Zoledronic Acid

advertisement
19th Annual NOCR Meeting
Session I: Breast Cancer
Bone Directed Therapy in Breast Cancer
Christy A Russell, MD
Keck School of Medicine
University of Southern California
Indications: Bisphosphonates vs
RANK-Ligand inhibitor
•Zoledronic acid is indicated for the treatment
of HCM and patients with multiple myeloma and
patients with documented bone metastases
from solid tumors, in conjunction with standard
antineoplastic therapy.
•Denosumab is indicated for the prevention of
skeletal related events in patients with bone
metastases from solid tumors.
Pathogenesis of Osteolytic Bone Metastases
Tumor Cells in Bone
Tumor-derived osteoclast
activating factors
• Parathyroid hormonerelated protein
• Interleukins -6, -8, -11
• Tumor necrosis factor
• Macrophage colonystimulating factor
(+)
(+)
Osteoclast
Bone-derived tumor
growth factors
• Transforming growth
factor 
• Insulin-like growth
factors
• Fibroblast growth factors
• Platelet-derived growth
factor
• Bone morphogenic
proteins
Bone
Derived from Roodman GD. N Engl J Med. 2004;350:1655-1664.
Pathogenesis of Osteoblastic Bone Metastases
Tumor Cells
Bone-derived
growth factors
Tumor-derived
osteoclast
activating
factors
• PTH-RP
• IL-6
(+)
Osteoclast
Osteoblast-derived
signals
(+)
Tumor-derived
osteoblast
growth factors
• ET-1
• TGF-
• FGF
• BMPs
• IGFs
Osteoblasts
Bone
Adapted with permission from Saad F et al. Eur Urol. 2004;45:26-34.
Mechanism of Bisphosphonate Inhibition of
Osteoclast Activity
Bisphosphonates
inhibit osteoclast
activity, and promote
osteoclast apoptosis1
X
Bisphosphonates may modulate
signaling from osteoblasts
to osteoclasts
 Increased OPG production2
 Decreased RANKL expression3
New bone
Bone
Bisphosphonates
are released locally
during bone resorption1
Bisphosphonates are
concentrated under
osteoclasts1
1. Reszka AA, Rodan GA. Curr Rheumatol Rep. 2003;5:65-74. 2. Viereck V et al. Biochem Biophys
Res Commun. 2002;291:680-686. 3. Pan B et al. J Bone Miner Res. 2004;19:147-154.
Receptor Activator of Nuclear Factor kB
Ligand (RANKL) and Osteoprotegerin (OPG)
Stromal cell/Osteoblast
Parathyroid hormone/
Parathyroid hormone–related protein
RANKL
1,25D3
PGE2
OPG
RANK
Interleukin-11
Osteoclast Precursor
Derived from Roodman GD. N Engl J Med. 2004;350:1655-1664.
Osteoclast
Breast Cancer
The Natural History of Bone
Metastases in Breast Cancer
• Pathologic fracture is the most common SRE in
patients with breast cancer
• Median onset is 11 mos from initial diagnosis of
bone metastases
• ~ 20% develop hypercalcemia after a median of
14 mos
• ~ 10% develop cord compression after a median
of 17 mos
Lipton A. Cancer. 2003;97:848-853.
Untreated Patients Experience Multiple
SREs
Skeletal Morbidity Rate*
4.5
4.0
4.00
3.70
3.5
3.0
2.71
2.5
2.0
1.47
1.5
1.0
0.5
0
SRE
SRE + HCM
Breast Cancer[1]
*Mean number of SRE per patient per yr.
Prostate CA[2]
NSCLC +
Other Solid
Tumors[3]
1. Lipton A, et al. Cancer. 2000;88:1082-1090. 2. Saad F. Clin Prostate Cancer. 2005;4:31-37.
3. Rosen LS, et al. Cancer. 2004;100:2613-2621.
FDA-Approved Agents for Prevention of
SREs in Metastatic Breast Cancer
Agent
Drug Class
Recommended Dose and Schedule
Zoledronic acid
Bisphosphonate
4 mg IV q3-4w
Pamidronate
Bisphosphonate
90 mg IV q3-4w
Denosumab
RANKL-targeted MAb
120 mg SQ q4w
• Both ASCO and NCCN recommend all 3 agents[1,2]
– No agent recommended over another
1. Van Poznak CH, et al. J Clin Oncol. 2011;29:1221-1227. 2. NCCN. Clinical practice guidelines in
oncology: breast cancer. v.2.2011.
Proportion of Breast Cancer Patients Having Skeletal-Related
Events (SREs) With Pamidronate
80
12 Months1
P=0.005
P=0.002
70
Patients (%)
60
50
40
24 Months2
P=0.49*
P=0.001†
56%
P<0.001
42%
52%
43%
33%
40%
29%
30
20
P=0.002
64%
51%
49%
43%
P≤0.001
19%
10
0
All SREs Radiation
to Bone
Fracture
Placebo
All SREs Radiation Fracture
to Bone
Pamidronate
*P=0.49 for nonvertebral fracture; †P=0.001 for vertebral fracture.
1. Hortobagyi GN et al. N Engl J Med. 1996;335:1785-1791. 2. Lipton A et al. Cancer. 2000;88:1082-1090.
Bisphosphonates Reduce SREs in Breast
Cancer
Study
Lipton et al[1]*
Treatment
Duration, Mos
Patients With
SRE, %
24
 Placebo
64
 Pamidronate
51
Rosen et al[2]
< .001
24
 Pamidronate
49
 Zoledronic acid
46
Kohno et al[3]
P Value
NS
12
 Placebo
50
 Zoledronic acid
30
*Includes HCM.
1. Lipton A, et al. Cancer. 2000;88:1082-1090. 2. Rosen LS, et al. Cancer. 2003;98:1735-1744.
3. Kohno N, et al. J Clin Oncol. 2005;23:3314-3321.
.003
Proportion of Patients With Bone
Metastases Without an SRE
Zoledronic Acid Significantly Delays Time to
First SRE Compared With Placebo
1.0
0.9
0.8
0.7
0.6
P = .004
0.5
0.4
0.3
0.2
0.1
0
Zoledronic acid 4 mg
Placebo
0
50
100
150
200
250
300
Days After Start of Study Drug
350
400
Kohno N, et al. SABCS 2004. Abstract 3060. Kohno N, et al. J Clin Oncol. 2005;23:3314-3321. Reprinted
with permission.
Zoledronic Acid vs Placebo in Stage IV
Breast Cancer With Bone Metastases
100
90
Zoledronic acid 4 mg (n = 114)
Placebo (n = 113)
80
Patients (%)
70
60
52.2
50
40
38.9
30.7
30
17.7
20
11.5
8.8
10
0
25.4
All SREs
Radiation to
bone
Kohno N, et al. J Clin Oncol. 2005;23:3314-3321.
Fractures
Events at 12 Mos
3.
5
Spinal cord
compression
2.6
8.8
HCM
Zoledronic Acid and Pamidronate in
Breast Cancer and Multiple Myeloma Patients With
Bone Metastases: 13-Month Data
100
47%
49%
40
20
Patients with no event (%)
Patients with SRE (%)
60
Zoledronic acid 4 mg
Pamidronate 90 mg
80
60
40
Median time (days)
Zoledronic acid: 373
Pamidronate:
363
20
0
0
Pamidronate Zoledronic
90 mg
acid 4 mg
All SREs
0
100 200 300 400 500
Time after start of drug (days)
SREs=skeletal-related events.
Adapted with permission from Rosen LS et al. Cancer J. 2001;7:377-387.
Zoledronic Acid vs Pamidronate in Breast Cancer and
Multiple Myeloma: 25-Month Data
Zoledronic acid
4 mg
Pamidronate
90 mg
P value
Proportion
with skeletal-related
event (SRE) (%)
47
51
0.243
Median time to SRE
(days)*
376
356
0.151
Mean skeletal
morbidity rate*
1.04
1.39
0.084
Multiple event analysis
(risk ratio)*
0.841
—
0.030
*Hypercalcemia of malignancy is included as an SRE.
Rosen LS et al. Cancer. 2003;98:1735-1744.
Zoledronic acid is More Effective than Pamidronate
In Reducing the Risk of Skeletal Complications
Multiple Event Analysis: Breast Cancer and Multiple Myeloma
Risk
reduction
P value
7%
.593
20%
.025
16%
.030
0.932
Multiple
myeloma
Breast
cancer
0.799
0.841
Total
0
0.2 0.4 0.6 0.8
1
1.2 1.4 1.6 1.8
Risk ratio (zoledronic acid 4 mg versus pam)
In favor of zoledronic acid
Rosen LS, et al. Cancer. 2003;98:1735-1744.
In favor of pamidronate
2
Patients without increase
(%)
Renal Profile of Pamidronate and Zoledronic Acid
in Patients With Metastatic Breast Cancer or
Multiple Myeloma
100
80
60
40
Zoledronic acid 4 mg
20
Pamidronate 90 mg
0
0
Zoledronic acid 4 mg
Pamidronate
90 mg
120
272
268
240
360
480
600
Time after start of study drug (days)
226
213
197
182
152
138
*Post–15-minute infusion amendment.
Adapted with permission from Berenson JR. The Oncologist. 2005;10:52-62.
81
73
720
68
59
30
27
3 Identical International, Randomized,
Double-Blind, Active-Controlled Trials
Enrollment Criteria
 Adults with breast, prostate,
or other solid tumors and
bone metastases or
multiple myeloma
 No current or previous IV
bisphosphonate
administration for treatment
of bone metastases
Denosumab 120 mg SC and Placebo IV*
q4w (n = 2862)
Supplemental Calcium and
Vitamin D Recommended
Zoledronic Acid 4 mg IV* and Placebo SC
q4w (n = 2861)
1° Endpoint
 Time to first on-study SRE (noninferiority)
2°
Endpoints
 Time to first on-study SRE (superiority)
 Time to first and subsequent on-study SRE (superiority)
*Per protocol and zoledronic acid label, IV product dose adjusted for baseline creatinine.
Denosumab vs Zoledronic Acid Pivotal
Phase III SRE Prevention Trials
In total, > 5700 patients with bone metastases
Study 136[1]
Breast cancer
(N = 2049)
Study 103[2]
Prostate cancer
(N = 1904)
Study 244[3]
Other solid tumors/MM
(N = 1779)
R
A
N
D
O
M
I
Z
A
T
I
O
N
Denosumab 120 mg SC q4w
+
Placebo IV q4w†
Supplemental calcium and vitamin D
Zoledronic Acid 4 mg IV q4w†
+
Placebo SC q4w
1. Stopeck AT, et al. J Clin Oncol. 2010;28:5132-5139. 2. Fizazi K, et al. Lancet. 2011;377:813-822.
3. Henry DH, et al. J Clin Oncol. 2011;29:1125-1132.
SRE Rate: Denosumab vs ZA in Breast
Cancer Patients With Bone Metastases
SREs per Patient per Yr
1.2
1.0
0.8
0.6
-22% (P = .004)
0.58
0.45
0.4
0.2
0
ZA
Stopeck AT, et al. J Clin Oncol. 2010;28:5132-5139.
Denosumab
Time to First On-Study SRE:
Extended Analysis
HR: 0.82 (95% CI: 0.71-0.95;
P = .0096, superiority)
Subjects Without SRE (%)
1.0
0.8
0.6
0.4
KM Estimate of
Median Mos
32.7
Denosumab
27.4
Zoledronic acid
0.2
0
0
Patients at Risk, n
Zoledronic acid 1020
Denosumab 1026
3
6
9
12
15
18
Study Mo
21
24
27
30
33
831
834
675
692
584
597
498
510
265
280
186
193
111
101
38
38
4
9
Stopeck A, et al. SABCS 2010. Abstract P6-14-01.
429
444
356
384
Cumulative Mean Number of SRE
Time to First and Subsequent On-Study
SRE* (Multiple Event Analysis)
Rate ratio: 0.77 (95% CI: 0.66-0.89;
P = .001†)
1.5
1.0
Total No. of
Events
0.5
474
608
Denosumab
Zoledronic acid
0
0
3
6
9
12
15 18
Mos
21
*Events that occurred at least 21 days apart. †Adjusted for multiplicity.
Stopeck AT, et al. J Clin Oncol. 2010;28:5132-5139.
24
27
30
Pooled Analysis: Time to First On-Study
SRE by Previous SRE History
With Previous SRE
Without Previous SRE
Zoledronic acid (n = 1091)
Denosumab (n = 1094)
Zoledronic acid (n = 1910)
Denosumab (n = 1912)
HR: 0.82 (95% CI: 0.70-0.96;
P = .015)
HR: 0.83 (95% CI: 0.72-0.97;
P = .021)
HR: 0.83 (95% CI: 0.74-0.92;
P < .001)
0
0
0
Zoledronic acid (n = 819)
Denosumab (n = 818)
Proportion of Patients
Without On-Study SRE
Overall
1.0
0.8
0.6
0.4
0.2
0
6
12
18
24
30
6
12
18
24
30
6
12
18
24
30
1910 1052 692 382 114
1912 1084 716 402 127
4
4
Study Mo
Risk Set, n
ZA
Dmab
819 425 266 145
818 411 266 144
36
48
0
1
n = number of patients randomized
Lipton A, et al. ASCO 2010. Abstract 9015.
1091 627 426 237 78
1094 673 450 258 79
4
3
Skeletal Complication Risk: Incremental
Benefits in Breast Cancer
No bisphosphonate
Pamidronate
64% risk at 2 yrs
~ 20% risk reduction
Zoledronic acid
Additional ~ 20%
risk reduction
64%
51%
34%
Denosumab
Additional 18%
risk reduction
27%
Lipton A, et al. Cancer. 2000;88:3033-3037. Rosen LS, et al. Cancer. 2003;100:36-43. Stopeck A, et al.
ECCO/ESMO 2009. Abstract 2LBA. Stopeck AT, et al. J Clin Oncol. 2010;28:5132-5139.
Between-Group Differences in AEs With
Unadjusted P < .05
Denosumab,
n (%) (n = 1020)
Pyrexia
Bone pain
Arthralgia
Anemia
Chills
Pain
Renal failure
Dyspepsia
Lumbar vertebral fracture
Alanine aminotransferase increased
Edema
Hypercalcemia
Metastases to spine
Skin hyperpigmentation
Hyperthermia
Bronchospasm
Blood urea increased
Renal failure acute
Toothache
Hypocalcemia
170
186
250
192
29
72
2
52
35
28
22
17
9
7
4
2
0
1
57
56
-10
-5
0
5
Risk Difference
Favors denosumab
Stopeck AT, et al. J Clin Oncol. 2010;28:5132-5139.
(16.7)
(18.2)
(24.5)
(18.8)
(2.8)
(7.1)
(0.2)
(5.1)
(3.4)
(2.7)
(2.2)
(1.7)
(0.9)
(0.7)
(0.4)
(0.2)
(0.0)
(0.1)
(5.6)
(5.5)
10
Favors zoledronic acid
Zoledronic Acid,
n (%) (n = 1013)
247
238
291
232
58
97
25
74
56
47
40
35
21
19
15
10
8
7
37
34
(24.4)
(23.5)
(28.7)
(22.9)
(5.7)
(9.6)
(2.5)
(7.3)
(5.5)
(4.6)
(3.9)
(3.5)
(2.1)
(1.9)
(1.5)
(1.0)
(0.8)
(0.7)
(3.7)
(3.4)
Adverse Events: From Extended Analysis
Event, n (%)
Zoledronic Acid
(n = 1013)
Denosumab
(n = 1020)
All adverse events
987 (97.4)
961 (96.2)
Serious adverse events
509 (50.2)
489 (47.9)
Adverse events related to renal toxicity
95 (9.4)
55 (5.4)
Osteonecrosis of the jaw*
18 (1.8)
26 (2.5)
Hypocalcemia (any)
37 (3.7)
62 (6.1)
12 (1.2)
18 (1.8)
286 (28.2)
109 (10.7)
 Hypocalcemia of grade 3 or 4†
Acute-phase reactions‡
*P = .2861
†No cases of hypocalcemia were grade 5 (fatal).
‡In the first 3 days after initial treatment.
Stopeck A, et al. SABCS 2010. Abstract P6-14-01.
ONJ Associated With Bone-Targeted
Therapy in Patients With Bone Metastases
All patients
(N = 5723)
Potential ONJ
(n = 276)
Positively adjudicated
for ONJ
(n = 89)
Zoledronic acid
(n = 37)
1.3%
Saad F, et al. Ann Oncol. 2012;23:1341-1347.
Denosumab
(n = 52)
1.8%
Integrated analysis of pivotal
denosumab SRE prevention trials
No significant difference
between groups (P = .13)
Associated Oral Events
n (%)
Zoledronic Acid
(n = 37)
Denosumab
(n = 52)
All
(N = 89)
Tooth extraction
24 (65)
30 (58)
54 (61)
Jaw pain
25 (68)
46 (88)
71 (80)
Local infection
17 (46)
26 (50)
43 (48)
Zoledronic Acid
(n = 37)
Denosumab
(n = 52)
All
(N = 89)
Mandible
31 (84)
34 (65)
65 (73)
Maxilla
5 (14)
15 (29)
20 (22)
Both
1 (3)
3 (6)
4 (4)
Location of ONJ
n (%)
Saad F, et al. Ann Oncol. 2012;23:1341-1347.
Systemic Risk Factors
Subjects With ONJ
Subjects Without ONJ
ZA
(n = 37)
Denosumab
(n = 52)
All
(N = 89)
ZA
(n = 2824)
Denosumab
(n = 2810)
All
(N = 5634)
Diabetes
11 (30)
9 (17)
20 (22)
431 (15)
443 (16)
874 (16)
Anemia
(Hb <10)
17 (46)
23 (44)
40 (45)
1185 (42)
1119 (40)
2304 (41)
Chemotherapy
agents
27 (73)
36 (69)
63 (71)
1950 (69)
1921 (68)
3871 (69)
Antiangiogenics
8 (22)
6 (12)
14 (16)
236 (8)
214 (8)
450 (8)
Corticosteroids
28 (76)
39 (75)
67 (75)
1786 (63)
1762 (63)
3548 (63)
n (%)
Saad F, et al. Ann Oncol. 2012;23:1341-1347.
Preventing and Managing ONJ
Risk factors
 Invasive dental procedures
 Poor oral hygiene or pre-existing dental disease
 Advanced malignancies, infections, concomitant therapies
Before bone-targeted
treatment
 Consider dental examination and preventive dentistry
in patients with active dental/jaw conditions
During treatment
 Avoid invasive dental procedures
 Maintain good oral hygiene
Suspected cases
 Refer to dentist or oral surgeon
 Extensive dental surgery may exacerbate
Denosumab. EPAR and summary of product characteristics; Amgen.
Zoledronic acid. Summary of product characteristics; Novartis.
Incidence of Hypocalcemia in the 3 Pivotal
Phase III Trials
Hypocalcemia Events, n (%)
Denosumab
(n = 2841)
Zoledronic Acid
(n = 2836)
Hypocalcemia
273 (9.6)
141 (5.0)
IV calcium Rx
104 (3.7)
47 (1.7)
SAE of hypocalcemia
41 (1.4)
18 (0.6)
 Grade 3*
72 (2.5)
33 (1.2)
 Grade 4*
16 (0.6)
5 (0.2)
*CTCAE grading, grade 3 < 7 mg/dL, grade 4 < 6 mg/dL;
No fatal events of hypocalcemia were reported
Body JJ, et al. Presentation from the 12th International Conference on Cancer-Induced Bone Disease,
November 15-17, 2012, Lyon, France.
Hypocalcemia in Relation to Calcium/Vit D
Supplementation With Denosumab
Patients, n
Incidence of
Hypocalcemia,* n (%)
Reported supplements
2461
213 (8.7)
Did not report supplements
380
60 (15.8)
*All AEs of hypocalcemia.
 Median time to hypocalcemia was 2.8 mos
 Most common in the first 6 mos of initiation of denosumab
therapy
Body JJ, et al. Presentation from the 12th International Conference on Cancer-Induced Bone Disease,
November 15-17, 2012, Lyon, France.
Hypocalcemia in Relation to Tumor Type
With Denosumab Treatment
Primary Tumor Type, n (%)
Patients With Hypocalcemia
Multiple myeloma (N = 86)
12 (14.0)
Prostate (N = 943)
121 (12.8)
Other solid tumors (N = 386)
48 (12.4)
Lung (N = 406)
35 (8.6)
Breast (N = 1020)
57 (5.6)
Body JJ, et al. Presentation from the 12th International Conference on Cancer-Induced Bone Disease,
November 15-17, 2012, Lyon, France.
Preventing and Managing Hypocalcaemia
Situation
Action
Pre-existing hypocalcemia or
vitamin D deficiency
Correct before starting bone-targeted
therapy
Start of bone-targeted therapy
Start daily oral supplements of
≥ 500 mg calcium and 400 IU vitamin D
Severe renal impairment (creatinine
clearance < 30 mL/min) or dialysis
Monitor calcium levels more frequently
Hypocalcemia on therapy
Additional short-term calcium
supplementation may be necessary
Denosumab. Summary of product characteristics; Amgen. Zoledronic acid. Summary of product
characteristics; Novartis. Block G, et al. Poster presentation at the National Kidney Foundation spring
clinical meeting 2010.
Question: What is the Maximum Time You
Provide Bone-Modifying Therapy
Guidelines and Duration of BoneTargeted Therapy
ESMO[1]
“The timing and optimal duration of bisphosphonate
treatment are unknown; benefit of duration beyond 2 yrs
has not been demonstrated . . . Long-term treatment
seems wise due to ongoing risk of skeletal events”
NCCN[2]
“Optimal schedule and duration are unknown . . . Limited
long-term safety data indicating bisphosphonate treatment
can continue beyond 2 yrs”
ASCO[3]
“Until evidence of substantial decline (clinical judgment) in
general performance status”
1. Cardoso F, et al. Ann Oncol. 2011;22(suppl 6) vi 25-30. 2. NCCN. Clinical practice guidelines in
oncology: breast cancer. v3.2012. 3. Van Poznak CH, et al. J Clin Oncol. 2011;29:1221-1227.
2-Yr Open-Label Extension Phase
Adults with
advanced
breast cancer
and confirmed
bone
metastases
Dmab 120 mg SC +
Placebo IV q4w
(n = 1026)
ZA 4 mg IV + Placebo
SC q4w
(n = 1020))
PA
r n
i a
m l
a y
r s
y i
s
Yes (89%) †
Superiority
of Dmab
over ZA
positive
risk:benefit
profile
Patient
choice for
openlabel
Dmab
(n = 752)
Dmab
120 mg SC
q4w 2 yrs
(n = 652)
2-yr survival
follow-up
q12w
Among patients previously receiving denosumab or zoledronic acid, 89% in each treatment
group chose to receive open-label denosumab
Cumulative median exposure to denosumab for the entire study (including blinded and
open-label treatment phases) was 19.1 mos (range: 0.1-59.8 mos, ie, ~ 5 yrs)
 216 patients received denosumab for ≥ 3 yrs
 76 patients received denosumab for ≥ 4 yrs
Stopeck AT, et al. SABCS 2011. Abstract P3-16-07.
AEs During Open-Label Treatment Phase
Event, n (%)
Dmab/Dmab
(n = 318)
ZA/Dmab
(n = 334)
All AEs
283 (89)
303 (91)
Serious AEs
126 (40)
133 (40)
ONJ
20 (6)
18 (5)
Hypocalcemia
12 (4)
9 (3)
Hypocalcemia, grade 3 or 4
4 (1)
3 (1)

No new safety signals were observed with up to ~ 5 yrs of monthly denosumab therapy

Incidence and pattern of AEs in patients who switched from zoledronic acid to
denosumab were similar to those observed in pts who continued with denosumab

Cumulative incidence of positively adjudicated ONJ was 4.7% for denosumab/
denosumab pts when administered for up to ~ 5 yrs and 3.5% for pts who switched from
zoledronic acid to denosumab

No neutralizing anti-denosumab antibodies were detected in either group
Stopeck AT, et al. SABCS 2011. Abstract P3-16-07.
BMA Optimal Interval
[TITLE]
[TITLE]
[TITLE]
[TITLE]
Management Summary
• Patients with bone metastases from breast cancer should be
offered therapy with a bone modifying agent in the absence of
contraindications
• BMA should be used as an adjunct to systemic therapy for the
underlying malignancy
• Appropriate bone modifying agents include subcutaneous
denosumab, IV pamidronate, and IV zoledronic acid
• For patients receiving a bisphosphonate, creatinine clearance must
be monitored and dose adjustments should be made as necessary
• The use of calcium and vitamin D supplements should be explored
in patients receiving bone modifying agents particularly with
denosumab use
• Routine dental care should be performed prior to initiation of a
bone modifying agent
• Continuation of the bone modifying agent for up to 2 years is
certainly acceptable though the optimal duration of therapy
remains unclear
Conclusions
• Bisphosphonates and denosumab are both
effective at
– Preventing SREs and HCM
– Palliating pain from bone mets
– Preventing the development of pain
• 2 distinct choices
– Different toxicity profiles
• Zoledronic acid: flulike symptoms, fevers, bone pains, renal
toxicity
• Denosumab: hypocalcemia
– Subcutaneous vs intravenous administration
Download