PSMA-Targeted Radionuclide Therapy for Prostate Cancer: Comparison
Please note this is a comparison between Version 2 by Nora Tang and Version 1 by Chalermrat Kaewput.

https://www.mdpi.com/2077-0383/11/10/2738There is now an increasing trend for targeting cancers to go beyond early diagnosis and actually improve Progression-Free Survival and Overall Survival. Identifying patients who might benefit from a particular targeted treatment is the main focus for Precision Medicine. Radiolabeled ligands can be used as predictive biomarkers which can confirm target expression by cancers using positron emission tomography (PET). The same ligand can subsequently be labeled with a therapeutic radionuclide for targeted radionuclide therapy. This combined approach is termed “Theranostics”. The prostate-specific membrane antigen (PSMA) has emerged as an attractive diagnostic and therapeutic target for small molecule ligands in prostate cancer. It can be labeled with either positron emitters for PET-based imaging or beta and alpha emitters for targeted radionuclide therapy. 

  • PSMA
  • prostate cancer
  • theranostics
  • PET/CT
  • radionuclide therapy

1. PSMA-Targeted Radioligand Therapy

1.1.

177

Lu-PSMA Radioligand Therapy

Lutetium-177 (177Lu) is a beta-emitting radioisotope with a half-life of 6.7 days. It has an average energy of 133.6 keV with a maximum penetration depth of <2 mm [101][1]. Because of its good physical properties and the possibility of post-treatment imaging, 177Lu labeled PSMA has been extensively studied and it has emerged as a novel treatment for mCRPC. Until recently, the majority of PSMA-targeting tracers involve urea-based agents (small molecule inhibitors) including 177Lu-PSMA-I&T (imaging and therapy) and 177Lu-PSMA-617, with the latter being preferred due to lower renal uptake [102,103][2][3]. A previous study of Ruigrok et al. revealed that although 177Lu-PSMA-617 and 177Lu-PSMA-I&T show similar binding characteristics in prostate tumors, 177Lu PSMA-I&T has a lower tumor-to-kidney ratio than 177Lu PSMA-617 [104][4].
Protecting the kidneys and salivary and lacrimal glands is the major challenge for PSMA-targeting radiotracers due to high uptake in these organs. Salivary gland toxicity has been documented as dose-limiting. This significantly reduced the quality of life of treated patients. Recently, there have been more reports and publications focusing on the safety and efficacy of 177Lu-PSMA radioligand therapy.
The systematic review and meta-analysis of Yadav et al. focused on 17 retrospective/prospective reports, totaling 744 patients treated with 177Lu PSMA-radioligand therapy and concluded that 75% of patients had a reduction of PSA levels, with 46% having a reduction of more than 50%. A radiographic partial remission was seen in 37%, median overall survival (OS) was 13.8 months, and median progression-free survival (PFS) was 11 months. The most common treatment-related side effects were myelosuppression, nephrotoxicity, and salivary gland toxicity (pain, swelling, and dry mouth) [21][5].
 
(a)
177Lu-PSMA-617
The LuPSMA trial (prospective single-arm, single-center, phase II trial) was conducted between August 2015 and December 2016. Forty men with PSMA-avid metastatic CRPC were treated with 177Lu-PSMA-617 (7.5 GBq/cycle). Seventeen (57%) of thirty patients had a reduction in PSA of 50% or more. There were no treatment-related deaths. The most common adverse effect associated with 177Lu-PSMA-617 was grade 1 dry mouth (87%), and grade 3 or 4 thrombocytopenia that may have been caused by 177Lu-PSMA-617 occurred in 13%. Objective response in LN or visceral disease was found in 82% of patients. Median PSA progression was 7.6 months, and median OS was 13.5 months [105][6].
The TheraP study (randomized phase 2 clinical trial at 11 centers in Australia) was conducted between February 2018 and September 2019 with 200 mCRPC patients. Treatment in the study was given to 98 (99%) of 99 men randomly selected to 177Lu-PSMA-617 (6.0–8.5 GBq intravenously every 6 weeks for up to 6 cycles) compared to 85 (84%) of 101 randomly assigned to cabazitaxel (20 mg/m2). PSA responses were more common among men in the 177Lu-PSMA-617 group than in the cabazitaxel group. Grade 3–4 adverse events occurred in 32 of 98 men (33%) in the 177Lu-PSMA-617 group versus 45 of 85 men (53%) in the cabazitaxel group. No deaths were found in the 177Lu-PSMA-617 group [17][7].
The VISION study was a phase 3 randomized trial from June 2018 to mid-October 2019. The study enrolled 831 mCRPC patients with a median follow-up of 20.9 months. 177Lu-PSMA-617 (7.4 GBq every 6 weeks × 6 cycles) combined standard of care (SOC) compared to SOC alone and revealed significant improvement in OS by a median of 4.0 months and PFS based on imaging was significantly longer. The incidence of grade 3 or above adverse events was higher with 177Lu-PSMA-617 than the other arm but the quality of life was not affected. Due to the favorable treatment outcome with low incidence of adverse events in this study, the promotion of 177Lu-PSMA-617 as a standard protocol in advanced PSMA-positive mCRPC is suggested [13][8].
The LuTectomy trial (open label, phase 1/2, non-randomized clinical trial) evaluated the dosimetry, efficacy, and toxicity of 177Lu-PSMA in men with high PSMA-expressing high-risk localized or locoregional advanced PCa who underwent radical prostatectomy (RP) and pelvic lymph node dissection (PLND). This study started enrollment in August 2020 and is expected to be completed in June 2023 (NCT04430192) [106][9].
The UpFrontPSMA trial (open label, randomized, phase 2 trial) of sequential 177Lu-PSMA 617 and docetaxel Versus docetaxel alone in 140 newly diagnosed metastatic PCa. The objective of this study was to evaluate response to treatment by measuring PSA levels and radiological response and safety of 177Lu-PSMA 617. This study started enrollment in April 2020 and is expected to be completed in April 2024 (NCT04343885) [107][10].
 
(b)
177Lu-PSMA-I&T
177Lu-labeled PSMA ligand (DOTAGA-(I-y) fk (Sub-KuE), also known as PSMA I&T, for “imaging and therapy”) is now considered essential for the treatment of advanced PCa [98][11]. PSMA-I&T was first developed in Germany in 2015. There are similar properties between 177Lu-PSMA-I&T and 177Lu-PSMA-617. In a study of 56 mCRPC patients who received an average dose of 5.76 GBq per cycle (total of 125 cycles), the PSA PFS was approximately 14 months with 59% of patients having >50% reduction in PSA levels [108][12].
In a previous large cohort study in 2019 involving100 patients treated with a total 319 cycles of 177Lu-PSMA-I&T (median 2 cycles, range 1–6), 6–8 weekly with mean activity of 7.4 GBq, PSA decreased ≥50% within 12 weeks of treatment. Longer PFS and OS was observed in 38 patients, PFS was 4.1 mo and OS was 12.9 mo. Hematologic grade 3/4 toxicities were anemia (9%), thrombocytopenia (4%), and neutropenia (6%). Grade 3/4 non-hematologic toxicities were not found [109][13].
The SPLASH trial (a phase 3, open-label, randomized study) evaluated the efficacy of 177Lu PSMA-I&T (AKA. 177Lu-PNT2002) versus abiraterone or enzalutamide in slowing the progression of radiographic findings in mCRPC patients. The study will begin with a safety and dosimetry arm involving 25 patients (Part 1) and Part 2 involves a randomized therapeutic phase in 390 patients. All patients will have long-term follow-up for at least 5 years, death, or loss to follow-up (Part 3). This study began enrollment in March 2021 with approximately 415 patients (NCT04647526) [110][14].
 
(c)
Combination of 177Lu-PSMA 617 with androgen receptor-axis-targeted therapies (ARAT)
ARAT was only approved for patients with metastatic castration. Systemic ARAT is now FDA-approved even in PCa without evidence of mCRPC. ARAT administration increases PSMA expression. In addition, ARAT may cause radiosensitization. It is hypothesized that combined 177Lu-PSMA radioligand treatment and ARAT might result in better tumor control in patients with CRPC [111][15].
The PSMAddition study is an international open-label, randomized, phase III study in 1126 metastatic hormone-sensitive PCa patients. The objective of this study was to evaluate the efficacy and safety of 177Lu-PSMA-617 combined with SOC, versus SOC alone. The SOC was determined as a combination of ARAT with ADT. Participants received approximately 7.4 GBq of 177Lu-PSMA-617, every 6 weeks for 6 planned cycles. The primary outcome was to assess radiographic PFS (rPFS) and estimated final OS analysis. This study started enrollment in June 2021 and is expected to be completed in December 2025 (NCT04720157) [112][16].
The ENZA-p trial (open label, randomized phase 2, multicenter) was to compare the efficacy and safety of 160 mg enzalutamide daily + 177Lu-PSMA-617 (up to 4 cycles of 7.5 GBq) versus enzalutamide alone in 160 mCRPC patients who were deemed at high-risk for early failure on enzalutamide monotherapy. This study started enrollment in August 2020 and is expected to be completed in June 2023 (NCT04419402) [113][17].
Even though there is a biological basis to promote combined ARAT and 177Lu-PSMA 617, key questions to answer will include the optimal dose, period of administration, and long-term safety of use of 177Lu-PSMA-617 in the early stage of subsequent lines of treatment.
 
(d)
Combination of 177Lu PSMA-617 with DNA damage repair inhibitor
Transcription active sites are often unstable and susceptible to breakage since the torsional stress and local depletion of nucleosomes allow DNA to be more accessible to damaging substances. A dedicated DNA damage response (DDR) is necessary to preserve genome integrity at the exposed sites. The DDR is a complex system of DNA damage sensor proteins, for example, the poly (ADP-ribose) polymerase 1 (PARP-1). It is important in repairing radiation-induced single-stranded DNA breaks, which permit cancer cells to become resistant to radiation [114][18]. Therefore, PARP-1 inhibition may cause radiosensitization when combined with radioligand treatment. PARP inhibitors, such as olaparib and rucaparib, have been described as effective against mCRPC [106,107][9][10].
LuPARP is an Australian phase 1, open-label, multicenter, dose-escalation and dose-expansion study to investigate the use of 177Lu-PSMA 617 with olaparib in 52 mCRPC patients who have progressed on novel AR targeted drugs and have PSMA-avid disease on imaging. Patients received fixed 7.4 GBq of 177Lu-PSMA-617 every 6 weeks plus olaparib. The primary endpoints were dose-limiting toxicity and maximum tolerated dose. Secondary endpoints were adverse events, rPFS, and OS. This study started enrollment in July 2019 and is expected to be completed in October 2022 (NCT03874884) [115][19].
 
(e)
Combination of 177Lu PSMA-617 with checkpoint inhibitor immunotherapy
Immunotherapy has changed the therapeutic strategy of many hematological and solid cancers. However, several phase I and II trials evaluating programmed death receptor 1 (PD-1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors have reported limited usage for mCRPC. Additionally, although sipuleucel-T represents the only FDA-approved cancer vaccine for mCRPC according to IMPACT trial results [116][20], its use is relatively limited in daily clinical practice. There are several ongoing clinical trials in various immunotherapy approaches either alone or combined with other therapies in mCRPC. Different mAb against PD-1, PD-L1, or CTLA-4 were tested for mCRP treatment with unsatisfactory results [117][21].
PRINCE is the phase Ib/II study of 177Lu-PSMA-617 combined with pembrolizumab for treatment in 37 mCRPC patients who have progressed on ARAT. Patients with PSMA-avid disease were enrolled to receive 177Lu-PSMA for up to 6 doses (starting at 8.5 GBq with a decrease of 0.5 GBq for each cycle) and pembrolizumab for up to 35 cycles (every 3 weeks). The primary objectives were PSA response rate and safety. Secondary objectives were rPFS, PSA-PFS, and OS. This study started recruitment in July 2019 and was completed in October 2021 (NCT03658447) [118][22].

1.2. Alpha-Emitting PSMA-Targeted Radioligand Therapy

For mCRPC treatment, multiple studies have confirmed that 177Lu-PSMA-617 has a favorable dosimetry and good objective response, including improvement in PSA levels and radiological findings [119,120][23][24]. However, approximately 30% of patients did not respond to 177Lu-labeled PSMA ligands. Targeted α-radiation therapy may be more effective for mCRPC treatment and has been reported in a limited number of patients to be effective in patients resistant to 177Lu-PSMA-617 therapy. Despite having good tolerability, high radioactivity accumulations in bone metastases which lie near to or within the red marrow, indicate that the actual absorbed dose to some domains of active marrow may be rather higher than estimated in the previous study due to spillover resulting in associated developmental risk factor for hematologic toxicity. Recent studies have shown that targeted α-radiation therapy is highly beneficial for mCRPC patients in this setting [121][25].
 
(a)
225Ac-PSMA-617
225Ac is an alpha emitter with a relatively long half-life (9.9 days) [122,123][26][27]. Actinium-225 (225Ac)-PSMA-617 has a significantly higher linear energy transfer (LET) compared to beta particles. With reference to a preliminary report from a single-center study, 225Ac-PSMA-617 (100 kBq/kg every two weeks) resulted in decreased PSA levels below measurable levels and showed complete response on imaging. No related hematologic toxicity was found. Xerostomia was the only clinical side effect mentioned [124][28]. In the previous study of Kratochwil et al. of fourteen PCa patients with metastasis, therapeutic activity of 100 kBq/kg of 225Ac PSMA-617 per cycle repeated every eight weeks demonstrated proper trade-off between toxicity and biochemical response. Low grade hematological toxicity was found in six patients and xerostomia was found in eight patients. Xerostomia was the dose-limiting factor with 100 kBq/kg considered the maximum tolerated dose [125][29]. A recent study of Sathekge M et al. assessed the therapeutic outcome of 225Ac-PSMA-617 in seventeen advanced PCa patients of which the results showed a good anti-tumoral effect assessed by serum PSA level and 68Ga-PSMA-PET/CT as seen in 94.1% of patients. A PSA decrease by ≥90% after treatment was found in 82.4% of patients. All patients experienced grade 1/2 xerostomia without severe symptoms [126][30]. Although targeted α-therapy with 225Ac-PSMA-617 is still considered experimental, it clearly has the potential to be of great benefit to advanced-stage PCa patients.
 
(b)
213Bi-labeled PSMA-617
Bismuth-213 (213Bi) is a short half-life (45.6 min) mixed alpha and beta emitting agent [123,127][27][31]. Sathekge et al. reported a 1st treatment case with 213Bi-PSMA-617 (two cycles with a cumulative activity of 592 MBq) in mCRPC patients who had progressed under conventional therapy gained PSMA-imaging response and biochemical response with a declined PSA level from 237 µg/L to 43 µg/L [128][32]. The previous study of Kratochwil et al. revealed that dosimetry of 213Bi-PSMA-617 is suitable for clinical application. However, when compared with 225Ac-PSMA-617, it suffers from higher perfusion-dependent off-target radiation, and a biological half-life of PSMA-617 in dose-limiting organs is longer than the physical half-life of 213Bi, making this agent a second-choice radiolabel for the targeted alpha therapy of PCa [129][33].

2. Anti-PSMA Radioimmunotherapy

J591 was the first monoclonal antibody (mAb) targeting the extracellular domain of PSMA that was derived from the original murine J591 (muJ591) by substituting epitopes of the B and T cell [130,131][34][35]. This led to differences in kinetics and biodistribution, the adverse events observed following two agents were often different. The prospective study of Tagawa concluded that the PSMA-targeted 177Lu antibody J591 was related with greater hematologic effects than with PSMA-617. However, 177Lu PSMA-617 is related with more non-hematological toxicity than 177Lu-J591. 177Lu-PSMA-617 was observed to be related with more PSA decline than 177Lu-J591, but no difference in OS was found on multivariable analysis [132][36]. The difference between mAb and Ligand are described in Table 41.
2 was defined as the single-dose maximum tolerated dose. The biologic activity was observed in four patients with >50% reduction in PSA levels for between 3 and 8 months. No patient developed a human anti-J591 antibody response to deimmunized J591 [133][37]. In a phase 2 study of Tagawa et al., single doses of 177Lu-J591 were studied in forty-seven patients with progression after hormonal treatment. A total of 10.6% experienced ≥50% reduction in PSA, 36.2% experienced ≥30% decline, and 59.6% experienced any PSA decline after single treatment. Median OS was approximately 22 vs. 12 months in mCRPC treated with a single-dose of 70 mCi/m2 and 65 mCi/m2, respectively. However, a higher dose resulted in a higher grade of thrombocytopenia and neutropenia [134][38]. In a phase 1/2 study, forty-nine mCRPC patients received 177Lu-J591 doses ranging between 20 and 45 mCi/m2 × 2 two weeks apart. The recommended doses at phase 2 were 40 mCi/m2 and 45 mCi/m2 × 2. Median OS was 42.3 months vs. 19.6 months in patients receiving higher vs. lower doses. The higher grade of thrombocytopenia and neutropenia were found in higher doses than lower doses [135][39].

2.2.

225

Ac-J591 Antibody

The reason for higher hematological toxicity for 177Lu-J591 is possibly because mAb is a large molecule with slower circulating clearance compared to smaller molecules, for example, PSMA-617. However, compared to PSMA-617, J591 shows no uptake in the salivary gland or kidneys [136][40]. Because of this character, it has been hypothesized that 225Ac-J591 can reduce the incidence of salivary and renal toxicity found in 225Ac-PSMA-617. In the previous phase 1 trial of Tagawa, twenty-two mCRPC patients received seven dose levels of 225Ac-J591, and only one patient who received 80 KBq/kg had grade 4 thrombocytopenia and anemia. Xerostomia was seen only in patients receiving 177Lu-PSMA-617 treatment. PSA reduction was observed in 64% with 41% of patients having ≥50% PSA reduction (Clinical trial information: NCT03276572) [137][41].

2.3.

227

TH-PSMA-TTC Antibody

PSMA-Targeted Thorium-227 Conjugate (PSMA-TTC) is a targeted alpha therapy for PCa. Thorium-227 (227Th) is a long half-life (18.7 days) α-particle emitter which is the parent nuclide of radium-223. 227Th-PSMA-TTC is a novel human antibody attached to 227Th and has shown very strong antitumor efficacy in animal models with PCa [138][42]. A phase I clinical trial of PSMA-TTC in mCRPC is currently ongoing based on promising preclinical data. It is planned with a recruitment of one hundred ninety-eight patients and is expected to be completed in September 2024 [139][43] (NCT03724747). Clinical trials of PSMA-targeted radioligand therapy is described in Table 52.

2.1.

177

Lu-J591 Antibody

J591 is typically labeled with lutetium-177 (177Lu-J591) to target PCa cells and deliver radioimmunotherapy. It has been studied extensively in phase I/II clinical trials. 177Lu-J591 was first studied in a phase I trial in 2005 in 35 mCRPC patients receiving up to three doses. Myelosuppression was a dose limited to 75 mCi/m2, and a dose level of 70-mCi/m

References

  1. Emmett, L.; Willowson, K.; Violet, J.; Shin, J.; Blanksby, A.; Lee, J. Lutetium 177 PSMA radionuclide therapy for men with prostate cancer: A review of the current literature and discussion of practical aspects of therapy. J. Med. Radiat. Sci. 2017, 64, 52–60.
  2. Zhang, H.; Koumna, S.; Pouliot, F.; Beauregard, J.M.; Kolinsky, M. PSMA Theranostics: Current Landscape and Future Outlook. Cancers 2021, 13, 4023.
  3. Ruigrok, E.A.M.; van Weerden, W.M.; Nonnekens, J.; de Jong, M. The future of PSMA-targeted radionuclide therapy: An overview of recent preclinical research. Pharmaceutics 2019, 11, 560.
  4. Ruigrok, E.A.; van Vliet, N.; Dalm, S.U.; de Blois, E.; van Gent, D.C.; Haeck, J.; de Ridder, C.; Stuurman, D.; Konijnenberg, M.W.; van Weerden, W.M.; et al. Extensive preclinical evaluation of lutetium-177-labeled PSMA-specific tracers for prostate cancer radionuclide therapy. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 1339–1350.
  5. Yadav, M.P.; Ballal, S.; Sahoo, R.K.; Dwivedi, S.N.; Bal, C. Radioligand Therapy with 177Lu-PSMA for Metastatic Castration-Resistant Prostate Cancer: A Systematic Review and Meta-Analysis. AJR 2019, 213, 275–285.
  6. Hofman, M.S.; Violet, J.; Hicks, R.J.; Ferdinandus, J.; Thang, S.P.; Akhurst, T.; Iravani, A.; Kong, G.; Kumar, A.R.; Murphy, D.G.; et al. -PSMA-617 radionuclide treatment in patients with metastatic castration-resistant prostate cancer (LuPSMA trial): A single-centre, single-arm, phase 2 study. Lancet Oncol. 2018, 19, 825–833.
  7. Hofman, M.; Emmett, L.; Sandhu, S.; Irvani, A.; Joshua, A.M.; Goh, J.C.; Pattison, D.A.; Tan, T.H.; Kirkwood, I.D.; Ng, S.; et al. 177Lu-PSMA-617 versus cabazitaxel in patients with metastatic castration-resistant prostate cancer (TheraP): A randomised, open-label, phase 2 trial. Lancet 2021, 397, 797–804.
  8. Sartor, O.; De Bono, J.; Chi, K.N.; Fizazi, K.; Herrmann, K.; Rahbar, K.; Tagawa, S.T.; Nordquist, L.T.; Vaishampayan, N.; El-Haddad, G.; et al. Lutetium-177-PSMA-617 for metastatic castration-resistant prostate cancer. N. Engl. J. Med. 2021, 385, 1091–1103.
  9. Dosimetry, Safety and Potential Benefit of 177Lu-PSMA-617 Prior to Prostatectomy (LuTectomy). Available online: https://clinicaltrials.gov/ct2/show/NCT04430192?term=Lutectomy&draw=2&rank=1 (accessed on 1 May 2022).
  10. In Men With Metastatic Prostate Cancer, What Is the Safety and Benefit of Lutetium-177 PSMA Radionuclide Treatment in Addition to Chemotherapy (UpFrontPSMA). Available online: https://clinicaltrials.gov/ct2/show/NCT04343885?term=UpFrontPSMA&draw=2&rank=1 (accessed on 1 May 2022).
  11. Alesini, D.; Iacovelli, R.; Palazzo, A.; Altavilla, A.; Risi, E.; Urbano, F.; Manai, C.; Passaro, A.; Magri, V.; Cortesi, E. Multimodality treatment of gynecomastia in patients receiving antiandrogen therapy for prostate cancer in the era of abiraterone acetate and new antiandrogen molecules. Oncology 2013, 84, 92–99.
  12. Weineisen, M.; Schottelius, M.; Simecek, J.; Baum, R.P.; Yildiz, A.; Beykan, S.; Kulkarni, H.R.; Lassmann, M.; Klette, I.; Eiber, M.; et al. 68Ga- and 177Lu-Labeled PSMA I&T: Optimization of a PSMA-Targeted Theranostic Concept and First Proof-of-Concept Human Studies. J. Nucl. Med. 2015, 56, 1169–1176.
  13. Baum, R.P.; Kulkarni, H.R.; Schuchardt, C.; Singh, A.; Wirtz, M.; Wiessalla, S.; Schottelius, M.; Mueller, D.; Klette, I.; Wester, H.J. 177Lu-Labeled Prostate-Specific Membrane Antigen Radioligand Therapy of Metastatic Castration-Resistant Prostate Cancer: Safety and Efficacy. J. Nucl. Med. 2016, 57, 1006–1013.
  14. Study Evaluating mCRPC Treatment Using PSMA -PNT2002 Therapy after Second-Line Hormonal Treatment (SPLASH). Available online: https://www.clinicaltrials.gov/ct2/show/NCT04647526 (accessed on 19 September 2021).
  15. Shah, H.; Vaishampayan, U. Therapy of Advanced Prostate Cancer: Targeting the Androgen Receptor Axis in Earlier Lines of Treatment. Target. Oncol. 2018, 13, 679–689.
  16. An International Prospective Open-Label, Randomized, Phase III Study Comparing 177Lu-PSMA-617 in Combination With Soc, Versus SoC Alone, in Adult Male Patients With mHSPC (PSMAddition). Available online: https://www.clinicaltrials.gov/ct2/show/NCT04720157 (accessed on 21 October 2021).
  17. Enzalutamide with Lu PSMA-617 versus Enzalutamide Alone in Men with Metastatic Castration-Resistant Prostate Cancer (ENZA-p). Available online: https://clinicaltrials.gov/ct2/show/NCT04419402?term=NCT04419402&draw=2&rank=1 (accessed on 1 May 2022).
  18. Wengner, A.M.; Scholz, A.; Haendler, B. Targeting DNA Damage Response in Prostate and Breast Cancer. Int. J. Mol. Sci. 2020, 21, 8273.
  19. 177Lu-PSMA-617 Therapy and Olaparib in Patients with Metastatic Castration Resistant Prostate Cancer (LuPARP). Available online: https://clinicaltrials.gov/ct2/show/NCT03874884 (accessed on 21 October 2021).
  20. Kantoff, P.W.; Higano, C.S.; Shore, N.D.; Berger, E.R.; Small, E.J.; Penson, D.F.; Redfern, C.H.; Ferrari, A.C.; Dreicer, R.; Sims, R.B.; et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 2010, 363, 411–422.
  21. Rizzo, A.; Mollica, V.; Cimadamore, A.; Santoni, M.; Scarpelli, M.; Giunchi, F.; Cheng, L.; Lopez-Beltran, A.; Fiorentino, M.; Montironi, R.; et al. Is There a Role for Immunotherapy in Prostate Cancer? Cells 2020, 9, 2051.
  22. PRINCE (PSMA-Lutetium Radionuclide Therapy and ImmuNotherapy in Prostate CancEr). Available online: https://clinicaltrials.gov/ct2/show/NCT03658447 (accessed on 21 October 2021).
  23. Delker, A.; Fendler, W.P.; Kratochwil, C.; Brunegraf, A.; Gosewisch, A.; Gildehaus, F.J.; Tritschler, S.; Stief, C.G.; Kopka, K.; Haberkorn, U.; et al. Dosimetry for 177Lu-DKFZ-PSMA-617: A new radiopharmaceutical for the treatment of metastatic prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, 42–51.
  24. Kabasakal, L.; AbuQbeitah, M.; Aygün, A.; Yeyin, N.; Ocak, M.; Demirci, E.; Toklu, T. Pre-therapeutic dosimetry of normal organs and tissues of 177Lu-PSMA-617 prostate-specific membrane antigen (PSMA) inhibitor in patients with castration-resistant prostate cancer. Eur. J. Nucl. Med. Mol Imaging 2015, 42, 1976–1983.
  25. Feuerecker, B.; Tauber, R.; Knorr, K.; Heck, M.; Beheshti, A.; Seidl, C.; Bruchertseifer, F.; Pickhard, A.; Gafita, A.; Kratochwil, C.; et al. Activity and Adverse Events of Actinium-225-PSMA-617 in Advanced Metastatic Castration-resistant Prostate Cancer After Failure of Lutetium-177-PSMA. Eur. Urol. 2021, 79, 343–350.
  26. Pommé, S.; Marouli, M.; Suliman, G.; Dikmen, H.; Van Ammel, R.; Jobbágy, V.; Dirican, A.; Stroh, H.; Paepen, J.; Bruchertseifer, F.; et al. Measurement of the 225Ac half-life. Appl. Radiat. Isot. 2012, 70, 2608–2614.
  27. Morgenstern, A.; Apostolidis, C.; Kratochwil, C.; Sathekge, M.; Krolicki, L.; Bruchertseifer, F. An Overview of Targeted Alpha Therapy with 225Actinium and 213Bismuth. Curr. Radiopharm. 2018, 11, 200–208.
  28. Kratochwil, C.; Bruchertseifer, F.; Giesel, F.L.; Weis, M.; Verburg, F.A.; Mottaghy, F.; Kopka, K.; Apostolidis, C.; Haberkorn, U.; Morgenstern, A. 225Ac-PSMA-617 for PSMA-targeted alpha-radiation therapy of metastatic castration resistant prostate cancer. J. Nucl. Med. 2016, 57, 1941–1944.
  29. Kratochwil, C.; Bruchertseifer, F.; Rathke, H.; Bronzel, M.; Apostolidis, C.; Weichert, W.; Haberkorn, U.; Giesel, F.L.; Morgenstern, A. Targeted alpha therapy of mCRPC with 225Actinium-PSMA-617: Dosimetry estimate and empirical dose finding. J. Nucl. Med. 2017, 58, 1624–1631.
  30. Sathekge, M.; Bruchertseifer, F.; Knoesen, O.; Reyneke, F.; Lawal, I.; Lengana, T.; Davis, C.; Mahapane, J.; Corbett, C.; Vorster, M.; et al. 225Ac-PSMA-617 in chemotherapy-naive patients with advancedprostate cancer: A pilot study. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 129–138.
  31. McDevitt, M.R.; Barendswaard, E.; Ma, D.; Lai, L.; Curcio, M.J.; Sgouros, G.; Ballangrud, Å.M.; Yang, W.H.; Finn, R.D.; Pellegrini, V.; et al. An alpha-particle emitting antibody (J591) for radioimmunotherapy of prostate cancer. Cancer Res. 2000, 60, 6095–6100.
  32. Sathekge, M.; Knoesen, O.; Meckel, M.; Modiselle, M.; Vorster, M.; Marx, S. 213Bi-PSMA-617 targeted alpha-radionuclide therapy in mCRPC. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 1099–1100.
  33. Kratochwil, C.; Schmidt, K.; Afshar-Oromieh, A.; Bruchertseifer, F.; Rathke, H.; Morgenstern, A.; Haberkorn, U.; Giesel, F.L. Targeted alpha therapy of mCRPC: Dosimetry estimate of 213Bismuth-PSMA-617. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 31–37.
  34. Liu, H.; Moy, P.; Kim, S.; Xia, Y.; Rajasekaran, A.; Navarro, V.; Knudsen, B.; Bander, N.H. Monoclonal antibodies to the extracellular domain of prostate-specific membrane antigen also react with tumor vascular endothelium. Cancer Res. 1997, 57, 3629–3634.
  35. Smith-Jones, P.M.; Vallabahajosula, S.; Goldsmith, S.J.; Navarro, V.; Hunter, C.J.; Bastidas, D.; Bander, N.H. In vitro characterization of radiolabeled monoclonal antibodies specific for the extracellular domain of prostate-specific membrane antigen. Cancer Res. 2000, 60, 5237–5243.
  36. Tagawa, S. AUA 2020: Comparison of Prostate-Specific Membrane Antigen-Targeted Radionuclide Therapy with Lutetium-177 via Antibody J591 vs. Small Molecule Ligand PSMA-617. Available online: https://www.urotoday.com/conference-highlights/aua-2020/aua-2020-prostate-cancer/122691-aua-2020-comparison-of-prostate-specific-membrane-antigen-targeted-radionuclide-therapy-with-lutetium-177-via-antibody-j591-vs-small-molecule-ligand-psma-617.html (accessed on 19 September 2021).
  37. Bander, N.; Milowsky, M.I.; Nanus, D.M.; Kostakoglu, L.; Vallabhajosula, S.; Goldsmith, S.J. Phase I trial of 177lutetium-labeled J591, a monoclonal antibody to prostate-specific membrane antigen, in patients with androgen-independent prostate cancer. J. Clin. Oncol. 2005, 23, 4591–4601.
  38. Tagawa, S.T.; Milowsky, M.I.; Morris, M.; Vallabhajosula, S.; Christos, P.; Akhtar, N.H.; Osborne, J.; Goldsmith, S.J.; Larson, S.; Taskar, N.P.; et al. Phase II study of Lutetium-177-labeled anti-prostate-specific membrane antigen monoclonal antibody J591 for metastatic castration-resistant prostate cancer. Clin. Cancer Res. 2013, 19, 5182–5191.
  39. Tagawa, S.T.; Vallabhajosula, S.; Christos, P.J.; Jhanwar, Y.S.; Batra, J.S.; Lam, L.; Osborne, J.; Beltran, H.; Molina, A.M.; Goldsmith, S.J.; et al. Phase 1/2 study of fractionated dose lutetium-177-labeled anti-prostate-specific membrane antigen monoclonal antibody J591 (177Lu-J591) for metastatic castration-resistant prostate cancer. Cancer 2019, 125, 2561–2569.
  40. Mateo, J.; Porta, N.; Bianchini, D.; McGovern, U.; Elliott, T.; Jones, R.; Syndikus, I.; Ralph, C.; Jain, S.; Varughese, M.; et al. Olaparib in patients with metastatic castration-resistant prostate cancer with DNA repair gene aberrations (TOPARP-B): A multicentre, open-label, randomised, phase 2 trial. Lancet Oncol. 2020, 21, 162–174.
  41. Tagawa, S.T.; Osborne, J.; Fernandez, E.; Thomas, C.; Niaz, M.J.; Ciriaco, A.; Vallabhajosula, S.; Vlachostergios, P.J.; Molina, A.M.; Sternberg, C.N.; et al. Phase I dose-escalation study of PSMA-targeted alpha emitter 225Ac-J591 in men with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2020, 38 (Suppl. S15), 5560.
  42. Hammer, S.; Hagemann, U.B.; Zitzmann-Kolbe, S.; Larsen, A.; Ellingsen, C.; Geraudie, S.; Grant, D.; Indrevoll, B.; Smeets, R.; von Ahsen, O.; et al. Preclinical Efficacy of a PSMA-Targeted Thorium-227 Conjugate (PSMA-TTC), a Targeted Alpha Therapy for Prostate Cancer. Clin. Cancer Res. 2020, 26, 1985–1996.
  43. Study to Evaluate the Safety, Tolerability, Pharmacokinetics, and Antitumor Activity of a Thorium-227 Labeled Antibody-chelator Conjugate Alone and in Combination with Darolutamide, in Patients with Metastatic Castration Resistant Prostate Cancer. Available online: https://clinicaltrials.gov/ct2/show/NCT03724747 (accessed on 19 September 2021).
More
Video Production Service