1
|
Abida W, Beltran H, Raychaudhuri R. State of the Art: Personalizing Treatment for Patients With Metastatic Castration-Resistant Prostate Cancer. Am Soc Clin Oncol Educ Book 2025; 45:e473636. [PMID: 40112242 DOI: 10.1200/edbk-25-473636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Until recently, the treatment of metastatic castration-resistant prostate cancer (mCRPC) relied exclusively on hormonal therapies and taxane chemotherapy. The advent of modern molecular profiling methods applied in the clinic, namely, next-generation sequencing and advanced positron emission tomography (PET) imaging, has allowed for the development of biomarker-driven therapeutics including anti-PD-L1 therapy for microsatellite instability-high or tumor mutation burden-high disease, poly(ADP-ribose) polymerase (PARP) inhibitors for patients with DNA damage repair mutations, and lutetium 177 vipivotide tetraxetan (177Lu-PSMA-617) for patients with prostate-specific membrane antigen (PSMA) PET-avid disease. While these targeted therapies have improved outcomes, there is an opportunity to refine biomarkers to optimize patient selection, understand resistance, and develop novel combination strategies. In addition, studies in the laboratory and in patient-derived samples have shown that a subset of mCRPC tumors lose expression of common prostate cancer markers such as prostate-specific antigen and PSMA because of lineage plasticity and the development of non-androgen receptor (AR)-driven disease. Non-AR-driven prostate cancer has been associated with aggressive behavior and poor prognosis, including in some cases histologic transformation to a poorly differentiated neuroendocrine prostate cancer (NEPC). The clinical management of NEPC typically follows the treatment paradigm for small cell lung cancer and increasingly relies on genomic and phenotypic characterization of disease, including loss of tumor suppressors and expression of cell surface markers such as DLL3. Therefore, both genomic subtyping and phenotypic subtyping are important to consider and can guide the clinical management of patients with advanced prostate cancer.
Collapse
Affiliation(s)
- Wassim Abida
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Ruben Raychaudhuri
- University of Washington and the Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
2
|
Kostos L, Buteau JP, Kong G, Tran B, Haskali MB, Fahey M, Crumbaker M, Emmett L, Hofman MS, Azad AA. Clinical Trial Protocol for LuCAB: A Phase I-II Trial Evaluating Cabazitaxel in Combination with [ 177Lu]Lu-PSMA-617 in Patients with Metastatic Castration-Resistant Prostate Cancer. J Nucl Med 2025; 66:572-578. [PMID: 39978808 DOI: 10.2967/jnumed.124.269252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/06/2025] [Indexed: 02/22/2025] Open
Abstract
[177Lu]Lu-prostate-specific membrane antigen (PSMA)-617 is a standard treatment for patients with metastatic castration-resistant prostate cancer (mCRPC) previously treated with docetaxel and an androgen receptor pathway inhibitor. However, for many, responses are short and progression is inevitable. Contributing factors to treatment resistance include molecular heterogeneity with variable PSMA expression, micrometastases that may not absorb sufficient radiation from 177Lu to result in cell death, and inherent or acquired radioresistance because of genomic alterations or the tumor microenvironment. Cabazitaxel is a radiosensitizer and may treat PSMA-negative disease that would otherwise evade targeting by [177Lu]Lu-PSMA-617. We hypothesize that the combination of [177Lu]Lu-PSMA-617 and cabazitaxel will be synergistic with an acceptable safety profile. Methods: This investigator-initiated phase I-II trial aims to evaluate the safety, tolerability, and preliminary efficacy of cabazitaxel and [177Lu]Lu-PSMA-617 in combination. Up to 38 patients with mCRPC will receive up to 6 doses of [177Lu]Lu-PSMA-617 administered intravenously every 6 wk at a fixed dose of 7.4 GBq. Cabazitaxel will be administered concurrently (dose range, 12.5-20 mg/m2) on day 2 and day 23 of each 6-wk cycle, with dose escalation determined using a traditional 3 + 3 design to establish the maximum tolerated or administered dose. Key eligibility criteria include a diagnosis of progressive mCRPC with PSMA-positive disease on PSMA PET/CT (SUVmax ≥ 15) and no sites of discordance on [18F]F-FDG PET/CT. Patients must have received prior docetaxel and an androgen receptor pathway inhibitor, have adequate bone marrow and organ function, and have an Eastern Cooperative Oncology Group performance status of 0 or 1. The primary objective is to assess for dose-limiting toxicities and determine the recommended phase II dose of cabazitaxel and [177Lu]Lu-PSMA-617 in combination. Secondary objectives include further safety evaluation through the measurement of the frequency and severity of adverse events, assessment of efficacy, and evaluation of changes in pain and health-related quality of life over the first 12 mo from treatment commencement. Plasma will be collected at baseline, during treatment, and at disease progression for circulating tumor DNA analysis, which will be correlated with clinical outcomes to identify potential biomarkers of treatment response or resistance. Conclusion: Enrollment commenced in August 2022, with anticipated completion in 2025.
Collapse
Affiliation(s)
- Louise Kostos
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - James P Buteau
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Grace Kong
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ben Tran
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Mohammad B Haskali
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Radiopharmaceutical Production and Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael Fahey
- Centre for Biostatistics and Clinical Trials, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Megan Crumbaker
- Department of Medical Oncology, Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Louise Emmett
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia; and
- St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Michael S Hofman
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Arun A Azad
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Stangl-Kremser J, Ricaurte-Fajardo A, Huicochea Castellanos S, Martinez-Fundichely A, Sun M, Osborne JR, Nauseef JT, Tagawa ST, Bander NH. Baseline Imaging Derived Factors of Response Following [225Ac]Ac-J591 Therapy in Metastatic Castration-Resistant Prostate Cancer: A Lesion Level Analysis. Prostate 2025; 85:502-509. [PMID: 39853781 DOI: 10.1002/pros.24853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/13/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025]
Abstract
PURPOSE Actinium-225 labeled prostate-specific membrane antigen (PSMA) targeted radionuclide therapy has emerged as a potential treatment option in the management of men with metastatic castrate-resistant prostate cancer (mCRPC). This study investigated molecular imaging-derived parameters and compared imaging response of lesions categorized by tumor site. METHODS Men with mCRPC treated with [225Ac]Ac-J591 from 2017 to 2022 at our center on two prospective trials (NCT03276572 and NCT04506567) with pre- and post-treatment [68Ga]Ga-PSMA-11 Positron Emission Tomography (PET) imaging studies available were included. SUVpeak of the 3 most- and 3 least-avid lesions of the tumor sites were manually assessed. The median change of the SUVpeak from pre- to post-treatment per tumor site was evaluated using the paired Wilcox test. An objective response (OR) in the follow-up image was defined as complete or partial response using PET Response Criteria in Solid Tumors (PERCIST) 1.0. RESULTS A total of 46 cases met the criteria for image review; most of them (n = 25, 54.3%) had more than one tumor site category. In total, 445 PSMA PET-positive lesions were assessed: 220 osseous, 163 nodal, 41 visceral, and 21 prostatic lesions. After treatment with [225Ac]Ac-J591, absolute SUVpeak values per tumor site declined significantly (p < 0.05) except for prostatic lesions (p = 1). The PERCIST-OR rate for osseous, nodal, visceral, and prostatic lesions was 53%, 28%, 56%, and 38%, respectively. CONCLUSION [225Ac]Ac-J591 is an active treatment in men with mCRPC. Tumor distribution patterns may influence treatment response and potentially prognosis. Our findings warrant further validation in a larger cohort but may be considered in treatment planning and trial design.
Collapse
Affiliation(s)
| | - Andres Ricaurte-Fajardo
- Division of Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, New York City, New York, USA
| | - Sandra Huicochea Castellanos
- Division of Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, New York City, New York, USA
| | - Alexander Martinez-Fundichely
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York City, New York, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York City, New York, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York City, New York, USA
| | - Michael Sun
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York City, New York, USA
| | - Joseph R Osborne
- Division of Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, New York City, New York, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York City, New York, USA
| | - Jones T Nauseef
- Meyer Cancer Center, Weill Cornell Medicine, New York City, New York, USA
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York City, New York, USA
| | - Scott T Tagawa
- Department of Urology, Weill Cornell Medicine, New York City, New York, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York City, New York, USA
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York City, New York, USA
| | - Neil H Bander
- Department of Urology, Weill Cornell Medicine, New York City, New York, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York City, New York, USA
| |
Collapse
|
4
|
von Eyben FE, Virgolini I, Baum R. Review on the Increasing Role for PSMA-Based Radioligand Therapy in Prostate Cancer. Cancers (Basel) 2024; 16:2520. [PMID: 39061160 PMCID: PMC11274522 DOI: 10.3390/cancers16142520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
In 2021, two randomized controlled trials (RCTs), TheraP and VISION, demonstrated that 177Lu-PSMA-617 as monotherapy was more effective for the decline of PSA than the comparator third-line treatments. METHODS Our review summarizes new RCTs that add to the use of radioligand therapy (RLT) for patients with high-risk prostate cancer (PCa). RESULTS Four past and present RCTs included 1081 patients. An RCT, ENZA-p, studied first-line treatment of patients with metastatic castration-resistant PCa (mCRPC). A combination of enzalutamide (ENZA) and 177Lu-PSMA-617 gave longer progression-free survival than ENZA as monotherapy. Other RCTs of patients with mCRPC, including the PSMAfore, and SPLASH trials, showed 177Lu-PSMA-617 as second-line treatment gave better progression-free survival than androgen receptor pathway inhibitors (combined p value < 6.9 × 10-6). CONCLUSIONS Patients with PCa gain if they are given PSMA-RLT early in the treatment of PCa and as part of combination therapies.
Collapse
Affiliation(s)
| | - Irene Virgolini
- Department of Nuclear Medicine, University Hospital Innsbruck, 6020 Innsbruck, Austria
| | - Richard Baum
- DKD Helios Clinic, 65 191 Frankfurth-Wiesbaden, Germany
| |
Collapse
|
5
|
Hindié E, Larouze A, Alcocer-Ávila M, Morgat C, Champion C. Palladium-103 ( 103Pd/ 103mRh), a promising Auger-electron emitter for targeted radionuclide therapy of disseminated tumor cells - absorbed doses in single cells and clusters, with comparison to 177Lu and 161Tb. Theranostics 2024; 14:4318-4330. [PMID: 39113794 PMCID: PMC11303077 DOI: 10.7150/thno.95436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
Early use of targeted radionuclide therapy (TRT) to eradicate disseminated tumor cells (DTCs) might offer cure. Selection of appropriate radionuclides is required. This work highlights the potential of 103Pd (T1/2 = 16.991 d) which decays to 103mRh (T1/2 = 56.12 min) then to stable 103Rh with emission of Auger and conversion electrons. Methods: The Monte Carlo track structure code CELLDOSE was used to assess absorbed doses in single cells (14-μm diameter; 10-μm nucleus) and clusters of 19 cells. The radionuclide was distributed on the cell surface, within the cytoplasm, or in the nucleus. Absorbed doses from 103Pd, 177Lu and 161Tb were compared after energy normalization. The impact of non-uniform cell targeting, and the potential benefit from dual-targeting was investigated. Additional results related to 103mRh, if used directly, are provided. Results: In the single cell, and depending on radionuclide distribution, 103Pd delivered 7- to 10-fold higher nuclear absorbed dose and 9- to 25-fold higher membrane dose than 177Lu. In the 19-cell clusters, 103Pd absorbed doses also largely exceeded 177Lu. In both situations, 161Tb stood in-between 103Pd and 177Lu. Non-uniform targeting, considering four unlabeled cells within the cluster, resulted in moderate-to-severe dose heterogeneity. For example, with intranuclear 103Pd, unlabeled cells received only 14% of the expected nuclear dose. Targeting with two 103Pd-labeled radiopharmaceuticals minimized dose heterogeneity. Conclusion: 103Pd, a next-generation Auger emitter, can deliver substantially higher absorbed doses than 177Lu to single tumor cells and cell clusters. This may open new horizons for the use of TRT in adjuvant or neoadjuvant settings, or for targeting minimal residual disease.
Collapse
Affiliation(s)
- Elif Hindié
- Service de Médecine Nucléaire, CHU de Bordeaux, Université de Bordeaux, UMR CNRS 5287, INCIA, F-33400, Talence, France
- Institut Universitaire de France, 1 rue Descartes, 75231 Paris cedex 05, France
| | - Alexandre Larouze
- Université de Bordeaux-CNRS-CEA, Centre Lasers Intenses et Applications, UMR 5107, 33405 Talence, France
| | - Mario Alcocer-Ávila
- Université de Bordeaux-CNRS-CEA, Centre Lasers Intenses et Applications, UMR 5107, 33405 Talence, France
| | - Clément Morgat
- Service de Médecine Nucléaire, CHU de Bordeaux, Université de Bordeaux, UMR CNRS 5287, INCIA, F-33400, Talence, France
| | - Christophe Champion
- Université de Bordeaux-CNRS-CEA, Centre Lasers Intenses et Applications, UMR 5107, 33405 Talence, France
| |
Collapse
|
6
|
Jewell K, Hofman MS, Ong JSL, Levy S. Emerging Theranostics for Prostate Cancer and a Model of Prostate-specific Membrane Antigen Therapy. Radiology 2024; 311:e231703. [PMID: 38563674 DOI: 10.1148/radiol.231703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
There is increasing demand worldwide to develop diagnostic and therapeutic (theranostic) markers for prostate cancer. One target of interest is prostate-specific membrane antigen (PSMA), a protein which is overexpressed in prostate cancer cells. Over the past decade, a growing body of literature has demonstrated that radiolabeled ligands that target PSMA show favorable clinical response and survival outcomes in patients with advanced prostate cancer. This focused review provides background to the development of PSMA as a target, an overview of key studies informing our current approach to radioligand-based imaging and therapy for prostate cancer, and a model for real-world implementation of PSMA theranostics based on an Australian experience.
Collapse
Affiliation(s)
- Kerry Jewell
- From the Department of Molecular Imaging and Therapeutic Nuclear Medicine and Department of Oncology, Prostate Theranostics and Imaging Centre of Excellence (ProsTIC); Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia (K.J., M.S.H., S.L.); University of Melbourne, Melbourne, Australia (M.S.H.); and Department of Nuclear Medicine, Fiona Stanley Hospital, Murdoch, Australia (J.S.L.O.)
| | - Michael S Hofman
- From the Department of Molecular Imaging and Therapeutic Nuclear Medicine and Department of Oncology, Prostate Theranostics and Imaging Centre of Excellence (ProsTIC); Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia (K.J., M.S.H., S.L.); University of Melbourne, Melbourne, Australia (M.S.H.); and Department of Nuclear Medicine, Fiona Stanley Hospital, Murdoch, Australia (J.S.L.O.)
| | - Jeremy S L Ong
- From the Department of Molecular Imaging and Therapeutic Nuclear Medicine and Department of Oncology, Prostate Theranostics and Imaging Centre of Excellence (ProsTIC); Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia (K.J., M.S.H., S.L.); University of Melbourne, Melbourne, Australia (M.S.H.); and Department of Nuclear Medicine, Fiona Stanley Hospital, Murdoch, Australia (J.S.L.O.)
| | - Sidney Levy
- From the Department of Molecular Imaging and Therapeutic Nuclear Medicine and Department of Oncology, Prostate Theranostics and Imaging Centre of Excellence (ProsTIC); Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia (K.J., M.S.H., S.L.); University of Melbourne, Melbourne, Australia (M.S.H.); and Department of Nuclear Medicine, Fiona Stanley Hospital, Murdoch, Australia (J.S.L.O.)
| |
Collapse
|
7
|
Belge Bilgin G, Bilgin C, Burkett BJ, Orme JJ, Childs DS, Thorpe MP, Halfdanarson TR, Johnson GB, Kendi AT, Sartor O. Theranostics and artificial intelligence: new frontiers in personalized medicine. Theranostics 2024; 14:2367-2378. [PMID: 38646652 PMCID: PMC11024845 DOI: 10.7150/thno.94788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/17/2024] [Indexed: 04/23/2024] Open
Abstract
The field of theranostics is rapidly advancing, driven by the goals of enhancing patient care. Recent breakthroughs in artificial intelligence (AI) and its innovative theranostic applications have marked a critical step forward in nuclear medicine, leading to a significant paradigm shift in precision oncology. For instance, AI-assisted tumor characterization, including automated image interpretation, tumor segmentation, feature identification, and prediction of high-risk lesions, improves diagnostic processes, offering a precise and detailed evaluation. With a comprehensive assessment tailored to an individual's unique clinical profile, AI algorithms promise to enhance patient risk classification, thereby benefiting the alignment of patient needs with the most appropriate treatment plans. By uncovering potential factors unseeable to the human eye, such as intrinsic variations in tumor radiosensitivity or molecular profile, AI software has the potential to revolutionize the prediction of response heterogeneity. For accurate and efficient dosimetry calculations, AI technology offers significant advantages by providing customized phantoms and streamlining complex mathematical algorithms, making personalized dosimetry feasible and accessible in busy clinical settings. AI tools have the potential to be leveraged to predict and mitigate treatment-related adverse events, allowing early interventions. Additionally, generative AI can be utilized to find new targets for developing novel radiopharmaceuticals and facilitate drug discovery. However, while there is immense potential and notable interest in the role of AI in theranostics, these technologies do not lack limitations and challenges. There remains still much to be explored and understood. In this study, we investigate the current applications of AI in theranostics and seek to broaden the horizons for future research and innovation.
Collapse
Affiliation(s)
| | - Cem Bilgin
- Department of Radiology, Mayo Clinic Rochester, MN, USA
| | | | - Jacob J. Orme
- Department of Oncology, Mayo Clinic Rochester, MN, USA
| | | | | | | | - Geoffrey B Johnson
- Department of Radiology, Mayo Clinic Rochester, MN, USA
- Department of Immunology, Mayo Clinic Rochester, MN, USA
| | | | - Oliver Sartor
- Department of Radiology, Mayo Clinic Rochester, MN, USA
- Department of Oncology, Mayo Clinic Rochester, MN, USA
- Department of Urology, Mayo Clinic Rochester, MN, USA
| |
Collapse
|
8
|
Corpetti M, Müller C, Beltran H, de Bono J, Theurillat JP. Prostate-Specific Membrane Antigen-Targeted Therapies for Prostate Cancer: Towards Improving Therapeutic Outcomes. Eur Urol 2024; 85:193-204. [PMID: 38104015 DOI: 10.1016/j.eururo.2023.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
CONTEXT Prostate-specific membrane antigen (PSMA) is a transmembrane glycoprotein overexpressed in most prostate cancers and exploited as a target for PSMA-targeted therapies. Different approaches to target PSMA-expressing cancer cells have been developed, showing promising results in clinical trials. OBJECTIVE To discuss the regulation of PSMA expression and the main PSMA-targeted therapeutic concepts illustrating their clinical development and rationalizing combination approaches with examples. EVIDENCE ACQUISITION We performed a detailed literature search using PubMed and reviewed the American Society of Clinical Oncology and European Society of Medical Oncology annual meeting abstracts up to September 2023. EVIDENCE SYNTHESIS We present an overarching description of the different strategies to target PSMA. The outcomes of PSMA-targeted therapies strongly rely on surface-bound PSMA expression. However, PSMA heterogeneity at different levels (interpatient and inter/intratumoral) limits the efficacy of PSMA-targeted therapies. We highlight the molecular mechanisms governing PSMA regulation, the understanding of which is crucial to designing therapeutic strategies aimed at upregulating PSMA expression. Thus far, homeobox B13 (HOXB13) and androgen receptor (AR) have emerged as critical transcription factors positively and negatively regulating PSMA expression, respectively. Furthermore, epigenetic regulation of PSMA has been also reported recently. In addition, many established therapeutic approaches harbor the potential to upregulate PSMA levels as well as potentiate DNA damage mediated by current radioligands. CONCLUSIONS PSMA-targeted therapies are rapidly advancing, but their efficacy is strongly limited by the heterogeneous expression of the target. A thorough comprehension of how PSMA is regulated will help improve the outcomes through increasing PSMA expression and will provide the basis for synergistic combination therapies. PATIENT SUMMARY Prostate-specific membrane antigen (PSMA) is overexpressed in most prostate cancers. PSMA-targeted therapies have shown promising results, but the heterogeneous expression of PSMA limits their efficacy. We propose to better elucidate the regulation of PSMA expression to increase the levels of the target and improve the therapeutic outcomes.
Collapse
Affiliation(s)
- Matteo Corpetti
- Institute of Oncology Research, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Cristina Müller
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland; Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Johann de Bono
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK
| | - Jean-Philippe Theurillat
- Institute of Oncology Research, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland.
| |
Collapse
|
9
|
Mader N, Schoeler C, Pezeshkpour N, Klimek K, Groener D, Happel C, Tselis N, Mandel P, Grünwald F, Sabet A. Intermittent Radioligand Therapy with 177Lu-PSMA-617 for Oligometastatic Castration-Resistant Prostate Cancer. Cancers (Basel) 2023; 15:4605. [PMID: 37760574 PMCID: PMC10527374 DOI: 10.3390/cancers15184605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
177Lu-PSMA-617 radioligand therapy (177Lu-PSMA-RLT) in patients with metastatic castration-resistant prostate cancer (mCRPC) currently consists of 4-6 cycles of 6.0-7.4 GBq of 177Lu-PSMA-617 each every 6-8 weeks. While safety and efficacy could be demonstrated in larger prospective trials irrespective of the tumor burden at 177Lu-PSMA RLT initiation, increased renal absorbed doses due to a reduced tumor sink effect in early responding, oligometastatic mCRPC patients pose difficulties. Response-adapted, dose distributing, intermittent treatment with up to six cycles has not been routinely performed, due to concerns about the potential loss of disease control. Treatment was discontinued in 19 early-responding patients with oligometastatic tumor burden after two (IQR 2-3) cycles of 177Lu-PSMA-RLT and 6.5 ± 0.7 GBq per cycle and resumed upon 68Ga-PSMA-11-PET/CT-based progression (according to the PCWG3 criteria). Subsequent treatment breaks were imposed if a PSMA-based imaging response could be achieved. A total of five (IQR 3-6) cycles reaching a cumulative activity of 32 ± 11 GBq were applied. A routine blood work-up including blood counts and liver and renal function was measured throughout the 177Lu-PSMA-RLT and follow-up to grade toxicity according to CTCAE v5.0 criteria. Survival outcome was calculated based on the Kaplan-Meier method. In total, treatment-free periods of 9 (IQR 6-17) cumulative months and the application of 177Lu-PSMA-RLT cycles over 16 (IQR 9-22) months could be achieved. Fifteen (84%) patients responded to subsequent cycles after the first treatment break and in 7/19 (37%) patients, intermittent 177Lu-PSMA-RLT consisted of ≥2 treatment breaks. The median PFS was 27 months (95% CI: 23-31) and overall survival was 45 months (95% CI: 28-62). No grade ≥3 hematological or renal toxicities could be observed during the 45 ± 21 months of follow-up. The cumulative mean renal absorbed dose was 16.7 ± 8.3 Gy and 0.53 ± 0.21 Gy/GBq. Intermittent radioligand therapy with 177Lu-PSMA-617 is feasible in early-responding patients with oligometastatic disease. A late onset of progression after subsequent cycles and the absence of significant toxicity warrants further investigation of the concept of intermittent treatment in selected patients.
Collapse
Affiliation(s)
- Nicolai Mader
- Department of Nuclear Medicine, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (N.M.); (C.S.); (N.P.); (K.K.); (D.G.); (C.H.); (F.G.)
| | - Christina Schoeler
- Department of Nuclear Medicine, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (N.M.); (C.S.); (N.P.); (K.K.); (D.G.); (C.H.); (F.G.)
| | - Niloufar Pezeshkpour
- Department of Nuclear Medicine, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (N.M.); (C.S.); (N.P.); (K.K.); (D.G.); (C.H.); (F.G.)
| | - Konrad Klimek
- Department of Nuclear Medicine, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (N.M.); (C.S.); (N.P.); (K.K.); (D.G.); (C.H.); (F.G.)
| | - Daniel Groener
- Department of Nuclear Medicine, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (N.M.); (C.S.); (N.P.); (K.K.); (D.G.); (C.H.); (F.G.)
| | - Christian Happel
- Department of Nuclear Medicine, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (N.M.); (C.S.); (N.P.); (K.K.); (D.G.); (C.H.); (F.G.)
| | - Nikolaos Tselis
- Department of Radiation Oncology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
| | - Philipp Mandel
- Department of Urology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
| | - Frank Grünwald
- Department of Nuclear Medicine, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (N.M.); (C.S.); (N.P.); (K.K.); (D.G.); (C.H.); (F.G.)
| | - Amir Sabet
- Department of Nuclear Medicine, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (N.M.); (C.S.); (N.P.); (K.K.); (D.G.); (C.H.); (F.G.)
| |
Collapse
|
10
|
Gutsche R, Gülmüs G, Mottaghy FM, Gärtner F, Essler M, von Mallek D, Ahmadzadehfar H, Lohmann P, Heinzel A. Multicentric 68Ga-PSMA PET radiomics for treatment response assessment of 177Lu-PSMA-617 radioligand therapy in patients with metastatic castration-resistant prostate cancer. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1234853. [PMID: 39355016 PMCID: PMC11440964 DOI: 10.3389/fnume.2023.1234853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/29/2023] [Indexed: 10/03/2024]
Abstract
Objective The treatment with 177Lutetium PSMA (177Lu-PSMA) in patients with metastatic castration-resistant prostate cancer (mCRPC) has recently been approved by the FDA and EMA. Since treatment success is highly variable between patients, the prediction of treatment response and identification of short- and long-term survivors after treatment could help tailor mCRPC diagnosis and treatment accordingly. The aim of this study is to investigate the value of radiomic parameters extracted from pretreatment 68Ga-PSMA PET images for the prediction of treatment response. Methods A total of 45 mCRPC patients treated with 177Lu-PSMA-617 from two university hospital centers were retrospectively reviewed for this study. Radiomic features were extracted from the volumetric segmentations of metastases in the bone. A random forest model was trained and validated to predict treatment response based on age and conventionally used PET parameters, radiomic features and combinations thereof. Further, overall survival was predicted by using the identified radiomic signature and compared to a Cox regression model based on age and PET parameters. Results The machine learning model based on a combined radiomic signature of three features and patient age achieved an AUC of 0.82 in 5-fold cross-validation and outperformed models based on age and PET parameters or radiomic features (AUC, 0.75 and 0.76, respectively). A Cox regression model based on this radiomic signature showed the best performance to predict overall survival (C-index, 0.67). Conclusion Our results demonstrate that a machine learning model to predict response to 177Lu-PSMA treatment based on a combination of radiomics and patient age outperforms a model based on age and PET parameters. Moreover, the identified radiomic signature based on pretreatment 68Ga-PSMA PET images might be able to identify patients with an improved outcome and serve as a supportive tool in clinical decision making.
Collapse
Affiliation(s)
- Robin Gutsche
- Institute of Neuroscience and Medicine (INM-4), Forschungszentrum Juelich, Juelich, Germany
- RWTH Aachen University, Aachen, Germany
| | | | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Florian Gärtner
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | - Markus Essler
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | - Dirk von Mallek
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | | | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-4), Forschungszentrum Juelich, Juelich, Germany
| | - Alexander Heinzel
- Institute of Neuroscience and Medicine (INM-4), Forschungszentrum Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
- Department of Nuclear Medicine, University Hospital Halle (Saale), Halle (Saale), Germany
| |
Collapse
|