CLINICAL PHARMACOLOGY
Mechanism Of Action
Xgeva binds to RANKL, a transmembrane or soluble protein
essential for the formation, function, and survival of osteoclasts, the cells
responsible for bone resorption, thereby modulating calcium release from bone.
Increased osteoclast activity, stimulated by RANKL, is a mediator of bone
pathology in solid tumors with osseous metastases. Similarly, giantcelltumors
of bone consist of stromal cells expressing RANKL and osteoclast-like giant
cells expressing RANK receptor, and signaling through the RANK receptorcontributes
to osteolysis and tumor growth. Xgeva prevents RANKL from activating its
receptor, RANK, on the surface of osteoclasts, their precursors, and
osteoclast-like giant cells.
Pharmacodynamics
In patients with breast cancer and bone metastases, the
median reduction in uNTx/Cr was 82% within 1 week following initiation of Xgeva
120 mg administered subcutaneously. In Studies 20050136, 20050244, and
20050103, the median reduction in uNTx/Cr from baseline to Month 3 was
approximately 80% in 2075 Xgeva-treated patients.
In a phase 3 study of patients with newly diagnosed
multiple myeloma who received SC doses of Xgeva 120 mg every 4 weeks (Q4W),
median reductions in uNTx/Cr of approximately 75% were observed by week 5.
Reductions in bone turnover markers were maintained, with median reductions of
74% to 79% for uNTx/Cr from weeks 9 to 49 of continued 120 mg Q4W dosing.
Pharmacokinetics
Following subcutaneous administration, bioavailability
was 62%. Denosumab displayed nonlinear pharmacokinetics at doses below 60mg,
but approximately dose-proportional increases in exposure at higher doses.
With multiple subcutaneous doses of 120mg once every
4weeks, up to 2.8-fold accumulation in serum denosumab concentrations was
observed and steady state was achieved by 6months. A mean (± standard
deviation) serum steady-state trough concentration of 20.5 (± 13.5)mcg/mL was
achieved by 6 months. The mean elimination half-life was 28 days.
In patients with newly diagnosed multiple myeloma who
received 120 mg every 4 weeks, denosumab concentrations appear to reach
steady-state by month 6. In patients with giant cell tumor of bone, after
administration of subcutaneous doses of 120mg once every 4weeks with additional
120mg doses on Days 8 and 15 of the first month of therapy, mean (± standard
deviation) serum trough concentrations on Day8, 15, and one month after the
first dose were 19.0 (± 24.1), 31.6 (± 27.3), 36.4(±20.6) mcg/mL, respectively.
Steady-state was achieved in 3 months after initiation of treatment with a mean
serum trough concentration of 23.4 (± 12.1) mcg/mL.
Special Populations
Body Weight
A population pharmacokinetic analysis was performed to
evaluate the effects of demographic characteristics. Denosumab clearance and
volume of distribution were proportional to body weight. The steady-state
exposure following repeat subcutaneous administration of 120mg every 4 weeks to
45 kg and 120 kg subjects were, respectively, 48% higher and 46% lower than
exposure of the typical 66 kg subject.
Age, Gender And Race
The pharmacokinetics of denosumab was not affected by
age, gender, and race.
Pediatrics
The pharmacokinetics of denosumab in pediatric patients
has not been assessed.
Hepatic Impairment
No clinical trials have been conducted to evaluate the
effect of hepatic impairment on the pharmacokinetics of denosumab.
Renal Impairment
In clinical trials of 87patients with varying degrees of
renal dysfunction, including patients on dialysis, thedegree of renal
impairmenthad no effect on the pharmacokinetics and pharmacodynamics of
denosumab [see Use In Specific Populations].
Drug Interactions
No formal drug-drug interaction trials have been
conducted with Xgeva. There was no evidence that various anticancer treatments
affected denosumab systemic exposure and pharmacodynamic effect. Serum
denosumab concentrations at 1 and 3 months and reductions in the bone turnover
marker uNTx/Cr (urinary N-terminal telopeptide corrected for creatinine) at
3months were similar in patients with and without prior intravenous
bisphosphonate therapy and were not altered by concomitant chemotherapy and/or
hormone therapy.
Animal Toxicology And/Or Pharmacology
Denosumab is an inhibitor of osteoclastic bone resorption
via inhibition of RANKL.
Because the biological activity of denosumab in animals
is specific to nonhuman primates, evaluation of genetically engineered
(knockout) mice or use of other biological inhibitors of the RANK/RANKL pathway,
OPG-Fc and RANK-Fc, provided additional information on the pharmacodynamic
properties of denosumab. RANK/RANKL knockout mice exhibited absence of lymph
node formation, as well as an absence of lactation due to inhibition of mammary
gland maturation (lobulo-alveolar gland development during pregnancy). Neonatal
RANK/RANKL knockout mice exhibited reduced bone growth and lack of tooth
eruption. A corroborative study in 2-week-old rats given the RANKLinhibitor
OPG-Fc also showed reduced bone growth, altered growth plates, and impaired
tooth eruption. These changes were partially reversible in this model when
dosing with the RANKL inhibitors was discontinued.
Clinical Trials
Bone Metastasis From Solid Tumors
The safety and efficacy of Xgeva for the prevention of
skeletal-related events in patients with bone metastases from solid tumors was
demonstrated in three international, randomized (1:1), double-blind,
active-controlled, noninferiority trials comparing Xgeva with zoledronic acid.
In all three trials, patients were randomized to receive 120 mg Xgeva
subcutaneously every 4 weeks or 4 mg zoledronic acid intravenously (IV) every
4weeks (dose adjusted for reduced renal function). Patients with creatinine
clearance less than 30 mL/min were excluded. In each trial, the main outcome
measure was demonstration of noninferiority of time to first skeletal-related
event (SRE) as compared to zoledronic acid. Supportive outcome measures were
superiority of time to first SRE and superiority of time to first and
subsequent SRE; testing for these outcome measures occurred if the main outcome
measure was statistically significant. An SRE was defined as any of the
following: pathologic fracture, radiation therapy to bone, surgery to bone, or
spinal cord compression.
Study 20050136 (NCT00321464) enrolled 2046 patients with
advanced breast cancer and bone metastasis. Randomization was stratified by a
history of prior SRE (yes or no), receipt of chemotherapy within 6weeks prior
to randomization (yes or no), prior oral bisphosphonate use (yes or no), and
region (Japan or other countries). Forty percent of patients had a previous
SRE, 40% received chemotherapy within 6weeks prior to randomization, 5%
received prior oral bisphosphonates, and 7% were enrolled from Japan. Median
age was 57 years, 80% of patients were White, and 99% of patients were women.
The median number of doses administered was 18 for denosumab and 17 for
zoledronic acid.
Study 20050244 (NCT00330759) enrolled 1776 adults with
solid tumors other than breast and castrate-resistant prostate cancer with bone
metastasis and multiple myeloma. Randomization was stratified by previous SRE
(yes or no), systemic anticancer therapy at time of randomization (yes or no),
and tumor type (non-small celllung cancer, myeloma, or other). Eighty-seven
percent were receiving systemic anticancer therapy at the time of
randomization, 52% had a previous SRE, 64% of patients were men, 87% were
White, and the median age was 60years. A total of 40% of patients had non-small
cell lung cancer, 10% had multiple myeloma, 9% had renal cell carcinoma, and 6%
had small cell lung cancer. Other tumor types each comprised less than 5% of
the enrolled population. The median number of doses administered was 7 for both
denosumab and zoledronic acid.
Study 20050103 (NCT00321620) enrolled 1901 men with
castrate-resistant prostate cancer and bone metastasis. Randomization was
stratified by previous SRE, PSA level(less than 10ng/mL or 10ng/mL or greater)
and receipt of chemotherapy within 6 weeks prior to randomization (yes or no).
Twenty-six percent of patients had a previous SRE, 15% of patients had PSA less
than 10ng/mL, and 14% received chemotherapy within 6 weeks prior to
randomization. Median age was 71 years and 86% of patients were White. The
median number of doses administered was 13 for denosumab and 11for zoledronic
acid.
Xgevadelayed the timeto first SRE following randomization
as compared to zoledronic acid in patients with breast or castrate-resistant
prostate cancer (CRPC) with osseous metastases (Table 2). In patients with bone
metastasis due to other solid tumors or lytic lesions due to multiple myeloma,
Xgeva was noninferior to zoledronic acid in delaying the time to first SRE
following randomization.
Overall survival and progression-free survival were
similar between arms in allthree trials.
Table 2: Efficacy Results for Xgeva Compared to
Zoledronic Acid
|
Study 20050136 Metastatic Breast Cancer |
Study 20050244 Metastatic Solid Tumors or Multiple Myeloma |
Study 20050103 Metastatic CRPCa |
Xgeva |
Zoledronic Acid |
Xgeva |
Zoledronic Acid |
Xgeva |
Zoledronic Acid |
N |
1026 |
1020 |
886 |
890 |
950 |
951 |
First On-study SRE |
Number of Patients who had SREs (%) |
315 (30.7) |
372 (36.5) |
278 (31.4) |
323 (36.3) |
341 (35.9) |
386 (40.6) |
Components of First SRE |
Radiation to Bone |
82 (8.0) |
119 (11.7) |
119 (13.4) |
144 (16.2) |
177 (18.6) |
203 (21.3) |
Pathological Fracture |
212 (20.7) |
238 (23.3) |
122 (13.8) |
139 (15.6) |
137 (14.4) |
143 (15.0) |
Surgery to Bone |
12 (1.2) |
8 (0.8) |
13 (1.5) |
19 (2.1) |
1 (0.1) |
4 (0.4) |
Spinal Cord Compression |
9 (0.9) |
7 (0.7) |
24 (2.7) |
21 (2.4) |
26 (2.7) |
36 (3.8) |
Median Time to SRE (months) |
NRb |
26.4 |
20.5 |
16.3 |
20.7 |
17.1 |
Hazard Ratio (95% CI) |
0.82 (0.71, 0.95) |
0.84 (0.71, 0.98) |
0.82 (0.71, 0.95) |
Noninferiority p-value |
< 0.001 |
< 0.001 |
< 0.001 |
Superiority p-valuec |
0.010 |
0.060 |
0.008 |
First and Subsequent SREd |
Mean Number/Patient |
0.46 |
0.60 |
0.44 |
0.49 |
0.52 |
0.61 |
Rate Ratio (95% CI) |
0.77 (0.66, 0.89) |
0.90 (0.77, 1.04) |
0.82 (0.71, 0.94) |
Superiority p-valuee |
0.001 |
0.145 |
0.009 |
a CRPC =castrate-resistant prostate cancer.
b NR = not reached.
c Superiority testing performed only after denosumab demonstrated to
be noninferior to zoledronic acid within trial.
d Allskeletal events postrandomization; new events defined by
occurrence≥ 21 days after preceding event.
e Adjusted p-values are presented. |
Multiple Myeloma
The efficacy of Xgeva for the
prevention of skeletal-related events in newly diagnosed multiple myeloma
patients with treatment through disease progression, was evaluated in Study
20090482 (NCT01345019), an international, randomized (1:1), double-blind, active-controlled,
noninferiority trial comparing Xgeva with zoledronic acid. In this trial,
patients were randomized to receive 120 mg Xgeva subcutaneously every 4 weeks
or 4 mg zoledronic acid intravenously (IV) every 4 weeks (dose adjusted for
reduced renal function). Patients with creatinine clearance less than 30 mL/min
were excluded. In this trial, the main efficacy outcome measure was
noninferiority of time to first skeletal-related event (SRE). Additional
efficacy outcome measures were superiority of time to first SRE, time to first
and subsequent SRE, and overall survival. An SRE was defined as any of the
following: pathologic fracture, radiation therapy to bone, surgery to bone, or
spinal cord compression.
Study 20090482 enrolled 1718 newly diagnosed multiple
myeloma patients with bone lesions. Randomization was stratified by a history
ofprior SRE (yes or no), the anti-myeloma agent being utilized/planned to be
utilized in first-line therapy (novel therapy-based or non-novel therapy-based
[novel therapies include bortezomib, lenalidomide, or thalidomide]), intent to
undergo autologous PBSC transplantation (yes or no), stage at diagnosis
(International Staging System I or II or III) and region Japan (yes or no). At
study enrollment, 96% of the patients were receiving or planning to receive
novel therapy based first-line anti-myeloma therapy, 55% of the patients
intended to undergo autologous PBSC transplantation, 61% of patients had a
previous SRE, 32% were at ISS stage I, 38% were at ISS stage II and 29% were at
ISS Stage III, and 2% were enrolled from Japan. Median age was 63 years, 82% of
patients were White, and 46% of patients were women. The median number of doses
administered was 16 for Xgeva and 15 for zoledronic acid.
Xgeva was noninferior to zoledronic acid in delaying the time
to first SRE following randomization (HR=0.98, 95% CI, 0.85-1.14). The results
for overall survival (OS) were comparable between Xgeva and zoledronic acid
treatment groups with a hazard ratio of 0.90 (95% CI: 0.70, 1.16).
Table 3: Efficacy Results for Xgeva Compared to
Zoledronic Acid
|
Study 20090482 Multiple Myeloma |
Xgeva |
Zoledronic Acid |
N |
859 |
859 |
First On-study SRE |
Number of Patients who had SREs (%) |
376(43.8) |
383(44.6) |
Components of First SRE |
Radiation to Bone |
47(5.5) |
62(7.2) |
Pathological Fracture |
342(39.8) |
338(39.3) |
Surgery to Bone |
37(4.3) |
48(5.6) |
Spinal Cord Compression |
6(0.7) |
4(0.5) |
Median Time to SRE (months)(95% CI) |
22.8(14.7, NEa) |
24(16.6, 33.3) |
Hazard Ratio (95% CI) |
0.98 (0.85, 1.14) |
aNE = not estimable |
Giant Cell Tumor Of Bone
The safety and efficacy of
Xgeva for the treatment of giant cell tumor of bone in adults or skeletally
mature adolescents were demonstrated in two open-label trials [Study20062004
(NCT00680992)and Study20040215 (NCT00396279)]that enrolled patients with histologically
confirmed measurable giant celltumor of bone that was either recurrent,
unresectable, or for which planned surgery was likely to result in severe
morbidity. Patients received 120 mg Xgeva subcutaneously every 4 weeks with
additional doses on Days 8 and 15 of the first cycle of therapy.
Study 20062004was a single arm, pharmacodynamic, and
proof of concept trial conducted in 37adult patients with unresectable or
recurrent giant cell tumor of bone. Patients were required to have
histologically confirmed giant cell tumor of bone and radiologic evidence of
measurable disease from a computed tomography (CT) or magnetic resonance
imaging (MRI) obtained within 28days prior to study enrollment. Patients
enrolled in Study 20062004underwent CTor MRI assessment of giant cell tumor of
bone at baseline and quarterly during Xgeva treatment.
Study 20040215was a parallel-cohort, proof of concept,
and safety trial conducted in 282adult or skeletally mature adolescent patients
with histologically confirmed giant cell tumor of bone and evidence of
measurable active disease. Study 20040215 enrolled 10 patients who were 13 -17
years of age [see Use In Specific Populations]. Patients enrolled into
one of three cohorts: Cohort 1 enrolled 170patients with surgically
unsalvageable disease (e.g., sacral or spinal sites of disease, or pulmonary
metastases); Cohort 2 enrolled 101 patients with surgically salvageable disease
where the investigator determined that the planned surgery was likely to result
in severe morbidity (e.g., joint resection, limb amputation, or
hemipelvectomy); Cohort 3 enrolled 11 patients who previously participated in
Study 20062004. Patients underwent imaging assessment of disease status at
intervals determined by their treating physician.
An independent review committee evaluated objective
response in 187 patients enrolled and treated in Study 20062004 and Study
20040215 for whom baseline and at least one post-baseline radiographic
assessment were available (27 of 37 patients enrolled in Study 20062004 and 160
of 270 patients enrolled in Cohorts1 and 2 of Study 20040215). The primary
efficacy outcome measure was objective response rate using modified Response
Evaluation Criteria in Solid Tumors (RECIST 1.1).
The overall objective response rate (RECIST1.1) was 25%
(95%CI: 19, 32). All responses were partial responses. The estimated median
time to response was 3 months. In the 47 patients with an objective response,
the median duration of follow-up was 20months (range: 2 to 44months), and 51%
(24/47) had a duration of response lasting at least 8 months. Three patients
experienced disease progression following an objective response.
Hypercalcemia Of Malignancy
The safety and efficacy of Xgeva was demonstrated in an open-label,
single-arm trial [Study 20070315 (NCT00896454)] that enrolled 33 patients with
hypercalcemia of malignancy (with or without bone metastases) refractory to
treatment with intravenous bisphosphonate therapy. Patients received Xgeva
subcutaneously every 4 weeks with additional 120 mg doses on Days 8 and 15 of
the first month of therapy.
In this trial, refractory hypercalcemia of malignancy was
defined as an albumin-corrected calcium of >12.5mg/dL (3.1mmol/L) despite
treatment with intravenous bisphosphonate therapy in 7-30days prior to
initiation of Xgeva therapy. The primary outcome measure was the proportion of
patients achieving a response, defined as corrected serum calcium (CSC) ≤
11.5 mg/dL (2.9 mmol/L), within 10 days after Xgeva administration. Efficacy
data are summarized in Figure 1 and Table 4. Concurrent chemotherapy did not
appear to affect response to Xgeva.
Figure 1: Corrected Serum Calcium by Visit in
Responders (Median and Interquartile Range)
Table 4: Efficacy in Patients with Hypercalcemia of Malignancy Refractory to Bisphosphonate Therapy
|
N = 33 |
Proportion (% ) (95% CI) |
All Responders (CSC ≤ 11.5 mg/dL) by Day 10 |
21 |
63.6 (45.1, 79.6) |
All Responders by Day 57 |
23 |
69.7 (51.3, 84.4) |
Complete Responders (CSC ≤ 10.8 mg/dL) by Day 10 |
12 |
36.4 (20.4, 54.9) |
All Complete Responders by Day 57 |
21 |
63.6 (45.1, 79.6) |
Median time to response (CSC
≤ 11.5mg/dL) was 9days (95%CI: 8, 19), and the median duration of
response was 104days (95%CI: 7, not estimable). Median time to complete
response (CSC ≤ 10.8mg/dL) was 23days (95%CI: 9, 36), and the median
duration of complete response was 34 days (95% CI: 1, 134).