Download presentation
Presentation is loading. Please wait.
1
CONVEGNO URO-ONCOLOGIA 7 Novembre 2015
Il controllo degli Eventi scheletrici Nel paziente con tumore alla prostata Metastatico alle ossa Fabio Fulfaro Ricercatore Universitario Policlinico Paolo Giaccone Palermo
2
METASTASI OSSEE epidemiologia
~30% dei pazienti con tumore metastatico sviluppa metastasi ossee clinicamente evidenti nel corso della malattia Un ulteriore 50% di queste localizzazioni può essere osservato al tavolo autoptico 80% di tutti i casi derivano da primitività mammaria, prostatica e polmonare
3
METASTASI OSSEE Incidenza
Primary Mieloma Rene Tiroide Mammella Polmone Vescica Melanoma Prostata % 65 – 75 75 60 20 – 25 > LITICHE > ADDENSANTI Coleman RE. Cancer Treat Rev. 2001;27:
8
Bone Markers in Osteolytic and Osteosclerotic Metastatic Bone Disease
60 500 50 400 40 300 Bone-specific alkaline phosphatase (ng/mL) 30 N-telopeptide (BCE/M Cr) 200 20 100 10 Lytic Blastic Mixed Lytic Blastic Mixed X-ray pattern X-ray pattern Lipton A. Semin Oncol. 2012;28:54-59.
11
Bone metastases can have serious and debilitating consequences SREs
Skeletal-related events (SREs) are defined as:1,2 A composite SRE endpoint is commonly used in clinical trials to evaluate the efficacy of bone-targeted agents2 Pathological fractures may be symptomatic or identified by imaging assessments Recently, symptomatic skeletal events (SSEs) has been used as an alternative study endpoint for skeletal complications3 Pathological fractures only included if clinically symptomatic Hypercalcaemia of malignancy is an additional potential complication of bone metastases4 Radiation to bone Pathological fracture Spinal cord compression Surgery to bone Bone metastases can result in serious and debilitating SREs.1 Since the late 1990s, SREs have been defined as spinal cord compression, radiation to bone, pathological fracture or surgery to bone.2 Spinal cord compression is considered an oncological emergency and suspected cases require urgent evaluation and treatment3 Radiotherapy to bone is often administered as palliative therapy for bone pain4 Pathological fractures can result when metastases destruct bone and reduce the load-bearing capacity; painful microfractures may develop into full fractures3 Surgery to bone may be required to stabilise pathological fractures or pre-empt the occurrence of fractures in vulnerable areas.3 A composite SRE endpoint is often used in clinical trials to evaluate the efficacy of bone-targeted agents to prevent skeletal complications in patients with bone metastases.2 Symptomatic skeletal events (SSEs) was used as an alternative skeletal endpoint in a recently reported Phase III trial of radium-223 in men with castration-resistant prostate cancer (CRPC) and bone metastases.5 The definition of SSE differs from SRE in that pathological fractures are required to be clinically apparent (symptomatic) whereas they may be identified by periodic radiological review to be classified as an SRE. Hypercalcaemia of malignancy is an additional potential complication of bone metastases.3 References Saad F, Gleason DM, Murray R, et al. Long-term efficacy of zoledronic acid for the prevention of skeletal complications in patients with metastatic hormone-refractory prostate cancer. J Natl Cancer Inst 2004;96:879–2. (Accessed August 2014). Coleman RE. Clinical features of metastatic bone disease and risk of skeletal morbidity. Clin Cancer Res 2006;12:6243s–9s. Chow E, Harris K, Fan G, et al. Palliative radiotherapy trials for bone metastases: a systematic review. J Clin Oncol 2007;25:1423–36. 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. 1. Saad F, et al. J Natl Cancer Inst 2004;96:879–82; 2. (Accessed August 2014); 3. Sartor O, et al. Lancet Oncol 2014;15:738−46; 4. Coleman RE, et al. Clin Cancer Res 2006;12:6243s−9s. 11
12
Cumulative incidence of on-study SREs
SREs are a common complication in patients with solid tumours and bone metastases Cumulative incidence of on-study SREs in patients with newly diagnosed bone metastases 1 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 6 12 18 24 30 36 Study month Cumulative incidence of on-study SREs Breast cancer (n = 621) Lung cancer (n = 477) Prostate cancer (n = 721) SREs are a common complication in patients with solid tumours and bone metastases.1 A review of medical records from two large US health systems was conducted to assess the risk of SREs (spinal cord compression, pathological fracture, radiation to bone, bone surgery) in patients with solid tumours (breast cancer: n = 621; lung cancer: n = 477; prostate cancer: n = 721) in routine clinical practice.1 Cumulative incidence of SREs in patients with newly diagnosed bone metastases was estimated, accounting for death as a competing risk.1 Intravenous (IV) bisphosphonates were received by 55.8%, 14.8% and 20.2% of patients with breast cancer, lung cancer and prostate cancer, respectively.1 More than one half of patients with bone metastases had evidence of SREs (breast cancer: 62.6%; lung cancer: 58.7%; prostate cancer: 51.7%) either at diagnosis of bone metastases or subsequently.1 Reference Oster G, Lamerato L, Glass AG, et al. Natural history of skeletal-related events in patients with breast, lung, or prostate cancer and metastases to bone: a 15-year study in two large US health systems. Support Care Cancer 2013;21:327986. Use of intrvenous bisphosphonates: breast cancer, 55.8%; lung cancer, 14.8%; and prostate cancer 20.2%. Oster G, et al. Support Care Cancer 2013;21:327986.
14
Skeletal Complications in Metastatic Bone Disease Are Significant
% of patients affected in PLACEBO arms of: Pamidronate trials ZOMETA® trials Disease Breast Myeloma Prostate Others Observation time 12 months 9 months 15 months 9 months Radiation to bone Fractures Hypercalcaemia of malignancy Surgery to bone Spinal cord compression
15
SREs are a substantial burden to patients and healthcare systems
Direct consequences to patients Pathological fracture1 Spinal cord compression1 Surgery to bone1 Radiation to bone for bone pain1 Indirect consequences to patients Poorer physical, functional and emotional status2 Reduced HRQoL2 Hospital visits/stays3 Health resource utilisation Increased treatment costs4 Increased hospital visits/stays3 SREs are a substantial burden to patients and healthcare systems. Direct consequences to patients include: the need for radiation to bone for bone pain; pathological fracture; spinal cord compression; and surgery to bone.1 Additional, indirect consequences to patients include negative effects on physical, functional and emotional status;2 the requirement to undergo hospital visits and stays;3 and an overall reduction in health-related quality of life (HRQoL).2 SREs are also associated with substantial healthcare resource utilisation due to higher treatment costs vs advanced cancer without SREs4 and the need for hospital visits and inpatient stays.3 References Costa L and Major PP. Effect of bisphosphonates on pain and quality of life in patients with bone metastases. Nat Clin Prac Oncol 2009;6:163–74. DePuy V, Anstrom KJ, Castel LD, et al. Effects of skeletal morbidities on longitudinal patient-reported outcomes and survival in patients with metastatic prostate cancer. Support Care Cancer 2007;15:869–76. Pocket RD, Castellano D, McEwan P, et al. The hospital burden of disease associated with bone metastases and skeletal-related events in patients with breast cancer, lung cancer, or prostate cancer in Spain. Eur J Cancer Care 2010;19:755–60. Delea T, McKiernan J, Brandman J, et al. Retrospective study of the effect of skeletal complications on total medical care costs in patients with bone metastases of breast cancer seen in typical clinical practice. J Support Oncol 2006;4:341–7. Impact on patients and healthcare systems 1. Costa L and Major PP. Nat Clin Prac Oncol 2009;6:163–74; 2. De Puy V, et al. Support Care Cancer 2007;15:869–76; 3. Pocket RD, et al. Eur J Cancer Care 2010;19:755–60; 4. Delea T, et al. J Support Oncol 2006;4:341–7. HRQoL, health-related quality of life.
16
LA MALATTIA METASTATICA OSSEA Obiettivi del Trattamento
Obiettivi primari Prevenire gli eventi scheletrici Ridurre al minimo l’invalidità Dare sollievo dal dolore Migliorare la funzionalità Migliorare la qualità di vita Limitare gli eventi avversi Migliorare l’accettabilità del paziente Considerare il suo stile di vita Ridurre i costi di gestione della terapia Benché i bifosfonati attualmente disponibili siano efficaci nel trattare e prevenire alcuni eventi scheletrici, la loro utilità clinica è limitata da problematiche di tollerabilità (in particolare la tollerabilità renale) e modalità di somministrazione. Recentemente, in pazienti trattati con alcuni bifosfonati è stato osservato un aumento del rischio di osteonecrosi mascellare/mandibolare. Permane il bisogno clinico di un trattamento che unisca a un'efficacia e ad una tollerabilità superiori una maggiore flessibilità di somministrazione.
17
Opzioni terapeutiche nella malattia metastatica ossea
Terapia locoregionale Terapia sistemica Radioterapia Chirurgia Interventistica (vertebroplastica) Ormonoterapia Chemioterapia Denosumab Terapia radiometabolica Terapia con bifosfonati
18
tra cellule neoplastiche e osteoclasti Liberazione di fattori
Interazioni tra cellule tumorali e osteoclasti: circolo vizioso o opportunità terapeutica? METASTASI OSSEE Interazioni tra cellule neoplastiche e osteoclasti Aumento del riassorbimento osseo Stimolazione degli osteoclasti Liberazione di fattori solubili ad azione attivante sugli osteoclasti (es. PTH-rP) OSTEOCLASTA Liberazione di fattori di stimolazione della crescita tumorale (es. TGF-b) Cell. Tumorale
19
Bifosfonati disponibili per la malattia metastatica ossea
1a generazione - Clodronato orale 2a generazione - Pamidronato endovena 3a generazione - Acido zoledronico endovena - Ibandronato endovena e orale Cristofanilli M, Hortobagyi GN. Cancer Control 1999; 6 (3):
20
Bifosfonati Trattamento dell’ipercalcemia maligna
Riduzione degli eventi traumatici patologici o incremento del tempo mediano al primo evento patologico Controllo del dolore osseo e riduzione dell’uso di analgesici Riduzione dei trattamenti radioterapici analgesici/preventivi Miglioramento della qualità della vita
21
Ac zoledronico: la riduzione del dolore metastatico osseo nel ca prostatico
1.2 1.0 0.8 0.6 0.4 0.2 Variazione media rispetto al basale Tempo trascorso nello studio (mesi) Ac zoledronico 4 mg (n=214) Placebo (n=208) P<0,05 Saad F et al. J Nat Cancer Inst , 19:
22
Sicurezza dei bifosfonati nella malattia metastatica ossea
importanza di assicurare una tollerabilità ottimale del trattamento con bifosfonati nella terapia palliativa Differenti profili di sicurezza dei bifosfonati Effetti indesiderati frequenti - eventi legati all’infusione (reazione di fase acuta) - tossicità /deterioramento funzione renale - effetti indesiderati gastrointestinali (gastrite, diarrea) - osteonecrosi della mandibola
23
Bone-targeted therapy was delayed in a substantial proportion of patients
Reasons for delaying treatment with bone-targeted agents † † The audit also showed that bone-targeted therapy was delayed in a substantial proportion of patients.1 The main reason for delaying bone-targeted treatment was that bone metastases were thought to be responding to systemic anti-tumour therapies (56% of total patient population).1 Additionally, bone-targeted treatment was delayed in 31% of patients due to safety concerns; within this group, these concerns included existing renal impairment (61%), dental health issues (28%) and poor risk/benefit ratio (34%).1 Reference 1. Casas A, Lebret T, Cavo M, et al. 146 study: cost of skeletal-related events by country. Support Care Cancer 2012;20(Suppl 1):1–283:abstract 368 (and poster). Data from a prospective, observational chart audit conducted in France, Germany, Italy, Spain and the UK in March 2010; data were available for 14,871 patients. Denosumab was not available at the time of the audit. †Includes poor life expectancy, poor performance status, etc Casas A, et al. Support Care Cancer 2012;20(Suppl 1):1–283:abstract 368 (and poster).
24
A substantial proportion of patients with bone metastases never received bone-targeted therapy for prevention of SREs Reasons why patients were expected never to receive bone-targeted agents (N = 1351) Despite the substantial burden of SREs to patients and healthcare systems, a substantial proportion of patients with bone metastases never receive bone-targeted therapy for the prevention of SREs. This slide shows data from a prospective European chart audit including 881 physicians who completed brief and detailed questionnaires on a total of 17,193 patients during a 2–3-week observational period in March It is important to note that denosumab was not available at the time of the audit. Of the 14,871 patients for whom data were available from the brief questionnaire, 17% were expected never to receive bone-targeted agents despite 72% having a moderate-to-high risk of SREs.1 In further sub-set analyses of the 1351 patients who were expected never to receive bone-targeted agents, short life expectancy (38%), renal issues (37%) and poor risk/benefit ratio (34%) were identified as the most common reasons why treatment with bone-target therapy was not considered.1 Reference 1. Casas A, Lebret, T Cavo M, et al. 146 study: cost of skeletal-related events by country. Support Care Cancer 2012;20(Suppl 1):1–283:abstract 368 (and poster). 17% of patients were expected never to receive bone-targeted agents despite 72% having a moderate-to-high SRE risk† Data from a prospective, observational chart audit conducted in France, Germany, Italy, Spain and the UK in March 2010; data were available for 14,871 patients. Denosumab was not available at the time of the audit. BM, bone metastases. †As determined by the treating physician. Casas A, et al. Support Care Cancer 2012;20(Suppl 1):1–283:abstract 368 (and poster).
25
Prevention of skeletal morbidity in metastatic bone disease
New ESMO clinical practice guidelines on bone health in cancer patients Prevention of skeletal morbidity in metastatic bone disease Guidance on bone-targeted treatment (denosumab or zoledronic acid) Initiation Commence at diagnoses of metastatic bone disease In all patients with breast cancer or CRPC, whether they are symptomatic or not In selected patients with advanced lung cancer, renal cancer and other solid tumours if life expectancy > 3 months and considered at high risk of SREs Continuation Continue indefinitely throughout the course of the disease Ongoing treatment is recommended for patients with progression of underlying bone metastases, a recent SRE and/or elevated bone resorption markers† The European Society for Medical Oncology (ESMO) has set out new clinical practice guidelines on bone health in patients with cancer. These guidelines highlight the value of bone-targeted treatments (denosumab or zoledronic acid) for the prevention of skeletal morbidity in metastatic bone disease.1 The initiation of therapy is recommended on diagnosis of metastatic bone disease in all patients with breast cancer or CRPC, irrespective of the presence of symptoms, in order to delay the first SRE and reduce subsequent complications from metastatic bone disease. Bone-targeted therapy is also recommended in selected patients with advanced lung cancer, renal cancer, and other solid tumours, if they have a life expectancy of more than 3 months and there is a high risk of SREs.1 It is recommended that bone-targeted treatment be continued throughout the course of the disease, particularly in patients with progression of underlying bone metastases, recent SREs and/or elevated bone resorption markers.1 Results from clinical trials evaluating potential clinical applications of bone markers (e.g. helping to identify patients at high risk for bone metastasis or bone lesion progression) are awaited to clarify the value of bone markers in clinical practice. Reference Coleman R, Body JJ, Aapro M, et al. Bone health in cancer patients: ESMO Clinical Practice Guidelines. Ann Oncol 2014 [Epub ahead of print]. †Results from clinical trials evaluating potential clinical applications of bone markers (e.g. helping to identify patients at high risk for bone metastasis or bone lesion progression) are awaited to identify the true value of bone markers in clinical practice. Coleman R, et al. Ann Oncol 2014 [Epub ahead of print].
26
Denosumab and prevention of skeletal-related events in adults with bone metastases
This slide set summarises key data on denosumab 120 mg subcutaneously (SC) every 4 weeks (Q4W) for skeletal-related event (SRE) prevention in adults with bone metastases from solid tumours. *▼ This medicinal product is subject to additional monitoring. All suspected adverse reactions should be reported. DMO-IHQ-AMG July-NP 26
27
Denosumab binds to RANK Ligand to block osteoclast differentiation and activation
‘Vicious cycle’ RANK Ligand Denosumab Osteoblasts In healthy adult bone, receptor activator of NF-κB (RANK) Ligand activity is tightly regulated to balance bone formation and resorption.1 However, in the presence of tumour cells, a vicious cycle of bone destruction and tumour growth may develop.2,3 Tumour cells that have invaded bone secrete factors that increase the expression of RANK Ligand by osteoblasts Increased expression of RANK Ligand upregulates osteoclast activity Excessive osteoclast activity drives increased bone resorption; in turn, this releases growth factors from the bone matrix that may perpetuate tumour activity This sequence of events drives a vicious cycle of bone destruction and tumour activity. Denosumab is a fully human monoclonal antibody that precisely binds to and sequesters free RANK Ligand, thus preventing activation of the RANK receptor and blocking the development of activated osteoclasts. Inhibiting RANK Ligand in this way mimics the endogenous action of osteoprotegerin (OPG) on the RANK/RANK Ligand pathway in inhibiting the formation, function and survival of bone-resorbing osteoclasts.13 Because bone destruction may lead to SREs, it can be hypothesised that targeting RANK Ligand with denosumab could prevent or delay SREs in patients with bone metastases. References Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and activation. Nature 2003;423:337–42. Roodman GD. Mechanisms of bone metastasis. N Engl J Med 2004;350:1655–64. Mundy GR. Metastasis to bone: causes, consequences and therapeutic opportunities. Nat Rev Cancer 2002;2:584–93. Osteoclast Growth factors Growth factors Ca2+ Tumour Adapted from Roodman GD. N Engl J Med 2004;350:1655–64; Mundy GR. Nat Rev Cancer 2002;2:584–93; McClung MR, et al. N Engl J Med 2006;354:821–31. RANK, receptor activator of NF-κB . 27
28
Pre-planned integrated analysis4 (N = 5723) Other solid tumours*/MM3
Three pivotal Phase III trials of denosumab vs zoledronic acid in patients with bone metastases from advanced cancer Primary endpoint: time to first on-study SRE (non-inferiority) Secondary endpoints: time to first on-study SRE (superiority); time to first and subsequent on-study SRE; safety and tolerability R A N D O M I S T Breast cancer1 Denosumab 120 mg SC Q4W + Placebo IV Q4W† Prostate cancer2 Pre-planned integrated analysis4 (N = 5723) Supplemental calcium and vitamin D Zoledronic acid 4 mg IV Q4W† + Placebo SC Q4W Other solid tumours*/MM3 Denosumab has been evaluated for SRE prevention in 3 large international, randomised, double-blind, double-dummy, active-controlled Phase III studies in patients with bone metastases (breast cancer, n = 2049; prostate cancer, n = 1904; other solid tumours or multiple myeloma, n = 1776).1−3 Patients were randomised to:1−3 Denosumab 120 mg SC and placebo IV Q4W or Zoledronic acid 4 mg IV and placebo SC Q4W. In line with the zoledronic acid prescribing information, agents administered IV (zoledronic acid or placebo) were dose-adjusted on the basis of baseline creatinine clearance ≤ 60 mL/min and held for renal function deterioration until serum creatinine returned to within 10% of baseline values. There was no requirement for dose adjustment with denosumab.1−3 Daily supplementation with calcium (≥ 500 mg) and vitamin D (≥ 400 IU) was strongly recommended.1−3 Key inclusion criteria were:1−3 Radiographic evidence of ≥ 1 bone metastasis or bone disease Eastern Cooperative Oncology Group (ECOG) performance status of 0, 1 or 2 Adequate organ function Life expectancy ≥ 6 months. Key exclusion criteria included the use of current or prior IV bisphosphonates for treatment of bone metastases.1−3 The primary endpoint was time to first on-study SRE (non-inferiority), with predefined secondary endpoints including time to first on-study SRE (superiority), time to first and subsequent on-study SRE, safety and tolerability. 1−3 Secondary endpoints were tested simultaneously with multiplicity adjustment (Hochberg procedure2) only after the primary endpoint was met (hierarchical order). This procedure is accepted by regulatory bodies When the primary endpoint was met, a superiority test on time to first on-study SRE was performed as a secondary endpoint.1−3 References Stopeck AT, Lipton A, Body JJ, et al. Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. J Clin Oncol 2010;28:5132–9. Fizazi K, Carducci M, Smith M, et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet 2011;377:813–22. Henry DH, Costa L, Goldwasser F, et al. Randomized, double-blind study of denosumab versus zoledronic acid in the treatment of bone metastases in patients with advanced cancer (excluding breast and prostate cancer) or multiple myeloma. J Clin Oncol 2011;29:1125−32. †Per protocol and Zometa® label, IV product dose adjusted for baseline creatinine clearance and subsequent dose intervals determined by serum creatinine. 1. Stopeck AT, et al. J Clin Oncol 2010;28:5132–9; 2. Fizazi K, et al. Lancet 2011;377:813–22; 3. Henry DH, et al. J Clin Oncol 2011;29:112532; 4. Lipton A, et al. Eur J Cancer 2012;48:3082–92. *Excluding breast and prostate. 28
29
Patients with a broad range of solid tumour types were enrolled
Baseline characteristic, n (%) or median Zoledronic acid (n = 2861) Denosumab (n = 2862) Women 1349 (47.2) 1316 (46.0) Age, years 63.0 ECOG status of 0 or 1 2546 (89.0) 2585 (90.3) Tumour type* Breast 1020 (35.7) 1026 (35.8) Prostate 951 (33.2) 950 (33.2) Non-small cell lung 352 (12.3) 350 (12.2) Multiple myeloma 93 (3.3) 87 (3.0) Renal 85 (3.0) 70 (2.4) Small cell lung 48 (1.7) 61 (2.1) Other 312 (10.9) 318 (11.1) Time from first bone metastasis to randomisation, months 2.30 2.17 Previous SRE† 1157 (40.4) 1112 (38.9) Patients with a broad range of solid tumour types were enrolled in the Phase III trials. In addition to the dedicated studies in breast and prostate cancer, the ‘other solid tumours and multiple myeloma’ study included a large number of patients with non-small cell lung cancer, renal carcinoma and small cell lung cancer, among others.1 Baseline characteristics, including age, sex, ECOG performance status, time from first bone metastasis to randomisation, and previous SRE were well balanced between the treatment groups.1 Approximately 40% of patients had experienced a previous SRE.1 Reference Lipton A, Siena S, Rader M, et al. Superiority of denosumab to zoledronic acid for prevention of skeletal-related events: a combined analysis of 3 pivotal, randomised, phase 3 trials. Eur J Cancer 2012;48:308292. Lipton A, et al. Eur J Cancer 2012;48:308292. *ECOG, Eastern Cooperative Oncology Group; †Based on randomisation. 29
30
Patients without SRE (%)
Time to first on-study SRE reached almost 28 months in patients on denosumab Time to first on-study SRE HR = 0.83 (95% CI, 0.76–0.90) P < (superiority) Time (months) Patients without SRE (%) 40 60 80 100 6 12 18 24 30 27.66 months 19.45 months 90 70 50 20 10 Time to first on-study SRE reached almost 28 months in patients on denosumab.1 The median time to first on-study SRE was months in patients on denosumab compared with months in patients on zoledronic acid − a difference of 8.21 months (P < 0.001, superiority).1 Denosumab was associated with a 17% risk reduction in first SRE over zoledronic acid (HR = 0.83; 95% CI, 0.76−0.90; P < 0.001, superiority).1 Reference Lipton A, Siena S, Rader M, et al. Superiority of denosumab to zoledronic acid for prevention of skeletal-related events: a combined analysis of 3 pivotal, randomised, phase 3 trials. Eur J Cancer 2012;48:308292. Denosumab Zoledronic acid (N = 5723) Lipton A, et al. Eur J Cancer 2012;48:308292. HR, hazard ratio. 30
31
Time to first and subsequent on-study SREs
Denosumab also significantly reduced the total number of SREs vs zoledronic acid Time to first and subsequent on-study SREs RR = 0.82 (95% CI, 0.75–0.89) P < (superiority) 3 6 9 12 15 18 21 24 27 30 33 36 Cumulative mean number of SREs per patient Time (months) 0.0 1.0 1.6 1.4 1.2 0.8 0.6 0.4 0.2 Total SREs 1360 1628 Denosumab Zoledronic acid (N = 5723) There were significantly fewer on-study SREs with denosumab vs zoledronic acid (1360 vs 1628 in total).1 Denosumab treatment was associated with an 18% risk reduction in first and subsequent SREs over zoledronic acid (P < 0.001, superiority). Reference Lipton A, Siena S, Rader M, et al. Superiority of denosumab to zoledronic acid for prevention of skeletal-related events: a combined analysis of 3 pivotal, randomised, phase 3 trials. Eur J Cancer 2012;48:308292. Lipton A, et al. Eur J Cancer 2012;48:308292. Events occurring at least 21 days apart (multiple event analysis). RR, rate ratio. 31
32
Denosumab delayed pain worsening by almost 2 months vs zoledronic acid
Time to moderate or severe pain (> 4 points) among patients with no or mild pain (0–4 points) at baseline (n = 2683)† HR = 0.83 (95% CI, 0.760.92) P < 0.001 mths Median time from no or mild pain to moderate or severe pain (months) 2683 (52%) of 5198 patients included in a pooled pain analysis reported no or mild pain at baseline (Brief Pain Inventory-Short Form [BPI-SF] worst pain score 0−4 points). Among these patients, denosumab delayed the median time to onset of moderate or severe pain (worst pain score > 4 points) by 1.8 months compared with zoledronic acid (6.5 vs 4.7 months) and reduced the risk of experiencing moderate or severe pain by 17% (HR, 0.83; 95% CI, 0.76–0.92; P < 0.001).1 Note that patients with multiple myeloma were excluded from the pooled pain analysis because denosumab is not indicated in patients with multiple myeloma.1 Reference von Moos R, Body JJ, Egerdie B, et al. Pain and health-related quality of life in patients with advanced solid tumours and bone metastases: integrated results from three randomized, double-blind studies of denosumab and zoledronic acid. Support Care Cancer 2013;21:3497507. von Moos R, et al. Support Care Cancer 2013;21:3497507. †BPISF worst pain score. Patients with MM were not included in this analysis.
33
Safety results of interest
Patient incidence, n (%) Zoledronic acid (n = 2836) Denosumab (n = 2841) Infectious AEs 1218 (42.9) 1233 (43.4) Infectious serious AEs 309 (10.9) 329 (11.6) Acute phase reactions (first 3 days) 572 (20.2) 246 (8.7) Cumulative rate of ONJ 37 (1.3) 52 (1.8) Year 1 15 (0.5) 22 (0.8) Year 2 28 (1.0) 51 (1.8) Hypocalcaemia 141 (5.0) 273 (9.6) New primary malignancy 18 (0.6) AEs leading to study discontinuation 280 (9.9) 270 (9.5) 572 (20.2) 246 (8.7) 37 (1.3) 52 (1.8) This slide shows safety results of interest in the pivotal Phase III SRE prevention trials. There were fewer acute phase reactions among patients on denosumab (8.7%) than zoledronic acid (20.2%).1 These were defined as a flu-like syndrome including pyrexia, chills, flushing, bone pain, arthralgias and myalgias, which have been associated with IV bisphosphonate use, as per the zoledronic acid prescribing information. Hypocalcaemia (corrected serum calcium [CSC] levels < 9.0 mg/dL) was more frequent with denosumab (9.6%) than zoledronic acid (5.0%).1 There was no significant difference between the two treatment groups in the cumulative rate of osteonecrosis of the jaw (ONJ).1 There was no pattern in the type of new malignancies diagnosed on study.1 Reference Lipton A, Siena S, Rader M, et al. Superiority of denosumab to zoledronic acid for prevention of skeletal-related events: a combined analysis of 3 pivotal, randomised, phase 3 trials. Eur J Cancer 2012;48:308292. 141 (5.0) 273 (9.6) Lipton A, et al. Eur J Cancer 2012;48:308292. ONJ, osteonecrosis of the jaw. 33
34
Higher rate of hypocalcaemia with denosumab vs zoledronic acid
In the pivotal Phase III trials most hypocalcaemia events were asymptomatic1,3−5 In the post-marketing setting, severe symptomatic hypocalcaemia (including fatal cases) has been reported, with most cases occurring in the first weeks of therapy2 The higher rate of hypocalcaemia with denosumab is consistent with the more potent antiresorptive effect vs zoledronic acid4 Hypocalcaemia1,2 Zoledronic acid (n = 2836) Denosumab (n = 2841) Overall (integrated analysis), % 5.0 9.6 Grade 3, %† 1.2 2.5 Grade 4, %† 0.2 0.6 Hypocalcaemia is a known side effect of drugs that reduce bone remodelling.1 In the pivotal Phase III SRE prevention trials, hypocalcaemia occurred more frequently with denosumab than zoledronic acid.2,3 Overall, hypocalcaemia was reported in 9.6% of patients on denosumab and 5% of patients on zoledronic acid Grade 3 or 4 hypocalcaemia was reported in 3.1% of patients on denosumab and 1.3% of patients on zoledronic acid Most hypocalcaemia events in patients on denosumab were asymptomatic.1,2,4,5 Of note, in the post-marketing setting, severe symptomatic hypocalcaemia (including fatal cases) has been reported.3 The higher rate of hypocalcaemia observed with denosumab compared with zoledronic acid is consistent with its more potent antiresorptive effect.5 References Fizazi K, Carducci M, Smith M, et al. A randomised, double-blind study of denosumab versus zoledronic acid in the treatment of bone metastases in men with castration-resistant prostate cancer. Lancet 2011;377:813–22. Lipton A, Fizazi K, Stopeck AT, et al. Superiority of denosumab to zoledronic acid for prevention of skeletal-related events: a combined analysis of 3 pivotal, randomized, phase 3 trials. Eur J Cancer 2012;48:3082–92. XGEVA® (denosumab) Summary of Product Characteristics. Amgen. Stopeck AT, Lipton A, Body JJ, et al. Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. J Clin Oncol 2010;28:5132–9. Henry DH, Costa L, Goldwasser F, et al. A randomized, double-blind study of denosumab versus zoledronic acid in the treatment of bone metastases in patients with advanced cancer (excluding breast and prostate cancer) or multiple myeloma. J Clin Oncol 2011;29:1125–32. †Grade 3 defined as: CSC < 7.0– 6.0 mg/dL; Ionised calcium < 0.9–0.8 mmol/L; hospitalisation indicated; Grade 4 defined as: CSC < 6.0 mg/dL; Ionised calcium < 0.8 mmol/L; life-threatening consequences. 1. Lipton A, et al. Eur J Cancer 2012;48:3082–92; 2. Denosumab (XGEVA®) Summary of Product Characteristics, Amgen; 3. Stopeck AT, et al. J Clin Oncol 2010;28:5132–9; 4. Henry DH, et al. J Clin Oncol 2011;29:1125–32; 5. Fizazi K, et al. Lancet 2011;377:813–22. 34
35
Minimising the risk of ONJ
Before starting denosumab, a dental examination with appropriate preventative dentistry is recommended Do not start denosumab in patients with an active dental jaw condition requiring surgery, or in patients who have not recovered following oral surgery Tell patients receiving denosumab to maintain good oral hygiene practices, receive routine dental check-ups, and immediately report any oral symptoms such as dental mobility, pain or swelling Risk factors include: invasive dental procedures, poor oral hygiene or other pre-existing dental disease, advanced malignancies, infections, older age, concomitant therapies (e.g. chemotherapy, corticosteroids, angiogenesis inhibitors, radiotherapy to the head and neck) smoking, and previous bisphosphonate therapy In patients with risk factors for ONJ, an individual benefit-risk assessment should be performed before initiating therapy This slide highlights guidance on minimising the risk of ONJ during denosumab treatment as outlined in the denosumab 120 mg Q4W SmPC.1 A dental examination with appropriate preventative dentistry is recommended before starting denosumab and treatment should not be initiated in patients with an active dental jaw condition requiring surgery, or in those who have not recovered following oral surgery.1 Routine dental check-ups should be performed. Additionally, patients should be educated about good oral hygiene and on the signs and symptoms of ONJ; they should be informed to report any oral symptoms immediately.1 Risk factors for ONJ have been identified and an individual benefit-risk assessment should be performed before starting therapy in patients with know risk factors.1 Reference 1. Denosumab (XGEVA®) Summary of Product Characteristics, Amgen. Denosumab (XGEVA®) Summary of Product Characteristics, Amgen.
36
Minimising the risk of hypocalcaemia
Pre-existing hypocalcaemia must be corrected prior to initiating therapy Supplementation with calcium and vitamin D is required in all patients unless hypercalcaemia is present ( mg Ca UI Vit D) Prior to the initial dose of denosumab Within 2 weeks after the initial dose If suspected symptoms of hypocalcaemia occur Monitoring of calcium levels should be conducted: Risk of hypocalcaemia increases with the degree of renal impairment Consider monitoring calcium levels more frequently in patients with risk factors for hypocalcaemia (e.g. patients with severe renal impairment, creatinine clearance < 30 ml/min), or if otherwise indicated based on the clinical condition of the patient Tell patients to report symptoms of hypocalcaemia If hypocalcaemia occurs, additional calcium supplementation and monitoring may be necessary This slide highlights guidance on minimising the risk of hypocalcaemia during denosumab treatment as outlined in the denosumab 120 mg Q4W Summary of Product Characteristics (SmPC). Reference 1. Denosumab (XGEVA®) Summary of Product Characteristics, Amgen. Denosumab (XGEVA®) Summary of Product Characteristics, Amgen.
37
Denosumab Versus Zoledronic Acid for Treatment of Bone Metastases in Men With Castration-Resistant Prostate Cancer: A Randomised, Double-Blind Study
38
Study Design: International, Randomised, Double-Blind, Active-Controlled Study
XGEVA™ PI 2010: 9,14 Fizazi K, et al. Lancet 2011;377: 815,Table 1 Key Inclusion Criteria Castration-resistant prostate cancer and 1 bone metastases Key Exclusion Criteria Current or prior IV bisphosphonate treatment N = 950 denosumab 120 mg SC and placebo IV Q4W XGEVA™ PI 2010: 2,2.1 Fizazi K, et al. Lancet 2011;377: 815,A,1 Fizazi K, et al. Lancet 2011;377: 814,B,1-2 Fizazi K, et al. Lancet 2011;377: 815,A,2 N = 951 zoledronic acid 4 mg IV* and placebo SC Q4W XGEVA™ PI 2010: 9,14 Fizazi K, et al. Lancet 2011;377: 813,Methods Key Points This was an international, randomized, double-blind, active-controlled trial comparing denosumab with zoledronic acid for the treatment of bone metastases in patients with castration-resistant prostate cancer The primary endpoint was time to first on-study SRE comparing denosumab with zoledronic acid for noninferiority Secondary efficacy endpoints, evaluated only if noninferiority was demonstrated, were superiority tests comparing denosumab and zoledronic acid for time to first on-study SRE and time to first and subsequent SRE(s) (multiple-event analysis) Background Eligible patients were men 18 years old with histologically confirmed prostate cancer and current or prior radiographic evidence of at least one bone metastasis and documented failure of at least 1 hormonal therapy Patients were randomized 1:1 to receive either SC injections of denosumab 120 mg and an IV placebo Q4W, or an IV infusion (lasting no less than 15 minutes) of zoledronic acid 4 mg and an SC injection of placebo Q4W Daily supplementation with calcium and vitamin D was strongly recommended Key exclusion criteria included current or prior IV bisphosphonate or oral bisphosphonate administration to treat bone metastasis References XGEVA™ (denosumab) prescribing information, Amgen. Fizazi K, Carducci M, Smith M, et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet. 2011;377: Primary Endpoint Time to first on-study skeletal-related event (SRE) (noninferiority) Secondary Endpoints Time to first on-study SRE (superiority) Time to first and subsequent on-study SRE(s) (superiority) Fizazi K, et al. Lancet 2011;377: 815,A,2 XGEVA™ PI 2010: 9,14 Fizazi K, et al. Lancet 2011;377: 814,A,2;B,1 *Per protocol and Zometa® label, IV product dose adjusted for baseline creatinine clearance and subsequent dose intervals determined by serum creatinine. No SC dose adjustments made due to increased serum creatinine. Fizazi K, et al. Lancet. 2011;377:813–822. Fizazi K, et al. Lancet 2011;377: 814,B,5; 815,A,1 XGEVA™ PI 2010: 2,2.1 Fizazi K, et al. Lancet 2011;377: 815,A,1 XGEVA™ PI 2010: 3,6.1 Fizazi K, et al. Lancet 2011;377: 814,B,1-2
39
Baseline Characteristics
Fizazi K, et al. Lancet 2011;377: 815,Table 1 Characteristic Denosumab (N = 950) Zoledronic Acid (N = 951) Median age, years (IQR) 71 (64–77) 71 (66–77) ECOG performance status of 0 or 1, n (%) 882 (93) 886 (93) Stratification Factors PSA at randomisation 10 g/L, n (%) 805 (85) 806 (85) Recent chemotherapy ( 6 weeks before randomisation), n (%) 132 (14) Previous SRE, n (%) 232 (24) 231 (24) Median time from diagnosis of bone metastasis to randomisation, months (IQR) 3.94 (1.22–15.67) 5.19 (1.31–16.10) Key Points A total of 1901 patients were randomized to receive denosumab (n = 950) or zoledronic acid (n = 951) Baseline age, race, and ECOG performance status variables were balanced between groups Background Randomization was stratified by previous SRE, PSA level ( 10 g/L vs 10 g/L), and chemotherapy for prostate cancer within 6 weeks before randomization Patients in the zoledronic acid arm had a slightly longer median time from bone metastases diagnosis to study randomization (denosumab, 3.94 months; zoledronic acid, 5.19 months) although the quartiles were similar Reference Fizazi K, Carducci M, Smith M, et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet. 2011;377: Fizazi K, et al. Lancet 2011;377: 815,Table 1 IQR = interquartile range. ECOG = Eastern Cooperative Oncology Group. PSA = prostate-specific antigen. Fizazi K, et al. Lancet. 2011;377:813–822.
40
Primary Endpoint: Time to First On-Study SRE
XGEVA™ PI, 2010: 11,Table 2 Fizazi K, et al. Lancet 2011;377: 816,Figure 2 HR = 0.82 (95% CI, 0.71–0.95) P (noninferiority) P = (superiority) 1.00 0.75 Proportion of Subjects Without SRE 0.50 Kaplan-Meier Estimate of Median Months 0.25 Key Points Denosumab significantly delayed the time to first on-study SRE by 18% compared with zoledronic acid (HR = 0.82; 95% CI, 0.71–0.95; P .001 for noninferiority and P = .008 for superiority)1 The median (95% CI) time to first on-study SRE was 20.7 months in the denosumab group and 17.1 months in the zoledronic acid group, a difference of 3.6 months1 Between-group divergence is evident beginning at 3 months after initiation of treatment2 References XGEVA™ (denosumab) prescribing information, Amgen. Fizazi K, Carducci M, Smith M, et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet. 2011;377: Denosumab Zoledronic acid 20.7 XGEVA™ PI, 2010: 11,Table 2 17.1 0.00 3 6 9 12 15 18 21 24 27 Fizazi K, et al. Lancet 2011;377: 816,Figure 2 Study Month Patients at Risk: Zoledronic acid 951 733 544 407 299 207 140 93 64 47 Denosumab 950 758 582 472 361 259 168 115 70 39 Fizazi K, et al. Lancet. 2011;377:813–822.
41
Cumulative Mean Number of SREs per Patient
Secondary Endpoint: Time to First and Subsequent On-Study SRE(s)* (Multiple-Event Analysis) XGEVA™ PI, 2010: 11,Table 2 Fizazi K, et al. Lancet 2011;377: 816,Figure 3 2.0 Rate ratio = 0.82 (95% CI, 0.71–0.94) P = (superiority) 1.8 1.6 1.4 1.2 Cumulative Mean Number of SREs per Patient 1.0 0.8 0.6 Key Points Denosumab significantly delayed the time to first and subsequent on-study SREs (rate ratio = 0.82; 95% CI, 0.71–0.94; adjusted P = .009)1 A total of 1078 events occurred, 494 in the denosumab treatment group, and 584 in the zoledronic acid treatment group2 References XGEVA™ (denosumab) prescribing information, Amgen. Fizazi K, Carducci M, Smith M, et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet. 2011;377: XGEVA™ PI, 2010: 11,Table 2 Events 0.4 Denosumab 494 Fizazi K, et al. Lancet 2011;377: 816,Figure 3 0.2 Zoledronic acid 584 0.0 3 6 9 12 15 18 21 24 27 30 33 36 Study Month *Events occurring at least 21 days apart. Fizazi K, et al. Lancet. 2011;377:813–822.
42
Summary of Adverse Events
Fizazi K, et al. Lancet 2011;377: 818,Table 4 Patient Incidence Denosumab (N = 943) n (%) Zoledronic Acid (N = 945) n (%) Infectious AEs* 402 (43) 375 (40) Acute phase reactions (first 3 days) 79 (8) 168 (18) Renal AEs† 139 (15) 153 (16) Cumulative rate of osteonecrosis of the jaw (ONJ)‡ 22 (2) 12 (1) Year 1 10 (1) 5 (1) Year 2 8 (1) Hypocalcaemia 121 (13) 55 (6) New primary malignancy 18 (2) Fizazi K, et al. Lancet 2011;377: 819,A,3 Fizazi K, et al. Lancet 2011;377: 818,Table 4 Key Point The incidence of AEs of interest was similar between the two study treatment groups Background During the first 3 days of treatment, AEs potentially associated with acute phase reactions occurred in 8% of denosumab and 18% of zoledronic acid patients AEs potentially associated with renal impairment occurred in 15% of the denosumab group and 16% of the zoledronic acid group Positively adjudicated ONJ occurred in 22 (2%) patients in the denosumab group and 12 (1%) patients in the zoledronic acid group (P = .09) AEs of hypocalcemia were reported in 121 (13%) patients in the denosumab group and 55 (6%) patients in the zoledronic acid group New primary malignancies were identified in 18 (2%) patients in the denosumab group and 10 (1%) patients in the zoledronic acid group Reference Fizazi K, Carducci M, Smith M, et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet. 2011;377: Fizazi K, et al. Lancet 2011;377: 819,A,3 Fizazi K, et al. Lancet 2011;377: 818,Table 4 *Based on Medical Dictionary for Regulatory Activities (MedDRA; version 12.1) system organ class categorization of infections and infestations. †Includes renal failure, increased blood creatinine, acute renal failure, renal impairment, increased blood urea, chronic renal failure, oliguria, hypercreatininaemia, anuria, azotemia, decreased creatinine renal clearance, decreased urine output, abnormal blood creatinine, proteinuria, decreased glomerular filtration rate, and nephritis. ‡P = 0.09. Fizazi K, et al. Lancet. 2011;377:813–822.
43
Summary Denosumab was superior to zoledronic acid in delaying the time to First SRE First and subsequent SRE(s) Notable adverse events occurring in both treatment groups included ONJ and hypocalcaemia Hypocalcaemia was more frequent in the denosumab arm Incidence of ONJ was similar between arms and not statistically significantly different Regarding Denosumab administration: Administered as a 120 mg SC injection once every 4 weeks No dose adjustment necessary for patients with renal impairment Fewer acute phase reactions in the denosumab arm Key Points The results of this study demonstrated that denosumab was superior to zoledronic acid in delaying time to first and subsequent SRE(s) in advanced prostate cancer patients with bone metastases Denosumab had several potentially beneficial characteristics
44
Radioisotopes Targets Bone Metastases
Naturally targets new bone growth in and around bone metastases Most acts as a calcium mimic Strontium-89 Samrarium-153 Radium-223 Ca Ra
45
Range of alpha-particle
Radium-223 (AlpharadinTM ) Based on alpha emitter Radium-223 ideal half-life of 11.4 days Excreted via small bowel Safe and easy to produce, deliver and handle Alpha Beta Relative particle mass 7000 1 Initial energy (MeV) 3-8 Range in tissue (µm) 40-90 LET (KeV/µm) 60-230 Charge +2 -1 Ion pairs/µm 5-20 DNA hits to kill cell 1-5 Range of alpha-particle Radium-223
46
ALSYMPCA (ALpharadin in SYMptomatic Prostate CAncer) Phase III Study Design
TREATMENT 6 injections at 4-week intervals PATIENTS R A N D OM I S E D 2:1 STRATIFICATION Confirmed symptomatic CRPC ≥ 2 bone metastases No known visceral metastases Post-docetaxel or unfit for docetaxel Radium-223 (50 kBq/kg) + Best standard of care Total ALP: < 220 U/L vs ≥ 220 U/L Bisphosphonate use: Yes vs No Prior docetaxel: Yes vs No Placebo (saline) + Best standard of care N = 922 Planned follow-up is 3 years Clinicaltrials.gov identifier: NCT
47
ALSYMPCA Overall Survival
100 HR 0.695; 95% CI, P = 90 80 70 60 Radium-223, n = 541 Median OS: 14.0 months % 50 40 30 Placebo, n = 268 Median OS: 11.2 months 20 10 Month 3 6 9 12 15 18 21 24 27 Radium- 223 541 450 330 213 120 72 30 Placebo 268 218 147 89 49 28 7 NEJM 2014
48
ALSYMPCA Secondary Endpoints: ALP and PSA
Hazard ratio 95% CI P-value Time to Total ALP progression (0.121 – 0.221) < Time to PSA progression (0.546 – 0.826) Radium-223 n (%) Placebo n (%) P-value Total ALP response (30% reduction) 165 (43) 4 (3) < 0.001 Total ALP normalisation* 83 (33) 1 (1) EORTC 2011 *In patients who had elevated total ALP at baseline.
49
ALSYMPCA Adverse Events
All Grades Grades 3 or 4 Radium-223 (n = 509) n (%) Placebo (n = 253) Haematologic Anaemia 136 (27) 69 (27) 54 (11) 29 (12) Neutropenia 20 (4) 2 (1) 9 (2) Thrombocytopenia 42 (8) 14 (6) 22 (4) 4 (2) Non-Haematologic Bone pain 217 (43) 147 (58) 89 (18) 59 (23) Diarrhoea 112 (22) 34 (13) 6 (1) 3 (1) Nausea 174 (34) 80 (32) 8 (2) Vomiting 88 (17) 32 (13) 10 (2) 6 (2) Constipation 46 (18) EORTC 2011
50
METANALISI TERAPIA RADIOMETABOLICA
Drug Design Development Therapy Tunio et al 2015
51
Tunio et al 2015
52
Tunio et al 2015
53
LINEE GUIDA AIOM 2014
54
GRAZIE PER L’ATTENZIONE
Similar presentations
© 2025 SlidePlayer.com Inc.
All rights reserved.