Anda di halaman 1dari 13

c h a p t e r

64
Hypercalcemia of Malignancy
Karena L. Swan and John J. Wysolmerski

CHAPTER OUTLINE
HISTORICAL INTRODUCTION,  1125 TREATMENT, 1131
NORMAL PHYSIOLOGY OF CALCIUM Hydration, 1131
METABOLISM, 1126 Inhibition of Bone Resorption,  1132
Parathyroid Hormone and the Calcium-Sensing Calcitonin, 1132
Receptor, 1126 Gallium Nitrate,  1133
Vitamin D,  1127 Mithramycin, 1133
Parathyroid Hormone–Related Protein,  1127 Bisphosphonates, 1133
MALIGNANCY-ASSOCIATED HYPERCALCEMIA,  1127 Denosumab, 1134
Humoral Hypercalcemia of Malignancy,  1128 Glucocorticoids, 1134
Ectopic Production of Parathyroid Hormone,  1128 Dialysis, 1134
Overproduction of 1,25-Dihydroxyvitamin D,  1129
Local Osteolytic Hypercalcemia,  1130

KEY POINTS
• H  ypercalcemia is a common complication of malignancy.
• Humoral hypercalcemia of malignancy is frequently caused by elevations in circulating
parathyroid hormone–related protein secreted by tumors not in the skeleton.
• Local osteolytic hypercalcemia is caused by local cytokine production by tumor cells
located within the bone microenvironment.
• Bone resorption is the key pathophysiologic mechanism by which excess calcium is
released into the circulation.
• Hypercalcemia of malignancy can be effectively treated by lowering bone resorption
and by treating the underlying malignancy if possible.
  

HISTORICAL INTRODUCTION
1960s supported this hypothesis by describing patients
Hypercalcemia is a frequent complication of cancer and with hypercalcemia who lacked bony metastases.6,7 These
can occur in 20% to 30% of patients during the course patients had squamous, renal, and genitourinary cancers as
of their malignant disease.1 It has been estimated that opposed to breast cancer or myeloma. Lafferty concluded
patients with this disorder have a 30-day mortality of that these tumors must secrete parathyroid hormone and
50%.2 The first reported case of malignancy-associated named this process pseudohyperparathyroidism.8
hypercalcemia (MAHC) was in the 1920s, coinciding At that point, it was thought that tumors could cause
with the development of an assay to measure serum cal- hypercalcemia through direct skeletal involvement or by
cium.3 In 1936, the first series of cases was published on acting on the skeleton in a humoral fashion. We now
patients with MAHC.4 The reported patients suffered refer to these conditions as two subtypes of MAHC. The
from myeloma and breast cancer with skeletal involve- first is local osteolytic hypercalcemia (LOH), and the sec-
ment, and their hypercalcemia was attributed to skeletal ond is humoral hypercalcemia of malignancy (HHM).
destruction caused by local tumor invasion. The humoral type generally predominates in clinical
In 1941, Fuller Albright suggested an alternative mech- practice.9,10
anism: that a circulating humoral factor might cause some As discussed later, in the 1980s investigators made
cases of MAHC.5 Studies published in the 1950s and great strides in understanding the pathophysiology of both
1125

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
1126 PART 6  PARATHYROID GLAND, CALCIOTROPIC HORMONES, AND BONE METABOLISM

LOC and HHM. First, it was appreciated that tumor cells circulation upon degradation and is excreted by the kid-
could make cytokines that stimulated the local produc- ney. Parathyroid secretory granules also digest the amino-
tion and activity of bone-resorbing osteoclasts within the terminal portion of PTH and release the C-­ terminal
bone microenvironment surrounding tumor cells. Second, portion into the circulation during hypercalcemic condi-
investigations into HHM led to the discovery of a new tions.13 PTH secretion is regulated by extracellular ion-
hormone, parathyroid hormone-related peptide (PTHrP), ized calcium concentrations, and small changes in ionized
which is evolutionarily related to parathyroid hormone calcium concentrations can result in large changes in PTH
and uses the same receptor. As a result, when tumors secretion.14-16
secrete PTHrP into the circulation, it acts like PTH to Parathyroid cells regulate PTH secretion by detect-
increase osteoclast differentiation and activity throughout ing changes in extracellular calcium concentrations
the skeleton. It has also become clear that two additional through a G-protein–coupled-receptor known as the
types of MAHC exist: 1,25(OH)2–vitamin D-induced calcium-­sensing receptor (CaSR).17-21 Calcium binds to
hypercalcemia in patients with lymphomas and true ecto- the calcium-sensing receptor and activates a downstream
pic production of parathyroid hormone. These will be signaling cascade that suppresses PTH synthesis and sub-
discussed further in this chapter. sequent secretion.18,19,22 Persistently elevated calcium
levels can result in degradation of PTH within parathy-
roid cells.23 The CaSR is also expressed in the kidney,
NORMAL PHYSIOLOGY OF CALCIUM METABOLISM where it regulates calcium handling by the renal tubules.
Calcium can be found in the body in two predominant Hypercalcemia activates the CaSR, resulting in suppres-
forms. First, calcium salts are key components of bone sion of renal calcium reabsorption24 and excretion of
hydroxyapatite, which provides structural integrity to excess calcium in the urine.
the skeleton so that this organ can bear weight, support The actions of PTH are mediated through a G-­protein–
locomotion, and protect internal organs. Second, soluble coupled-receptor known as the type 1 PTH/PTHrP
calcium ions in the extracellular fluid (ECF) and cytosol receptor (PTH1R), because it responds to both PTH
contribute to a multitude of biochemical reactions, signal- and PTHrP. Activation of the receptor by PTH activates
ing cascades, and electrical systems. In the adult human, several signaling cascades, but the best documented has
there are about 1000 g of calcium, of which 99% is found been the adenylyl cyclase pathway. In the kidney, PTH
in bone.11 Thus, only 1% of calcium is found in the ECF acts on proximal tubular cells to inhibit reabsorption
and soft tissues. Skeletal and ECF calcium are exchange- of phosphate through removal and degradation of the
able allowing bone to provide calcium to the ECF, when sodium-phosphate co-transporters 2a and 2c (NPT2a
calcium concentrations are low and also to store excess and NPT2c) from the luminal membranes.25 It also stim-
calcium when needed. Ionized or free calcium represents ulates the activity of renal 1α hydroxylase in proximal
approximately 50% of the circulating pool, and the tubular cells and inhibits 24-hydroxylase, which results
remaining fraction is bound to serum proteins. Calcium in an increase in the biologically active 1,25 (OH)2 vita-
is largely bound to albumin in the circulation, but can min D.26 PTH increases the reabsorption of calcium in
also be complexed with citrate or sulfate.11 The ionized the kidney through reclaiming calcium from the glo-
fraction is the physiologically important one, but it is dif- merular filtrate. There is also a PTH-independent para-
ficult to measure, as it requires anaerobic conditions for cellular process linked to the reabsorption of sodium. By
collection and handling. Therefore, clinically total serum increasing the positive charges on the luminal side of the
calcium is most commonly used as an indirect measure- tubule, PTH promotes calcium reabsorption in the corti-
ment of the ionized fraction, although some medical cen- cal thick ascending loop of Henle. In the distal tubule,
ters now routinely measure the ionized fraction. calcium channels are inserted into apical membranes to
The parathyroid glands, kidneys, skeleton, and gut are stimulate the basolateral sodium-calcium exchanges.27 In
the organs involved in calcium homeostasis through the the skeleton, PTH activates bone turnover and releases
action of parathyroid hormone (PTH), vitamin D, and stored calcium. It does this by stimulating transport of
PTH-related protein (PTHrP). These interactions will be calcium across bone lining cells from a pool of calcium at
described in further detail. the surface of bone. It also stimulates both bone resorp-
tion and bone formation. PTH increases the number and
Parathyroid Hormone and activity of bone-forming osteoblasts and bone-resorbing
the Calcium-Sensing Receptor osteoclasts, increases the size of the osteoblast precur-
PTH is produced from the four parathyroid glands sor pool,28 increases the bone-forming activity of mature
located near the upper and lower poles of the thyroid osteoblasts, and promotes the release of colony-stimulat-
gland. These glands are derived from the fourth and third ing factor 1 and receptor activator of nuclear factor κB
branchial pouches, respectively, and can be found any- (NF-κB) ligand (RANKL), which stimulate the formation
where along the path of embryologic migration. of new osteoclasts and activate mature osteoclasts.29 PTH
PTH is an 84–amino acid protein synthesized as a inhibits osteoblast production of osteoprotegerin, which
single-chain, pre-pro parathyroid peptide. This peptide is is a soluble decoy receptor for RANKL that inhibits
cleaved twice to generate the biologically active form of osteoclast development. The ultimate effects of PTH on
the hormone, PTH 1-84.12 PTH has a very short half- bone turnover depend on the pattern of PTH exposure.
life (minutes) and is degraded by the liver and kidney. Continued long-term elevations in PTH stimulate net
The C-terminal fragment of PTH is released into the bone resorption while intermittent PTH stimulation can

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
64  HYPERCALCEMIA OF MALIGNANCY 1127

result in net bone formation. See Chapter 56 for further its development or functioning. Many different hormones
discussion on PTH. and growth factors regulate the transcription and/or
stability of PTHrP mRNA. As with PTH, the CaSR has
Vitamin D been found to regulate PTHrP gene expression in many
In the skin, keratinocytes absorb ultraviolet light and cells.41,42
convert 7-dehydrocholesterol to vitamin D3.30 This is The amino terminus of PTHrP is remarkably similar to
the biologically inert form of vitamin D, and it must be that of PTH. The two peptides share 8 of the first 13 amino
hydroxylated in the liver by vitamin D 25-hydroxylase acids and a high degree of predicted secondary structure
to 25-hydroxyvitamin D3.31 The second hydroxylation over the next 21 amino acids. These common sequences
occurs in the kidney, when 25-hydroxyvitamin D3 is allow both proteins to bind and activate the same PTH/
converted to 1,25 (OH)2 vitamin D3, by 25 (OH)D-1α PTHrP receptor (PTH1R). The vast majority of studies
hydroxylase (1α hydroxylase).31 This enzymatic pro- in vitro suggest that this receptor binds PTHrP and PTH
cess in the kidney is stimulated by PTH and phosphate with equal affinity and that both activate the receptor in
and inhibited by hypercalcemia and FGF23. The 1α an identical manner. However, the human PTH1R may
hydroxylase enzyme is also expressed in immune cells, respond slightly differently to PTH and PTHrP. There
especially activated macrophages. Therefore, 1,25(OH)2 appear to be physical differences in the binding of the two
vitamin D3 can be produced by macrophages, classically peptides to different conformational states of the recep-
in granulomas. tor, so that the duration of cAMP production is shorter
Vitamin D is a steroid hormone that interacts with for PTHrP 1 to 36 than for PTH 1 to 34.43 Nevertheless,
specific nuclear receptors to regulate gene expression. when PTHrP enters the systemic circulation, it can mimic
The vitamin D receptor (VDR) is a member of the steroid the effects of PTH and cause hypercalcemia in HHM.44
hormone receptor family of ligand-binding transcription PTHrP has been found in at least some cells of almost
factors.31-33 When 1,25(OH)2 vitamin D is present, the all organs, and a variety of functions have been ascribed
vitamin D receptor heterodimerizes with the retinoid acid to PTHrP. The best documented are its roles in the
x receptor and binds to specific DNA sequences within the growth plate during the development of endochondral
target gene.34,35 1,25(OH)2 vitamin D stimulates calcium bone, the development of the mammary gland, its ability
absorption from the diet by stimulating expression of to relax smooth muscle in the vasculature and uterus, and
TRPV6, an apical membrane calcium channel that allows its stimulation of pancreatic islet cell proliferation. These
calcium influx into intestinal epithelial cells by enhanc- functions are discussed at greater length in Chapter 57.
ing production of mediators, such as calbindin D. This Pertinent to its role in causing hypercalcemia in cancer, it
facilitates calcium movement through the cytoplasm by is worth highlighting PTHrP’s function during lactation.
stimulating the calcium pump, Ca ATPase 1b (PMCA1b), Milk production requires a great deal of calcium, much of
which moves calcium across the basolateral membrane of which comes from the maternal skeleton. As a result, lac-
gut enterocytes, and by enhancing paracellular calcium tating women lose 5% to 8% of their bone mass over the
absorption through a change in charge selectivity of the first 6 months of full-time nursing, and lactating rodents
tight junctions, possibly through changes in Claudin 2 can lose 25% to 30% of their skeletal mass in as little
and 12 levels.34 At pathologic levels, 1,25 (OH)2 Vitamin as 12 to 14 days. Bone loss during this time is mediated,
D can increase bone resorption by binding to the VDR in part, by the secretion of PTHrP from breast epithelial
on osteoblasts and stimulating osteoblastic production cells into the systemic circulation. This is the only time
of RANKL, leading to osteoclast activation and bone that PTHrP circulates in normal individuals in apprecia-
resorption.36,37 See Chapter 59 for detailed treatment of ble amounts and, like PTH, during lactation it increases
vitamin D chemistry and biology. bone resorption to liberate skeletal calcium stores. This
PTH-like function during reproductive cycles is probably
Parathyroid Hormone–Related Protein the principal evolutionary pressure that helped to main-
Parathyroid hormone-related protein (PTHrP) is a tain PTHrP’s ability to share the same receptor with PTH,
“cousin” of PTH; both peptides are derived from related even though, in every other aspect, the two peptides func-
genes that together with TIP-39 define a small gene fam- tion much differently. Since HHM exists because PTHrP
ily. PTHrP was initially identified as a cause of MAHC. and PTH share the same receptor, ultimately, this evolu-
Much research has now shown that PTHrP is a widely tionary pressure also explains why hypercalcemia occurs
expressed cytokine that is involved in a variety of physi- in many malignancies.45
ologic functions. PTHrP is discussed in greater length
in Chapter 57. What follows here is a short discussion MALIGNANCY-ASSOCIATED HYPERCALCEMIA
enabling the reader to appreciate its pathophysiologic
role in humoral hypercalcemia of malignancy (see later). Historically, MAHC has been subdivided into two main
In humans, PTHrP is encoded by a single-copy gene categories. The first occurs in patients with few or no bone
containing 8 exons and at least 3 promoters located on metastases but a diffuse upregulation of bone resorption.
the short arm of chromosome 12.38-40 Alternative splic- These tumors produce a humoral factor(s) (usually PTHrP)
ing at the 3′ end of the gene gives rise to three different that circulates and causes increased osteoclast numbers
classes of mRNAs coding for specific translation prod- and activity. This type of MAHC has been referred to as
ucts of 139, 141, or 173 amino acids. PTHrP mRNA has humoral hypercalcemia of malignancy (HHM). The second
been found in almost every organ at some time during main category includes tumors with extensive osteolytic

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
1128 PART 6  PARATHYROID GLAND, CALCIOTROPIC HORMONES, AND BONE METABOLISM

bone metastases. In these tumors, the metastatic cells Although PTH and PTHrP act on the same receptor,
within the skeleton produce local paracrine factors that several biochemical aspects of HHM differ from changes
increase osteoclastic bone resorption around the tumor seen in primary hyperparathyroidism. First, as noted ear-
deposits, but not systemically. These tumors require an lier, renal calcium clearance is higher in HHM than it is
extensive skeletal tumor burden to cause hypercalcemia. in hyperparathyroidism. Second, although PTHrP levels
are elevated, 1,25(OH)2 vitamin D levels are suppressed
Humoral Hypercalcemia of Malignancy in HHM, whereas the elevations in PTH in primary hyper-
HHM is the more common of the two types of MAHC, parathyroidism result in increased 1,25(OH)2 vitamin D
representing 75% to 80% of cases in consecutive series of levels. Finally, bone histomorphometric studies in patients
patients with MAHC.9,10 Classically, it occurs most com- with HHM demonstrate very elevated rates of bone resorp-
monly in squamous, renal, and genitourinary malignan- tion but suppressed bone formation rates.51 In contrast,
cies, some breast cancers, and human T-cell leukemia virus typically in primary hyperparathyroidism, rates of both
(HTLV) 1–associated adult T-cell leukemia/lymphomas.46 bone formation and bone resorption are elevated. Similar
However, almost all tumor types have been reported to differences in bone formation rates in HHM as compared to
occasionally cause HHM. It is difficult to determine the hyperparathyroidism are noted when biochemical markers
exact incidence of HHM in patients with different types of of bone turnover such as total deoxypyridinoline (Dpyd),
cancer as the frequency generally increases as cancer pro- pyridinoline cross-linked carboxyterminal telopeptide of
gresses. For instance, approximately 15% of patients with type 1 collagen (ICTP), serum osteocalcin (OC), and serum
renal cell carcinoma were shown to have hypercalcemia bone type alkaline phosphatase (B-ALP) were measured.52
prior to treatment.47 In contrast, up to 50% of patients It is still not entirely clear why elevations in PTHrP produce
with squamous cell tumors of the head and neck followed different effects on the kidney and bone cells if both pep-
for several years eventually developed HHM.48 In fact, tides act on the same receptor. Nonetheless, studies by Hor-
hypercalcemia is typically a complication of advanced can- witz and colleagues revealed that infusions of PTHrP are
cer, and the prognosis is poor in patients with HHM, with less effective at stimulating 1,25(OH)2 vitamin D produc-
reports suggesting a median survival of 2 to 3 months.49,50 tion than are infusions of PTH.53 Interestingly, in the same
HHM is most commonly caused by a tumor, distant studies PTH and PTHrP stimulated renal tubular reabsorp-
from the skeleton, releasing PTHrP into the systemic circu- tion of calcium equivalently.53-55 As for the uncoupling of
lation. PTHrP is often a normal product of the cells of ori- bone formation from bone resorption, tumor production
gin of the tumor, but in the setting of malignancy, PTHrP of IL-6 has been implicated in human squamous carcinoma
production is upregulated by tumor cells, and the tumors cells that also secrete PTHrP, and this cytokine may accel-
evade the normal barriers that usually keep PTHrP within erate osteoclastic bone resorption and suppress osteoblastic
the local tissue environment. As a result, PTHrP enters the function.56 Finally, although PTHrP and PTH appear to act
systemic circulation and gains access to the PTHR1 pool in equivalently in many experimental systems, recent studies
the skeleton and kidney that are normally reserved for PTH. have documented differences in the way the two peptides
This is associated with a characteristic pattern of biochemi- bind to the PTH1R, thus providing a potential mechanism
cal abnormalities first defined by Stewart and colleagues in through which the two proteins might have tissue-specific
1980.9 Patients typically demonstrate elevated serum cal- differences in signaling that would explain their divergent
cium levels associated with suppressed PTH levels and low effects on 1,25 vitamin D production and the stimulation
1,25 (OH)2 vitamin D levels. As a result, calcium clear- of bone formation by osteoblasts.43,44,57,58
ance in the kidneys is increased and gut calcium absorp-
tion is decreased, contributing to a profound negative Ectopic Production of Parathyroid Hormone
calcium balance. Serum phosphorus levels are low-normal Although HHM was originally thought to be secondary to
or low with a decreased TmP/GFR. Nephrogenous cAMP ectopic PTH production, it is quite rare that MAHC is actu-
(NcAMP) excretion rates are elevated, reflecting activation ally caused by ectopic production of PTH. Approximately
of the renal proximal tubular PTH receptor despite the 18 cases exist in the literature of ectopic hyperparathy-
reduction in circulating PTH levels. The elevated NcAMP roidism caused by a nonparathyroid tumor. These tumors
levels in HHM formed the basis for biochemical assays include neuroendocrine tumors, hepatocellular carcinoma,
that resulted in the purification of PTHrP from tumors and medullary thyroid cancer, ovarian carcinoma, small-cell
the cloning of the PTHrP gene in the 1980s. lung carcinoma, squamous carcinoma, rhabdoid tumor of
Hypercalcemia in HHM results from the release of cal- the kidney, endometrial adenosquamous carcinoma, and
cium from the skeleton at a rate that exceeds the kidney’s thymoma.59-73 Immunoradiometric two-site assays for
ability to excrete the excess load (Fig. 64-1). Patients ini- intact PTH and PTHrP allow for the determination as to
tially develop hypercalciuria but eventually develop dehy- whether PTH or PTHrP is causing hypercalcemia in malig-
dration due to an osmotic diuresis combined with the effects nancy. There is no cross-reactivity between PTH and PTHrP
of calcium to induce a form of nephrogenic diabetes insipi- in these assays.59 Nussbaum and coworkers described a
dus due to activation of the CaSR on the distal tubule and patient with ovarian carcinoma, in which genetic evaluation
collecting duct. Once patients become dehydrated, serum of the tumor cells revealed DNA amplification and rear-
calcium levels typically rise rapidly and patients develop rangement in the upstream regulatory region of the PTH
symptoms of lethargy, anorexia, and confusion from the gene. This resulted in increased PTH expression.59 Van-
elevated ionized calcium levels that only worsen the dehy- Houten and associates described a patient with a high-grade
dration and accelerate the rise in serum calcium levels. neuroendocrine carcinoma of the pancreas found to have

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
64  HYPERCALCEMIA OF MALIGNANCY 1129

ectopic PTH production. Genetic evaluation of these tumor source of the 1α-hydroxylase was the tumor-associated
cells revealed transactivation of the PTH gene promoter macrophages and not the lymphoma cells themselves.
associated with hypomethylation of the PTH gene locus, This would suggest that the tumor cells make some para-
indicative of re-activation of PTH gene transcription in the crine factor that upregulates 1α-hydroxylase expression in
tumor cells.70 macrophages, although it is not clear whether lymphoma
cells can also sometimes express 1α-hydroxylase. Unlike
Overproduction of 1,25-Dihydroxyvitamin D the kidney, there is no systemic feedback inhibition of the
Another rare cause of HHM is the overproduction of tumor-associated 1α-hydroxylase activity, but 1,25(OH)2
1,25(OH)2 vitamin D by lymphomas, both Hodgkin’s vitamin D production is responsive to 25 vitamin D levels.
and non-Hodgkin’s types.74-78 In this setting, local acti- Hypercalcemia in these patients is largely secondary to
vation of 1α-hydroxylase activity within or around the the hyperabsorption of calcium in the intestine, although
tumor results in the overproduction of the active form of high 1,25 vitamin D levels can also directly cause bone
vitamin D. In at least one carefully studied patient, the resorption (see Fig. 64-1). Patients typically have normal

NORMAL HOMEOSTASIS
Oral Ca intake
1000 mg

500 mg
ECF Ca2+
150 mg
1000 mg
GI tract 0 mg Skeleton
Stimulated Serum Ca
by 1,25 9.5 mg/dL
500 mg
vit D

Filtered load of Ca in kidney


10,000 mg

Distal tubular calcium reabsorption is


stimulated by PTH
850 mg

150 mg
A Kidney
HUMORAL HYPERCALCEMIA OF MALIGNANCY

Oral Ca intake
Skeleton

Decreased ↑ PTHrP levels


absorption results in
Serum Ca
GI tract uncoupling of
10.5->14 mg/dL
↓ levels bone formation
1,25 vit D and resorption.

Higher filtered load of


Ca in kidney

↓ PTH levels
↑ PTHrP levels leads to enhanced
Increased renal retention of Ca
excretion
Greater, but insufficient Ca excretion
B Kidney
Figure 64-1  Comparison of normal calcium fluxes with MAHC. A, The central box represents extracellular fluid (ECF), which contains approxi-
mately 1000 mg of calcium. This compartment interfaces with the gastrointestinal tract, the kidney, and the skeleton. The fluxes into and out of the
ECF are measured in milligrams per day. There is a zero calcium balance under homeostatic conditions. B, In humoral hypercalcemia of malignancy,
elevated circulating PTHrP levels result in an uncoupling of bone resorption from bone formation and a net output of calcium from bone. There is
decreased intestinal absorption of calcium secondary to reduced 1,25 vitamin D levels. The kidney filters an increased load of calcium, and PTHrP
acts on the kidney to enhance renal retention of calcium. However, this is insufficient to reduce serum calcium concentrations to normal levels.
Continued

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
1130 PART 6  PARATHYROID GLAND, CALCIOTROPIC HORMONES, AND BONE METABOLISM

LOCAL OSTEOLYTIC HYPERCALCEMIA

Oral Ca intake
Skeleton

Decreased
absorption ↑ local release of
Serum Ca factors by
GI tract
10.5->14 mg/dL skeletal
↓ levels metastasis leads
1,25 vit D to uncoupling of
bone formation
and resorption.
Higher filtered load of
Ca in kidney

↓ PTH levels
↑ excretion of urinary calcium
Increased
excretion
Greater, but insufficient Ca excretion
C Kidney
1,25 VITAMIN D MEDIATED HYPERCALCEMIA

Oral Ca intake
Skeleton

Increased
absorption ↑↑ 1,25 vit D may
Serum Ca
GI tract lead to increased
10.5->14 mg/dL
↑↑ 1,25 bone resorption
vit D

Higher filtered load of


Ca in kidney

↓ PTH levels
↑ excretion of urinary calcium
Decreased
excretion
Greater, but insufficient Ca excretion
D Kidney
Figure 64-1, cont’d  C, In local osteolytic hypercalcemia, local release of factors from skeletal metastases leads to the local uncoupling of bone
resorption from formation. If there are enough metastases, there is a net output of calcium from bone. There is decreased intestinal absorption of
calcium secondary to reduced 1,25 vitamin D levels. The kidney filters an increased load of calcium, but this is insufficient to reduce serum calcium
concentrations to normal levels. D, In 1,25 vitamin D-mediated hypercalcemia, there is a marked increase in intestinal calcium absorption, and there
also may be an increase in bone resorption. The kidney filters an increased load of calcium, but this is insufficient to reduce serum calcium concentra-
tions to normal levels. (A, Adapted from Stewart AF. Normal physiology of bone and mineral homeostasis. In Andreoli TE, et al. (eds.). Andreoli and
Carpenter’s Cecil essentials of medicine. Philadelphia: Elsevier Saunders; 2010.)

serum phosphate levels and increased renal calcium excre- the stimulation of osteolysis around the bone metastases
tion. PTH levels are appropriately suppressed in the face with release of calcium into the systemic circulation. In
of hypercalcemia, and PTHrP concentrations are unmea- order to develop hypercalcemia, patients usually must
sureable.77 However, hypercalcemia can also be caused have extensive skeletal metastases. Classically, the most
by lymphomas that secrete PTHrP. Therefore, measuring common tumors presenting with this syndrome include
PTHrP levels is clinically important in this setting. myeloma, breast cancer, and lymphoma.
The biochemical findings in the disorder are hypercal-
Local Osteolytic Hypercalcemia cemia with reduced PTH and 1,25(OH)2 vitamin D lev-
Local osteolytic hypercalcemia (LOH) is the second most els, similar to HHM. However, in this condition, patients
common form of MAHC composing approximately 20% have normal serum phosphorus concentrations and a
to 30% of patients in consecutive series of patients with normal renal phosphate threshold. In contrast to HHM,
MAHC.9,10,79 In patients with LOH, the hypercalcemia PTHrP levels are undetectable, so nephrogeneous cAMP
is caused by tumor growth within bone, which leads to (NcAMP) levels are reduced secondary to the suppressed

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
64  HYPERCALCEMIA OF MALIGNANCY 1131

PTH levels. Urinary calcium excretion is increased as a are typically osteolytic, and if the skeletal burden is great
result of an increased filtered load of calcium found in enough, enough calcium can be released from the skeleton
hypercalcemic conditions and a decreased distal tubular to overwhelm the body’s adaptive mechanisms, resulting in
calcium reabsorption in the setting of lower PTH levels. hypercalcemia. It is difficult to ascertain the current inci-
Intestinal absorption of calcium is also reduced because of dence of hypercalcemia in patients with breast cancer and
the decreased circulating 1,25(OH)2 vitamin D levels. As bone metastases. Older studies suggested that 10% to 15%
with HHM, this results in a net negative calcium balance. of all patients with breast cancer developed hypercalcemia,
The pathophysiology of LOH is an extension of the patho- the majority of whom had bone metastases. More recent
physiology of osteolytic bone metastases. In order for tumor studies (usually drug trials) group together hypercalcemia,
cells to survive and grow in bone, they influence the behavior pathologic fracture, radiation treatment for impending frac-
of normal bone cells to remodel the bone microenvironment, ture, and bone pain under the term skeletal-related events
often activating bone resorption and causing osteolysis. This (SRE) and do not often report the incidence of hypercalce-
leads to the release of factors from the bone matrix that, mia separately. In these trials, the control group of patients
in turn, stimulate further growth of the tumor cells, causing with bone metastases had an SRE rate of approximately
more osteolysis. This vicious cycle of peri-tumor osteolysis 60%. The use of prophylactic bisphosphonates is associated
and tumor growth has been the focus of many studies. In with an aggregate 15% reduction in SREs, so it is likely
short, it appears that tumor cells produce soluble factors that the widespread use of these medications in patients
that lead to increased osteoclast recruitment, differentiation, with breast cancer and bone metastases has reduced the
and activity.80-83 Some tumors also elaborate factors that incidence of hypercalcemia by at least a similar amount.95
inhibit osteoblast-mediated bone formation. As a result, the Like myeloma cells, breast cancer cells have been shown
tumors cause progressive bone destruction, allowing them to produce a variety of factors that induce local bone resorp-
to expand within the skeleton. If the number and size of the tion (Fig. 64-2). One of the best studied of these factors is
osteolytic metastases is great enough, then the release of cal- PTHrP. It is well documented that PTHrP is produced by a
cium from the skeleton will exceed the kidney’s ability to number of primary breast carcinomas and that this some-
excrete the filtered load, and patients will develop clinical times leads to classical HHM.96 However, animal models
hypercalcemia, usually associated with renal insufficiency have suggested that localized PTHrP production by breast
(see Fig. 64-1). As opposed to HHM, bone resorption does tumor cells contributes to the osteolysis associated with
not happen throughout the skeleton in LOH, just around the skeletal metastases.97,98 These studies have suggested that, in
individual tumor deposits. The general pathophysiology of response to the bone microenvironment, breast cancer cells
bone metastases has been reviewed in great detail previously. metastatic to the skeleton produce more PTHrP than cells
In this setting, we will only briefly review some of the main in the primary tumor.98,99 TGF-β, released at sites of bone
cytokines that have been implicated in driving the vicious resorption, has been shown to upregulate PTHrP produc-
cycle of osteolysis, but the reader is referred to other reviews tion through a signaling pathway involving the transcription
for a more complete discussion of this topic.84-86 factor Gli2.100 PTHrP, in turn, increases the production of
One of the tumors that frequently causes extensive RANKL and decreases the production of OPG, increasing
bone destruction and hypercalcemia is myeloma. Forty osteoclast numbers and activity.99 This sets up a feedforward
years ago, Mundy described an osteoclast-activating fac- loop between PTHrP production and bone resorption, con-
tor (OAF) that was produced by myeloma cells.87 Many tributing to a vicious cycle of osteolysis. In addition, unlike
subsequent studies have identified a series of cytokines in normal breast cells, CaSR signaling in breast cancer cells
produced by myeloma cells that have the ability to cause stimulates PTHrP production and can synergize with the
osteolysis. These include interleukin 1α (IL-1α), IL-1β, effects of TGF-β.101,102 Finally, the mechanical stiffness of
tumor necrosis factor α (TNF-α), TNF-β (lymphotoxin), bone tissue may also increase PTHrP production.103 Thus,
transforming growth factor (TGF-α), MIP-1α (macrophage several aspects of the bone microenvironment conspire to
inflammatory protein-1α), IL-3, IL-6, and CD-47.88 Many increase PTHrP production in skeletal metastases. How-
of these cytokines increase osteoclast differentiation and ever, PTHrP is not the only factor important in this pro-
activity by increasing the production of receptor activator cess. Breast cancer cells have been shown to produce other
of NF-κB ligand (RANKL) by osteoblasts.89-91 RANKL is osteolytic factors, such as IL-6, IL-11, prostaglandin E2,
the dominant factor produced by osteoblasts to regulate Jagged 1, macrophage colony-stimulating factor (MCSF),
the production and activity of osteoclasts, and this factor and macrophage-stimulating protein (MSP).83 MCSF and
acts through a receptor, RANK, found on osteoclast pro- MSP may act directly to stimulate osteoclast production and
genitors and mature osteoclasts. In recent years, it has also activity, while the other factors act with PTHrP to stimulate
become clear that suppression of bone formation is also RANKL production by osteoblasts.83,104,105 As depicted in
important in the pathogenesis of myeloma bone disease. Figure 64-2, the growth of osteolytic metastases depends
This appears to be related to the secretion of Dickkopf1 on a vicious cycle of bone resorption and tumor cell prolif-
(DKK-1), an inhibitor of the WNT signaling pathway, eration that is fed by the release of growth factors from the
by myeloma cells. Inhibition of Wnt signaling suppresses bone matrix, the breast cancer cells, and normal bone cells.
osteoblast activity and bone formation, augmenting the
local osteolysis and release of calcium from the bone.92-94 TREATMENT
Breast cancer is the second tumor type classically asso-
ciated with LOH. Bone metastases are common in breast Hydration
cancer, occurring in up to 70% of patients with advanced Hypercalcemia is associated with polyuria. In addition,
disease. As in myeloma, bone metastases from breast cancers hypercalcemic patients also suffer from reduced oral intake,

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
1132 PART 6  PARATHYROID GLAND, CALCIOTROPIC HORMONES, AND BONE METABOLISM

Osteoblasts
Angiogenesis
↓ OB differentiation invasive growth
↑ OC activity bone resorption
immunosuppression
Active
Latent TGF-β
TGF-β
Cancer cells

Osteoclasts
Osteoclast
precursors IL-6

PTHrP
IL-11
CTGF
↑ RANKL
↓ OPG Jagged

Bone
matrix

Osteoblasts
Figure 64-2  Osteolytic bone metastasis in breast cancer. In skeletal metastases, the tumor cells make factors, including PTHrP, that upregulate
osteoclastic bone resorption. This results in the release of TGF-β from the bone matrix, which acts on cancer cells to further stimulate the production
of osteolytic factors, such as PTHrP, connective tissue growth factor (CTGF), IL-6, and IL-11. These factors increase the RANKL/OPG expression
ratio in bone stromal cells such as osteoblasts, resulting in osteoclastogenesis. Moreover, active TGF-β stimulates Jagged 1 expression in cancer cells,
which in turn stimulates Notch signaling in osteoclasts and osteoblasts, resulting in increased osteoclastogenesis and production of the cytokine IL-6
by osteoblast (blue dotted line), which stimulates proliferation of tumor cells. TGF-β also inhibits osteoblast differentiation. Thus, once osteolysis
is established, a vicious cycle of bone resorption and tumor cell proliferation is fed by the release of growth factors from bone matrix, tumor cells,
and bone cells. (Reproduced with permission from Buijs JT, Stayrook KR, Guise TA. The role of TGF-beta in bone metastasis: novel therapeutic
perspectives. Bonekey Rep. 2012 1:96.)

due to anorexia, nausea, and vomiting. These factors lead and colleagues reviewed the literature on this topic and
to dehydration and a reduction in GFR.106 An impair- argued that routine use of loop diuretics in the treatment
ment in renal function, in turn, limits the ability to excrete of MAHC was not helpful.108 Clearly, if loop diuretics are
the increased calcium load coming usually from bone and given before adequate volume repletion, they run the risk of
accelerates the development of hypercalcemia. As a result, prolonging dehydration and may actually impair the excre-
the first goal of therapy is to correct volume depletion tion of a calcium load on this basis. Therefore, if diuretics
(Fig. 64-3). It has typically been recommended to hydrate are to be used, their administration must be delayed until
patients with 0.9% NS (normal saline) at 200 to 500 mL/ volume status has been fully restored. In addition, they are
hour intravenously. The initial rate and duration of fluid valuable in managing hypercalcemic patients who develop
administration should be determined by clinical signs of fluid overload after aggressive fluid resuscitation.108
dehydration, the duration and severity of hypercalcemia,
and underlying medical conditions, especially cardiovascu- Inhibition of Bone Resorption
lar disease. After initial volume repletion and establishment The majority of patients with MAHC are hypercalcemic
of adequate urine output, fluids can be continued at >100 because of excessive bone resorption. Therefore, the main-
mL/hour, but it is important to carefully monitor vital signs stay of therapy is the administration of powerful antiresorp-
and watch for signs of fluid overload, especially in cases tive agents, primarily bisphosphonates and denosumab.
with existing comorbidities such as congestive heart failure However other agents, such as calcitonin, gallium nitrate,
(CHF).106,107 While intravenous fluid administration can and mithramycin have also been described to be helpful in
transiently lower serum calcium concentrations on the order lowering calcium levels. In the sections that follow, we will
of 1 to 1.5 mg/dL, additional therapies are required to lower discuss the available agents for inhibiting bone resorption,
calcium further and to maintain the lower calcium levels. concentrating on the most clinically useful and available
It has been common practice to add loop diuretics, such agents, pamidronate, zolendronic acid, and Denosumab.
as furosemide at a dose of 20 to 40 mg intravenously, after
rehydration has been achieved to promote calcium excre- Calcitonin
tion through a forced saline diuresis. The concept is to take Calcitonin is a peptide hormone secreted by the parafol-
advantage of the fact that sodium delivery to the distal tubule licular cells of the thyroid gland that acts to inhibit osteo-
promotes urinary calcium excretion and helps to accelerate clast activity. It has been utilized as an antiresorptive
calcium clearance by the kidneys.1,106 However, LeGrand therapy for both hypercalcemia of malignancy and Paget’s

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
64  HYPERCALCEMIA OF MALIGNANCY 1133

SUGGESTED TREATMENT OF MALIGNANCY ASSOCIATED HYPERCALCEMIA

Intravenous saline *Consider calcitonin


0.9% NS 200–500 ml/hr 4–8 IU/kg SQ/IM every
[Determined by the volume status of the patient and the 12 hours × 48–72 hrs
duration and severity of hypercalcemia] In conditions of altered
consciousness or
Ca >15 mg/dL

Furosemide
20–40 mg IV
[After rehydration has been achieved]

Pamidronate Denosumab
60–90 mg IV over a 2 hour period 120 mg SQ weekly for 4 weeks
in 50–200 ml of saline or and then every 4 weeks
5% dextrose in water

Or
*Consider glucocorticoids
Zoledronate For example, prednisone 60 mg
4 mg IV over a 15 minute period orally daily for 10 days
in 50 ml of saline or In cases of lymphoma or elevated
5% dextrose in water 1,25 vitamin D levels Figure 64-3  Suggested treatment for
malignancy-associated hypercalcemia.

disease.106,107 It also has been shown to decrease the renal treatment with etidronate, 28 of 34 patients (82%) who
tubular resorption of calcium, thus promoting renal cal- received gallium nitrate achieved normocalcemia, and the
cium excretion.109,110 The benefits of calcitonin are its rapid median duration of normocalcemia was 8 days.113
action, but its limitations have been the rapid development
of tachyphylaxis and its relatively modest calcium-lowering Mithramycin
actions. Salmon calcitonin has a longer half-life in the cir- Mithramycin, otherwise known as plicamycin, is an antibi-
culation than human calcitonin, and it is given as a subcu- otic agent that was initially used to treat testicular neoplasms
taneous or intramuscular injection at a dose of 4 to 8 IU and was found to lower serum calcium levels.115 It was
per kilogram every 12 hours.1 Administration of calcitonin utilized for the treatment of hypercalcemia of malignancy
results in a rapid reduction in serum calcium concentrations prior to the introduction of bisphosphonates. It interferes
with a maximal response within 12 to 24 hours. However, with osteoclastic function and can reduce serum calcium
the reductions are small (approximately 1 mg/dL), and the levels within 24 hours.116 It is given as a single dose of 25
effects are usually lost within 48 to 96 hours, probably due mcg/kg of body weight over a 4- to 6-hour period in saline.1
to the downregulation of calcitonin receptors.111 Neverthe- Its side effects include thrombocytopenia, platelet defects
less, it can be a valuable temporizing agent that can rap- resulting in bleeding diathesis, azotemia, proteinuria, and
idly lower calcium levels acutely while waiting for the more hepatic toxicity.117 It is rarely, if ever, used currently.
prolonged onset of other antiresorptive agents. This can be
an important initial therapy for patients in danger of dying Bisphosphonates
from extreme elevations in serum calcium levels. Bisphosphonates are the most commonly used medica-
tions to treat MAHC in the United States. These drugs
Gallium Nitrate are analogues of pyrophosphates, which resist enzymatic
Gallium nitrate was initially evaluated as a cancer chemo- hydrolysis and have a high affinity for hydroxyapatite.118
therapeutic, and patients receiving this treatment devel- Bisphosphonates concentrate at areas of high bone turn-
oped a positive calcium balance with reduced urinary over and inhibit osteoclast function. Most patients are
calcium excretion.112 The hypocalcemic effects result treated either with pamidronate (aminohydroxy-propyl-
from the inhibition of bone resorption.113 It is given as a idene bisphosphonate) or zoledronate (zolendronic acid),
continuous intravenous infusion of 100 to 200 mg/m2 of both potent nitrogen-containing bisphosphonates that
body surface area over a 24-hour period for 5 consecutive interfere with protein prenylation through the inhibition
days.1,106 Gallium nitrate can cause nephrotoxicity, and it of the mevalonate pathway enzyme, farnesyl disphosphate
is important that adequate urine output be maintained on synthase, which is downstream of HMG-CoA reductase.
this therapy.114 In one study comparing gallium nitrate These medications inhibit osteoclast function by inducing
treatment to treatment with calcitonin, 18 of 24 patients apoptosis.118 The standard dose of pamidronate is 60 to 90
receiving gallium nitrate achieved normocalcemia, and mg given intravenously over a 2-hour period in a solution
the median duration of normocalcemia was 6 days.114 of 50 to 200 mL of saline or 5% dextrose in water.1,119
In another study comparing gallium nitrate therapy to Zoledronate is given as a 4-mg intravenous infusion over

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
1134 PART 6  PARATHYROID GLAND, CALCIOTROPIC HORMONES, AND BONE METABOLISM

15 minutes in a solution of 50 mL of saline or 5% dex- treatment option in patients with impaired renal function,
trose in water.1 The serum calcium level normalizes in who cannot tolerate bisphosphonates. Hypocalcemia is
most patients, but the response can take 3 to 7 days and one of the main adverse effects of denosumab. Patients in
last from 1 to 3 weeks. In one study examining the dose- the Freedom trial also had a slightly increased incidence of
response to treatment with pamidronate in patients with skin infections as compared to patients treated with pla-
MAHC, corrected serum calcium normalized in 61% of cebo.125 In the denosumab group, 0.3% reported serious
patients receiving 60 mg and 100% of patients receiving adverse events of cellulitis, as compared with <0.1% in the
90 mg.119 The mean duration of normalization in these two placebo group (P = .002).125
groups was 13.3 and 10.8 days in the 60-mg and 90-mg Denosumab has been shown to prevent SREs or hyper-
treatment groups, respectively.119 One study, which was calcemia of malignancy in phase III studies of advanced
a pooled analysis from two randomized, double-blind, malignancies.126-128 Compared with monthly infusions of
parallel-group trials, showed that zolendronate was more zolendronate, administration of denosumab was associ-
effective at normalizing serum calcium levels in patients ated with an 18% reduction in the risk for SREs127 and a
with HCM, with a significantly longer time to relapse than 70% success rate in normalization of serum calcium levels
pamidronate.120,121 in a small study.129 It also appears that Denosumab may
In this study, two treatment doses of zoledronic acid, 4 be useful in patients with MAHC who do not respond to
mg and 8 mg, were compared to 90 mg of pamidronate. The bisphosphonates. In a recent study by Hu and coworkers,
complete response rates were 88.4%, 86.7%, and 69.7%, patients with hypercalcemia of malignancy refractory to
respectively.121 The corrected serum calcium normalized bisphosphonate treatment received subcutaneous deno-
by day 4 in 50% of the zoledronic acid-treated patients and sumab on days 1, 8, 15, and 29, and then every 4 weeks
33.3% of the pamidronate-treated patients.121 The median at a dose of 120 mg given as a subcutaneous injection.130
duration of complete response was 32 and 43 days in those In this study, by day 10, 12/15 patients had responded
treated with 4 mg or 8 mg of zoledronic acid, respectively, with albumin-corrected serum calcium levels (CSC) <11.5
and 18 days in those treated with pamidronate.121 mg/dL, and the median duration of response was 26 days.
The dosing interval is every 3 to 4 weeks, depending on In 10 of 15 patients, the CSC was <10.8 mg/dL.130
the recurrence of hypercalcemia. There may be a reduction
in efficacy with repeated dosing. The intravenous bisphos- Glucocorticoids
phonates are excreted unchanged by the kidneys and must Glucocorticoids can be an effective treatment for hyper-
be adjusted for impaired renal function. The most com- calcemia associated with an overproduction of 1,25(OH)2
mon adverse effects are renal failure and 24 to 48 hours vitamin D in patients with lymphoma.106 This is likely
of transient flulike symptoms, including fever, aches, and due to the ability of glucocorticoids to suppress local
chills precipitated by the initial infusion. The fever is often cytokine production resulting in the suppression of 1,25
mild and usually occurs only with the first infusion. vitamin D production through a reduction in vitamin D
In addition to the acute treatment of MAHC, bisphos- 1α-hydroxylase activity.107 Treatment can consist of 60 mg/
phonates are now routinely given to patients with day of prednisone for 10 days.1 This treatment can interfere
myeloma and breast cancer in order to prevent the occur- with some chemotherapeutic agents, and treatment can be
rence of SREs, including hypercalcemia. A recent com- associated with hypokalemia, hyperglycemia, hyperten-
prehensive meta-analaysis demonstrated that treatment sion, Cushing’s syndrome, and immunosuppression.1
of women with breast cancer and bone metastases with
regular infusions of bisphosphonates reduced the over- Dialysis
all rate of SRE’s by 15% in randomized trials and also Patients with hypercalcemia and acute kidney injury with
delayed the onset of the first SRE in patients.95 However, oliguria may require dialysis to lower their calcium lev-
this treatment did not prolong survival, and adjuvant els. In these patients, a saline diuresis is not possible and
treatment of breast cancer without bone metastases was could lead to significant fluid overload. Hemodialysis or
not associated with enhanced survival.122 peritoneal dialysis utilizing a very low calcium or cal-
cium-free dialysate is implemented in such situations.106
Denosumab It can result in a rapid decrease in serum total calcium
Denosumab is the most recent antiresorptive therapy to be levels.131
approved for the treatment of MAHC. It is a humanized
monoclonal antibody that binds to RANKL and prevents it Acknowledgement
from binding to the receptor RANK on osteoclast precur- This work was supported by NIH Grants DK55501 and
sors and mature osteoclasts. This leads to the inhibition of CA153702 to JW and NIH Grant CA153702-09S1 to KS.
the differentiation, activation, and function of osteoclasts   
which, in turn, leads to a reduction in bone resorption.123 It • For your free Expert Consult eBook with biblio-

has a profound suppressive effect on biochemical markers graphic citations as well as the ability to take notes,
of bone resorption and is the most powerful antiresorptive highlight important content, search the full text, and
agent currently available.124 Denosumab is cleared through more, visit http://www.ExpertConsult.Inkling.com.
the reticuloendothelial system, independent from the kid-
neys.123 The lack of renal toxicity makes it an attractive

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
REFERENCES 27. Friedman PA, Gesek FA. Calcium transport in renal epithelial
cells. Am J Physiol. 1993;264(2 Pt 2):F181–F198.
1. Stewart AF. Clinical practice. Hypercalcemia associated with can- 28. Nishida S, et al. Increased bone formation by intermittent para-
cer. N Engl J Med. 2005;352(4):373–379. thyroid hormone administration is due to the stimulation of pro-
2. Ralston SH, et al. Cancer-associated hypercalcemia: morbid- liferation and differentiation of osteoprogenitor cells in bone mar-
ity and mortality. Clinical experience in 126 treated patients. row. Bone. 1994;15(6):717–723.
Ann Intern Med. 1990;112(7):499–504. 29. Teitelbaum SL, Ross FP. Genetic regulation of osteoclast develop-
3. Zondek H, Petow H, Siebert W. Die Bedeutung der Calciumb- ment and function. Nat Rev Genet. 2003;4(8):638–649.
estimmung im Blute furdie Diagnose der Niereninsuffizienz. 30. MacLaughlin JA, Anderson RR, Holick MF. Spectral character of
Z Klin Med. 1923;99:129–138. sunlight modulates photosynthesis of previtamin D3 and its pho-
4. Gutman A, Tyson T, Gutman E. Serum calcium, inorganic phos- toisomers in human skin. Science. 1982;216(4549):1001–1003.
phorus and phosphatase activity in hyperparathyroidism, Paget’s 31. Bikle D, Adams J, Christakos S. Vitamin D production, metabo-
disease, multiple myeloma and neoplastic disease of the bones. lism, mechanisms of action, and clinical requirements. In: Rosen C,
Arch Intern Med. 1936;57:379–413. Compston J, Lian J, eds. Primer on the metabolic bone diseases
5.  Case Records of the Massachusetts General Hospital (Case and disorders of mineral metabolism. Washington, DC: The
27461). N Engl J Med. 1941;225:789–791. American Society for Bone and Mineral Research; 2008:141–149.
6. Plimpton C, Gelhorn A. Hypercalcemia in malignant disease with- 32. Haussler M, et al. In: Feldman D, Pike J, Adams J, Vitamin D,
out evidence of bone destruction. Am J Med. 1956;21:750. eds. Nuclear vitamin D receptor: natural ligands, molecular struc-
7. Connor T, Thomas W, Howard J. The etiology of hypercalcemia ture-function, and transcriptional control of vital genes. 3rd ed.
associated with lung carcinoma. J Clin Invest. 1956;35:597. Waltham, MA: Academic Press; 2011:137–170.
8. Lafferty F. Pseudohyperparathyroidism. Medicine. 1966;45:247. 33. Pike J, Meyer M, Lee S. In: Feldman D, Pike J, Adams J, Vita-
9. Stewart AF, et al. Biochemical evaluation of patients with cancer- min D, eds. The vitamin D receptor: biochemical, molecular, bio-
associated hypercalcemia: evidence for humoral and nonhumoral logical, and genomic era investigations. Waltham, MA: Academic
groups. N Engl J Med. 1980;303(24):1377–1383. Press; 2011:97–135.
10. Burtis WJ, et al. Immunochemical characterization of circulating 34. Fleet JC, Schoch RD. Molecular mechanisms for regulation of
parathyroid hormone-related protein in patients with humoral hy- intestinal calcium absorption by vitamin D and other factors.
percalcemia of cancer. N Engl J Med. 1990;322(16):1106–1112. Crit Rev Clin Lab Sci. 2010;47(4):181–195.
11. Wysolmerski J, Insogna K. The parathyroid glands, hyper- 35. Fleet JC, et al. Vitamin D and cancer: a review of molecular mech-
calcemia, and hypocalcemia. In: Goldman L, Schafer A, eds. anisms. Biochem J. 2012;441(1):61–76.
Goldman’s Cecil Medicine. 24th ed. Philadelphia: Elsevier 36. Takeda S, et al. Stimulation of osteoclast formation by 1,25-dihy-
Saunders; 2012:1591–1601. droxyvitamin D requires its binding to vitamin D receptor (VDR)
12. Wiren KM, Potts Jr JT, Kronenberg HM. Importance of the pro- in osteoblastic cells: studies using VDR knockout mice. Endocri-
peptide sequence of human preproparathyroid hormone for signal nology. 1999;140(2):1005–1008.
sequence function. J Biol Chem. 1988;263(36):19771–19777. 37. Kearns AE, Khosla S, Kostenuik PJ. Receptor activator of nuclear
13. D’Amour P, et al. Acute regulation of circulating parathyroid factor kappaB ligand and osteoprotegerin regulation of bone re-
hormone (PTH) molecular forms by calcium: utility of PTH frag- modeling in health and disease. Endocr Rev. 2008;29(2):155–192.
ments/PTH(1-84) ratios derived from three generations of PTH 38. Mangin M, et al. An identification of a cDNA encoding a parathy-
assays. J Clin Endocrinol Metab. 2006;91(1):283–289. roid hormone-like peptide from a human tumor associated with
14. Mayer GP, Hurst JG. Sigmoidal relationship between parathy- humoral hypercalcemia of malignancy. Proc Natl Acad Sci U S A.
roid hormone secretion rate and plasma calcium concentration in 1988;85:597–601.
calves. Endocrinology. 1978;102(4):1036–1042. 39. Philbrick WM, et al. Defining the roles of parathyroid hormone-
15. Brown EM. Four-parameter model of the sigmoidal relationship related protein in normal physiology. Physiol Rev. 1996;76:
between parathyroid hormone release and extracellular calcium 127–173.
concentration in normal and abnormal parathyroid tissue. J Clin 40. Suva LJ, et al. A parathyroid hormone-related protein implicat-
Endocrinol Metab. 1983;56(3):572–581. ed in malignant hypercalcemia: cloning and expression. Science.
16. Brenner B, Stein H. Divalent ion homeostasis. Contemp Issues 1987;237:893–896.
Nephrol. 1997;2:779–799. 41. Chattopadhyay N. Effects of calcium-sensing receptor on the se-
17. Brown EM, et al. Cloning and characterization of an extracel- cretion of parathyroid hormone-related peptide and its impact on
lular Ca(2+)-sensing receptor from bovine parathyroid. Nature. humoral hypercalcemia of malignancy. Am J Physiol Endocrinol
1993;366(6455):575–580. Metab. 2006;290(5):E761–E770.
18. Brown EM, MacLeod RJ. Extracellular calcium sensing and extra- 42. VanHouten J, et al. The calcium-sensing receptor regulates mam-
cellular calcium signaling. Physiol Rev. 2001;81(1):239–297. mary gland parathyroid hormone-related protein production and
19. Chattopadhyay N, Mithal A, Brown EM. The calcium-sensing calcium transport. J Clin Invest. 2004;113(4):598–608.
receptor: a window into the physiology and pathophysiology of 43. Dean T, et al. Altered selectivity of parathyroid hormone (PTH)
mineral ion metabolism. Endocr Rev. 1996;17(4):289–307. and PTH-related protein (PTHrP) for distinct conformations
20. Riccardi D, Kemp PJ. The calcium-sensing receptor beyond ex- of the PTH/PTHrP receptor. Mol Endocrinol. 2008;22(1):
tracellular calcium homeostasis: conception, development, adult 156–166.
physiology, and disease. Annu Rev Physiol. 2012;74:271–297. 44. Stewart AF, et al. Biochemical evaluation of patients with cancer-
21. Hofer AM, Brown EM. Extracellular calcium-sensing and signal- associated hypercalcemia: evidence for humoral and nonhumoral
ling. Nat Rev Mol Cell Biol. 2003;4(7):530–538. groups. N Engl J Med. 1980;303:1377–1383.
22. Chen RA, Goodman WG. Role of the calcium-sensing receptor 45. VanHouten JN, et al. Mammary-specific deletion of parathyroid
in parathyroid gland physiology. Am J Physiol Renal Physiol. hormone-related protein preserves bone mass during lactation.
2004;286(6):F1005–F1011. J Clin Invest. 2003;112(9):1429–1436.
23. Habener JF, Rosenblatt M, Potts Jr JT. Parathyroid hormone: bio- 46. Watanabe T, et al. Constitutive expression of parathyroid
chemical aspects of biosynthesis, secretion, action, and metabo- hormone-related protein gene in human T cell leukemia virus
lism. Physiol Rev. 1984;64(3):985–1053. type 1 (HTLV-1) carriers and adult T cell leukemia patients
24. Kantham L, et al. The calcium-sensing receptor (CaSR) defends that can be trans-activated by HTLV-1 tax gene. J Exp Med.
against hypercalcemia independently of its regulation of para- 1990;172(3):759–765.
thyroid hormone secretion. Am J Physiol Endocrinol Metab. 47. Palapattu GS, Kristo B, Rajfer J. Paraneoplastic syndromes in
2009;297(4):E915–E923. urologic malignancy: the many faces of renal cell carcinoma.
25. Murer H, Forster I, Biber J. The sodium phosphate cotransporter Rev Urol. 2002;4(4):163–170.
family SLC34. Pflugers Arch. 2004;447(5):763–767. 48. Alsirafy SA, Sroor MY, Al-Shahri MZ. Hypercalcemia in advanced
26. Bisello A, Friedman P. PTH and PTHrP actions on kidney and head and neck squamous cell carcinoma: prevalence and potential
bone. Princ of Bone Biol. 2008;1:665–712. impact on palliative care. J Support Oncol. 2009;7(5):154–157.

1134.e1

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
1134.e2 REFERENCES

49. Le Tinier F, et al. Cancer-associated hypercalcaemia in squamous- 71. Mahoney EJ, et al. Life-threatening hypercalcemia from a hepato-
cell malignancies: a survival and prognostic factor analysis. Int J cellular carcinoma secreting intact parathyroid hormone: localiza-
Oral Maxillofac Surg. 2011;40(9):938–942. tion by sestamibi single-photon emission computed tomographic
50. Penel N, et al. Cancer-associated hypercalcemia treated with in- imaging. Endocr Pract. 2006;12(3):302–306.
travenous diphosphonates: a survival and prognostic factor analy- 72. Demura M, et al. Ectopic production of parathyroid hormone
sis. Support Care Cancer. 2008;16(4):387–392. in a patient with sporadic medullary thyroid cancer. Endocr J.
51. Stewart AF, et al. Quantitative bone histomorphometry in humor- 2010;57(2):161–170.
al hypercalcemia of malignancy: uncoupling of bone cell activity. 73. Kandil E, et al. Ectopic secretion of parathyroid hormone in a
J Clin Endocrinol Metab. 1982;55(2):219–227. neuroendocrine tumor: a case report and review of the literature.
52. Nakayama K, et al. Differences in bone and vitamin D metabo- Int J Clin Exp Med. 2011;4(3):234–240.
lism between primary hyperparathyroidism and malignancy- 74. Adams JS, et al. Vitamin D metabolite-mediated hypercalcemia
associated hypercalcemia. J Clin Endocrinol Metab. 1996;81(2): and hypercalciuria patients with AIDS- and non-AIDS-associated
607–611. lymphoma. Blood. 1989;73(1):235–239.
53. Horwitz MJ, et al. Continuous PTH and PTHrP infusion causes 75. Breslau NA, et al. Hypercalcemia associated with increased serum
suppression of bone formation and discordant effects on 1,25(OH)2 calcitriol levels in three patients with lymphoma. Ann Intern Med.
vitamin D. J Bone Miner Res. 2005;20(10):1792–1803. 1984;100(1):1–6.
54. Syed MA, et al. Parathyroid hormone-related protein-(1-36) stim- 76. Rosenthal N, et al. Elevations in circulating 1,25-dihydroxyvita-
ulates renal tubular calcium reabsorption in normal human volun- min D in three patients with lymphoma-associated hypercalcemia.
teers: implications for the pathogenesis of humoral hypercalcemia J Clin Endocrinol Metab. 1985;60(1):29–33.
of malignancy. J Clin Endocrinol Metab. 2001;86(4):1525–1531. 77. Seymour JF, Gagel RF. Calcitriol: the major humoral mediator of
55. Horwitz MJ, et al. Direct comparison of sustained infusion hypercalcemia in Hodgkin’s disease and non-Hodgkin’s lympho-
of human parathyroid hormone-related protein-(1-36) [hP- mas. Blood. 1993;82(5):1383–1394.
THrP-­ (1-36)] versus hPTH-(1-34) on serum calcium, plasma 78. Mundy GR, et al. Bone-resorbing activity in supernatants from
1,25-­dihydroxyvitamin D concentrations, and fractional calcium lymphoid cell lines. N Engl J Med. 1974;290(16):867–871.
excretion in healthy human volunteers. J Clin Endocrinol Metab. 79. Budayr AA, et al. Increased serum levels of a parathyroid hor-
2003;88(4):1603–1609. mone-like protein in malignancy-associated hypercalcemia.
56. Nagai Y, et al. Role of interleukin-6 in uncoupling of bone Ann Intern Med. 1989;111(10):807–812.
in vivo in a human squamous carcinoma coproducing parathyroid 80. Yoneda T, Sasaki A, Mundy GR. Osteolytic bone metastasis in
hormone-related peptide and interleukin-6. J Bone Miner Res. breast cancer. Breast Cancer Res Treat. 1994;32(1):73–84.
1998;13(4):664–672. 81. Clohisy DR, et al. Human breast cancer induces osteoclast acti-
57. Datta NS, Abou-Samra AB. PTH and PTHrP signaling in osteo- vation and increases the number of osteoclasts at sites of tumor
blasts. Cell Signal. 2009;21(8):1245–1254. osteolysis. J Orthop Res. 1996;14(3):396–402.
58. Cupp ME, et al. Parathyroid hormone (PTH) and PTH-related 82. Pederson L, et al. Identification of breast cancer cell line-derived
peptide domains contributing to activation of different PTH paracrine factors that stimulate osteoclast activity. Cancer Res.
receptor-mediated signaling pathways. J Pharmacol Exp Ther. 1999;59(22):5849–5855.
2013;345(3):404–418. 83. Winding B, et al. Human breast cancer cells induced angiogenesis,
59. Nussbaum SR, Gaz RD, Arnold A. Hypercalcemia and ectopic recruitment, and activation of osteoclasts in osteolytic metastasis.
secretion of parathyroid hormone by an ovarian carcinoma with J Cancer Res Clin Oncol. 2000;126(11):631–640.
rearrangement of the gene for parathyroid hormone. N Engl J 84. Buijs JT, Stayrook KR, Guise TA. The role of TGF-beta in
Med. 1990;323(19):1324–1328. bone metastasis: novel therapeutic perspectives. Bonekey Rep.
60. Botea V, Edelson GW, Munasinghe RL. Hyperparathyroidism, 2012;1:96.
hypercalcemia, and calcified brain metastatic lesions in a patient 85. Papachristou DJ, Basdra EK, Papavassiliou AG. Bone metasta-
with small cell carcinoma demonstrating positive immunostain for ses: molecular mechanisms and novel therapeutic interventions.
parathyroid hormone. Endocr Pract. 2003;9(1):40–44. Med Res Rev. 2012;32(3):611–636.
61. Samaan NA, et al. Ectopic parathyroid hormone production 86. Eyob H, Ekiz HA, Welm AL. RON promotes the metastatic
by a squamous carcinoma of the tonsil. Arch Otolaryngol. spread of breast carcinomas by subverting antitumor immune
1983;109(2):91–94. responses. Oncoimmunology 2(9):e25670. 2013.
62. Mayes LC, et al. Hypercalcemia associated with immunoreactive 87. Mundy GR, et al. Evidence for the secretion of an osteoclast stimu-
parathyroid hormone in a malignant rhabdoid tumor of the kid- lating factor in myeloma. N Engl J Med. 1974;291(20):1041–1046.
ney (rhabdoid Wilms’ tumor). Cancer. 1984;54(5):882–884. 88. Mundy GR. Hypercalcemic factors other than parathyroid
63. Arps H, et al. Pancreatic endocrine carcinoma with ectopic PTH- hormone-related protein. Endocrinol Metab Clin North Am.
production and paraneoplastic hypercalcaemia. Virchows Arch A 1989;18(3):795–806.
Pathol Anat Histopathol. 1986;408(5):497–503. 89. Shimizu H, et al. A case of myeloma with hypercalcemia caused by
64. Buller R, et al. Paraneoplastic hypercalcemia associated with ad- high serum concentrations of both parathyroid hormone-related
enosquamous carcinoma of the endometrium. Gynecol Oncol. peptide (PTHrP) and macrophage inflammatory protein-1alpha
1991;40(1):95–98. (MIP-1alpha). Intern Med. 2011;50(24):2993–2996.
65. Koyama Y, et al. Intact PTH-producing hepatocellular carcinoma 90. Gao JL, et al. Structure and functional expression of the human
treated by transcatheter arterial embolization. Abdom Imaging. macrophage inflammatory protein 1 alpha/RANTES receptor.
1999;24(2):144–146. J Exp Med. 1993;177(5):1421–1427.
66. Uchimura K, et al. Lung cancer associated with hypercalce- 91. Abe M, et al. Role for macrophage inflammatory protein (MIP)-
mia induced by concurrently elevated parathyroid hormone 1alpha and MIP-1beta in the development of osteolytic lesions in
and parathyroid hormone-related protein levels. Metabolism. multiple myeloma. Blood. 2002;100(6):2195–2202.
2002;51(7):871–875. 92. Heider U, et al. Serum concentrations of DKK-1 decrease in pa-
67. Yoshimoto K, et al. Ectopic production of parathyroid hormone tients with multiple myeloma responding to anti-myeloma treat-
by small cell lung cancer in a patient with hypercalcemia. J Clin ment. Eur J Haematol. 2009;82(1):31–38.
Endocrinol Metab. 1989;68(5):976–981. 93. Zhou F, Meng S, Song H, Claret FX. Dickkopf-1 is a key regula-
68. Strewler GJ, et al. Production of parathyroid hormone by a ma- tor of myeloma bone disease: Opportunities and challenges for
lignant nonparathyroid tumor in a hypercalcemic patient. J Clin therapeutic intervention. Blood Rev. 2013;27(6):261–267.
Endocrinol Metab. 1993;76(5):1373–1375. 94. Tian E, et al. The role of the Wnt-signaling antagonist DKK1 in
69. Rizzoli R, et al. A thymoma as a cause of true ectopic hyperpara- the development of osteolytic lesions in multiple myeloma. N Engl
thyroidism. J Clin Endocrinol Metab. 1994;79(3):912–915. J Med. 2003;349(26):2483–2494.
70. VanHouten JN, et al. Hypercalcemia of malignancy due to ectopic 95. Wong MH, Stockler MR, Pavlakis N. Bisphosphonates and oth-
transactivation of the parathyroid hormone gene. J Clin Endocri- er bone agents for breast cancer. Cochrane Database Syst Rev.
nol Metab. 2006;91(2):580–583. 2012;2:CD003474.

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
REFERENCES 1134.e3

96. Isales C, Carcangiu ML, Stewart AF. Hypercalcemia in breast cancer. 115. Perlia CP, et al. Mithramycin treatment of hypercalcemia. Cancer.
Reassessment of the mechanism. Am J Med. 1987;82(6):1143–1147. 1970;25(2):389–394.
97. Guise TA, et al. Evidence for a causal role of parathyroid hor- 116. Ralston SH, et al. Comparison of aminohydroxypropylidene
mone-related protein in the pathogenesis of human breast cancer- diphosphonate, mithramycin, and corticosteroids/calcito-
mediated osteolysis. J Clin Invest. 1996;98(7):1544–1549. nin in treatment of cancer-associated hypercalcaemia. Lancet.
98. Yin JJ, et al. TGF-beta signaling blockade inhibits PTHrP secre- 1985;2(8461):907–910.
tion by breast cancer cells and bone metastases development. 117. Stewart AF. Therapy of malignancy-associated hypercalcemia:
J Clin Invest. 1999;103(2):197–206. 1983. Am J Med. 1983;74(3):475–480.
99. Akhtari M, et al. Biology of breast cancer bone metastasis. Cancer 118. Zaidi M, et al. Osteoclastogenesis, bone resorption, and osteo-
Biol Ther. 2008;7(1):3–9. clast-based therapeutics. J Bone Miner Res. 2003;18(4):599–609.
100. Johnson RW, et al. TGF-beta promotion of Gli2-induced expres- 119. Nussbaum SR, et al. Single-dose intravenous therapy with
sion of parathyroid hormone-related protein, an important osteo- pamidronate for the treatment of hypercalcemia of malig-
lytic factor in bone metastasis, is independent of canonical Hedge- nancy: comparison of 30-, 60-, and 90-mg dosages. Am J Med.
hog signaling. Cancer Res. 2011;71(3):822–831. 1993;95(3):297–304.
101. Mamillapalli R, et al. Switching of G-protein usage by the calci- 120. Cheer SM, Noble S. Zoledronic acid. Drugs. 2001;61(6):
um-sensing receptor reverses its effect on parathyroid hormone- 799–805. discussion 806.
related protein secretion in normal versus malignant breast cells. 121. Major P, et al. Zoledronic acid is superior to pamidronate in
J Biol Chem. 2008;283(36):24435–24447. the treatment of hypercalcemia of malignancy: a pooled analy-
102. Sanders JL, et al. Extracellular calcium-sensing receptor expres- sis of two randomized, controlled clinical trials. J Clin Oncol.
sion and its potential role in regulating parathyroid hormone-­ 2001;19(2):558–567.
related peptide secretion in human breast cancer cell lines. 122. Coleman RE, et al. Breast-cancer adjuvant therapy with zoledron-
Endocrinology. 2000;141(12):4357–4364. ic acid. N Engl J Med. 2011;365(15):1396–1405.
103. Ruppender NS, et al. Matrix rigidity induces osteolytic gene expres- 123. Baron R, Ferrari S, Russell RG. Denosumab and bisphosphonates:
sion of metastatic breast cancer cells. PloS One 5(11):e15451. 2010. different mechanisms of action and effects. Bone. 2011;48(4):
104. Thomas RJ, et al. Breast cancer cells interact with osteoblasts 677–692.
to support osteoclast formation. Endocrinology. 1999;140(10): 124. Boikos SA, Hammers HJ. Denosumab for the treatment of
4451–4458. bisphosphonate-refractory hypercalcemia. J Clin Oncol.
105. Welm AL, et al. The macrophage-stimulating protein pathway 2012;30(29):e299.
promotes metastasis in a mouse model for breast cancer and 125. Cummings SR, et al. Denosumab for prevention of fractures
predicts poor prognosis in humans. Proc Natl Acad Sci U S A. in postmenopausal women with osteoporosis. N Engl J Med.
2007;104(18):7570–7575. 2009;361(8):756–765.
106. Rosner MH, Dalkin AC. Onco-nephrology: the pathophysiology 126. Stopeck AT, et al. Denosumab compared with zoledronic acid
and treatment of malignancy-associated hypercalcemia. Clin J Am for the treatment of bone metastases in patients with advanced
Soc Nephrol. 2012;7(10):1722–1729. breast cancer: a randomized, double-blind study. J Clin Oncol.
107. Clines GA. Mechanisms and treatment of hypercalcemia of ma- 2010;28(35):5132–5139.
lignancy. Curr Opin Endocrinol Diabetes Obes. 2011;18(6): 127. Fizazi K, et al. Denosumab versus zoledronic acid for treatment
339–346. of bone metastases in men with castration-resistant prostate can-
108. LeGrand SB, Leskuski D, Zama I. Narrative review: furosemide cer: a randomised, double-blind study. Lancet. 2011;377(9768):
for hypercalcemia: an unproven yet common practice. Ann Intern 813–822.
Med. 2008;149(4):259–263. 128. Henry DH, et al. Randomized, double-blind study of denosum-
109. Cochran M, et al. Renal effects of calcitonin. Br Med J. ab versus zoledronic acid in the treatment of bone metastases
1970;1(5689):135–137. in patients with advanced cancer (excluding breast and pros-
110. Hosking DJ, Gilson D. Comparison of the renal and skeletal ac- tate cancer) or multiple myeloma. J Clin Oncol. 2011;29(9):
tions of calcitonin in the treatment of severe hypercalcaemia of 1125–1132.
malignancy. Q J Med. 1984;53(211):359–368. 129. Dietzek A, Connelly K, Cotugno M, et al. Denosumab in hyper-
111. Binstock ML, Mundy GR. Effect of calcitonin and glutocorticoids calcemia of malignancy: a case series. J Oncol Pharm Pract. 2014
in combination on the hypercalcemia of malignancy. Ann Intern Jan 10. Epub ahead of print.
Med. 1980;93(2):269–272. 130. Hu MI, et al. Denosumab for patients with persistent or relapsed
112. Warrell RP, Alcock NW, Bockman RS. Gallium nitrate inhibits hypercalcemia of malignancy despite recent bisphosphonate treat-
accelerated bone turnover in patients with bone metastases. J Clin ment. J Natl Cancer Inst. 2013;105(18):1417–1420.
Oncol. 1987;5(2):292–298. 131. Camus C, et al. Calcium free hemodialysis: experience in the treat-
113. Warrell Jr RP, et al. A randomized double-blind study of gallium ment of 33 patients with severe hypercalcemia. Intensive Care
nitrate compared with etidronate for acute control of cancer-­ Med. 1996;22(2):116–121.
related hypercalcemia. J Clin Oncol. 1991;9(8):1467–1475. 132. Stewart AF. Normal physiology of bone and mineral homestasis.
114. Warrell Jr RP, et al. Gallium nitrate for acute treatment of cancer- In: Andreoli TE, et al., eds. Andreoli and Carpenter’s Cecil essen-
related hypercalcemia. A randomized, double-blind comparison tials of medicine. Philadelphia: Elsevier Saunders; 2010.
to calcitonin. Ann Intern Med. 1988;108(5):669–674.

Descargado para VICENTE DAVID AGUILERA MOREIRA (vicentedavid92@hotmail.com) en Universidad de las Americas de ClinicalKey.es por Elsevier en mayo 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.

Anda mungkin juga menyukai