ArticlePDF Available

Bone-targeted radium-223 in symptomatic, hormone-refractory prostate cancer: A randomised, multicentre, placebo-controlled phase II study

Authors:

Abstract and Figures

The alpha-emitter radium-223 ((223)Ra) is a bone-seeking radionuclide studied as a new treatment for patients with bone metastases from hormone-refractory prostate cancer. We aimed to study mature outcomes from a randomised, multicentre, phase II study of (223)Ra. Patients with hormone-refractory prostate cancer and bone pain needing external-beam radiotherapy were assigned to four intravenous injections of (223)Ra (50 kBq/kg, 33 patients) or placebo (31 patients), given every 4 weeks. Primary endpoints were change in bone-alkaline phosphatase (ALP) concentration and time to skeletal-related events (SREs). Secondary endpoints included toxic effects, time to prostate-specific-antigen (PSA) progression, and overall survival. All tests were done at a 5% significance level, based on intention to treat. Median relative change in bone-ALP during treatment was -65.6% (95% CI -69.5 to -57.7) and 9.3% (3.8-60.9) in the (223)Ra group and placebo groups, respectively (p<0.0001, Wilcoxon ranked-sums test). Hazard ratio for time to first SRE, adjusted for baseline covariates, was 1.75 (0.96-3.19, p=0.065, Cox regression). Haematological toxic effects did not differ significantly between two groups. No patient discontinued (223)Ra because of treatment toxicity. Median time to PSA progression was 26 weeks (16-39) versus 8 weeks (4-12; p=0.048) for (223)Ra versus placebo, respectively. Median overall survival was 65.3 weeks (48.7-infinity) for (223)Ra and 46.4 weeks (32.1-77.4) for placebo (p=0.066, log rank). The hazard ratio for overall survival, adjusted for baseline covariates was 2.12 (1.13-3.98, p=0.020, Cox regression). (223)Ra was well tolerated with minimum myelotoxicity, and had a significant effect on bone-ALP concentrations. Larger clinical trials are warranted to study (223)Ra on the prevention of SREs and on overall survival in patients with hormone-refractory prostate cancer. Bone-targeting properties of (223)Ra could also potentially be used for treating skeletal metastasis from other primary cancers.
Content may be subject to copyright.
http://oncology.thelancet.com Vol 8 July 2007
587
Articles
Bone-targeted radium-223 in symptomatic, hormone-
refractory prostate cancer: a randomised, multicentre,
placebo-controlled phase II study
Sten Nilsson, Lars Franzén, Christopher Parker, Christopher Tyrrell, René Blom, Jan Tennvall, Bo Lennernäs, Ulf Petersson, Dag C Johannessen,
Michael Sokal, Katharine Pigott, Jeff rey Yachnin, Michael Garkavij, Peter Strang, Johan Harmenberg, Bjørg Bolstad, Øyvind S Bruland
Summary
Background The alpha-emitter radium-223 (223Ra) is a bone-seeking radionuclide studied as a new treatment for
patients with bone metastases from hormone-refractory prostate cancer. We aimed to study mature outcomes from a
randomised, multicentre, phase II study of 223Ra.
Methods Patients with hormone-refractory prostate cancer and bone pain needing external-beam radiotherapy were
assigned to four intravenous injections of 223Ra (50 kBq/kg, 33 patients) or placebo (31 patients), given every 4 weeks.
Primary endpoints were change in bone-alkaline phosphatase (ALP) concentration and time to skeletal-related events
(SREs). Secondary endpoints included toxic eff ects, time to prostate-specifi c-antigen (PSA) progression, and overall
survival. All tests were done at a 5% signifi cance level, based on intention to treat.
Findings Median relative change in bone-ALP during treatment was –65⋅6% (95% CI –69⋅5 to –57⋅7) and 9⋅3%
(3⋅8–60⋅9) in the 223Ra group and placebo groups, respectively (p<0⋅0001, Wilcoxon ranked-sums test). Hazard ratio
for time to fi rst SRE, adjusted for baseline covariates, was 1⋅75 (0⋅96–3⋅19, p=0⋅065, Cox regression). Haematological
toxic eff ects did not diff er signifi cantly between two groups. No patient discontinued 223Ra because of treatment
toxicity. Median time to PSA progression was 26 weeks (16–39) versus 8 weeks (4–12; p=0⋅048) for 223Ra versus placebo,
respectively. Median overall survival was 65⋅3 weeks (48⋅7–∞) for 223Ra and 46⋅4 weeks (32⋅1–77⋅4) for placebo
(p=0⋅066, log rank). The hazard ratio for overall survival, adjusted for baseline covariates was 2⋅12 (1⋅13–3⋅98,
p=0⋅020, Cox regression).
Interpretation 223Ra was well tolerated with minimum myelotoxicity, and had a signifi cant eff ect on bone-ALP
concentrations. Larger clinical trials are warranted to study 223Ra on the prevention of SREs and on overall survival in
patients with hormone-refractory prostate cancer. Bone-targeting properties of 223Ra could also potentially be used for
treating skeletal metastasis from other primary cancers.
Introduction
Hormone-refractory prostate cancer has a propensity to
involve the bone marrow at an early stage. Subsequently,
this involvement leads to the development of
symptomatic skeletal metastases with pain, spinal-cord
compression, pathological fracture, and pancytopenia.
Bone-targeted treatments such as bisphosphonates (eg,
zoledronate) and bone-seeking radioisotopes (eg, 89Sr
and ethylenediaminetetramethylene phosphonate
(EDTMP)-153Sm) are commonly used to delay skeletal
disease progression and relieve pain. Zoledronate
reduces the risk of skeletal-related events, but does not
extend survival.1 The beta-emitting radioisotopes 89Sr and
EDTMP-153Sm have been shown to improve pain control
in men with symptomatic, hormone-refractory prostate
cancer, with myelo suppression as their dose-limiting
toxicity.2,3
Beta-emitting radioisotopes produce relatively low-
energy radiation with a track length in tissues of up to
several mm. By contrast, alpha-emitters produce high,
linear energy transfer (LET) radiation with a range of
less than 100 µm. Compared with 89Sr and EDTMP-153Sm,
a bone-seeking alpha-emitter might therefore have an
increased anti-tumour eff ect, by virtue of the densely
ionising abilities of high-LET radiation, but with relative
sparing of the bone marrow because of its short-track
length.
Radium-223 (223Ra) is a bone-seeking alpha-emitter
with a half-life of 11⋅4 days that has been studied
extensively in preclinical animal models. In mice, the
biodistribution of 223Ra has been shown to correspond
with that of 89Sr, targeting of the bony skeleton with
retention of its daughter isotopes in the bone matrix.4
Modelling of the dose deposition in relation to tumour
deposits in the bone marrow suggests a substantial
reduction in dose to the healthy bone marrow with 223Ra
compared with 89Sr.4 In a rat model of breast cancer, 223Ra
showed pronounced anti-tumour eff ects in the absence
of bone marrow toxic eff ects.5 After 67 days’ follow-up
period, two of fi ve animals treated with at least 100 kBq/kg
223Ra survived, whereas none did in the control group. In
this model, treatment with bisphosphonates gave no
survival benefi t.
A phase I trial of one intravenous injection of 223Ra was
done recently in 25 patients with metastatic bone disease
(15 with hormone-refractory prostate cancer, ten with
Lancet Oncol 2007; 8: 587–94
Published Online
June 3, 2007
DOI:10.1016/S1470-
2045(07)70147-X
See Refl ection and Reaction
page 564
Karolinska Hospital,
Stockholm, Sweden
(Prof S Nilsson MD,
J Yachnin MD); Karolinska
Institute, Stockholm, Sweden
(Prof S Nilsson, Prof P Strang MD,
J Harmenberg MD);
Länssjukhuset Sundsvall-
Härnösand, Sundsvall, Sweden
(L Franzén MD); Institute of
Cancer Research and Royal
Marsden Hospital, Sutton, UK
(C Parker FRCR); Plymouth
General Hospital, Plymouth,
UK (C Tyrrell FRCR); University
Hospital Linköping, Linköping,
Sweden (R Blom MD);
University Hospital Lund,
Lund, Sweden
(Prof J Tennvall MD,
M Garkavij MD); Sahlgrenska
University Hospital,
Gothenburg, Sweden
(B Lennernäs MD);
Centrallasarettet i Västerås,
Västerås, Sweden
(U Petersson MD); Haukeland
University Hospital, Bergen,
Norway (D C Johannessen MD);
Nottingham City Hospital,
Nottingham, UK
(M Sokal FRCR); Royal Free
Hospital, London, UK
(K Pigott FRCR); Algeta ASA,
Oslo, Norway (J Harmenberg,
B Bolstad MSc); University of
Oslo, Norwegian Radium
Hospital, Oslo, Norway
(Prof Ø S Bruland MD)
Correspondence to:
Dr Christopher Parker, Institute
of Cancer Research and Royal
Marsden Hospital, Sutton, UK
chris.parker@icr.ac.uk
Articles
588
http://oncology.thelancet.com Vol 8 July 2007
breast cancer).6 Doses of 50–250 kBq/kg were well
tolerated, with grade 3 leucopenia (Common Terminology
Criteria for Adverse Events [CTCAE], version 2.0) in three
of 25 patients and no grade 2+ thrombocytopenia. No
dose-limiting toxic eff ects were recorded. Gastrointestinal
adverse events, most commonly diarrhoea (10 of 25 patients;
highest CTCAE grade 2), were most frequent, especially in
the highest dose groups. Preferential uptake was seen in
skeletal metastases compared with healthy bone, with
excretion mainly through the intestinal tract. Preliminary
evidence of effi cacy was seen with substantial reductions
in serum alkaline phosphatase (ALP) concentrations and
improved pain control across the dose levels.6 Preliminary
data suggested that the eff ect on ALP was similar across
doses, and the lowest tested dose (50 kBq/kg) was selected
for future studies.
In a randomised, double-blind, placebo-controlled,
multicentre phase II study, we aimed to investigate the
eff ect of repeated 223Ra doses in men with symptomatic,
hormone-refractory prostate cancer. Main study objectives
were the effi cacy of 223Ra treatment with respect to the
reduction in bone-specifi c ALP (bone-ALP) concentration,
and time to occurrence of skeletal-related events (SREs).
Methods
Patients
Eligible patients had histologically or cytologically confi rmed
adenocarcinoma of the prostate; multiple bone metastases
or one painful lesion with two consecutive rising amounts
of serum prostate-specifi c antigen (PSA); Eastern
Cooperative Oncology Group performance status 0–2; life
expectancy of longer than 3 months; adequate
haematological (neutrophils ≥1⋅5×109/L; platelets ≥100×109/
L; haemoglobin >100 g/L), renal (creatinine <1⋅5×upper
limit of normal), and hepatic (normal bilirubin [within
institutional limits], aspartate aminotransferase and alanine
aminotransferase <2⋅5×upper limit of normal) function;
and bone pain needing external-beam radiotherapy. All
patients had either bilateral orchidectomy or continued
treatment on a luteinising-hormone-releasing-hormone
agonist throu gh out the study. We excluded patients if they
had another currently active malignant disease; had
received chemo therapy, immunotherapy, or external-beam
radiotherapy within the past 6 weeks; had a change in
hormonal treatment within 6 weeks before study drug use,
bisphosphonates within 3 months; or had any previous
systemic radiotherapy with strontium, samarium, or
rhenium. All patients gave written informed consent.
Procedures
Patients due to receive local-fi eld, external-beam
radiotherapy to relieve pain from bone metastases were
assigned to receive either four repeated monthly
injections of 50 kBq/kg 223Ra (Alpharadin, Algeta ASA,
Oslo, Norway) or repeated injections of saline (fi gure 1).
223Ra was supplied to the hospitals as a ready-to-use
solution for injection, with little shielding needed since
the isotope is an alpha-emitter. Randomisation was
done centrally with a random number generator and
stratifi ed according to study centre. For masked
treatment allocation, an individual from the nuclear
medicine department at every centre was responsible
for study treatment preparation and labelling.
Researchers remained masked to treatment allocation.
Treatment lasted for 12 weeks, during which four
injections were given at 4-week intervals, with the fi rst
injection given at the time of external-beam radiotherapy
and no later than 7 days afterwards. External-beam
radiotherapy was given to the most painful site, with
appropriate margins, and to an area not exceeding
400 cm2. Either one fraction of 8 Gy or a fractionated
course of 20 Gy (4 Gy×5) over 1 week, or 30 Gy (3 Gy×10)
over 2 weeks was permitted.
Panel: Defi nitions of SREs
25% increase in pain severity index compared with baseline after day 15 (during the
rst 16 weeks of the study, the pain increase had to be confi rmed with a second
assessment). Patients indicating pain severity on Brief Pain Inventory forms8
(scale 0–10), with index defi ned as mean score of four questions on pain severity
[worst, least, average, and current])
Increased analgesic consumption (analgesia classifi ed according to World Health
Organization ladder for cancer pain [L0=no analgesia, L1=non-opioids, L2=weak
opioids, L3=strong opioids]; increase defi ned as change to higher level or ≥50%
increase in strong opioids, with temporary increases during fi rst 2 weeks after fi rst
injection excluded)
Neurological symptoms secondary to skeletal manifestations of prostate cancer
New pathological bone fractures (vertebral and non-vertebral)
• Tumour-related orthopaedic surgical intervention
Subsequent external-beam radiation to relieve skeletal pain
Use of radioisotopes to relieve new skeletal-related symptoms
Use of corticosteroids for skeletal pain palliation
Use of chemotherapy, bisphosphonates, or hormones to treat progression of skeletal
disease
4 injections
q 4 weeks
W12 W16
Bone markers
PSA
N=64
Local external-beam
radiotherapy
Patients with
hormone-refractory
prostate cancer
50 kBq/kg 223Ra every 4 weeks
Saline (placebo) every 4 weeks
M6 M9 M18 M24
M12
Study
unmasked
SREs
Pain
Bone markers
PSA
Safety
Survival
Follow-up
of long-term
toxic effects
and survival
Randomise
Treatment
Figure 1: Overall summary of study treatment
W=week. M=month.
Articles
http://oncology.thelancet.com Vol 8 July 2007
589
Patients were monitored every 2 weeks until 4 weeks
after the last injection, and then at 6, 9, and 12 months.
Every visit included clinical history, concomitant
medication, physical examination, pain and analgesic
assessment, adverse event recording, and blood tests
(including bone-ALP, PSA, full blood count, and markers
of bone turnover). Patients were followed for survival and
long-term toxic eff ects at 18 and 24 months.
Bubley described PSA-confi rmed response and
progression in 1999.7 Confi rmed PSA response was a 50%
reduction from baseline and confi rmed with a second
measurement at least 4 weeks later. PSA progression was
an increase of 25% from nadir in patients without a
confi rmed PSA response and 50% in those with a con-
rmed PSA response. SREs were defi ned as one of a
specifi c set of events listed in the panel.
Patients who began non-study treatment (eg, hormonal
therapy, external-beam radiotherapy) for progressive
disease during the study were allowed to continue with
the study drug and be analysed for safety. Effi cacy analyses
were done with and without censoring at the time of
commencement of non-study treatments (corticosteroids,
33 allocated to ²²³Ra treatment
33 received at least one ²²³Ra treatment
5 did not receive all four ²²³Ra treatments
2 with progressive disease or
investigator decision
2 with cardiac disease
1 with confusion
31 allocated to placebo treatment
31 received at least one placebo treatment
10 did not receive all four placebo treatments
4 with progressive disease
4 patient preference
1 with cardiac disease
1 with confusion
64 patients with hormone-refractory
prostrate cancer randomised
29 patients in study at week 16 22 patients in study at week 16
4 patients withdrawn
before week 16
9 patients withdrawn
before week 16
19 patients in study at month 12 12 patients in study at month 12
10 patients withdrawn
between week 16 and
month 12 because of
disease progression or
death
10 patients withdrawn
between week 16 and
month 12 because of
disease progression or
death
14 patients in study at month 18 5 patients in study at month 18
5 patients withdrawn
between month 12 and
month 18 because of
disease progression or
death
7 patients withdrawn
between month 12 and
month 18 because of
disease progression or
death
Figure 2: Study profi le
All patients followed for survival irrespective of study status (ie, in study or withdrawn). Information on the number screened was not obtained.
²²³Ra (n=33) Placebo (n=31)
Age, years 73, 73 (57–88) 72, 72 (60–84)
Haemoglobin, g/L 126, 125 (100–153) 129, 126 (99–149)
PSA, ng/mL 167, 511 (10–6000) 233, 480 (1–4002)
Bone-ALP, ng/mL 57, 121 (13–1145) 68, 132 (11–706)
Total ALP, U/L 228, 437 (80–3047) 279, 501 (51–2280)
Albumin, g/L 40, 39 (28–46) 38, 39 (30–47)
Lactate dehydrogenase (U/L) 348, 351 (154–750) 345, 426 (144–1284)
ECOG performance status
09 6
118 20
26 5
Extent of disease
<6 metastases 12 7
6–20 metastases 10 13
>20 metastases 11 11
Pain severity index 3·50, 3·88 (1·00–7·75) 4·00, 3·78 (0·75–7·75)
Data are number of patients (median, mean, range) in intention-to-treat population
Table 1: Baseline patient characteristics
Articles
590
http://oncology.thelancet.com Vol 8 July 2007
external-beam radiotherapy, cytotoxic chemotherapy, add-
itional radionuclides, hormones, and bisphosphonates).
The study design was approved by all the research ethics
committees from the study jurisdictions and centres.
In addition to time to SREs, bone-ALP was chosen as a
primary endpoint because it is used in clinical practice
as a marker of disease extent and prognosis, and because
a bone-targeted therapy would probably not be eff ective
in treating hormone-refractory prostate cancer without
greatly aff ecting bone-ALP. Secondary endpoints
included safety, serum markers of bone turnover (total
ALP, procollagen I N propeptide [PINP], C-terminal
crosslinking telopeptide of type I collagen [S-CTX-I], and
type I collagen crosslinked C-telopeptide [ICTP]), serum
PSA, and overall survival.
Statistical methods
The study was designed to have 80% power to detect an
absolute diff erence at 5% signifi cance level of at least
15% between treatment and control groups with respect
to the mean change in bone ALP from baseline to 4 weeks
after the last injection. Although medians are reported in
the results and are more useful for clinicians, no standard
techniques exist for calculating sample size from
medians, therefore, means were used for this purpose.
Sample size calculations were based on the normal
distribution with the following formula and rounded up:
n=(2*SD2*(Zα/2+Zβ)2/meandiff 2 +Zα/2/4) (Zα/2=value of
normal distribution [1⋅96] for α/2 [α=0⋅05], and Zβ=value
of normal distribution [0⋅84] for 1-power [power=0⋅8]).
We planned a sample size of 60 patients (30 in each
group). Patients who received at least one dose of study
drug were included in the analyses, and all results were
analysed by intention to treat. All tests of signifi cance
were done at the 5% signifi cance level and no adjustment
for multiplicity was done for secondary variables, since
the present study was explorative in nature. We expected
that the relative change of bone ALP and other variables
would not be normally distributed, and would contain
non-positive data, so standard transformation methods
would not be possible. Therefore, we analysed the
diff erence between groups of relative change of bone
ALP and other variables by use of the Wilcoxon ranked-
sums test. For the analysis of time to event, we used a
log-rank test (unadjusted) and a Cox proportional
hazards model (adjusted for covariates). Covariates
were baseline values of albumin, ECOG performance
status, haemoglobin, lactate dehydrogenase, PSA, total
ALP, and age. Proportions were analysed with the
Fisher’s exact test. We used screening values for missing
data at baseline, and a last-value-carried-forward
approach for missing laboratory values. For relative
change and time-to-PSA-progression, we excluded two
patients in the placebo group without any post-baseline
values.
Role of the funding source
This study was sponsored by Algeta ASA (Oslo, Norway)
in collaboration with TFS Trial Form Support (Lund,
Sweden; monitoring and database); Harrison Clinical
Research (Ely, UK; monitoring); and Statisticon AB
(Uppsala, Sweden; statistical advice and analysis). BB
and JH are employees of Algeta. BB was involved in the
study design, and BB and JH contributed to the analysis
²²³Ra (n=33) Placebo (n=30*)
1234 1234
Platelets 6 0 0 0 4 0 1 0
Neutrophils 5 2 1 0 0 0 0 0
White blood cells 9 1 1 0 3 0 0 0
Haemoglobin 26 4 1 0 19 6 0 1
*One patient was not evaluable for toxic eff ects. Numbers in second row are toxicity graded with CTCAE version 3.0.
Data are number of patients.
Table 2: Worst grade for haematological toxic eff ects during treatment
223Ra Placebo
Mild Moderate Severe Mild Moderate Severe
Confusional State 1 0 0 0 0 0
Vomiting 0 1 0 0 0 0
Pseudomonas infection 0 0 1* 0 0 0
Exacerbated chronic
obstructive airways disease
001*000
Pneumonia 0 1 1† 0 0 0
Sepsis 0 0 1† 0 0 1‡
Dehydration 0 0 1† 0 0 0
Spinal cord compression 0 1 0 0 0
Chest pain 0 1 0 0 0 0
Myocardial infarction 0 0 0 0 0
Tumour fl are 001§001
Ataxia 0 0 0 0 0 1
Subdural haematoma 0 0 0 0 0 1
Deep-vein thrombosis 0 0 0 0 1 0
Perineal pain 0 0 0 0 0 1
Bone pain 0 0 0 0 1‡
Atrial fi brillation 0 0 0 0 0 1‡
Adrenal disorder 0 0 0 0 0 1
Fall 0 0 0 0 0 1**
Cachexia 0 0 0 0 1** 0
Haemoglobin decreased 0 0 0 1 0 0
Pyrexia 0 0 0 0 1†† 0
Malignant neoplasm 0 0 0 0 0 1††
Hypocalcaemia 0 0 0 0 1 0
Intestinal obstruction 0 0 0 0 0 1
Mild=transient and easily tolerated. Moderate=causes discomfort and interrupts usual activities. Severe=considerable
interference with usual activities, and might be incapacitating or life threatening. *One patient had pseudomonas
infection and exacerbated chronic obstructive airways disease. †One patient had pneumonia, sepsis, and dehydration.
‡One patient had atrial fi brillation, sepsis, and bone pain. ¶One patient had bone pain and spinal cord compression
§One patient had myocardial infarction and tumour fl are.**One patient had a fall and cachexia. ††One patient had
pyrexia and malignant neoplasm. When a serious adverse event was reported more then once by the same patient, the
highest intensity is listed.
Table 3: Serious adverse events
Articles
http://oncology.thelancet.com Vol 8 July 2007
591
and interpretation of the data. Algeta had no other role in
the collection, analysis, or interpretation of the data. The
corresponding author had full access to all the raw data
and the fi nal responsibility to submit for publication. JH,
PS, and SN also had access to the raw data.
Results
64 patients were recruited in 11 centres in Sweden,
Norway, and the UK between Feb 11, 2004, and May 3,
2005 (fi gure 2). 33 patients were assigned external-
beam radiotherapy and 223Ra, and 31 to external-beam
radiotherapy and placebo. Table 1 shows the baseline
values of bone-ALP, haemoglobin, albumin, PSA, age,
extent of disease on bone scan,9 and pain score. We
recorded no signifi cant diff erence in external-beam
radiotherapy dose fractionation at baseline between the
study groups (223Ra, median 8 Gy [range 6–30]; placebo,
8 Gy [8–30]). All patients have been followed for at least
18 months (range 18–24).
All 64 patients received external-beam radiotherapy and
at least one injection of study drug. 28 patients in the
223Ra group and 21 in the placebo group completed all
four injections of study drug. Figure 2 shows the reasons
for not completing all four treatments. During treatment
(from week –1 to week 16), corticosteroids were begun in
24 patients (12 223Ra, 12 placebo), anti-androgens in two
223Ra patients, oestrogen in one placebo patient, and
bisphosphonate in two placebo patients.
The extent of haematological toxic eff ects was at a
minimum (table 2). Thrombocytopenia (CTCAE grade
2+) was not seen in the 223Ra group but recorded in one
placebo patient. We recorded grade 2+ neutropenia in
three patients given 223Ra and none given placebo.
Neutropenia was reversible and most commonly seen
during the fi rst 4 weeks of treatment, with no evidence of
cumulative myelotoxic eff ects recorded. We saw no
substantial diff erences in haematological toxic eff ects
between the two groups, and no patient discontinued
223Ra because of treatment-related toxic eff ects.
12 serious adverse events were reported in eight
patients receiving 223Ra, compared with 19 serious adverse
events in 14 patients receiving placebo (table 3). One
patient’s vomiting needed hospital care 6 h after the fi rst
223Ra injection, which was deemed related to the study
drug. This patient received subsequent injections with
223Ra without further vomiting. The investigators deemed
that vomiting was treatment-related; but they were
uncertain whether sepsis (reported in one patient in each
study group) and tumour fl are (one patient in the 223Ra
group) were treatment-related (one patient in the placebo
group had tumour fl are, but this was deemed not
treatment-related). All other serious adverse events were
deemed unrelated to treatment.
Table 4 lists adverse events that occurred in more than
15% of patients. Apart from constipation, no statistically
signifi cant diff erence existed between treatment groups.
Constipation was mild or moderate in all but one patient.
Median time to onset of constipation was 4 weeks (range
1–12) in the 223Ra group, and actual time of onset from
baseline was 7 and 10 weeks in the two patients in the
placebo group. Unfortunately, no data for duration of
constipation were available.
Median relative change in bone-ALP from baseline to
4 weeks after last study injection was –65⋅6% (95% CI
−200
−100
0
100
200
300
400
²²³Ra
Placebo
Relative change in bone−ALP (%)
Figure 3: Distribution of % change in serum bone-ALP from baseline to 4 weeks after last study drug
Individual results sorted by eff ect size.
223Ra (n=33) Placebo (n=31)
Mild Moderate Severe Total Mild Moderate Severe Total
Diarrhoea 6 3 0 9 5 5 0 10
Constipation 6 5 1 12 0 2 0 2
Vomiting 4 4 0 8 3 3 0 6
Nausea 6 3 0 9 6 3 1 10
Fatigue 3 5 0 8 7 0 0 7
Bone pain 2 7 1 10 7 7 2 16
Myalgia 3 2 0 5 3 1 0 4
Tumour fl are 2 3 1 6 1 5 1 7
Anaemia 0 5 0 5 2 3 2 7
Data are number of patients. Mild=transient and easily tolerated. Moderate=causes discomfort and interrupts usual
activities. Severe=considerable interference with usual activities, and might be incapacitating or life threatening.
Table 4: Adverse events in more than 15% of study population during treatment (reported by at least
nine patients)
223Ra Placebo p*
Bone-ALP –65·6% (–92·2 to 124·9) 9·3% (–77·4 to 384·1) <0·0001
Total ALP –46·2% (–89·3 to 102·5) 30·7% (–75·4 to 212·9) <0·0001
PINP –63·2% (–93·7 to 151·0) 38·3% (–72·5 to 602·8) <0·0001
CTX-I –31·4% (–74·3 to 143·3) 31·7% (–57·5 to 395·8) 0·002
ICTP 14·6% (–54·6 to 158·9) 43·2% (–56·3 to 242·1) 0·011
Data are median (range). *Wilcoxon ranked-sums test.
Table 5: Relative change in serum markers of bone metabolism from baseline to 4 weeks after last study
injection
Articles
592
http://oncology.thelancet.com Vol 8 July 2007
–69⋅5 to –57⋅7) in the 223Ra group and 9⋅3% (3⋅8–60⋅9) in
the placebo group (p<0⋅0001, Wilcoxon ranked-sums test).
Figure 3 shows distribution of the change in bone-ALP.
Table 5 shows median relative change in bone-ALP and the
other serum markers of bone metabolism. Compared with
the placebo group, the 223Ra group had a signifi cant
reduction in all fi ve markers (bone-ALP, total-ALP, PINP,
CTX-I, and ICTP).
Median time to fi rst SRE was 14 weeks (95% CI 9–30) in
the 223Ra group and 11 weeks (5–25) in the placebo group
(p=0⋅257, log rank; fi gure 4). Hazard ratio for time to fi rst
SRE adjusted for baseline covariates was 1⋅75 (95% CI
0⋅96–3⋅19, p=0⋅065, Cox regression) where the placebo
group is the reference. By week 16, 17 patients in the 223Ra
group had 34 SREs in total, compared with 18 patients
who had 44 SREs in the placebo group (p=0⋅625, Fisher’s
exact test). If we excluded pain and analgesic consumption
from the SRE defi nitions, six patients in the 223Ra group
had nine SREs in total, compared with 11 patients in the
placebo group who had 21 SREs (p=0⋅159). By week 52,
26 patients in each group had had at least one SRE.
Median relative change in PSA from baseline to 4 weeks
after last study injection was –23⋅8% (range –98⋅6 to
545⋅6) in the 223Ra group and 44⋅9% (–91⋅3 to 563⋅5) in
the placebo group (p=0⋅003, Wilcoxon ranked-sums test;
gure 5). A confi rmed PSA response of more than 50%
was seen in 11 of 31 patients assigned 223Ra and fi ve of
28 assigned placebo (p=0⋅153, Fisher’s exact test). Median
time to PSA progression was 26 weeks (95% CI 16–39)
for 223Ra compared with 8 weeks (4–12) for placebo
(p=0⋅048, log rank).
Censoring for concomitant treatment that might aff ect
PSA did not change the overall results, since most
treatments were started after the PSA endpoints were
reached (PSA response or progression). After censoring
of concomitant treatments, nine patients who were
assigned 223Ra showed a confi rmed PSA response of at
least 50% compared with two assigned placebo (p=0⋅045,
Fisher’s exact test). The three censored patients in the
placebo group and two in the experimental group with
confi rmed 50% PSA response had all started cortico-
steroids. Median time to PSA progression remained at
26 weeks for 223Ra-treated patients compared with 8 weeks
for placebo (p=0⋅040, log rank).
Median overall survival was 65⋅3 weeks (95%
CI 48⋅7–∞) for 223Ra and 46⋅4 weeks (32⋅1–77⋅4) weeks
for placebo (p=0⋅066, log rank; fi gure 6). Hazard ratio for
survival adjusted for baseline covariates was 2⋅12 (95%
CI 1⋅13–3⋅98, p=0⋅020, Cox regression), indicating an
increased risk of death in the placebo group. At
18 months’ follow-up, 15 patients assigned 223Ra survived
compared with eight assigned placebo.
Discussion
In our randomised study of patients with symptomatic,
hormone-refractory prostate cancer, 223Ra was well
tolerated with little or no myelotoxic eff ect, and showed
promising evidence of effi cacy. One primary endpoint
was met, with a signifi cant eff ect on bone-ALP 4 weeks
after last treatment. Effi cacy data in this small study
suggested a potential benefi cial eff ect of 223Ra on SRE
risk, time to PSA progression, and overall survival. The
good toxicity profi le seen for 223Ra will allow future
studies to use increased doses and extended treatment
periods.
This study shows promising evidence for the safety
and tolerability of 223Ra 50 kBq/kg at 4 weekly intervals.
0·0
0·2
0·4
0·6
0·8
1·0
Proportion without SRE
Numbers at risk
²²³Ra
Placebo
05 504540353025201510
Time (weeks)
33 23 4567811131519
31 23 11245781315
²²³Ra
Placebo
Figure 4: Survival function of time to fi rst SRE
Special instructions (PLEASE MARK WITH RED SPOT IF URGENT)
$£¥µπΩαβχδεγηκλμτ§+±×÷<>
Ref number
Editor
Author
Created by Helen
²²³Ra
Placebo
−200
0
200
400
600
Relative change in PSA (%)
Figure 5: Distribution of % change in serum PSA from baseline to 4 weeks after last study drug
Individual results sorted by eff ect size.
Articles
http://oncology.thelancet.com Vol 8 July 2007
593
The lack of haematological toxic eff ects contrasts with
those in previous studies of beta-emitting radioisotopes
in the treatment of hormone-refractory prostate cancer.
For example, 89Sr was studied in a trial of similar design
(n=126),10 in which grade 2+ thrombocytopenia was
seen in 41 (61%) of 67 patients assigned 89Sr versus six
(10%) of 58 assigned placebo. Notably, grade 4
thrombocytopenia was seen in seven (10%) patients in
the 89Sr group versus one (2%) in the placebo group.
89Sr was shown to signifi cantly reduce alkaline
phosphatase and PSA concentrations, but no eff ect was
seen on overall survival. On the basis of the present
study, 223Ra at 50 kBq/kg given every 4 weeks seems to
be at least as eff ective as conventional radioisotopes,
but with fewer haematological toxic eff ects. This result
supports the hypothesis that, compared with beta-
emitters such as 89Sr and HEDP-153Sm, the short-range,
high LET alpha radiation from 223Ra has an increased
anti-tumour eff ect, with relative sparing of the bone
marrow.11 Since myelosuppressive chemotherapy is
now a standard treatment for hormone-refractory
prostate cancer and pancytopenia is a feature of
progressive disease, this result represents a major
advantage for the future development of 223Ra. Docetaxel
is the standard fi rst-line chemotherapy for hormone-
refractory prostate cancer.12 In view of the favourable
toxicity profi le of 223Ra in the present trial, combination
therapy with docetaxel could be considered, either
sequentially or concurrently. Furthermore, whereas
89Sr and HEDP-153Sm are used for pain palliation, and
repeat treatment is not possible until bone-marrow
recovery, the toxicity profi le of 223Ra will allow repeat
treatment of asymptomatic patients and could have a
disease-modifying eff ect.
For non-haematological toxic eff ects, constipation was
the only adverse event seen substantially more often in
the 223Ra group than in the placebo group, for which the
explanation is unclear. Constipation was mild or
moderate in all but one patient, and no clear temporal
relation was recorded with study drug treatment.
Notably, the phase I study6 showed that 223Ra was
associated with diarrhoea and not constipation.
A signifi cant benefi t for 223Ra was recorded with
respect to all serum bone markers. These endpoints are
not in themselves clinically meaningful, but suggest
that 223Ra has a real biological eff ect on bone metabolism.
Favourable trends were seen in all PSA endpoints,
including a signifi cant eff ect on time to PSA progression,
which is consistent with the hypothesis that eff ective
treatment of bone metastases can substantially delay
overall disease progression in hormone-refractory
prostate cancer.
Interpretation of the clinically meaningful endpoints
is restricted by the sample size, and by the fact that all
patients received external-beam radiotherapy at
baseline. Too few SREs were recorded for fi rm
conclusions to be drawn regarding the clinical
eff ectiveness of 223Ra. However, in the pivotal trial of
zoledronate,13 the proportion of patients having an SRE
fell from 44% to 33%, a relative reduction of 25%.
The overall survival data in this study of only
64 patients should be interpreted with caution. The
eff ects of known baseline prognostic factors on time to
SRE, time to PSA progression, and overall survival time
have been tested using a Cox proportional hazards
model, which, if anything, increased rather than
decreased the treatment eff ect of 223Ra. Furthermore,
our small randomised trial is the third to show a
signifi cant advantage in overall survival for radioisotope
treatment in hormone-refractory prostate cancer.14,15
Thus, the survival benefi t of 223Ra in this trial could be a
genuine treatment eff ect. However, the benefi ts of 223Ra
could be due to an imbalance in important but unknown
baseline characteristics (eg, doubling time of
pretreatment PSA). Effi cacy data might also be
confounded by concomitant use of other anticancer
treatments. However, the main results remain unaltered
or strengthened after patients were censored at the time
of receiving concomitant treatments. Therefore the
benefi ts of 223Ra seen in this study are unlikely to be
explained by concomitant treatments. Further studies
of 223Ra should explore the potential for escalation of
dose and for increased duration of treatment by more
than four injections. The bone-targeting properties of
223Ra could also be applicable to the treatment of skeletal
metastasis from other primary cancers.
0·0
0·2
0·4
0·6
0·8
1·0
Proportion alive
Numbers at risk
0 10 1009080706050403020
Time (weeks)
33 32 779161820252931
31 30 335111314192227
²²³Ra
Placebo
²²³Ra
Placebo
Figure 6: Overall survival
Articles
594
http://oncology.thelancet.com Vol 8 July 2007
Contributors
Ø
B, PS, BB, JY, and SN contributed to study design. SN, LF, CP, CT, RB,
JT, BL, UP, DJ, MS, KP, JY, and MG contributed to data collection.
Ø
B,
CP, BB, JH, PS, and SN had access to the raw data, and contributed to
analysis and interpretation of the data. CP wrote the fi rst draft of the
manuscript and had fi nal editorial control. All authors contributed to
revision of the manuscript.
Confl icts of interest
CP,
Ø
B, and PS are external consultants of Algeta ASA, Norway
(sponsors of the study and manufacturers of 223Ra). BB and JH are
employees of Algeta ASA.
Ø
B, LF, BB, and JH own stock or options in
Algeta ASA. CP received payment from Algeta ASA for writing the
manuscript. No other confl ict of interest has been declared.
Acknowledgments
We thank our Nuclear Medicine colleagues and the Central Laboratory in
Synarc (Lyon, France) for analysing serum markers of bone metabolism
and PSA, and Marcus Thuresson for statistical support.
References
1 Saad F, Gleason DM, Murray R, et al. Long-term effi cacy of
zoledronic acid for the prevention of skeletal complications in
patients with metastatic hormone-refractory prostate cancer.
J Natl Cancer Inst 2004; 96: 879–82.
2 Finlay IG, Mason MD, Shelley M. Radioisotopes for the palliation of
metastatic bone cancer: a systematic review. Lancet Oncol 2005;
6: 392–400.
3 Lewington V. Bone-seeking radionuclides for therapy. J Nucl Med
2005; 46 (suppl 1): 38–47S.
4 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: 252–59.
5 Henriksen G, Breistol K, Bruland OS, et al. Signifi cant antitumor
eff ect from bone-seeking, alpha-particle-emitting (223)Ra
demonstrated in an experimental skeletal metastases model.
Cancer Res 2002; 62: 3120–25.
6 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: 4451–59.
7 Bubley GJ, Carducci M, Dahut W, et al. Eligibility and response
guidelines for phase II clinical trials in androgen-independent
prostate cancer: recommendations from the Prostate-Specifi c
Antigen Working Group. J Clin Oncol 1999; 17: 3461–67.
8 Klepstad P, Loge JH, Borchgrevink PC, et al. The Norwegian brief
pain inventory questionnaire: translation and validation in cancer
pain patients. J Pain Symptom Manage 2002; 24: 517–25.
9 Soloway MS, Hardeman SW, Hickey D, et al. Stratifi cation of
patients with metastatic prostate cancer based on extent of disease
on initial bone scan. Cancer 1988; 61: 195–202.
10 Porter AT, McEwan AJB, Powe JE, et al. Results of a randomized
phase-III trial to evaluate the effi cacy of strontium-89 adjuvant to
local fi eld external beam irradiation in the management of
endocrine resistant metastatic prostate cancer. Int J Radiat Oncol
Biol Phys 1993; 25: 805–13.
11 Bruland OS, Nilsson S, Fisher DR, Larsen RH. High-linear energy
transfer irradiation targeted to skeletal metastases by the
alpha-emitter 223Ra: adjuvant or alternative to conventional
modalities? Clin Cancer Res 2006; 12: 6250–57s.
12 Tannock IF, de Wit R, Berry WR, et al. Docetaxel plus prednisone or
mitoxantrone plus prednisone for advanced prostate cancer.
N Engl J Med 2004; 351: 1502–12.
13 Saad F, Gleason DM, Murray R, et al. A randomized,
placebo-controlled trial of zoledronic acid in patients with hormone-
refractory metastatic prostate carcinoma. J Natl Cancer Inst 2002;
94: 1458–68.
14 Tu SM, Millikan RE, Mengistu B, et al. Bone-targeted therapy for
advanced androgen-independent carcinoma of the prostate: a
randomised phase II trial. Lancet 2001; 357: 336–41.
15 Palmedo H, Manka-Waluch A, Albers P, et al. Repeated
bone-targeted therapy for hormone-refractory prostate carcinoma:
randomized phase II trial with the new, high-energy radio-
pharmaceutical rhenium-188 hydroxyethylidene diphos phonate.
J Clin Oncol 2003; 21: 2869–75.
... A maximum of ten cycles was deemed sufficient to assess efficacy and to monitor safety. Assessment of bone markers is in line with the endpoints used in previous trials on Radium-223-dichloride [15,16]. The phase I trial implemented 3 mg/kg. ...
... Bone metastases are the main cause of death [4]. A bone-focused radionuclide treatment can curb disease progression and prolong OS [11,15]. The rationale behind the development of ODX is to effectively treat bone metastases with a novel mode of action. ...
... The reduction in B-ALP, S-P1NP and S-Osteocalcin is in line with a secondary effect to the reduced osteoclast and tumour cell activities. The reduction in bone marker levels is in the same range as those seen after Radium-223-dichloride in symptomatic mCRPC [15] and asymptomatic mCRPC patients [22]. The ODX study was conducted during a period before BPAs were commonly recommended as part of the standard treatment of mCRPC [6e10]. ...
Article
Full-text available
Abstract Aims: This study aimed to assess the efficacy and safety of ODX, a novel, cyto- toxic, bone-targeting drug candidate, in castration-resistant prostate cancer bone metastatic disease. Methods: Patients with progressive disease were randomised to ten cycles of ODX, intrave- nous infusion Q2W (3, 6, and 9 mg/kg, respectively). The primary objective was to asses the relative change from baseline in bone alkaline phosphatase (B-ALP) and serum-amino- terminal-propeptide of Type I procollagen (S-P1NP) at 12 weeks. The inclusion criteria selected were broad, and a double-blind design was used to ensure objective recruitment of patients for the assessment of efficacy. None of the patients received bone-protecting agents during the ODX treatment period. Results: Fifty-five 21,20 and 14) patients were randomised to ODX (3, 6 and 9 mg/kg), respec- tively. The lower number of patients in arm 3 was due to too low a recruitment rate towards the end of the study. The median treatment time were 14, 13 and 14 weeks, respectively. The decrease in B-ALP at 12 weeks in study arms 3, 6 and 9 mg/kg was seen in 6/15 (40%), 8/12 (67%) and 5/12 (42%) patients, respectively, whereas the corresponding numbers for P1NP were 8/15 (53%), 8/12 (67%), and 4/12 (33%), respectively. The median decrease in B-ALP and P1NP at 12 weeks for study arms 3, 6 and 9 mg/kg were 37%, 14% and 43%, respectively, and 51%, 40% and 64%, respectively. The decrease in serum C-terminal telopeptide at 12 weeks was seen in the vast majority of patients and in about one-third of patients in bone scan index. ODX was well tolerated, and no drug-related serious adverse events occurred. There were no significant differences between study arms regarding efficacy and safety. Conclusions: ODX was well tolerated and demonstrated inhibitory effects on markers related to the vicious cycle in bone at all three doses. The reduction in metastatic burden, assessed with bone scan index, supports this finding. Studies with continued ODX treatment until dis- ease progression are being planned (ClinicalTrials.gov Identifier: NCT02825628).
... In the ALSYMPCA trial, 47% showed an ALP response and 16% a PSA response (≥30% reduction from baseline) [2]. It has also been demonstrated that ALP, a biomarker of osteoblast activity, is superior to PSA as a biomarker of Ra-223 efficacy [10,[18][19][20]; thus, that has emerged as the leading biomarker in treated patients. An increase in PSA can be the result of the development of lymph node or visceral metastasis, which is not affected by Ra-223. ...
... Whether ALP change is an accurate biomarker for predicting survival remains controversial. One study demonstrated that an ALP response ≥30% or ≥10% reduction from the baseline value in patients receiving Ra-223 therapy can be a good predictor of OS [21], whereas three others found that ALP response after Ra-223 was not effective for OS prediction [10,12,19]. In another study, Sartor et al. [22] showed that ALP decline at 12 weeks after the first Ra-223 administration was correlated with OS, though the results did not meet the statistical requirements to indicate significance. ...
Article
Full-text available
Simple Summary This study was a retrospective investigation of a Japanese cohort of 205 metastatic castration-resistant prostate cancer (mCRPC) patients who received Ra-223 in 14 hospitals between July 2016 and August 2020 and for whom bone scintigraphy before and after the radium-223 treatment was available. Following treatment, alkaline phosphatase (ALP) decline (%ALP < 0%) was noted in 72.2% (148/205), automated bone scan index (aBSI) decline (%aBSI < 0%) in 52.7% (108/205), and PSA decline (%PSA < 0%) in 27.8% (57/205). Furthermore, a reduction in both aBSI and ALP was seen in 87 (42.4%), a reduction in only ALP was seen in 61 (29.8%), a reduction in only aBSI was seen in 21 (10.2%), and in both aBSI and ALP increasing/stable (≥0%) was seen in 36 (17.6%) patients. Multiparametric analysis showed changes in PSA (HR 4.30, 95% CI 2.32–8.77, p < 0.0001), aBSI (HR 2.22, 95%CI 1.43–3.59, p = 0.0003), and ALP (HR 2.06, 95%CI 1.35–3.14, p = 0.0008) as significant prognostic factors for OS. For mCRPC patients treated with Ra-223, aBSI change is useful as an imaging biomarker for treatment response assessment and survival prediction. Abstract To evaluate the usefulness of change in the automated bone scan index (aBSI) value derived from bone scintigraphy findings as an imaging biomarker for the assessment of treatment response and survival prediction in metastatic castration-resistant prostate cancer (mCRPC) patients treated with Ra-223. This study was a retrospective investigation of a Japanese cohort of 205 mCRPC patients who received Ra-223 in 14 hospitals between July 2016 and August 2020 and for whom bone scintigraphy before and after radium-223 treatment was available. Correlations of aBSI change, with changes in the serum markers alkaline phosphatase (ALP) and prostate-specific antigen (PSA) were evaluated. Additionally, the association of those changes with overall survival (OS) was assessed using the Cox proportional-hazards model and Kaplan–Meier curve results. Of the 205 patients enrolled, 165 (80.5%) completed six cycles of Ra-223. Following treatment, ALP decline (%ALP < 0%) was noted in 72.2% (148/205), aBSI decline (%aBSI < 0%) in 52.7% (108/205), and PSA decline (%PSA < 0%) in 27.8% (57/205). Furthermore, a reduction in both aBSI and ALP was seen in 87 (42.4%), a reduction in only ALP was seen in 61 (29.8%), a reduction in only aBSI was seen in 21 (10.2%), and in both aBSI and ALP increasing/stable (≥0%) was seen in 36 (17.6%) patients. Multiparametric analysis showed changes in PSA [hazard ratio (HR) 4.30, 95% confidence interval (CI) 2.32–8.77, p < 0.0001], aBSI (HR 2.22, 95%CI 1.43–3.59, p = 0.0003), and ALP (HR 2.06, 95%CI 1.35–3.14, p = 0.0008) as significant prognostic factors for OS. For mCRPC patients treated with Ra-223, aBSI change is useful as an imaging biomarker for treatment response assessment and survival prediction.
... Encouragingly, most patients reported pain palliation by 8 wk; on the basis of these promising results, subsequent trials proceeded. Phase II trials on patients with CRPC and bone metastases demonstrated acceptable safety and tolerability (42). Nilsson et al. (42) enrolled 64 patients to receive either 223 Ra-dichloride (50 kBq/kg every 4 wk) or placebo, with a primary endpoint of total alkaline phosphatase and time to skeleton-related events. ...
... Phase II trials on patients with CRPC and bone metastases demonstrated acceptable safety and tolerability (42). Nilsson et al. (42) enrolled 64 patients to receive either 223 Ra-dichloride (50 kBq/kg every 4 wk) or placebo, with a primary endpoint of total alkaline phosphatase and time to skeleton-related events. This trial reported a significant response to alkaline phosphatase (265.6% vs. 9.3%, P , 0.0001) and delayed skeleton-related events (hazard ratio [HR], 1.75; 95% CI, 0.96-3.19; ...
Article
Radiopharmaceutical therapy is an emerging treatment modality that has demonstrated increasing importance as a significant component in the treatment of cancer. Prostate cancer (PCa) remains one of the commonest solid-organ tumors and is associated with significant societal burdens. Despite significant disease heterogeneity, PCa remains an ideal candidate for radiopharmaceutical therapy because of the prolonged disease course, metastatic disease tropism, and sensitivity to radiation therapy. To date, advanced PCa remains one of the most successful arenas for the development and approval of radiopharmaceutical agents. In this review, we aim to summarize the complex processes required to obtain regulatory approval for a novel agent and highlight the limitations and hurdles specific to the approval of radiopharmaceutical agents. In advanced PCa, we outline the importance of a framework for trial design with respect to defining disease state and acceptable outcome measures-as recommended by the Prostate Cancer Clinical Trials Working Group (PCWG). Finally, using the principles mandated by the Food and Drug Administration approval process and the framework provided by the PCWG, we outline experience with the successful approval of the radiopharmaceutical agents 223Ra and 177Lu-PSMA-617.
... Accordingly, some experts have concluded that the excess fractures were not secondary to the combination of 223 Ra-Cl 2 and abiraterone per se, but to the concomitant steroid course required to offset abiraterone's inhibition of glucocorticoid synthesis and maintain homeostasis in the adrenocorticotropic hormonemineralocorticoid axis (56). Prednisone/prednisolone alters bone turnover and suppresses osteoblast differentiation and activity (57), and it may have an interactive effect with 223 Ra-Cl 2 , which suppresses alkaline phosphatase, a marker of osteoblast activity (58,59). Future trials may investigate the use of smaller steroid doses or alternative combinations not requiring steroids; for the time being, no combination therapy involving 223 Ra-Cl 2 is proven safe or superior to monotherapy. ...
... Unlike b-emitters, a-emitters characterized by high energy (5-9 MeV) (10) and a short pathlength (50-100 mm (10) and 100 mm in tissue (11)) have demonstrated anticancer potential by reducing tumor burden and serum prostate-specific antigen levels (12)(13)(14)(15). The prototypical a-emitting particle used for radiotherapeutics is 225 Ac, which has a half-life of 10 d (10). ...
Article
The protein expression of the prostate-specific membrane antigen correlates with unfavorable or aggressive histologic features of prostate cancer, resulting in use as a diagnostic PET imaging radiotracer and therapeutic target. Here, we discuss the methods to develop 225Ac-DOTA-J591, an α-labeled compound targeting an extracellular epitope of prostate-specific membrane antigen, which is currently being studied in early clinical trials. In addition, we review quality control, radiation safety measures, and clinical considerations before administration of this radioimmunotherapeutic agent.
... The results of the study bear correlation to those obtained in other studies like the phase II study of radium-223 chloride for palliation of painful bone metastases [23] [24] [25], which showed that reduction in platelet, white blood cell and neutrophil counts tend to occur in the first two weeks following radium treatment and later returned to baseline levels. ...
Research
Full-text available
Background: Radiation pharmacokinetics and pharmacodynamics of Radium point to bone as its site of primary uptake and action. It is therefore very important to investigate the hematological and other toxicity of the radiopharmaceutical Radium-223 dichloride in the wake of its introduction for treatment of bone metastases. Methods: Five Patients with post-chemotherapy castrate resistant prostate cancer with bone metastases received 50kBq/ kg body weight of Radium-223 Dichloride injection every 4 weeks for 5 cycles. 1. Screening 2. Treatment (including an End of Treatment Visit) 3. Follow-up Results: The ranges of measured parameters for 4 patients at the end of 5th cycle (20th week) were within normal limits: Hb : Wk 20 = 110-126g/l, pre-treatment = 110-136g/l (normal range 120-170) Wbc: Wk 20 = 3.3-6.2x109/l, Pre-treatment=3.5-8.9x109/l (normal range 4.5-10) Platelets: Wk 20 = 156-241x109/l, pre-treatment = 206-302x109/l (normal range 150-400). Alkaline Phosphatase: Wk 20 = 43-128IU/l, Pre-treatment = 57-253IU/l (normal Range 40-120). The 5th Patient developed bone marrow failure in the 12th week, with concurrent flaring of his alkaline phosphatase value. Conclusion: The results of this study suggest that Radium-223 dichloride at dose of 50kbq/kg is safe for palliative treatment of bone pains in metastatic bone diseases. The 5th patient's bone marrow failure cause may be attributed to several factors such as 1. Direct effect of the drug on bone tissues 2. Drug induced allergic response 3. myelosuppression worsened by concom-itant radiotherapy. Research Article
... Radium-223 is an α-emitter with preferential uptake into areas of increased bone formation, a characteristic that prolongs the time to SRE events and increases overall survival [130]. The rate of myelosuppression using radium-223 is small, with only mild thrombocytopenia being observed [131]. ...
Article
Full-text available
Bladder cancer (BCa) is the 10th most common and 13th most deadly malignancy worldwide. About 5% of BCa patients present initially with metastatic disease, with bone being the most diagnosed site for distant metastasis. The overall one-year survival of patients with BCa is 84%, whereas it is only 21% in patients with bone metastasis (BM). Metastasis of BCa cells to bone occurs by epithelial-to-mesenchymal transition, angiogenesis, intravasation, extravasation, and interactions with the bone microenvironment. However, the mechanism of BCa metastasis to the bone is not completely understood; it needs a further preclinical model to completely explain the process. As different imaging mechanisms, PET-CT cannot replace a radionuclide bone scan or an MRI for diagnosing BM. The management of BCa patients with BM includes chemotherapy, immunotherapy, targeted therapy, antibody-drug conjugates, bisphosphonates, denosumab, radioisotopes, and surgery. The objective of these treatments is to inhibit disease progression, improve overall survival, reduce skeletal-related events, relieve pain, and improve the quality of life of patients.
Chapter
Full-text available
Article
Full-text available
This review describes the basic principles of radiometal-theranostics and its dawn based on the development of the positron-emitting ⁸⁶ Y and ⁸⁶ Y-labeled radiopharmaceuticals to quantify biodistribution and dosimetry of ⁹⁰ Y-labeled analogue therapeutics. The nuclear and inorganic development of ⁸⁶ Y (including nuclear and cross section data, irradiation, radiochemical separation and recovery) led to preclinical and clinical evaluation of ⁸⁶ Y-labeled citrate and EDTMP complexes and yielded organ radiation doses in terms of mGy/MBq ⁹⁰ Y. The approach was extended to [ 86/90 Y]Y-DOTA-TOC, yielding again yielded organ radiation doses in terms of mGy/MBq ⁹⁰ Y. The review further discusses the consequences of this early development in terms of further radiometals that were used ( ⁶⁸ Ga, ¹⁷⁷ Lu etc.), more chelators that were developed, new biological targets that were addressed (SSTR, PSMA, FAP, etc.) and subsequent generations of radiometal-theranostics that resulted out of that.
Article
Full-text available
A large proportion of the practice of radiotherapy in the management of metastatic adenocarcinoma of the prostate is associated with palliation of pain from osseous metastases and improving quality of life. Strontium-89 is a systemic radionuclide that has clinical efficacy in the palliation of pain from bony metastases. The study was a Phase-3 randomized placebo control trial performed in eight Canadian Cancer Centers to evaluate the effectiveness of strontium-89 as an adjunct to local field radiotherapy. Patients with endocrine refractory metastatic prostate cancer received local field radiotherapy and either strontium-89 as a single injection of 10.8 mCi or placebo. One hundred twenty-six patients were recruited. No significant differences in survival or in relief of pain at the index site were noted. Intake of analgesics over time demonstrated a significant reduction in the arm treated with strontium-89. Progression of pain as measured by sites of new pain or the requirement for radiotherapy showed statistically significant differences between the arms in favor of strontium-89. Tumor markers including prostate specific antigen, acid phosphatase, and alkaline phosphatase were also reduced in patients receiving strontium-89. A Quality-of-Life analysis was performed as a multivariate data set and demonstrated an overall superiority of strontium-89 with alleviation of pain and improvement in physical activity being statistically significant. Toxicity was evaluated and demonstrated increased hematological toxicity in the group receiving strontium-89. It is concluded that the addition of strontium-89 is an effective adjuvant therapy to local field radiotherapy reducing progression of disease as evidenced by new sites of pain and the requirement of further radiotherapy and improving quality-of-life and need for analgesic support in this group of patients. 24 refs., 7 figs., 2 tabs.
Article
Full-text available
Prostate-specific antigen (PSA) is a glycoprotein that is found almost exclusively in normal and neoplastic prostate cells. For patients with metastatic disease, changes in PSA will often antedate changes in bone scan. Furthermore, many but not all investigators have observed an association between a decline in PSA levels of 50% or greater and survival. Since the majority of phase II clinical trials for patients with androgen-independent prostate cancer (AIPC) have used PSA as a marker, we believed it was important for investigators to agree on definitions and values for a minimum set of parameters for eligibility and PSA declines and to develop a common approach to outcome analysis and reporting. We held a consensus conference with 26 leading investigators in the field of AIPC to define these parameters. We defined four patient groups: (1) progressive measurable disease, (2) progressive bone metastasis, (3) stable metastases and a rising PSA, and (4) rising PSA and no other evidence of metastatic disease. The purpose of determining the number of patients whose PSA level drops in a phase II trial of AIPC is to guide the selection of agents for further testing and phase III trials. We propose that investigators report at a minimum a PSA decline of at least 50% and this must be confirmed by a second PSA value 4 or more weeks later. Patients may not demonstrate clinical or radiographic evidence of disease progression during this time period. Some investigators may want to report additional measures of PSA changes (ie, 75% decline, 90% decline). Response duration and the time to PSA progression may also be important clinical end point. Through this consensus conference, we believe we have developed practical guidelines for using PSA as a measurement of outcome. Furthermore, the use of common standards is important as we determine which agents should progress to randomized trials which will use survival as an end point.
Article
Full-text available
The therapeutic efficacy of the alpha-particle-emitting radionuclide (223)Ra (t(1/2) = 11.4 days) in the treatment against experimental skeletal metastases in rats was addressed. Biodistribution studies, involving measurement of (223)Ra in bone marrow samples, were performed in rats after i.v. injection. To study the therapeutic effect of (223)Ra, an experimental skeletal metastases model in nude rats was used. Animals that had received 10(6) MT-1 human breast cancer cells were treated with (223)Ra doses in the range of 6-30 kBq after 7 days. The biodistribution experiment demonstrated that (223)Ra was selectively concentrated in bone as compared with soft tissues. The femur content of (223)Ra was 800 +/- 56% of injected dose per gram tissue times gram body weight (b.w.; mean +/- SD) 1 day after the injection and 413 +/- 23% of injected dose per gram tissue times gram b.w. at 14 days. The femur:kidney ratio increased from (5.9 +/- 2.0).10(2) at 1 day to (7.2 +/- 3.0).10(2) at 14 days, whereas the femur:liver ratio increased from (6.2 +/- 0.2).10(2) to (9.1 +/- 6.6).10(2). Femur:spleen ratio increased from (8.1 +/- 0.3).10(2) at 1 day to (6.4 2.2).10(3) at 14 days. The femoral bone:marrow ratio was 6.5 +/- 2.1 after day 1 and larger than 15 at day 14. All of the tumor-bearing control animals had to be sacrificed because of tumor-induced paralysis 20-30 days after injection with tumor cells, whereas the rats treated with > or =10 kBq of (223)Ra had a significantly increased symptom-free survival (P < 0.05). Also 36% (5 of 14) of rats treated with 11 kBq and 40% (2 of 5) of rats treated with 10 kBq were alive beyond the 67-day follow-up period. No signs of bone marrow toxicity or b.w. loss were observed in the groups of treated animals. The significant antitumor effect of (223)Ra at doses that are tolerated by the bone marrow is most likely linked to the intense and highly localized radiation dose from alpha-particles at the bone surfaces. The results of this study indicate that (223)Ra should be additionally studied as a potential bone marrow-sparing treatment of cancers involving the skeleton.
Article
Full-text available
Bone metastases are a common cause of morbidity in patients with prostate carcinoma. We studied the effect of a new bisphosphonate, zoledronic acid, which blocks bone destruction, on skeletal complications in prostate cancer patients with bone metastases. Patients with hormone-refractory prostate cancer and a history of bone metastases were randomly assigned to a double-blind treatment regimen of intravenous zoledronic acid at 4 mg (N = 214), zoledronic acid at 8 mg (subsequently reduced to 4 mg; 8/4) (N = 221), or placebo (N = 208) every 3 weeks for 15 months. Proportions of patients with skeletal-related events, time to the first skeletal-related event, skeletal morbidity rate, pain and analgesic scores, disease progression, and safety were assessed. All statistical tests were two-sided. Approximately 38% of patients who received zoledronic acid at 4 mg, 28% who received zoledronic acid at 8/4 mg, and 31% who received placebo completed the study. A greater proportion of patients who received placebo had skeletal-related events than those who received zoledronic acid at 4 mg (44.2% versus 33.2%; difference = -11.0%, 95% confidence interval [CI] = -20.3% to -1.8%; P =.021) or those who received zoledronic acid at 8/4 mg (38.5%; difference versus placebo = -5.8%, 95% CI = -15.1% to 3.6%; P =.222). Median time to first skeletal-related event was 321 days for patients who received placebo, was not reached for patients who received zoledronic acid at 4 mg (P =.011 versus placebo), and was 363 days for those who received zoledronic acid at 8/4 mg (P =.491 versus placebo). Compared with urinary markers in patients who received placebo, urinary markers of bone resorption were statistically significantly decreased in patients who received zoledronic acid at either dose (P =.001). Pain and analgesic scores increased more in patients who received placebo than in patients who received zoledronic acid, but there were no differences in disease progression, performance status, or quality-of-life scores among the groups. Zoledronic acid at 4 mg given as a 15-minute infusion was well tolerated, but the 8-mg dose was associated with renal function deterioration. Zoledronic acid at 4 mg reduced skeletal-related events in prostate cancer patients with bone metastases.
Article
Background: Prostate carcinoma is linked to osteoblastic metastasis. We therefore investigated the value of bone-targeted consolidation therapy in selected patients with advanced androgen-independent carcinoma of the prostate. Methods: 103 patients received induction chemotherapy, consisting of ketoconazole and doxorubicin alternating with estramustine and vinblastine. After two or three cycles of induction chemotherapy, we randomly assigned 72 patients who were clinically stable or responders to receive doxorubicin with or without strontium-89 (Sr-89) every week for 6 weeks. Findings: Overall 62 of the 103 (60%, 95% CI 50-70) patients had a 50% or greater reduction in serum prostate-specific antigen concentration that was maintained for at least 8 weeks, and 43 (42%, 32-52) had an 80% or greater reduction. 49 (52%) patients with bone pain at registration had complete resolution of pain. After follow-up of 67 patients until death, the estimated median survival for all 103 patients was 17.5 months (range 0.5-37.7). For the 36 patients randomly assigned to receive Sr-89 and doxorubicin, the median survival time was 27.7 months (4.9-37.7), and for the 36 who received doxorubicin alone it was 16.8 months (4.4-34.2) (p=0.0014). The hazard ratio was 2.76 (95% CI 1.44-5.29). Interpretation: Bone-targeted consolidation therapy consisting of one dose of Sr-89 plus doxorubicin once a week for 6 weeks, when given to patients with stable or responding advanced androgen-independent carcinoma of the prostate after induction chemotherapy, improved overall survival.
Article
Most patients with metastatic prostate cancer will have metastasis to bone. Such patients are best monitored by serial radionuclide bone scans. One hundred sixty six men with bone metastasis from prostate cancer who received androgen deprivation therapy had their pretreatment bone scans reviewed using a semiquantitative grading system based upon the extent of disease (EOD) observed on the scan. The EOD on the scan correlated with survival. The 2-year survival rates for EOD I to IV were 94%, 74%, 68%, and 40%, respectively. The survival of patients in categories EOD I and IV significantly differed from the other categories. Men with metastatic prostate cancer entered into trials designed to evaluate the impact of treatment on survival should be stratified based upon the EOD on the bone scan. This analysis also indicates that patients in the EOD IV category have a particularly poor prognosis and may be candidates for alternative treatments.
Article
Purpose: A large proportion of the practice of radiotherapy in the management of metastatic adenocarcinoma of the prostate is associated with palliation of pain from osseous metastases and improving quality of life. Radiation therapy is well known to be effective in treating painful sites and may also be effective in reducing the propensity for adjuvantly treated disease to become symptomatic. Strontium-89 is a systemic radionuclide that has clinical efficacy in the palliation of pain from bony metastases. Methods and materials: The study was a Phase-III randomized placebo control trial performed in eight Canadian Cancer Centers to evaluate the effectiveness of strontium-89 as an adjunct to local field radiotherapy. Patients with endocrine refractory metastatic prostate cancer received local field radiotherapy and either strontium-89 as a single injection of 10.8 mCi or placebo. Results: One hundred twenty-six patients were recruited. No significant differences in survival or in relief of pain at the index site where noted. Intake of analgesics over time demonstrated a significant reduction in the arm treated with strontium-89. Progression of pain as measured by sites of new pain or the requirement for radiotherapy showed statistically significant differences between the arms in favor of strontium-89. Tumor makers including prostate specific antigen, acid phosphatase, and alkaline phosphatase were also reduced in patients receiving strontium-89. A Quality-of-Life analysis was performed as a multivariate data set and demonstrated an overall superiority of strontium-89 with alleviation of pain and improvement in physical activity being statistically significant. Toxicity was evaluated and demonstrated increased hematological toxicity in the group receiving strontium-89. Conclusions: It is concluded that the addition of strontium-89 is an effective adjuvant therapy to local field radiotherapy reducing progression of disease as evidenced by new sites of pain and the requirement of further radiotherapy and improving quality-of-life and need for analgesic support in this group of patients.