touchONCOLOGY touchONCOLOGY
Genitourinary Cancer, Prostate Cancer
Read Time: 13 mins

Targeted Alpha Therapy – Optimising Survival in Advanced Prostate Cancer

Copy Link
Published Online: Dec 21st 2018 European Oncology & Haematology. 2018;14(Suppl 3);3–10
Authors: Johann S De Bono
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

Targeted alpha therapy (TAT) delivers high-energy alpha particles to cancer cells and the tumour microenvironment to treat metastatic castration-resistant prostate cancer (mCRPC) and other cancers. Radium-223 is the first approved TAT in this indication but optimal sequencing with it and other treatments such as sipuleucel-T, cabazitaxel, docetaxel, abiraterone and enzalutamide remains a challenge. Radium-223 selectively binds to areas of increased bone turnover, including metastases, and delivers a powerful dose of radiation with minimal exposure to healthy tissue. Data from clinical studies, especially the pivotal ALpharadin in SYMPtomatic Prostate Cancer (ALSYMPCA) trial (n=921) has shown that radium-223 significantly increases median overall survival (OS), reduces risk of death in mCRPC compared with standard of care and was well tolerated. The ERA-223 study (ClinicalTrials.gov Identifier: NCT02043678; n=806) is currently investigating simultaneous treatment with radium-223 and abiraterone/prednisone, but as the study has produced a safety signal that the treatment increased skeletal fractures and decreased OS, a label change has been issued by the European Medicines Agency in which radium-223 (Xofigo®; Bayer, Leverkusen, Germany) is contraindicated for combination use with abiraterone and prednisone. Nevertheless, radium-223 remains a valuable treatment in mCRPC, especially in suitable sequence with other treatments. In addition, in vitro studies indicate that radium-233 increases anti-tumour activity when used in combination with immunotherapy agents or DNA repair pathway inhibitors. Other promising developments in TAT include the use of isotopes such as thorium-227 or actinium-225 linked to monoclonal antibodies or small peptides/ligands that target specific tumour-associated antigens or other sites. Full data from ongoing studies such as ERA-223 will advance understanding of how radium-223 should be used in mCRPC and how it can further improve outcomes.

Keywords

Alpha-emitter, targeted alpha therapy, radium-223, metastatic, prostate cancer, metastatic castration-resistant prostate cancer, combinations

Article:

Targeted alpha therapy (TAT) is the delivery of high-energy alpha particles to cancer cells and the tumour microenvironment to treat cancer. After a survival benefit was shown for radium-223 versus standard of care in patients with metastatic castration-resistant prostate cancer (mCRPC), radium-223 became the first approved TAT.1–3 In the European Society for Medical Oncology (ESMO) guidelines for urogenital cancer, radium-223 is currently recommended for men with bone-predominant, symptomatic mCRPC without visceral metastases.4

Since the development of radium-223, the therapeutic landscape has changed; several other compounds have also shown a survival advantage in patients with mCRPC. Therefore, the current treatment challenge is how best to sequence radium-223 to optimise survival.5,6 In a satellite symposium, an expert panel explored the place of radium-223 in the treatment of mCRPC based on mechanisms of action, clinical trial experience and case presentations in current clinical settings. Since this symposium took place, the ERA-223 study (ClinicalTrials.gov Identifier: NCT02043678; n=806), which was investigating the simultaneous initiation of radium-223 and abiraterone/prednisone, was unblinded based on a recommendation from the Independent Data Monitoring Committee. The data from the unblinded study showed a safety signal in the treatment arm with regards to both skeletal fractures and overall survival (OS). Based on this data, the European Medicines Agency (EMA) has issued a label change contraindicating the combination of radium-223 and abiraterone/prednisone.7 Despite these recent findings of an experimental combination, TAT remains a valuable and effective treatment in mCRPC with the potential to improve outcomes in many patients with various other treatment combinations under clinical investigation.

Treatment landscape in metastatic castration-resistant prostate cancer

For many years, docetaxel was the only treatment for mCRPC with a demonstrated survival benefit; however, more recently several compounds have been approved for this indication including abiraterone, enzalutamide, radium-223, sipuleucel-T (US only) and cabazitaxel. The approval of these compounds in mCRPC was based on clinical trials showing modest gains in absolute survival both before and after docetaxel, with consistent hazard ratios (HRs) of the newer compounds ranging between 0.63 and 0.78 depending on how early in the disease course treatment was given.8,9

The second-generation androgen pathway inhibitors abiraterone and enzalutamide were initially approved for patients with symptomatic, progressive mCRPC following docetaxel chemotherapy after demonstrating improved OS versus placebo and standard of care.10–13 Building on this experience, abiraterone and enzalutamide were assessed in asymptomatic or mildly symptomatic metastatic disease, and were shown to improve OS versus standard of care, thus extending the use of both compounds to before docetaxel in mCRPC.14,15 At the same time, the approval of radium-223 was supported by clinical data in patients with mCRPC demonstrating an OS benefit and delayed onset of symptomatic skeletal events for patients treated with radium-223 compared with placebo and standard of care.16,17 The survival benefit was observed for radium-223 versus standard of care when given to patients with mCRPC and no visceral metastases either as an initial treatment or second-line after docetaxel chemotherapy. Patients with metastatic prostate cancer with high volume disease may benefit from docetaxel chemotherapy while they are hormone-naive, but in mCRPC docetaxel is often reserved for patients with progressive symptomatic disease after receiving abiraterone/enzalutamide and radium-223 treatment.18–20

Radium-223 – mechanisms of action

Radium-223 is an example of a mechanism-mediated TAT. This involves the selective targeting of tumour cells in specific organs or tissues based on inherent chemical properties.5,21,22 As a calcium mimetic, the alpha-emitting radioisotope radium-223 is taken up by bone where it selectively binds to areas of increased bone turnover, including bone metastases. It is administered intravenously and travels from the bloodstream to deliver a radioactive ‘payload’ to tumour cells and other cells in the bone microenvironment.2,3,5,6 With radium-223, 95% of energy emission is via cell-killing alpha particles, but the accompanying 1% gamma emission is enough to observe distribution with a gamma camera as demonstrated in clinical trials. Gamma imaging after radium-223, which is not required for everyday practice, shows that a powerful dose of radiation is delivered directly to the sources of bone metastases.2,23–25 Alpha particles form ionising radiation at a short range (50–100 μm), which effectively destroys small clusters of isolated cancer cells, with minimal exposure to healthy tissue.26 Off-target exposure is also minimised as <2% of radium-223 absorbed into hydroxyapatite of bone becomes dislodged.27 The advantages of alpha-emitters over beta-emitters include a shorter range, higher linear energy transfer and the ability to cause double-stranded DNA breaks that result in cell cycle arrest and cell death (Table 1).26,28–35

Tumour growth and bone remodelling in mCRPC involves a vicious cycle of abnormal osteoblastic bone growth in prostate cancer-derived bone metastases, and paracrine factors from prostate cancer cells that affect osteoblasts and osteoclasts.36,37 The resulting abnormal bone remodelling stimulates osteoblasts to secrete factors that promote the growth of bone metastases, which further stimulates osteoclast activity. The mechanisms of action by which radium-223 disrupts this vicious cycle have been demonstrated in a mouse model of osteolytic breast cancer and, more recently, in two mouse xenograft models of advanced prostate cancer.29,30 In the mouse xenograft models, radium-223 inhibited tumour-induced osteoblastic bone growth, protected normal bone architecture and reduced bone volume.29 In tumour-bearing tibiae, double-stranded DNA breaks increased in tumour cells at 24 and 72 hours after one injection of radium-223 compared with the vehicle control, and double-stranded DNA breaks were increased in osteoblasts and osteoclasts at 72 hours.29

Potential synergies between the mechanisms of action of radium-223 and those of various other anti-cancer agents are of particular interest. These could conceivably create highly effective treatment combinations in prostate and other cancer types.38

Radium-223 – pivotal trial data

The approval of radium-223 in the US and EU in 2013 was based on the pivotal ALpharadin in SYMPtomatic Prostate Cancer (ALSYMPCA) trial (n=921), which demonstrated an OS benefit compared with the standard of care in patients with bone predominant mCRPC.16,17 In this trial, patients with mCRPC received radium-223 before or after docetaxel, with the choice of initial treatment based on whether a patient was fit enough for chemotherapy. The final results from the trial showed that the risk of death was reduced by 30% for radium-233 compared with standard of care, and the median OS for radium-223 and standard of care was 14.9 months and 11.3 months, respectively – a 30% improvement of 3.6 months (HR: 0.70; p<0.001; Figure 1).16,31–5 In addition, the median time to first symptomatic skeletal event was 15.6 months with radium-223 versus 9.8 months with standard of care (HR: 0.66). Radium-223 also reduced the need for external beam radiation therapy for bone pain (HR: 0.67) and the risk of spinal cord compression (HR: 0.52) compared with standard of care.39

In the ALSYMPCA trial, substantial declines in prostate-specific antigen (PSA) levels were not demonstrated in the radium-223 group, and PSA levels were not a useful measure of the response to treatment.40 However, declines in total alkaline phosphatase (tALP) were observed in 87% of the radium-223 group by week 12, compared with 23% of the standard of care group, and the dynamic profile of tALP was suggestive of a rapid and progressive inhibition of ALP production by osteoblasts. Although tALP levels correlated with OS, the biomarker was not established as a surrogate of disease progression.40

The 3-year safety follow up of the ALSYMPCA trial showed that radium-223 was well tolerated with a low incidence of grade 3 or 4 myelosuppression.41 The haematological treatment-emergent adverse events anaemia, neutropenia and thrombocytopenia were 13%, 2% and 7%, respectively in the radium-223 group, and 13%, 1% and 2%, respectively in the standard of care group. The incidence of myelosuppression remained low, and radium-223 continued to be well tolerated with no new safety signals.41

Contemporary use of radium-223

At the Advanced Prostate Cancer Consensus Conference in March 2017 a panel of about 60 experts was asked their opinion on the optimal method of treating advanced prostate cancer based on the current literature. Panellists with clinical experience of using radium-223 provided consensus regarding the measurement of biomarkers, imaging for staging and treatment monitoring.42

The majority of panellists supported measuring PSA every 3–4 weeks (40%) or every 2–4 months (40%), and measuring tALP every 3–4 weeks (47%) or every 2–4 months (35%) during radium-223 treatment (Figure 2). However, based on the ALSYMPCA trial, it was agreed that there were no biomarkers for response to treatment in mCRPC. When asked about preferred methods of imaging for staging and treatment monitoring during radium-223 treatment, the panel opted for computed tomography (CT) and bone scintigraphy (75%), and next-generation imaging (23%) such as whole-body magnetic resonance imaging (MRI) with diffusion imaging and/or positron emission tomography (PET)/CT with prostate cancer-specific PET radiotracers, choline or fluciclovine. In terms of timing of imaging, panellists chose from the following options: 1) imaging every 3–4 months during and after treatment (41%); 2) 6 months after the completion of radium-223 treatment and every 3–4 months thereafter (27%); or 3) 6 months after the completion of radium-223 treatment and follow-up imaging at clinical or biochemical progression (24%).

The panel was asked for their preferred approach to treating men who are receiving radium-223 for mCRPC and have disease progression outside of the bone but had not previously received a second-generation androgen pathway inhibitor. Of the available options, 52% chose to add abiraterone or enzalutamide, 20% chose to add abiraterone or enzalutamide only in selected patients, and 26% chose to stop treatments including radium-223.42 It should be noted that this voting occurred before the 27 July 2018 label guidance from the European Medicines Agency (EMA)’s Committee for Medicinal Products for Human Use, which considered data from the phase III ERA-223 study (ClinicalTrials.gov Identifier: NCT02043678; n=806).43 Based on this data, the EMA Pharmacovigilance Risk Assessment Committee (PRAC) ruled that in the EU, a combination of radium-223 and abiraterone (Zytiga®, Janssen, Beerse, Belgium) with prednisone/prednisolone should be stopped and a contraindication has been added to the label.7

Case presentations

The use of radium-223 in a contemporary setting has been described in case studies of men at different stages of advanced prostate cancer. In each case described below, favourable biochemical and clinical responses were observed in patients who received radium-223 that was layered on to treatment with a second-generation androgen inhibitor. The treatment approach in these case examples was different than that studied in ERA-223, in which radium-223 and abiraterone plus prednisone were commenced at the same time in all patients who were randomised to receive this combination. The case examples differed as the combination of the two drug classes occurred upon disease progression. Nonetheless, the PRAC recommendation has contraindicated any combination of radium-223 and abiraterone/prednisone.

Abiraterone added to radium-223 in advanced prostate cancer

A 76-year-old male was diagnosed with metastatic advanced prostate cancer and was started on androgen deprivation therapy and denosumab in March 2012; in December 2012, rising PSA levels indicated castration resistance. In January 2013, docetaxel was started resulting in a PSA decline of ≥50% that was sustained for 3 months. In September 2013, the patient developed increasing pelvic pain and had rising PSA levels and was re-staged using a bone and CT scan, and found to have stable disease. MRI of the long-spine excluded significant epidural disease. In October 2013, the patient was started on radium-223 under the expanded access programme.

With each infusion of radium-223, both PSA and tALP increased within the normal ranges. In December 2013, after three doses of radium-223, on CT scan the prostate tumour had increased in size with possible rectal wall infiltration; on bone scan there was possible necrosis of the jaw, which was later confirmed. Abiraterone was layered on to radium-223, which resulted in a rapid decrease in PSA, decreased prostate size and no new bone lesions. The patient continued with abiraterone for 3 years, after which his disease progressed and he received second-line cabazitaxel.

The clinical observations in the case presentation suggest that in men receiving radium-223 CT should be considered after three doses or if soft tissue progression is suspected. PSA and tALP should be monitored in patients receiving radium-223, although they are not markers of disease progression.

Radium-223 layered on after progression on enzalutamide in asymptomatic metastatic castration-resistant prostate cancer

A 61-year-old male presented with T3 prostate cancer and a PSA of 500 ng/ml after responding to androgen deprivation therapy for 9 years. The patient received eight cycles of docetaxel and maintained reasonable control for 4 months, until at 5 months PSA levels started to increase. The patient was re-staged and found to have asymptomatic bone metastases and no visceral involvement. The patient received six cycles of enzalutamide and after an initial period of stability PSA levels increased. Radium-223 and denusomab were layered on to enzalutamide. Within 3 months of starting radium-223, the patient’s PSA levels decreased to 0.1 ng/ml and 18 months later (currently) the patient was still responding to treatment (Figure 3).

Radium-223 layered on to enzalutamide in rapidly progressing symptomatic metastatic castration-resistant prostate cancer

A 67-year-old male was very ill when he presented with nocturia and long-term worsening back pain. His sister had previously died of neutropenic sepsis during chemotherapy for breast cancer. The patient’s bone scan showed diffuse bone involvement and stage T3 prostate cancer with a PSA of 2,000 ng/ml and alkaline phosphatase (ALP) 6,000 U/l. The patient started hormone therapy and his back-pain continued for which analgesia was needed; the patient refused docetaxel. After 3 months of androgen deprivation therapy, the patient’s PSA nadir fell from 2,000 to 50 ng/ml; tALP started to rise rapidly so zoledronic acid was started, to which the tALP levels responded well, decreasing from about 6,000 to 1,000 U/l. After a further 3 months, the patient had advanced high-volume prostate cancer and rising PSA levels, for which he was receiving androgen deprivation therapy and zoledronic acid, and he was continuing to refuse cytotoxic chemotherapy. Enzalutamide was started and external radiation to the pelvis was given to control pain. For 2 months of enzalutamide treatment, pain remained controlled and PSA levels continued to increase above 300 ng/ml with stable tALP, before a rapid progression of biochemical markers by 3 months. The patient then received radium-223 which was layered on to enzalutamide, zoledronic acid and androgen deprivation therapy, and after six cycles, PSA declined to about 100 ng/ml with a good pain response (Figure 4). The patient remained pain free but died 12 months after starting radium-223 after developing rapidly progressive disease.

Developments in targeted alpha therapy

Radium-223 is currently the only TAT that is approved for clinical use in mCRPC. In addition to this indication, it is currently under investigation in breast cancer (phase II [ClinicalTrials.gov Identifiers: NCT02258464, NCT02258451, NCT02366130]) and multiple myeloma (phase I [ClinicalTrials.gov Identifier: NCT02928029]). Radium-223 and other TATs are also under investigation in various combinations and indications (Table 2).

An alternative experimental approach to mCRPC therapy is to give radium-233 and immunotherapy in combination. These different treatments may act synergistically and reverse the immunosuppressive response affecting osteoclasts within bone metastases. In an in vitro study, human prostate, breast and lung carcinoma cell lines were exposed to sub-lethal levels of radium-223, and within 96 hours each tumour type had increased cytotoxic T lymphocyte-mediated lysis of tumour cells; that is, radium-223 did not kill the tumour cells – rather it induced the production of cytotoxic T lymphocytes.44 Lysis of each tumour type by CD8+ cytotoxic T lymphocytes was accompanied by augmented protein expression of major histocompatibility compex I (MHC-I) and other molecules that mediate antigen presentation. These data suggest that radium-223 can induce immunogenic modulation in tumour cells that survive irradiation.44

Further interesting potential combinations with radium-223 include various inhibitors of DNA repair pathways, including poly (ADP-ribose) polymerase (PARP) inhibitors, and drugs that target kinases (e.g. ATR, ATM, DNA-PK). These agents may be potent radiosensitisers, producing synergistic anti-tumour activity with TAT.45,46 At a recent American Association for Cancer Research Annual Meeting, data were presented showing that in an in vivo model mimicking mCRPC (intratibial LNCaP-Luc prostate cancer model), there was a synergistic anti-tumour effect between the ATR kinase inhibitor BAY 1895344 and radium-223. This was demonstrated by substantially lower levels of serum PSA and procollagen I N-terminal peptide when this combination was added compared with BAY 1895344 or radium-223 alone or vehicle (Figure 5).47

The flexibility of targeting mechanisms with some alpha-emitters allows for a potential range of applications across various tumour types using novel delivery methods.48 One such approach is to conjugate alpha-emitters to monoclonal antibodies or antibody fragments that target tumour cells.26 The potential of this strategy has been demonstrated in various studies, including an animal model that used a conjugate of thorium-227 to trastuzumab to specifically target ovarian cancer cells.49 A further in vivo study used thorium-227, which was conjugated to an anti-CD33 antibody, and was shown to bind with high affinity to the sialic acid receptor CD33. This approach provides a means of selectively targeting alpha doses to myeloid cells in the treatment of acute myeloid leukemia.50

An additional novel delivery method is created by conjugating the alpha-emitter to small molecules or peptides to target tumour cells. Ligands, synthetic protein scaffolds (e.g. affibodies) and substrate analogues (e.g. peptides) can be used as targeting agents.26 Other investigational compounds such as actinium-225 are chelated to a peptide that binds to the prostate-specific membrane antigen (PSMA) enzyme on target prostatic cells, thereby delivering alpha particles to the tumour.26,43

A further interesting example of TAT in mCRPC treatment is the use of PSMA-617-targeted actinium-225. This treatment is currently being investigated in mCRPC patients as part of an off-trial compassionate access programme. In this programme, two patients achieved a complete response on imaging, with PSA levels falling from 2,923 ng/ml to <0.1 ng/ml despite widespread metastatic disease.51,52 This suggested that actinium-225–PSMA-617 has the potential to benefit patients with advanced prostate cancer. No haematological toxicity has been reported, but it should be noted that six patients (43%) had grade 1 xerostomia, and two patients (14%) had grade 2 xerostomia.

Overall, these advances in the targeted delivery of radionuclides and the increased availability of alpha-emitters have led to a number of recent clinical trials of TAT in prostate cancer, as well as other cancers such as breast and multiple myeloma. Given the flexibility of targeted delivery methods and the direct cytotoxic effects of alpha-emitters, TAT could potentially change the treatment paradigm in several cancer indications.

Conclusions

The proof-of-concept that TAT improves OS with minimal toxicity in patients with advanced prostate cancer was first established during the clinical development of radium-223. More recently, several compounds from other classes have been approved in mCRPC that have changed the treatment landscape and expanded the possibilities for sequencing and combining treatments with different mechanisms of action. Physician experience from real-world clinical practice suggests that, in certain circumstances, radium-223 and the second-generation androgen pathway inhibitors may have potentially synergistic mechanisms of action. In the case presentations, layering radium-223 onto enzalutamide or abiraterone upon clinical progression (or layering on abiraterone to radium-223) provided a good clinical response in men with advanced prostate cancer. However, the use of these combinations has been the subject of a contraindication issued by the EMA, which recommended against any combination of radium-223 with abiraterone and prednisone.7 It should be noted that in the case examples presented above, radium-223 was layered onto the novel anti-hormonal treatment upon progression, whereas in the ERA-223 study patients were given the combination from the outset, before progression occurred (ClinicalTrials.gov Identifier: NCT02043678)43. It is unknown whether toxicities or potential efficacy arising from layering are different from those arising from combining therapies at initiation of treatment, or if there is a difference between abiraterone plus prednisone and enzalutamide when given in combination. Current clinical trials are not comparing these different treatment regimens, so may not fully resolve this issue. Nevertheless, data from ERA-223 and other ongoing studies, as well as greater experience from real-world clinical practice, should better define the optimal use of radium-233 with other treatments in the management of patients with mCRPC and may improve understanding of its role in managing this increasingly prevalent disease.

Article Information:
Disclosure

Johann de Bono reports personal fees from AstraZeneca,
Astellas, Bayer, Celgene, Daiichi-Sankyo, Genentech, Genmab,
GlaxoSmithKline, Janssen, Merck Sharp & Dohme, Merck Serono, Pfizer,
Roche, Sanofi-Aventis and Terumo outside the submitted work.

Review Process

This article reports the proceedings of a sponsored
satellite symposium and, as such, has not been subject to this journal’s
usual peer-review process, but was reviewed for scientific accuracy by
the symposium speaker and Editorial Board before publication.

Authorship

The named author meets the International Committee
of Medical Journal Editors (ICMJE) criteria for authorship of this
manuscript, takes responsibility for the integrity of the work as a whole,
and has given final approval for the version to be published.

Correspondence

Johann de Bono, The Institute of Cancer Research,
Drug Development Unit, Sycamore House, 15 Cotswold Road,
Sutton, Surrey, SM2 5NG, UK. E: johann.de-bono@icr.ac.uk

Support

The publication of this article was supported by Bayer, who were given the opportunity to review the article for scientific accuracy before submission. Any resulting changes were made at the expert reviewer’s discretion.

Access

This article is published under the Creative Commons
Attribution Non-commercial License, which permits any non-commercial
use, distribution, adaptation, and reproduction provided the original
author and source are given appropriate credit. © The Author 2018.

Acknowledgements

Medical writing support was provided by James Gilbart of
Touch Medical Media and funded by Bayer. This article reports the proceedings of
a sponsored satellite symposium held at the Congress of the European Society for
Medical Oncology (ESMO) 2017 annual meeting, Madrid, Spain, 8–12 September
2017, and has been updated to ensure the content is timely at the time of press.

Received

2018-10-03T00:00:00

References

  1. Allen BJ, Huang CY, Clarke RA. Targeted alpha anticancer therapies: update and future prospects. Biologics. 2014;8:255–67.
  2. Bayer AG. Xofigo 1100 kBq/mL solution for injection. Summary of product characteristics. Available at: www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/002653/WC500156172.pdf (accessed 15 October 2017).
  3. Bayer HealthCare Pharmaceuticals. Xofigo (radium Ra 223 dichloride) Injection, for intravenous use. Prescribing Information. 2016. Available at: www.accessdata.fda.gov/drugsatfda_docs/label/2013/203971lbl.pdf (accessed 15 October 2017).
  4. ESMO Guidelines Committee. Prostate cancer eUpdate. Ann Oncol. 2016;27(Suppl 5):v146–7.
  5. O’Sullivan JM. Radium-223 from bench to bedside – future directions for targeted alpha therapy. European Oncology & Haematology. 2018;14(Suppl 2):3–9.
  6. Parker C, Heidenreich A, Nilsson S, Shore N. Current approaches to incorporation of radium-223 in clinical practice. Prostate Cancer Prostatic Dis. 2018;21:37–47.
  7. European Medicines Agency. Prostate cancer medicine Xofigo must not be used with Zytiga and prednisone/prednisolone - Ongoing clinical study shows increased risk of death and fractures with the combination - EMA/176752/2018, 2018, 2018. Available at: www.ema.europa.eu/docs/en_GB/document_library/Referrals_document/Xofigo_20/Under_evaluation/WC500245467.pdf (accessed 15 April 2018).
  8. Picciotto M, Franchina T, Russo A, et al. Emerging role of radium-223 in the growing therapeutic armamentarium of metastatic castration-resistant prostate cancer. Expert Opin Pharmacother. 2017;18:899–908.
  9. Zhao S, Yu EY, Castrate-resistant prostate cancer: post-docetaxel management. Curr Opin Urol. 2013;23:201–7.
  10. Caffo O, Veccia A, Kinspergher S, Maines F. Abiraterone acetate and its use in the treatment of metastatic prostate cancer: a review. Future Oncol. 2018;14:431–42.
  11. de Bono JS, Logothetis CJ, Molina A, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. 2011;364:1995–2005.
  12. Nadal R, Bellmunt J. The evolving role of enzalutamide on the treatment of prostate cancer. Future Oncol. 2016;12:607–16.
  13. Ryan CJ, Smith MR, de Bono JS, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med. 2013;368:138–48.
  14. Beer TM, Armstrong AJ, Rathkopf DE, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 2014;371:424–33.
  15. Scher HI, Fizazi K, Saad F, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367:1187–97.
  16. Parker C, Nilsson S, Heinrich D, et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med. 2013;369:213–23.
  17. 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. 2014;15:738–46.
  18. National Comprehensive Cancer Network. Guidelines for patients with prostate cancer, 2018. Available at: www.nccn.org/patients/guidelines/prostate/index.html (accessed 10 December 2018).
  19. Parker C, Gillessen S, Heidenreich A, et al. Cancer of the prostate: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26 Suppl 5:v69–77.
  20. Puente J, Grande E, Medina A, et al. Docetaxel in prostate cancer: a familiar face as the new standard in a hormone-sensitive setting. Ther Adv Med Oncol. 2017;9:307–18.
  21. Marques IA, Neves AR, Abrantes AM, et al. Targeted alpha therapy using Radium-223: From physics to biological effects. Cancer Treat Rev. 2018;68:47–54.
  22. Nuhn P, De Bono JS, Fizazi K, et al. Update on systemic prostate cancer therapies: management of metastatic castration-resistant prostate cancer in the era of precision oncology. Eur Urol. 2018; pii: S0302-2838(18)30246-X. [Epub ahead of print].
  23. Flux GD. Imaging and dosimetry for radium-223: the potential for personalized treatment. Br J Radiol. 2017;90:20160748.
  24. Nilsson S, Larsen RH, Fossa SD, et al. First clinical experience with alpha-emitting radium-223 in the treatment of skeletal metastases. Clin Cancer Res. 2005;11:4451–9.
  25. Wright CL, Monk JP 3rd, Murrey DA Jr, Hall NC. Real-time scintigraphic assessment of intravenous radium-223 administration for quality control. Biomed Res Int. 2015;2015:324708.
  26. Dekempeneer Y, Keyaerts M, Krasniqi A, et al. Targeted alpha therapy using short-lived alpha-particles and the promise of nanobodies as targeting vehicle. Expert Opin Biol Ther. 2016;16:1035–47.
  27. Henriksen G, Fisher DR, Roeske JC, et al. Targeting of osseous sites with alpha-emitting 223Ra: comparison with the beta-emitter 89Sr in mice. J Nucl Med. 2003;44:252–9.
  28. Brechbiel MW. Targeted alpha-therapy: past, present, future? Dalton Trans. 2007;4918–28.
  29. 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. 2017;23:4335–46.
  30. Suominen MI, Rissanen JP, Käkönen R, et al. Survival benefit with radium-223 dichloride in a mouse model of breast cancer bone metastasis. J Natl Cancer Inst. 2013;105:908–16.
  31. Ceder J, Elgqvist J. Targeting prostate cancer stem cells with alpha-particle therapy. Front Oncol. 2016;6:273.
  32. Elgqvist J, Frost S, Pouget JP, Albertsson P. The potential and hurdles of targeted alpha therapy - clinical trials and beyond. Front Oncol. 2014;3:324.
  33. GE Healthcare, Medi-Physics Inc. Metastron (strontium-89 chloride injection). Prescribing Information, 2013. Available at: www.accessdata.fda.gov/drugsatfda_docs/label/2013/020134s012lbl.pdf (19 December 2018).
  34. Lantheus Medical Imaging, Inc. Quadramet (samarium-153 lexidronam) Prescribing Information, 2017. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/020570s008lbl.pdf (accessed 19 December 2018).
  35. Hoskin P, Sartor O, O’Sullivan JM, et al. Efficacy and safety of radium-223 dichloride in patients with castration-resistant prostate cancer and symptomatic bone metastases, with or without previous docetaxel use: a prespecified subgroup analysis from the randomised, double-blind, phase 3 ALSYMPCA trial. Lancet Oncol. 2014;15:1397–406.
  36. Cook LM, Shay G, Araujo A, Lynch CC. Integrating new discoveries into the “vicious cycle” paradigm of prostate to bone metastases. Cancer Metastasis Rev. 2014;33:511–25.
  37. Keller ET, Brown J. Prostate cancer bone metastases promote both osteolytic and osteoblastic activity. J Cell Biochem. 2004;91:718–29.
  38. 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. 2018;16:149–54.
  39. De Luca R, Costa RP, Tripoli V, et al. The clinical efficacy of radium-223 for bone metastasis in patients with castration-resistant prostate cancer: an Italian clinical experience. Oncology. 2018;94:161–6.
  40. Sartor O, Coleman RE, Nilsson S, et al. An exploratory analysis of alkaline phosphatase, lactate dehydrogenase, and prostate-specific antigen dynamics in the phase 3 ALSYMPCA trial with radium-223. Ann Oncol. 2017;28:1090–7.
  41. 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. 2017;73:427–35.
  42. Gillessen S, Attard G, Beer TM, et al. Management of patients with advanced prostate cancer: the report of the Advanced Prostate Cancer Consensus Conference APCCC 2017. Eur Urol. 2017;73:178–211.
  43. Umbricht CA, Benesova M, Schmid RM, et al. 44Sc-PSMA-617 for radiotheragnostics in tandem with 177Lu-PSMA-617-preclinical investigations in comparison with 68Ga-PSMA-11 and 68Ga-PSMA-617. EJNMMI Res. 2017;7:9.
  44. Malamas AS, Gameiro SR, Knudson KM, Hodge JW. Sublethal exposure to alpha radiation (223Ra dichloride) enhances various carcinomas’ sensitivity to lysis by antigen-specific cytotoxic T lymphocytes through calreticulin-mediated immunogenic modulation. Oncotarget. 2016;7:86937–47.
  45. Marechal A, Zou L. DNA damage sensing by the ATM and ATR kinases. Cold Spring Harb Perspect Biol. 2013;5: pii: a012716.
  46. O’Connor MJ. Targeting the DNA damage response in cancer. Mol Cell. 2015;60:547–60.
  47. Wengner AM, Siemeister G, Luecking U, et al. Synergistic in vivo activity of the ATR inhibitor BAY 1895344 in combination with the targeted alpha therapy radium-223 dichloride in a preclinical tumor model mimicking bone metastatic castration-resistant prostate cancer (mCRPC). Presented at: American Association for Cancer Research (AACR) Annual Meeting, Washington, DC, USA, 14–18 April 2018; Poster 838. Available at: www.pharmatest.com/files/4815/2526/1411/Final_Wengner_AACR_2018_poster_838_website.pdf (accessed 19 December 2018).
  48. UroToday-GUOncToday. ESMO 2017: The future of targeted alpha therapy, 2017. Available at: www.urotoday.com/conference-highlights/esmo-2017/esmo-2017-prostate-cancer/98343-esmo-2017-the-future-of-targeted-alpha-therapy.html (accessed 21 June 2018).
  49. Heyerdahl H, Abbas N, Sponheim K, et al. Targeted alpha therapy with 227Th-trastuzumab of intraperitoneal ovarian cancer in nude mice. Curr Radiopharm. 2013;6:106–16.
  50. Hagemann UB, Wickstroem K, Wang E, et al. In vitro and in vivo efficacy of a novel CD33-targeted thorium-227 conjugate for the treatment of acute myeloid leukemia. Mol Cancer Ther. 2016;15:2422–31.
  51. Kratochwil C, Bruchertseifer F, Rathke H, et al. Targeted alpha-therapy of metastatic castration-resistant prostate cancer with (225)Ac-PSMA-617: dosimetry estimate and empiric dose finding. J Nucl Med. 2017;58:1624–31.
  52. Zustovich F, Barsanti R. Targeted α therapies for the treatment of bone metastases. Int J Mol Sci. 2018;19: pii: E74.

Further Resources

Share this Article
Related Content In Prostate Cancer
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72