Download as doc, pdf, or txt
Download as doc, pdf, or txt
You are on page 1of 32

Onco-Nephrology: The

Pathophysiology and Treatment of


Malignancy-Associated
Hypercalcemia

Mitchell H. Rosner and Alan C. Dalkin


CJASN October 2012, 7 (10) 1722-1729; DOI: https://doi.org/10.2215/CJN.02470312

 Article
 Figures & Data
 Info & Metrics
  PDF

Summary

Hypercalcemia complicates the course of 10%–30% of all patients with malignancies


and can be a sign of very poor prognosis and advanced malignancy. Prompt recognition
of the nonspecific signs and symptoms of hypercalcemia and institution of therapy can
be lifesaving, affording the opportunity to address the underlying etiology. The
mechanisms of malignancy-associated hypercalcemia generally fall into three
categories: humoral hypercalcemia due to secreted factors (such as parathyroid-related
hormone), local osteolysis due to tumor invasion of bone, and absorptive hypercalcemia
due to excess vitamin D produced by malignancies. The mainstays of therapy for
hypercalcemia are aggressive intravenous volume expansion with saline,
bisphosphonate therapy, and perhaps loop diuretics. Adjunctive therapy may include
calcitonin and corticosteroids. In refractory cases, gallium nitrate and perhaps
denosumab are alternatives. In patients presenting with severe AKI, hemodialysis with a
low-calcium bath can be effective. In most cases, therapy normalizes calcium levels and
allows for palliation or curative therapy of the malignancy.

Introduction

Hypercalcemia (defined as a serum calcium level above the upper limit of the normal
reference range of 10.5 mg/dl or 2.5 mmol/L, 40%–45% of which is ionized calcium) can
complicate the course of 10%–30% of all patients with cancer (1–4). The incidence of
hypercalcemia is low at the initial presentation of cancer (1%–5%) but increases
dramatically in patients with advanced disease and a poor prognosis (5). In fact, the
median duration of survival for patients with malignancy-associated hypercalcemia is
only 2–6 months from the onset of hypercalcemia (3,5).
The symptoms of hypercalcemia are nonspecific, often develop gradually, and may
resemble the symptoms of the underlying malignancy and its treatment. These
nonspecific symptoms may lead to a delay in diagnosis, with increased morbidity and
mortality. Clinical presentation is influenced by the rate of onset as well as the severity of
hypercalcemia. Typical symptoms include nausea, vomiting, constipation, abdominal
pain, anorexia, weight loss, bone pain, polyuria, fatigue, and weakness. In patients with
more severe degrees of hypercalcemia (serum calcium > 14 mg/dl), neurologic
symptoms such as confusion and even coma may become more apparent. These
patients require hospitalization and urgent therapeutic intervention (6).

Given the presence of concomitant cachexia and malnutrition in this population, it is also
critical to correct the serum calcium for a low albumin level or measure an ionized serum
calcium level in order to assess the true degree of hypercalcemia (7). Although ionized
calcium is more sensitive than albumin-corrected total calcium in the diagnosis of
hypercalcemia of malignancy, the clinical usefulness of this measurement is unclear. In
at least one study, slightly increased ionized calcium levels did not predict the
development of frank hypercalcemia in patients with solid malignant tumors (8,9). Thus,
evaluation of total calcium concentration in light of changes in bound versus ionized
calcium is essential in order to obtain a complete patient assessment.

Mechanisms of Malignancy-Associated Hypercalcemia

Perturbations of calcium regulation in patients with hypercalcemia of malignancy fall into


three general categories (Figure 1). First, most common are humoral factors secreted in
a paracrine or endocrine fashion (humoral hypercalcemia of malignancy). Second, in
patients with substantial tumor burden metastatic to bone, the local effects of tumor can
directly facilitate bone dissolution and calcium mobilization (local osteolysis). Third, less
commonly, absorptive hypercalcemia results from excess vitamin D activation by the
neoplasm. The cellular basis for the former two mechanisms includes changes in the
activity and balance at the level of the bone-remodeling unit.
 Download figure
 Open in new tab
 Download Powerpoint
Figure 1.
Mechanism of malignancy-associated hypercalcemia. PTHrP, parathyroid hormone–
related hormone.
Bone turnover involves the highly coordinated activity of two distinct types of cells (10).
The osteoblast, or bone-forming cell, is derived from a mesenchymal stem cell and is
closely related to adipocytes, chondrocyte, myocytes, and fibroblasts. In contrast, the
osteoclast, or bone-removing cell, is derived from stems cells populating the monocyte
and macrophage lineage. Although the specifics remain incompletely understood,
communication between osteoblasts and osteoclasts primarily involves the receptor
activator of nuclear factor κ B (RANK) ligand (RANKL) signaling pathway (10–12). RANK
is a receptor expressed on osteoclast precursor cells. The naturally occurring ligand,
RANKL, is produced by osteoblasts and drives proliferation and differentiation of the
osteoclasts into mature, multinucleated units. In addition, the osteoblast cell produces
osteoprotegerin (OPG), a decoy receptor that binds to and inactivates RANKL.

The cellular regulation of RANK, RANKL, and OPG expression is incompletely


understood, but the osteoblast is the focal point for the integration of endocrine and
paracrine signals that alter bone remodeling. For example, osteoblast cells express
estrogen receptors that when occupied with ligand can reduce RANKL and increase
OPG (13). Directly related to the hypercalcemia of malignancy, osteoblasts also express
the cell surface receptor for parathyroid hormone (PTH) and parathyroid hormone–
related hormone (PTHrP), PTH1R (14). Both PTH and PTHrP stimulate PTH1R, which,
in turn, increases osteoblast activity and RANKL signaling to the osteoclast. OPG
expression may decline as well. In sum, PTH/PTHrP signaling results in an increased
bone turnover with a greater increase in bone resorption than formation, resulting in a
net efflux of calcium and hypercalcemia from the bone microenvironment.

Tumors that commonly produce PTHrP include squamous cell carcinomas of the lung,
cervix, and esophagus; certain lymphomas; renal cell carcinoma; and adenocarcinoma
of the breast, prostate, and ovary (15–17). In addition, several case reports have
documented PTHrP secretion in other malignancies. Ectopic secretion of PTH secretion
is less common than that of PTHrP. The number of cases in which tumor-related
production of PTH has been verified are few but include pulmonary, thyroid, ovarian, and
pancreatic malignancies (18–20).

Local osteolysis as the basis for hypercalcemia occurs most frequently in widely
metastatic disease, and the degree of hypercalcemia correlates with the extent of tumor
burden. Local osteolysis is most commonly seen in patients with metastatic breast and
lung cancers (21,22). Probably due to a common pathophysiology, multiple myeloma,
usually extensive in nature, can present with significant areas of osteolysis and
hypercalcemia (23). Underlying the release of calcium from the bone microenvironment
is increased osteoclast activity, probably due to PTHrP and other factors that can
increase resorption. As a result of the paracrine nature of this condition, circulating levels
of PTHrP may be “normal” or only slightly above normal, in contrast to levels in patients
with humoral hypercalcemia due to PTHrP.
Vitamin D has numerous physiologic actions, including the enhancement of calcium and
phosphate absorption from the intestinal tract. Stored vitamin D (25-[OH]D) is 1-
hydroxylated in the kidney to the active compound 1,25-(OH)2D, which in turn
acts via the vitamin D receptor. PTH actively drives the 1-hydroxylase step at the kidney,
and states of hyperparathyroidism, including ectopic PTH from malignancies, are
associated with elevated 1,25-(OH)2D levels. Of note, despite its action through the
common PTH1R, PTHrP is a poor stimulus for 1α-hydroxylation compared with PTH,
and levels of active vitamin D are often low or normal in patients with humoral
hypercalcemia of malignancy (24). Important for the understanding of hypercalcemia, the
1-hydroxylase enzyme is expressed in tissues outside of the kidney, including
macrophages and some neoplastic tissues. Patients with Hodgkin lymphoma and non-
Hodgkin lymphoma, as well as multiple myeloma, have been described as having
vitamin D–mediated hypercalcemia (25). In these individuals, 1,25-(OH)2D levels, along
with calcium levels, are high while PTH is suppressed from the negative feedback to the
parathyroid cells. Similarly, measurements of bone turnover markers are low in vitamin
D–mediated hypercalcemia because the reduction in PTH results in a diminution of
osteoblast and osteoclast activity.

Laboratory Investigation

Given that the malignancy is usually advanced by the time hypercalcemia develops, a
thorough history and physical examination can often lead to the correct diagnosis with
limited laboratory evaluation. Measurement of intact PTH through an immunoradiometric
or immunochemoluminescent assay will confirm a PTH-independent process. In this
case, the PTH levels will be suppressed (often <20 pg/ml) and should prompt further
testing to distinguish among the multiple possible causes in patients with malignancy.
This would include measurement of PTHrP, 1,25-(OH)2D levels, serum and urine protein
electrophoresis, assessment of serum free light chains, and possibly imaging studies
(such as a skeletal survey). Less common causes of hypercalcemia in malignancy
include drugs (e.g., retinoic acid [26]) and, rarely, parathyroid carcinomas producing
excessive PTH.

Therapy for Malignancy-Associated Hypercalcemia

Patients presenting with acute and symptomatic hypercalcemia require prompt therapy
that rapidly reduces the serum calcium level, restores the GFR, and leads to longer-term
normalization of the serum calcium level. An algorithmic approach is shown in Figure 2.
 Download figure
 Open in new tab
 Download Powerpoint
Figure 2.
Treatment algorithm for malignancy-associated hypercalcemia. IV, intravenous.

Intravenous Fluid and Diuretics

Hypercalcemia leads to a decrease in the GFR through a combination of the natriuretic


effects of high serum levels of calcium and renal vasoconstriction (27). In fact, polyuria is
the most common renal manifestation of hypercalcemia, and it is thought that the
impaired urinary concentrating ability is due to activation of the calcium-sensing receptor
in the thick ascending limb of the loop of Henle (28). Activation of the calcium-sensing
receptor leads to decreased resorption of sodium and chloride in the loop of Henle,
resulting in decreased countercurrent multiplication and decreased ability to concentrate
the urine. In addition, activation of the calcium-sensing receptor in the collecting duct
blunts the response of this segment to the actions of arginine vasopressin (29). Thus,
patients presenting with hypercalcemia are usually profoundly volume depleted.
Furthermore, patients usually will present with poor oral intake and may have had
nausea and vomiting that further worsen the volume depletion. The decrease in GFR
leads to impaired calcium clearance and, thus, restoration of extracellular fluid volume,
and GFR is a key goal of therapy.

Thus, initial therapy should begin with intravenous (IV) volume expansion with 0.9%
saline at 200–500 ml per hour. Volume expansion will lead to an increase in GFR and an
increase in urine calcium excretion. The goal should be establishing an adequate urine
output (>75 ml per hour). In milder cases of hypercalcemia, this therapy can be sufficient
to restore normal calcium levels (30). However, the effect of repleting the extracellular
volume is transient, and other therapies are required. Careful monitoring of the patient’s
vital signs and laboratory values must occur during the acute phase of volume repletion,
along with vigilance for evidence of volume overload. Hypernatremia may be seen
during this phase of therapy because of hypercalcemia-induced nephrogenic diabetes
insipidus and may require changing from isotonic to hypotonic fluid therapy (31).
A common practice is to add a loop diuretic to saline therapy in an effort to increase
urine calcium excretion (forced saline diuresis). LeGrand and colleagues reviewed the
evidence-base for this approach (32). By inhibiting the sodium-potassium-chloride
(NKCC2) transporter in the thick ascending limb of the loop of Henle, a loop diuretic
decreases the lumen positive charge that normally drives calcium reabsorption; thus, a
loop diuretic increases urine calcium excretion and theoretically should be useful in
therapy for hypercalcemia (33,34). However, LeGrand et al. could find only nine articles
that documented the use of furosemide in the treatment of hypercalcemia, the last of
which was published in 1983. These studies comprised a total of 37 patients. The
average dosage of furosemide was 1120 mg given over 24 hours, and serum calcium
normalized in 14 of 39 episodes of hypercalcemia; however, normalization was rapid
(<12 hours) in only two cases. Lower dosages of furosemide (40–60 mg/d) did not
achieve normalization of the serum calcium. Most important, monitoring in these patients
was intensive and included aggressive replacement of hourly urine output losses, and
secondary electrolyte disorders, such as hypernatremia, hypophosphatemia,
hypomagnesemia, and metabolic acidosis, were seen. Thus, the routine use of loop
diuretics in the therapy for hypercalcemia cannot be recommended, and their use should
be restricted to patients who develop fluid overload while receiving aggressive volume
resuscitation.

Bisphosphonates

High-potency IV bisphosphonates are effective agents for the treatment of malignancy-


associated hypercalcemia. The three high-potency bisphosphonates are pamidronate,
zoledronate, and ibandronate (dosing guidelines are shown in Table 1). Because the IV
bisphosphonates are excreted unchanged by the kidneys through glomerular filtration,
their dosing must be adjusted for impaired renal function to avoid toxicity (35,36).
Mechanistically, bisphosphonates decrease bone resorption through extracellular and
intracellular mechanisms (37–41). In the extracellular space, bisphosphonates bind to
calcium phosphate and stabilize the bone matrix, while intracellularly these agents inhibit
osteoclast activity, in part through inhibition of the mevalonate pathway (37–39).
Furthermore, bisphosphonates impair cellular adenosine triphosphate–dependent
metabolic pathways, disrupt the osteoclast cytoskeleton, and induce apoptosis (40,41).

 View inline
 View popup
Table 1.
Dosing guidelines for intravenous bisphosphonates in the treatment of acute hypercalcemia
Randomized clinical trials with pamidronate, zoledronate, and ibandronate have all
shown efficacy in the treatment of hypercalcemia of malignancy (42–44). However, the
three bisphosphonates differ in terms of their potency and risk for toxicity. Nussbaum et
al. demonstrated a dose response using pamidronate, with 40%, 61%, and 100% of
patients achieving normocalcemia with 30, 60, and 90 mg, respectively (45). Similar
efficacy of pamidronate is seen over a dosing span ranging from 2 to 24 hours (46).
Thus, a practical approach is to administer pamidronate, 60–90 mg, over 2–6 hours, with
the longer dosing time reserved for patients with lower GFRs and the higher doses for
patients with more severe hypercalcemia. The efficacy of pamidronate (and probably
other bisphosphonates) is influenced by the mechanism of hypercalcemia (more
effective in cases of hypercalcemia associated with bone metastases); the level of
hypercalcemia (higher doses required to treat more severe hypercalcemia); and, in
patients with humoral hypercalcemia, the level of PTHrP (higher levels of PTHrP are
associated with a lower response rate) (47,48). The fact that patients with higher levels
of PTHrP show less of a hypocalcemic response to bisphosphonates is probably due to
the continued actions of PTHrP on the kidney to decrease calcium excretion, an effect
that is not modulated by bisphosphonates.

Zoledronate is 100 times more potent than pamidronate, and two parallel randomized
trials evaluated the efficacy of 4 or 8 mg of zoledronate versus 90 mg of pamidronate in
restoring normocalcemia by day 10 after treatment (49). Both doses of zoledronate were
superior to pamidronate, with serum calcium normalizing by day 10 in 88%, 87%, and
70% of patients receiving 4 and 8 mg of zoledronate and 90 mg of pamidronate,
respectively (49). Moreover, the mean duration of complete response was 32, 43, and
18 days, respectively, in the three treatment groups (49).

Ibandronate is also effective in treating malignancy-associated hypercalcemia (44,50).


Doses of 2, 4, and 6 mg had success rates of 50%, 76%, and 77%, respectively (44).
Compared with pamidronate, the duration of response is longer with ibandronate (50).
However, ibandronate is not approved by the Food and Drug Administration (FDA) to
treat malignancy-associated hypercalcemia.

The most common side effect associated with these agents is infusion-related fever,
which is usually mild (51). However, bisphosphonate-associated nephrotoxicity affecting
both glomerular and tubular structures has been described in both case series and
randomized clinical trials (35). In terms of glomerular toxicity, bisphosphonates (most
commonly, pamidronate and, only rarely, zoledronate) have been associated with
collapsing focal segmental glomerulosclerosis (FSGS), noncollapsing FSGS, and
minimal-change disease in several case series (52–56). The most common lesion is
collapsing FSGS, which is seen with high-dose IV pamidronate often in patients with
underlying multiple myeloma and usually after repeated doses (35). In clinical trials,
glomerular toxicity of the bisphosphonates was not reported (35). In some cases, the
nephrotic syndrome resolves after cessation of the drug, but some patients may
progress to ESRD, especially if the glomerular lesion is collapsing FSGS.

Tubular injury (nephrotoxic acute tubular necrosis) associated with bisphosphonates


(mainly, zoledronate) has also been described in both case reports and clinical trials
(57–59). In 2003, the FDA Adverse Event Reporting System experience with
bisphosphonates and AKI revealed 72 cases associated with IV zoledronate (60). In this
cohort, AKI developed approximately 2 months after initiation of therapy and after a
mean of 2.4 doses of zoledronate. Risk factors for AKI included advanced cancer,
previous bisphosphonate therapy, and use of nonsteroidal anti-inflammatory agents.
Most patients did not fully recover to baseline renal function, and 37.5% of patients
required dialysis (60).

On the basis of the renal toxicity of bisphosphonates, the American Society of Clinical
Oncology developed dosing and monitoring guidelines (Table 1) and recommended that
zoledronate be avoided in patients with a creatinine clearance <30 ml/min (61).
Furthermore, serum creatinine should be monitored before each dose of bisphosphonate
and the drug withheld in patients with worsening of renal function. Urine albumin
excretion should be monitored at 3- to 6-month intervals, and the drug should be held if
albuminuria develops (61).

Interestingly, ibandronate appears to have minimal to no renal toxicity, even in patients


with underlying CKD (62–64). However, more experience with this drug for the treatment
of malignancy-associated hypercalcemia is needed before firm conclusions on its renal
safety can be made.

An increasingly recognized complication of bisphosphonate therapy is osteonecrosis of


the jaw (ONJ) (65,66). The precise etiology is not known, and the most common
precipitating factors include dental extraction, long-term sequential use of IV
pamidronate or zoledronate, smoking, and poor dental health (67). The absolute risk for
ONJ is unknown, although it is estimated to occur in <1:10,000 treated individuals and
may be seen even less often in patients receiving shorter-term treatment. Diagnosis is
based on clinical criteria that include pain and evidence of local infection. Management
is largely supportive, with occasional need for surgical debridement. The condition can
be prevented through good dental hygiene and avoidance of invasive dental procedures
during bisphosphonate therapy (68).

Gallium Nitrate

Gallium nitrate is approved for the treatment of cancer-related hypercalcemia. The


cellular basis for this action is unknown but appears to involve a reduction in osteoclast
activity and bone remodeling (69–72). Clinically, gallium nitrate is administered by a
continuous infusion at a dosage of 200 mg/m2 per day over 5 consecutive days. Reports
comparing gallium to bisphosphonates (73,74) and calcitonin (75) have shown equal or
perhaps even superior efficacy in decreasing calcium levels. Adverse effects from
treatment include hypocalcemia and hypophosphatemia, the latter being observed in
nearly 80% of patients. In addition, renal impairment has been reported, and hence
gallium nitrate is not recommended for use when the serum creatinine concentration
exceeds 2.5 mg/dl. Adequate fluid resuscitation is essential before initiation of treatment
with gallium, and BUN and creatinine must be followed closely during treatment. Patients
receiving gallium have reported nausea, vomiting, lethargy, altered mental status, and
both diarrhea and constipation.

Calcitonin

Calcitonin is a peptide hormone secreted by the parafollicular C cells of the thyroid


gland. It inhibits osteoclast bone resorption and increases urinary calcium excretion.
Synthetic calcitonin (derived from salmon) is administered as 4–8 U/kg intramuscularly
or subcutaneously every 6–8 hours. Calcitonin has a rapid onset of effect, leading to
reductions in serum calcium within 2–6 hours after dosing. The calcium-lowering effects
of calcitonin are modest and transient, with a mean duration of effect of 2–4 days (76).
Furthermore, repeated administration of calcitonin results in downregulation of the
calcitonin receptors on osteoclasts and dimunition in effect (tachyphylaxis) (77). Overall,
its efficacy is poor, inducing normocalcemia in just one third of patients (78). Calcitonin’s
major role may be as an adjunctive agent with bisphosphonates in the treatment of
severe, symptomatic hypercalcemia when calcium levels must be reduced rapidly (79).
Side effects are minimal; nausea, vomiting, and injection site pain are the most common.
Overall, there is little evidence that calcitonin use has better efficacy over volume
repletion and bisphosphonate therapy alone.

Corticosteroids

Corticosteroids are effective agents in the treatment of hypercalcemia associated with


malignancies that overproduce calcitriol. This is due to their effect in inhibiting 1α-
hydroxylase conversion of 25-hydroxyvitamin D to calcitriol. Typically, these patients
have underlying Hodgkin or non-Hodgkin lymphoma (80). The efficacy of corticosteroids
is based on case reports and thus the dosing guidelines are uncertain; however, a
reasonable regimen is IV hydrocortisone, 200–300 mg/d, for 3–5 days (79). Typically,
calcium levels are slow to decrease with corticosteroid treatment alone. Patients who do
respond with normalization of their serum calcium can be transitioned to maintenance
oral prednisone, 10–30 mg/d (81).

Hemodialysis

For patients with acute hypercalcemia and significant AKI (especially in the setting of
oliguria), saline-induced diuresis may not be feasible and may lead to volume overload.
In these circumstances, hemodialysis using a very-low-calcium dialysate (≤1 mmol/L) is
an option (82). For refractory cases of hypercalcemia, several case reports describe the
use of citrate anticoagulation to chelate calcium along with continuous venovenous
hemodiafiltration (83,84).

Denosumab

Given the important role RANKL in the pathogenesis of bone metastases and
secondarily in leading to hypercalcemia, inhibition of this pathway is an attractive means
of potentially treating forms of malignancy-associated hypercalcemia due to bone
calcium release. Denosumab is a fully human monoclonal antibody that binds to RANKL
and inhibits osteoclast maturation, activation, and function (85). It is FDA approved for
preventing skeletal-related events in patients with solid malignancy, as well preventing
bone fractures in postmenopausal women with osteoporosis. In clinical trials,
denosumab has shown efficacy in preventing skeletal-related events (such as bone
metastases) in patients with prostate, breast, and other solid-organ cancers, as well as
multiple myeloma (85). Denosumab is well tolerated, with arthralgias as the most
common side effect (86). Because RANKL is expressed on B and T cells, there was
some concern that denosumab would increase the risk for serious infections. The FDA
has reported only a slightly increased incidence of serious infections (4.1% for
denosumab and 3.3% for placebo), with no increase in opportunistic infections seen
(86). Of note, denosumab has been associated with a similar incidence of osteonecrosis
of the jaw as zoledronic acid (87). Denosumab is not cleared through renal mechanisms
and does not require dosing adjustments for renal impairment.

In clinical trials in patients with malignancies, the most common laboratory abnormality
was hypocalcemia due to inhibition of bone resorption (88–90). This is not surprising
given the mechanism of action of denosumab and leads to the obvious question of
whether this drug can be used to treat malignancy-associated hypercalcemia. Hu et
al. studied patients with bisphosphonate-refractory malignancy-associated
hypercalcemia (91). These patients had solid malignancies or myeloma and elevated
corrected serum calcium levels (>12.5 mg/dl) despite recent IV bisphosphonate therapy.
Patients received denosumab, 120 mg subcutaneously, every 4 weeks, with additional
loading doses of 120 mg on days 8 and 15 of the first month. The primary end point was
reaching a response defined as a corrected serum calcium level ≤11.5 mg/dl within the
first 10 days after denosumab dosing. An interim analysis of this study (n=15) revealed
that 80% of patients achieved a response by day 10, with a median response time of 8
days and a response duration of 28 days. Furthermore, an ad hoc pooled analysis of the
clinical trials with denosumab (88–90) revealed that compared with zoledronic acid,
denosumab significantly delayed the time to first on-study episodes of hypercalcemia
(92). More work will be required to understand the role of denosumab in the treatment of
malignancy-associated hypercalcemia, but it holds much promise as a safe, more
convenient (subcutaneous versus IV) therapy.

Summary

Patients with underlying malignancy frequently encounter hypercalcemia. Indeed,


hypercalcemia carries a poor prognosis. Multiple cellular mechanisms underlie the
increase in calcium concentration, and a proper evaluation to elucidate the cause is
essential to guide treatment. The mainstay of initial management is aggressive fluid
resuscitation, with the addition of loop diuretics if volume overload is present. In addition,
antiresorptive agents, primarily the bisphosphonates, can aid in the treatment of these
patients. Although the physician must bear in mind potential pitfalls with the use of these
medications, prompt and assertive treatment of hypercalcemia in patients with cancer
can be successful and life-saving.

Disclosures
None.

Footnotes


Published online ahead of print. Publication date available at www.cjasn.org.


 Copyright © 2012 by the American Society of Nephrology

References

. 1.↵

1. Sargent JTS, 

2. Smith OP

: Haematological emergencies managing hypercalcaemia in adults and children with haematological disorders. Br J
Haematol 149: 465–477, 2010
CrossRefPubMedGoogle Scholar
. 2.

1. Stewart AF

: Clinical practice. Hypercalcemia associated with cancer. N Engl J Med 352: 373–379, 2005


CrossRefPubMedGoogle Scholar
. 3.↵

1. Ralston SH, 

2. Gallacher SJ, 

3. Patel U, 

4. Campbell J, 

5. Boyle IT

: Cancer-associated hypercalcemia: morbidity and mortality. Clinical experience in 126 treated patients. Ann Intern
Med 112: 499–504, 1990
CrossRefPubMedGoogle Scholar
. 4.↵

1. Burt ME, 

2. Brennan MF

: Incidence of hypercalcemia and malignant neoplasm. Arch Surg 115: 704–707, 1980


CrossRefPubMedGoogle Scholar
. 5.↵

1. Vassilopoulou-Sellin R, 

2. Newman BM, 

3. Taylor SH, 

4. Guinee VF

: Incidence of hypercalcemia in patients with malignancy referred to a comprehensive cancer


center. Cancer 71: 1309–1312, 1993
CrossRefPubMedGoogle Scholar
. 6.↵

1. Edelson GW, 

2. Kleerekoper M

: Hypercalcemic crisis. Med Clin North Am 79: 79–92, 1995


PubMedGoogle Scholar
. 7.↵

1. Labriola L, 

2. Wallemacq P, 

3. Gulbis B, 

4. Jadoul M

: The impact of the assay for measuring albumin on corrected (‘adjusted’) calcium concentrations. Nephrol Dial
Transplant 24: 1834–1838, 2009
Abstract/FREE Full TextGoogle Scholar
. 8.↵

1. Ijaz A, 

2. Mehmood T, 

3. Qureshi AH, 

4. Anwar M, 

5. Dilawar M, 

6. Hussain I, 

7. Khan FA, 

8. Khan DA, 

9. Hussain S, 

10. Khan IA
: Estimation of ionized calcium, total calcium and albumin corrected calcium for the diagnosis of hypercalcaemia of
malignancy. J Coll Physicians Surg Pak 16: 49–52, 2006
PubMedGoogle Scholar
. 9.↵

1. Riancho JA, 

2. Arjona R, 

3. Sanz J, 
4. Olmos JM, 

5. Valle R, 

6. Barceló JR, 

7. González-Macías J

: Is the routine measurement of ionized calcium worthwhile in patients with cancer? Postgrad Med J 67: 350–
353, 1991
Abstract/FREE Full TextGoogle Scholar
. 10.↵

1. Mellis DJ, 

2. Itzstein C, 

3. Helfrich MH, 

4. Crockett JC

: The skeleton: a multi-functional complex organ: the role of key signalling pathways in osteoclast differentiation and
in bone resorption. J Endocrinol 211: 131–143, 2011
Abstract/FREE Full TextGoogle Scholar
. 11.

1. Hsu H, 

2. Lacey DL, 

3. Dunstan CR, 

4. Solovyev I, 

5. Colombero A, 

6. Timms E, 

7. Tan HL, 

8. Elliott G, 

9. Kelley MJ, 

10. Sarosi I, 
11. Wang L, 
12. Xia XZ, 
13. Elliott R, 
14. Chiu L, 
15. Black T, 
16. Scully S, 
17. Capparelli C, 
18. Morony S, 
19. Shimamoto G, 
20. Bass MB, 
21. Boyle WJ
: Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by
osteoprotegerin ligand. Proc Natl Acad Sci USA 96: 3540–3545, 1999
Abstract/FREE Full TextGoogle Scholar
. 12.↵

1. Hofbauer LC, 

2. Schoppet M

: Clinical implications of the osteoprotegerin/RANKL/RANK system for bone and vascular


diseases. JAMA 292: 490–495, 2004
CrossRefPubMedGoogle Scholar
. 13.↵

1. Eghbali-Fatourechi G, 

2. Khosla S, 

3. Sanyal A, 

4. Boyle WJ, 

5. Lacey DL, 

6. Riggs BL

: Role of RANK ligand in mediating increased bone resorption in early postmenopausal women. J Clin
Invest 111: 1221–1230, 2003
CrossRefPubMedGoogle Scholar
. 14.↵

1. Abou-Samra AB, 

2. Jüppner H, 

3. Force T, 

4. Freeman MW, 

5. Kong XF, 

6. Schipani E, 

7. Urena P, 

8. Richards J, 

9. Bonventre JV, 

10. Potts JT Jr., 
11. et al
: Expression cloning of a common receptor for parathyroid hormone and parathyroid hormone-related peptide from
rat osteoblast-like cells: A single receptor stimulates intracellular accumulation of both cAMP and inositol
trisphosphates and increases intracellular free calcium. Proc Natl Acad Sci USA 89: 2732–2736, 1992
Abstract/FREE Full TextGoogle Scholar
. 15.↵

1. Sone S, 

2. Yano S

: Molecular pathogenesis and its therapeutic modalities of lung cancer metastasis to bone. Cancer Metastasis
Rev 26: 685–689, 2007
CrossRefPubMedGoogle Scholar
. 16.
1. Grill V, 

2. Ho P, 

3. Body JJ, 

4. Johanson N, 

5. Lee SC, 

6. Kukreja SC, 

7. Moseley JM, 

8. Martin TJ

: Parathyroid hormone-related protein: Elevated levels in both humoral hypercalcemia of malignancy and
hypercalcemia complicating metastatic breast cancer. J Clin Endocrinol Metab 73: 1309–1315, 1991
CrossRefPubMedGoogle Scholar
. 17.↵

1. Sourbier C, 

2. Massfelder T

: Parathyroid hormone-related protein in human renal cell carcinoma. Cancer Lett 240: 170–182, 2006


CrossRefPubMedGoogle Scholar
. 18.↵

1. Yoshimoto K, 

2. Yamasaki R, 

3. Sakai H, 

4. Tezuka U, 

5. Takahashi M, 

6. Iizuka M, 

7. Sekiya T, 

8. Saito S

: Ectopic production of parathyroid hormone by small cell lung cancer in a patient with hypercalcemia. J Clin
Endocrinol Metab 68: 976–981, 1989
CrossRefPubMedGoogle Scholar
. 19.

1. Iguchi H, 

2. Miyagi C, 

3. Tomita K, 

4. Kawauchi S, 

5. Nozuka Y, 

6. Tsuneyoshi M, 

7. Wakasugi H

: Hypercalcemia caused by ectopic production of parathyroid hormone in a patient with papillary adenocarcinoma of
the thyroid gland. J Clin Endocrinol Metab 83: 2653–2657, 1998
CrossRefPubMedGoogle Scholar
. 20.↵
1. Nussbaum SR, 

2. Gaz RD, 

3. Arnold A

: Hypercalcemia and ectopic secretion of parathyroid hormone by an ovarian carcinoma with rearrangement of the
gene for parathyroid hormone. N Engl J Med 323: 1324–1328, 1990
PubMedGoogle Scholar
. 21.↵

1. Chen YC, 

2. Sosnoski DM, 

3. Mastro AM

: Breast cancer metastasis to the bone: Mechanisms of bone loss. Breast Cancer Res 12: 215–226, 2010
CrossRefPubMedGoogle Scholar
. 22.↵

1. Clines GA

: Mechanisms and treatment of hypercalcemia of malignancy. Curr Opin Endocrinol Diabetes Obes 18: 339–


346, 2011
CrossRefPubMedGoogle Scholar
. 23.↵

1. Abildgaard N, 

2. Brixen K, 

3. Eriksen EF, 

4. Kristensen JE, 

5. Nielsen JL, 

6. Heickendorff L

: Sequential analysis of biochemical markers of bone resorption and bone densitometry in multiple
myeloma. Haematologica 89: 567–577, 2004
Abstract/FREE Full TextGoogle Scholar
. 24.↵

1. Schilling T, 

2. Pecherstorfer M, 

3. Blind E, 

4. Leidig G, 

5. Ziegler R, 

6. Raue F

: Parathyroid hormone-related protein (PTHrP) does not regulate 1,25-dihydroxyvitamin D serum levels in
hypercalcemia of malignancy. J Clin Endocrinol Metab 76: 801–803, 1993
CrossRefPubMedGoogle Scholar
. 25.↵

1. Seymour JF, 

2. Gagel RF

: Calcitriol: the major humoral mediator of hypercalcemia in Hodgkin’s disease and non-Hodgkin’s
lymphomas. Blood 82: 1383–1394, 1993
FREE Full TextGoogle Scholar
. 26.↵

1. Hatake K, 

2. Uwai M, 

3. Ohtsuki T, 

4. Tomizuka H, 

5. Izumi T, 

6. Yoshida M, 

7. Miura Y

: Rare but important adverse effects of all-trans retinoic acid in acute promyelocytic leukemia and their
management. Int J Hematol 66: 13–19, 1997
CrossRefPubMedGoogle Scholar
. 27.↵

1. Castelli I, 

2. Steiner LA, 

3. Kaufmann MA, 

4. Drop LJ

: Renovascular responses to high and low perfusate calcium steady-state experiments in the isolated perfused rat
kidney with baseline vascular tone. J Surg Res 61: 51–57, 1996
CrossRefPubMedGoogle Scholar
. 28.↵

1. Hebert SC, 

2. Brown EM

: The scent of an ion: Calcium-sensing and its roles in health and disease. Curr Opin Nephrol Hypertens 5: 45–
53, 1996
CrossRefPubMedGoogle Scholar
. 29.↵

1. Khanna A

: Acquired nephrogenic diabetes insipidus. Semin Nephrol 26: 244–248, 2006


CrossRefPubMedGoogle Scholar
. 30.↵

1. Sleeboom HP, 

2. bijvoet OLM, 

3. Van Oosterom AT, 

4. Gleed JH, 

5. O'Riordan JL

: Comparison of intravenous (3-amino-1-hydroxypropylidene)-1-1-bisphosphonate and volume repletion in tumour-


induced hypercalcemia. Lancet 2: 239–243, 1983
PubMedGoogle Scholar
. 31.↵

1. Kacprowicz RF, 
2. Lloyd JD

: Electrolyte complications of malignancy. Hematol Oncol Clin North Am 24: 553–565, 2010


CrossRefPubMedGoogle Scholar
. 32.↵

1. LeGrand SB, 

2. Leskuski D, 

3. Zama I

: Narrative review: Furosemide for hypercalcemia: An unproven yet common practice. Ann Intern Med 149: 259–
263, 2008
CrossRefPubMedGoogle Scholar
. 33.↵

1. Nielsen SP, 

2. Andersen O, 

3. Steven KE

: Magnesium and calcium metabolism during prolonged furosemide (Lasix) administration to normal rats. Acta
Pharmacol Toxicol (Copenh) 27: 469–479, 1969
PubMedGoogle Scholar
. 34.↵

1. Toft H, 

2. Roin J

: Effect of frusemide administration on calcium excretion. BMJ 1: 437–438, 1971


Abstract/FREE Full TextGoogle Scholar
. 35.↵

1. Perazella MA, 

2. Markowitz GS

: Bisphosphonate nephrotoxicity. Kidney Int 74: 1385–1393, 2008


CrossRefPubMedGoogle Scholar
. 36.↵

1. Lin JH

: Bisphosphonates: A review of their pharmacokinetic properties. Bone 18: 75–85, 1996


CrossRefPubMedGoogle Scholar
. 37.↵

1. Russell RGG, 

2. Rogers MJ

: Bisphosphonates: From the laboratory to the clinic and back again. Bone 25: 97–106, 1999


CrossRefPubMedGoogle Scholar
. 38.

1. Amin D, 

2. Cornell SA, 

3. Gustafson SK, 

4. Needle SJ, 

5. Ullrich JW, 
6. Bilder GE, 

7. Perrone MH

: Bisphosphonates used for the treatment of bone disorders inhibit squalene synthase and cholesterol
biosynthesis. J Lipid Res 33: 1657–1663, 1992
AbstractGoogle Scholar
. 39.↵

1. Rogers MJ, 

2. Gordon S, 

3. Benford HL, 

4. Coxon FP, 

5. Luckman SP, 

6. Monkkonen J, 

7. Frith JC

: Cellular and molecular mechanisms of action of bisphosphonates. Cancer 88[Suppl]: 2961–2978, 2000


CrossRefPubMedGoogle Scholar
. 40.↵

1. Hiroi-Furuya E, 

2. Kameda T, 

3. Hiura K, 

4. Mano H, 

5. Miyazawa K, 

6. Nakamaru Y, 

7. Watanabe-Mano M, 

8. Okuda N, 

9. Shimada J, 

10. Yamamoto Y, 
11. Hakeda Y, 
12. Kumegawa M
: Etidronate (EHDP) inhibits osteoclastic-bone resorption, promotes apoptosis and disrupts actin rings in isolate-
mature osteoclasts. Calcif Tissue Int 64: 219–223, 1999
CrossRefPubMedGoogle Scholar
. 41.↵

1. Mackie PS, 

2. Fisher JL, 

3. Zhou H, 

4. Choong PF

: Bisphosphonates regulate cell growth and gene expression in the UMR 106-01 clonal rat osteosarcoma cell
line. Br J Cancer 84: 951–958, 2001
CrossRefPubMedGoogle Scholar
. 42.↵
1. Purohit OP, 

2. Radstone CR, 

3. Anthony C, 

4. Kanis JA, 

5. Coleman RE

: A randomised double-blind comparison of intravenous pamidronate and clodronate in the hypercalcaemia of
malignancy. Br J Cancer 72: 1289–1293, 1995
CrossRefPubMedGoogle Scholar
. 43.

1. Body JJ, 

2. Lortholary A, 

3. Romieu G, 

4. Vigneron AM, 

5. Ford J

: A dose-finding study of zoledronate in hypercalcemic cancer patients. J Bone Miner Res 14: 1557–1561, 1999
CrossRefPubMedGoogle Scholar
. 44.↵

1. Pecherstorfer M, 

2. Herrmann Z, 

3. Body JJ, 

4. Manegold C, 

5. Degardin M, 

6. Clemens MR, 

7. Thürlimann B, 

8. Tubiana-Hulin M, 

9. Steinhauer EU, 

10. van Eijkeren M, 


11. Huss HJ, 
12. Thiébaud D
: Randomized phase II trial comparing different doses of the bisphosphonate ibandronate in the treatment of
hypercalcemia of malignancy. J Clin Oncol 14: 268–276, 1996
AbstractGoogle Scholar
. 45.↵

1. Nussbaum SR, 

2. Younger J, 

3. Vandepol CJ, 

4. Gagel RF, 

5. Zubler MA, 
6. Chapman R, 

7. Henderson IC, 

8. Mallette LE

: Single-dose intravenous therapy with pamidronate for the treatment of hypercalcemia of malignancy: Comparison
of 30-, 60-, and 90-mg dosages. Am J Med 95: 297–304, 1993
CrossRefPubMedGoogle Scholar
. 46.↵

1. Dodwell DJ, 

2. Howell A, 

3. Morton AR, 

4. Daley-Yates PT, 

5. Hoggarth CR

: Infusion rate and pharmacokinetics of intravenous pamidronate in the treatment of tumour-induced


hypercalcaemia. Postgrad Med J 68: 434–439, 1992
Abstract/FREE Full TextGoogle Scholar
. 47.↵

1. Gurney H, 

2. Grill V, 

3. Martin TJ

: Parathyroid hormone-related protein and response to pamidronate in tumor-induced


hypercalcemia. Lancet 341: 1611–1613, 1993
CrossRefPubMedGoogle Scholar
. 48.↵

1. Walls J, 

2. Ratcliffe WA, 

3. Howell A, 

4. Bundred NJ

: Response to intravenous bisphosphonate therapy in hypercalcaemic patients with and without bone metastases:
the role of parathyroid hormone-related protein. Br J Cancer 70: 169–172, 1994
PubMedGoogle Scholar
. 49.↵

1. Major P, 

2. Lortholary A, 

3. Hon J, 

4. Abdi E, 

5. Mills G, 

6. Menssen HD, 

7. Yunus F, 

8. Bell R, 

9. Body J, 
10. Quebe-Fehling E, 
11. Seaman J
: Zoledronic acid is superior to pamidronate in the treatment of hypercalcemia of malignancy: A pooled analysis of
two randomized, controlled clinical trials. J Clin Oncol 19: 558–567, 2001
Abstract/FREE Full TextGoogle Scholar
. 50.↵

1. Pecherstorfer M, 

2. Steinhauer EU, 

3. Rizzoli R, 

4. Wetterwald M, 

5. Bergström B

: Efficacy and safety of ibandronate in the treatment of hypercalcemia of malignancy: A randomized multicentric
comparison to pamidronate. Support Care Cancer 11: 539–547, 2003
CrossRefPubMedGoogle Scholar
. 51.↵

1. Zojer N, 

2. Keck AV, 

3. Pecherstorfer M

: Comparative tolerability of drug therapies for hypercalcaemia of malignancy. Drug Saf 21: 389–406, 1999


CrossRefPubMedGoogle Scholar
. 52.↵

1. Markowitz GS, 

2. Appel GB, 

3. Fine PL, 

4. Fenves AZ, 

5. Loon NR, 

6. Jagannath S, 

7. Kuhn JA, 

8. Dratch AD, 

9. D’Agati VD

: Collapsing focal segmental glomerulosclerosis following treatment with high-dose pamidronate. J Am Soc
Nephrol 12: 1164–1172, 2001
Abstract/FREE Full TextGoogle Scholar
. 53.

1. Barri YM, 

2. Munshi NC, 

3. Sukumalchantra S, 

4. Abulezz SR, 

5. Bonsib SM, 

6. Wallach J, 
7. Walker PD

: Podocyte injury associated glomerulopathies induced by pamidronate. Kidney Int 65: 634–641, 2004


CrossRefPubMedGoogle Scholar
. 54.

1. Desikan R, 

2. Veksler Y, 

3. Raza S, 

4. Stokes B, 

5. Sabir T, 

6. Li ZJ, 

7. Jagannath S

: Nephrotic proteinuria associated with high-dose pamidronate in multiple myeloma. Br J Haematol 119: 496–


499, 2002
CrossRefPubMedGoogle Scholar
. 55.

1. Markowitz GS, 

2. Fine PL, 

3. D’agati VD

: Nephrotic syndrome after treatment with pamidronate. Am J Kidney Dis 39: 1118–1122, 2002


CrossRefPubMedGoogle Scholar
. 56.↵

1. Bodmer M, 

2. Amico P, 

3. Mihatsch MJ, 

4. Haschke M, 

5. Kummer O, 

6. Krähenbühl S, 

7. Mayr M

: Focal segmental glomerulosclerosis associated with long-term treatment with zoledronate in a myeloma
patient. Dial Transplant 22: 2366–2370, 2007
CrossRefGoogle Scholar
. 57.↵

1. Markowitz GS, 

2. Fine PL, 

3. Stack JI, 

4. Kunis CL, 

5. Radhakrishnan J, 

6. Palecki W, 

7. Park J, 
8. Nasr SH, 

9. Hoh S, 

10. Siegel DS, 
11. D’Agati VD
: Toxic acute tubular necrosis following treatment with zoledronate (Zometa). Kidney Int 64: 281–289, 2003
CrossRefPubMedGoogle Scholar
. 58.

1. Rosen LS, 

2. Gordon D, 

3. Kaminski M, 

4. Howell A, 

5. Belch A, 

6. Mackey J, 

7. Apffelstaedt J, 

8. Hussein M, 

9. Coleman RE, 

10. Reitsma DJ, 
11. Seaman JJ, 
12. Chen BL, 
13. Ambros Y
: Zoledronic acid versus pamidronate in the treatment of skeletal metastases in patients with breast cancer or
osteolytic lesions of multiple myeloma: A phase III, double-blind, comparative trial. Cancer J 7: 377–387, 2001
PubMedGoogle Scholar
. 59.↵

1. Rosen LS, 

2. Gordon D, 

3. Tchekmedyian S, 

4. Yanagihara R, 

5. Hirsh V, 

6. Krzakowski M, 

7. Pawlicki M, 

8. de Souza P, 

9. Zheng M, 

10. Urbanowitz G, 
11. Reitsma D, 
12. Seaman JJ
: Zoledronic acid versus placebo in the treatment of skeletal metastases in patients with lung cancer and other solid
tumors: A phase III, double-blind, randomized trial—the Zoledronic Acid Lung Cancer and Other Solid Tumors Study
Group. J Clin Oncol 21: 3150–3157, 2003
Abstract/FREE Full TextGoogle Scholar
. 60.↵

1. Chang JT, 

2. Green L, 

3. Beitz J

: Renal failure with the use of zoledronic acid. N Engl J Med 349: 1676–1679, discussion 1676–1679, 2003
CrossRefPubMedGoogle Scholar
. 61.↵

1. Kyle RA, 

2. Yee GC, 

3. Somerfield MR, 

4. Flynn PJ, 

5. Halabi S, 

6. Jagannath S, 

7. Orlowski RZ, 

8. Roodman DG, 

9. Twilde P, 

10. Anderson K
; American Society of Clinical Oncology: American Society of Clinical Oncology 2007 clinical practice guideline
update on the role of bisphosphonates in multiple myeloma. J Clin Oncol 25: 2464–2472, 2007
Abstract/FREE Full TextGoogle Scholar
. 62.↵

1. Heidenreich A, 

2. Elert A, 

3. Hofmann R

: Ibandronate in the treatment of prostate cancer associated painful osseous metastases. Prostate Cancer Prostatic
Dis 5: 231–235, 2002
CrossRefPubMedGoogle Scholar
. 63.

1. Body JJ, 

2. Diel IJ, 

3. Tripathy D, 

4. Bergstrom B

: Intravenous ibandronate does not affect time to renal function deterioration in patients with skeletal metastases
from breast cancer: Phase III trial results. Eur J Cancer Care (Engl) 15: 299–302, 2006
CrossRefPubMedGoogle Scholar
. 64.↵

1. Bergner R, 

2. Henrich DM, 
3. Hoffmann M, 

4. et al

: Renal safety of ibandronate in multiple myeloma patients with renal deterioration. Cancer Treat Rev 31[suppl
1]: S45, 2005
Google Scholar
. 65.↵

1. Carter GD, 

2. Goss AN

: Bisphosphonates and avascular necrosis of the jaws. Aust Dent J 48: 268, 2003


PubMedGoogle Scholar
. 66.↵

1. Hellstein JW, 

2. Kalmar JR

: For the dental patient. Osteoporosis medications and the risk of developing jaw complications. J Am Dent
Assoc 138: 692, 2007
FREE Full TextGoogle Scholar
. 67.↵

1. Carneiro E, 

2. Vibhute P, 

3. Montazem A, 

4. Som PM

: Bisphosphonate-associated mandibular osteonecrosis. AJNR Am J Neuroradiol 27: 1096–1097, 2006


Abstract/FREE Full TextGoogle Scholar
. 68.↵

American Dental Association Council on Scientific Affairs: Dental management of patients receiving oral
bisphosphonate therapy: Expert panel recommendations. J Am Dent Assoc 137: 1144–1150, 2006
.
Abstract/FREE Full TextGoogle Scholar
.

. 69.↵

1. Repo MA, 

2. Bockman RS, 

3. Betts F, 

4. Boskey AL, 

5. Alcock NW, 

6. Warrell RP Jr.

: Effect of gallium on bone mineral properties. Calcif Tissue Int 43: 300–306, 1988


PubMedGoogle Scholar
. 70.

1. Bockman RS

: Studies on the mechanism of action of gallium nitrate. Semin Oncol 18[Suppl 5]: 21–25, 1991


PubMedGoogle Scholar
. 71.

1. Warrell RP Jr., 

2. Bockman RS, 

3. Coonley CJ, 

4. Isaacs M, 

5. Staszewski H

: Gallium nitrate inhibits calcium resorption from bone and is effective treatment for cancer-related hypercalcemia. J
Clin Invest 73: 1487–1490, 1984
PubMedGoogle Scholar
. 72.↵

1. Hall TJ, 

2. Chambers TJ

: Gallium inhibits bone resorption by a direct effect on osteoclasts. Bone Miner 8: 211–216, 1990


CrossRefPubMedGoogle Scholar
. 73.↵

1. Cvitkovic F, 

2. Armand J-P, 

3. Tubiana-Hulin M, 

4. Rossi JF, 

5. Warrell RP Jr.

: Randomized, double-blind, phase II trial of gallium nitrate compared with pamidronate for acute control of cancer-
related hypercalcemia. Cancer J 12: 47–53, 2006
CrossRefPubMedGoogle Scholar
. 74.↵

1. Warrell RP Jr., 

2. Murphy WK, 

3. Schulman P, 

4. O’Dwyer PJ, 

5. Heller G

: A randomized double-blind study of gallium nitrate compared with etidronate for acute control of cancer-related
hypercalcemia. J Clin Oncol 9: 1467–1475, 1991
AbstractGoogle Scholar
. 75.↵

1. Warrell RP Jr., 

2. Israel R, 

3. Frisone M, 

4. Snyder T, 

5. Gaynor JJ, 

6. Bockman RS
: Gallium nitrate for acute treatment of cancer-related hypercalcemia. A randomized, double-blind comparison to
calcitonin. Ann Intern Med 108: 669–674, 1988
CrossRefPubMedGoogle Scholar
. 76.↵

1. Huang CL, 

2. Sun L, 

3. Moonga BS, 

4. Zaidi M

: Molecular physiology and pharmacology of calcitonin. Cell Mol Biol (Noisy-le-grand) 52: 33–43, 2006


PubMedGoogle Scholar
. 77.↵

1. Purdue BW, 

2. Tilakaratne N, 

3. Sexton PM

: Molecular pharmacology of the calcitonin receptor. Receptors Channels 8: 243–255, 2002


CrossRefPubMedGoogle Scholar
. 78.↵

1. Davidson TG

: Conventional treatment of hypercalcemia of malignancy. Am J Health Syst Pharm 58[Suppl 3]: S8–S15, 2001


Abstract/FREE Full TextGoogle Scholar
. 79.↵

1. Mathur M, 

2. Sykes JA, 

3. Saxena VR, 

4. et al

: Treatment of acute lymphoblastic leukemia-induced extreme hypercalcemia with pamidronate and


calcitonin. Pediatr Crit Care Med 4: 252–255, 2003
CrossRefGoogle Scholar
. 80.↵

1. Bilezikian JP

: Management of acute hypercalcemia. N Engl J Med 326: 1196–1203, 1992


CrossRefPubMedGoogle Scholar
. 81.↵

1. Santarpia L, 

2. Koch CA, 

3. Sarlis NJ

: Hypercalcemia in cancer patients: Pathobiology and management. Horm Metab Res 42: 153–164, 2010


CrossRefPubMedGoogle Scholar
. 82.↵

1. Argilés A, 

2. Kerr PG, 
3. Canaud B, 

4. Flavier JL, 

5. Mion C

: Calcium kinetics and the long-term effects of lowering dialysate calcium concentration. Kidney Int 43: 630–
640, 1993
CrossRefPubMedGoogle Scholar
. 83.↵

1. Srámek V, 

2. Novák I, 

3. Matĕjovic M, 

4. Rokyta R, 

5. Nalos M, 

6. Hora P, 

7. Pittrová H

: Continuous venovenous hemodiafiltration (CVVHDF) with citrate anticoagulation in the treatment of a patient with
acute renal failure, hypercalcemia, and thrombocytopenia. Intensive Care Med 24: 262–264, 1998
CrossRefPubMedGoogle Scholar
. 84.↵

1. Kindgen-Milles D, 

2. Kram R, 

3. Kleinekofort W, 

4. Morgera S

: Treatment of severe hypercalcemia using continuous renal replacement therapy with regional citrate
anticoagulation. ASAIO J 54: 442–444, 2008
CrossRefPubMedGoogle Scholar
. 85.↵

1. Castellano D, 

2. Sepulveda JM, 

3. García-Escobar I, 

4. Rodriguez-Antolín A, 

5. Sundlöv A, 

6. Cortes-Funes H

: The role of RANK-ligand inhibition in cancer: the story of denosumab. Oncologist 16: 136–145, 2011


Abstract/FREE Full TextGoogle Scholar
. 86.↵

Background document for meeting of advisory committee for reproductive health drugs. FDA report, August 13,
2009. Available at http://www.fda.gov. Accessed 2/29/2012
.
Google Scholar
.

. 87.↵
1. Saad F, 

2. Brown JE, 

3. Van Poznak C, 

4. Ibrahim T, 

5. Stemmer SM, 

6. Stopeck AT, 

7. Diel IJ, 

8. Takahashi S, 

9. Shore N, 

10. Henry DH, 
11. Barrios CH, 
12. Facon T, 
13. Senecal F, 
14. Fizazi K, 
15. Zhou L, 
16. Daniels A, 
17. Carriere P, 
18. Dansey R
: Incidence, risk factors, and outcomes of osteonecrosis of the jaw: Integrated analysis from three blinded active-
controlled phase III trials in cancer patients with bone metastases. Ann Oncol 23: 1341–1347, 2012
Abstract/FREE Full TextGoogle Scholar
. 88.↵

1. Stopeck AT, 

2. Lipton A, 

3. Body J-J, 

4. Steger GG, 

5. Tonkin K, 

6. de Boer RH, 

7. Lichinitser M, 

8. Fujiwara Y, 

9. Yardley DA, 

10. Viniegra M, 
11. Fan M, 
12. Jiang Q, 
13. Dansey R, 
14. Jun S, 
15. Braun A
: Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast
cancer: a randomized, double-blind study. J Clin Oncol 28: 5132–5139, 2010
Abstract/FREE Full TextGoogle Scholar
. 89.

1. Henry DH, 

2. Costa L, 

3. Goldwasser F, 

4. Hirsh V, 

5. Hungria V, 

6. Prausova J, 

7. Scagliotti GV, 

8. Sleeboom H, 

9. Spencer A, 

10. Vadhan-Raj S, 
11. von Moos R, 
12. Willenbacher W, 
13. Woll PJ, 
14. Wang J, 
15. Jiang Q, 
16. Jun S, 
17. Dansey R, 
18. Yeh H
: Randomized, double-blind study of denosumab versus zoledronic acid in the treatment of bone metastases in
patients with advanced cancer (excluding breast and prostate cancer) or multiple myeloma. J Clin Oncol 29: 1125–
1132, 2011
Abstract/FREE Full TextGoogle Scholar
. 90.↵

1. Fizazi K, 

2. Carducci M, 

3. Smith M, 

4. Damião R, 

5. Brown J, 

6. Karsh L, 

7. Milecki P, 

8. Shore N, 

9. Rader M, 

10. Wang H, 
11. Jiang Q, 
12. Tadros S, 
13. Dansey R, 
14. Goessl C
: Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate
cancer: A randomised, double-blind study. Lancet 377: 813–822, 2011
CrossRefPubMedGoogle Scholar
. 91.↵

1. Hu MI, 

2. Gucalp R, 

3. Insogna K, 

4. et al

: Denosumab for treatment of hypercalcemia of malignancy in patients with solid tumors or hematological
malignancies refractory to IV bisphosphonates: a single-arm multicenter study. Presented at the 53rd American
Society of Henatology Annual Meeting and Exposition, San Diego, CA, December 10–13, Abstract 2483
Google Scholar
. 92.↵

1. Diel IJ, 

2. Body J-J, 

3. Stopeck A, 

4. et al

: Effect of denosumab treatment on prevention of hypercalcemia of malignancy in cancer patients with metastatic
bone disease. Presented at the European Multidisciplinary Cancer Congress, Stockholm, Sweden, September 23–
27, 2011
Google Scholar
View Abstract

You might also like