1
|
Fallah J, Mulkey F, Fiero MH, Gittleman H, Song C, Puthiamadathil J, Amatya A, Agrawal S, Vellanki P, Suzman DL, Singh H, Amiri-Kordestani L, Mishra-Kalyani P, Pazdur R, Kluetz PG. Equipoise Lost? Trial Conduct Challenges in an Era of Breakthrough Therapies. J Clin Oncol 2024; 42:3783-3787. [PMID: 39288354 DOI: 10.1200/jco-24-01200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
FDA Oncology Center's @Falleh_Fallah and colleagues discuss loss of equipoise and other trial conduct challenges in an era of breakthrough therapies - via @JCO_ASCO.
Collapse
Affiliation(s)
- Jaleh Fallah
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
| | - Flora Mulkey
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
| | - Mallorie H Fiero
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
| | - Haley Gittleman
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
| | - Chi Song
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
| | - Jeevan Puthiamadathil
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
| | - Anup Amatya
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
| | - Sundeep Agrawal
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
| | - Paz Vellanki
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
| | - Daniel L Suzman
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
| | - Harpreet Singh
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
- Oncology Center of Excellence (OCE), US Food and Drug Administration, Silver Spring, MD
| | - Laleh Amiri-Kordestani
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
- Oncology Center of Excellence (OCE), US Food and Drug Administration, Silver Spring, MD
| | - Pallavi Mishra-Kalyani
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
| | - Richard Pazdur
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
- Oncology Center of Excellence (OCE), US Food and Drug Administration, Silver Spring, MD
| | - Paul G Kluetz
- Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD
- Oncology Center of Excellence (OCE), US Food and Drug Administration, Silver Spring, MD
| |
Collapse
|
2
|
Kramer CS, Zhang J, Baum RP. Extraordinary therapeutic effect of PSMA radioligand therapy in treatment-refractory progressive metastatic prostate cancer with the transketolase inhibitor benfo-oxythiamine as a radiosensitizer-A case report. Front Med (Lausanne) 2024; 11:1462234. [PMID: 39444815 PMCID: PMC11496175 DOI: 10.3389/fmed.2024.1462234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024] Open
Abstract
Herein we report, for the first time, the therapeutic response of a prostate cancer patient with the thiamine antagonist benfo-oxythiamine (B-OT) added to prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (PRLT). The patient was initially diagnosed as pT3b pN0 (0/7) M0 L0 V0 R0 G3, Gleason score 5 + 5 = 10, with an initial prostate-specific antigen (PSA) level of 4.05 ng/ml. Shortly after radical prostatectomy, 68Ga-PSMA positron emission tomography/computed tomography (PET/CT) revealed PSMA-positive lymph node metastases. Despite treatment with androgen deprivation therapy, external beam radiation therapy, palliative chemotherapy, and five cycles of PRLT (177Lu-PRLT or TANDEM-PRLT, respectively), the patient experienced progression in PSA levels as well as in PSMA PET/CT. Due to the intense PSMA expression, 177Lu-PRLT with 177Lu-PSMA-I&T was resumed for another 4 cycles (cycles 6th to 9th) and the patient was additionally treated with the thiamine antagonist benfo-oxythiamine. It was hypothesized that B-OT acts as a radiosensitizer by interfering with the repair of damaged DNA. B-OT-PRLT was well-tolerated and no substantial changes in laboratory results were observed. Additionally, the patient reported significant improvement in clinical symptoms. Post-treatment 177Lu-PSMA single-photon computed tomography (SPECT)/CT after the 7th cycle (and after 2 cycles of B-OT-PRLT) revealed regression of metastases compared to the post-treatment SPECT/CT after the 6th cycle. Before the 8th cycle, PSMA PET/CT showed a mixed response following prior uncontrollable cancer progression. Moreover, the PSA level showed a significant decline after one cycle of B-OT-PRLT. Although the patient had experienced massive progression before the first cycle of B-OT-PRLT, he survived for an additional 12 months. This case supports the hypothesis that B-OT-PRLT could overcome radiation resistance in prostate cancer patients who do not initially respond to 177Lu- or 225Ac-PRLT.
Collapse
Affiliation(s)
- Carsten S. Kramer
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, Wiesbaden, Germany
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Clinical Imaging Research Centre, National University of Singapore, Singapore, Singapore
| | - Richard P. Baum
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, Wiesbaden, Germany
| |
Collapse
|
3
|
Galbiati A, Bocci M, Ravazza D, Mock J, Gilardoni E, Neri D, Cazzamalli S. Preclinical Evaluation of 177Lu-OncoFAP-23, a Multivalent FAP-Targeted Radiopharmaceutical Therapeutic for Solid Tumors. J Nucl Med 2024; 65:1604-1610. [PMID: 39266289 DOI: 10.2967/jnumed.124.268200] [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: 06/07/2024] [Accepted: 08/05/2024] [Indexed: 09/14/2024] Open
Abstract
Fibroblast activation protein (FAP) is abundantly expressed in the stroma of most human solid tumors. Clinical-stage radiolabeled FAP ligands are increasingly used as tools for the detection of various cancer lesions. To unleash the full therapeutic potential of FAP-targeting agents, ligands need to remain at the tumor site for several days after administration. We recently described the discovery of OncoFAP, a high-affinity small organic ligand of FAP with a rapid accumulation in tumors and low uptake in healthy tissues in cancer patients. Trimerization of OncoFAP provided a derivative (named TriOncoFAP, or OncoFAP-23) with improved FAP affinity. In this work, we evaluated the tissue biodistribution profile and the therapeutic performance of OncoFAP-23 in tumor-bearing mice. Methods: OncoFAP-23 was radiolabeled with the theranostic radionuclide 177Lu. Preclinical experiments were conducted on mice bearing SK-RC-52.hFAP (BALB/c nude mice) or CT-26.hFAP (BALB/c mice) tumors. 177Lu-OncoFAP and 177Lu-FAP-2286 were included in the biodistribution study as controls. Toxicologic evaluation was performed on Wistar rats and CD1 mice by injecting high doses of OncoFAP-23 or its cold-labeled counterpart, respectively. Results: 177Lu-OncoFAP-23 emerged for its best-in-class biodistribution profile, high and prolonged tumor uptake (i.e., ∼16 percentage injected dose/g at 96 h), and low accumulation in healthy organs, which correlates well with its potent single-agent anticancer activity at low levels of administered radioactivity. Combination treatment with the tumor-targeted interleukin 2 (L19-IL2, a clinical-stage immunocytokine) further expands the therapeutic window of 177Lu-OncoFAP-23 by potentiating its in vivo antitumor activity. Proteomics studies revealed a potent tumor-directed immune response on treatment with the combination. OncoFAP-23 and natLu-OncoFAP-23 exhibited a favorable toxicologic profile, without showing any side effects or signs of toxicity. Conclusion: OncoFAP-23 presents enhanced tumor uptake and tumor retention and low accumulation in healthy organs, findings that correspond to a strongly improved in vivo antitumor efficacy. The data presented in this work support the clinical development of 177Lu-OncoFAP-23 for the treatment of FAP-positive solid tumors.
Collapse
Affiliation(s)
| | - Matilde Bocci
- R&D Department, Philochem AG, Otelfingen, Switzerland
| | | | | | | | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland; and
- Philogen S.p.A., Siena, Italy
| | | |
Collapse
|
4
|
Ma J, Zhang S, Yang N, Shang J, Gao X, Chen J, Wei H, Li Y, Zeng H, Xu H, Wang J, Liang SH, Wang R, Hu K, Wang L. Discovery of a highly specific radiolabeled antibody targeting B-cell maturation antigen: Applications in PET imaging of multiple myeloma. Eur J Nucl Med Mol Imaging 2024:10.1007/s00259-024-06907-3. [PMID: 39259226 DOI: 10.1007/s00259-024-06907-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024]
Abstract
PURPOSE Multiple myeloma (MM) is characterized by the uncontrolled proliferation of monoclonal plasma cells (PC) in the bone marrow (BM). B-cell maturation antigen (BCMA) is predominantly expressed in malignant plasma cells, and associated with the proliferation, survival, and progression of various myeloma cells. Given these important roles, BCMA emerges as an ideal target antigen for MM therapy. However, effective stratification of patients who may benefit from targeted BCMA therapy and real-time monitoring the therapeutic efficacy poses significant clinical challenge. This study aims to develop a BCMA targeted diagnostic modality, and preliminarily explore its potential value in the radio-immunotherapy of MM. EXPERIMENTAL DESIGN Using zirconium-89 (89Zr, t1/2 = 78.4 h) for labeling the BCMA-specific antibody, the BCMA-targeting PET tracer [89Zr]Zr-DFO-BCMAh230430 was prepared. The EC50 values of BCMAh230430 and DFO-BCMAh230430 were determined by ELISA assay. BCMA expression was assessed in four different tumor cell lines (MM.1S, RPMI 8226, BxPC-3, and KYSE520) through Western blot and flow cytometry. In vitro binding affinity was determined by cell uptake studies of [89Zr]Zr-DFO-BCMAh230430 in these tumor cell lines. For in vivo evaluation, PET imaging and ex vivo biodistribution studies were conducted in tumor-bearing mice to evaluate imaging performance and systemic distribution of [89Zr]Zr-DFO-BCMAh230430. Immunochemistry analysis was performed to detect BCMA expression in tumor tissues, confirming the specificity of our probe. Furthermore, we explored the anti-tumor efficacy of Lutetium-177 labeled BCMA antibody, [177Lu]Lu-DTPA-BCMAh230430, in tumor bearing-mice to validate its radioimmunotherapy potential. RESULTS The radiolabeling of [89Zr]Zr-DFO-BCMAh230430 and [177Lu]Lu-DTPA-BCMAh230430 showed satisfactory radiocharacteristics, with a radiochemical purity exceeding 99%. ELISA assay results revealed closely aligned EC50 values for BCMAh230430 and DFO-BCMAh230430, which are 57 pM and 67 pM, respectively. Western blot and flow cytometry analyses confirmed the highest BCMA expression level. Cell uptake data indicated that MM.1S cells had a total cellular uptake (the sum of internalization and surface binding) of 38.3% ± 1.53% for [89Zr]Zr-DFO-BCMAh230430 at 12 h. PET imaging of [89Zr]Zr-DFO-BCMAh230430 displayed radioactive uptake of 7.71 ± 0.67%ID/g in MM.1S tumors and 4.13 ± 1.21%ID/g in KYSE520 tumors at 168 h post-injection (n = 4) (P < 0.05), consistent with ex vivo biodistribution studies. Immunohistochemical analysis of tumor tissues confirmed higher BCMA expression in MM.1S tumors xenograft compared to KYSE520 tumors. Notably, [177Lu]Lu-DTPA-BCMAh230430 showed some anti-tumor efficacy, evidenced by slowed tumor growth. Furthermore, no significant difference in body weight was observed in MM.1S tumor-bearing mice over 14 days of administration with or without [177Lu]Lu-DTPA-BCMAh230430. CONCLUSIONS Our study has successfully validated the essential role of [89Zr]Zr-DFO-BCMAh230430 in non-invasively monitoring BCMA status in MM tumors, showing favorable tumor uptake and specific binding affinity to MM tumors. Furthermore, our research revealed, as a proof-of-concept, the effectiveness of [177Lu]Lu-DTPA-BCMAh230430 in radioimmunotherapy for MM tumors. In conclusion, we present a novel BCMA antibody-based radiotheranostic modality that holds promise for achieving efficient and precise MM diagnostic and therapy.
Collapse
Affiliation(s)
- Jie Ma
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Nianhui Yang
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jingjie Shang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Xin Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jiahui Chen
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yinlong Li
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA
| | - Hui Zeng
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jinghao Wang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research On Chronic Disease, Guangzhou, 510630, China
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
- The Guangzhou Key Laboratory of Basic and Translational Research On Chronic Disease, Guangzhou, 510630, China.
| |
Collapse
|
5
|
Ells Z, Grogan TR, Czernin J, Dahlbom M, Calais J. Dosimetry of [ 177Lu]Lu-PSMA-Targeted Radiopharmaceutical Therapies in Patients with Prostate Cancer: A Comparative Systematic Review and Metaanalysis. J Nucl Med 2024; 65:1264-1271. [PMID: 38960712 PMCID: PMC11294071 DOI: 10.2967/jnumed.124.267452] [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: 01/16/2024] [Accepted: 04/29/2024] [Indexed: 07/05/2024] Open
Abstract
Novel theranostic approaches using radiopharmaceuticals targeting prostate-specific membrane antigen (PSMA) have emerged for treating metastatic castration-resistant prostate cancer. The physical properties and commercial availability of 177Lu make it one of the most used radionuclides for radiopharmaceutical therapy (RPT). In this literature review, we aimed at comparing the dosimetry of the most used [177Lu]Lu-PSMA RPT compounds. Methods: This was a systematic review and metaanalysis of [177Lu]Lu-PSMA RPT (617, I&T, and J591) dosimetry in patients with prostate cancer. Absorbed doses in Gy/GBq for each organ at risk (kidney, parotid and submandibular glands, bone marrow, liver, and lacrimal glands) and for tumor lesions (bone and nonbone lesions) were extracted from included articles. These were used to estimate the pooled average absorbed dose of each agent in Gy/GBq and in Gy/cycle, normalized to the injected activity (per cycle) used in the VISION (7.4 GBq), SPLASH (6.8 GBq), and PROSTACT trials (5.8 GBq). Results: Twenty-nine published articles comprising 535 patients were included in the metaanalysis. The pooled doses (weighted average across studies) of [177Lu]Lu-PSMA-617 and [177Lu]Lu-PSMA-I&T were 4.04 Gy/GBq (17 studies, 297 patients) and 4.70 Gy/GBq (10 studies, 153 patients) for the kidney (P = 0.10), 5.85 Gy/GBq (14 studies, 216 patients) and 2.62 Gy/GBq (5 studies, 86 patients) for the parotids (P < 0.01), 5.15 Gy/GBq (5 studies, 81 patients) and 4.35 Gy/GBq (1 study, 18 patients) for the submandibular glands (P = 0.56), 11.03 Gy/GBq (6 studies, 121 patients) and 19.23 Gy/GBq (3 studies, 53 patients) for the lacrimal glands (P = 0.20), 0.24 Gy/GBq (12 studies, 183 patients) and 0.19 Gy/GBq (4 studies, 68 patients) for the bone marrow (P = 0.31), and 1.11 Gy/GBq (9 studies, 154 patients) and 0.56 Gy/GBq (4 studies, 56 patients) for the liver (P = 0.05), respectively. Average tumor doses tended to be higher for [177Lu]Lu-PSMA-617 than for [177Lu]Lu-PSMA-I&T in soft tissue tumor lesions (4.19 vs. 2.94 Gy/GBq; P = 0.26). Dosimetry data of [177Lu]Lu-J591 were limited to one published study of 35 patients with reported absorbed doses of 1.41, 0.32, and 2.10 Gy/GBq to the kidney, bone marrow, and liver, respectively. Conclusion: In this metaanalysis, there was no significant difference in absorbed dose between [177Lu]Lu-PSMA-I&T and [177Lu]Lu-PSMA-617. There was a possible trend toward a higher kidney dose with [177Lu]Lu-PSMA-I&T and a higher tumor lesion dose with [177Lu]Lu-PSMA-617. It remains unknown whether this finding has any clinical impact. The dosimetry methodologies were strikingly heterogeneous among studies, emphasizing the need for standardization.
Collapse
Affiliation(s)
- Zachary Ells
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California; and
| | - Tristan R Grogan
- Department of Medicine Statistics Core, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California; and
| | - Magnus Dahlbom
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California; and
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California; and
| |
Collapse
|
6
|
Vergnaud L, Dewaraja YK, Giraudet AL, Badel JN, Sarrut D. A review of 177Lu dosimetry workflows: how to reduce the imaging workloads? EJNMMI Phys 2024; 11:65. [PMID: 39023648 DOI: 10.1186/s40658-024-00658-8] [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: 01/25/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024] Open
Abstract
177 Lu radiopharmaceutical therapy is a standardized systemic treatment, with a typical dose of 7.4 GBq per injection, but its response varies from patient to patient. Dosimetry provides the opportunity to personalize treatment, but it requires multiple post-injection images to monitor the radiopharmaceutical's biodistribution over time. This imposes an additional imaging burden on centers with limited resources. This review explores methods to lessen this burden by optimizing acquisition types and minimizing the number and duration of imaging sessions. After summarizing the different steps of dosimetry and providing examples of dosimetric workflows for177 Lu -DOTATATE and177 Lu -PSMA, we examine dosimetric workflows based on a reduced number of acquisitions, or even just one. We provide a non-exhaustive description of simplified methods and their assumptions, as well as their limitations. Next, we detail the specificities of each normal tissue and tumors, before reviewing dose-response relationships in the literature. In conclusion, we will discuss the current limitations of dosimetric workflows and propose avenues for improvement.
Collapse
Affiliation(s)
- Laure Vergnaud
- CREATIS; CNRS UMR 5220; INSERM U 1044, Université de Lyon; INSA-Lyon; Université Lyon 1, Lyon, France.
| | - Yuni K Dewaraja
- Department of Radiology, University of Michigan, Ann Arbor, USA
| | | | - Jean-Noël Badel
- CREATIS; CNRS UMR 5220; INSERM U 1044, Université de Lyon; INSA-Lyon; Université Lyon 1, Lyon, France
- Centre de lutte contre le cancer Léon Bérard, Lyon, France
| | - David Sarrut
- CREATIS; CNRS UMR 5220; INSERM U 1044, Université de Lyon; INSA-Lyon; Université Lyon 1, Lyon, France
- Centre de lutte contre le cancer Léon Bérard, Lyon, France
| |
Collapse
|
7
|
Karimzadeh A, Schatz L, Sauer M, Apostolova I, Buchert R, Klutmann S, Lehnert W. Organ and tumor dosimetry including method simplification for [ 177Lu]Lu-PSMA-I&T for treatment of metastatic castration resistant prostate cancer. EJNMMI Phys 2024; 11:63. [PMID: 39017988 PMCID: PMC11255161 DOI: 10.1186/s40658-024-00668-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Internal dosimetry in individual patients is essential for safe and effective radioligand therapy. Multiple time point imaging for accurate dosimetry is time consuming and hence can be demanding for nuclear medicine departments as well as patients. The objectives of this study were (1) to assess absorbed doses to organs at risk and tumor lesions for [177Lu]Lu-PSMA-I&T using whole body SPECT imaging and (2) to investigate possible simplified dosimetry protocols. METHODS This study included 16 patients each treated with 4 cycles of [177Lu]Lu-PSMA-I&T. They underwent quantitative whole body SPECT/CT imaging (3 bed positions) at four time points (TP) comprising 2 h, 24 h, 48 h and 72-168 h post-injection (p.i.). Full 3D dosimetry (reference method) was performed for all patients and dose cycles for organs at risk (kidneys, parotid glands and submandibular glands) and up to ten tumor lesions per patient (resulting in 90 lesions overall). The simplified dosimetry methods (SM) included (1) generating time activity curves for subsequent cycles using a single TP of imaging applying the kinetics of dose cycle 1, and for organs at risk also (2) simple extrapolation from dose cycle 1 and (3) from both, dose cycle 1 and 2. RESULTS Normalized absorbed doses were 0.71 ± 0.32 mGy/MBq, 0.28 ± 0.12 mGy/MBq and 0.22 ± 0.08 mGy/MBq for kidneys, parotid glands and submandibular glands, respectively. Tumor doses decreased from 3.86 ± 3.38 mGy/MBq in dose cycle 1 to 2.01 ± 2.65 mGy/MBq in dose cycle 4. Compared to the full dosimetry approach the SM 1 using single TP imaging at 48 h p.i. resulted in the most accurate and precise results for the organs at risk in terms of absorbed doses per cycle and total cumulated dose. For tumor lesions better results were achieved using the fourth TP (≥ 72 h p.i.). CONCLUSION Simplification of safety dosimetry protocols is possible for [177Lu]Lu-PSMA-I&T therapy. If tumor dosimetry is of interest a later imaging TP (≥ 72 h p.i.) should be used/added to account for the slower kinetics of tumors compared to organs at risk.
Collapse
Affiliation(s)
- Amir Karimzadeh
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Linus Schatz
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Markus Sauer
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ivayla Apostolova
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ralph Buchert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Susanne Klutmann
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Wencke Lehnert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| |
Collapse
|
8
|
Książek I, Ligęza A, Drzymała F, Borek A, Miszczyk M, Francuz MR, Matsukawa A, Yanagisawa T, Fazekas T, Zapała Ł, Rajwa P. Role of Lutetium Radioligand Therapy in Prostate Cancer. Cancers (Basel) 2024; 16:2433. [PMID: 39001495 PMCID: PMC11240688 DOI: 10.3390/cancers16132433] [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: 06/02/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Theranostics utilize ligands that chelate radionuclides and selectively bind with cancer-specific membrane antigens. In the case of prostate cancer (PCa), the state-of-the-art lutetium-177-PSMA combines the radioactive β-emitter 177Lu with Vipivotide Tetraxetan, a prostate-specific membrane antigen (PSMA)-binding ligand. Several studies have been conducted, and the therapy is not without adverse effects (e.g., xerostomia, nausea, and fatigue); however, few events are reported as severe. The available evidence supports the use of 177Lu-PSMA in selected metastatic castration-resistant prostate cancer patients, and the treatment is considered a standard of care in several clinical scenarios. Emerging research shows promising results in the setting of hormone-sensitive prostate cancer; however, evidence from high-quality controlled trials is still missing. In this review, we discuss the available evidence for the application of 177Lu-PSMA in the management of PCa patients.
Collapse
Affiliation(s)
- Ignacy Książek
- Department of Urology, Medical University of Silesia, 41-800 Zabrze, Poland; (I.K.); (A.B.)
| | - Artur Ligęza
- Department of Urology, Medical University of Silesia, 41-800 Zabrze, Poland; (I.K.); (A.B.)
| | - Franciszek Drzymała
- Department of Urology, Medical University of Silesia, 41-800 Zabrze, Poland; (I.K.); (A.B.)
| | - Adam Borek
- Department of Urology, Medical University of Silesia, 41-800 Zabrze, Poland; (I.K.); (A.B.)
| | - Marcin Miszczyk
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dąbrowa Górnicza, Poland
| | | | - Akihiro Matsukawa
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
- Department of Urology, Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Takafumi Yanagisawa
- Department of Urology, Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Tamás Fazekas
- Department of Urology, Semmelweis University, 1085 Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
| | - Łukasz Zapała
- Clinic of General, Oncological and Functional Urology, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Paweł Rajwa
- Department of Urology, Medical University of Silesia, 41-800 Zabrze, Poland; (I.K.); (A.B.)
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
9
|
Turner JH. Theranostic Innovation by Humane N-of-One Cancer Care in Real-World Patients. Cancer Biother Radiopharm 2024; 39:323-329. [PMID: 38324047 DOI: 10.1089/cbr.2023.0198] [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] [Indexed: 02/08/2024] Open
Abstract
Patients with relapsed or refractory metastatic cancer unresponsive to standard therapies have motivated nuclear physicians to develop innovative radioligands, precisely targeted to tumor molecular receptors, for effective treatment of specific advanced malignancies. Individual practitioners in departments of nuclear medicine across the world have performed first-in-human studies on compassionate patient usage N-of-One protocols. These physician-sponsored studies then evolved into early-phase clinical trials and obtained real-world data to demonstrate real-world evidence of effectiveness in prolonging survival and enhancing quality of life of many so-called "End-Stage" cancer patients. Virtually all the therapeutic radiopharmaceuticals in current clinical oncology have been discovered and developed into effective specific treatments of targetable cancers by individual doctors in the course of their hospital practice. Pharma industry was not involved until many years later when performance of mandated Phase 3 randomized controlled trials became necessary to achieve regulatory agency approval. This article traces the history of several novel theranostic agents developed from compassionate N-of-One studies by hospital physicians over the past 36 years. It acknowledges the collegiality and collaboration of individual nuclear medicine specialists, worldwide, in pioneering effective humane therapy of particular advanced cancers unresponsive to conventional treatments.
Collapse
Affiliation(s)
- J Harvey Turner
- Department of Nuclear Medicine, The University of Western Australia, Fiona Stanley Fremantle Hospitals Group, Murdoch, Australia
| |
Collapse
|
10
|
Al-Ibraheem A, Abdlkadir AS, Sweedat DA, Maus S, Al-Rasheed U, Salah S, Khriesh F, Juaidi D, Abu Dayek D, Istatieh F, Anwar F, Asrawi A, Abufara A, Al-Rwashdeh M, Abu-Hijlih R, Sharaf B, Ghanem R, Abdel-Razeq H, Mansour A. From Despair to Hope: First Arabic Experience of 177Lu-PSMA and 161Tb-PSMA Therapy for Metastatic Castration-Resistant Prostate Cancer. Cancers (Basel) 2024; 16:1974. [PMID: 38893095 PMCID: PMC11171070 DOI: 10.3390/cancers16111974] [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: 04/23/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
The objective of this retrospective study is to assess the effectiveness and safety of two beta-emitting prostate-specific membrane antigen (PSMA) radioligands, [177Lu]Lu and [161Tb]Tb, in heavily treated patients with metastatic castration-resistant prostate cancer (mCRPC). A total of 148 cycles of beta-emitting PSMA radioligand therapy were given to 53 patients at a specialized cancer care center in Amman, Jordan. This treatment was offered following the exhaustion of all prior treatment modalities. Approximately half of the cases (n = 26) demonstrated an initial partial response to PSMA radioligand therapy. Moreover, roughly one-fourth of the patients (n = 13) exhibited a sustained satisfactory biochemical response, which qualified them to receive a total of six PSMA radioligand therapy cycles and maintain continued follow-up for additional treatment cycles. This was reflected by an adequate prostate-specific antigen (PSA) decline and a concomitant partial response evident on [68Ga]Ga-PSMA positron emission tomography/computed tomography imaging. A minority of patients (n= 18; 34%) experienced side effects. Generally, these were low-grade and self-limiting toxicities. This study endorses previous research evidence about PSMA radioligand therapy's safety and efficacy. It also provides the first clinical insight from patients of Arab ethnicity. This should facilitate and promote further evidence, both regionally and internationally.
Collapse
Affiliation(s)
- Akram Al-Ibraheem
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Al-Jubeiha, Amman 11941, Jordan; (A.S.A.); (D.A.S.); (U.A.-R.); (D.J.); (D.A.D.); (F.I.)
- School of Medicine, University of Jordan, Al-Jubeiha, Amman 11942, Jordan
| | - Ahmed Saad Abdlkadir
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Al-Jubeiha, Amman 11941, Jordan; (A.S.A.); (D.A.S.); (U.A.-R.); (D.J.); (D.A.D.); (F.I.)
| | - Deya’ Aldeen Sweedat
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Al-Jubeiha, Amman 11941, Jordan; (A.S.A.); (D.A.S.); (U.A.-R.); (D.J.); (D.A.D.); (F.I.)
| | - Stephan Maus
- Department of Nuclear Medicine, Saarland University Medical Center, D-66421 Homburg, Germany;
| | - Ula Al-Rasheed
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Al-Jubeiha, Amman 11941, Jordan; (A.S.A.); (D.A.S.); (U.A.-R.); (D.J.); (D.A.D.); (F.I.)
| | - Samer Salah
- Department of Medicine, King Hussein Cancer Center (KHCC), Amman 11941, Jordan; (S.S.); (A.A.); (M.A.-R.); (B.S.); (H.A.-R.)
| | - Fadi Khriesh
- Department of Nuclear Medicine, Klinikum Fulda, Pacelliallee 4, 36039 Fulda, Germany;
| | - Diyaa Juaidi
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Al-Jubeiha, Amman 11941, Jordan; (A.S.A.); (D.A.S.); (U.A.-R.); (D.J.); (D.A.D.); (F.I.)
| | - Dina Abu Dayek
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Al-Jubeiha, Amman 11941, Jordan; (A.S.A.); (D.A.S.); (U.A.-R.); (D.J.); (D.A.D.); (F.I.)
| | - Feras Istatieh
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Al-Jubeiha, Amman 11941, Jordan; (A.S.A.); (D.A.S.); (U.A.-R.); (D.J.); (D.A.D.); (F.I.)
| | - Farah Anwar
- Department of Nuclear Medicine, Warith International Cancer Institute, Karbala 56001, Iraq;
| | - Aisha Asrawi
- Department of Nursing, King Hussein Cancer Center (KHCC), Amman 11941, Jordan;
| | - Alaa Abufara
- Department of Medicine, King Hussein Cancer Center (KHCC), Amman 11941, Jordan; (S.S.); (A.A.); (M.A.-R.); (B.S.); (H.A.-R.)
| | - Mohammad Al-Rwashdeh
- Department of Medicine, King Hussein Cancer Center (KHCC), Amman 11941, Jordan; (S.S.); (A.A.); (M.A.-R.); (B.S.); (H.A.-R.)
| | - Ramiz Abu-Hijlih
- Department of Radiation Oncology, King Hussein Cancer Center (KHCC), Amman 11941, Jordan;
| | - Baha’ Sharaf
- Department of Medicine, King Hussein Cancer Center (KHCC), Amman 11941, Jordan; (S.S.); (A.A.); (M.A.-R.); (B.S.); (H.A.-R.)
| | - Rami Ghanem
- Department of Surgery, King Hussein Cancer Center (KHCC), Amman 11941, Jordan;
| | - Hikmat Abdel-Razeq
- Department of Medicine, King Hussein Cancer Center (KHCC), Amman 11941, Jordan; (S.S.); (A.A.); (M.A.-R.); (B.S.); (H.A.-R.)
| | - Asem Mansour
- Department of Diagnostic Radiology, King Hussein Cancer Center (KHCC), Amman 11941, Jordan;
| |
Collapse
|
11
|
Ovruchesky E, Pan E, Guer M, Elliott A, Siva S, Ravi P, McGregor B, Bagrodia A, Derweesh I, Barata P, Heath EI, Antonarakis ES, Darabi S, Hoon DSB, Mortazavi A, Choueiri TK, Nabhan C, Wei S, McKay RR. Characterization of FOLH1 Expression in Renal Cell Carcinoma. Cancers (Basel) 2024; 16:1855. [PMID: 38791934 PMCID: PMC11119455 DOI: 10.3390/cancers16101855] [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: 04/09/2024] [Revised: 04/26/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
PURPOSE Given the emergence of PSMA-targeted diagnostic agents and therapeutics, we sought to investigate patterns of FOLH1 expression in RCC and their impacts on RCC outcomes. METHODS We conducted a pooled multi-institutional analysis of patients with RCC having undergone DNA and RNA next-generation sequencing. FOLH1-high/low expression was defined as the ≥75th/<25th percentile of RNA transcripts per million (TPM). Angiogenic, T-effector, and myeloid expression signatures were calculated using previously defined gene sets. Kaplan-Meier estimates were calculated from the time of tissue collection or therapy start. RESULTS We included 1,724 patients in the analysis. FOLH1 expression was significantly higher in clear cell (71%) compared to non-clear cell RCC tumors (19.0 versus 3.3 TPM, p < 0.001) and varied by specimen site (45% primary kidney/55% metastasis, 13.6 versus 9.9 TPM, p < 0.001). FOLH1 expression was correlated with angiogenic gene expression (Spearman = 0.76, p < 0.001) and endothelial cell abundance (Spearman = 0.76, p < 0.001). While OS was similar in patients with FOLH1-high versus -low ccRCC, patients with FOLH1-high clear cell tumors experienced a longer time on cabozantinib treatment (9.7 versus 4.6 months, respectively, HR 0.57, 95% CI 0.35-0.93, p < 0.05). CONCLUSIONS We observed differential patterns of FOLH1 expression based on histology and tumor site in RCC. FOLH1 was correlated with angiogenic gene expression, increased OS, and a longer duration of cabozantinib treatment.
Collapse
Affiliation(s)
- Eric Ovruchesky
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| | - Elizabeth Pan
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| | - Melis Guer
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| | - Andrew Elliott
- Department of Clinical and Translational Research, Caris Life Sciences, Inc., Phoenix, AZ 85040, USA
| | - Shankar Siva
- Division of Radiation Oncology and Cancer Imaging, Department of Oncology, Peter MacCallum Cancer Centre, the University of Melbourne, Melbourne, VIC 3052, Australia
| | - Praful Ravi
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Bradley McGregor
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Aditya Bagrodia
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| | - Ithaar Derweesh
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| | - Pedro Barata
- Department of Hematology and Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Elisabeth I. Heath
- Department of Oncology, Wayne State University, Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Emmanuel S. Antonarakis
- Department of Hematology and Oncology, University of Minnesota, Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - Sourat Darabi
- Clinical Genomics, Hoag Family Cancer Institute, Newport Beach, CA 92663, USA
| | - Dave S. B. Hoon
- Department of Translational Molecular Medicine, Saint John’s Cancer Institute, Providence Health Systems, Santa Monica, CA 90404, USA
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, The Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Toni K. Choueiri
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Chadi Nabhan
- Department of Clinical and Translational Research, Caris Life Sciences, Inc., Phoenix, AZ 85040, USA
| | - Shuanzeng Wei
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Rana R. McKay
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| |
Collapse
|
12
|
Houédé N, Hebert K. Combining enzalutamide and [ 177Lu]Lu-PSMA-617 in metastatic castration-resistant prostate cancer. Lancet Oncol 2024; 25:531-533. [PMID: 38621399 DOI: 10.1016/s1470-2045(24)00179-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/17/2024]
Affiliation(s)
- Nadine Houédé
- Département d'Oncologie Médicale, Institut de Cancérologie du Gard, CHU de Nîmes, 30900 Nîmes, France; Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Montpellier, France.
| | - Kevin Hebert
- Département de Médecine Nucléaire, Institut du Cancer de Montpellier and Université de Montpellier, Montpellier, France
| |
Collapse
|
13
|
Grob D, Privé BM, Muselaers CHJ, Mehra N, Nagarajah J, Konijnenberg MW, Peters SMB. Bone marrow dosimetry in low volume mHSPC patients receiving Lu-177-PSMA therapy using SPECT/CT. EJNMMI Phys 2024; 11:34. [PMID: 38568428 PMCID: PMC10991600 DOI: 10.1186/s40658-024-00636-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Bone marrow toxicity in advanced prostate cancer patients who receive [177Lu]Lu-PSMA-617 is a well-known concern. In early stage patients; e.g. low volume metastatic hormone sensitive prostate cancer (mHSPC) patients, prevention of late bone marrow toxicity is even more crucial due to longer life expectancy. To date, bone marrow dosimetry is primarily performed using blood sampling. This method is time consuming and does not account for possible active bone marrow uptake. Therefore other methodologies are investigated. We calculated the bone marrow absorbed dose for [177Lu]Lu-PSMA-617 in mHSPC patients using SPECT/CT imaging and compared it to the blood sampling method as reference. METHODS Eight mHSPC patients underwent two cycles (3 and 6 GBq) of [177Lu]Lu-PSMA-617 therapy. After each cycle, five time point (1 h, 1 day, 2 days, 3 days, 7 days) SPECT/CT was performed at kidney level. Bone marrow dosimetry was performed using commercial software by drawing ten 1.5 cm diameter spheres in the lowest ten vertebrae to determine the time-integrated activity. Simplified protocols using only 2 imaging time points and 3 vertebrae were also compared. Blood-based dosimetry was based on the blood sampling method according to the EANM guideline. RESULTS Mean bone marrow absorbed dose was significantly different (p < 0.01) for the imaging based method (25.4 ± 8.7 mGy/GBq) and the blood based method (17.2 ± 3.4 mGy/GBq), with an increasing absorbed dose ratio between both methods over time. Bland Altman analysis of both simplification steps showed that differences in absorbed dose were all within the 95% limits of agreement. CONCLUSION This study showed that bone marrow absorbed dose after [177Lu]Lu-PSMA-617 can be determined using an imaging-based method of the lower vertebrae, and simplified using 2 time points (1 and 7 days) and 3 vertebrae. An increasing absorbed dose ratio over time between the imaging-based method and blood-based method suggests that there might be specific bone marrow binding of [177Lu]Lu-PSMA-617.
Collapse
Affiliation(s)
- Dagmar Grob
- Department of Medical Imaging, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Healthcare and Information Technology, Slingeland Hospital, Doetinchem, The Netherlands
| | - Bastiaan M Privé
- Department of Medical Imaging, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Radiation Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - James Nagarajah
- Department of Medical Imaging, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Mark W Konijnenberg
- Department of Medical Imaging, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Steffie M B Peters
- Department of Medical Imaging, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
14
|
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
|
15
|
Eisazadeh R, Mirshahvalad SA, Schwieghofer-Zwink G, Hehenwarter L, Rendl G, Gampenrieder S, Greil R, Pirich C, Beheshti M. Pre-treatment 68 Ga-PSMA-11 PET/CT Prognostic Value in Predicting Response to 177Lu-PSMA-I&T Therapy and Patient Survival. Mol Imaging Biol 2024; 26:360-369. [PMID: 38360991 DOI: 10.1007/s11307-024-01900-6] [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: 11/09/2023] [Revised: 01/15/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
PURPOSE To assess the prognostic value of pre-treatment [68Ga]Ga-PSMA-11 PET/CT and other baseline clinical characteristics in predicting prostate cancer (PCa) patients response to [177Lu]Lu-PSMA (PSMA-I&T), as well as patient survival. PROCEDURES In this retrospective study, 81 patients who received [177Lu]Lu-PSMA-I&T between October 2018 and January 2023 were reviewed. Eligible patients had metastatic castration-resistant PCa, underwent pre-treatment [68Ga]Ga-PSMA-11 PET/CT, and had serum prostate-specific antigen (PSA) levels available. On PET/CT images, SUVmax, SULmax, SUVpeak, and SULpeak of the most-avid tumoral lesion, as well as SUVmean of the parotid gland (P-SUVmean) and liver (L-SUVmean), were measured. Also, whole-body PSMA tumour volume (PSMA-TV) and total lesion PSMA (TL-PSMA) were calculated. To interpret treatment response after [177Lu]Lu-PSMA-I&T, a composite of PSA values and [68Ga]Ga-PSMA-11 PET/CT findings were considered. The outcomes were dichotomised into progressive versus controlled (stable disease or partial response) disease. Then, the association of baseline parameters with patient response was evaluated. Also, survival analyses were performed to assess baseline parameters in predicting overall survival. RESULTS Sixty patients (age:73 ± 8, PSA:185 ± 371) were included. Patients received at least one cycle of [177Lu]Lu-PSMA therapy (median = 4). Overall, half of the patients showed disease progression. In the progressive versus controlled disease evaluation, the highest SULmax, as well as SUVmax and SULmax to both backgrounds (L-SUVmean and P-SUVmean), were significantly correlated with the outcome (p-values < 0.05). In the multivariate analysis, only SULmax to the L-SUVmean remained significant (p-value = 0.038). The best cut-off was 8 (AUC = 0.71). With a median follow-up of 360 days, 11 mortal events were documented. In the multivariate survival analysis, only SULmax to P-SUVmean (cut-off = 2.4; p-value = 0.043) retained significance (hazard ratio = 4.0). CONCLUSIONS A greater level of PSMA uptake, specifically higher tumour-to-background uptake in the hottest lesion, may hold substantial prognostic significance, considering both [177Lu]Lu-PSMA-I&T response and patient survival. These ratios may have the potential to be used for PCa patient selection for radioligand therapy.
Collapse
Affiliation(s)
- Roya Eisazadeh
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University Salzburg, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
| | - Seyed Ali Mirshahvalad
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University Salzburg, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
- Joint Department of Medical Imaging, University Medical Imaging Toronto (UMIT), University Health Network, Mount Sinai Hospital & Women's College Hospital; University of Toronto, Toronto, ON, Canada
| | - Gregor Schwieghofer-Zwink
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University Salzburg, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
| | - Lukas Hehenwarter
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University Salzburg, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
| | - Gundula Rendl
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University Salzburg, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
| | - Simon Gampenrieder
- Department of Internal Medicine III With Haematology, Medical Oncology, and Oncologic Center, University Hospital, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Richard Greil
- Department of Internal Medicine III With Haematology, Medical Oncology, and Oncologic Center, University Hospital, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University Salzburg, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University Salzburg, Muellner Hauptstrasse 48, 5020, Salzburg, Austria.
| |
Collapse
|
16
|
Denmeade SR. Resolute Progress Down a Long and Winding Road Leads to the Promised Land of Prostate-Specific Membrane Antigen-Based Therapies for Prostate Cancer. J Clin Oncol 2024; 42:852-856. [PMID: 38181307 DOI: 10.1200/jco.23.02310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 01/07/2024] Open
|
17
|
Yazdani E, Karamzadeh-Ziarati N, Cheshmi SS, Sadeghi M, Geramifar P, Vosoughi H, Jahromi MK, Kheradpisheh SR. Automated segmentation of lesions and organs at risk on [ 68Ga]Ga-PSMA-11 PET/CT images using self-supervised learning with Swin UNETR. Cancer Imaging 2024; 24:30. [PMID: 38424612 PMCID: PMC10903052 DOI: 10.1186/s40644-024-00675-x] [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: 12/04/2023] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA) PET/CT imaging is widely used for quantitative image analysis, especially in radioligand therapy (RLT) for metastatic castration-resistant prostate cancer (mCRPC). Unknown features influencing PSMA biodistribution can be explored by analyzing segmented organs at risk (OAR) and lesions. Manual segmentation is time-consuming and labor-intensive, so automated segmentation methods are desirable. Training deep-learning segmentation models is challenging due to the scarcity of high-quality annotated images. Addressing this, we developed shifted windows UNEt TRansformers (Swin UNETR) for fully automated segmentation. Within a self-supervised framework, the model's encoder was pre-trained on unlabeled data. The entire model was fine-tuned, including its decoder, using labeled data. METHODS In this work, 752 whole-body [68Ga]Ga-PSMA-11 PET/CT images were collected from two centers. For self-supervised model pre-training, 652 unlabeled images were employed. The remaining 100 images were manually labeled for supervised training. In the supervised training phase, 5-fold cross-validation was used with 64 images for model training and 16 for validation, from one center. For testing, 20 hold-out images, evenly distributed between two centers, were used. Image segmentation and quantification metrics were evaluated on the test set compared to the ground-truth segmentation conducted by a nuclear medicine physician. RESULTS The model generates high-quality OARs and lesion segmentation in lesion-positive cases, including mCRPC. The results show that self-supervised pre-training significantly improved the average dice similarity coefficient (DSC) for all classes by about 3%. Compared to nnU-Net, a well-established model in medical image segmentation, our approach outperformed with a 5% higher DSC. This improvement was attributed to our model's combined use of self-supervised pre-training and supervised fine-tuning, specifically when applied to PET/CT input. Our best model had the lowest DSC for lesions at 0.68 and the highest for liver at 0.95. CONCLUSIONS We developed a state-of-the-art neural network using self-supervised pre-training on whole-body [68Ga]Ga-PSMA-11 PET/CT images, followed by fine-tuning on a limited set of annotated images. The model generates high-quality OARs and lesion segmentation for PSMA image analysis. The generalizable model holds potential for various clinical applications, including enhanced RLT and patient-specific internal dosimetry.
Collapse
Affiliation(s)
- Elmira Yazdani
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences, Tehran, 14155-6183, Iran
- Fintech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Seyyed Saeid Cheshmi
- Department of Computer and Data Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran
| | - Mahdi Sadeghi
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences, Tehran, 14155-6183, Iran.
- Fintech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Parham Geramifar
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Habibeh Vosoughi
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Nuclear Medicine and Molecular Imaging Department, Imam Reza International University, Razavi Hospital, Mashhad, Iran
| | - Mahmood Kazemi Jahromi
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences, Tehran, 14155-6183, Iran
- Fintech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Saeed Reza Kheradpisheh
- Department of Computer and Data Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
18
|
Zang J, Wang G, Zhao T, Liu H, Lin X, Yang Y, Shao Z, Wang C, Chen H, Chen Y, Zhu Z, Miao W, Chen X, Zhang J. A phase 1 trial to determine the maximum tolerated dose and patient-specific dosimetry of [ 177Lu]Lu-LNC1003 in patients with metastatic castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 2024; 51:871-882. [PMID: 37864592 DOI: 10.1007/s00259-023-06470-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/03/2023] [Indexed: 10/23/2023]
Abstract
PURPOSE This translational study aimed to determine the maximum tolerated dose (MTD), safety, dosimetry, and therapeutic efficacy of 177Lu-PSMA-EB-01 (denoted as [177Lu]Lu-LNC1003) in patients with metastatic castration-resistant prostate cancer (mCRPC). METHODS A total of 13 patients with mCRPC were recruited in this study. A standard 3 + 3 dose escalation protocol was performed. The following dose levels were ultimately evaluated: 1.11, 1.85, and 2.59 GBq/cycle. Patients received [177Lu]Lu-LNC1003 therapy for up to two cycles at a 6-week interval. RESULTS Patients received fractionated doses of [177Lu]Lu-LNC1003 ranging from 1.11 to 2.59 GBq per cycle. Myelosuppression was dose-limiting at 2.59 GBq, and 1.85 GBq was determined to be the MTD. The total-body effective dose for 177Lu-LNC1003 was 0.35 ± 0.05 mSv/MBq. The salivary glands were found to receive the highest estimated radiation dose, which was calculated to be 3.61 ± 2.83 mSv/MBq. The effective doses of kidneys and red bone marrow were 1.88 ± 0.35 and 0.22 ± 0.04 mSv/MBq, respectively. The tumor mean absorbed doses for bone and lymph node metastases were 8.52 and 9.51 mSv/MBq. Following the first treatment cycle, PSA decline was observed in 1 (33.3%), 4 (66.7%), and 2 (50.0%) patients at dose levels 1 (1.11 GBq), 2 (1.85 GBq), and 3 (2.59 GBq), respectively. Compared with the baseline serum PSA value, 1 (33.3%) at dose level 1 and 4 (66.6%) patients at dose level 2, presented a PSA decline after the second treatment cycle. CONCLUSION This phase 1 trial revealed that the MTD of [177Lu]Lu-LNC1003 is 1.85 GBq. The treatment with multiple cycles at the dose of 1.11 GBq /cycle and 1.85 GBq /cycle was well tolerated. [177Lu]Lu-LNC1003 has higher tumor effective doses in bone and lymph nodes metastases while the absorbed dose in the red bone marrow should be closely monitored in future treatment studies with higher doses and multiple cycles. The frequency of administration also needs to be further explored to assess the efficacy and side effects of [177Lu]Lu-LNC1003 treatment. TRIAL REGISTRATION 177Lu-PSMA-EB-01 in patients with metastatic castration-resistant prostate cancer (NCT05613738, Registered 14 November 2022). URL of registry https://classic. CLINICALTRIALS gov/ct2/show/NCT05613738.
Collapse
Affiliation(s)
- Jie Zang
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian Province, China
- Department of Nuclear Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian Province, China
| | - Guochang Wang
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian Province, China
- Department of Nuclear Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian Province, China
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Tianzhi Zhao
- Departments of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Huipan Liu
- Department of Nuclear Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Sichuan Province, Luzhou, 646000, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, Sichuan Province, China
| | - Xiuting Lin
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian Province, China
- Department of Nuclear Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian Province, China
| | - Yun Yang
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian Province, China
- Department of Nuclear Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian Province, China
| | - Zezhong Shao
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian Province, China
- Department of Nuclear Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian Province, China
| | - Chao Wang
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian Province, China
- Department of Nuclear Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian Province, China
| | - Haojun Chen
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Cancer Center, First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Yue Chen
- Department of Nuclear Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Sichuan Province, Luzhou, 646000, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, Sichuan Province, China
| | - Zhaohui Zhu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China.
| | - Weibing Miao
- Department of Nuclear Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian Province, China.
- Department of Nuclear Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian Province, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian Province, China.
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Departments of Surgery, Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117597, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117597, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Singapore.
| | - Jingjing Zhang
- Departments of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
| |
Collapse
|
19
|
Unterrainer LM, Calais J, Bander NH. Prostate-Specific Membrane Antigen: Gateway to Management of Advanced Prostate Cancer. Annu Rev Med 2024; 75:49-66. [PMID: 38285513 DOI: 10.1146/annurev-med-081522-031439] [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] [Indexed: 01/31/2024]
Abstract
Prostate-specific membrane antigen (PSMA) as a transmembrane protein is overexpressed by prostate cancer (PC) cells and is accessible for binding antibodies or low-molecular-weight radioligands due to its extracellular portion. Successful targeting of PSMA began with the development of humanized J591 antibody. Due to their faster clearance compared to antibodies, small-molecule radioligands for targeted imaging and therapy of PC have been favored in recent development efforts. PSMA positron emission tomography (PET) imaging has higher diagnostic performance than conventional imaging for initial staging of high-risk PC and biochemical recurrence detection/localization. However, it remains to be demonstrated how to integrate PSMA PET imaging for therapy response assessment and as an outcome endpoint measure in clinical trials. With the recent approval of 177Lu-PSMA-617 by the US Food and Drug Administration for metastatic castration-resistant PC progressing after chemotherapy, the high value of PSMA-targeted therapy was confirmed. Compared to standard of care, PSMA-based radioligand therapy led to a better outcome and a higher quality of life. This review, focusing on the advanced PC setting, provides an overview of different approved and nonapproved PSMA-targeted imaging and therapeutic modalities and discusses the future of PSMA-targeted theranostics, also with an outlook on non-radiopharmaceutical-based PSMA-targeted therapies.
Collapse
Affiliation(s)
- Lena M Unterrainer
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA; ,
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA; ,
| | - Neil H Bander
- Department of Urology, Weill Cornell Medicine, New York, NY, USA;
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
20
|
Ravi Kumar AS, Hofman MS. Unraveling the Impact of 177Lu-PSMA Radioligand Therapy on Renal Impairment: Distinguishing Causation from Correlation. J Nucl Med 2024; 65:85-86. [PMID: 37973187 DOI: 10.2967/jnumed.123.266638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/01/2023] [Indexed: 11/19/2023] Open
Affiliation(s)
- Aravind S Ravi Kumar
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Centre Melbourne, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Centre Melbourne, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
de la Fuente-Mendoza JE, Azorín-Vega EP, Mendoza-Nava HJ, Rodríguez-Martínez G, Rodríguez-Dorantes M. Estimation of the relative biological effectiveness (RBE) of the Lu-DOTA-iPSMA177<!--Q1:CorrectlyacknowledgingtheprimaryfundersandgrantIDsofyourresearchisimportanttoensurecompliancewithfunderpolicies.Pleasemakesurethatfundersarementionedaccordingly.--> radiopharmaceutical. Appl Radiat Isot 2023; 202:111065. [PMID: 37879244 DOI: 10.1016/j.apradiso.2023.111065] [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: 02/24/2023] [Revised: 08/28/2023] [Accepted: 10/05/2023] [Indexed: 10/27/2023]
Abstract
Relative biological effectiveness is a radiobiological parameter relevant in radiotherapy planning and useful in evaluating the physiological impact of radiation in different tissues. Targeted radionuclide therapy allows the selective and specific deposition of higher radiation doses in a noninvasive way and without collateral effects through the administration of radiopharmaceuticals. Lu-DOTA-177(hydrazinylnicotinoyl-Lys-(Nal)-NH-CO-NH-Glu) also called Lu-iPSMA177 is a third generation radiopharmaceutical composed by a peptide that recognizes the prostate-specific membrane antigen (PSMA), a membrane protein overexpressed in several types of cancer and that mediates the radiopharmaceutical's recognition of cancer cells. The present study reports radiobiological parameters of Lu-iPSMA177 and demonstrates the superiority of targeted radiopharmaceuticals over external radiotherapy treatment options in terms of their relative biological effectiveness. The relative biological effectiveness value of 1.020±0.003 for the LINAC, estimated by fitting the linear-quadratic model equation to the resulting survival curves, was like those of 1.25±0.04,1.060±0.005and1.00±0.04 obtained by an alternative method in relation to the mean lethal doses at 90, 80 or 60 survival percent respectively. While the relative biological effectiveness values of 5.65±0.13,4.72±0.27and2.87±0.19 estimated for Lu-iPSMA177 were significantly higher than those for the LINAC. The results confirm that the biological effect produced by the deposition of a radiation absorbed dose delivered by the LINAC can be induced with a quarter of that dose using Lu-iPSMA177 due to the energy distribution, dose-rate and energy fluence.
Collapse
|
22
|
Sudo H, Tsuji AB, Sugyo A, Harada Y, Nagayama S, Katagiri T, Nakamura Y, Higashi T. Head-to-head comparison of three chelates reveals DOTAGA promising for 225 Ac labeling of anti-FZD10 antibody OTSA101. Cancer Sci 2023; 114:4677-4690. [PMID: 37781962 PMCID: PMC10728013 DOI: 10.1111/cas.15978] [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: 05/11/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 10/03/2023] Open
Abstract
To select the most suitable chelate for 225 Ac radiolabeling of the anti-FZD10 antibody OTSA101, we directly compared three chelates: S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane tetraacetic acid (p-SCN-Bn-DOTA), 2,2',2″-(10-(1-carboxy-4-((4-isothiocyanatobenzyl)amino)-4-oxobutyl)-1,4,7,10-tetraazacyclododecane-1,4,7-triyl) triacetic acid (p-SCN-Bn-DOTAGA), and 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid mono-N-hydroxysuccinimide ester (DO3A-NHS-ester). We evaluated the binding affinity of the chelate-conjugated OTSA101 antibodies, as well as the labeling efficiency and stability in murine serum of 225 Ac-labeled OTSA101 as in vitro properties. The biodistribution, intratumoral distribution, absorbed doses, and therapeutic effects of the chelate-conjugated OTSA101 antibodies were assessed in the synovial sarcoma mouse model SYO-1. Of the three conjugates, DOTAGA conjugation had the smallest impact on the binding affinity (p < 0.01). The labeling efficiencies of DOTAGA-OTSA101 and DO3A-OTSA101 were 1.8-fold higher than that of DOTA-OTSA101 (p < 0.01). The stabilities were similar between 225 Ac-labeled DOTA-OTSA101, DOTAGA-OTSA101, and DO3A-OTSA101in serum at 37 and 4°C. The dosimetric analysis based on the biodistribution revealed significantly higher tumor-absorbed doses by 225 Ac-labeled DOTA-OTSA101 and DOTAGA-OTSA101 compared with 225 Ac-DO3A-OTSA101 (p < 0.05). 225 Ac-DOTAGA-OTSA101 exhibited the highest tumor-to-bone marrow ratio, with bone marrow being the dose-limiting tissue. The therapeutic and adverse effects were not significantly different between the three conjugates. Our findings indicate that among the three evaluated chelates, DOTAGA appears to be the most promising chelate to produce 225 Ac-labeled OTSA101 with high binding affinity and high radiochemical yields while providing high absorbed doses to tumors and limited absorbed doses to bone marrow.
Collapse
Affiliation(s)
- Hitomi Sudo
- Department of Molecular Imaging and TheranosticsNational Institutes for Quantum Science and Technology (QST)ChibaJapan
| | - Atsushi B. Tsuji
- Department of Molecular Imaging and TheranosticsNational Institutes for Quantum Science and Technology (QST)ChibaJapan
| | - Aya Sugyo
- Department of Molecular Imaging and TheranosticsNational Institutes for Quantum Science and Technology (QST)ChibaJapan
| | | | | | - Toyomasa Katagiri
- Division of Genome MedicineInstitute of Advanced Medical Sciences, Tokushima UniversityTokushimaJapan
- National Institutes of Biomedical Innovation, Health and NutritionIbarakiJapan
| | - Yusuke Nakamura
- National Institutes of Biomedical Innovation, Health and NutritionIbarakiJapan
| | - Tatsuya Higashi
- Department of Molecular Imaging and TheranosticsNational Institutes for Quantum Science and Technology (QST)ChibaJapan
| |
Collapse
|
23
|
Feuerecker B, Gafita A, Langbein T, Tauber R, Seidl C, Bruchertseifer F, Gschwendt JE, Weber WA, D’Alessandria C, Morgenstern A, Eiber M. Comparative Analysis of Morphological and Functional Effects of 225Ac- and 177Lu-PSMA Radioligand Therapies (RLTs) on Salivary Glands. Int J Mol Sci 2023; 24:16845. [PMID: 38069166 PMCID: PMC10706561 DOI: 10.3390/ijms242316845] [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: 08/31/2023] [Revised: 10/24/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Most Prostate Specific Membrane Antigens (PSMAs) targeting small molecules accumulate in the salivary glands (SGs), raising concerns about SG toxicity, especially after repeated therapies or therapy with 225Ac-labeled ligands. SG toxicity is assessed clinically by the severity of patient-reported xerostomia, but this parameter can be challenging to objectively quantify. Therefore, we explored the feasibility of using SG volume as a biomarker for toxicity. In 21 patients with late-stage metastatic resistant prostate cancer (mCRPC), the PSMA volume and ligand uptake of SG were analyzed retrospectively before and after two cycles of 177Lu-PSMA (LuPSMA; cohort A) and before and after one cycle of 225Ac-PSMA-617 (AcPSMA, cohort B). Mean Volume-SG in cohort A was 59 ± 13 vs. 54 ± 16 mL (-10%, p = 0.4), and in cohort B, it was 50 ± 13 vs. 40 ± 11 mL (-20%, p = 0.007), respectively. A statistically significant decrease in the activity concentration in the SG was only observed in group B (SUVmean: 9.2 ± 2.8 vs. 5.3 ± 1.8, p < 0.0001; vs. A: SUVmean: 11.2 ± 3.3 vs. 11.1 ± 3.5, p = 0.8). SG volume and PSMA-ligand uptake are promising markers to monitor the SG toxicity after a PSMA RLT.
Collapse
Affiliation(s)
- Benedikt Feuerecker
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, 81675 München, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partnersite München, 69124 Heidelberg, Germany
- Department of Radiology, University Hospital, LMU Munich, 81377 München, Germany
- Department of Radiology, School of Medicine, Technical University of Munich, 81675 München, Germany
| | - Andrei Gafita
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Thomas Langbein
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, 81675 München, Germany
| | - Robert Tauber
- Department of Urology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, 81675 München, Germany
| | - Christof Seidl
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, 81675 München, Germany
| | | | - Jürgen E. Gschwendt
- Department of Urology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, 81675 München, Germany
| | - Wolfgang A. Weber
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, 81675 München, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partnersite München, 69124 Heidelberg, Germany
| | - Calogero D’Alessandria
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, 81675 München, Germany
| | - Alfred Morgenstern
- European Commission, Joint Research Centre (JRC), 76344 Karlsruhe, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, 81675 München, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partnersite München, 69124 Heidelberg, Germany
| |
Collapse
|
24
|
Pouget JP, Chan TA, Galluzzi L, Constanzo J. Radiopharmaceuticals as combinatorial partners for immune checkpoint inhibitors. Trends Cancer 2023; 9:968-981. [PMID: 37612188 PMCID: PMC11311210 DOI: 10.1016/j.trecan.2023.07.014] [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: 07/04/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of multiple cancer types. However, only a fraction of patients with cancer responds to ICIs employed as stand-alone therapeutics, calling for the development of safe and effective combinatorial regimens to extend the benefits of ICIs to a larger patient population. In addition to exhibiting a good safety and efficacy profile, targeted radionuclide therapy (TRT) with radiopharmaceuticals that specifically accumulate in the tumor microenvironment has been associated with promising immunostimulatory effects that (at least in preclinical cancer models) provide a robust platform for the development of TRT/ICI combinations. We discuss preclinical and clinical findings suggesting that TRT stands out as a promising partner for the development of safe and efficient combinatorial regimens involving ICIs.
Collapse
Affiliation(s)
- Jean-Pierre Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France.
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Centre, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| | - Julie Constanzo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| |
Collapse
|
25
|
Hohberg M, Reifegerst M, Drzezga A, Wild M, Schmidt M. Prediction of Response to 177Lu-PSMA Therapy Based on Tumor-to-Kidney Ratio on Pretherapeutic PSMA PET/CT and Posttherapeutic Tumor-Dose Evaluation in mCRPC. J Nucl Med 2023; 64:1758-1764. [PMID: 37652546 DOI: 10.2967/jnumed.122.264953] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 07/25/2023] [Indexed: 09/02/2023] Open
Abstract
The aim of this study was to analyze the absorbed dose of 177Lu-PSMA in osseous versus lymphatic metastases in patients with metastatic castration-resistant prostate cancer across therapy cycles and to relate those data to therapeutic success. In addition, pretherapeutic prostate-specific membrane antigen (PSMA) PET/CT was evaluated for its ability to predict response behavior. Methods: The study comprised 30 patients with metastatic castration-resistant prostate cancer, each receiving at least 3 cycles of 177Lu-PSMA therapy. Prostate-specific antigen (PSA) values between baseline and 6 wk after the third therapy cycle were used to classify the patients as responders (PSA decline ≥ 50%) or nonresponders (unchanged or increasing PSA level). Quantitative SPECT/CT images were acquired 24, 48, and 168 h after application of 177Lu-PSMA. The absorbed dose for tumor lesions was calculated with dosimetry software. From the pretherapeutic PET/CT scan, the tumor-to-kidney uptake ratio was determined for different SUVs. Results: Regardless of patient response, the kidneys received a mean dose of 0.55 ± 0.20 Gy/GBq per cycle. In the first therapy cycle, the lymph node lesions received a mean dose of 3.73 ± 1.65 Gy/GBq in responders and 1.86 ± 1.25 Gy/GBq in nonresponders (P < 0.01). For bone lesions, the respective mean doses were 3.47 ± 2.00 Gy/GBq and 1.48 ± 0.95 Gy/GBq (P < 0.01). When successive therapy cycles were compared, the mean dose was found to have been reduced from the first to the second cycle by 27% for lymph nodes and by 33% for bone lesions. A significant difference (P < 0.01) in the ratio of lymph node and bone lesion uptake to kidney uptake between responders and nonresponders could be deduced from the pretherapeutic PET/CT scan. Conclusion: Significantly higher doses were achieved for lymph node and bone lesions in responders. The highest absorbed dose, for both lymphatic and osseous lesions, was achieved in the first cycle, decreasing in the second therapy cycle thereafter despite unchanged therapy activities. It may be possible to estimate the response to therapy from the ratio of tumor uptake to kidney uptake obtained from the pretherapeutic PSMA PET/CT scans.
Collapse
Affiliation(s)
- Melanie Hohberg
- Department of Nuclear Medicine and Cancer Center Cologne, University Hospital of Cologne, Cologne, Germany
| | - Manuel Reifegerst
- Department of Nuclear Medicine and Cancer Center Cologne, University Hospital of Cologne, Cologne, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine and Cancer Center Cologne, University Hospital of Cologne, Cologne, Germany
| | - Markus Wild
- Department of Nuclear Medicine and Cancer Center Cologne, University Hospital of Cologne, Cologne, Germany
| | - Matthias Schmidt
- Department of Nuclear Medicine and Cancer Center Cologne, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
26
|
Okuhata K, Monzen H, Nakamura Y, Takai G, Nagano K, Nakamura K, Kubo K, Hosono M. Effectiveness of shielding materials against 177Lu gamma rays and the corresponding distance relationship. Ann Nucl Med 2023; 37:629-634. [PMID: 37596439 DOI: 10.1007/s12149-023-01860-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/07/2023] [Indexed: 08/20/2023]
Abstract
OBJECTIVE The purpose of this study is to determine the dose reduction of different shielding materials at various distances from a 177Lu photon radiation source. METHODS Two protective aprons with lead equivalent thicknesses of 0.25 mm and 0.35 mm and tungsten-containing rubber (TCR) were used as shielding materials. A vial containing 177Lu was sealed in a lead container so that a narrow beam went out through a 3 mm-diameter hole. The dose rate was measured at distances of 0, 10, 50, 100, and 200 cm from the source using a NaI scintillation survey meter to obtain the rate of dose reduction. TCR was tested with thicknesses ranging from 0.3 to 1.0 mm at 0.1 mm intervals and from 1.0 to 4.0 mm at 0.5 mm intervals. RESULTS At distances of 0, 10, 50, 100, and 200 cm, the dose reduction for the lead equivalent thickness of 0.25 mm were 32.7%, 54.5%, 93.1%, 97.9%, and 99.6%, respectively; and for the lead equivalent thickness of 0.35 mm were 53.4%, 70.6%, 95.6%, 98.9%, and 99.6%, respectively. Without any shielding, the dose rate decreased by 34.4% at 10 cm and by 88.8% at 50 cm from the radiation source. The dose reduction for the TCR thickness of 3.5 mm was 89.8% at 0 cm and 93.3% at 10 cm. The TCR thickness of 0.4 mm provided a dose reduction comparable to or greater than that of the 0.25 mm lead equivalent, whereas the TCR thickness of 1.0 mm or greater provided a dose reduction comparable to that of the 0.35 mm lead equivalent. CONCLUSIONS Achieving a reduction of 95% or more requires the 0.25 mm lead equivalent for a distance of 100 cm, the 0.35 mm lead equivalent for 50 cm, the TCR thickness of 0.3 mm for 100 cm, or the TCR thickness of 0.9 mm for 50 cm. Without wearing a protective apron, a reduction of approximately 95% is observed at distances greater than 100 cm. These findings would be useful for medical staff engaging in related activities.
Collapse
Affiliation(s)
- Katsuya Okuhata
- Department of Radiology, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-ku, Osaka-shi, Osaka, 5530003, Japan.
| | - Hajime Monzen
- Department of Medical Physics, Graduate School of Medical Sciences, Kindai University, 377-2 Onohigashi, Osakasayama-shi, Osaka, 5898511, Japan
| | - Yasunori Nakamura
- Department of Radiology, University Hospital, Kyoto Prefectural University of Medicine, 465 Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 6028566, Japan
| | - Go Takai
- Department of Radiology, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-ku, Osaka-shi, Osaka, 5530003, Japan
| | - Keiji Nagano
- Department of Radiology, Kansai Electric Power Hospital, 2-1-7 Fukushima, Fukushima-ku, Osaka-shi, Osaka, 5530003, Japan
| | - Kenji Nakamura
- Department of Medical Physics, Graduate School of Medical Sciences, Kindai University, 377-2 Onohigashi, Osakasayama-shi, Osaka, 5898511, Japan
| | - Kazuki Kubo
- Department of Medical Physics, Graduate School of Medical Sciences, Kindai University, 377-2 Onohigashi, Osakasayama-shi, Osaka, 5898511, Japan
| | - Makoto Hosono
- Department of Radiation Oncology, Faculty of Medicine, Kindai University, 377-2 Onohigashi, Osakasayama-shi, Osaka, 5898511, Japan
| |
Collapse
|
27
|
Uygur E, Sezgin C, Parlak Y, Karatay KB, Arikbasi B, Avcibasi U, Toklu T, Barutca S, Harmansah C, Sozen TS, Maus S, Scher H, Aras O, Gumuser FG, Muftuler FZB. The Radiolabeling of [161Tb]-PSMA-617 by a Novel Radiolabeling Method and Preclinical Evaluation by In Vitro/In Vivo Methods. RESEARCH SQUARE 2023:rs.3.rs-3415703. [PMID: 37961521 PMCID: PMC10635383 DOI: 10.21203/rs.3.rs-3415703/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Prostate cancer (PC) is the most common type of cancer in elderly men, with a positive correlation with age. As resistance to treatment has developed, particularly in the progressive stage of the disease and in the presence of microfocal multiple bone metastases, new generation radionuclide therapies have emerged. Recently, [161Tb], a radiolanthanide introduced for treating micrometastatic foci, has shown great promise for treating prostate cancer. Results In this study, Terbium-161 [161Tb]Tb was radiolabeled with prostate-specific membrane antigen (PSMA)-617 ([161Tb]-PSMA-617) and the therapeutic efficacy of the radiolabeled compound investigated in vitro and in vivo. [161Tb]-PSMA-617 was found to have a radiochemical yield of 97.99 ± 2.01% and was hydrophilic. [161Tb]-PSMA-617 was also shown to have good stability, with a radiochemical yield of over 95% up to 72 hours. In vitro, [161Tb]-PSMA-617 showed a cytotoxic effect on LNCaP cells but not on PC-3 cells. In vivo, scintigraphy imaging visualized the accumulation of [161Tb]-PSMA-617 in the prostate, kidneys, and bladder. Conclusions The results suggest that [161Tb]-PSMA-617 can be an effective radiolabeled agent for the treatment of PSMA positive foci in prostate cancer.
Collapse
Affiliation(s)
- Emre Uygur
- Manisa Celal Bayar University: Manisa Celal Bayar Universitesi
| | | | - Yasemin Parlak
- Manisa Celal Bayar University: Manisa Celal Bayar Universitesi
| | - Kadriye Busra Karatay
- Ege University Institute of Nuclear Sciences: Ege Universitesi Nukleer Bilimler Enstitusu
| | | | - Ugur Avcibasi
- Manisa Celal Bayar Üniversitesi: Manisa Celal Bayar Universitesi
| | | | - Sabri Barutca
- Adnan Menderes Üniversitesi Tıp Fakültesi: Adnan Menderes Universitesi Tip Fakultesi
| | | | | | - Stephan Maus
- Saarland University Hospital and Saarland University Faculty of Medicine: Universitatsklinikum des Saarlandes und Medizinische Fakultat der Universitat des Saarlandes
| | - Howard Scher
- Memorial Sloan-Kettering Cancer Center Inpatient Hospital and Main Campus: Memorial Sloan Kettering Cancer Center
| | - Omer Aras
- Memorial Sloan-Kettering Cancer Center Inpatient Hospital and Main Campus: Memorial Sloan Kettering Cancer Center
| | | | | |
Collapse
|
28
|
Cytawa W, Hendel R, Tomasik B, Weinzierl FX, Bley T, Jassem J, Schirbel A, Buck AK, Bundschuh RA, Hartrampf PE, Werner RA, Lapa C. Early biochemical and radiographic response after one cycle of [ 177Lu]Lu-PSMA I&T radioligand therapy in metastatic castration-resistant prostate cancer patients. Eur J Nucl Med Mol Imaging 2023; 50:3765-3776. [PMID: 37474735 PMCID: PMC10547638 DOI: 10.1007/s00259-023-06326-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/27/2023] [Indexed: 07/22/2023]
Abstract
PURPOSE The aim of this study was to investigate very early radiographic PSMA PET response after one cycle of [177Lu]Lu-PSMA I&T radioligand therapy (RLT) of metastatic castration-resistant prostate cancer (mCRPC) and to assess its role in predicting overall response and survival. METHODS This retrospective study enrolled 40 mCRPC patients who were treated with a median of 3 (2-9) [177Lu]Lu-PSMA I&T RLT cycles. Biochemical response was based on the relative change of serum PSA according to PCWG3 criteria, while radiographic response referred to the relative change of PSMA-derived total viable tumor volumes expressed as total lesion PSMA (TLP). RESULTS After one cycle of RLT, biochemical partial response (PR) was seen in 8/40 (20.0%), stable disease (SD) in 22/40 (55.0%), and progressive disease (PD) in 10/40 (25%) patients. In PSMA PET, very early molecular PR was observed in 12 (30.0%), SD in 19 (47.5%), and PD in 9 (22.5%) subjects. The PSA and TLP nadir were achieved after a median of 1 (1-5) and 2 (1-6) cycles, respectively. Nineteen (47.5%) patients showed overall biochemical PR, 11 (27.5%) had SD, and 10 (25%) experienced PD. In PSMA-directed PET, 4 patients experienced molecular complete response (CR), 24 (60.0%) had PR, 4 (10.0%) SD, and 8 (20.0%) PD. Early biochemical or radiographic response was not associated with longer overall survival (OS). Overall biochemical responders had a nearly significantly longer median OS (22.7 months) than non-responders (14.4 months, p = 0.08). Early PSA progression was associated with shorter OS (12.2 months), compared to biochemical SD/PR (18.7 months, p = 0.09). CONCLUSION In this retrospective cohort, there was no association between early PSMA PET radiographic response and overall survival; hence, treatment should not be prematurely discontinued. In contrast, early PSA progression after one cycle of [177Lu]Lu-PSMA I&T RLT was an indicator of overall progression and poor clinical outcome.
Collapse
Affiliation(s)
- Wojciech Cytawa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- Department of Nuclear Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Robin Hendel
- Department of Radiology, University Hospital Würzburg, Würzburg, Germany
| | - Bartłomiej Tomasik
- Department of Oncology and Radiotherapy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Thorsten Bley
- Department of Radiology, University Hospital Würzburg, Würzburg, Germany
| | - Jacek Jassem
- Department of Oncology and Radiotherapy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Ralph A Bundschuh
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
| | - Philipp E Hartrampf
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany.
| |
Collapse
|
29
|
Volpe F, Nappi C, Piscopo L, Zampella E, Mainolfi CG, Ponsiglione A, Imbriaco M, Cuocolo A, Klain M. Emerging Role of Nuclear Medicine in Prostate Cancer: Current State and Future Perspectives. Cancers (Basel) 2023; 15:4746. [PMID: 37835440 PMCID: PMC10571937 DOI: 10.3390/cancers15194746] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Prostate cancer is the most frequent epithelial neoplasia after skin cancer in men starting from 50 years and prostate-specific antigen (PSA) dosage can be used as an early screening tool. Prostate cancer imaging includes several radiological modalities, ranging from ultrasonography, computed tomography (CT), and magnetic resonance to nuclear medicine hybrid techniques such as single-photon emission computed tomography (SPECT)/CT and positron emission tomography (PET)/CT. Innovation in radiopharmaceutical compounds has introduced specific tracers with diagnostic and therapeutic indications, opening the horizons to targeted and very effective clinical care for patients with prostate cancer. The aim of the present review is to illustrate the current knowledge and future perspectives of nuclear medicine, including stand-alone diagnostic techniques and theragnostic approaches, in the clinical management of patients with prostate cancer from initial staging to advanced disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Michele Klain
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80138 Naples, Italy; (F.V.); (C.N.); (L.P.); (E.Z.); (C.G.M.); (A.P.); (M.I.); (A.C.)
| |
Collapse
|
30
|
Abouzayed A, Seitova K, Lundmark F, Bodenko V, Oroujeni M, Tolmachev V, Rosenström U, Orlova A. 177Lu-labeled PSMA targeting therapeutic with optimized linker for treatment of disseminated prostate cancer; evaluation of biodistribution and dosimetry. Front Oncol 2023; 13:1221103. [PMID: 37829345 PMCID: PMC10565663 DOI: 10.3389/fonc.2023.1221103] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/07/2023] [Indexed: 10/14/2023] Open
Abstract
Introduction Prostate specific membrane antigen (PSMA), highly expressed in metastatic castration-resistant prostate cancer (mCRPC), is an established therapeutic target. Theranostic PSMA-targeting agents are widely used in patient management and has shown improved outcomes for mCRPC patients. Earlier, we optimized a urea-based probe for radionuclide visualization of PSMA-expression in vivo using computer modeling. With the purpose to develop a targeting agent equally suitable for radionuclide imaging and therapy, the agent containing DOTA chelator was designed (BQ7876). The aim of the study was to test the hypothesis that 177Lu-labeled BQ7876 possesses target binding and biodistribution properties potentially enabling its use for radiotherapy. Methods BQ7876 was synthesized and labeled with Lu-177. Specificity and affinity of [177Lu]Lu-BQ7876 to PSMA-expressing PC3-pip cells was evaluated and its processing after binding to cells was studied. Animal studies in mice were performed to assess its biodistribution in vivo, target specificity and dosimetry. [177Lu]Lu-PSMA-617 was simultaneously evaluated for comparison. Results BQ7876 was labeled with Lu-177 with radiochemical yield >99%. Its binding to PSMA was specific in vitro and in vivo when tested in antigen saturation conditions as well as in PSMA-negative PC-3 tumors. The binding of [177Lu]Lu-BQ7876 to living cells was characterized by rapid association, while the dissociation included a rapid and a slow phase with affinities KD1 = 3.8 nM and KD2 = 25 nM. The half-maximal inhibitory concentration for natLu-BQ7876 was 59 nM that is equal to 61 nM for natLu-PSMA-617. Cellular processing of [177Lu]Lu-BQ7876 was accompanied by slow internalization. [177Lu]Lu-BQ7876 was cleared from blood and normal tissues rapidly. Initial elevated uptake in kidneys decreased rapidly, and by 3 h post injection, the renal uptake (13 ± 3%ID/g) did not differ significantly from tumor uptake (9 ± 3%ID/g). Tumor uptake was stable between 1 and 3 h followed by a slow decline. The highest absorbed dose was in kidneys, followed by organs and tissues in abdomen. Discussion Biodistribution studies in mice demonstrated that targeting properties of [177Lu]Lu-BQ7876 are not inferior to properties of [177Lu]Lu-PSMA-617, but do not offer any decisive advantages.
Collapse
Affiliation(s)
- Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Kamila Seitova
- Scientific and Research Laboratory of Chemical and Pharmaceutical Research, Siberian State Medical University, Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
| | - Fanny Lundmark
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Vitalina Bodenko
- Scientific and Research Laboratory of Chemical and Pharmaceutical Research, Siberian State Medical University, Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Affibody AB, Solna, Sweden
| | - Vladimir Tolmachev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ulrika Rosenström
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
31
|
Varaprasad GL, Gupta VK, Prasad K, Kim E, Tej MB, Mohanty P, Verma HK, Raju GSR, Bhaskar L, Huh YS. Recent advances and future perspectives in the therapeutics of prostate cancer. Exp Hematol Oncol 2023; 12:80. [PMID: 37740236 PMCID: PMC10517568 DOI: 10.1186/s40164-023-00444-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 09/10/2023] [Indexed: 09/24/2023] Open
Abstract
Prostate cancer (PC) is one of the most common cancers in males and the fifth leading reason of death. Age, ethnicity, family history, and genetic defects are major factors that determine the aggressiveness and lethality of PC. The African population is at the highest risk of developing high-grade PC. It can be challenging to distinguish between low-risk and high-risk patients due to the slow progression of PC. Prostate-specific antigen (PSA) is a revolutionary discovery for the identification of PC. However, it has led to an increase in over diagnosis and over treatment of PC in the past few decades. Even if modifications are made to the standard PSA testing, the specificity has not been found to be significant. Our understanding of PC genetics and proteomics has improved due to advances in different fields. New serum, urine, and tissue biomarkers, such as PC antigen 3 (PCA3), have led to various new diagnostic tests, such as the prostate health index, 4K score, and PCA3. These tests significantly reduce the number of unnecessary and repeat biopsies performed. Chemotherapy, radiotherapy, and prostatectomy are standard treatment options. However, newer novel hormone therapy drugs with a better response have been identified. Androgen deprivation and hormonal therapy are evolving as new and better options for managing hormone-sensitive and castration-resistant PC. This review aimed to highlight and discuss epidemiology, various risk factors, and developments in PC diagnosis and treatment regimens.
Collapse
Affiliation(s)
- Ganji Lakshmi Varaprasad
- Department of Biological Sciences and Bioengineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea
| | - Vivek Kumar Gupta
- Department of Biological Sciences and Bioengineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea
| | - Kiran Prasad
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Eunsu Kim
- Department of Biological Sciences and Bioengineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea
| | - Mandava Bhuvan Tej
- Department of Health Care Informatics, Sacred Heart University, 5151 Park Avenue, Fair Fields, CT, 06825, USA
| | - Pratik Mohanty
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lungs Health and Immunity, Helmholtz Zentrum, 85764, Neuherberg, Munich, Germany
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul, 04620, Republic of Korea.
| | - Lvks Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| | - Yun Suk Huh
- Department of Biological Sciences and Bioengineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea.
| |
Collapse
|
32
|
He M, Cao Y, Chi C, Zhao J, Chong E, Chin KXC, Tan NZV, Dmitry K, Yang G, Yang X, Hu K, Enikeev M. Unleashing novel horizons in advanced prostate cancer treatment: investigating the potential of prostate specific membrane antigen-targeted nanomedicine-based combination therapy. Front Immunol 2023; 14:1265751. [PMID: 37795091 PMCID: PMC10545965 DOI: 10.3389/fimmu.2023.1265751] [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: 07/23/2023] [Accepted: 09/04/2023] [Indexed: 10/06/2023] Open
Abstract
Prostate cancer (PCa) is a prevalent malignancy with increasing incidence in middle-aged and older men. Despite various treatment options, advanced metastatic PCa remains challenging with poor prognosis and limited effective therapies. Nanomedicine, with its targeted drug delivery capabilities, has emerged as a promising approach to enhance treatment efficacy and reduce adverse effects. Prostate-specific membrane antigen (PSMA) stands as one of the most distinctive and highly selective biomarkers for PCa, exhibiting robust expression in PCa cells. In this review, we explore the applications of PSMA-targeted nanomedicines in advanced PCa management. Our primary objective is to bridge the gap between cutting-edge nanomedicine research and clinical practice, making it accessible to the medical community. We discuss mainstream treatment strategies for advanced PCa, including chemotherapy, radiotherapy, and immunotherapy, in the context of PSMA-targeted nanomedicines. Additionally, we elucidate novel treatment concepts such as photodynamic and photothermal therapies, along with nano-theragnostics. We present the content in a clear and accessible manner, appealing to general physicians, including those with limited backgrounds in biochemistry and bioengineering. The review emphasizes the potential benefits of PSMA-targeted nanomedicines in enhancing treatment efficiency and improving patient outcomes. While the use of PSMA-targeted nano-drug delivery has demonstrated promising results, further investigation is required to comprehend the precise mechanisms of action, pharmacotoxicity, and long-term outcomes. By meticulous optimization of the combination of nanomedicines and PSMA ligands, a novel horizon of PSMA-targeted nanomedicine-based combination therapy could bring renewed hope for patients with advanced PCa.
Collapse
Affiliation(s)
- Mingze He
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Yu Cao
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Changliang Chi
- Department of Urology, First Hospital of Jilin University, Changchun, China
| | - Jiang Zhao
- Department of Urology, Xi’an First Hospital, Xi’an, China
| | - Eunice Chong
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Ke Xin Casey Chin
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Nicole Zian Vi Tan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Korolev Dmitry
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Guodong Yang
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Xinyi Yang
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Kebang Hu
- Department of Urology, First Hospital of Jilin University, Changchun, China
| | - Mikhail Enikeev
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
33
|
He M, Cao Y, Chi C, Zhao J, Chong E, Chin KXC, Tan NZV, Dmitry K, Yang G, Yang X, Hu K, Enikeev M. Unleashing novel horizons in advanced prostate cancer treatment: investigating the potential of prostate specific membrane antigen-targeted nanomedicine-based combination therapy. Front Immunol 2023; 14. [DOI: https:/doi.org/10.3389/fimmu.2023.1265751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024] Open
Abstract
Prostate cancer (PCa) is a prevalent malignancy with increasing incidence in middle-aged and older men. Despite various treatment options, advanced metastatic PCa remains challenging with poor prognosis and limited effective therapies. Nanomedicine, with its targeted drug delivery capabilities, has emerged as a promising approach to enhance treatment efficacy and reduce adverse effects. Prostate-specific membrane antigen (PSMA) stands as one of the most distinctive and highly selective biomarkers for PCa, exhibiting robust expression in PCa cells. In this review, we explore the applications of PSMA-targeted nanomedicines in advanced PCa management. Our primary objective is to bridge the gap between cutting-edge nanomedicine research and clinical practice, making it accessible to the medical community. We discuss mainstream treatment strategies for advanced PCa, including chemotherapy, radiotherapy, and immunotherapy, in the context of PSMA-targeted nanomedicines. Additionally, we elucidate novel treatment concepts such as photodynamic and photothermal therapies, along with nano-theragnostics. We present the content in a clear and accessible manner, appealing to general physicians, including those with limited backgrounds in biochemistry and bioengineering. The review emphasizes the potential benefits of PSMA-targeted nanomedicines in enhancing treatment efficiency and improving patient outcomes. While the use of PSMA-targeted nano-drug delivery has demonstrated promising results, further investigation is required to comprehend the precise mechanisms of action, pharmacotoxicity, and long-term outcomes. By meticulous optimization of the combination of nanomedicines and PSMA ligands, a novel horizon of PSMA-targeted nanomedicine-based combination therapy could bring renewed hope for patients with advanced PCa.
Collapse
|
34
|
Inderjeeth A, Iravani A, Subramaniam S, Conduit C, Sandhu S. Novel radionuclide therapy combinations in prostate cancer. Ther Adv Med Oncol 2023; 15:17588359231187202. [PMID: 37547444 PMCID: PMC10399256 DOI: 10.1177/17588359231187202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/21/2023] [Indexed: 08/08/2023] Open
Abstract
Prostate cancer remains the commonest cancer diagnosed in males and a leading cause of cancer-related death. Men with metastatic castration-resistant prostate cancer (mCRPC) who have progressed on chemotherapy and androgen receptor pathway inhibitors (ARPI) have limited treatment options, significant morbidity, and poor outcomes. Prostate-specific membrane antigen (PSMA)-directed radionuclide therapy (RNT) is emerging as an efficacious and well-tolerated therapy; however, disease progression is universal. Several ongoing RNT trials focus on combination strategies to improve efficacy and durability of treatment response, including combinations with ARPIs, chemotherapy, immunotherapy, and targeted therapies. Further, efforts are underway to expand the role of PSMA-directed RNT to earlier stages of disease including hormone-sensitive and localized prostate cancer. In this review, we discuss the rationale and ongoing RNT combination therapeutic trials in prostate cancer and summarize the efficacy and toxicity associated with RNT.
Collapse
Affiliation(s)
- Andrisha–Jade Inderjeeth
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Amir Iravani
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Shalini Subramaniam
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- Department of Medical Oncology, Bankstown-Lidcombe Hospital, Bankstown, NSW, Australia
| | - Ciara Conduit
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Sir Peter MacCallum Cancer Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | | |
Collapse
|
35
|
Shi Y, Feng Y, Xu L, Li W, Guan L, Zuo R, Liu S, Pang H, Wang Z. The value of gallium-68 prostate-specific membrane antigen PET/CT and 2-[18F]fluoro-2-deoxy-D-glucose PET/CT in the detection of thyroid cancer lesions: a prospective head-to-head comparison. Br J Radiol 2023:bjr.20230291. [PMID: 38904463 DOI: 10.1259/bjr.20230291] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/17/2023] [Accepted: 06/04/2023] [Indexed: 07/03/2023] Open
Abstract
OBJECTIVE Thyroid cancer is increasing in incidence. Prostate-specific membrane antigen (PSMA) targeted radionuclide imaging and treatment demonstrated remarkable value in prostate cancer patients. Studies have shown that PSMA is also expressed in thyroid cancer. Our purpose is to evaluate the clinical usefulness of [68Ga]Ga-PSMA-11 PET/CT for the diagnosis of thyroid cancer. METHODS We enrolled 23 DTC and 17 RAIR-DTC patients prospectively. All patients underwent [68Ga]Ga-PSMA-11 PET/CT and 2-[18F]FDG PET/CT. PSMA expression was determined by immunohistochemistry on histological samples of lymphatic metastasis of 12 patients. We compared the detection rates and semi-quantitative parameters between [68Ga]Ga-PSMA-11PET/CT and 2-[18F]FDG PET/CT. RESULTS A total of 72 lesions were detected. Detection rates of DTC and RAIR-DTC by [68Ga]Ga-PSMA-11 PET/CT were lower than those by 2-[18F]FDG PET/CT (60.00% vs. 90.00%, P = .004; 59.38% vs. 96.88%). Compared with DTC, RAIR-DTC had higher semi-quantitative parameters of 2-[18F]FDG PET/CT. There was no significant difference in semi-quantitative parameters of [68Ga]Ga-PSMA-11 PET/CT between DTC and RAIR-DTC. Immunohistochemistry showed a significantly higher PSMA expression for RAIR-DTC than for DTC. However, there was no significant correlation between PSMA expression and SUVmax on 68Ga-PSMA [68Ga]Ga-PSMA-11 PET/CT. CONCLUSIONS [68Ga]Ga-PSMA-11 PET/CT can detect thyroid cancer metastases but its detection rate was lower than that of 2-[18F]FDG PET/CT. There was a difference in PSMA expression levels between DTC and RAIR-DTC, but the difference was not reflected on [68Ga]Ga-PSMA-11 PET/CT. ADVANCES IN KNOWLEDGE [68Ga]Ga-PSMA-11 PET/CT has potential value in the diagnosis of thyroid cancer. [68Ga]Ga-PSMA-11 PET/CT could screen out patients who may benefit from PSMA-targeted radionuclide therapy.
Collapse
Affiliation(s)
- Yangrui Shi
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuyue Feng
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lu Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wenbo Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lili Guan
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rui Zuo
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shuang Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hua Pang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhengjie Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
36
|
Parghane RV, Basu S. PSMA-targeted radioligand therapy in prostate cancer: current status and future prospects. Expert Rev Anticancer Ther 2023; 23:959-975. [PMID: 37565281 DOI: 10.1080/14737140.2023.2247562] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/19/2023] [Accepted: 08/09/2023] [Indexed: 08/12/2023]
Abstract
INTRODUCTION The prostate-specific membrane antigen (PSMA) targeted radioligand therapy (PRLT) for the treatment of metastatic castration-resistant prostate cancer (mCRPC) patients has generated significant interest among the oncologic community, with several publications documenting good response rates and survival benefits with low toxicity profiles. AREAS COVERED Indications, patient preparation, dose administration, post-treatment imaging, dosimetry, and side effect profiles of 177Lu-PSMA-617 are discussed in this article. We also discuss results from prospective studies, major retrospective studies, meta-analyses, clinical trials, and mentioned major ongoing clinical trials on PRLT. We have also portrayed our own experiences and future perspectives on PRLT. EXPERT OPINION For PRLT, PSMA-617 and PSMA-I&T molecules have revolutionized the theranostic approach in the management of advanced prostate cancer, with solid backing from several published articles showing favorable outcomes and an excellent safety profile of 177Lu-PSMA-617. Improvement in quality of life and survival was seen in the majority of mCRPC patients after 177Lu-PSMA-617 PRLT. Patients with good performance status, asymptomatic, only lymph node metastases, high PSMA expressing lesions, and no discordant FDG avid lesions have a longer survival after 177Lu-PSMA-617 PRLT than patients with poor performance status, symptomatic, hepatic, brain, and skeletal metastases, discordant PSMA, and FDG-avid lesions. Docetaxel and cabazitaxel are approved treatments for mCRPC patients. 177Lu-PSMA-617 is approved as a third-line systemic treatment for mCRPC patients with failure to respond to androgen receptor pathway inhibitors and docetaxel therapy. PRLT is a safe and effective alternative to cabazitaxel (third-line systemic treatment), but it has a higher cost. 177Lu-PSMA-617 could be a more efficient therapeutic option for mCRPC patients as first-line or combined therapy, and it may be a useful therapeutic option for the treatment of metastatic hormone-sensitive prostate cancer (mHSPC) patients. Several clinical studies and clinical trials on PRLT are currently underway. In the future, the results of these trials will be helpful in evolving treatment strategies for prostate cancer patients.
Collapse
Affiliation(s)
- Rahul V Parghane
- Radiation Medicine Centre (BARC), Tata Memorial Hospital Annexe, Mumbai, India
- Radiation Medicine Centre (BARC), Homi Bhabha National Institute, Mumbai, India
| | - Sandip Basu
- Radiation Medicine Centre (BARC), Tata Memorial Hospital Annexe, Mumbai, India
- Radiation Medicine Centre (BARC), Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
37
|
Kratochwil C, Fendler WP, Eiber M, Hofman MS, Emmett L, Calais J, Osborne JR, Iravani A, Koo P, Lindenberg L, Baum RP, Bozkurt MF, Delgado Bolton RC, Ezziddin S, Forrer F, Hicks RJ, Hope TA, Kabasakal L, Konijnenberg M, Kopka K, Lassmann M, Mottaghy FM, Oyen WJG, Rahbar K, Schoder H, Virgolini I, Bodei L, Fanti S, Haberkorn U, Hermann K. Joint EANM/SNMMI procedure guideline for the use of 177Lu-labeled PSMA-targeted radioligand-therapy ( 177Lu-PSMA-RLT). Eur J Nucl Med Mol Imaging 2023; 50:2830-2845. [PMID: 37246997 PMCID: PMC10317889 DOI: 10.1007/s00259-023-06255-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/25/2023] [Indexed: 05/30/2023]
Abstract
Prostate-specific membrane antigen (PSMA) is expressed by the majority of clinically significant prostate adenocarcinomas, and patients with target-positive disease can easily be identified by PSMA PET imaging. Promising results with PSMA-targeted radiopharmaceutical therapy have already been obtained in early-phase studies using various combinations of targeting molecules and radiolabels. Definitive evidence of the safety and efficacy of [177Lu]Lu-PSMA-617 in combination with standard-of-care has been demonstrated in patients with metastatic castration-resistant prostate cancer, whose disease had progressed after or during at least one taxane regimen and at least one novel androgen-axis drug. Preliminary data suggest that 177Lu-PSMA-radioligand therapy (RLT) also has high potential in additional clinical situations. Hence, the radiopharmaceuticals [177Lu]Lu-PSMA-617 and [177Lu]Lu-PSMA-I&T are currently being evaluated in ongoing phase 3 trials. The purpose of this guideline is to assist nuclear medicine personnel, to select patients with highest potential to benefit from 177Lu-PSMA-RLT, to perform the procedure in accordance with current best practice, and to prepare for possible side effects and their clinical management. We also provide expert advice, to identify those clinical situations which may justify the off-label use of [177Lu]Lu-PSMA-617 or other emerging ligands on an individual patient basis.
Collapse
Affiliation(s)
- Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, 45147, Essen, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Klinikum Rechts Der Isar, Technical University Munich (TUM), 81675, Munich, Germany
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Oncology, Sir Peter MacCallum, University of Melbourne, Melbourne, VIC, Australia
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital Sydney, Darlinghurst, Australia
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA, USA
| | - Joseph R Osborne
- Department of Radiology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Amir Iravani
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Phillip Koo
- Division of Diagnostic Imaging, Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| | - Liza Lindenberg
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Richard P Baum
- Curanosticum Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, Wiesbaden, Germany
| | - Murat Fani Bozkurt
- Hacettepe University Faculty of Medicine, Department of Nuclear Medicine, Ankara, Turkey
| | - Roberto C Delgado Bolton
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), Logroño (La Rioja), Spain
| | - Samer Ezziddin
- Department of Nuclear Medicine, Saarland University Medical Center, Homburg, Germany
| | - Flavio Forrer
- Department of Radiology and Nuclear Medicine, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Rodney J Hicks
- The University of Melbourne Department of Medicine, St Vincent's Hospital, Melbourne, Australia
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging / Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Levent Kabasakal
- Department of Nuclear Medicine, Cerrahpasa Medical Faculty, Istanbul University- Cerrahpasa, Istanbul, Turkey
| | - Mark Konijnenberg
- Radiology & Nuclear Medicine Department, Erasmus MC, Rotterdam, The Netherlands
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Technical University Dresden, School of Science, Faculty of Chemistry and Food Chemistry; German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Michael Lassmann
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH Aachen University Medical Faculty, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Wim J G Oyen
- Department of Biomedical Sciences, Humanitas University, and Humanitas Clinical and Research Centre, Department of Nuclear Medicine, Milan, Italy
- Department of Radiology and Nuclear Medicine, Rijnstate Hospital, Arnhem, the Netherlands
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Muenster, Muenster, Germany
| | - Heiko Schoder
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Lisa Bodei
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stefano Fanti
- Division of Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Ken Hermann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, 45147, Essen, Germany
| |
Collapse
|
38
|
Patell K, Kurian M, Garcia JA, Mendiratta P, Barata PC, Jia AY, Spratt DE, Brown JR. Lutetium-177 PSMA for the treatment of metastatic castrate resistant prostate cancer: a systematic review. Expert Rev Anticancer Ther 2023; 23:731-744. [PMID: 37194261 DOI: 10.1080/14737140.2023.2213892] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/10/2023] [Indexed: 05/18/2023]
Abstract
INTRODUCTION Metastatic castrate resistant prostate cancer (mCPRC) remains an aggressive form of prostate cancer that no longer responds to traditional hormonal treatment alone. Despite the advent of novel anti-androgen medications, many patients continue to progress, and as a result, there is a growing need for additional treatment options. AREAS COVERED Lutetium-177 (177Lu) - PSMA-617 has become one of the new frontline treatment options for refractory metastatic castrate resistant prostate cancer after the failure of novel anti-androgen therapy and chemotherapy. Lu-177 has been used in real-world prospective trials and is now becoming utilized in newer phase III clinical trials. Here, we present a comprehensive overview of the current literature, covering retrospective studies, prospective studies, and clinical trials that established Lutetium-177-PSMA-617 (177Lu-PSMA-617) for the treatment of mCRPC. EXPERT OPINION 177Lu - PSMA-617 has been approved for treatment of mCRPC based on positive phase III studies. While this treatment is tolerable and effective, biomarkers are necessary to determine which patients will benefit. In the future, radioligand treatments will likely be utilized in earlier lines of therapy and potentially in combination with other prostate cancer treatments.
Collapse
Affiliation(s)
- Kanchi Patell
- Deparment of Internal Medicine, Division of Medical Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - Matthew Kurian
- Deparment of Internal Medicine, Division of Medical Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - Jorge A Garcia
- Deparment of Internal Medicine, Division of Medical Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Prateek Mendiratta
- Deparment of Internal Medicine, Division of Medical Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Pedro C Barata
- Deparment of Internal Medicine, Division of Medical Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Angela Y Jia
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Jason R Brown
- Deparment of Internal Medicine, Division of Medical Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| |
Collapse
|
39
|
Parker D, Zambelli J, Lara MK, Wolf TH, McDonald A, Lee E, Abou-Elkacem L, Gordon EJ, Baum RP. Case Report: Long-term complete response to PSMA-targeted radioligand therapy and abiraterone in a metastatic prostate cancer patient. Front Oncol 2023; 13:1192792. [PMID: 37188199 PMCID: PMC10175697 DOI: 10.3389/fonc.2023.1192792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
Despite decades of research and clinical trials, metastatic castration-resistant prostate cancer (mCRPC) remains incurable and typically fatal. Current treatments may provide modest increases in progression-free survival but can come with significant adverse effects and are disaggregated from the diagnostic imaging needed to fully assess the spread of metastatic disease. A theranostic approach, using radiolabeled ligands that target the cell surface protein PSMA, simplifies the visualization and disease treatment process by enabling both to use similar agents. Here, we describe an exemplary case wherein a gentleman in his 70s with mCRPC on diagnosis was treated with 177Lu-PSMA-617 and abiraterone, and remains disease-free to date, over five years later.
Collapse
Affiliation(s)
- David Parker
- Private Health Management, Los Angeles, CA, United States
| | | | | | | | - Amber McDonald
- Private Health Management, Los Angeles, CA, United States
| | - Erica Lee
- Private Health Management, Los Angeles, CA, United States
| | | | - Eva J. Gordon
- Private Health Management, Los Angeles, CA, United States
| | | |
Collapse
|
40
|
Broggio D, Célier D, Michel C, Isambert A. Contact restriction time after common nuclear medicine therapies: spreadsheet implementation based on conservative retention function and individual measurements. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2023; 43:021504. [PMID: 36927533 DOI: 10.1088/1361-6498/acc4d1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/16/2023] [Indexed: 06/18/2023]
Abstract
The increasing use of new radiopharmaceuticals invites us to reconsider some radiation protection issues, such as the contact restriction time that limits public exposure by nuclear medicine patients. Contact restriction time should be patient specific and conservative, and its assessment made easy for clinicians. Here a method is proposed based on conservative estimation of the whole-body retention function and at least one measurement of the patient's dose rate. Recommended values of the retention function are given for eight therapies:131I (Graves' disease, remnant ablation, patient follow-up, meta-iodobenzylguanidine),177Lu-prostate-specific membrane antigen and177Lu-DOTATATE therapies, and90Y and166Ho microsphere injection of the liver. The patient line source model for scaling dose rate from one distance to another is included in the restriction time calculation. The method is benchmarked against published values and the influence of the dose rate scaling and whole-body retention function illustrated. A spreadsheet is provided, along with the source code, with recommended values for the eight therapies. The recommended values can be changed as well as the dose rate scaling function, and other radiopharmaceuticals can be included in the spreadsheet provided retention functions are defined.
Collapse
Affiliation(s)
- D Broggio
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SDOS/LEDI, F-92260 Fontenay-aux-Roses, France
| | - D Célier
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SER/UEM, F-92260 Fontenay-aux-Roses, France
| | - C Michel
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SER/UEM, F-92260 Fontenay-aux-Roses, France
| | - A Isambert
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SER/UEM, F-92260 Fontenay-aux-Roses, France
| |
Collapse
|
41
|
Kidney absorbed radiation doses for [ 177 Lu]Lu-PSMA-617 and [ 177 Lu]Lu-PSMA-I&T determined by 3D clinical dosimetry. Nucl Med Commun 2023; 44:270-275. [PMID: 36597884 PMCID: PMC9994818 DOI: 10.1097/mnm.0000000000001658] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE For prostate-specific membrane antigen-directed radioligand therapy (PSMA-RLT), [ 177 Lu]Lu-PSMA-617 and [ 177 Lu]Lu-PSMA-I&T are the currently preferred compounds. Recent preclinical studies suggested ~30x higher kidney absorbed dose for [ 177 Lu]Lu-PSMA-I&T compared to [ 177 Lu]Lu-PSMA-617, which may lead to an increased risk of kidney toxicity. We performed two single-centre, prospective dosimetry studies with either [ 177 Lu]Lu-PSMA-617 or [ 177 Lu]Lu-PSMA-I&T, using an identical dosimetry protocol. We evaluated the absorbed doses of both 177 Lu-labelled radioligands in human kidneys. METHODS 3D SPECT/computed tomography (CT) imaging of the kidneys was performed after PSMA-RLT in cancer patients with PSMA-positive disease and an adequate glomerular filtration rate (≥50 mL/min). Ten metastatic hormone-sensitive prostate cancer patients (mHSPC) were treated with [ 177 Lu]Lu-PSMA-617 and 10 advanced salivary gland cancer (SGC) patients were treated with [ 177 Lu]Lu-PSMA-I&T. SPECT/CT imaging was performed at five timepoints (1 h, 24 h, 48 h, 72 h, and 168 h post-injection). In mHSPC patients, SPECT/CT imaging was performed after cycles 1 and 2 (cumulative activity: 9 GBq) and in SGC patients only after cycle 1 (activity: 7.4 GBq). Kidney absorbed dose was calculated using organ-based dosimetry. RESULTS The median kidney absorbed dose was 0.49 Gy/GBq (range: 0.34-0.66) and 0.73 Gy/GBq (range: 0.42-1.31) for [ 177 Lu]Lu-PSMA-617 and [ 177 Lu]Lu-PSMA-I&T, respectively (independent samples t test; P = 0.010). CONCLUSION This study shows that the kidney absorbed dose for [ 177 Lu]Lu-PSMA-617 and [ 177 Lu]Lu-PSMA-I&T differs, with a ~1.5x higher median kidney absorbed dose for [ 177 Lu]Lu-PSMA-I&T. This difference in the clinical setting is considerably smaller than observed in preclinical studies and may not hamper treatments with [ 177 Lu]Lu-PSMA-I&T.
Collapse
|
42
|
Rathore R, Rangrej SB, Kieme I, Carvalho V, King K, Amadou Y, McKinley J, Masawi A. Treatment of Advanced Metastatic Prostate Cancer Using Molecular-Targeted Therapy: Radioligand Lutetium-177 Prostate-Specific Membrane Antigen. Cureus 2023; 15:e37778. [PMID: 37214073 PMCID: PMC10195000 DOI: 10.7759/cureus.37778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 05/23/2023] Open
Abstract
This study investigates the predicting factors of the biochemical response and survival of patients with advanced metastatic prostate cancer who underwent therapy with radioligand lutetium-177 (177Lu)-prostate-specific membrane antigen (PSMA), often referred to as [177Lu]Lu-PSMA. This study is a review of the previous literature. This study included articles published in the last 10 years in the English language. According to the literature review, treatment with [177Lu]Lu-PSMA has a positive impact on prostate-specific antigen (PSA) within the first cycle and a negative impact on lymph node metastasis. There is a plausible positive impact on PSA after multiple cycles and performance status and a negative impact on visceral metastasis. In conclusion, the reviews show that treatment with [177Lu]Lu-PSMA in patients with castration-resistant prostate cancer is beneficial in reducing PSA and metastasis.
Collapse
Affiliation(s)
- Rajni Rathore
- Pharmacology and Therapeutics, Saint James School of Medicine, Arnos Vale, VCT
| | - Shahid B Rangrej
- Anatomy/Research, Saint James School of Medicine, Arnos Vale, VCT
| | - Ian Kieme
- Medical School, Saint James School of Medicine, Arnos Vale, VCT
| | | | - Katie King
- Medical School, Saint James School of Medicine, Arnos Vale, VCT
| | - Yacoubou Amadou
- Medical School, Saint James School of Medicine, Arnos Vale, VCT
| | - John McKinley
- Medical School, Saint James School of Medicine, Arnos Vale, VCT
| | - Audrey Masawi
- Medical School, Saint James School of Medicine, Arnos Vale, VCT
| |
Collapse
|
43
|
PSMA Receptor-Based PET-CT: The Basics and Current Status in Clinical and Research Applications. Diagnostics (Basel) 2023; 13:diagnostics13010158. [PMID: 36611450 PMCID: PMC9818911 DOI: 10.3390/diagnostics13010158] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 01/05/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA) is a 100 kD, 750 amino acid (AA) long type II transmembrane glycoprotein that has a short N-terminal intracellular domain with 19 AA, 24 AA transmembrane proteins and a large C-terminal extracellular domain with 707 AA. PSMA has been mapped to chromosome 11p 11-12 in the region of the folate hydrolase gene (FOLH1) and has no known natural ligand. The protein possesses enzymatic activity-glutamate carboxypeptidase II (GCP-II)-and is thought to have role in folate uptake (FOLH1 gene). 'PSMA' expression, although significantly up-regulated in prostate carcinoma (more in high-risk and aggressive variants), is not exclusive for it and is noted in various other benign and malignant conditions, especially in the neovasculature. Currently, PSMA PET-CT is approved for high-risk and biochemically recurrent prostate carcinoma (PCa), and in patient selection for PSMA based theranostics. This review aims to highlight the clinical evolution of the PSMA molecule and PSMA PET-CT as a diagnostic modality, various indications of PSMA PET-CT, the appropriateness criteria for its use, pitfalls and artefacts, and other uses of PSMA PET apart from prostate carcinoma.
Collapse
|
44
|
Shin D, Ha S, O JH, Rhew SA, Yoon CE, Kwon HJ, Moon HW, Park YH, Park SY, Park C, Chi DY, Yoo IR, Lee JY. A Single Dose of Novel PSMA-Targeting Radiopharmaceutical Agent [ 177Lu]Ludotadipep for Patients with Metastatic Castration-Resistant Prostate Cancer: Phase I Clinical Trial. Cancers (Basel) 2022; 14:cancers14246225. [PMID: 36551710 PMCID: PMC9777064 DOI: 10.3390/cancers14246225] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/10/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
[177Lu]Ludotadipep, which enables targeted delivery of beta-particle radiation to prostate tumor cells, had been suggested as a promising therapeutic option for mCRPC. From November 2020 to March 2022, a total of 30 patients were enrolled for single dose of [177Lu]Ludotadipep RPT, 6 subjects in each of the 5 different activity groups of 1.9 GBq, 2.8 GBq, 3.7 GBq, 4.6 GBq, and 5.6 GBq. [177Lu]Ludotadipep was administered via venous injection, and patients were hospitalized for three days to monitor for any adverse effects. Serum PSA levels were followed up at weeks 1, 2, 3, 4, 6, 8, and 12, and PSMA PET/CT with [18F]Florastamin was obtained at baseline and again at weeks 4 and 8. The subjects required positive PSMA PET/CT prior to [177Lu]Ludotadipep administration. Among the 29 subjects who received [177Lu]Ludotadipep, 36 treatment emergent adverse events (TEAEs) occurred in 17 subjects (58.6%) and 4 adverse drug reactions (ADRs) in 3 subjects (10.3%). Of the total 24 subjects who had full 12-week follow-up data, 16 (66.7%) showed decrease in PSA of any magnitude, and 9 (37.5%) showed a decrease in PSA by 50% or greater. A total of 5 of the 24 patients (20.8%) showed disease progression (PSA increase of 25% or higher from the baseline) at the 12th week following single dose of [177Lu]Ludotadipep. These data thus far suggest that [177Lu]Ludotadipep could be a promising RPT agent with low toxicity in mCRPC patients who have not been responsive to conventional treatments.
Collapse
Affiliation(s)
- Dongho Shin
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seunggyun Ha
- Department of Nuclear Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Joo Hyun O
- Department of Nuclear Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seung ah Rhew
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Chang Eil Yoon
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hyeok Jae Kwon
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hyong Woo Moon
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Yong Hyun Park
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sonya Youngju Park
- Department of Nuclear Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Chansoo Park
- Research Institute of Labeling, FutureChem Co., Ltd., Seoul 04793, Republic of Korea
| | - Dae Yoon Chi
- Research Institute of Labeling, FutureChem Co., Ltd., Seoul 04793, Republic of Korea
| | - Ie Ryung Yoo
- Department of Nuclear Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Correspondence: (I.R.Y.); (J.Y.L.); Tel./Fax: +82-2-2258-1401 (I.R.Y.); +82-2-2258-1401 (J.Y.L.)
| | - Ji Youl Lee
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Correspondence: (I.R.Y.); (J.Y.L.); Tel./Fax: +82-2-2258-1401 (I.R.Y.); +82-2-2258-1401 (J.Y.L.)
| |
Collapse
|
45
|
Oh SW, Suh M, Cheon GJ. Current Status of PSMA-Targeted Radioligand Therapy in the Era of Radiopharmaceutical Therapy Acquiring Marketing Authorization. Nucl Med Mol Imaging 2022; 56:263-281. [PMID: 36425273 PMCID: PMC9679068 DOI: 10.1007/s13139-022-00764-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/11/2022] [Accepted: 07/11/2022] [Indexed: 10/15/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA) is highly expressed in PCa, which gradually increases in high-grade tumors, metastatic tumors, and tumors nonresponsive to androgen deprivation therapy. PSMA has been a topic of interest during the past decade for both diagnostic and therapeutic targets. Radioligand therapy (RLT) utilizes the delivery of radioactive nuclides to tumors and tumor-associated targets, and it has shown better efficacy with minimal toxicity compared to other systemic cancer therapies. Nuclear medicine has faced a new turning point claiming theranosis as the core of academic identity, since new RLTs have been introduced to clinics through the official new drug development processes for approval from the Food and Drug Administration (FDA) or European Medical Agency. Recently, PSMA targeting RLT was approved by the US FDA in March 2022. This review introduces PSMA RLT focusing on ongoing clinical trials to enhance our understanding of nuclear medicine theranosis and strive for the development of new radiopharmaceuticals.
Collapse
Affiliation(s)
- So Won Oh
- Department of Nuclear Medicine, Seoul National University Boramae Medical Center, Seoul, 07061 Korea
| | - Minseok Suh
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, 03080 Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080 Korea
| |
Collapse
|
46
|
van der Gaag S, Bartelink IH, Vis AN, Burchell GL, Oprea-Lager DE, Hendrikse H. Pharmacological Optimization of PSMA-Based Radioligand Therapy. Biomedicines 2022; 10:3020. [PMID: 36551776 PMCID: PMC9775864 DOI: 10.3390/biomedicines10123020] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/09/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Prostate cancer (PCa) is the most common malignancy in men of middle and older age. The standard treatment strategy for PCa ranges from active surveillance in low-grade, localized PCa to radical prostatectomy, external beam radiation therapy, hormonal treatment and chemotherapy. Recently, the use of prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT) for metastatic castration-resistant PCa has been approved. PSMA is predominantly, but not exclusively, expressed on PCa cells. Because of its high expression in PCa, PSMA is a promising target for diagnostics and therapy. To understand the currently used RLT, knowledge about pharmacokinetics (PK) and pharmacodynamics (PD) of the PSMA ligand and the PSMA protein itself is crucial. PK and PD properties of the ligand and its target determine the duration and extent of the effect. Knowledge on the concentration-time profile, the target affinity and target abundance may help to predict the effect of RLT. Increased specific binding of radioligands to PSMA on PCa cells may be associated with better treatment response, where nonspecific binding may increase the risk of toxicity in healthy organs. Optimization of the radioligand, as well as synergistic effects of concomitant agents and an improved dosing strategy, may lead to more individualized treatment and better overall survival.
Collapse
Affiliation(s)
- Suzanne van der Gaag
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| | - Imke H. Bartelink
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - André N. Vis
- Department of Urology, Prostate Cancer Network Amsterdam, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - George L. Burchell
- Medical Library, VU University, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Daniela E. Oprea-Lager
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| | - Harry Hendrikse
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
47
|
Hertz B, Watabe T, Baum RP. Celebrating 80 years anniversary of radioiodine for use in thyroid cancer and perspectives for theranostics. Ann Nucl Med 2022; 36:1007-1009. [DOI: 10.1007/s12149-022-01806-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022]
|
48
|
Kara A. An optimization study for targeted alpha therapy: Ion behaviours and dose calculations within ICRU-compact bone tissue. Appl Radiat Isot 2022; 191:110552. [DOI: 10.1016/j.apradiso.2022.110552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/27/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
|
49
|
Response to [ 177 Lu]Lu-PSMA radioligand therapy in metastatic castration-resistant prostate cancer patients presenting with only lymph node metastases. Nucl Med Commun 2022; 43:1113-1120. [PMID: 36120814 PMCID: PMC9575560 DOI: 10.1097/mnm.0000000000001611] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE [ 177 Lu]Lu-PSMA radioligand therapy (PSMA-RLT) is a promising therapy for patients with metastatic castration-resistant prostate cancer (mCRPC) and offers a survival benefit particularly to patients with only lymph node metastases. We therefore sought to evaluate the clinical outcome of this therapy in such a cohort. METHODS Of all prostate cancer patients admitted to our department between September 2015 and March 2019 to receive 1-4 courses of PSMA-RLT (each course consisted of three cycles of highly standardized PSMA-RLT every 4 weeks), only 10 consecutive men were found to have nodal metastases only and were analyzed retrospectively. RESULTS Nine out of 10 patients responded to their first PSMA-RLT course with a mean prostate-specific antigen (PSA) decline of 71.8 ± 25.2%, seven of them demonstrated a PSA decline of ≥50%. Collectively, seven of eight patients responded to further PSMA-RLT courses with a total PSA reduction of 59.8 ± 30.0%, five of which showed a PSA reduction of ≥50%. One patient experienced complete remission. Median progression-free survival was 85 weeks (range 14-255 weeks) and median overall survival was not reached during the median observation time of 209 weeks (30-298 weeks). Univariate Cox-regression identified initial PSA decline as the only predictive parameter for progression-free survival ( P = 0.047). CONCLUSION mCRPC patients with only lymph node metastases showed favorable survival and excellent response to PSMA-RLT, leading to transient partial remission of the disease in most of them.
Collapse
|
50
|
Sun J, Huangfu Z, Yang J, Wang G, Hu K, Gao M, Zhong Z. Imaging-guided targeted radionuclide tumor therapy: From concept to clinical translation. Adv Drug Deliv Rev 2022; 190:114538. [PMID: 36162696 DOI: 10.1016/j.addr.2022.114538] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 09/03/2022] [Accepted: 09/11/2022] [Indexed: 01/24/2023]
Abstract
Since the first introduction of sodium iodide I-131 for use with thyroid patients almost 80 years ago, more than 50 radiopharmaceuticals have reached the markets for a wide range of diseases, especially cancers. The nuclear medicine paradigm also shifts from solely molecular imaging or radionuclide therapy to imaging-guided radionuclide therapy, which is deemed a vital component of precision cancer therapy and an emerging medical modality for personalized medicine. The imaging-guided radionuclide therapy highlights the systematic integration of targeted nuclear diagnostics and radionuclide therapeutics. Regarding this, nuclear imaging serves to "visualize" the lesions and guide the therapeutic strategy, followed by administration of a precise patient specific dose of radiotherapeutics for treatment according to the absorbed dose to different organs and tumors calculated by dosimetry tools, and finally repeated imaging to predict the prognosis. This strategy leads to significantly enhanced therapeutic efficacy, improved patient outcomes, and manageable adverse events. In this review, we provide an overview of imaging-guided targeted radionuclide therapy for different tumors such as advanced prostate cancer and neuroendocrine tumors, with a focus on development of new radioligands and their preclinical and clinical results, and further discuss about challenges and future perspectives.
Collapse
Affiliation(s)
- Juan Sun
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhenyuan Huangfu
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Jiangtao Yang
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China.
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan.
| | - Mingyuan Gao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|