Onco-Nephrology: The Pathophysiology and Treatment of Malignancy-Associated Hypercalcemia

Mitchell H. Rosner* and Alan C. Dalkin†

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 *Division of Nephrology and underlying etiology. The mechanisms of malignancy-associated hypercalcemia generally fall into three cate- †Division of gories: humoral hypercalcemia due to secreted factors (such as parathyroid-related hormone), local osteolysis Endocrinology, due to tumor invasion of , and absorptive hypercalcemia due to excess vitamin D produced by malignancies. The University of Virginia mainstays of therapy for hypercalcemia are aggressive intravenous volume expansion with saline, bisphosphonate Health System, therapy, and perhaps loop diuretics. Adjunctive therapy may include and corticosteroids. In refractory Charlottesville, Virginia cases, and perhaps denosumab are alternatives. In patients presenting with severe AKI, Correspondence: Dr. hemodialysis with a low- bath can be effective. In most cases, therapy normalizes calcium levels and Mitchell H. Rosner, allows for palliation or curative therapy of the malignancy. Division of Clin J Am Soc Nephrol 7: 1722–1729, 2012. doi: 10.2215/CJN.02470312 Nephrology, Box 800133, University of Virginia Health System, Introduction measurement is unclear. In at least one study, slightly Charlottesville, VA Hypercalcemia (defined as a serum calcium level increased ionized calcium levels did not predict the 22908. Email: mhr9r@ above the upper limit of the normal reference range development of frank hypercalcemia in patients virginia.edu of 10.5 mg/dl or 2.5 mmol/L, 40%–45% of which is with solid malignant tumors (8,9). Thus, evaluation ionized calcium) can complicate the course of 10%– of total calcium concentration in light of changes in 30% of all patients with cancer (1–4). The incidence of bound versus ionized calcium is essential in order to hypercalcemia is low at the initial presentation of can- obtain a complete patient assessment. cer (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 – Mechanisms of Malignancy-Associated malignancy-associated hypercalcemia is only 2 6 Hypercalcemia months from the onset of hypercalcemia (3,5). Perturbations of calcium regulation in patients with fi The symptoms of hypercalcemia are nonspeci c, hypercalcemia of malignancy fall into three general often develop gradually, and may resemble the symp- categories (Figure 1). First, most common are hu- toms of the underlying malignancy and its treatment. moral factors secreted in a paracrine or endocrine fi These nonspeci c symptoms may lead to a delay in fashion (humoral hypercalcemia of malignancy). Sec- diagnosis, with increased morbidity and mortality. ond, in patients with substantial tumor burden met- fl Clinical presentation is in uenced by the rate of onset astatic to bone, the local effects of tumor can directly as well as the severity of hypercalcemia. Typical symp- facilitate bone dissolution and calcium mobilization toms include nausea, vomiting, constipation, abdominal (local osteolysis). Third, less commonly, absorptive pain, anorexia, weight loss, bone pain, polyuria, fatigue, hypercalcemia results from excess vitamin D activa- and weakness. In patients with more severe degrees of tion by the neoplasm. The cellular basis for the former . hypercalcemia (serum calcium 14 mg/dl), neurologic two mechanisms includes changes in the activity and symptoms such as confusion and even coma may be- balance at the level of the bone-remodeling unit. come more apparent. These patients require hospitali- Bone turnover involves the highly coordinated zation and urgent therapeutic intervention (6). activity of two distinct types of cells (10). The osteo- Given the presence of concomitant cachexia and blast, or bone-forming cell, is derived from a mesen- malnutrition in this population, it is also critical to chymal stem cell and is closely related to adipocytes, correct the serum calcium for a low albumin level or chondrocyte, myocytes, and fibroblasts. In contrast, measure an ionized serum calcium level in order to the , or bone-removing cell, is derived assess the true degree of hypercalcemia (7). Although from stems cells populating the monocyte and mac- ionized calcium is more sensitive than albumin- rophage lineage. Although the specifics remain in- corrected total calcium in the diagnosis of hypercal- completely understood, communication between cemia of malignancy, the clinical usefulness of this osteoblasts and primarily involves the

1722 Copyright © 2012 by the American Society of Nephrology www.cjasn.org Vol 7 October, 2012 Clin J Am Soc Nephrol 7: 1722–1729, October, 2012 Hypercalcemia of Malignancy, Rosner and Dalkin 1723

Figure 1. | Mechanism of malignancy-associated hypercalcemia. PTHrP, parathyroid hormone–related hormone. receptor activator of nuclear factor k B (RANK) ligand extensive in nature, can present with significant areas of os- (RANKL) signaling pathway (10–12). RANK is a receptor teolysis and hypercalcemia (23). Underlying the release of expressed on osteoclast precursor cells. The naturally oc- calcium from the bone microenvironment is increased oste- curring ligand, RANKL, is produced by osteoblasts and oclast activity, probably due to PTHrP and other factors that drives proliferation and differentiation of the osteoclasts can increase resorption. As a result of the paracrine nature of into mature, multinucleated units. In addition, the osteo- this condition, circulating levels of PTHrP may be “normal” blast cell produces osteoprotegerin (OPG), a decoy recep- or only slightly above normal, in contrast to levels in patients tor that binds to and inactivates RANKL. with humoral hypercalcemia due to PTHrP. The cellular regulation of RANK, RANKL, and OPG Vitamin D has numerous physiologic actions, including expression is incompletely understood, but the osteoblast is the enhancement of calcium and phosphate absorption the focal point for the integration of endocrine and paracrine from the intestinal tract. Stored vitamin D (25-[OH]D) is signals that alter bone remodeling. For example, osteoblast 1-hydroxylated in the kidney to the active compound 1,25- cells express estrogen receptors that when occupied with (OH)2D, which in turn acts via the vitamin D receptor. PTH ligand can reduce RANKL and increase OPG (13). Directly actively drives the 1-hydroxylase step at the kidney, and related to the hypercalcemia of malignancy, osteoblasts also states of hyperparathyroidism, including ectopic PTH from express the cell surface receptor for parathyroid hormone malignancies, are associated with elevated 1,25-(OH)2Dlev- (PTH) and parathyroid hormone–related hormone (PTHrP), els. Of note, despite its action through the common PTH1R, PTH1R (14). Both PTH and PTHrP stimulate PTH1R, which, PTHrP is a poor stimulus for 1a-hydroxylation compared in turn, increases osteoblast activity and RANKL signaling with PTH, and levels of active vitamin D are often low or to the osteoclast. OPG expression may decline as well. In normal in patients with humoral hypercalcemia of malig- sum, PTH/PTHrP signaling results in an increased bone nancy (24). Important for the understanding of hypercalce- turnover with a greater increase in than for- mia, the 1-hydroxylase enzyme is expressed in tissues mation, resulting in a net efflux of calcium and hypercalce- outside of the kidney, including macrophages and some mia from the bone microenvironment. neoplastic tissues. Patients with Hodgkin lymphoma and Tumors that commonly produce PTHrP include squa- non-Hodgkin lymphoma, as well as multiple myeloma, mous cell carcinomas of the lung, cervix, and esophagus; have been described as having vitamin D–mediated hyper- certain lymphomas; renal cell carcinoma; and adenocarci- calcemia (25). In these individuals, 1,25-(OH)2D levels, along noma of the breast, prostate, and ovary (15–17). In addi- with calcium levels, are high while PTH is suppressed from tion, several case reports have documented PTHrP the negative feedback to the parathyroid cells. Similarly, secretion in other malignancies. Ectopic secretion of PTH measurements of bone turnover markers are low in vitamin secretion is less common than that of PTHrP. The number D–mediated hypercalcemia because the reduction in PTH of cases in which tumor-related production of PTH has results in a diminution of osteoblast and osteoclast activity. been verified are few but include pulmonary, thyroid, ovarian, and pancreatic malignancies (18–20). Laboratory Investigation Local osteolysis as the basis for hypercalcemia occurs Given that the malignancy is usually advanced by the most frequently in widely metastatic disease, and the degree time hypercalcemia develops, a thorough history and of hypercalcemia correlates with the extent of tumor burden. physical examination can often lead to the correct diagnosis Local osteolysis is most commonly seen in patients with with limited laboratory evaluation. Measurement of intact metastatic breast and lung cancers (21,22). Probably due to a PTH through an immunoradiometric or immunochemolu- common pathophysiology, multiple myeloma, usually minescent assay will confirm a PTH-independent process. 1724 Clinical Journal of the American Society of Nephrology

In this case, the PTH levels will be suppressed (often Volume expansion will lead to an increase in GFR and an ,20 pg/ml) and should prompt further testing to distin- increase in urine calcium excretion. The goal should be es- guish among the multiple possible causes in patients with tablishing an adequate urine output (.75 ml per hour). In malignancy. This would include measurement of PTHrP, milder cases of hypercalcemia, this therapy can be sufficient 1,25-(OH)2D levels, serum and urine protein electrophoresis, to restore normal calcium levels (30). However, the effect of assessment of serum free light chains, and possibly imaging repleting the extracellular volume is transient, and other ther- studies (such as a skeletal survey). Less common causes of apies are required. Careful monitoring of the patient’svital hypercalcemia in malignancy include drugs (e.g., retinoic signs and laboratory values must occur during the acute acid [26]) and, rarely, parathyroid carcinomas producing phase of volume repletion, along with vigilance for evidence excessive PTH. of volume overload. Hypernatremia may be seen during this phase of therapy because of hypercalcemia-induced nephro- genic diabetes insipidus and may require changing from iso- Therapy for Malignancy-Associated Hypercalcemia tonic to hypotonic fluid therapy (31). Patients presenting with acute and symptomatic hyper- A common practice is to add a loop diuretic to saline calcemia require prompt therapy that rapidly reduces the therapy in an effort to increase urine calcium excretion serum calcium level, restores the GFR, and leads to longer- term normalization of the serum calcium level. An algo- (forced saline diuresis). LeGrand and colleagues reviewed rithmic approach is shown in Figure 2. 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 Intravenous Fluid and Diuretics Hypercalcemia leads to a decrease in the GFR through a decreases the lumen positive charge that normally drives combination of the natriuretic effects of high serum levels calcium reabsorption; thus, a loop diuretic increases urine of calcium and renal vasoconstriction (27). In fact, polyuria calcium excretion and theoretically should be useful in is the most common renal manifestation of hypercalcemia, therapy for hypercalcemia (33,34). However, LeGrand fi and it is thought that the impaired urinary concentrating et al. could nd only nine articles that documented the ability is due to activation of the calcium-sensing receptor use of furosemide in the treatment of hypercalcemia, the in the thick ascending limb of the loop of Henle (28). Ac- last of which was published in 1983. These studies tivation of the calcium-sensing receptor leads to decreased comprised a total of 37 patients. The average dosage of resorption of sodium and chloride in the loop of Henle, furosemide was 1120 mg given over 24 hours, and serum resulting in decreased countercurrent multiplication and calcium normalized in 14 of 39 episodes of hypercalcemia; , decreased ability to concentrate the urine. In addition, ac- however, normalization was rapid ( 12 hours) in only – tivation of the calcium-sensing receptor in the collecting two cases. Lower dosages of furosemide (40 60 mg/d) duct blunts the response of this segment to the actions of did not achieve normalization of the serum calcium. arginine vasopressin (29). Thus, patients presenting with Most important, monitoring in these patients was inten- hypercalcemia are usually profoundly volume depleted. sive and included aggressive replacement of hourly urine Furthermore, patients usually will present with poor oral output losses, and secondary electrolyte disorders, such as intake and may have had nausea and vomiting that further hypernatremia, hypophosphatemia, hypomagnesemia, worsen the volume depletion. The decrease in GFR leads to and metabolic acidosis, were seen. Thus, the routine use impaired calcium clearance and, thus, restoration of extra- of loop diuretics in the therapy for hypercalcemia cannot cellular fluid volume, and GFR is a key goal of therapy. be recommended, and their use should be restricted to Thus, initial therapy should begin with intravenous (IV) patients who develop fluid overload while receiving ag- volume expansion with 0.9% saline at 200–500 ml per hour. gressive volume resuscitation.

Figure 2. | Treatment algorithm for malignancy-associated hypercalcemia. IV, intravenous. Clin J Am Soc Nephrol 7: 1722–1729, October, 2012 Hypercalcemia of Malignancy, Rosner and Dalkin 1725

Bisphosphonates associated with a lower response rate) (47,48). The fact that High-potency IV bisphosphonates are effective agents patients with higher levels of PTHrP show less of a hypocal- for the treatment of malignancy-associated hypercalcemia. cemic response to bisphosphonates is probably due to the The three high-potency bisphosphonates are pamidronate, continued actions of PTHrP on the kidney to decrease cal- zoledronate, and ibandronate (dosing guidelines are cium excretion, an effect that is not modulated by bisphosph- shown in Table 1). Because the IV bisphosphonates are onates. excreted unchanged by the kidneys through glomerular Zoledronate is 100 times more potent than pamidronate, filtration, their dosing must be adjusted for impaired and two parallel randomized trials evaluated the efficacy of renal function to avoid toxicity (35,36). Mechanistically, 4 or 8 mg of zoledronate versus 90 mg of pamidronate in bisphosphonates decrease bone resorption through extra- restoring normocalcemia by day 10 after treatment (49). cellular and intracellular mechanisms (37–41). In the extra- Both doses of zoledronate were superior to pamidronate, cellular space, bisphosphonates bind to calcium phosphate with serum calcium normalizing by day 10 in 88%, 87%, and stabilize the bone matrix, while intracellularly these and 70% of patients receiving 4 and 8 mg of zoledronate agents inhibit osteoclast activity, in part through inhi- and 90 mg of pamidronate, respectively (49). Moreover, bition of the mevalonate pathway (37–39). Furthermore, the mean duration of complete response was 32, 43, bisphosphonates impair cellular adenosine triphosphate– and 18 days, respectively, in the three treatment dependent metabolic pathways, disrupt the osteoclast groups (49). cytoskeleton, and induce apoptosis (40,41). Ibandronate is also effective in treating malignancy- Randomized clinical trials with pamidronate, zoledronate, associated hypercalcemia (44,50). Doses of 2, 4, and 6 mg and ibandronate have all shown efficacy in the treatment of had success rates of 50%, 76%, and 77%, respectively hypercalcemia of malignancy (42–44). However, the three (44). Compared with pamidronate, the duration of re- bisphosphonates differ in terms of their potency and risk sponse is longer with ibandronate (50). However, for toxicity. Nussbaum et al. demonstrated a dose response ibandronate is not approved by the Food and Drug Ad- using pamidronate, with 40%, 61%, and 100% of patients ministration (FDA) to treat malignancy-associated hy- achieving normocalcemia with 30, 60, and 90 mg, respec- percalcemia. tively (45). Similar efficacy of pamidronate is seen over a The most common side effect associated with these agents dosing span ranging from 2 to 24 hours (46). Thus, a prac- is infusion-related fever, which is usually mild (51). How- tical approach is to administer pamidronate, 60–90 mg, over ever, bisphosphonate-associated nephrotoxicity affecting 2–6 hours, with the longer dosing time reserved for patients both glomerular and tubular structures has been described with lower GFRs and the higher doses for patients with in both case series and randomized clinical trials (35). In more severe hypercalcemia. The efficacy of pamidronate terms of glomerular toxicity, bisphosphonates (most com- (and probably other bisphosphonates) is influenced by the monly, pamidronate and, only rarely, zoledronate) have mechanism of hypercalcemia (more effective in cases of hy- been associated with collapsing focal segmental glomerulo- percalcemia associated with bone metastases); the level of sclerosis (FSGS), noncollapsing FSGS, and minimal-change hypercalcemia (higher doses required to treat more severe disease in several case series (52–56). The most common hypercalcemia); and, in patients with humoral hypercalce- lesion is collapsing FSGS, which is seen with high-dose IV mia, the level of PTHrP (higher levels of PTHrP are pamidronate often in patients with underlying multiple

Table 1. Dosing guidelines for intravenous bisphosphonates in the treatment of acute hypercalcemia

Bisphosphonate per CrCl Dose and Infusion Rate Dosing Interval

CrCl .60 ml/min pamidronate 90 mg over 2–3 hr Every 3–4wk zoledronate 4 mg over 15 min Every 3–4wk ibandronate Not FDA approved CrCl 30–60 ml/min pamidronate 60–90 mg over 2–3 hr Every 3–4wk Reserve higher doses for more severe hypercalcemia zoledronate Every 3–4wk CrCl 50–60 ml/min 3.5 mg CrCl 40–49 ml/min 3.3 mg CrCl 30–39 ml/min 3 mg ibandronate Not FDA approved CrCl , 30 ml/min pamidronate 60–90 mg over 4–6 hr Every 3–4wk Reserve higher doses for more severe hypercalcemia zoledronate Not recommended Not recommended ibandronate Not FDA approved

Data obtained from reference 61. CrCl, clearance; FDA, Food and Drug Administration. 1726 Clinical Journal of the American Society of Nephrology

myeloma and usually after repeated doses (35). In clinical addition, renal impairment has been reported, and hence trials, glomerular toxicity of the bisphosphonates was not gallium nitrate is not recommended for use when the se- reported (35). In some cases, the nephrotic syndrome re- rum creatinine concentration exceeds 2.5 mg/dl. Adequate solves after cessation of the drug, but some patients may fluid resuscitation is essential before initiation of treatment progress to ESRD, especially if the glomerular lesion is col- with gallium, and BUN and creatinine must be followed lapsing FSGS. closely during treatment. Patients receiving gallium have Tubular injury (nephrotoxic acute tubular necrosis) as- reported nausea, vomiting, lethargy, altered mental status, sociated with bisphosphonates (mainly, zoledronate) has and both diarrhea and constipation. also been described in both case reports and clinical trials – (57 59). In 2003, the FDA Adverse Event Reporting System Calcitonin experience with bisphosphonates and AKI revealed 72 Calcitonin is a peptide hormone secreted by the para- cases associated with IV zoledronate (60). In this cohort, follicular C cells of the thyroid gland. It inhibits osteoclast AKI developed approximately 2 months after initiation of bone resorption and increases urinary calcium excretion. therapy and after a mean of 2.4 doses of zoledronate. Risk Synthetic calcitonin (derived from salmon) is administered factors for AKI included advanced cancer, previous as 4–8 U/kg intramuscularly or subcutaneously every 6–8 bisphosphonate therapy, and use of nonsteroidal anti- hours. Calcitonin has a rapid onset of effect, leading to fl in ammatory agents. Most patients did not fully recover reductions in serum calcium within 2–6 hours after dosing. to baseline renal function, and 37.5% of patients required The calcium-lowering effects of calcitonin are modest and dialysis (60). transient, with a mean duration of effect of 2–4 days (76). On the basis of the renal toxicity of bisphosphonates, the Furthermore, repeated administration of calcitonin results American Society of Clinical Oncology developed dosing in downregulation of the calcitonin receptors on osteoclasts and monitoring guidelines (Table 1) and recommended and dimunition in effect (tachyphylaxis) (77). Overall, its that zoledronate be avoided in patients with a creatinine efficacy is poor, inducing normocalcemia in just one third , clearance 30 ml/min (61). Furthermore, serum creatinine of patients (78). Calcitonin’s major role may be as an adjunc- should be monitored before each dose of bisphosphonate tive agent with bisphosphonates in the treatment of severe, and the drug withheld in patients with worsening of renal symptomatic hypercalcemia when calcium levels must be function. Urine albumin excretion should be monitored at reduced rapidly (79). Side effects are minimal; nausea, vom- 3- to 6-month intervals, and the drug should be held if iting, and injection site pain are the most common. Overall, albuminuria develops (61). there is little evidence that calcitonin use has better efficacy Interestingly, ibandronate appears to have minimal to no over volume repletion and bisphosphonate therapy alone. renal toxicity, even in patients with underlying CKD (62– 64). However, more experience with this drug for the treat- ment of malignancy-associated hypercalcemia is needed Corticosteroids Corticosteroids are effective agents in the treatment of before firm conclusions on its renal safety can be made. hypercalcemia associated with malignancies that overpro- An increasingly recognized complication of bisphospho- duce calcitriol. This is due to their effect in inhibiting nate therapy is osteonecrosis of the jaw (ONJ) (65,66). The 1a-hydroxylase conversion of 25-hydroxyvitamin D to cal- precise etiology is not known, and the most common pre- citriol. Typically, these patients have underlying Hodgkin cipitating factors include dental extraction, long-term se- or non-Hodgkin lymphoma (80). The efficacy of cortico- quential use of IV pamidronate or zoledronate, smoking, steroids is based on case reports and thus the dosing and poor dental health (67). The absolute risk for ONJ is guidelines are uncertain; however, a reasonable regimen unknown, although it is estimated to occur in ,1:10,000 is IV hydrocortisone, 200–300 mg/d, for 3–5days(79). treated individuals and may be seen even less often in Typically, calcium levels are slow to decrease with corti- patients receiving shorter-term treatment. Diagnosis is based costeroid treatment alone. Patients who do respond with on clinical criteria that include pain and evidence of local normalization of their serum calcium can be transitioned infection. Management is largely supportive, with occa- to maintenance oral prednisone, 10–30 mg/d (81). sional need for surgical debridement. The condition can be prevented through good dental hygiene and avoidance of invasive dental procedures during bisphosphonate Hemodialysis fi therapy (68). For patients with acute hypercalcemia and signi cant AKI (especially in the setting of oliguria), saline-induced diuresis Gallium Nitrate may not be feasible and may lead to volume overload. In Gallium nitrate is approved for the treatment of cancer- these circumstances, hemodialysis using a very-low-calcium # related hypercalcemia. The cellular basis for this action is dialysate ( 1 mmol/L) is an option (82). For refractory unknown but appears to involve a reduction in osteoclast cases of hypercalcemia, several case reports describe the activity and bone remodeling (69–72). Clinically, gallium use of citrate anticoagulation to chelate calcium along with fi nitrate is administered by a continuous infusion at a dos- continuous venovenous hemodia ltration (83,84). age of 200 mg/m2 per day over 5 consecutive days. Re- ports comparing gallium to bisphosphonates (73,74) and Denosumab calcitonin (75) have shown equal or perhaps even superior Given the important role RANKL in the pathogenesis of efficacy in decreasing calcium levels. Adverse effects from bone metastases and secondarily in leading to hypercalcemia, treatment include hypocalcemia and hypophosphatemia, inhibition of this pathway is an attractive means of poten- the latter being observed in nearly 80% of patients. In tially treating forms of malignancy-associated hypercalcemia Clin J Am Soc Nephrol 7: 1722–1729, October, 2012 Hypercalcemia of Malignancy, Rosner and Dalkin 1727

due to bone calcium release. Denosumab is a fully human Disclosures monoclonal antibody that binds to RANKL and inhibits os- None. teoclast 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 References 1. Sargent JTS, Smith OP: Haematological emergencies managing postmenopausal women with osteoporosis. In clinical trials, fi in adults and children with haematological denosumab has shown ef cacy in preventing skeletal-related disorders. Br J Haematol 149: 465–477, 2010 events (such as bone metastases) in patients with prostate, 2. Stewart AF: Clinical practice. Hypercalcemia associated with breast, and other solid-organ cancers, as well as multiple cancer. N Engl J Med 352: 373–379, 2005 myeloma (85). Denosumab is well tolerated, with arthralgias 3. Ralston SH, Gallacher SJ, Patel U, Campbell J, Boyle IT: Cancer- associated hypercalcemia: morbidity and mortality. Clinical expe- as the most common side effect (86). Because RANKL is rience in 126 treated patients. Ann Intern Med 112: 499–504, 1990 expressed on B and T cells, there was some concern that 4. Burt ME, Brennan MF: Incidence of hypercalcemia and malig- denosumab would increase the risk for serious infections. nant neoplasm. Arch Surg 115: 704–707, 1980 The FDA has reported only a slightly increased incidence 5. Vassilopoulou-Sellin R, Newman BM, Taylor SH, Guinee VF: Incidence of hypercalcemia in patients with malignancy referred of serious infections (4.1% for denosumab and 3.3% for pla- to a comprehensive cancer center. Cancer 71: 1309–1312, 1993 cebo), with no increase in opportunistic infections seen (86). 6. Edelson GW, Kleerekoper M: Hypercalcemic crisis. Med Clin Of note, denosumab has been associated with a similar in- North Am 79: 79–92, 1995 cidence of osteonecrosis of the jaw as zoledronic acid (87). 7. Labriola L, Wallemacq P, Gulbis B, Jadoul M: The impact of the Denosumab is not cleared through renal mechanisms and assay for measuring albumin on corrected (‘adjusted’) calcium concentrations. Nephrol Dial Transplant 24: 1834–1838, 2009 does not require dosing adjustments for renal impairment. 8. Ijaz A, Mehmood T, Qureshi AH, Anwar M, Dilawar M, Hussain In clinical trials in patients with malignancies, the most I, Khan FA, Khan DA, Hussain S, Khan IA: Estimation of ionized common laboratory abnormality was hypocalcemia due to calcium, total calcium and albumin corrected calcium for the inhibition of bone resorption (88–90). This is not surprising diagnosis of hypercalcaemia of malignancy. J Coll Physicians Surg Pak 16: 49–52, 2006 given the mechanism of action of denosumab and leads to 9. Riancho JA, Arjona R, Sanz J, Olmos JM, Valle R, Barcelo´ JR, the obvious question of whether this drug can be used to Gonza´lez-Macı´as J: Is the routine measurement of ionized cal- treat malignancy-associated hypercalcemia. Hu et al. stud- cium worthwhile in patients with cancer? Postgrad Med J 67: ied patients with bisphosphonate-refractory malignancy- 350–353, 1991 associated hypercalcemia (91). These patients had solid 10. Mellis DJ, Itzstein C, Helfrich MH, Crockett JC: The skeleton: a multi-functional complex organ: the role of key signalling malignancies or myeloma and elevated corrected serum pathways in osteoclast differentiation and in bone resorption. calcium levels (.12.5 mg/dl) despite recent IV bisphospho J Endocrinol 211: 131–143, 2011 nate therapy. Patients received denosumab, 120 mg sub- 11. Hsu H, Lacey DL, Dunstan CR, Solovyev I, Colombero A, Timms cutaneously, every 4 weeks, with additional loading doses E, Tan HL, Elliott G, Kelley MJ, Sarosi I, Wang L, Xia XZ, Elliott R, Chiu L, Black T, Scully S, Capparelli C, Morony S, Shimamoto G, of 120 mg on days 8 and 15 of the first month. The primary fi Bass MB, Boyle WJ: Tumor necrosis factor receptor family end point was reaching a response de ned as a corrected member RANK mediates osteoclast differentiation and activation serum calcium level #11.5 mg/dl within the first 10 days induced by osteoprotegerin ligand. Proc Natl Acad Sci USA 96: after denosumab dosing. An interim analysis of this study 3540–3545, 1999 (n515) revealed that 80% of patients achieved a response 12. Hofbauer LC, Schoppet M: Clinical implications of the osteoprote- gerin/RANKL/RANK system for bone and vascular diseases. JAMA by day 10, with a median response time of 8 days and a 292: 490–495, 2004 response duration of 28 days. Furthermore, an ad hoc 13. Eghbali-Fatourechi G, Khosla S, Sanyal A, Boyle WJ, Lacey DL, pooled analysis of the clinical trials with denosumab Riggs BL: Role of RANK ligand in mediating increased bone re- (88–90) revealed that compared with zoledronic acid, de- sorption in early postmenopausal women. J Clin Invest 111: 1221–1230, 2003 nosumab significantly delayed the time to first on-study 14. Abou-Samra AB, Ju¨ppner H, Force T, Freeman MW, Kong XF, episodes of hypercalcemia (92). More work will be re- Schipani E, Urena P, Richards J, Bonventre JV, Potts JT Jr, et al: quired to understand the role of denosumab in the treat- Expression cloning of a common receptor for parathyroid ment of malignancy-associated hypercalcemia, but it holds hormone and parathyroid hormone-related peptide from rat much promise as a safe, more convenient (subcutaneous osteoblast-like cells: A single receptor stimulates intracellular accumulation of both cAMP and inositol trisphosphates and versus IV) therapy. increases intracellular free calcium. Proc Natl Acad Sci USA 89: 2732–2736, 1992 Summary 15. Sone S, Yano S: Molecular pathogenesis and its therapeutic mo- Patients with underlying malignancy frequently encounter dalities of lung cancer metastasis to bone. Cancer Metastasis Rev 26: 685–689, 2007 hypercalcemia. Indeed, hypercalcemia carries a poor prog- 16. Grill V, Ho P, Body JJ, Johanson N, Lee SC, Kukreja SC, Moseley nosis. Multiple cellular mechanisms underlie the increase in JM, Martin TJ: Parathyroid hormone-related protein: Elevated calcium concentration, and a proper evaluation to elucidate levels in both humoral hypercalcemia of malignancy and hy- the cause is essential to guide treatment. The mainstay of percalcemia complicating metastatic breast cancer. J Clin initial management is aggressive fluid resuscitation, with the Endocrinol Metab 73: 1309–1315, 1991 17. Sourbier C, Massfelder T: Parathyroid hormone-related protein in addition of loop diuretics if volume overload is present. In human renal cell carcinoma. Cancer Lett 240: 170–182, 2006 addition, antiresorptive agents, primarily the bisphospho- 18. Yoshimoto K, Yamasaki R, Sakai H, Tezuka U, Takahashi M, nates, can aid in the treatment of these patients. Although the Iizuka M, Sekiya T, Saito S: Ectopic production of parathyroid physician must bear in mind potential pitfalls with the use of hormone by small cell lung cancer in a patient with hypercal- cemia. J Clin Endocrinol Metab 68: 976–981, 1989 these medications, prompt and assertive treatment of hyper- 19. Iguchi H, Miyagi C, Tomita K, Kawauchi S, Nozuka Y, Tsuneyoshi calcemia in patients with cancer can be successful and life- M, Wakasugi H: Hypercalcemia caused by ectopic produc- saving. tion of parathyroid hormone in a patient with papillary 1728 Clinical Journal of the American Society of Nephrology

adenocarcinoma of the thyroid gland. J Clin Endocrinol Metab 43. Body JJ, Lortholary A, Romieu G, Vigneron AM, Ford J: A dose- 83: 2653–2657, 1998 finding study of zoledronate in hypercalcemic cancer patients. 20. Nussbaum SR, Gaz RD, Arnold A: Hypercalcemia and ectopic J Bone Miner Res 14: 1557–1561, 1999 secretion of parathyroid hormone by an ovarian carcinoma with 44. Pecherstorfer M, Herrmann Z, Body JJ, Manegold C, Degardin M, rearrangement of the gene for parathyroid hormone. N Engl J Med Clemens MR, Thu¨rlimann B, Tubiana-Hulin M, Steinhauer EU, 323: 1324–1328, 1990 van Eijkeren M, Huss HJ, Thie´baud D: Randomized phase II trial 21. Chen YC, Sosnoski DM, Mastro AM: Breast cancer metastasis to comparing different doses of the bisphosphonate ibandronate in the bone: Mechanisms of bone loss. Breast Cancer Res 12: 215– the treatment of hypercalcemia of malignancy. J Clin Oncol 14: 226, 2010 268–276, 1996 22. Clines GA: Mechanisms and treatment of hypercalcemia of ma- 45. Nussbaum SR, Younger J, Vandepol CJ, Gagel RF, Zubler MA, lignancy. Curr Opin Endocrinol Diabetes Obes 18: 339–346, 2011 Chapman R, Henderson IC, Mallette LE: Single-dose intravenous 23. Abildgaard N, Brixen K, Eriksen EF, Kristensen JE, Nielsen JL, therapy with pamidronate for the treatment of hypercalcemia of Heickendorff L: Sequential analysis of biochemical markers of malignancy: Comparison of 30-, 60-, and 90-mg dosages. Am J bone resorption and bone densitometry in multiple myeloma. Med 95: 297–304, 1993 Haematologica 89: 567–577, 2004 46. Dodwell DJ, Howell A, Morton AR, Daley-Yates PT, Hoggarth 24. Schilling T, Pecherstorfer M, Blind E, Leidig G, Ziegler R, Raue F: CR: Infusion rate and pharmacokinetics of intravenous pamidr- Parathyroid hormone-related protein (PTHrP) does not regulate onate in the treatment of tumour-induced hypercalcaemia. 1,25-dihydroxyvitamin D serum levels in hypercalcemia of ma- Postgrad Med J 68: 434–439, 1992 lignancy. J Clin Endocrinol Metab 76: 801–803, 1993 47. Gurney H, Grill V, Martin TJ: Parathyroid hormone-related pro- 25. Seymour JF, Gagel RF: Calcitriol: the major humoral mediator of tein and response to pamidronate in tumor-induced hypercal- hypercalcemia in Hodgkin’s disease and non-Hodgkin’s lym- cemia. Lancet 341: 1611–1613, 1993 phomas. Blood 82: 1383–1394, 1993 48. Walls J, Ratcliffe WA, Howell A, Bundred NJ: Response to in- 26. Hatake K, Uwai M, Ohtsuki T, Tomizuka H, Izumi T, Yoshida M, travenous bisphosphonate therapy in hypercalcaemic patients Miura Y: Rare but important adverse effects of all-trans retinoic with and without bone metastases: the role of parathyroid hor- acid in acute promyelocytic leukemia and their management. Int mone-related protein. Br J Cancer 70: 169–172, 1994 J Hematol 66: 13–19, 1997 49. Major P,Lortholary A, Hon J, Abdi E, Mills G, Menssen HD, Yunus 27. Castelli I, Steiner LA, Kaufmann MA, Drop LJ: Renovascular re- F, Bell R, Body J, Quebe-Fehling E, Seaman J: Zoledronic acid is sponses to high and low perfusate calcium steady-state experi- superior to pamidronate in the treatment of hypercalcemia of ments in the isolated perfused rat kidney with baseline vascular malignancy: A pooled analysis of two randomized, controlled tone. JSurgRes61: 51–57, 1996 clinical trials. J Clin Oncol 19: 558–567, 2001 28. Hebert SC, Brown EM: The scent of an ion: Calcium-sensing and 50. Pecherstorfer M, Steinhauer EU, Rizzoli R, Wetterwald M, its roles in health and disease. Curr Opin Nephrol Hypertens 5: Bergstro¨m B: Efficacy and safety of ibandronate in the treatment 45–53, 1996 of hypercalcemia of malignancy: A randomized multicentric 29. Khanna A: Acquired nephrogenic diabetes insipidus. Semin comparison to pamidronate. Support Care Cancer 11: 539–547, Nephrol 26: 244–248, 2006 2003 30. Sleeboom HP, bijvoet OLM, Van Oosterom AT, Gleed JH, 51. Zojer N, Keck AV, Pecherstorfer M: Comparative tolerability of O’Riordan JL: Comparison of intravenous (3-amino-1-hydrox- drug therapies for hypercalcaemia of malignancy. Drug Saf 21: ypropylidene)-1-1-bisphosphonate and volume repletion in tu- 389–406, 1999 mour-induced hypercalcemia. Lancet 2: 239–243, 1983 52. Markowitz GS, Appel GB, Fine PL, Fenves AZ, Loon NR, 31. Kacprowicz RF, Lloyd JD: Electrolyte complications of malig- Jagannath S, Kuhn JA, Dratch AD, D’Agati VD: Collapsing focal nancy. Hematol Oncol Clin North Am 24: 553–565, 2010 segmental glomerulosclerosis following treatment with high- 32. LeGrand SB, Leskuski D, Zama I: Narrative review: Furosemide dose pamidronate. J Am Soc Nephrol 12: 1164–1172, 2001 for hypercalcemia: An unproven yet common practice. Ann In- 53. Barri YM, Munshi NC, Sukumalchantra S, Abulezz SR, Bonsib SM, tern Med 149: 259–263, 2008 Wallach J, Walker PD: Podocyte injury associated glomer- 33. Nielsen SP, Andersen O, Steven KE: and calcium me- ulopathies induced by pamidronate. Kidney Int 65: 634–641, 2004 tabolism during prolonged furosemide (Lasix) administration to 54. Desikan R, Veksler Y, Raza S, Stokes B, Sabir T, Li ZJ, Jagannath S: normal rats. Acta Pharmacol Toxicol (Copenh) 27: 469–479, 1969 Nephrotic proteinuria associated with high-dose pamidronate in 34. Toft H, Roin J: Effect of frusemide administration on calcium multiple myeloma. Br J Haematol 119: 496–499, 2002 excretion. BMJ 1: 437–438, 1971 55. Markowitz GS, Fine PL, D’agati VD: Nephrotic syndrome after 35. Perazella MA, Markowitz GS: Bisphosphonate nephrotoxicity. treatment with pamidronate. Am J Kidney Dis 39: 1118–1122, Kidney Int 74: 1385–1393, 2008 2002 36. Lin JH: Bisphosphonates: A review of their pharmacokinetic 56. Bodmer M, Amico P, Mihatsch MJ, Haschke M, Kummer O, properties. Bone 18: 75–85, 1996 Kra¨henbu¨hl S, Mayr M: Focal segmental glomerulosclerosis as- 37. Russell RGG, Rogers MJ: Bisphosphonates: From the laboratory sociated with long-term treatment with zoledronate in a mye- to the clinic and back again. Bone 25: 97–106, 1999 loma patient. Dial Transplant 22: 2366–2370, 2007 38. Amin D, Cornell SA, Gustafson SK, Needle SJ, Ullrich JW, Bilder 57. Markowitz GS, Fine PL, Stack JI, Kunis CL, Radhakrishnan J, GE, Perrone MH: Bisphosphonates used for the treatment of Palecki W, Park J, Nasr SH, Hoh S, Siegel DS, D’Agati VD: Toxic bone disorders inhibit squalene synthase and cholesterol bio- acute tubular necrosis following treatment with zoledronate synthesis. J Lipid Res 33: 1657–1663, 1992 (Zometa). Kidney Int 64: 281–289, 2003 39. Rogers MJ, Gordon S, Benford HL, Coxon FP, Luckman SP, 58. Rosen LS, Gordon D, Kaminski M, Howell A, Belch A, Mackey J, Monkkonen J, Frith JC: Cellular and molecular mechanisms of Apffelstaedt J, Hussein M, Coleman RE, Reitsma DJ, Seaman JJ, action of bisphosphonates. Cancer 88[Suppl]: 2961–2978, 2000 Chen BL, Ambros Y: Zoledronic acid versus pamidronate in the 40. Hiroi-Furuya E, Kameda T, Hiura K, Mano H, Miyazawa K, treatment of skeletal metastases in patients with breast cancer or Nakamaru Y, Watanabe-Mano M, Okuda N, Shimada J, osteolytic lesions of multiple myeloma: A phase III, double-blind, Yamamoto Y, Hakeda Y, Kumegawa M: Etidronate (EHDP) in- comparative trial. Cancer J 7: 377–387, 2001 hibits osteoclastic-bone resorption, promotes apoptosis and 59. Rosen LS, Gordon D, Tchekmedyian S, Yanagihara R, Hirsh V, disrupts actin rings in isolate-mature osteoclasts. Calcif Tissue Int Krzakowski M, Pawlicki M, de Souza P, Zheng M, Urbanowitz G, 64: 219–223, 1999 Reitsma D, Seaman JJ: Zoledronic acid versus placebo in the 41. Mackie PS, Fisher JL, Zhou H, Choong PF: Bisphosphonates treatment of skeletal metastases in patients with lung cancer and regulate cell growth and gene expression in the UMR 106-01 other solid tumors: A phase III, double-blind, randomized trial— clonal rat osteosarcoma cell line. Br J Cancer 84: 951–958, 2001 the Zoledronic Acid Lung Cancer and Other Solid Tumors Study 42. Purohit OP, Radstone CR, Anthony C, Kanis JA, Coleman RE: A Group. J Clin Oncol 21: 3150–3157, 2003 randomised double-blind comparison of intravenous pamidro- 60. Chang JT, Green L, Beitz J: Renal failure with the use of nate and clodronate in the hypercalcaemia of malignancy. zoledronic acid. N Engl J Med 349: 1676–1679, discussion Br J Cancer 72: 1289–1293, 1995 1676–1679, 2003 Clin J Am Soc Nephrol 7: 1722–1729, October, 2012 Hypercalcemia of Malignancy, Rosner and Dalkin 1729

61. Kyle RA, Yee GC, Somerfield MR, Flynn PJ, Halabi S, Jagannath S, 80. Bilezikian JP: Management of acute hypercalcemia. N Engl J Med Orlowski RZ, Roodman DG, Twilde P, Anderson K; American 326: 1196–1203, 1992 Society of Clinical Oncology: American Society of Clinical On- 81. Santarpia L, Koch CA, Sarlis NJ: Hypercalcemia in cancer pa- cology 2007 clinical practice guideline update on the role of tients: Pathobiology and management. Horm Metab Res 42: 153– bisphosphonates in multiple myeloma. J Clin Oncol 25: 2464– 164, 2010 2472, 2007 82. Argile´s A, Kerr PG, Canaud B, Flavier JL, Mion C: Calcium ki- 62. Heidenreich A, Elert A, Hofmann R: Ibandronate in the treatment netics and the long-term effects of lowering dialysate calcium of prostate cancer associated painful osseous metastases. Pros- concentration. Kidney Int 43: 630–640, 1993 tate Cancer Prostatic Dis 5: 231–235, 2002 83. Sra´mek V, Nova´kI,Matĕjovic M, Rokyta R, Nalos M, Hora P, 63. Body JJ, Diel IJ, Tripathy D, Bergstrom B: Intravenous ibandronate Pittrova´ H: Continuous venovenous hemodiafiltration does not affect time to renal function deterioration in patients (CVVHDF) with citrate anticoagulation in the treatment of a with skeletal metastases from breast cancer: Phase III trial results. patient with acute renal failure, hypercalcemia, and thrombo- Eur J Cancer Care (Engl) 15: 299–302, 2006 cytopenia. Intensive Care Med 24: 262–264, 1998 64. Bergner R, Henrich DM, Hoffmann M, et al: Renal safety of 84. Kindgen-Milles D, Kram R, Kleinekofort W, Morgera S: Treatment of ibandronate in multiple myeloma patients with renal de- severe hypercalcemia using continuous renal replacement therapy terioration. Cancer Treat Rev 31[suppl 1]: S45, 2005 with regional citrate anticoagulation. ASAIO J 54: 442–444, 2008 65. Carter GD, Goss AN: Bisphosphonates and avascular necrosis of 85. Castellano D, Sepulveda JM, Garcı´a-Escobar I, Rodriguez- the jaws. Aust Dent J 48: 268, 2003 Antolı´n A, Sundlo¨v A, Cortes-Funes H: The role of RANK-ligand 66. Hellstein JW, Kalmar JR: For the dental patient. Osteoporosis inhibition in cancer: the story of denosumab. Oncologist 16: medications and the risk of developing jaw complications. JAm 136–145, 2011 Dent Assoc 138: 692, 2007 86. Background document for meeting of advisory committee for 67. Carneiro E, Vibhute P, Montazem A, Som PM: Bisphosphonate- reproductive health drugs. FDA report, August 13, 2009. Avail- associated mandibular osteonecrosis. AJNR Am J Neuroradiol able at http://www.fda.gov. Accessed 2/29/2012 27: 1096–1097, 2006 87. Saad F, Brown JE, Van Poznak C, Ibrahim T, Stemmer SM, Stopeck 68. American Dental Association Council on Scientific Affairs: AT, Diel IJ, Takahashi S, Shore N, Henry DH, Barrios CH, Facon T, Dental management of patients receiving oral bisphosphonate Senecal F, Fizazi K, Zhou L, Daniels A, Carriere P, Dansey R: therapy: Expert panel recommendations. JAmDentAssoc137: Incidence, risk factors, and outcomes of osteonecrosis of the jaw: 1144–1150, 2006 Integrated analysis from three blinded active-controlled phase III 69. Repo MA, Bockman RS, Betts F, Boskey AL, Alcock NW, Warrell trials in cancer patients with bone metastases. Ann Oncol 23: RP Jr: Effect of gallium on bone mineral properties. Calcif Tissue 1341–1347, 2012 Int 43: 300–306, 1988 88. Stopeck AT, Lipton A, Body J-J, Steger GG, Tonkin K, de Boer RH, 70. Bockman RS: Studies on the mechanism of action of gallium Lichinitser M, Fujiwara Y, Yardley DA, Viniegra M, Fan M, Jiang Q, nitrate. Semin Oncol 18[Suppl 5]: 21–25, 1991 Dansey R, Jun S, Braun A: Denosumab compared with zoledronic 71. Warrell RP Jr, Bockman RS, Coonley CJ, Isaacs M, Staszewski H: acid for the treatment of bone metastases in patients with advanced Gallium nitrate inhibits calcium resorption from bone and is ef- breast cancer: a randomized, double-blind study. J Clin Oncol 28: fective treatment for cancer-related hypercalcemia. J Clin Invest 5132–5139, 2010 73: 1487–1490, 1984 89. Henry DH, Costa L, Goldwasser F, Hirsh V,Hungria V,Prausova J, 72. Hall TJ, Chambers TJ: Gallium inhibits bone resorption by a direct Scagliotti GV, Sleeboom H, Spencer A, Vadhan-Raj S, von Moos effect on osteoclasts. Bone Miner 8: 211–216, 1990 R, Willenbacher W, Woll PJ, Wang J, Jiang Q, Jun S, Dansey R, 73. Cvitkovic F, Armand J-P, Tubiana-Hulin M, Rossi JF, Warrell RP Jr: Yeh H: Randomized, double-blind study of denosumab versus Randomized, double-blind, phase II trial of gallium nitrate zoledronic acid in the treatment of bone metastases in patients compared with pamidronate for acute control of cancer-related with advanced cancer (excluding breast and prostate cancer) or hypercalcemia. Cancer J 12: 47–53, 2006 multiple myeloma. J Clin Oncol 29: 1125–1132, 2011 74. Warrell RP Jr, Murphy WK, Schulman P, O’Dwyer PJ, Heller G: A 90. Fizazi K, Carducci M, Smith M, Damia˜o R, Brown J, Karsh L, randomized double-blind study of gallium nitrate compared with Milecki P,Shore N, Rader M, Wang H, Jiang Q, Tadros S, Dansey R, etidronate for acute control of cancer-related hypercalcemia. Goessl C: Denosumab versus zoledronic acid for treatment of bone J Clin Oncol 9: 1467–1475, 1991 metastases in men with castration-resistant prostate cancer: A 75. Warrell RP Jr, Israel R, Frisone M, Snyder T, Gaynor JJ, Bockman randomised, double-blind study. Lancet 377: 813–822, 2011 RS: Gallium nitrate for acute treatment of cancer-related hyper- 91. Hu MI, Gucalp R, Insogna K, et al: Denosumab for treatment of calcemia. A randomized, double-blind comparison to calcito- hypercalcemia of malignancy in patients with solid tumors or nin. Ann Intern Med 108: 669–674, 1988 hematological malignancies refractory to IV bisphosphonates: 76. Huang CL, Sun L, Moonga BS, Zaidi M: Molecular physiology a single-arm multicenter study. Presented at the 53rd American and pharmacology of calcitonin. Cell Mol Biol (Noisy-le-grand) Society of Henatology Annual Meeting and Exposition, San 52: 33–43, 2006 Diego, CA, December 10–13, Abstract 2483 77. Purdue BW, Tilakaratne N, Sexton PM: Molecular pharmacol- 92. Diel IJ, Body J-J, Stopeck A, et al: Effect of denosumab treatment ogy of the calcitonin receptor. Receptors Channels 8: 243–255, on prevention of hypercalcemia of malignancy in cancer patients 2002 with metastatic bone disease. Presented at the European Multi- 78. Davidson TG: Conventional treatment of hypercalcemia of ma- disciplinary Cancer Congress, Stockholm, Sweden, September lignancy. Am J Health Syst Pharm 58[Suppl 3]: S8–S15, 2001 23–27, 2011 79. Mathur M, Sykes JA, Saxena VR, et al: Treatment of acute lym- phoblastic leukemia-induced extreme hypercalcemia with pa- midronate and calcitonin. Pediatr Crit Care Med 4: 252–255, Published online ahead of print. Publication date available at www. 2003 cjasn.org.