Abstract
Theranostics in men with metastatic castration-resistant prostate cancer (mCRPC) has been developed to target bone and the tumor itself. Currently, bone-directed targeted alpha therapy with radium-223 (223Ra) is the only theranostic agent proven to prolong survival in men with mCRPC who have symptomatic bone metastases and no known visceral metastases. The clinical utility and therapeutic success of 223Ra has encouraged the development of other tumor-targeting theranostic agents in mCRPC, primarily targeting prostate-specific membrane antigen (PSMA) with radioligand therapy (RLT). There is increasing evidence of promising response rates and a low toxicity profile with 177Lu-labeled PSMA RLT in patients with mCRPC. A phase III randomized study of 177Lu-labeled PSMA RLT has completed accrual and is awaiting results as to whether the drug improves radiographic progression-free survival and overall survival in men with mCRPC receiving standard of care treatments. Additional early clinical trials are investigating the role of tumor-directed targeted alpha therapy with radiotracers such as 225Ac. In this article, we review the current status of theranostics in prostate cancer, discussing the challenges and opportunities of combination therapies with more conventional agents such as androgen receptor inhibitors, cytotoxic chemotherapy, and immunotherapy.
Introduction
Prostate cancer is the second most common cancer in men, with an incidence increasing globally by 40% over the last 14 years1 and 1.3 million new cases diagnosed worldwide in 2018.2 Globally, prostate cancer is the fifth-leading cause of cancer-related mortality.1,3 However, evolving therapeutic approaches have altered the clinical course for men with metastatic prostate cancer with improvements in both survival and quality of life.
Typical first-line therapy for metastatic disease is a testosterone-reducing agent with androgen deprivation therapy (ADT), frequently combined with a novel androgen receptor (AR) axis inhibitor such as abiraterone or chemotherapy with docetaxel.4–6 Most patients have an initial response to these regimens; however, the disease eventually progresses to metastatic castration-resistant prostate cancer (mCRPC). At this point, other strategies must be employed to improve survival, including cytotoxic chemotherapy, AR-directed therapies, therapeutic vaccine therapy, and, more recently, theranostic agents.7–13
Theranostic agents target disease-specific structures (eg, proteins or antigens) expressed in a patient’s cancer or host organ for metastatic disease. These structures can be used as targets for ligands with specific radioisotopes for imaging and subsequently for therapy. These agents can not only be disease-specific but also can be varied to suit the desired sites of disease, namely in prostate cancer, tumor or bone.
Theranostic agents when used therapeutically can be combined with alpha- or beta-emitting radioisotopes. Beta emitters were initially used in the development of novel theranostic agents. They are longer range, low-energy agents (linear energy transfer [LET]: 0.3 keV/μm; tissue range: 2000–11,500 μm) that enhance cytotoxicity to tumor cells rather than delivering a lethal dose to a targeted single cancer cell due to their low LET,14 which results mainly in single-strand DNA breaks. However, due to their long tissue range, electrons can travel further to multiple cells and increase overall dose to tumor—the so-called crossfire effect—but can also potentially deliver dose to normal tissue. A newer approach in theranostics is to use short-range, high-energy alpha emitters (LET: 100keV/μm; tissue range: 20–80 μm), which are highly cytotoxic agents that travel over short distances. Upon travel into cell nuclei, they cause DNA damage,14 mainly as double-strand breaks. As a result, alpha radiation is thought to allow treatment of microscopic disease while sparing surrounding normal tissue.15
In this article, we provide an overview of the current and future molecular therapy strategies used for treatment of prostate cancer.
Bone-Targeting Strategies
Metastatic prostate cancer primarily occurs within bone. Normal bone structural integrity is maintained in a complex bone microenvironment by continuous balanced bone reabsorption by osteoclasts and bone formation by osteoblasts.16 Metastatic prostate cancer cells interact via signaling pathways with osteoblasts, osteocytes, osteoclasts, bone marrow stem cells, and hematopoietic cells within the bone microenvironment, disrupting normal tightly regulated osteoblastic and osteoclastic activity.17,18 Growth factors released by prostate cancer cells in bone disrupt the balanced relationship between osteoblasts and osteoclasts, leading to proliferation of new disorganized woven bone. Osteoblasts also express growth factors (eg, vascular endothelial growth factor), which promote prostate cancer cell growth and survival.19 A bidirectional positive feedback loop between tumor cells and the bone microenvironment leads to characteristic osteoblastic bone metastasis formation.18 This cycle of dysregulated bone metabolism leads to pathological bone morphology and physiology, which in turn leads to bone pain, fractures, and increased mortality.20,21
Beta-emitting bone-seeking radiopharmaceuticals, such as strontium-89-chloride (89Sr) and samarium-153-EDTMP (153Sm), have long been used as palliative agents in the setting of painful bone metastases, including prostate cancer, with reported improvement in pain and quality of life.22 However, to date, these agents have not been tested in formal comparative studies powered to demonstrate a clinical benefit.22 Nonetheless, preliminary data suggest that these agents might offer benefits relative to standard agents.23,24
Radium-223 (223Ra) is a targeted alpha-particle therapy (TAT) approved by the US Food and Drug Administration (FDA) in 201325 and European Medicines Agency (EMA)26 in 2018 for the treatment of patients with mCRPC and bone metastases following the ALSYMPCA (ALpharadin in SYMP-tomatic Prostate Cancer) trial.9 ALSYMPCA demonstrated that 223Ra (6 injections at 55 kBq/kg every 4 weeks) prolonged overall survival (OS) in men with mCRPC when added to standard anticancer therapies vs patients receiving best standard of care and placebo (median 14.9 vs 11.3 months receiving placebo; hazard ratio [HR] 0.70, 95% confidence interval [CI] 0.58–0.83; P < 0.001). Patients in the 223Ra arm also demonstrated longer time to first symptomatic skeletal event (median 15.6 vs 9.8 months; HR 0.66, 95% CI 0.52–0.83; P < 0.001) than those in the placebo arm.9,27 No benefit was conferred for increasing number of doses or administering higher doses of 223Ra.28
Animal models of prostate cancer bone metastases treated with 223Ra have been difficult to develop due to the complexity of the metastatic cascade, including immunologic response and tumor heterogenitity29 and were only recently developed in 2017,30 post approval of 223Ra by the FDA. Thus, preclinical studies of bone metastasis models in other cancers were used for initial experiences in mCRPC, which demonstrated that 223Ra targets bone metastasis by replacing calcium in hydroxyapatite complexes in woven bone surrounding prostate cancer metastasis, leading to osteoblast-mediated new bone formation at sites of metastatic lesions.31,32 223Ra decays to produce alpha particles with high LET, traveling over a short distance of ≤100 μm, which leads to localized cytotoxicity, likely inducing double-strand breakage of DNA in adjacent tumor cells, osteoblasts, and osteoclasts and disrupting the positive-feedback loops between these cells.33
Preclinical studies have demonstrated that 223Ra causes inhibition of tumor growth, pathological bone reaction, and stabilization of the normal bone structure. In addition, as 223Ra causes irreparable damage of tumor cell DNA and cell death, the feedback loop is disrupted and becomes less reliant on active cell proliferation, possibly offering an advantage for killing tumors with a low proliferation rate or dormant micrometastases.31 Also, TAT may minimize damage of surrounding normal tissue when compared to beta-emitters or external beam radiotherapy due to the short distance travelled by high LET, comparable to 3–6 cell diameters. In mouse models, 223Ra deposited at the bone surface (not within the tumor) and skeletal accumulation was dependent on local blood vessel density, suggesting that local vascular supply is necessary for effective delivery of TAT.34
It is thought that 223Ra may enhance immune system activity by inhibiting immunosuppression and altering cancer cells’ phenotypes, therefore making them susceptible to immune-mediated cell killing.35 In vitro models in human prostate and breast cancer cell lines have shown that 223Ra enhances T cell-mediated lysis of tumor cells through antigen-specific CD8+ cytotoxic T lymphocytes and may augment immune responses via the stimulation of interferon genes pathway.36
TAT is thought to be less prone to development of tumor resistance mechanisms, such as hypoxia, since it induces direct double-strand DNA damage, which is independent of oxygen. This is in comparison to oxygen-dependent activation of alternate signaling pathways used in AR-axis inhibitors and chemotherapy agents and oxygen-dependent DNA damage caused by external beam radiotherapy.
223Ra in Clinical Practice
The American and European guidelines regarding indications for treatment with 223Ra are conflicting. The National Comprehensive Cancer Network guidelines (2019) recommend treatment with 223Ra in men with mCRPC, symptomatic bone metastases, and no visceral metastases,37 while the American Urological Association guidelines (2018) recommend 223Ra for patients with mCRPC and symptomatic bone metastases, good performance status, no previous docetaxel chemotherapy, and no known visceral disease.38 The European Association of Urology guidelines (2020) recommend 223Ra as a potential life-prolonging treatment for men with mCRPC and progression on docetaxel chemotherapy.39
223Ra was well tolerated during the ALSYMPCA trial and demonstrated a favorable short-term safety profile with improvements in quality-of-life measures and low rates of myelosuppression9,40,41 as well as low cumulative incidence rates for hematologic and nonhematologic adverse events.42 The ALSYMPCA trial included follow-up of men with mCRPC and symptomatic bone metastases for up to 3 years from first injection of 223Ra to assess for association with possible induction of secondary malignancy, such as acute myelogenous leukemia, myelodysplastic syndrome, or new primary bone cancer.42 The 3-year follow-up did not find an association between 223Ra treatment and occurrence of second malignancies and there was a similar low incidence rate of nontreatment-related malignancies in both the 223Ra and placebo arms.42
Long-term safety is being evaluated further through the ongoing Radium-223 alpha Emitter Agent in non-intervention Safety Study in mCRPC popUlation for long-teRm Evaluation (REASSURE) trial, a global, prospective, single-arm observational study of patients with mCRPC and bone metastases treated with 223Ra in routine clinical practice.43 REASSURE is evaluating the long-term safety and occurrence of primary second malignancies during a 7-year follow-up period and to date, the rate and degree of severity of treatment-related adverse events seen in follow-up in both this trial43 and the ALSYMPCA trial42 are not substantially different from those observed in the original ALSYMPCA trial.9 The primary safety concern raised by 223Ra concerns the use of the drug in combination with the novel AR-inhibitor abiraterone as a first-line therapy, which has been associated with increased risk of bone fractures; this is described in greater detail later in this review.44,45
Currently, 223Ra is administered according to standard fixed doses based on patient weight. However, theranostic agents are unique in their ability to allow imaging in vivo of the uptake and retention of the agent, which enables calculation of absorbed radiation doses and radiation exposure in tumor and normal tissues, thus maximizing the antitumor effects of the therapy while sparing its effect on normal tissues46 and potentially providing more personalized dosing.
223Ra decay produces a series of 6 products before becoming a stable lead molecule, with 28.3 MeV of emitted energy—95% comes from alpha emissions, 3.2% from beta particles, and <2% from gamma emissions. Gamma emissions, although low signal, allow the potential for quantitative imaging of 223Ra.47,48 However, microscopic energy deposition within the bone matrix also presents challenges for internal dosimetry in the bone marrow and skeleton.48
Small studies have demonstrated a correlation between absorbed dose and local lesion response to 223Ra. Pacilio et al showed a correlation between the uptake of 223Ra in a metastatic lesion and the scintigraphy agent technetium-99m-methylene diphosphonate (99mTc MDP) in nine patients with mCRPC and bone metastases; thus, conventional bone scintigraphy might help to define lesion extent and response.47,49
Several microdosimetric approaches are being developed, with potential application in clinical imaging and dosimetry.46 With 223Ra, a trabecular marrow cavity model allows for calculation of the fraction of marrow volume receiving a cytotoxic absorbed dose, which may enable improved estimates of the dose of radiation delivered by radium and could potentially be applied to other TATs in the future.
Combination Therapies
Following the reassuring outcomes in terms of safety and tolerability in clinical practice, 223Ra has been proposed for use in combination with other agents, with many prospective trials ongoing.
First-line treatment for newly diagnosed metastatic disease typically employs ADT, to which the majority of patients have an initial response; however, with time the disease progresses to mCRPC but interestingly remains dependent on the AR-signaling pathway.50 Novel hormonal agents (eg, abiraterone, a selective steroidal inhibitor of cytochrome P450 c17 (CYP17), a key enzyme for testosterone and estrogen biosynthesis,51,52 and enzalutamide, a nonsteroidal antiandrogen) inhibit the AR axis system. Large randomized phase III clinical trials have demonstrated that these agents prolong the lives of men with mCRPC.10–12,37,53,54
Due to different mechanisms of action and nonoverlapping safety profiles, combining 223Ra with abiraterone or enzalutamide was proposed. Preliminary data on combination strategies suggested that 223Ra could be safely combined with abiraterone or enzalutamide.55–57 The subsequent phase III Evaluation of Radium-223 dichloride in combination with Abiraterone in castration-resistant prostate cancer (ERA-223) trial evaluated the efficacy and safety of adding 223Ra treatment or placebo to abiraterone plus prednisone or prednisolone in patients with asymptomatic or minimally symptomatic mCRPC,58 with primary analysis revealing no statistically significant difference in OS between the groups (30.7 months [223Ra group] vs 33.3 months 1 [placebo group]; HR 1.195, 95% CI 0.950–1.505; P = 0.13).58 However, patients in the 223Ra combination arm had an unexpected increased incidence of fractures (29% vs 11%). This study led the EMA in 2018 to contraindicate the combination use of 223Ra with abiraterone and prednisolone or prednisone.44,45
Among other hypothesized etiologies, the increased risk of fractures in the combination 223Ra group may be due to concurrent steroid use, as potentially harmful effects of glucocorticoids and changes within the bone microenvironment have previously been demonstrated.59 In the ERA-223 trial, concurrent use of bone health agents (BHAs), including bisphosphonates (zoledronic acid) and denosumab from baseline, was associated with a lower incidence of fractures in both the 223Ra and placebo groups (15% and 7%, respectively) compared with patients who did not receive BHAs (37% and 15%, respectively),58 with similar findings seen in the ALSYMPCA trial, in which patients treated with 223Ra in combination with BHAs appeared to have a superior outcome to those who received 223Ra without BHAs in terms of symptomatic skeletal events.9,27 Preliminary results from the ongoing randomized phase III PEACE trial comparing enzalutamide to combination enzalutamide and 223Ra in asymptomatic or mildly symptomatic mCRPC patients60 (NCT02194842) have shown that enzalutamide in combination with 223Ra increases fracture risk by 33% but that the risk is almost completely eliminated by continuous administration of BHA, commencing at least 6 weeks before the first dose of 223Ra.61 Accrual is ongoing.
Another agent currently being studied in combination with 223Ra is docetaxel, an antimitotic chemotherapy agent. Docetaxel has been proven to improve survival when used in combination with ADT in newly diagnosed patients with metastatic castration-naïve disease.4,5 Several factors support the combination of 223Ra with docetaxel, as chemotherapy has been shown to have a radiosensitizing effect through different mechanisms, such as reducing DNA repair, increasing vulnerability of hypoxic cells to cytotoxic agents, inhibiting pro-survival pathways, and reducing ability of tumor cells to repopulate during radiotherapy; these mechanisms result in an additive cytotoxic effect in combination with radiotherapy.62,63
The potential benefits of combining radiopharmaceuticals with chemotherapy in patients with mCRPC have previously been demonstrated in clinical studies of chemotherapy in combination with palliative beta particle-emitting agents64,65; however, no data currently exist on radiosensitizing effects when combining chemotherapy with TAT. The rationale for combining bone-targeted alpha therapy with chemotherapy is provided by the concept of simultaneous targeting of the tumor and bony compartment of the disease, with each agent independently prolonging survival and possibly augmenting the other via cross-sensitization.66 A randomized phase I/II clinical trial showed that 223Ra (55 kBq/kg every 6 weeks, n = 36) plus docetaxel (60 mg/m2 every 3 weeks) might enhance antitumor activity compared to docetaxel alone (75 mg/m2 every 3 weeks, n = 17).67 The 223Ra combination arm was well tolerated without greater adverse effects than in the docetaxel arm and demonstrated more durable suppression of prostate-specific antigen (PSA) and alkaline phosphatase.67–69 On the basis of these encouraging results, a phase III trial of this treatment combination is currently recruiting patients (NCT03574571).70 Additional prospective trials are ongoing with 223Ra in combination with other novel agents, including immune checkpoint inhibitors (NCT02814669).71–73
Tumor-Directed Imaging and Therapy
Prostate-specific membrane antigen (PSMA), first discovered in 1993,74 is a 750-amino acid type II transmembrane glycoprotein with folate hydrolase activity. It is produced in the cell membranes of prostate epithelial cells and is highly upregulated in the vast majority of prostate carcinomas. PSMA expression is also upregulated in high-grade, metastatic, and castration-resistant prostate cancer. However, PSMA is not only specific to prostate epithelial cells but is also expressed in other tissues including salivary glands, proximal renal tubules, duodenal mucosa, and neuroendocrine cells in colonic crypts. It remains unclear why PSMA is upregulated in prostate cancer as no known natural ligand exists; however, PSMA ligands undergo constitutive internalization by PSMA, making it an attractive imaging and molecular target for prostate cancer.
PSMA imaging was initially based on anti-PSMA antibodies, with the first clinically viable radiotracer, J591, described in 1997.75 J591, which accumulates in PSMA-expressing PC cells by binding to the extracellular domain of PSMA, can be labeled with 111In for scintigraphy or 89Zr for positron emission tomography (PET). Although large molecule radiolabeled antibodies demonstrate good tumor detection, the practicality of antibody imaging can be limited by long delays between injection and imaging and slow clearance from circulation.
PSMA imaging for diagnosis and staging of PC became more feasible with the development of small-molecule ligands, the majority of which contain glutamine urea-lysine dimers. Over the last 3 decades, urea-based inhibitors76 and PSMA inhibitors by phosphonamidothionate derivatives of glutamic acid77 were developed, enabling the development of clinically available small-molecule ligands. Small-molecule ligands, which also internalize into prostate cancer cells upon binding to PSMA, offer an advantage over large-molecule monoclonal antibodies, with increased uptake and higher percentage of specific binding to PSMA, as well as increased permeability into solid tumors. They also demonstrate more rapid tissue distribution and tissue clearance than monoclonal antibodies.
The small-molecule PSMA ligands 123I-MIP-1972 and 123I-MIP-1095 (Molecular Insight Pharmaceuticals, Cambridge, MA) were first used in humans in 2008 for diagnosis with scintigraphy,78 with subsequent development of 99mTc-MIP-1404 and 99mTc-MIP-1405 in 2010.79,80 Subsequently, PSMA-labeled PET radiotracers were developed with 68Ga-PSMA-11 used clinically for diagnosis since 2011, primarily for detecting recurrence or metastatic disease in the setting of biochemical recurrence following primary treatment. 68Ga-PSMA-11 is not currently FDA-approved but is widely used in Europe and Australia.
Studies have shown that 68Ga-PSMA-11 PET/CT has high sensitivity and specificity compared to alternative imaging methods used for detection of recurrent PC.81,82 The probability of detecting tumor lesions increases with increased PSA levels, with a reported detection rate of about 50% at PSA <0.5 ng/mL and 80% at PSA >1.0 ng/mL.82 However, even with high PSA values (>10.0 ng/mL), not all patients have a positive 68Ga-PSMA-11 PET/CT. Ongoing use of ADT at the time of study has been significantly associated with positive 68Ga-PSMA-11 PET/CT. Additional PSMA-labeled radiotracers in use or under investigation include 18F-PSMA, 18F-DCFPyl, 18F-rh-PSMA-7, and 18F-si-PSMA-7, with similar promising results. However, as PSMA is not specific to prostate cancer, and as clinical experience with 68Ga-PSMA-11 PET/CT has increased, more reports of PSMA-avid, nonprostate benign and malignant lesions have been published.
Since antibodies and small-molecule ligands are internalized once they bind to PSMA, these molecules were also determined to be an attractive target for radiopharmaceutical therapy for prostate cancer. Again, radiolabeled large monoclonal antibodies (90Y or 177Lu-PSMA-J591) were developed initially, with phase I and II clinical trials showing encouraging results.83–85 However, due to molecule size and resultant poor permeability in solid tumors as well as slow clearance from circulation, these molecules can deliver increased absorbed doses to red marrow. Hematologic toxicity can result, sometimes irreversible. Tagawa et al, who treated 47 patients with 177Lu-PSMA-J591, reported grade 4 thrombocytopenia in 46.8% of patients with 29.8% requiring platelet transfusions and grade 4 neutropenia in 25.5% of patients.83 However, more recent phase I/II trials have shown that a fractionated regimen of 177Lu-J591 allows for higher cumulative radiation dosing with greater decrease in PSA levels and longer survival compared to a single-dose regimen.86
As with imaging, small-molecule inhibitors of PSMA have also been used in radiopharmaceutical therapy79; the first agent used in clinical practice was 131I-MIP-1095 (Molecular Insight Pharmaceuticals, Inc. Cambridge, MA), which showed promising results with symptomatic relief, PSA decrease of >50% in up to 70.6% patients treated, and median time to PSA progression of 126 days. 131I-MIP-1095 also demonstrated milder hematologic toxicity compared with radiolabeled antibodies, with Afshar-Oromieh et al reporting no significant hematologic toxicity as per WHO CTC grading system in a study of 34 patients with mCRPC treated with up to 3 cycles of 131I-MIP-1095.87 However, xerostomia was a significant side effect with 131I-MIP-1095. Grade 1 xerostomia was seen in 70.6% of patients, with grade of xerostomia increasing with number of cycles, although it was self-limited in the majority of patients.88 While 131I is predominantly a beta emitter (606 keV; 90%), it also has relatively high gamma emission (364 keV, 10%) and a long half-life of 8.02 days, which makes it a less ideal radiopharmaceutical from a radiation safety point of view, compared to lower-energy emitters such as 177Lu (half-life of 6.7 days;(Figure).
Figure.
(A) Mechanism of action of radium-223 (223Ra). 223Ra substitutes for calcium in hydroxyapatite complexes in woven bone surrounding prostate cancer metastatic lesions. 223Ra emits high-energy alpha particles over a range of ≤100 μm. The alpha radiation leads to localized cytotoxicity through the induction of DNA double-strand breaks in adjacent tumor cells, osteoblasts and osteoclasts, which disrupts the positive-feedback loops between these cells, leading to inhibition of tumor growth and pathological bone reaction and stabilization of the normal bone microenvironment. (B) Disease-specific theranostic agents, many being used in combination with other anticancer agents targeting distant metastatic disease, are in being used in routine clinical practice or are under preclinical or clinical investigation in mCRPC.
177Lu-PSMA-617
Initial studies of the use of 177Lu-PSMA-617 in patient cohorts heavily pretreated with agents including ADT, next-generation AR-axis targeting agents, and docetaxel demonstrated favorable therapeutic response with mild side effects; for example, in the initial phase I trial with 10 patients who received 1 cycle of RLT, 70% of patients demonstrated a decline in PSA, with 5 patients demonstrating a decline of >50% at 8 weeks and only 1 patient suffering grade 3/4 hematologic toxicity.89 Ahmadzadehfar et al demonstrated similar results in a slightly larger cohort of patients who received up to 2 cycles of 177Lu-PSMA-617.90 Baum et al showed even more promising results with 177Lu-PSMA-I&T in a cohort of patients with early-stage castration resistance and less prior chemotherapy exposure (25% of patients vs >75% in aforementioned studies), with reported PSA declines of >50% in 80% of patients and longer median OS of 13.7 months.91 The largest cohort published to date is a German multicenter retrospective analysis of 145 patients with mCRPC that mirrored the results of earlier trials, with PSA decline seen in 60% of patients and PSA decline >50% seen in 45% of patients.92 However, this study determined that the presence of visceral metastases, especially liver metastases and/or alkaline phosphatase >220 U/l, was a predictor of a poorer response.
Studies have suggested a clinical benefit with RLT; Rahbar et al demonstrated an estimated median survival in a group treated with 177Lu-PSMA-617 of 29.4 weeks compared to 19.7 weeks in a historical control group (HR: 0.44; 95% CI: 0.20–0.95; P = 0.031),92 but survival benefit has not yet been proven. Subsequent studies that analyzed OS in patients receiving up to 8 cycles of 177Lu-PSMA-617 RLT determined that any PSA decline after the first cycle was a significant prognostic marker of survival (68 vs 33 weeks and 59 vs 28 weeks, respectively), with a PSA decline of ≥21%, the optimal parameter for predicting improved OS in the multivariate analysis.93 However, the same study also showed that in patients who do not respond to the first cycle of RLT, further therapy cycles should still be performed, since nearly one-third of the patients showed a delayed response after additional therapy cycles.93
177Lu-PSMA-617 has been well tolerated to date.92,94 Hematologic side effects have been minimal, with grade 3/4 toxicities including anemia, leukopenia, and thrombocytopenia reported in 10%, 3%, and 4% of patients, respectively, treated with 177Lu-PSMA-617. Use of 177Lu-PSMA-617 RLT after radionuclide therapy with 223Ra dichloride has not demonstrated a higher probability of hematologic toxicity.95 Although there is specific renal binding of PSMA ligands, to date, no grade 3/4 renal toxicities have been reported after RLT.96 Eight percent of patients reported mild or transient xerostomia.
Although several 177Lu-PSMA-617 trials have demonstrated favorable safety and therapeutic response,89,91,92 these trials are limited as they are predominantly retrospective phase I/II in design. Also, PSA is only a surrogate marker for post-treatment effects, and an improved PSA response is not necessarily predictive of longer progression-free survival (PFS) and OS.97,98 To date, only 1 prospective single-center phase II trial has been performed; 30 mCRPC patients who previously received abiraterone acetate, enzalutamide, or taxol chemotherapy received up to 4 cycles of 177Lu-PSMA-617.94 This group demonstrated a PSA decline ≥50% in 57% of patients, with an objective response in nodal or visceral disease reported in 14 of 17 (82%) patients with measurable disease. Additionally, 11 (37%) patients experienced a 10-point or greater improvement in global health score by the end of cycle 2.
The VISION trial is a prospective, multicenter, randomized phase III trial open to accrual that is examining whether adding 177Lu-PSMA-617 RLT to standard-of-care therapy (SOCs) in mCRPC patients is more effective than SOCs alone99 (NCT03511664). The study endpoints are radiographic PFS and OS. Patients randomized to receive RLT will receive 7.4 GBq (±10%) 177Lu-PSMA-617 intravenously every 6 weeks for a maximum of 6 cycles. After 4 cycles, patients will be assessed for (1) evidence of response, (2) residual disease, and (3) tolerance to RLT and if the criteria for all 3 assessments are met, the patient may receive an additional 2 cycles of RLT. The trial is currently accruing, and results are pending; if they are positive, this may lead to regulatory approval for RLT in mCRPC. However, the VISION trial does have potential limitations—for example, patients on chemotherapy or 223Ra were not considered SOC due to safety concerns and the trial does not compare 177Lu-PSMA-617 RLT to SOCs alone.
The TheraP phase II trial, however, is comparing 177Lu-PSMA-617 with the second-line chemotherapy agent cabazitaxel in patients with mCRPC who have progressed on ADT and first-line chemotherapy with endpoints of PSA response rate, pain response, PFS, quality of life, and frequency and severity of adverse events100 (NCT03392428). Accrual is complete and results are pending.
Future Developments
Combination therapies with 177Lu-PSMA-617 and other RLTs—for example, 177Lu-PSMA-R2 and 177Lu-PSMA-I&T—are being investigated as potential treatment strategies, as further outlined in Table.
Table.
Summary of Selected Ongoing Clinical Trials Using Theranostic Agents in mCRPC
| Agent | Mechanism of Action | Trial Design | Primary Endpoint | Estimated Completion Date | Clinical Trial ID |
|---|---|---|---|---|---|
|
| |||||
| Alpha emitters | |||||
| 223Ra + Enzalutamide (vs enzalutamide only) “PEACE III” | AR inhibitor | Randomized Phase III | Radiographic PFS | December 2025 | NCT02194842 |
| 223Ra + Docetaxel (vs Docetaxel only) | Microtubule inhibitor | Randomized Phase III | Overall survival | June 2023 | NCT03574571 |
| 223Ra + Pembrolizumab (vs 223Ra only) | PD-1 inhibitor | Randomized Phase II | Extent of immune cell infiltration | June 2024 | NCT03093428 |
| 223Ra + Olaparib (vs 223Ra only) | PARP inhibitor | Randomized Phase I/II | - MTD of olaparib and 223Ra - Radiographic PFS |
April 2020 | NCT03317392 |
| 225Ac-J591 | Monoclonal Antibody | Phase I | - Change in the number of subjects with DLT - MTD |
July 2024 | NCT03276572 |
| 225Ac-PSMA | Radioligand | Phase I | Serum PSA | December 2021 | NCT04225910 |
| Beta-emitters | |||||
| 177Lu-PSMA-617 + SOC (vs SOC only) “VISION” | Radioligand | Randomized Phase III | Overall survival | June 2021 | NCT03511664 |
|
177Lu-PSMA-617 vs Cabazitaxel “TheraP” |
Radioligand | Randomized Phase II | PSA response rate | January 2021 | NCT03392428 |
| 177Lu-PSMA-R2 | Radioligand | Phase I/II | - Incidence of DLT during first cycle - PSA response rate |
June 2022 | NCT03490838 |
| 177Lu-PSMA-I&T | Radioligand | Phase I | Serum PSA | August 2020 | NCT04188587 |
|
177Lu-PSMA-617 + Pembrolizumab “PRINCE” |
Radioligand + PD-1 inhibitor | Phase Ib/II | - Adverse Events - PSA response |
October 2021 | NCT03658447 |
|
177Lu-PSMA-617 + Olaparib “LuPARP” |
Radioligand + PARP inhibitor | Phase I | - DLT - MTD - Recommend Phase II dose |
October 2022 | NCT03874884 |
| 177Lu-PSMA-617 + idronoxil “LuPin” | Radioligand + Radiosensitizing agent | Phase I/II | - Toxicity profile - Anti-cancer efficacy |
Not provided | ACTRN12618001073291 |
| 177Lu-PSMA-617 + 177Lu-J591 | Radioligand + Monoclonal Ab | Phase I/II | - DLT of combination therapy - Cumulative MTD - PSA decline |
June 2022 | NCT03545165 |
| 227Th-BAY2315497 | Monoclonal Ab | Phase I | MTD | November 2022 | NCT03724747 |
mCRPC, metastatic castrate resistant prostate cancer; PSA, prostate specific antigen; PSMA, prostate specific membrane antigen; 223Ra, radium-223; 177Lu, luteteim-177; 225Ac, actinium-225; 227Th, thorium-227; AR, androgen receptor; PD-1, programmed cell death 1; PARP, poly (ADP-ribose) polymerase; Ab, antibody; PFS, progression-free survival; MTD, maximum tolerated dose; DLT, dose limiting toxicity; SOC, best standard of care.
However, up to 30% of patients are nonresponders or develop resistance to 177Lu-PSMA-617. Previously, studies in patients with neuroendocrine tumors refractory to beta-emitting RLT (90Y or 177Lu-somatostatin receptor analogs) have demonstrated a subsequent response to TAT with 213Bi-DOTATAC.101 Because TATs have a shorter tissue-penetration range, they have a theoretical advantage in terms of hematologic toxicity, especially in patients with diffuse-type bone marrow infiltration, as seen in widespread bone metastases. Thus, TATs have been proposed as possible tumor-targeting agents in mCRPC. To date, 225Ac-PSMA-617 has been the most studied PSMA-targeted TAT, with phase I and II clinical trials studying its use predominantly in heavily pretreated patients with agents including ADT, enzalutamide, and docetaxel.
Kratochwil et al retrospectively reviewed 40 patients with mCRPC who received 3 cycles of 225Ac-PSMA-617 (100 kBq/kg) at 8-week intervals following failure on SOC therapy.102 They reported ≥50% PSA decline in 60% of patients, exceeding the biochemical response rates of 177Lu-PSMA-617. In addition, complete response with regard to PSA and PSMA PET/CT was achieved in 13% of 225Ac-PSMA-617 patients; complete response with 177Lu-PSMA RLT was reported in only 1% of patients, even with less advanced disease. They reported a median PFS of 7 months and an OS of 12 months within their cohort. However, as mentioned previously, PSA is only a surrogate marker for response and improvement in PSA does not necessarily predict longer PFS and OS in patients receiving 225Ac-PSMA-617 vs 177Lu-PSMA.
Reported side effects of 225Ac-PSMA-617 have been more concerning than with 177Lu-PSMA-617—most notably, xerostomia, with one-third of patients requesting to discontinue therapy due to this side effect in 1 study.103 The use of 225Ac-PSMA-617 in combination with or following 177Lu-PSMA-617 RLT has been limited. Khreish et al reported their experience of treating 20 patients with 1 cycle of tandem 225Ac-PSMA-617 and 177Lu-PSMA-617 without safety concerns and interestingly with reported rates of xerostomia less than with 225Ac-PSMA-617 monotherapy,104 although this finding is limited, as it is a subjective metric. Feuerecker et al reported the use of 225Ac-PSMA-617 in a cohort of 15 patients following failure of 177Lu-PSMA-617 RLT, demonstrating a PSA decline in two-thirds of patients with one-third of patients showing a decline of ≥50%.105 However, xerostomia was reported in all patients as well as significant hematologic toxicities: 27% of patients with grade 3/4 anemia and 13% of patients suffering grade 3 thrombocytopenia.
Although 225Ac as an alpha-emitting radioisotope for RLT shows early promising results, its development as a RLT may also be limited due to its scarcity. Annually, only 63 GBq (1.7 Ci) of 225Ac is produced globally, primarily derived from the build-up of 229Th through the decay of 233U stockpiles available from three sources (Canada, Germany, and Russia), which would facilitate 225Ac-PSMA-617 RLT for <1000 patients per year.106
Other TATs have been proposed in patients with mCRPC who have failed SOCs. A first-in-human treatment concept with 213Bi-PSMA-617 has recently been reported in a patient with progressive mCRPC.107 Following 2 cycles of therapy (accumulative activity of 592 MBq), the patient demonstrated both a biochemical (PSA decline >80%) and radiological response on 68Ga-PSMA PET/CT. However, further research is required to study the efficacy and safety of this therapy. Additionally, an ongoing phase I trial is investigating Thorium-227 BAY2315497, an alpha emitter conjugated to an antibody specific for PSMA, in patients with mCRPC108 (NCT03724747).
Humanized IgG1 antibody hu11B6, which internalizes into prostate and prostate cancer cells by binding to the catalytic cleft of human kallikrein 2 (hK2), a prostate-specific enzyme directed by the AR pathway, has also been explored.109 Preclinical studies investigating 225Ac-hu11B6110 and 177Lu-hu11B6109 can deliver therapeutic absorbed doses to prostate cancer xenografts with transient hematologic side effects, with both radiotracers demonstrating a therapeutic mechanism, a feed-forward mechanism coupled to the AR pathway driving DNA damage or clonal lineage selection. There is an ongoing pilot trial of this agent to establish targeting, as a prelude to a phase I therapeutic trial111 (NCT04116164).
Finally, targeting of the fibroblast activation protein is currently being explored for different tumor types using PET imaging with the fibroblast activation protein inhibitor (FAPI), 68Ga-FAPI-04, with early case reports of FAPI expression in mCRPC.112,113 The first therapeutic applications of 90Y-FAPI-04 have been applied in a patient with metastatic breast cancer, but this could potentially be an emerging imaging and therapeutic strategy in mCRPC.114
Conclusion
Theranostic agents in the treatment of prostate cancer were initially developed for targeting bone-specific disease but more recently, agents are being developed to target the tumor itself. 223Ra TAT remains an important component of the treatment paradigm for mCRPC, as to date, it is the only agent that has a proven survival benefit. 223Ra acts through multiple mechanisms to remodel pathologic bone cells, including DNA damage to tumor cells, and activates the cellular immune defenses within the bone microenvironment, which is important for understanding its role in combination with other agents such as cytotoxic chemotherapy and immunotherapy, for which initial preclinical trials are promising. However, the results of randomized controlled trials are required before 223Ra combinations can be used in clinical practice, especially in light of the unexpected findings of the ERA-223 trial, despite the convincing preclinical rationale for combining 223Ra with abiraterone to treat mCRPC.
The recent development of tumor-specific RLT is an exciting advancement for the role of theranostics in the treatment of mCRPC. Although early-phase I and II trials have been promising, the role of 177Lu-PSMA-617 will be determined by the VISION trial; if successful, it will cement the role of theranostics and alter treatment paradigms, with PSMA-based imaging used to identify patients likely to benefit from treatment with 177Lu-PSMA-617. However, if the results of 177Lu-PSMA-617 are not successful, it will represent a substantial drawback for the field of theranostics.
Finally, tumor-specific TATs are in development which, due to their inherent properties, may offer therapeutic gain while minimizing damage to normal tissue. TATs may also allow targeting of small volume disease. Although initial experiences seem promising, with both biochemical and radiological responses, randomized controlled trials are again required to determine treatment efficacy. Studies are also required to determine in what specific situations RLT should be alpha- or beta-directed, or whether a combination of both may confer additional benefit.
Footnotes
Conflicts of Interest: None.
References
- 1.Allemani C, Matsuda T, Di Carlo V, et al. : Global surveillance of trends in cancer survival 2000−14 (CONCORD-3): Analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet 391:1023–1075, 2018. 10.1016/S0140-6736(17)33326-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Prostate Cancer Statistics | World Cancer Research Fund. Available at: https://www.wcrf.org/dietandcancer/cancer-trends/prostate-cancer-statistics. Accessed February 13, 2020.
- 3.Reinmuth N, Stumpf P, Stumpf A, et al. : Characteristics of lung cancer after a previous malignancy. Respir Med 108:910–917, 2014. 10.1016/j.rmed.2014.02.015 [DOI] [PubMed] [Google Scholar]
- 4.Sweeney CJ, Chen Y-H, Carducci M, et al. : Chemohormonal therapy in metastatic hormone-sensitive prostate cancer. N Engl J Med 373:737–746, 2015. 10.1056/NEJMoa1503747 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.James ND, Sydes MR, Clarke NW, et al. : Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer (STAMPEDE): Survival results from an adaptive, multiarm, multistage, platform randomised controlled trial. Lancet 387:1163–1177, 2016. 10.1016/S0140-6736(15)01037-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Fizazi K, Tran N, Fein L, et al. : Abiraterone plus prednisone in metastatic, castration-sensitive prostate cancer. N Engl J Med 377:352–360, 2017. 10.1056/NEJMoa1704174 [DOI] [PubMed] [Google Scholar]
- 7.Tannock IF, de Wit R, Berry WR, et al. : Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med 351:1502–1512, 2004. 10.1056/NEJMoa040720 [DOI] [PubMed] [Google Scholar]
- 8.De Bono JS, Oudard S, Ozguroglu M, et al. : Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: A randomised open-label trial. Lancet 376:1147–1154, 2010. 10.1016/S0140-6736(10)61389-X [DOI] [PubMed] [Google Scholar]
- 9.Parker C, Nilsson S, Heinrich D, et al. : Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 369:213–223, 2013. 10.1056/NEJMoa1213755 [DOI] [PubMed] [Google Scholar]
- 10.Ryan CJ, Smith MR, De Bono JS, et al. : Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med 368:138–148, 2013. 10.1056/NEJMoa1209096 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Beer TM, Armstrong AJ, Rathkopf DE, et al. : Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med 371:424–433, 2014. 10.1056/NEJMoa1405095 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Scher HI, Fizazi K, Saad F, et al. : Increased survival with enzalutamide in prostate cancer after chemotherapy. Cabot RC, Harris NL, Rosenberg ES, et al. , eds. N Engl J Med 367:1187–1197, 2012. 10.1056/NEJMoa1207506 [DOI] [PubMed] [Google Scholar]
- 13.Kantoff PW, Higano CS, Shore ND, et al. : Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 363:411–422, 2010. 10.1056/NEJMoa1001294 [DOI] [PubMed] [Google Scholar]
- 14.Marcu L, Bezak E, Allen BJ: Global comparison of targeted alpha vs targeted beta therapy for cancer: In vitro, in vivo and clinical trials. Crit Rev Oncol Hematol 2018. 10.1016/j.critrevonc.2018.01.001 [DOI] [PubMed] [Google Scholar]
- 15.Allen BJ: A comparative evaluation of Ac225 vs Bi213 as therapeutic radioisotopes for targeted alpha therapy for cancer. Australas Phys Eng Sci Med 40:369–376, 2017. 10.1007/s13246-017-0534-6 [DOI] [PubMed] [Google Scholar]
- 16.Hammer S, Hagemann UB, Zitzmann-Kolbe S, et al. : Abstract 844: Preclinical activity of PSMA-TTC, a targeted alpha therapeutic in patient-derived prostate cancer models. Cancer Res 78:844., 2018. 10.1158/1538-7445.am2018-844. American Association for Cancer Research (AACR) [DOI] [Google Scholar]
- 17.Body JJ, Casimiro S, Costa L: Targeting bone metastases in prostate cancer: Improving clinical outcome. Nat Rev Urol 12:340–356, 2015. 10.1038/nrurol.2015.90 [DOI] [PubMed] [Google Scholar]
- 18.Casimiro S, Guise TA, Chirgwin J: The critical role of the bone microenvironment in cancer metastases. Mol Cell Endocrinol 310:71–81, 2009. 10.1016/j.mce.2009.07.004 [DOI] [PubMed] [Google Scholar]
- 19.Ottewell PD: The role of osteoblasts in bone metastasis. J bone Oncol 5:124–127, 2016. 10.1016/j.jbo.2016.03.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hagiwara M, Delea TE, Saville MW, et al. : Healthcare utilization and costs associated with skeletal-related events in prostate cancer patients with bone metastases. Prostate Cancer Prostatic Dis 16:23–27, 2013. 10.1038/pcan.2012.42 [DOI] [PubMed] [Google Scholar]
- 21.Oefelein MG, Ricchiuti V, Conrad W, et al. : Skeletal fractures negatively correlate with overall survival in men with prostate cancer. J Urol 168:1005–1007, 2002. 10.1097/01.ju.0000024395.86788.cc [DOI] [PubMed] [Google Scholar]
- 22.Jong van Dodewaard-de JM, Oprea-Lager DE, Hooft L, et al. : Radiopharmaceuticals for palliation of bone pain in patients with castration-resistant prostate cancer metastatic to bone: A systematic review. Eur Urol 70:416–426, 2016. 10.1016/j.eururo.2015.09.005 [DOI] [PubMed] [Google Scholar]
- 23.Tu SM, Delpassand ES, Jones D, et al. : Strontium-89 combined with doxorubicin in the treatment of patients with androgen-independent prostate cancer. Urol Oncol 2:191–197, 1996. 10.1016/S1078-1439(97)00013-6 [DOI] [PubMed] [Google Scholar]
- 24.Sciuto R, Festa A, Pasqualoni R, et al. : Metastatic bone pain palliation with 89-Sr and 186-Re-HEDP in breast cancer patients. Breast Cancer Res Treat 66:101–109, 2001. 10.1023/A:1010658522847 [DOI] [PubMed] [Google Scholar]
- 25.Xofigo (radium Ra 223 dichloride) Injection | Enhanced Reader. mozextension://4f227677-c1f9-a741-9dae-9a28f7e15797/enhanced-reader.html?openApp&pdf=https%3A%2F%2Fwww.accessdata.fda.gov%2Fdrugsatfda_docs%2Flabel%2F2013%2F203971lbl.pdf. Accessed February 14, 2020.
- 26.Xofigo | European Medicines Agency. https://www.ema.europa.eu/en/medicines/human/EPAR/xofigo. Accessed February 14, 2020.
- 27.Sartor O, Coleman R, Nilsson S, et al. : Effect of radium-223 dichloride on symptomatic skeletal events in patients with castration-resistant prostate cancer and bone metastases: Results from a phase 3, double-blind, randomised trial. Lancet Oncol 15:738–746, 2014. 10.1016/S1470-2045(14)70183-4 [DOI] [PubMed] [Google Scholar]
- 28.Sternberg CN, Saad F, Graff JN, et al. : A randomized phase 2 study investigating 3 dosing regimens of radium-223 dichloride (Ra-223) in bone metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol 36(15_suppl):5008, 2018. 10.1200/jco.2018.36.15_suppl.5008 [DOI] [Google Scholar]
- 29.Berish RB, Ali AN, Telmer PG, et al. : Translational models of prostate cancer bone metastasis. Nat Rev Urol 15:403–421, 2018. 10.1038/s41585-018-0020-2 [DOI] [PubMed] [Google Scholar]
- 30.Suominen MI, Fagerlund KM, Rissanen JP, et al. : Radium-223 inhibits osseous prostate cancer growth by dual targeting of cancer cells and bone microenvironment in mouse models. Clin Cancer Res 23:4335–4346, 2017. 10.1158/1078-0432.CCR-16-2955 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Bruland ØS, Nilsson S, Fisher DR, et al. : High-linear energy transfer irradiation targeted to skeletal metastases by the a-emitter 223Ra: Adjuvant or alternative to conventional modalities? Clin Cancer Res 12 (20 PART 2):6250s–6257s, 2006. 10.1158/1078-0432.CCR-06-0841 [DOI] [PubMed] [Google Scholar]
- 32.Henriksen G, Breistøl K, Bruland ØS, et al. : Significant antitumor effect from bone-seeking, alpha-particle-emitting (223)Ra demonstrated in an experimental skeletal metastases model. Cancer Res 62:3120–3125, 2002 [PubMed] [Google Scholar]
- 33.Iagaru AH, Mittra E, Colletti PM, et al. : Bone-targeted imaging and radionuclide therapy in prostate cancer. J Nucl Med 57(Suppl 3):19S–24S, 2016. 10.2967/jnumed.115.170746 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Abou DS, Ulmert D, Doucet M, et al. : Whole-body and microenvironmental localization of radium-223 in naïve and mouse models of prostate cancer metastasis. J Natl Cancer Inst 108, 2016. 10.1093/jnci/djv380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hodge JW, Guha C, Neefjes J, et al. : Synergizing radiation therapy and immunotherapy for curing incurable cancers: Opportunities and challenges. Oncology 22:1064–1070, 2008 [PMC free article] [PubMed] [Google Scholar]
- 36.Barber GN: STING: Infection, inflammation and cancer. Nat Rev Immunol 15:760–770, 2015. 10.1038/nri3921 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.NCCN—Evidence-Based Cancer Guidelines, Oncology Drug Compendium, Oncology Continuing Medical Education. Prostate cancer version Available at:https://www.nccn.org/. Accessed February 14, 2020.
- 38.Prostate Cancer: Castration Resistant Guideline—American Urological Association. Available at:https://www.auanet.org/guidelines/prostate-cancer-castration-resistant-guideline. Accessed February 14, 2020.
- 39.EAU Guidelines: Prostate Cancer | Uroweb. Available at:https://uro-web.org/guideline/prostate-cancer/. Accessed February 14, 2020.
- 40.Nilsson S, Cislo P, Sartor O, et al. : Patient-reported quality-of-life analysis of radium-223 dichloride from the phase III ALSYMPCA study. Ann Oncol 27:868–874, 2016. 10.1093/annonc/mdw065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Vogelzang NJ, Coleman RE, Michalski JM, et al. : Hematologic safety of radium-223 dichloride: Baseline prognostic factors associated with myelosuppression in the ALSYMPCA trial.. Clin Genitourin Cancer 15:42–52, 2017. 10.1016/j.clgc.2016.07.027. Elsevier Ince8 [DOI] [PubMed] [Google Scholar]
- 42.Parker CC, Coleman RE, Sartor O, et al. : Three-year safety of radium-223 dichloride in patients with castration-resistant prostate cancer and symptomatic bone metastases from phase 3 randomized alpharadin in symptomatic prostate cancer trial. Eur Urol 73:427–435, 2018. 10.1016/j.eururo.2017.06.021 [DOI] [PubMed] [Google Scholar]
- 43.Dizdarevic S, Petersen PM, Essler M, et al. : Interim analysis of the REASSURE (Radium-223 alpha Emitter Agent in non-intervention Safety Study in mCRPC popUlation for long-teRm Evaluation) study: Patient characteristics and safety according to prior use of chemotherapy in routine clinical practice. Eur J Nucl Med Mol Imaging 46:1102–1110, 2019. 10.1007/s00259-019-4261-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Xofigo | European Medicines Agency. Available at:https://www.ema.europa.eu/en/medicines/human/referrals/xofigo. Accessed February 14, 2020.
- 45.O’Sullivan JM, Heinrich D, James ND, et al. : The case against the European Medicines Agency’s change to the label for radium-223 for the treatment of metastatic castration-resistant prostate cancer. Eur Urol 75:e51–e52, 2019. 10.1016/j.eururo.2018.11.003 [DOI] [PubMed] [Google Scholar]
- 46.Sgouros G, Hobbs RF: Dosimetry for radiopharmaceutical therapy. Semin Nucl Med 44:172–178, 2014. 10.1053/j.semnuclmed.2014.03.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Pacilio M, Ventroni G, De Vincentis G, et al. : Dosimetry of bone metastases in targeted radionuclide therapy with alpha-emitting 223Ra-dichloride. Eur J Nucl Med Mol Imaging 43:21–33, 2016. 10.1007/s00259-015-3150-2 [DOI] [PubMed] [Google Scholar]
- 48.Flux GD: Imaging and dosimetry for radium-223: The potential for personalized treatment. Br J Radiol 90, 2017:20160748. 10.1259/bjr.20160748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Akabani G, Kennel SJ, Zalutsky MR: Microdosimetric analysis of a-particle-emitting targeted radiotherapeutics using histological images. J Nucl Med 44:792–805, 2003 [PubMed] [Google Scholar]
- 50.Knudsen KE, Scher HI: Starving the addiction: New opportunities for durable suppression of AR signaling in prostate cancer. Clin Cancer Res 15:4792–4798, 2009. 10.1158/1078-0432.CCR-08-2660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Attard G, Belldegrun AS, De Bono JS: Selective blockade of androgenic steroid synthesis by novel lyase inhibitors as a therapeutic strategy for treating metastatic prostate cancer. BJU Int 96:1241–1246, 2005. 10.1111/j.1464-410X.2005.05821.x [DOI] [PubMed] [Google Scholar]
- 52.Crona DJ, Milowsky MI, Whang YE: Androgen receptor targeting drugs in castration-resistant prostate cancer and mechanisms of resistance. Clin Pharmacol Ther 98:582–589, 2015. 10.1002/cpt.256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.De Bono JS, Logothetis CJ, Molina A, et al. : Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med 364:1995–2005, 2011. 10.1056/NEJMoa1014618 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Parker C, Gillessen S, Heidenreich A, et al. : Cancer of the prostate: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 26:v69–v77, 2015. 10.1093/annonc/mdv222 [DOI] [PubMed] [Google Scholar]
- 55.Saad F, Carles J, Gillessen S, et al. : Radium-223 and concomitant therapies in patients with metastatic castration-resistant prostate cancer: An international, early access, open-label, single-arm phase 3b trial. Lancet Oncol 17:1306–1316, 2016. 10.1016/S1470-2045(16)30173-5 [DOI] [PubMed] [Google Scholar]
- 56.Sartor O, Vogelzang NJ, Sweeney C, et al. : Radium_223 safety, efficacy, and concurrent use with abiraterone or enzalutamide: First U.S. experience from an expanded access program. Oncologist 23:193–202, 2018. 10.1634/theoncologist.2017-0413 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Shore ND, Tutrone RF, Mariados NF, et al. : eRADicAte: A prospective evaluation combining radium-223 dichloride and abiraterone acetate plus prednisone in patients with castration-resistant prostate cancer. Clin Genitourin Cancer 16:149–154, 2018. 10.1016/j.clgc.2017.10.022 [DOI] [PubMed] [Google Scholar]
- 58.Smith M, Parker C, Saad F, et al. : Addition of radium-223 to abiraterone acetate and prednisone or prednisolone in patients with castration-resistant prostate cancer and bone metastases (ERA 223): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 20:408–419, 2019. 10.1016/S1470-2045(18)30860-X [DOI] [PubMed] [Google Scholar]
- 59.Ton FN, Gunawardene SC, Lee H, et al. : Effects of low-dose prednisone on bone metabolism. J Bone Miner Res 20:464–470, 2005. 10.1359/JBMR.041125 [DOI] [PubMed] [Google Scholar]
- 60.Phase III Radium 223 mCRPC-PEACE III—Full text view—ClinicalTrials.gov. Available at:https://clinicaltrials.gov/ct2/show/NCT02194842. Accessed February 14, 2020.
- 61.Tombal BF, Loriot Y, Saad F, et al. : Decreased fracture rate by mandating bone-protecting agents in the EORTC 1333/PEACE III trial comparing enzalutamide and Ra223 versus enzalutamide alone: An interim safety analysis. J Clin Oncol 37(15_suppl):5007, 2019. 10.1200/jco.2019.37.15_suppl.5007 [DOI] [Google Scholar]
- 62.Seiwert TY, Salama JK, Vokes EE: The concurrent chemoradiation paradigm—General principles. Nat Clin Pract Oncol 4:86–100, 2007. 10.1038/ncponc0714 [DOI] [PubMed] [Google Scholar]
- 63.Perrotti M, Doyle T, Kumar P, et al. : Phase I/II trial of docetaxel and concurrent radiation therapy in localized high risk prostate cancer (AGUSG 03–10). Urol Oncol Semin Orig Investig 26:276–280, 2008. 10.1016/j.urolonc.2007.04.003 [DOI] [PubMed] [Google Scholar]
- 64.Morris MJ, Pandit-Taskar N, Carrasquillo J, et al. : Phase I study of samarium-153 lexidronam with docetaxel in castration-resistant metastatic prostate cancer. J Clin Oncol 27:2436–2442, 2009. 10.1200/JCO.2008.20.4164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Fizazi K, Beuzeboc P, Lumbroso J, et al. : Phase II trial of consolidation docetaxel and samarium-153 in patients with bone metastases from castration-resistant prostate cancer. J Clin Oncol 27:2429–2435, 2009. 10.1200/JCO.2008.18.9811 [DOI] [PubMed] [Google Scholar]
- 66.Bentzen SM, Harari PM, Bernier J: Exploitable mechanisms for combining drugs with radiation: Concepts, achievements and future directions. Nat Clin Pract Oncol 4:172–180, 2007. 10.1038/ncponc0744 [DOI] [PubMed] [Google Scholar]
- 67.Morris MJ, Loriot Y, Sweeney CJ, et al. : Radium-223 in combination with docetaxel in patients with castration-resistant prostate cancer and bone metastases: A phase 1 dose escalation/randomised phase 2a trial. Eur J Cancer 114:107–116, 2019. 10.1016/j.ejca.2019.04.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Morris MJ, Loriot Y, Sweeney C, et al. : Updated results: A phase I/IIa randomized trial of radium-223 + docetaxel versus docetaxel in patients with castration-resistant prostate cancer and bone metastases. J Clin Oncol 34(15_suppl):5075, 2016. 10.1200/jco.2016.34.15_suppl.5075 [DOI] [Google Scholar]
- 69.Morris MJ, Loriot Y, Fizazi K, et al. : Effects of radium-223 (Ra-223) with docetaxel versus docetaxel alone on bone biomarkers in patients with bone-metastatic castration-resistant prostate cancer (CRPC): A phase I/IIa clinical trial. J Clin Oncol 35(6_suppl):154, 2017. 10.1200/jco.2017.35.6_suppl.154 [DOI] [Google Scholar]
- 70.A study to test Radium-223 with docetaxel in patients with prostate cancer—Full text view—ClinicalTrials.gov. Available at:https://clinicaltrials.gov/ct2/show/NCT03574571. Accessed February 16, 2020.
- 71.Study evaluating the addition of pembrolizumab to Radium-223 in mCRPC—Full text view—ClinicalTrials.gov. Available at:https://clinicaltrials.gov/ct2/show/NCT03093428. Accessed February 16, 2020.
- 72.De Bono JS, Goh JC, Ojamaa K, et al. : KEYNOTE-199: Pembrolizumab (pembro) for docetaxel-refractory metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol 36(15_suppl):5007, 2018. 10.1200/jco.2018.36.15_suppl.5007 [DOI] [Google Scholar]
- 73.Safety and tolerability of atezolizumab (ATZ) in combination with Radium-223 Dichloride (R-223-D) in metastatic castrate-resistant prostate cancer (CRPC) progressed following treatment with an androgen pathway inhibitor—Full text view—ClinicalTrials.gov. Available at:https://clinicaltrials.gov/ct2/show/NCT02814669. Accessed April 6, 2020.
- 74.Israeli RS, Powell CT, Corr JG, et al. : Expression of the prostate-specific membrane antigen1. Cancer Res 54:1807–1811, 1994 [PubMed] [Google Scholar]
- 75.Liu H, Moy P, Kim S, et al. : Monoclonal antibodies to the extracellular domain of prostate-specific membrane antigen also react with tumor vascular endothelium. Cancer Res 57:3629–3634, 1997 [PubMed] [Google Scholar]
- 76.Kozikowski AP, Nan F, Conti P, et al. : Design of remarkably simple, yet potent urea-based inhibitors of glutamate carboxypeptidase II (NAA-LADase) [1]. J Med Chem 44:298–301, 2001. 10.1021/jm000406m [DOI] [PubMed] [Google Scholar]
- 77.Rodriguez CE, Lu H, Dinh TT, et al. : Competitive inhibition of a glutamate carboxypeptidase by phosphonamidothionate derivatives of glutamic acid. Bioorganic Med Chem Lett 9:1415–1418, 1999. 10.1016/S0960-894X(99)00198-5 [DOI] [PubMed] [Google Scholar]
- 78.Maresca KP, Hillier SM, Femia FJ, et al. : A series of halogenated heterodimeric inhibitors of prostate specific membrane antigen (PSMA) as radiolabeled probes for targeting prostate cancer. J Med Chem 52:347–357, 2009. 10.1021/jm800994j [DOI] [PubMed] [Google Scholar]
- 79.Hillier SM, Maresca KP, Lu G, et al. : 99mTc-labeled small-molecule inhibitors of prostate-specific membrane antigen for molecular imaging of prostate cancer. J Nucl Med 54:1369–1376, 2013. 10.2967/jnumed.112.116624 [DOI] [PubMed] [Google Scholar]
- 80.Lu G, Maresca KP, Hillier SM, et al. : Synthesis and SAR of 99mTc/Relabeled small molecule prostate specific membrane antigen inhibitors with novel polar chelates. Bioorganic Med Chem Lett 23:1557–1563, 2013. 10.1016/j.bmcl.2012.09.014 [DOI] [PubMed] [Google Scholar]
- 81.Fendler WP, Calais J, Eiber M, et al. : Assessment of 68Ga-PSMA-11 PET accuracy in localizing recurrent prostate cancer: A prospective single-arm clinical trial. JAMA Oncol 5:856–863, 2019. 10.1001/jamaoncol.2019.0096. American Medical Association [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Afshar-Oromieh A, Holland-Letz T, Giesel FL, et al. : Diagnostic performance of 68Ga-PSMA-11 (HBED-CC) PET/CT in patients with recurrent prostate cancer: Evaluation in 1007 patients. Eur J Nucl Med Mol Imaging 44:1258–1268, 2017. 10.1007/s00259-017-3711-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Tagawa ST, Milowsky MI, Morris M, 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 19:5182–5191, 2013. 10.1158/1078-0432.CCR-13-0231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Vallabhajosula S, Goldsmith SJ, Kostakoglu L, et al. : Radioimmunotherapy of prostate cancer using90Y- and 177Lu-lebeled J591 monoclonal antibodies: Effect of multiple treatments on myelotoxicity.. Clin Cancer Res 11, 2005. 10.1158/1078-0432.CCR-1004-0023 [DOI] [PubMed] [Google Scholar]
- 85.Bander NH, Milowsky MI, Nanus DM, et al. : 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 23:4591–4601, 2005. 10.1200/JCO.2005.05.160 [DOI] [PubMed] [Google Scholar]
- 86.Tagawa ST, Vallabhajosula S, Christos PJ, 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 125:2561–2569, 2019. 10.1002/cncr.32072 [DOI] [PubMed] [Google Scholar]
- 87.Afshar-Oromieh A, Babich JW, Kratochwil C, et al. : The rise of PSMA ligands for diagnosis and therapy of prostate cancer. J Nucl Med 57 (Suppl 3):79S–89S, 2016. 10.2967/jnumed.115.170720 [DOI] [PubMed] [Google Scholar]
- 88.Afshar-Oromieh A, Haberkorn U, Zechmann C, et al. : Repeated PSMA-targeting radioligand therapy of metastatic prostate cancer with 131I-MIP-1095. Eur J Nucl Med Mol Imaging 44:950–959, 2017. 10.1007/s00259-017-3665-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ahmadzadehfar H, Rahbar K, Kürpig S, et al. : Early side effects and first results of radioligand therapy with 177Lu-DKFZ-617 PSMA of castrate-resistant metastatic prostate cancer: A two-centre study. EJNMMI Res 5:114, 2015. 10.1186/s13550-015-0114-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ahmadzadehfar H, Eppard E, Kürpig S, et al. : Therapeutic response and side effects of repeated radioligand therapy with 177Lu-PSMA-DKFZ-617 of castrate-resistant metastatic prostate cancer. Oncotarget 7:12477–12488, 2016. 10.18632/oncotarget.7245 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Baum RP, Kulkarni HR, Schuchardt C, et al. : 177Lu-labeled prostate-specific membrane antigen radioligand therapy of metastatic castration-resistant prostate cancer: Safety and efficacy. J Nucl Med 57:1006–1013, 2016. 10.2967/jnumed.115.168443 [DOI] [PubMed] [Google Scholar]
- 92.Rahbar K, Ahmadzadehfar H, Kratochwil C, et al. : German multicenter study investigating 177Lu-PSMA-617 Radioligand therapy in advanced prostate cancer patients. J Nucl Med 58:85–90, 2017. 10.2967/jnumed.116.183194 [DOI] [PubMed] [Google Scholar]
- 93.Rahbar K, Bögeman M, Yordanova A, et al. : Delayed response after repeated 177Lu-PSMA-617 radioligand therapy in patients with metastatic castration resistant prostate cancer. Eur J Nucl Med Mol Imaging 45:243–246, 2018. 10.1007/s00259-017-3877-z [DOI] [PubMed] [Google Scholar]
- 94.Hofman MS, Violet J, Hicks RJ, et al. : [177 Lu]-PSMA-617 radionuclide treatment in patients with metastatic castration-resistant prostate cancer (LuPSMA trial): A single-centre, single-arm, phase 2 study. Lancet Oncol 19:825–833, 2018. 10.1016/S1470-2045(18)30198-0 [DOI] [PubMed] [Google Scholar]
- 95.Ahmadzadehfar H, Zimbelmann S, Yordanova A, et al. : Radioligand therapy of metastatic prostate cancer using 177Lu-PSMA-617 after radiation exposure to 223Ra-dichloride. Oncotarget 8:55567–55574, 2017. 10.18632/oncotarget.15698 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Yordanova A, Becker A, Eppard E, et al. : The impact of repeated cycles of radioligand therapy using [177Lu]Lu-PSMA-617 on renal function in patients with hormone refractory metastatic prostate cancer. Eur J Nucl Med Mol Imaging 44:1473–1479, 2017. 10.1007/s00259-017-3681-9 [DOI] [PubMed] [Google Scholar]
- 97.Kelly WK, Scher HI, Mazumdar M, et al. : Prostate-specific antigen as a measure of disease outcome in metastatic hormone-refractory prostate cancer. J Clin Oncol 11:607–615, 1993. 10.1200/JCO.1993.11.4.607 [DOI] [PubMed] [Google Scholar]
- 98.Williams S: Surrogate endpoints in early prostate cancer research. Transl Androl Urol 7:472–482, 2018. 10.21037/tau.2018.05.10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Study of 177Lu-PSMA-617 in metastatic castrate-resistant prostate cancer—Full text view—clinicalTrials.gov. Available at:https://clinical-trials.gov/ct2/show/NCT03511664?term=VISION&cond=Prostate+Cancer&draw=2&rank=1. Accessed February 14, 2020.
- 100.Hofman M, Emmett L, Violet JA, et al. : TheraP: A randomized phase II trial of [177 Lu]-PSMA-617 theranostic versus cabazitaxel in progressive metastatic castration-resistant prostate cancer. J Clin Oncol 37 (7_suppl):TPS332, 2019. 10.1200/jco.2019.37.7_-suppl.tps332 [DOI] [PubMed] [Google Scholar]
- 101.Kratochwil C, Giesel FL, Bruchertseifer F, et al. : 213Bi-DOTATOC receptor-targeted alpha-radionuclide therapy induces remission in neuroendocrine tumours refractory to beta radiation: A first-in-human experience. Eur J Nucl Med Mol Imaging 41:2106–2119, 2014. 10.1007/s00259-014-2857-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Kratochwil C, Bruchertseifer F, Giesel FL, et al. : 225Ac-PSMA-617 for PSMA-targeted a-radiation therapy of metastatic castration-resistant prostate cancer. J Nucl Med 57:1941–1944, 2016. 10.2967/jnumed.116.178673 [DOI] [PubMed] [Google Scholar]
- 103.Kratochwil C, Bruchertseifer F, Rathke H, et al. : Targeted a-therapy of metastatic castration-resistant prostate cancer with 225Ac-PSMA-617: Dosimetry estimate and empiric dose finding. J Nucl Med 58:1624–1631, 2017. 10.2967/jnumed.117.191395 [DOI] [PubMed] [Google Scholar]
- 104.Khreish F, Ebert N, Ries M, et al. : 225Ac-PSMA-617/177Lu-PSMA-617 tandem therapy of metastatic castration-resistant prostate cancer: pilot experience. Eur J Nucl Med Mol Imaging 47:721–728, 2020. 10.1007/s00259-019-04612-0 [DOI] [PubMed] [Google Scholar]
- 105.Feuerecker B, Knorr K, Beheshti A, et al. : Safety and efficacy of Ac-225-PSMA-617 in mCRPC after failure of Lu-177-PSMA. J Med Imaging Radiat Sci 50:S20–S21, 2019. 10.1016/j.jmir.2019.03.06631648963 [DOI] [Google Scholar]
- 106.Robertson AKH, Ramogida CF, Schaffer P, et al. : Development of 225 Ac radiopharmaceuticals: TRIUMF perspectives and experiences. Curr Radiopharm 11:156–172, 2018. 10.2174/1874471011666180416161908 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Sathekge M, Knoesen O, Meckel M, et al. : 213Bi-PSMA-617 targeted alpha-radionuclide therapy in metastatic castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 44:1099–1100, 2017. 10.1007/s00259-017-3657-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Study to evaluate the safety, tolerability, pharmacokinetics, and antitumor activity of a thorium-227 labeled antibody-chelator conjugate, in patients with metastatic castration resistant prostate cancer—Full text view—ClinicalTrials.gov. Available at:https://clinicaltrials.gov/ct2/show/NCT03724747?cond=Thorium-227+BAY2315497%2C&draw=2&rank=1. Accessed February 14, 2020. [Google Scholar]
- 109.Timmermand OV, Elgqvist J, Beattie KA, et al. : Preclinical efficacy of hK2 targeted [177Lu]hu11B6 for prostate cancer theranostics. Theranostics 9:2129–2142, 2019. 10.7150/thno.31179 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.McDevitt MR, Thorek DLJ, Hashimoto T, et al. : Feed-forward alpha particle radiotherapy ablates androgen receptor-addicted prostate cancer. Nat Commun 9, 2018. 10.1038/s41467-018-04107-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Safety and targeting of anti-hk2 antibody in mCRPC—Full text view—ClinicalTrials.gov. Available at:https://clinicaltrials.gov/ct2/show/NCT04116164. Accessed April 7, 2020. [Google Scholar]
- 112.Kratochwil C, Flechsig P, Lindner T, et al. : 68 Ga-FAPI PET/CT: tracer uptake in 28 different kinds of cancer. J Nucl Med 60:801–805, 2019. 10.2967/jnumed.119.227967 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Khreish F, Rosar F, Kratochwil C, et al. : Positive FAPI-PET/CT in a metastatic castration-resistant prostate cancer patient with PSMA-negative/FDG-positive disease. Eur J Nucl Med Mol Imaging 1–2, 2019. 10.1007/s00259-019-04623-x [DOI] [PubMed] [Google Scholar]
- 114.Lindner T, Loktev A, Altmann A, et al. : Development of quinoline-based theranostic ligands for the targeting of fibroblast activation protein. J Nucl Med 59:1415–1422, 2018. 10.2967/jnumed.118.210443 [DOI] [PubMed] [Google Scholar]

