You are currently viewing a new version of our website. To view the old version click .
Cancers
  • Review
  • Open Access

20 November 2025

PSMA Theranostics in Prostate Cancer and Beyond: Current and Future Perspectives

,
,
,
,
and
1
Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
2
University of Cambridge, Cambridge CB2 1TN, UK
3
Department of Surgery, University of Melbourne, Melbourne, VIC 3010, Australia
4
Department of Urology, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia
This article belongs to the Section Clinical Research of Cancer

Simple Summary

Prostate-specific membrane antigen (PSMA) has changed the diagnosis and treatment of prostate cancer through the development of highly targeted imaging and radioligand therapy. This review summarises the normal function of PSMA and its role in cancer. We highlight the limitations of PSMA as a sole biomarker and discuss emerging genomic, circulating, and imaging biomarkers that can complement PSMA-based theranostics. The scope of PSMA use is being expanded by quantitative imaging, artificial intelligence, and liquid biopsies which enable prompt assessment of disease biology and treatment response. Furthermore, PSMA expression on blood vessels extends its potential beyond prostate cancer into other malignancies. Collectively, these highlight the role of PSMA in an evolving, biomarker-driven approach to personalised and precision oncology.

Abstract

Prostate-specific membrane antigen (PSMA) is a type II transmembrane glycoprotein that has become central to prostate cancer (PCa) diagnostics and treatment. Beyond its enzymatic role in folate and glutamate metabolism, PSMA is upregulated in advanced PCa, where it contributes to angiogenesis, tumour progression, and therapeutic resistance. This review integrates current understanding of PSMA biology with an emphasis on the role of PSMA expression and the hallmarks of cancer-proliferative signalling, metabolic adaptation, and evasion of cell death. While PSMA has revolutionised theranostic strategies in PCa, its utility as a sole biomarker is limited in select cases such as neuroendocrine differentiation and discordant disease biology. To address these challenges, we highlight emerging biomarkers and novel imaging markers that complement PSMA, including genomic alterations, circulating tumour markers, and exosomal microRNAs. Advances in radiomics and dual-tracer positron emission tomography (PET) further refine patient selection by capturing aggressive low-PSMA phenotypes. Furthermore, PSMA-PET is showing promise in other malignancies, including renal cell carcinoma (RCC) and glioblastoma multiforme (GBM), where neovasculature expression may extend its theranostic applications beyond PCa. By situating PSMA within this broader biomarker landscape, we outline opportunities for theranostic integration, including predictive models, combination therapies and expansion into non-prostate malignancies. Understanding the biology of PSMA in conjunction with novel biomarkers provides a framework for optimising theranostic applications and advancing personalised cancer care.

1. Introduction

The emergence of molecular imaging and targeted therapy—theranostics—has transformed, and expanded, the field of precision oncology. Prostate-specific membrane antigen (PSMA) is central to this, enabling sensitive disease localisation with PSMA-positron emission tomography/computed tomography (PSMA-PET/CT) and lesion-directed radioligand therapy (RLT) using α-β-emitting agents for patients with prostate cancer (PCa) [1,2]. Randomised trials including TheraP and VISION have validated these approaches [2,3]. Nonetheless, PSMA biology, imaging interpretation, and therapeutic applications remain dynamic with not all lesions and patients responding equally. The future of precision oncology will depend on the integration of complementary biomarkers to refine patient selection, predict therapy response, and overcome resistant disease. This narrative review synthesises the current understanding of PSMA biology and its theranostic translation, incorporating recent developments in quantitative imaging, biomarkers, emerging molecular correlates, and exploring the role of PSMA beyond PCa.

2. PSMA Biology

2.1. Genetics and Structural Profile

PSMA is encoded by folate hydrolase 1 (FOLH1) on chromosome 11p11.2, producing a 750 amino acid type II transmembrane glycoprotein functioning as a zinc metalloenzyme [4,5,6]. PSMA exists as a homodimer, with each monomer containing a central binding cavity with two zinc ions and entrance accommodating glutamate moieties [7]. PSMA catalyses the hydrolysis of polyglutamated folates which regulates folate and glutamate metabolism [8]. These catalytic properties are important components of metabolic pathways often upregulated in malignancy [9]. In the context of RLT, ligand binding triggers clathrin-mediated endocytosis facilitating delivery of radio-labelled payloads and concentration within cancer cells [10]. Cryoelectron microscopy and crystallographic analyses have refined understanding of PSMA’s binding pocket, with the identification of key residues that interact with the glutamate-containing motifs present in most RLT [11]. These structural insights guide the rational design of next-generation ligands with improved tumour retention and reduced off-target salivary gland and marrow uptake.

2.2. Regulation, Expression, and Functional Roles

PSMA expression is tightly regulated by androgen-receptor (AR) signalling, chromatin accessibility, and methylation status of chromosome 11p [12]. Along the PCa disease continuum, there is dynamic modulation with upregulation occurring during the transition from hormone-sensitive to castration-resistant states, and increased expression following AR-pathway inhibitor (ARPI) administration [13]. Androgen deprivation induces PSMA transcriptional upregulation whilst neuroendocrine differentiation can suppress PSMA expression, as neuroendocrine cells are largely devoid of PSMA [12]. Both inter- and intra-lesional, spatial, and temporal heterogeneity can complicate interpretation and therapy planning.
PSMA expression is modulated at a chromatin level. Looping between enhancing and promoter regions, differential methylation, and histone-modification patterns can either activate or silence FOLH1 transcription, explaining why some high-grade tumours are PSMA-negative despite aggressive histology [14]. DNA-methylation loss at regulatory CpG (5′-C-phosphate-G-3′) foci and histone-acetylation-gain polymorphisms have been linked to PSMA-high phenotypes, whereas neuroendocrine differentiation is associated with promoter hypermethylation, chromatin compaction, and reduced PSMA expression [14,15]. PSMA expression may also be influenced by microenvironmental factors including hypoxia, oxidative stress, and cytokine signalling, which may upregulate angiogenic factors and PSMA, including Hypoxia-inducible factor-1 (HIF-1α) [14,16,17].

2.3. Functional Roles

PSMA contributes to tumour progression through metabolic remodelling and angiogenesis, enhancing tumour cell invasion and migration via activation of focal-adhesion kinase and integrin-signalling pathways [4]. Recent reports have demonstrated that PSMA is expressed not only in PCa epithelium but also in the endothelial cells of tumour neovasculature across multiple malignancies supporting its exploitation as a vascular theranostic target [18,19,20]. The dual localisation of PSMA in both the epithelial tumour cell and surrounding neovasculature suggests a dual biologic function—sustaining the cancer metabolism and remodelling the vascular niche to sustain increased metabolic activity.

3. Clinical Role of PSMA

3.1. Imaging Biomarkers

PSMA-PET/CT has outperformed conventional imaging for both initial staging and biochemical-recurrence (BCR) localisation, with multiple trials supporting its superiority [1,2]. The proPSMA trial established that PSMA-PET/CT has a 27% higher diagnostic accuracy than combined CT and bone scan for primary staging, identifying additional nodal and distant metastases in approximately 33% of patients [1]. Meta-analyses have confirmed that standardised uptake volume (SUV)max and total PSMA-positive tumour volume correlate with Gleason grade, prostate-specific antigen (PSA) levels, and risk of BCR [21,22]. The utility of SUVmax was explored with the recent post hoc analysis and development of a PRIMARY score, aiding diagnosis of PCa [23].
PSMA states occur in approximately 3–5% of intermediate-to-high-risk cases despite aggressive histology, highlighting biological divergence and heterogeneity [24]. Nonetheless, quantitative PSMA metrics now serve as imaging biomarkers guiding management—baseline uptake predicts outcomes of ARPI administration , chemotherapy, and RLT [25,26,27].
PSMA-PET/CT demonstrates dynamic regulation. Uptake tends to increase with disease progression—from localised hormone-sensitive states (HSPC) to metastatic castration-resistance PCa (mCRPC) [1,12,15,28]. This reflects both cellular upregulation of PSMA and clonal selection under androgen deprivation across the PCa continuum [12]. ARPIs, such as Enzalutamide, transiently augment PSMA expression and enhance imaging contrast, while neuroendocrine differentiation suppresses uptake and poses challenges for therapeutic targeting [29,30,31,32].
Increasingly, PSMA-PET/CT is integrated into radiotherapy planning and response monitoring. Radiotherapy guided by biology utilises PET signal intensity to tailor dose planning, while changes in uptake post-therapy provide response surrogates which can provide increasingly personalised care [33,34].

3.2. Radioligand Therapy

Following TheraP and subsequently the VISION trials, [177Lu]Lu-PSMA-617 is an option for patients with highly PSMA-avid mCRPC [3]. The phase II TheraP trial confirmed that [177Lu]Lu-PSMA-617 had a higher PSA ≥ 50% response rates (66% vs. 37%) and fewer grade-3 adverse events compared to Cabazitaxel, but no overall-survival difference [3]. The phase III VISION trial demonstrated that [177Lu]Lu-PSMA-617 significantly improved overall survival (median 15.3 vs. 11.3 months) and radiological progression-free survival (8.7 vs. 3.4 months) versus standard of care [2]. Although the incidence of adverse events was higher in the treatment arm (52.7% vs. 38.0%), quality of life was not impacted [2]. α-emitters strategies including 225Ac-PSMA-617 and 212Pb-PSMA are being explored to overcome resistance in those patients with bulky disease states, but validation in larger studies is necessary [35,36,37,38].
Combination regimes are emerging—PSMA-RLT with poly ADP ribose polymerase (PARP) inhibition (LuPARP trial), Docetaxel (UpFrontPSMA) are being explored [39,40]. The interim results from the first dual-tracer RLT trial (AlphaBet) were recently published demonstrating a reduction in PSA of at least 50% in approximately 50% of patients, with grade-3 events only reported in five patients [41]. To explore the synergistic benefit of [177Lu]Lu-PSMA-617 and surgery, men with high-risk localised PCa were given upfront [177Lu]Lu-PSMA-617 prior to radical prostatectomy in the LuTectomy study, demonstrating low rates of BCR and low rates of adverse events on follow-up [21,42].
Quantitative imaging metrics inform therapy eligibility and response prediction. Baseline SUVmax correlates with both PSA response and survival following RLT, with patients with extensive PSMA disease demonstrating attenuated benefit to RLT, underscoring the importance of multimodal biomarker assessment [43,44].
Monitoring of therapy and dynamic adaptation based on patient response is an emerging space. Serial PSMA-PET/CT assessment facilitates dynamic evaluation of treatment efficacy, with new PSMA lesions suggesting clonal escape. To manage these complexities, adaptive dosage algorithms, adjusting cumulative [177Lu]Lu-PSMA-617 activity based on residual uptake, are being examined to individualise treatment cycles [43,45,46]. Recently, standardised response frameworks such as Response Evaluation Criteria in PSMA Imaging (RECIP) have been proposed to integrate software-based quantitative assessment of PSMA+ total tumour volume [47]. Gaftia et al. found excellent agreement between visual and quantitative RECIP, further demonstrating that RECIP progressive disease was associated with significantly shorter overall survival compared with non-progressive disease [48]. However, these adaptive strategies warrant validation in larger, prospective trials. Overall, RLT toxicity profiles remain favourable—xerostomia and fatigue are the most common, while haematological toxicity remains uncommon and typically reversible. Protective strategies such as salivary-gland cooling are under active investigation [49]. Overall, RLT toxicity profiles remain favourable—xerostomia and fatigue are the most common, while haematological toxicity remains uncommon, and typically reversible. Protective strategies such as salivary-gland cooling are under active investigation and require validation in larger studies [49].

3.3. Determinants of Response

PSMA uptake depends on receptor density, perfusion, cellular internalisation kinetics, and the tumour microenvironment (TME) (Figure 1) [12]. Recent meta-analyses suggest that SUVmax independently predicts progression-free and overall survival post-RLT in the mCRPC setting [26,43,50]. However, discordant FDG+/PSMA lesions signify dedifferentiation and poor response, warranting dual-tracer imaging [51,52,53].
Figure 1. Determinants of PSMA uptake and response. DDR: DNA damage response.

4. Emerging Biomarkers to Complement PSMA

4.1. Limitations of PSMA as a Sole Biomarker

Despite its clinical success, PSMA is an imperfect universal marker. Heterogeneity of expression, neuroendocrine differentiation/transformation, and transcriptional suppression through epigenetic reprogramming can lead to false-negative imaging [12,13]. Off-target uptake in salivary glands and kidneys limits contrast uptake and may result in adverse effects. Small-volume disease may evade detection due to partial-volume effects and may be mitigated by artificial intelligence (AI) and machine learning (ML)—particularly deep learning-based reconstruction and partial-volume correction networks which work to enhance spatial resolution and quantitative accuracy [54].

4.2. Genomic and Molecular Markers

Defects in DNA damage response (DDR) genes including breast cancer type 1/2 susceptibility gene (BRCA1/2), ataxia-telangiectasia mutated (ATM), checkpoint kinase 2 (CHEK2), and partner and localiser of BRCA2 (PLAB2) are common in advanced disease, occurring in approximately 12% of metastatic PCa cases, and may upregulate PSMA expression via replication stress and metabolic demand [55,56]. These alterations underpin combination strategies pairing RLT with PARP inhibition and are currently being investigated in large trials such as LuPARP [39]. Phosphatase and tension homolog (PTEN) loss activates downstream growth signalling. Dual inhibition of PSMA and growth signalling stimulated by PTEN loss represent a promising therapeutic opportunity, exploiting PSMA-induced nutrient uptake while blocking downstream growth pathways [53,57,58]. The AR-V7 splice variant confers resistance to ARPIs and identification of AR-V7-positive patients can prioritise PSMA-RLT or combination approaches.
Further subtype-specific PSMA biology has been elucidated with transcriptional profiling. Erythroblastosis virus E26 (ETS)-fusion-positive tumours, such as transmembrane-protease and serine-2-ETS-related gene (TMPRSS2-ERG), demonstrate stronger FOLH1 promote activity [12,56]. Integration of these molecular subtype findings with PSMA imaging signatures could help refine patient stratification for RLT [59]. Epigenetic modifications are now emerging as biomarkers. Histone-deacetylase inhibitors and demethylating agents can re-induce FOLH1 expression in low-expression PSMA tumours, potentially making them susceptible to PSMA-targeted radioligands, but this remains validated only in vitro and requires further exploration [14].

4.3. Circulating Biomarkers

Liquid biopsies provide longitudinal insight. Circulating tumour DNA (ctDNA) mirrors mutational burden and clonal evolution, while circulating tumour cell (CTC) enumeration and phenotyping can track tumoral heterogeneity [60,61]. ctDNA profiling can help detect DDR and PTEN alterations that predict PSMA-RLT responsiveness, and serial ctDNA sampling may provide insight into emergent mutations that may signal developing RLT resistance. Quantification of total ctDNA may serve as a biomarker of advanced PCa [62,63]. Phenotyping and CTC enumeration provide some prognostic value—high baseline CTC counts correlate with shorter progression-free survival, and detection of PSMA+ CTCs can complement imaging in the assessment of intra-lesional heterogeneity [64]. Therapeutic response can be predicted by measuring exosomal miRNA such as miR-141 as well as mRNA cargo reflecting FOLH1, DDR, or AR-V7 status [65]. The integration of exosomal and PSMA-PET/CT data could enable real-time adaptive therapeutic approaches—escalating RLT with miRNA spikes or de-escalating when ctDNA clears.

4.4. Imaging Biomarkers

Radiomics derived from PSMA-PET/CT, including texture, entropy, and total lesion volume, correlate strongly with outcomes beyond SUVmax [13,66]. There exist multiple other targets that may complement the use of PSMA-tracer imaging in challenging circumstances of low-uptake disease states (Table 1). These alternative molecular targets illustrate a shift toward multi-target theranostics, replacing single-antigen approaches to address tumour heterogeneity which is common in advanced disease states. While these radiomics show promise, implementation into routine clinical decision-making faces challenges due to a lack of cross-centre standardisation, small cohorts, and variations in reconstruction protocols across centres. Thus, there is need for multi-institutional radiomic repositories and data harmonisation frameworks to resolve these limitations [67]. Furthermore, data privacy and interoperability are essential for any deployment of large-scale AI models.
Table 1. Biomarkers under investigation for targeting.

4.5. Tumour Microenvironment Markers

PSMA expression interacts with the TME and can influence theranostic response. Hypoxia reduces β-particle effectiveness and may induce PSMA downregulation through metabolic reprogramming and vascular endothelial growth factor (VEGF)-driven angiogenesis [53,78,79]. To address these limitations, hypoxia-PET tracers, such as 18F-FAZA and 64Cu-ATSM, are used to guide patient selection and RLT [80,81]. Stromal and angiogeneic factors—VEGF and HIF-1α—correlate with PSMA uptake on endothelial cells [16,82]. Given that PSMA is expressed on the endothelial cells of tumour neovasculature, these angiogeneic pathways may be exploited to amplify and concentrate ligand delivery, explaining the strong uptake in highly vascular tumours such as renal cell carcinoma (RCC) and glioblastoma multiforme (GBM). RLT has a complex interplay with the tumour microenvironment, with a recent review by Eapen et al. suggesting that beyond PSMA+-cell death, [177Lu]Lu-PSMA-617 treatment may stimulate a systemic immune response which may be harnessed to produce long-lasting cancer immunity [17].

4.6. Immune Markers

Immune checkpoint biomarkers represent another relevant pathway of exploration. High PSMA expression co-exists with increased programmed death ligand 1 (PD-L1) expression in subsets of men with PCa, supporting the need for combined RLT and immunotherapy strategies [83,84]. There are now early-phase trials, such as PRINCE, that have shown tolerability and potential additive efficacy of RLT and Pembrolizumab [85].
The future of PSMA-based theranostics, and more broadly RLT depends on a multimodal approach. Combining genomic factors (DDR, PTEN, AR-V7 variants, PSMA mRNA), radiomic factors (SUVmax-based metrics), and liquid biopsy data (ctDNA/CTCs/exosome counts) can generate composite risk models to guide individualised treatment algorithms. AI may augment these approaches and provide robust means to predict RLT response and toxicity with high accuracy. These frameworks will move PSMA theranostics into a new era of dynamic, personalised precision medicine.

5. PSMA Theranostics Beyond PCa

PSMA is expressed on the endothelium of tumour neovasculature in non-PCa malignancies, reflecting the role of the enzyme in angiogenic states and highlighting its utility beyond the prostate. This provides promise for enabling vascular-targeted imaging in a multitude of tumours.

5.1. Renal Cell Carcinoma (RCC)

PSMA is highly expressed in the endothelial cells of clear-cell RCC neovasculature [86]. In a recent systematic review by Sadaghiani et al., PSMA-PET/CT demonstrated promise in the restaging setting, but the review was limited by small patient numbers and heterogeneity necessitating further research [87]. In several prospective series, 68Ga-PSMA-11 and 18F-DCFPyL PET identified nodal and pulmonary metastases missed by conventional imaging, with uptake intensity correlating with histologic grade [88,89]. Prospective trials are now under way to examine [177Lu]Lu-PSMA RLT in metastatic RCC [90,91].
The role of PSMA-PET/CT may extend beyond detection of occult disease in RCC. In their study, Khaleel et al. assessed the correlation between FOLH1 expression and gene expression signature scores that corresponded to the TME [91]. Increased FOLH1 expression was significantly associated with higher TME angiogenesis, and expression predicted progression-free survival in patients with metastatic clear-cell RCC treated with the tyrosine kinase inhibitor Sunitinib [91]. This suggests that PSMA-PET/CT imaging could be utilised as a non-invasive marker to guide systemic therapy options and predict the treatment response to VEGF-inhibiting agents in patients with metastatic clear-cell RCC.

5.2. Salivary-Gland Tumours

Adenoid cystic carcinoma (ACC) of the salivary glands often exhibits strong PSMA uptake in tumour cells and neovasculature [92]. 68Ga-PSMA PET identifies primary and metastatic ACC lesions with high contrast [92]. Although a small pilot study, Wang et al. found that amongst patients diagnosed with ACC, 68Ga-PSMA PET revealed more PET+ extrapulmonary tumours than 18F-FDG PET, but fewer PET+ pulmonary lesions, suggesting that a combination of both imaging modalities may be optimal in patients with ACC [93]. Nonetheless, prospective multi-centre studies are necessary to validate these findings. Prospective case series have reported mixed efficacy profiles in patients treated with [177Lu]Lu-PSMA therapy for ACC, but treatment has been well tolerated with rapid relief of tumour-associated discomfort [94,95].

5.3. Glioblastoma Multiforme (GBM)

GBMs express PSMA in tumour microvessels and in reactive astrocytes [82]. Immunohistochemistry studies have demonstrated PSMA staining in GBMs, but this has not correlated with tracer uptake [96]. 68Ga-PSMA PET detects regions of active glioma with higher specificity than conventional imaging [97]. Given that PSMA expression in endothelial-targeted PSMA-RLT acts as vascular-targeted radiotherapeutic agent. Earlier clinical experience with [177Lu]Lu-PSMA in recurrent GBM has demonstrated feasibility and safety, with stabilisation of disease in pre-treated patients, but requires validation in larger series [98,99].

5.4. Translational Insights

Across non-PCa malignancies, PSMA expression tends to be localised to proliferating endothelial cells within the architecture of neovasculature, rather than the malignant epithelial pool. This expands the functioning definition of PSMA from a prostate-specific antigen to a pan-angiogenic marker of tumour microvasculature. These findings provide promising insights for the future of oncological therapy. PSMA ligands may act as vascular-targeted radiotherapeutics, providing selective ablation of tumour blood supply.

5.5. Clinical Challenges

Despite promising early results, several challenges remain: (1) Heterogenous PSMA expression across tumour types even within a single lesion necessitates companion imaging to accurately select patients. (2) Physiological uptake in off-target organs complicates imaging interpretation and dosimetry. (3) Randomised trials are lacking with most data derived from small single-centre cohorts with an undefined state for appropriate dosimetry in non-prostate malignancies. (4) Ligand optimisation is needed to increase vascular endothelial penetration and limit off-target binding. Nevertheless, a new theme of PSMA marking pathological angiogenesis has emerged, positioning it as a promising target for vascular theranostics. Further studies may build on these findings by utilising a multi-tracer approach to capture both the tumour and surrounding neovasculature, providing a holistic map of the tumour, perfusion, and metabolism.

6. Future Directions

A future direction in PSMA theranostics is the integration of geonomic, imaging, and circulating biomarkers into multimodal predictive frameworks. These models should include a combination of PSMA-PET metrics with ctDNA, DDR gene status, and transcriptomic signatures to guide appropiate patient selection for therapy [63,100].

6.1. Quantitative and AI-Driven Imaging

The evolution of PSMA theranostics is being driven by ongoing exploration of quantitative imaging metrics including SUVmax, PSMA-derived tumour volume, and total lesion PSMA uptake [13]. These serve as imaging biomarkers to predict and evaluate therapy response and may be combined with genomic features to form a hybrid framework linking PSMA-PET/CT phenotypes with genomic features. AI and ML models are transforming PSMA-PET interpretation from solely qualitative to predictive analytical tools [101].

6.2. Adaptive and Combination Therapy

Traditionally, PSMA-RLT protocols have used fixed cycles, but adaptive dosing guided by imaging and biomarkers is a promising approach on the horizon. Serial PSMA-PET and liquid biopsy may enable real-time therapy adaptation, with rising ctDNA or the appearance of PSMA- lesions triggering escalation to α-emitter therapy, or a switch to combination regimes. On the other hand, biochemical and imaging responses could justify treatment de-escalation, minimise toxicity, and reduce costs. The future of PSMA-based therapy is combinatorial, integrating molecularly matched agents to overcome resistant clones. Sequential α/β-emitter regimes are gaining traction [41]. Recently, Kluge et al. examined the relationship between cell-free DNA levels and PSMA-positive tumour volume, finding a weak correlation [62]. This is contrary to Amseian et al., who found no correlation in their prospective study, emphasising the need for larger, prospective studies [102].

6.3. Next-Generation Ligands and Isotopes

Ligand engineering aims to optimise tumour retention and clearance kinetics, minimising off-target internalisation. Modifications to linker length, charge, and hydrophilicity have yielded 64Cu-PSMA-CM ligands with improved albumin binding and extended circulation time and more are in development to improve binding to PSMA and minimise off-target toxicity [103]. Bispecific ligands combining GRPR or FAP motifs are being validated, aiming to overcome inter-lesional heterogeneity by binding multiple antigens simultaneously, ensuring radioligand delivery even when a single antigenic target is downregulated [68,104]

6.4. Future Outlooks

The next horizon is the integration of imaging, molecular, and clinical data in predictive models. In their models, Gafita et al. developed a normogram that combined PSMA-PET/CT-derived tumour volume, SUVmean and baseline clinical features to predict overall survival following [177Lu]Lu-PSMA-617 therapy [43]. Similarly, Pan et al. used a dual-tracer approach to identify PSMA/FDG+ discordant disease in a multi-centre study to develop nomograms and predict poor RLT response [105].
Despite these initial results, validation is necessary to ensure cross-centre reproducibility. Standardisation of reconstructive protocols, such as European Association of Nuclear Medicine (EANM)/EANM Research Ltd. (EARL)-compliant imaging, is essential to ensure consistentcy across institutions [106]. Given significant concern surrounding privacy, federated-learning approaches enable collaborative model training without direct data sharing [107]. Adherence to reporting standards such as the Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis + AI (TRIPOD+AI) will improve transparency of studies developing prediction models [108]. Collectively, these strategies support the development of reproducible predictive models that can guide adaptive PSMA theranostics.
The convergence of imaging, therapy, and AI marks the beginning of a new era in theranostics, with PSMA remaining at the cornerstone of this transformation, defining the next generation of precision oncology. In the next decade, PSMA-directed RLT may evolve from a salvage option for mCRPC to an earlier-line, combination-based, cross-tumour modality. Nonetheless, it is important that these resources are distributed equally across healthcare services as they emerge to avoid inequalities [109,110].

7. Conclusions

PSMA has ushered in a new era of successful, targeted, molecular theranostics, but PSMA expression is dynamic and context-dependent. Quantitative imaging metrics, genomic signatures, and microenvironmental cues can refine patient selection and guide combination approaches. The future or precision oncology will integrate PSMA with a constellation of biomarkers to personalise theranostic strategies. Moreover, PSMA’s endothelial expression across multiple malignancies positions it as the gateway to a new era of biomarker-driven pan-cancer theranostic strategy. Nonetheless, future research is needed to standardise imaging and molecular criteria to define PSMA- disease, as well as integration of multi-omic signature into decision algorithms and prospective trials to confirm their utility in appropriate patient selection and adaptive dosing regimens.

Author Contributions

K.S.: Conceptualisation, writing—original draft preparation, writing—review and editing. D.C.: Writing—review and editing. D.H.: Writing—review and editing. D.G.M.: Supervision, writing—review and editing. N.L.: Supervision. M.P. and D.G.M.: Supervision, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. 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] [PubMed]
  2. Sartor, O.; de Bono, J.; Chi, K.N.; Fizazi, K.; Herrmann, K.; Rahbar, K.; Tagawa, S.T.; Nordquist, L.T.; Vaishampayan, N.; El-Haddad, G.; et al. Lutetium-177-PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2021, 385, 1091–1103. [Google Scholar] [CrossRef] [PubMed]
  3. Hofman, M.S.; Emmett, L.; Sandhu, S.; Iravani, A.; Joshua, A.M.; Goh, J.C.; Pattison, D.A.; Tan, T.H.; Kirkwood, I.D.; Ng, S.; et al. [(177)Lu]Lu-PSMA-617 versus cabazitaxel in patients with metastatic castration-resistant prostate cancer (TheraP): A randomised, open-label, phase 2 trial. Lancet 2021, 397, 797–804. [Google Scholar] [CrossRef] [PubMed]
  4. O’Keefe, D.S.; Bacich, D.J.; Huang, S.S.; Heston, W.D.W. A Perspective on the Evolving Story of PSMA Biology, PSMA-Based Imaging, and Endoradiotherapeutic Strategies. J. Nucl. Med. 2018, 59, 1007–1013. [Google Scholar] [CrossRef]
  5. O’Keefe, D.S.; Bacich, D.J.; Heston, W.D. Comparative analysis of prostate-specific membrane antigen (PSMA) versus a prostate-specific membrane antigen-like gene. Prostate 2004, 58, 200–210. [Google Scholar] [CrossRef]
  6. Rawlings, N.D.; Barrett, A.J. Structure of membrane glutamate carboxypeptidase. Biochim. Biophys. Acta 1997, 1339, 247–252. [Google Scholar] [CrossRef]
  7. Machulkin, A.E.; Petrov, S.A.; Bodenko, V.; Larkina, M.S.; Plotnikov, E.; Yuldasheva, F.; Tretyakova, M.; Bezverkhniaia, E.; Zyk, N.Y.; Stasyuk, E.; et al. Synthesis and Preclinical Evaluation of Urea-Based Prostate-Specific Membrane Antigen-Targeted Conjugates Labeled with (177)Lu. ACS Pharmacol. Transl. Sci. 2024, 7, 1457–1473. [Google Scholar] [CrossRef]
  8. Ghosh, A.; Heston, W.D. Effect of carbohydrate moieties on the folate hydrolysis activity of the prostate specific membrane antigen. Prostate 2003, 57, 140–151. [Google Scholar] [CrossRef]
  9. Yao, V.; Parwani, A.; Maier, C.; Heston, W.D.; Bacich, D.J. Moderate expression of prostate-specific membrane antigen, a tissue differentiation antigen and folate hydrolase, facilitates prostate carcinogenesis. Cancer Res. 2008, 68, 9070–9077. [Google Scholar] [CrossRef]
  10. Ghosh, A.; Heston, W.D. Tumor target prostate specific membrane antigen (PSMA) and its regulation in prostate cancer. J. Cell Biochem. 2004, 91, 528–539. [Google Scholar] [CrossRef]
  11. Schäfer, M.; Bauder-Wüst, U.; Roscher, M.; Motlová, L.; Kutilová, Z.; Remde, Y.; Klika, K.D.; Graf, J.; Bařinka, C.; Benešová-Schäfer, M. Structure-Activity Relationships and Biological Insights into PSMA-617 and Its Derivatives with Modified Lipophilic Linker Regions. ACS Omega 2025, 10, 7077–7090. [Google Scholar] [CrossRef] [PubMed]
  12. Bakht, M.K.; Beltran, H. Biological determinants of PSMA expression, regulation and heterogeneity in prostate cancer. Nat. Rev. Urol. 2025, 22, 26–45. [Google Scholar] [CrossRef] [PubMed]
  13. Al Saffar, H.; Chen, D.C.; Delgado, C.; Ingvar, J.; Hofman, M.S.; Lawrentschuk, N.; Perera, M.; Murphy, D.G.; Eapen, R. The Current Landscape of Prostate-Specific Membrane Antigen (PSMA) Imaging Biomarkers for Aggressive Prostate Cancer. Cancers 2024, 16, 939. [Google Scholar] [CrossRef] [PubMed]
  14. Sayar, E.; Patel, R.A.; Coleman, I.M.; Roudier, M.P.; Zhang, A.; Mustafi, P.; Low, J.Y.; Hanratty, B.; Ang, L.S.; Bhatia, V.; et al. Reversible epigenetic alterations mediate PSMA expression heterogeneity in advanced metastatic prostate cancer. JCI Insight 2023, 8, e162907. [Google Scholar] [CrossRef]
  15. Sheehan, B.; Guo, C.; Neeb, A.; Paschalis, A.; Sandhu, S.; de Bono, J.S. Prostate-specific Membrane Antigen Biology in Lethal Prostate Cancer and its Therapeutic Implications. Eur. Urol. Focus. 2022, 8, 1157–1168. [Google Scholar] [CrossRef]
  16. Vlachostergios, P.J.; Karathanasis, A.; Dimitropoulos, K.; Zachos, I.; Tzortzis, V. High PSMA expression is associated with immunosuppressive tumor microenvironment in clear cell renal cell carcinoma. Precis. Clin. Med. 2024, 7, pbae010. [Google Scholar] [CrossRef]
  17. Eapen, R.S.; Williams, S.G.; Macdonald, S.; Keam, S.P.; Lawrentschuk, N.; Au, L.; Hofman, M.S.; Murphy, D.G.; Neeson, P.J. Neoadjuvant lutetium PSMA, the TIME and immune response in high-risk localized prostate cancer. Nat. Rev. Urol. 2024, 21, 676–686. [Google Scholar] [CrossRef]
  18. Wang, G.; Li, L.; Wang, J.; Zang, J.; Chen, J.; Xiao, Y.; Fan, X.; Zhu, L.; Kung, H.F.; Zhu, Z. Head-to-head comparison of [(68)Ga]Ga-P16-093 and 2-[(18)F]FDG PET/CT in patients with clear cell renal cell carcinoma: A pilot study. Eur. J. Nucl. Med. Mol. Imaging 2023, 50, 1499–1509. [Google Scholar] [CrossRef]
  19. Pellegrino, S.; Fonti, R. A look into the future: The role of PSMA beyond prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 2023, 51, 278–280. [Google Scholar] [CrossRef]
  20. Van de Wiele, C.; Sathekge, M.; de Spiegeleer, B.; De Jonghe, P.J.; Debruyne, P.R.; Borms, M.; Beels, L.; Maes, A. PSMA expression on neovasculature of solid tumors. Histol. Histopathol. 2020, 35, 919–927. [Google Scholar] [CrossRef]
  21. Mazzone, E.; Thomson, A.; Chen, D.C.; Cannoletta, D.; Quarta, L.; Pellegrino, A.; Gandaglia, G.; Moon, D.; Eapen, R.; Lawrentschuk, N.; et al. The Role of Prostate-specific Membrane Antigen Positron Emission Tomography for Assessment of Local Recurrence and Distant Metastases in Patients with Biochemical Recurrence of Prostate Cancer After Definitive Treatment: A Systematic Review and Meta-analysis. Eur. Urol. 2025, 88, 129–141. [Google Scholar] [CrossRef]
  22. Mazzone, E.; Cannoletta, D.; Quarta, L.; Chen, D.C.; Thomson, A.; Barletta, F.; Stabile, A.; Moon, D.; Eapen, R.; Lawrentschuk, N.; et al. A Comprehensive Systematic Review and Meta-analysis of the Role of Prostate-specific Membrane Antigen Positron Emission Tomography for Prostate Cancer Diagnosis and Primary Staging before Definitive Treatment. Eur. Urol. 2025, 87, 654–671. [Google Scholar] [CrossRef] [PubMed]
  23. Emmett, L.; Papa, N.; Buteau, J.; Ho, B.; Liu, V.; Roberts, M.; Thompson, J.; Moon, D.; Sheehan-Dare, G.; Alghazo, O.; et al. The PRIMARY Score: Using Intraprostatic (68)Ga-PSMA PET/CT Patterns to Optimize Prostate Cancer Diagnosis. J. Nucl. Med. 2022, 63, 1644–1650. [Google Scholar] [CrossRef] [PubMed]
  24. Veerman, H.; Donswijk, M.; Bekers, E.; Olde Heuvel, J.; Bodar, Y.J.L.; Boellaard, T.N.; van Montfoort, M.L.; van Moorselaar, R.J.A.; Oprea-Lager, D.E.; van Leeuwen, P.J.; et al. The clinical characteristics of patients with primary non-prostate-specific membrane antigen-expressing prostate cancer on preoperative positron emission tomography/computed tomography. BJU Int. 2022, 129, 314–317. [Google Scholar] [CrossRef] [PubMed]
  25. Shagera, Q.A.; Karfis, I.; Kristanto, P.; Spyridon, S.; Diamand, R.; Santapau, A.; Peltier, A.; Roumeguère, T.; Flamen, P.; Artigas, C. 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. [Google Scholar] [CrossRef]
  26. Yanagisawa, T.; Matsukawa, A.; Rajwa, P.; Miszczyk, M.; Fazekas, T.; Pradere, B.; Miyajima, K.; Enei, Y.; Cormio, A.; Dematteis, A.; et al. Prognostic factors of PSMA-targeted radioligand therapy in metastatic castration-resistant prostate cancer: A systematic review and meta-analysis. Prostate Cancer Prostatic Dis. 2025. [Google Scholar] [CrossRef]
  27. Di Franco, M.; Mei, R.; Garcia, C.; Fanti, S. Treatment response assessment in mCRPC: Is PSMA-PET/CT going to take the lead? Ther. Adv. Med. Oncol. 2024, 16, 17588359241258367. [Google Scholar] [CrossRef]
  28. Emmett, L.; Yin, C.; Crumbaker, M.; Hruby, G.; Kneebone, A.; Epstein, R.; Nguyen, Q.; Hickey, A.; Ihsheish, N.; O’Neill, G.; et al. Rapid Modulation of PSMA Expression by Androgen Deprivation: Serial (68)Ga-PSMA-11 PET in Men with Hormone-Sensitive and Castrate-Resistant Prostate Cancer Commencing Androgen Blockade. J. Nucl. Med. 2019, 60, 950–954. [Google Scholar] [CrossRef]
  29. Emmett, L.; Subramaniam, S.; Crumbaker, M.; Nguyen, A.; Joshua, A.M.; Weickhardt, A.; Lee, S.T.; Ng, S.; Francis, R.J.; Goh, J.C.; et al. [(177)Lu]Lu-PSMA-617 plus enzalutamide in patients with metastatic castration-resistant prostate cancer (ENZA-p): An open-label, multicentre, randomised, phase 2 trial. Lancet Oncol. 2024, 25, 563–571. [Google Scholar] [CrossRef] [PubMed]
  30. Rosar, F.; Neher, R.; Burgard, C.; Linxweiler, J.; Schreckenberger, M.; Hoffmann, M.A.; Bartholoma, M.; Khreish, F.; Ezziddin, S. Upregulation of PSMA Expression by Enzalutamide in Patients with Advanced mCRPC. Cancers 2022, 14, 1696. [Google Scholar] [CrossRef]
  31. van der Gaag, S.; Vis, A.N.; Bartelink, I.H.; Koppes, J.C.C.; Hodolic, M.; Hendrikse, H.; Oprea-Lager, D.E. Exploring the Flare Phenomenon in Patients with Castration-Resistant Prostate Cancer: Enzalutamide-Induced PSMA Upregulation Observed on PSMA PET. J. Nucl. Med. 2025, 66, 373–376. [Google Scholar] [CrossRef]
  32. Iravani, A.; Mitchell, C.; Akhurst, T.; Sandhu, S.; Hofman, M.S.; Hicks, R.J. Molecular Imaging of Neuroendocrine Differentiation of Prostate Cancer: A Case Series. Clin. Genitourin. Cancer 2021, 19, e200–e205. [Google Scholar] [CrossRef]
  33. Vlachostergios, P.J.; Zachos, I.; Tzortzis, V. Biomarkers in Prostate-Specific Membrane Antigen Theranostics. Diagnostics 2021, 11, 1108. [Google Scholar] [CrossRef]
  34. Gaudreault, M.; Chang, D.; Hardcastle, N.; Jackson, P.; Kron, T.; Hanna, G.G.; Hofman, M.S.; Siva, S. Utility of Biology-Guided Radiotherapy to De Novo Metastases Diagnosed During Staging of High-Risk Biopsy-Proven Prostate Cancer. Front. Oncol. 2022, 12, 854589. [Google Scholar] [CrossRef]
  35. Kratochwil, C.; Bruchertseifer, F.; Giesel, F.L.; Weis, M.; Verburg, F.A.; Mottaghy, F.; Kopka, K.; Apostolidis, C.; Haberkorn, U.; Morgenstern, A. 225Ac-PSMA-617 for PSMA-Targeted α-Radiation Therapy of Metastatic Castration-Resistant Prostate Cancer. J. Nucl. Med. 2016, 57, 1941–1944. [Google Scholar] [CrossRef] [PubMed]
  36. Sathekge, M.; Bruchertseifer, F.; Knoesen, O.; Reyneke, F.; Lawal, I.; Lengana, T.; Davis, C.; Mahapane, J.; Corbett, C.; Vorster, M.; et al. (225)Ac-PSMA-617 in chemotherapy-naive patients with advanced prostate cancer: A pilot study. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 129–138. [Google Scholar] [CrossRef] [PubMed]
  37. Rosar, F.; Krause, J.; Bartholoma, M.; Maus, S.; Stemler, T.; Hierlmeier, I.; Linxweiler, J.; Ezziddin, S.; Khreish, F. Efficacy and Safety of [(225)Ac]Ac-PSMA-617 Augmented [(177)Lu]Lu-PSMA-617 Radioligand Therapy in Patients with Highly Advanced mCRPC with Poor Prognosis. Pharmaceutics 2021, 13, 722. [Google Scholar] [CrossRef]
  38. Puttick, S.; Griffiths, M.; Pattison, D.; Hanson, A.; Latter, M.; Kuan, K.; Taylor, S.; Tieu, W.; Kryza, T.; Meyrick, D.; et al. Development of a Novel 212Pb-based Targeted Alpha Therapy for metastatic Castration-Resistant Prostate Cancer. J. Nucl. Med. 2024, 65 (Suppl. 2), 242474. [Google Scholar]
  39. Sandhu, S.; Joshua, A.M.; Emmett, L.; Crumbaker, M.; Bressel, M.; Huynh, R.; Banks, P.D.; Wallace, R.; Hamid, A.; Inderjeeth, A.J.; et al. LuPARP: Phase 1 trial of 177Lu-PSMA-617 and olaparib in patients with metastatic castration resistant prostate cancer (mCRPC). J. Clin. Oncol. 2023, 41, 5005. [Google Scholar] [CrossRef]
  40. Hallqvist, A.; Brynjarsdóttir, E.; Krantz, T.; Sjögren, M.; Svensson, J.; Bernhardt, P. (177)Lu-DOTATATE in Combination with PARP Inhibitor Olaparib Is Feasible in Patients with Somatostatin-Positive Tumors: Results from the LuPARP Phase I Trial. J. Nucl. Med. 2025, 66, 707–712. [Google Scholar] [CrossRef]
  41. Kostos, L.; Buteau, J.P.; Xie, J.; Cardin, A.; Akhurst, T.; Alipour, R.; Au, L.; Chan, J.; Chin, K.Y.; Emmerson, B.; et al. Lutetium-177 [177Lu]Lu-PSMA-I&T plus radium-223 in patients with metastatic castration-resistant prostate cancer (AlphaBet): An interim analysis of the investigator-initiated, single-centre, single-arm, phase 1/2 trial. Lancet Oncol. 2025, 26, 1479–1488. [Google Scholar] [CrossRef] [PubMed]
  42. Eapen, R.S.; Buteau, J.P.; Jackson, P.; Mitchell, C.; Oon, S.F.; Alghazo, O.; McIntosh, L.; Dhiantravan, N.; Scalzo, M.J.; O’Brien, J.; et al. Administering [(177)Lu]Lu-PSMA-617 Prior to Radical Prostatectomy in Men with High-risk Localised Prostate Cancer (LuTectomy): A Single-centre, Single-arm, Phase 1/2 Study. Eur. Urol. 2024, 85, 217–226. [Google Scholar] [CrossRef] [PubMed]
  43. Gafita, A.; Calais, J.; Grogan, T.R.; Hadaschik, B.; Wang, H.; Weber, M.; Sandhu, S.; Kratochwil, C.; Esfandiari, R.; Tauber, R.; et al. Nomograms to predict outcomes after (177)Lu-PSMA therapy in men with metastatic castration-resistant prostate cancer: An international, multicentre, retrospective study. Lancet Oncol. 2021, 22, 1115–1125. [Google Scholar] [CrossRef]
  44. Emmett, L.; Papa, N.; Subramaniam, S.; Crumbaker, M.; Nguyen, A.; Joshua, A.M.; Sandhu, S.; Weickhardt, A.; Lee, S.T.; Ng, S.; et al. Prognostic and predictive value of baseline PSMA-PET total tumour volume and SUVmean in metastatic castration-resistant prostate cancer in ENZA-p (ANZUP1901): A substudy from a multicentre, open-label, randomised, phase 2 trial. Lancet Oncol. 2025, 26, 1168–1177. [Google Scholar] [CrossRef]
  45. Murthy, V.; Gafita, A.; Thin, P.; Nguyen, K.; Grogan, T.; Shen, J.; Drakaki, A.; Rettig, M.; Czernin, J.; Calais, J. Prognostic Value of End-of-Treatment PSMA PET/CT in Patients Treated with (177)Lu-PSMA Radioligand Therapy: A Retrospective, Single-Center Analysis. J. Nucl. Med. 2023, 64, 1737–1743. [Google Scholar] [CrossRef] [PubMed]
  46. Violet, J.; Jackson, P.; Ferdinandus, J.; Sandhu, S.; Akhurst, T.; Iravani, A.; Kong, G.; Kumar, A.R.; Thang, S.P.; Eu, P.; et al. Dosimetry of (177)Lu-PSMA-617 in Metastatic Castration-Resistant Prostate Cancer: Correlations Between Pretherapeutic Imaging and Whole-Body Tumor Dosimetry with Treatment Outcomes. J. Nucl. Med. 2019, 60, 517–523. [Google Scholar] [CrossRef]
  47. Gafita, A.; Rauscher, I.; Weber, M.; Hadaschik, B.; Wang, H.; Armstrong, W.R.; Tauber, R.; Grogan, T.R.; Czernin, J.; Rettig, M.B.; 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. [Google Scholar] [CrossRef]
  48. Gafita, A.; Djaileb, L.; Rauscher, I.; Fendler, W.P.; Hadaschik, B.; Rowe, S.P.; Herrmann, K.; Calais, J.; Rettig, M.; Eiber, M.; et al. Response Evaluation Criteria in PSMA PET/CT (RECIP 1.0) in Metastatic Castration-resistant Prostate Cancer. Radiology 2023, 308, e222148. [Google Scholar] [CrossRef]
  49. van Kalmthout, L.W.M.; Lam, M.; de Keizer, B.; Krijger, G.C.; Ververs, T.F.T.; de Roos, R.; Braat, A. Impact of external cooling with icepacks on (68)Ga-PSMA uptake in salivary glands. EJNMMI Res. 2018, 8, 56. [Google Scholar] [CrossRef]
  50. Chen, D.C.; Buteau, J.P.; Papa, N.; Akhurst, T.; Alipour, R.; Bollampally, N.; Cardin, A.; Eifer, M.; Casanueva Eliceiry, S.; Jackson, P.; et al. Prognostic Value of Initial Imaging and PSA Change with [177Lu]Lu-PSMA-617 Radiopharmaceutical Therapy in Patients with Metastatic Castration-Resistant Prostate Cancer: A ProsTIC Registry Analysis. J. Nucl. Med. 2025. [Google Scholar] [CrossRef]
  51. Michalski, K.; Ruf, J.; Goetz, C.; Seitz, A.K.; Buck, A.K.; Lapa, C.; Hartrampf, P.E. Prognostic implications of dual tracer PET/CT: PSMA ligand and [(18)F]FDG PET/CT in patients undergoing [(177)Lu]PSMA radioligand therapy. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 2024–2030. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, H.; Koumna, S.; Pouliot, F.; Beauregard, J.M.; Kolinsky, M. PSMA Theranostics: Current Landscape and Future Outlook. Cancers 2021, 13, 4023. [Google Scholar] [CrossRef] [PubMed]
  53. Heidegger, I.; Kesch, C.; Kretschmer, A.; Tsaur, I.; Ceci, F.; Valerio, M.; Tilki, D.; Marra, G.; Preisser, F.; Fankhauser, C.D.; et al. Biomarkers to personalize treatment with 177Lu-PSMA-617 in men with metastatic castration-resistant prostate cancer—A state of the art review. Ther. Adv. Med. Oncol. 2022, 14, 17588359221081922. [Google Scholar] [CrossRef] [PubMed]
  54. Azimi, M.S.; Kamali-Asl, A.; Ay, M.R.; Zeraatkar, N.; Hosseini, M.S.; Sanaat, A.; Dadgar, H.; Arabi, H. Deep learning-based partial volume correction in standard and low-dose positron emission tomography-computed tomography imaging. Quant. Imaging Med. Surg. 2024, 14, 2146–2164. [Google Scholar] [CrossRef]
  55. Pritchard, C.C.; Mateo, J.; Walsh, M.F.; De Sarkar, N.; Abida, W.; Beltran, H.; Garofalo, A.; Gulati, R.; Carreira, S.; Eeles, R.; et al. Inherited DNA-Repair Gene Mutations in Men with Metastatic Prostate Cancer. N. Engl. J. Med. 2016, 375, 443–453. [Google Scholar] [CrossRef]
  56. Paschalis, A.; Sheehan, B.; Riisnaes, R.; Rodrigues, D.N.; Gurel, B.; Bertan, C.; Ferreira, A.; Lambros, M.B.K.; Seed, G.; Yuan, W.; et al. Prostate-specific Membrane Antigen Heterogeneity and DNA Repair Defects in Prostate Cancer. Eur. Urol. 2019, 76, 469–478. [Google Scholar] [CrossRef]
  57. Sugawara, T.; Nevedomskaya, E.; Heller, S.; Bohme, A.; Lesche, R.; von Ahsen, O.; Grunewald, S.; Nguyen, H.M.; Corey, E.; Baumgart, S.J.; et al. Dual targeting of the androgen receptor and PI3K/AKT/mTOR pathways in prostate cancer models improves antitumor efficacy and promotes cell apoptosis. Mol. Oncol. 2024, 18, 726–742. [Google Scholar] [CrossRef]
  58. Maes, J.; Gesquière, S.; De Spiegeleer, A.; Maes, A.; Van de Wiele, C. Prostate-Specific Membrane Antigen Biology and Pathophysiology in Prostate Carcinoma, an Update: Potential Implications for Targeted Imaging and Therapy. Int. J. Mol. Sci. 2024, 25, 9755. [Google Scholar] [CrossRef]
  59. Rupp, N.J.; Freiberger, S.N.; Ferraro, D.A.; Laudicella, R.; Heimer, J.; Muehlematter, U.J.; Poyet, C.; Moch, H.; Eberli, D.; Rüschoff, J.H.; et al. Immunohistochemical ERG positivity is associated with decreased PSMA expression and lower visibility in corresponding [(68)Ga]Ga-PSMA-11 PET scans of primary prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 2024, 52, 305–313. [Google Scholar] [CrossRef]
  60. Wyatt, A.W.; Azad, A.A.; Volik, S.V.; Annala, M.; Beja, K.; McConeghy, B.; Haegert, A.; Warner, E.W.; Mo, F.; Brahmbhatt, S.; et al. Genomic Alterations in Cell-Free DNA and Enzalutamide Resistance in Castration-Resistant Prostate Cancer. JAMA Oncol. 2016, 2, 1598–1606. [Google Scholar] [CrossRef]
  61. Zainfeld, D.; Goldkorn, A. Liquid Biopsy in Prostate Cancer: Circulating Tumor Cells and Beyond. Cancer Treat. Res. 2018, 175, 87–104. [Google Scholar] [CrossRef]
  62. Kluge, K.; Einspieler, H.; Haberl, D.; Spielvogel, C.; Stoiber, S.; Vraka, C.; Papp, L.; Wunsch, S.; Egger, G.; Kramer, G.; et al. Examining the Relationship and Prognostic Significance of Cell-Free DNA Levels and the PSMA-Positive Tumor Volume in Men with Prostate Cancer: A Retrospective-Prospective [(68)Ga]Ga-PSMA-11 PET/CT Study. J. Nucl. Med. 2024, 65, 63–70. [Google Scholar] [CrossRef]
  63. Kwan, E.M.; Hofman, M.S.; Ng, S.W.S.; Emmett, L.; Sandhu, S.; Buteau, J.P.; Iravani, A.; Joshua, A.M.; Francis, R.J.; Subhash, V.; et al. Circulating tumour DNA fraction as a predictor of treatment efficacy in a randomized phase 2 trial of [177Lu]Lu-PSMA-617 (LuPSMA) versus cabazitaxel in metastatic castration-resistant prostate cancer (mCRPC) progressing after docetaxel (TheraP ANZUP 1603). J. Clin. Oncol. 2024, 42, 5055. [Google Scholar] [CrossRef]
  64. Vlachostergios, P.J. Circulating tumor cell-based PSMA and PSA expression as a predictive and prognostic tool in prostate cancer. Transl. Cancer Res. 2025, 14, 1511–1515. [Google Scholar] [CrossRef] [PubMed]
  65. Li, Z.; Ma, Y.Y.; Wang, J.; Zeng, X.F.; Li, R.; Kang, W.; Hao, X.K. Exosomal microRNA-141 is upregulated in the serum of prostate cancer patients. Onco Targets Ther. 2016, 9, 139–148. [Google Scholar] [CrossRef] [PubMed]
  66. Zamboglou, C.; Carles, M.; Fechter, T.; Kiefer, S.; Reichel, K.; Fassbender, T.F.; Bronsert, P.; Koeber, G.; Schilling, O.; Ruf, J.; et al. Radiomic features from PSMA PET for non-invasive intraprostatic tumor discrimination and characterization in patients with intermediate- and high-risk prostate cancer—A comparison study with histology reference. Theranostics 2019, 9, 2595–2605. [Google Scholar] [CrossRef]
  67. Filippi, L.; Urso, L.; Bianconi, F.; Palumbo, B.; Marzola, M.C.; Evangelista, L.; Schillaci, O. Radiomics and theranostics with molecular and metabolic probes in prostate cancer: Toward a personalized approach. Expert. Rev. Mol. Diagn. 2023, 23, 243–255. [Google Scholar] [CrossRef]
  68. Liolios, C.; Schäfer, M.; Haberkorn, U.; Eder, M.; Kopka, K. Novel Bispecific PSMA/GRPr Targeting Radioligands with Optimized Pharmacokinetics for Improved PET Imaging of Prostate Cancer. Bioconjugate Chem. 2016, 27, 737–751. [Google Scholar] [CrossRef]
  69. Hoberuck, S.; Michler, E.; Wunderlich, G.; Lock, S.; Holscher, T.; Froehner, M.; Braune, A.; Ivan, P.; Seppelt, D.; Zophel, K.; et al. 68Ga-RM2 PET in PSMA- positive and -negative prostate cancer patients. Nuklearmedizin 2019, 58, 352–362. [Google Scholar] [CrossRef]
  70. Ruigrok, E.A.M.; Verhoeven, M.; Konijnenberg, M.W.; de Blois, E.; de Ridder, C.M.A.; Stuurman, D.C.; Bertarione, L.; Rolfo, K.; de Jong, M.; Dalm, S.U. Safety of [(177)Lu]Lu-NeoB treatment: A preclinical study characterizing absorbed dose and acute, early, and late organ toxicity. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 4440–4451. [Google Scholar] [CrossRef]
  71. Li, W.; Jiang, Z.; Cui, N.; Li, J.; Cheng, L.; Liu, W.; Li, J.; Wang, K. Superiority of FAPI-PET/CT for examining multiple malignant tumors: A retrospective study. Am. J. Cancer Res. 2023, 13, 4547–4559. [Google Scholar]
  72. Sollini, M.; Kirienko, M.; Gelardi, F.; Fiz, F.; Gozzi, N.; Chiti, A. State-of-the-art of FAPI-PET imaging: A systematic review and meta-analysis. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 4396–4414. [Google Scholar] [CrossRef] [PubMed]
  73. Xiang, J.; Wang, R.; Wang, J.; Peng, X.; Wang, Y.; Zhu, Z.; Chen, X.; Zhang, J. Preliminary Safety, Biodistribution, and Dosimetry of Fibroblast activation protein and Integrin αvβ3 dual targeting radioligand 177Lu-DOTA-FAPI-RGD: First-in-Human Results. J. Nucl. Med. 2025, 66 (Suppl. 1), 251663. [Google Scholar]
  74. Kelly, W.K.; Danila, D.C.; Lin, C.C.; Lee, J.L.; Matsubara, N.; Ward, P.J.; Armstrong, A.J.; Pook, D.; Kim, M.; Dorff, T.B.; et al. Xaluritamig, a STEAP1 × CD3 XmAb 2+1 Immune Therapy for Metastatic Castration-Resistant Prostate Cancer: Results from Dose Exploration in a First-in-Human Study. Cancer Discov. 2024, 14, 76–89. [Google Scholar] [CrossRef] [PubMed]
  75. Kelly, W.; Danila, D.; Lin, C.C.; Lee, J.L.; Matsubara, N.; Ward, P.; Armstrong, A.J.; Pook, D.; Kim, M.; Dorff, T.; et al. Interim results from a phase I study of AMG 509 (xaluritamig), a STEAP1 x CD3 XmAb 2+1 immune therapy, in patients with metastatic castration-resistant prostate cancer (mCRPC). Ann. Oncol. 2023, 34, S953–S954. [Google Scholar] [CrossRef]
  76. Zhai, B.-T.; Tian, H.; Sun, J.; Zou, J.-B.; Zhang, X.-F.; Cheng, J.-X.; Shi, Y.-J.; Fan, Y.; Guo, D.-Y. Urokinase-type plasminogen activator receptor (uPAR) as a therapeutic target in cancer. J. Transl. Med. 2022, 20, 135. [Google Scholar] [CrossRef]
  77. Azam, A.; Kurbegovic, S.; Carlsen, E.A.; Andersen, T.L.; Larsen, V.A.; Law, I.; Skjøth-Rasmussen, J.; Kjaer, A. Prospective phase II trial of [(68)Ga]Ga-NOTA-AE105 uPAR-PET/MRI in patients with primary gliomas: Prognostic value and Implications for uPAR-targeted Radionuclide Therapy. EJNMMI Res. 2024, 14, 100. [Google Scholar] [CrossRef]
  78. Shi, Z.; Hu, C.; Zheng, X.; Sun, C.; Li, Q. Feedback loop between hypoxia and energy metabolic reprogramming aggravates the radioresistance of cancer cells. Exp. Hematol. Oncol. 2024, 13, 55. [Google Scholar] [CrossRef]
  79. Weiner, A.B.; Agrawal, R.; Wang, N.K.; Sonni, I.; Li, E.V.; Arbet, J.; Zhang, J.J.H.; Proudfoot, J.A.; Hong, B.H.; Davicioni, E.; et al. Molecular Hallmarks of Prostate-specific Membrane Antigen in Treatment-naive Prostate Cancer. Eur. Urol. 2024, 86, 579–587. [Google Scholar] [CrossRef]
  80. Minagawa, Y.; Shizukuishi, K.; Koike, I.; Horiuchi, C.; Watanuki, K.; Hata, M.; Omura, M.; Odagiri, K.; Tohnai, I.; Inoue, T.; et al. Assessment of tumor hypoxia by 62Cu-ATSM PET/CT as a predictor of response in head and neck cancer: A pilot study. Ann. Nucl. Med. 2011, 25, 339–345. [Google Scholar] [CrossRef]
  81. Mapelli, P.; Picchio, M. 18F-FAZA PET imaging in tumor hypoxia: A focus on high-grade glioma. Int. J. Biol. Markers 2020, 35, 42–46. [Google Scholar] [CrossRef]
  82. Yuile, A.; Lee, A.; Moon, E.A.; Hudson, A.; Kastelan, M.; Miller, S.; Chan, D.; Wei, J.; Back, M.F.; Wheeler, H.R. PSMA Expression Correlates with Improved Overall Survival and VEGF Expression in Glioblastoma. Biomedicines 2023, 11, 1148. [Google Scholar] [CrossRef]
  83. Altunay, B.; Schäfer, L.; Morgenroth, A.; Peña, Q.; Lammers, T.; Saar, M.; Mottaghy, F.M.; Lütje, S. Combining PSMA-Targeted Radiopharmaceutical Therapy with Immunotherapy. J. Nucl. Med. 2025, 66, 1522–1527. [Google Scholar] [CrossRef]
  84. Wang, F.; Wu, L.; Yin, L.; Shi, H.; Gu, Y.; Xing, N. Combined treatment with anti-PSMA CAR NK-92 cell and anti-PD-L1 monoclonal antibody enhances the antitumour efficacy against castration-resistant prostate cancer. Clin. Transl. Med. 2022, 12, e901. [Google Scholar] [CrossRef] [PubMed]
  85. Sandhu, S.; Joshua, A.M.; Emmett, L.; Spain, L.A.; Horvath, L.; Crumbaker, M.; Anton, A.; Wallace, R.; Pasam, A.; Bressel, M.; et al. PRINCE: Phase I trial of 177Lu-PSMA-617 in combination with pembrolizumab in patients with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2022, 40, 5017. [Google Scholar] [CrossRef]
  86. Binzaqr, S.; Kryza, D.; Giraudet, A.L.; Bernhard, J.C.; Gross-Goupil, M.; Yacoub, M.; Margue, G.; Hindié, E.; Morgat, C. Prostate-specific membrane antigen (PSMA) expression in primary and metastatic renal cell cancer (UroCCR-65 study). EJNMMI Res. 2025, 15, 38. [Google Scholar] [CrossRef] [PubMed]
  87. Sadaghiani, M.S.; Baskaran, S.; Gorin, M.A.; Rowe, S.P.; Provost, J.C.; Teslenko, I.; Bilyk, R.; An, H.; Sheikhbahaei, S. Utility of PSMA PET/CT in Staging and Restaging of Renal Cell Carcinoma: A Systematic Review and Metaanalysis. J. Nucl. Med. 2024, 65, 1007–1012. [Google Scholar] [CrossRef]
  88. Li, Y.; Zheng, R.; Zhang, Y.; Huang, C.; Tian, L.; Liu, R.; Liu, Y.; Zhang, Z.; Han, H.; Zhou, F.; et al. Special issue “The advance of solid tumor research in China”: 68Ga-PSMA-11 PET/CT for evaluating primary and metastatic lesions in different histological subtypes of renal cell carcinoma. Int. J. Cancer 2023, 152, 42–50. [Google Scholar] [CrossRef]
  89. Gorin, M.A.; Rowe, S.P.; Hooper, J.E.; Kates, M.; Hammers, H.J.; Szabo, Z.; Pomper, M.G.; Allaf, M.E. PSMA-Targeted (18)F-DCFPyL PET/CT Imaging of Clear Cell Renal Cell Carcinoma: Results from a Rapid Autopsy. Eur. Urol. 2017, 71, 145–146. [Google Scholar] [CrossRef]
  90. Kryza, D.; Vinceneux, A.; Bidaux, A.S.; Garin, G.; Tatu, D.; Cropet, C.; Badel, J.N.; Perol, D.; Giraudet, A.L. A multicentric, single arm, open-label, phase I/II study evaluating PSMA targeted radionuclide therapy in adult patients with metastatic clear cell renal cancer (PRadR). BMC Cancer 2024, 24, 163. [Google Scholar] [CrossRef]
  91. Khaleel, S.; Perera, M.; Papa, N.; Kuo, F.; Golkaram, M.; Rappold, P.; Kotecha, R.R.; Coleman, J.; Russo, P.; Motzer, R.; et al. Gene expression of prostate-specific membrane antigen (FOLH1) in clear cell renal cell carcinoma predicts angiogenesis and response to tyrosine kinase inhibitors. Urol. Oncol. 2025, 43, 192.e21–192.e28. [Google Scholar] [CrossRef]
  92. Tan, B.F.; Tan, W.C.C.; Wang, F.Q.; Lechner, M.; Schartinger, V.H.; Tan, D.S.W.; Loke, K.S.H.; Nei, W.L. PSMA PET Imaging and Therapy in Adenoid Cystic Carcinoma and Other Salivary Gland Cancers: A Systematic Review. Cancers 2022, 14, 3585. [Google Scholar] [CrossRef]
  93. Wang, G.; Zhou, M.; Zang, J.; Jiang, Y.; Chen, X.; Zhu, Z.; Chen, X. A pilot study of 68 Ga-PSMA-617 PET/CT imaging and 177Lu-EB-PSMA-617 radioligand therapy in patients with adenoid cystic carcinoma. EJNMMI Res. 2022, 12, 52. [Google Scholar] [CrossRef] [PubMed]
  94. van Ruitenbeek, N.J.; Uijen, M.J.M.; Driessen, C.M.L.; Peters, S.M.B.; Privé, B.M.; van Engen-van Grunsven, A.C.H.; Konijnenberg, M.W.; Gotthardt, M.; Nagarajah, J.; van Herpen, C.M.L. Lutetium-177-PSMA therapy for recurrent/metastatic salivary gland cancer: A prospective pilot study. Theranostics 2024, 14, 5388–5399. [Google Scholar] [CrossRef] [PubMed]
  95. Klein Nulent, T.J.W.; van Es, R.J.J.; Willems, S.M.; Braat, A.; Devriese, L.A.; de Bree, R.; de Keizer, B. First experiences with (177)Lu-PSMA-617 therapy for recurrent or metastatic salivary gland cancer. EJNMMI Res. 2021, 11, 126. [Google Scholar] [CrossRef] [PubMed]
  96. van Lith, S.A.M.; Pruis, I.J.; Tolboom, N.; Snijders, T.J.; Henssen, D.; ter Laan, M.; te Dorsthorst, M.; Leenders, W.P.J.; Gotthardt, M.; Nagarajah, J.; et al. PET Imaging and Protein Expression of Prostate-Specific Membrane Antigen in Glioblastoma: A Multicenter Inventory Study. J. Nucl. Med. 2023, 64, 1526. [Google Scholar] [CrossRef]
  97. Şahin, M.; Akgun, E.; Sirolu, S.; Can, G.; Sayman, H.B.; Oner Dincbas, F. Is there any additional benefit of (68)Ga-PSMA PET on radiotherapy target volume definition in patients with glioblastoma? Br. J. Radiol. 2022, 95, 20220049. [Google Scholar] [CrossRef]
  98. Kumar, A.; Ballal, S.; Yadav, M.P.; ArunRaj, S.T.; Haresh, K.P.; Gupta, S.; Damle, N.A.; Garg, A.; Tripathi, M.; Bal, C. 177Lu-/68Ga-PSMA Theranostics in Recurrent Glioblastoma Multiforme: Proof of Concept. Clin. Nucl. Med. 2020, 45, e512–e513. [Google Scholar] [CrossRef]
  99. Ghaedian, T.; Alipour, A.; Rakhsha, A.; Nasrollahi, H.; Ghaedian, M.; Andalibi, S.; Saffarian, A. Excellent Response of Glioblastoma Multiforme to [177Lu] Lu-PSMA Therapy. Clin. Nucl. Med. 2022. [Google Scholar] [CrossRef]
  100. Torquato, S.; Pallavajjala, A.; Goldstein, A.; Toro, P.V.; Silberstein, J.L.; Lee, J.; Nakazawa, M.; Waters, I.; Chu, D.; Shinn, D.; et al. Genetic Alterations Detected in Cell-Free DNA Are Associated With Enzalutamide and Abiraterone Resistance in Castration-Resistant Prostate Cancer. JCO Precis. Oncol. 2019, 3, 1–14. [Google Scholar] [CrossRef]
  101. Liu, J.; Sandhu, K.; Woon, D.T.S.; Perera, M.; Lawrentschuk, N. The Value of Artificial Intelligence in Prostate-Specific Membrane Antigen Positron Emission Tomography: An Update. Semin. Nucl. Med. 2025, 55, 371–376. [Google Scholar] [CrossRef]
  102. Amseian, G.; Figueras, M.; Mases, J.; Mengual, L.; Ribal, M.J.; Quintero, K.; Pages, R.; Ingelmo-Torres, M.; Roldan, F.L.; Caratini, R.; et al. cfDNA fragmentation patterns correlate with tumor burden measured via PSMA PET/CT volumetric parameters in patients with biochemical recurrence of prostate cancer. EJNMMI Res. 2024, 14, 124. [Google Scholar] [CrossRef] [PubMed]
  103. Ren, Y.; Liu, T.; Liu, C.; Guo, X.; Wang, F.; Zhu, H.; Yang, Z. An Albumin-Binding PSMA Ligand with Higher Tumor Accumulation for PET Imaging of Prostate Cancer. Pharmaceuticals 2022, 15, 513. [Google Scholar] [CrossRef] [PubMed]
  104. Sallam, M.; Nguyen, N.T.; Sainsbury, F.; Kimizuka, N.; Muyldermans, S.; Benešová-Schäfer, M. PSMA-targeted radiotheranostics in modern nuclear medicine: Then, now, and what of the future? Theranostics 2024, 14, 3043–3079. [Google Scholar] [CrossRef] [PubMed]
  105. Pan, J.; Zhang, T.; Chen, S.; Bu, T.; Zhao, J.; Ni, X.; Shi, B.; Gan, H.; Wei, Y.; Wang, Q.; et al. Nomogram to predict the presence of PSMA-negative but FDG-positive lesion in castration-resistant prostate cancer: A multicenter cohort study. Ther. Adv. Med. Oncol. 2024, 16, 17588359231220506. [Google Scholar] [CrossRef]
  106. Aide, N.; Lasnon, C.; Veit-Haibach, P.; Sera, T.; Sattler, B.; Boellaard, R. EANM/EARL harmonization strategies in PET quantification: From daily practice to multicentre oncological studies. Eur. J. Nucl. Med. Mol. Imaging 2017, 44 (Suppl. 1), 17–31. [Google Scholar] [CrossRef]
  107. Sheller, M.J.; Edwards, B.; Reina, G.A.; Martin, J.; Pati, S.; Kotrotsou, A.; Milchenko, M.; Xu, W.; Marcus, D.; Colen, R.R.; et al. Federated learning in medicine: Facilitating multi-institutional collaborations without sharing patient data. Sci. Rep. 2020, 10, 12598. [Google Scholar] [CrossRef]
  108. Collins, G.S.; Moons, K.G.M.; Dhiman, P.; Riley, R.D.; Beam, A.L.; Van Calster, B.; Ghassemi, M.; Liu, X.; Reitsma, J.B.; van Smeden, M.; et al. TRIPOD+AI statement: Updated guidance for reporting clinical prediction models that use regression or machine learning methods. BMJ 2024, 385, e078378. [Google Scholar] [CrossRef]
  109. Sandhu, K.; Lim, S.; Lawrentschuk, N.; Murphy, D.; Perera, M. Utilisation of PSMA-PET in Australia following government subsidisation: Trends in primary staging and biochemical recurrence. Prostate Int. 2025, in press. [CrossRef]
  110. Sandhu, K.; Perera, M.; Lawrentschuk, N. Lutetium-177 PSMA—The new snake oil? An Australian experience. BJU Int. 2025, 136, 212–213. [Google Scholar] [CrossRef]
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.

Article Metrics

Citations

Article Access Statistics

Multiple requests from the same IP address are counted as one view.