33:681–688.
    OpenUrlPubMed
  62. 62.↵
    1. Yadav MP,
    2. Ballal S,
    3. Sahoo RK,
    4. Tripathi M,
    5. Seth A,
    6. Bal C
    . Efficacy and safety of 225Ac-PSMA-617 targeted alpha therapy in metastatic castration-resistant prostate cancer patients. Theranostics. 2020;10:9364–9377.
    OpenUrlCrossRefPubMed
  63. 63.↵
    1. Bieth M,
    2. Kronke M,
    3. Tauber R,
    4. et al
    . Exploring new multimodal quantitative imaging indices for the assessment of osseous tumor burden in prostate cancer using 68Ga-PSMA PET/CT. J Nucl Med. 2017;58:1632–1637.
    OpenUrlAbstract/FREE Full Text
  64. 64.↵
    1. Ozdemir Y,
    2. Torun N,
    3. Guler OC,
    4. et al
    . Local control and vertebral compression fractures following stereotactic body radiotherapy for spine metastases. J Bone Oncol. 2019;15:100218.
    OpenUrlPubMed
  65. 65.↵
    1. Heinzel A,
    2. Boghos D,
    3. Mottaghy FM,
    4. et al
    . 68Ga-PSMA PET/CT for monitoring response to 177Lu-PSMA-617 radioligand therapy in patients with metastatic castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging. 2019;46:1054–1062.
    OpenUrlPubMed
  66. 66.↵
    1. Seitz AK,
    2. Rauscher I,
    3. Haller B,
    4. et al
    . Preliminary results on response assessment using 68Ga-HBED-CC-PSMA PET/CT in patients with metastatic prostate cancer undergoing docetaxel chemotherapy. Eur J Nucl Med Mol Imaging. 2018;45:602–612.
    OpenUrlPubMed
  67. 67.↵
    1. Mittlmeier LM,
    2. Unterrainer M,
    3. Rodler S,
    4. et al
    . 18F-PSMA-1007 PET/CT for response assessment in patients with metastatic renal cell carcinoma undergoing tyrosine kinase or checkpoint inhibitor therapy: preliminary results. Eur J Nucl Med Mol Imaging. 2021;48:2031–2037.
    OpenUrlPubMed
  68. 68.↵
    1. Khreish F,
    2. Wiessner M,
    3. Rosar F,
    4. et al
    . Response assessment and prediction of progression-free survival by 68Ga-PSMA-11 PET/CT based on tumor-to-liver ratio (TLR) in patients with mCRPC undergoing 177Lu-PSMA-617 radioligand therapy. Biomolecules. 2021;11:1099.
    OpenUrlPubMed
  69. 69.↵
    1. Rosar F,
    2. Hau F,
    3. Bartholoma M,
    4. et al
    . Molecular imaging and biochemical response assessment after a single cycle of [225Ac]Ac-PSMA-617/[177Lu]Lu-PSMA-617 tandem therapy in mCRPC patients who have progressed on [177Lu]Lu-PSMA-617 monotherapy. Theranostics. 2021;11:4050–4060.
    OpenUrlCrossRefPubMed
  70. 70.↵
    1. Grubmuller B,
    2. Senn D,
    3. Kramer G,
    4. et al
    . Response assessment using 68Ga-PSMA ligand PET in patients undergoing 177Lu-PSMA radioligand therapy for metastatic castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging. 2019;46:1063–1072.
    OpenUrlPubMed
  71. 71.↵
    1. Gupta M,
    2. Choudhury PS,
    3. Rawal S,
    4. Goel HC,
    5. Rao SA
    . Evaluation of RECIST, PERCIST, EORTC, and MDA criteria for assessing treatment response with Ga68-PSMA PET-CT in metastatic prostate cancer patient with biochemical progression: a comparative study. Nucl Med Mol Imaging. 2018;52:420–429.
    OpenUrlPubMed
  72. 72.↵
    1. Gupta M,
    2. Choudhury PS,
    3. Rawal S,
    4. Goel HC,
    5. Rao SA
    . Evaluation of response in patients of metastatic castration resistant prostate cancer undergoing systemic radiotherapy with lutetium177-prostate-specific membrane antigen: a comparison between response evaluation criteria in solid tumors, positron-emission tomography response criteria in solid tumors, European organization for research and treatment of cancer, and MDA criteria assessed by gallium 68-prostate-specific membrane antigen positron-emission tomography-computed tomography. Urol Ann. 2019;11:155–162.
    OpenUrlPubMed
  73. 73.↵
    1. Grubmuller B,
    2. Rasul S,
    3. Baltzer P,
    4. et al
    . Response assessment using [68Ga]Ga-PSMA ligand PET in patients undergoing systemic therapy for metastatic castration-resistant prostate cancer. Prostate. 2020;80:74–82.
    OpenUrlPubMed
  74. 74.↵
    1. Ke ZB,
    2. Chen SM,
    3. Chen JY,
    4. et al
    . Head-to-head comparisons of [68Ga]Ga-PSMA-11 PET/CT, multiparametric MRI, and prostate-specific antigen for the evaluation of therapeutic responses to neoadjuvant chemohormonal therapy in high-risk non-metastatic prostate cancer patients: a prospective study. Eur J Nucl Med Mol Imaging. 2023;50:1240–1251.
    OpenUrlPubMed
  75. 75.↵
    1. Hartrampf PE,
    2. Heinrich M,
    3. Seitz AK,
    4. et al
    . Metabolic tumour volume from PSMA PET/CT scans of prostate cancer patients during chemotherapy: do different software solutions deliver comparable results? J Clin Med. 2020;9:1390.
    OpenUrlPubMed
  76. 76.↵
    1. Michalski K,
    2. Klein C,
    3. Brueggemann T,
    4. Meyer PT,
    5. Jilg CA,
    6. Ruf J
    . Assessing response to [177Lu]PSMA radioligand therapy using modified PSMA PET progression criteria. J Nucl Med. 2021;62:1741–1746.
    OpenUrlAbstract/FREE Full Text
  77. 77.↵
    1. Gafita A,
    2. Djaileb L,
    3. Rauscher I,
    4. et al
    . Response Evaluation Criteria in PSMA PET/CT (RECIP 1.0) in metastatic castration-resistant prostate cancer. Radiology. 2023;308:e222148.
    OpenUrlPubMed
  78. 78.↵
    1. Kendrick J,
    2. Francis RJ,
    3. Hassan GM,
    4. et al
    . Prognostic utility of RECIP 1.0 with manual and AI-based segmentations in biochemically recurrent prostate cancer from [68Ga]Ga-PSMA-11 PET images. Eur J Nucl Med Mol Imaging. 2023;50:4077–4086.
    OpenUrlPubMed
  79. 79.↵
    1. Shagera QA,
    2. Gil T,
    3. Barraco E,
    4. et al
    . Evaluating response to radium-223 using 68Ga-PSMA PET/CT imaging in patients with metastatic castration-resistant prostate cancer. Ann Nucl Med. 2025;39:208–216.
    OpenUrlPubMed
  80. 80.↵
    1. Sartor O,
    2. de Bono J,
    3. Chi KN,
    4. et al
    .; VISION Investigators. Lutetium-177-PSMA-617 for metastatic castration-resistant prostate cancer. N Engl J Med. 2021;385:1091–1103.
    OpenUrlCrossRefPubMed
  81. 81.↵
    1. Kratochwil C,
    2. Fendler WP,
    3. Eiber M,
    4. et al
    . EANM procedure guidelines for radionuclide therapy with 177Lu-labelled PSMA-ligands (177Lu-PSMA-RLT). Eur J Nucl Med Mol Imaging. 2019;46:2536–2544.
    OpenUrlCrossRefPubMed
  82. 82.↵
    1. Murthy V,
    2. Gafita A,
    3. Thin P,
    4. et al
    . Prognostic value of end-of-treatment PSMA PET/CT in patients treated with 177Lu-PSMA radioligand therapy: a retrospective, single-center analysis. J Nucl Med. 2023;64:1737–1743.
    OpenUrlAbstract/FREE Full Text
  83. 83.↵
    1. Jackson P,
    2. Hofman M,
    3. McIntosh L,
    4. Buteau JP,
    5. Ravi Kumar A
    . Radiation dosimetry in 177Lu-PSMA-617 therapy. Semin Nucl Med. 2022;52:243–254.
    OpenUrlPubMed
  84. 84.↵
    1. John N,
    2. Pathmanandavel S,
    3. Crumbaker M,
    4. et al
    . 177Lu-PSMA SPECT quantitation at 6 weeks (dose 2) predicts short progression-free survival for patients undergoing 177Lu-PSMA-I&T therapy. J Nucl Med. 2023;64:410–415.
    OpenUrlAbstract/FREE Full Text
  85. 85.↵
    1. Song H,
    2. Leonio MI,
    3. Ferri V,
    4. et al
    . Same-day post-therapy imaging with a new generation whole-body digital SPECT/CT in assessing treatment response to [177Lu]Lu-PSMA-617 in metastatic castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging. 2024;51:2784–2793.
    OpenUrlPubMed
  86. 86.↵
    1. Neubauer MC,
    2. Nicolas GP,
    3. Bauman A,
    4. et al
    .; all investigators on behalf of the SSNM Therapy Working Group. Early response monitoring during [177Lu]Lu-PSMA I&T therapy with quantitated SPECT/CT predicts overall survival of mCRPC patients: subgroup analysis of a Swiss-wide prospective registry study. Eur J Nucl Med Mol Imaging. 2024;51:1185–1193.
    OpenUrlPubMed
  87. 87.↵
    1. Wang Y,
    2. Galante JR,
    3. Haroon A,
    4. et al
    . The future of PSMA PET and WB MRI as next-generation imaging tools in prostate cancer. Nat Rev Urol. 2022;19:475–493.
    OpenUrlPubMed
  88. 88.
    1. Zhou J,
    2. Gou Z,
    3. Wu R,
    4. Yuan Y,
    5. Yu G,
    6. Zhao Y
    . Comparison of PSMA-PET/CT, choline-PET/CT, NaF-PET/CT, MRI, and bone scintigraphy in the diagnosis of bone metastases in patients with prostate cancer: a systematic review and meta-analysis. Skeletal Radiol. 2019;48:1915–1924.
    OpenUrlPubMed
  89. 89.
    1. Padhani AR,
    2. Tunariu N,
    3. Perez-Lopez R,
    4. Tombal B,
    5. Lecouvet FE
    . Evaluating prostate cancer bone metastases response with whole-body MRI: what we know and still need to know. Eur Radiol. 2024;34:7564–7566.
    OpenUrlPubMed
  90. 90.↵
    1. Van Nieuwenhove S,
    2. Van Damme J,
    3. Padhani AR,
    4. et al
    . Whole-body magnetic resonance imaging for prostate cancer assessment: current status and future directions. J Magn Reson Imaging. 2022;55:653–680.
    OpenUrlPubMed
  91. 91.↵
    1. Lecouvet FE,
    2. El Mouedden J,
    3. Collette L,
    4. et al
    . Can whole-body magnetic resonance imaging with diffusion-weighted imaging replace Tc 99m bone scanning and computed tomography for single-step detection of metastases in patients with high-risk prostate cancer? Eur Urol. 2012;62:68–75.
    OpenUrlCrossRefPubMed
  92. 92.↵
    1. Van Damme J,
    2. Tombal B,
    3. Collette L,
    4. et al
    . Comparison of 68Ga-prostate specific membrane antigen (PSMA) positron emission tomography computed tomography (PET-CT) and whole-body magnetic resonance imaging (WB-MRI) with diffusion sequences (DWI) in the staging of advanced prostate cancer. Cancers (Basel). 2021;13:5286.
    OpenUrlPubMed
  93. 93.↵
    1. Tombal B,
    2. Rezazadeh A,
    3. Therasse P,
    4. Van Cangh PJ,
    5. Vande Berg B,
    6. Lecouvet FE
    . Magnetic resonance imaging of the axial skeleton enables objective measurement of tumor response on prostate cancer bone metastases. Prostate. 2005;65:178–187.
    OpenUrlCrossRefPubMed
  94. 94.↵
    1. Lecouvet FE,
    2. Chabot C,
    3. Taihi L,
    4. Kirchgesner T,
    5. Triqueneaux P,
    6. Malghem J
    . Present and future of whole-body MRI in metastatic disease and myeloma: how and why you will do it. Skeletal Radiol. 2024;53:1815–1831.
    OpenUrlPubMed
  95. 95.↵
    1. Padhani AR,
    2. Lecouvet FE,
    3. Tunariu N,
    4. et al
    . Metastasis reporting and data system for prostate cancer: practical guidelines for acquisition, interpretation, and reporting of whole-body magnetic resonance imaging-based evaluations of multiorgan involvement in advanced prostate cancer. Eur Urol. 2017;71:81–92.
    OpenUrlCrossRefPubMed
  96. 96.↵
    1. Lecouvet FE,
    2. Talbot JN,
    3. Messiou C,
    4. Bourguet P,
    5. Liu Y,
    6. de Souza NM
    , EORTC Imaging Group. Monitoring the response of bone metastases to treatment with magnetic resonance imaging and nuclear medicine techniques: a review and position statement by the European Organisation for Research and Treatment of Cancer imaging group. Eur J Cancer. 2014;50:2519–2531.
    OpenUrlCrossRefPubMed
  97. 97.↵
    1. Ceci F,
    2. Castellucci P,
    3. Graziani T,
    4. et al
    . 11C-choline PET/CT in castration-resistant prostate cancer patients treated with docetaxel. Eur J Nucl Med Mol Imaging. 2016;43:84–91.
    OpenUrlCrossRefPubMed
  98. 98.↵
    1. Van Damme J,
    2. Tombal B,
    3. Michoux N,
    4. et al
    . Value of whole-body magnetic resonance imaging using the MET-RADS-P criteria for assessing the response to intensified androgen deprivation therapy in metastatic hormone-naïve and castration-resistant prostate cancer. Eur Urol Oncol. November 5, 2024 [Epub ahead of print].
  99. 99.↵
    1. Halabi S,
    2. Kelly WK,
    3. Ma H,
    4. et al
    . Meta-analysis evaluating the impact of site of metastasis on overall survival in men with castration-resistant prostate cancer. J Clin Oncol. 2016;34:1652–1659.
    OpenUrlAbstract/FREE Full Text
  100. 100.↵
    1. Evans MJ,
    2. Smith-Jones PM,
    3. Wongvipat J,
    4. et al
    . Noninvasive measurement of androgen receptor signaling with a positron-emitting radiopharmaceutical that targets prostate-specific membrane antigen. Proc Natl Acad Sci U S A. 2011;108:9578–9582.
    OpenUrlAbstract/FREE Full Text
  101. 101.↵
    1. Hope TA,
    2. Truillet C,
    3. Ehman EC,
    4. et al
    . 68Ga-PSMA-11 PET imaging of response to androgen receptor inhibition: first human experience. J Nucl Med. 2017;58:81–84.
    OpenUrlAbstract/FREE Full Text
  102. 102.↵
    1. Aggarwal R,
    2. Wei X,
    3. Kim W,
    4. et al
    . Heterogeneous flare in prostate-specific membrane antigen positron emission tomography tracer uptake with initiation of androgen pathway blockade in metastatic prostate cancer. Eur Urol Oncol. 2018;1:78–82.
    OpenUrlPubMed
  103. 103.↵
    1. Markowski MC,
    2. Velho PI,
    3. Eisenberger MA,
    4. et al
    . Detection of early progression with 18F-DCFPyL PET/CT in men with metastatic castration-resistant prostate cancer receiving bipolar androgen therapy. J Nucl Med. 2021;62:1270–1273.
    OpenUrlAbstract/FREE Full Text
  104. 104.↵
    1. Parker C,
    2. Nilsson S,
    3. Heinrich D,
    4. et al
    .; ALSYMPCA Investigators. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med. 2013;369:213–223.
    OpenUrlCrossRefPubMed
  105. 105.↵
    1. Bennett R,
    2. Li E,
    3. Ho AY,
    4. et al
    . PSMA PET imaging for evaluation of recurrent or persistent prostate cancer after primary prostate radiation. Journal of Clinical Oncology. 2024;42:35–35.
    OpenUrl
  106. 106.↵
    1. Aydin AM,
    2. Haberal B,
    3. Artykov M,
    4. Bilen CY,
    5. Yazici S
    . Clinicopathological predictors of positive 68Ga-PSMA-11 PET/CT in PSA-only recurrence of localized prostate cancer following definitive therapy. Ann Nucl Med. 2019;33:326–332.
    OpenUrlPubMed
  107. 107.↵
    1. Fendler WP,
    2. Calais J,
    3. Eiber M,
    4. et al
    .; PSMA PET Reader Group. False positive PSMA PET for tumor remnants in the irradiated prostate and other interpretation pitfalls in a prospective multi-center trial. Eur J Nucl Med Mol Imaging. 2021;48:501–508.
    OpenUrlPubMed
  108. 108.↵
    1. Crook J,
    2. Malone S,
    3. Perry G,
    4. Bahadur Y,
    5. Robertson S,
    6. Abdolell M
    . Postradiotherapy prostate biopsies: what do they really mean—results for 498 patients. Int J Radiat Oncol Biol Phys. 2000;48:355–367.
    OpenUrlPubMed
  109. 109.↵
    1. Zelefsky MJ,
    2. Goldman DA,
    3. Reuter V,
    4. et al
    . Long-term implications of a positive posttreatment biopsy in patients treated with external beam radiotherapy for clinically localized prostate cancer. J Urol. 2019;201:1127–1133.
    OpenUrlPubMed
  110. 110.↵
    1. Crook JM,
    2. Malone S,
    3. Perry G,
    4. et al
    . Twenty-four-month postradiation prostate biopsies are strongly predictive of 7-year disease-free survival: results from a Canadian randomized trial. Cancer. 2009;115:673–679.
    OpenUrlCrossRefPubMed
  111. 111.↵
    1. Bergamin S,
    2. Eade T,
    3. Kneebone A,
    4. et al
    . Interim results of a prospective prostate-specific membrane antigen-directed focal stereotactic reirradiation trial for locally recurrent prostate cancer. Int J Radiat Oncol Biol Phys. 2020;108:1172–1178.
    OpenUrlPubMed
  112. 112.↵
    1. Wang F,
    2. Liu C,
    3. Vidal I,
    4. et al
    . Comparison of multiple segmentation methods for volumetric delineation of primary prostate cancer with prostate-specific membrane antigen-targeted 18F-DCFPyL PET/CT. J Nucl Med. 2024;65:87–93.
    OpenUrlAbstract/FREE Full Text
  113. 113.↵
    1. Gafita A,
    2. Menard AR,
    3. Solnes LB
    . Diagnostic radiopharmaceuticals: a sustainable path to the improvement of patient care. J Nucl Med. 2025;66:186.
    OpenUrlFREE Full Text
  114. 114.↵
    1. Khalil MM
    1. Ballinger JR,
    2. Koziorowski J
    . Regulation of PET radiopharmaceuticals production in Europe. In: Khalil MM, ed. Basic Science of PET Imaging. Springer International Publishing; 2017:127–143.
  115. 115.↵
    1. Leung KH,
    2. Rowe SP,
    3. Sadaghiani MS,
    4. et al
    . Deep semisupervised transfer learning for fully automated whole-body tumor quantification and prognosis of cancer on PET/CT. J Nucl Med. 2024;65:643–650.
    OpenUrlAbstract/FREE Full Text
  116. 116.
    1. Leung KH,
    2. Rowe SP,
    3. Leal JP,
    4. et al
    . Deep learning and radiomics framework for PSMA-RADS classification of prostate cancer on PSMA PET. EJNMMI Res. 2022;12:76.
    OpenUrlPubMed
  117. 117.↵
    1. Li Y,
    2. Imami MR,
    3. Zhao L,
    4. et al
    . An automated deep learning-based framework for uptake segmentation and classification on PSMA PET/CT imaging of patients with prostate cancer. J Imaging Inform Med. 2024;37:2206–2215.
    OpenUrlPubMed
  • Received for publication February 3, 2025.
  • Accepted for publication April 21, 2025.
PreviousNext
Back to top

In this issue

Journal of Nuclear Medicine: 66 (6)
Journal of Nuclear Medicine
Vol. 66, Issue 6
June 1, 2025
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Complete Issue (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Journal of Nuclear Medicine.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Treatment Response Evaluation in Prostate Cancer Using PSMA PET/CT
(Your Name) has sent you a message from Journal of Nuclear Medicine
(Your Name) thought you would like to see the Journal of Nuclear Medicine web site.
Citation Tools
Treatment Response Evaluation in Prostate Cancer Using PSMA PET/CT
Andrei Gafita, Jennifer A. Schroeder, Francesco Ceci, Jorge D. Oldan, Amir H. Khandani, Frederic E. Lecouvet, Lilja B. Solnes, Steven P. Rowe
Journal of Nuclear Medicine Jun 2025, jnumed.124.268071; DOI: 10.2967/jnumed.124.268071

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Treatment Response Evaluation in Prostate Cancer Using PSMA PET/CT
Andrei Gafita, Jennifer A. Schroeder, Francesco Ceci, Jorge D. Oldan, Amir H. Khandani, Frederic E. Lecouvet, Lilja B. Solnes, Steven P. Rowe
Journal of Nuclear Medicine Jun 2025, jnumed.124.268071; DOI: 10.2967/jnumed.124.268071
Twitter logo Facebook logo LinkedIn logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One
Bookmark this article

Jump to section

  • Article
    • Abstract
    • ROLE OF PSMA-TARGETED IMAGING IN PC
    • THERAPEUTIC RESPONSE EVALUATION IN PC
    • PET FOR RESPONSE EVALUATION
    • PSMA PET–SPECIFIC CRITERIA FOR RESPONSE EVALUATION IN PC
    • PSMA PET FOR RESPONSE EVALUATION DURING PSMA THERANOSTICS
    • PSMA PET FOR RESPONSE EVALUATION WITH NON-PSMA–TARGETED THERAPIES
    • POTENTIAL ROLE OF PSMA PET IN TREATMENT RESPONSE FOR LOCAL THERAPIES
    • REGULATORY AND REIMBURSEMENT LANDSCAPE
    • CONCLUSION
    • Footnotes
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Approaches to Imaging Immune Activation Using PET
  • Large Language Models and Large Multimodal Models in Medical Imaging: A Primer for Physicians
Show more Continuing Education

Similar Articles

Keywords

  • PSMA
  • prostate-specific membrane antigen
  • prostate cancer
  • radiopharmaceutical therapy
  • response assessment
  • PPP
  • RECIP
SNMMI

© 2025 SNMMI

Powered by HighWire